Immune Netw. 2016 Feb;16(1):13-25. English.
Published online Feb 25, 2016.
Copyright © 2016 The Korean Association of Immunologists
Review

Induction of CD4+ Regulatory and Polarized Effector/helper T Cells by Dendritic Cells

Manfred B. Lutz
    • Institute of Virology and Immunobiology, University of Würzburg, 97078 Würzburg, Germany.
Received November 28, 2015; Revised January 15, 2016; Accepted January 18, 2016.

This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Dendritic cells (DCs) are considered to play major roles during the induction of T cell immune responses as well as the maintenance of T cell tolerance. Naive CD4+ T cells have been shown to respond with high plasticity to signals inducing their polarization into effector/helper or regulatory T cells. Data obtained from in vitro generated bone-marrow (BM)-derived DCs as well as genetic mouse models revealed an important but not exclusive role of DCs in shaping CD4+ T cell responses. Besides the specialization of some conventional DC subsets for the induction of polarized immunity, also the maturation stage, activation of specialized transcription factors and the cytokine production of DCs have major impact on CD4+ T cells. Since in vitro generated BM-DCs show a high diversity to shape CD4+ T cells and their high similarity to monocyte-derived DCs in vivo, this review reports data mainly on BM-DCs in this process and only touches the roles of transcription factors or of DC subsets, which have been discussed elsewhere. Here, recent findings on 1) the conversion of naive into anergic and further into Foxp3 regulatory T cells (Treg) by immature DCs, 2) the role of RelB in steady state migratory DCs (ssmDCs) for conversion of naive T cells into Foxp3+ Treg, 3) the DC maturation signature for polarized Th2 cell induction and 4) the DC source of IL-12 for Th1 induction are discussed.

Keywords
Dendritic cells; CD4+ T cells; Polarization; Regulatory T cells; Anergy

INTRODUCTION

Adaptive immune responses are characterized by the activation and differentiation of CD4+ T cells into distinct effector T helper (Th) cell subsets but also induced regulatory T cells (iTregs). The latter support pre-existing thymus-derived, natural regulatory T cells (nTregs) to allow but at the same time also control the effector cell responses against pathogens and prevent autoreactive T cells from autoimmune attack. While effector CD4+ T cell responses are characterized by the polarization of naive T cells into Th1, Th2, Th9 or Th17 subsets, iTregs of either Foxp3+ or Foxp3 IL-10+ (Tr1) subtypes can be polarized from either naive T cells or already polarized effector Th1 or Th2 cells (1, 2, 3).

It appears that repetitive stimulation of CD4+ Th1 or Th2 cell leads to a loss of their effector cytokine production but an increase and finally dominance of IL-10 release as shown in mice (1) but also with cultured human T cell clones (4). The induction of Th17 effector or Foxp3+ iTregs from naive T cells has been described to depend on specific cytokines. While IL-6 is necessary for generation of both T cell phenotypes, TGF-β plays a pivotal role in generating Foxp3+ iTregs while in its absence Th17 cell development is observed (5). However, it remains unclear which type of DCs and DC-derived factors would induce all of these tolerogenic (Foxp3+ iTregs or Foxp3 IL-10+ Tr1 cells) or immunogenic T cell polarization programs (Th1, Th2 and Th17 cells). While recent efforts concentrated on the role of conventional DC subsets and their transcription factors for CD4+ T cell polarization (6, 7, 8), not much has been investigated on the DC phenotype or maturation stage such as non-migratory immature or migratory semi-mature or migratory fully mature DCs. Since quantitative aspects of T cell stimulation like peptide concentrations, TCR signal intensity or quality and quantity of costimulation play considerable roles for Th polarization (9, 10), we believe that studying DC maturity may also help to contribute to the understanding of this aspect in Th cell polarization (11) and regulatory T cell induction (3). The strength of DC maturity/activation, represented by the surface expression of MHC and diverse costimulatory molecules as well as cytokines (12), will also be translated into the strength of T cell stimulation (13).

For the priming of Th cells in vivo DCs play a central role due to their antigen presentation capacity together with highly expressed costimulatory molecules and the production of pro-inflammatory cytokines. One additional key feature of DCs is their migratory capacity from infection sites to the draining lymph nodes. DC migration requires coordinate mechanisms of soluble and matrix-associated CCL19 and CCL21 chemokines recognized by the receptor CCR7 (14, 15). Antigen capture and migration of blood DCs into lymphoid organs has been observed during immune responses but is less well understood (16). Under inflammatory conditions, monocyte-derived DCs infiltrating into atherosclerotic plaques may direct i/nTreg expansion by secretion of CCL17 chemokine (17). Since CCR7-dependent migration of DCs also occurs under steady state conditions (14), the question remained whether in mice such ssmDCs induce tolerance in naive CD4+ T cells by inducing anergy, as observed in vitro (18), or induce deletion, as observed for CD8+ T cells (19), or by converting the naive cells into iTregs.

For the latter it remained to be determined, whether i) Foxp3+ iTregs would be induced as shown by using an osmotic mini-pump system (20) or whether Tr1 cells would result by employing endogenous tolerizing migratory DCs in an asthma model found by others (21) or as we observed by adoptive transfer of TNF-matured DCs in the experimental autoimmune encephalomyelitis (EAE) model (22). Finally, the question remained whether anergic T cells were stably anergic and non-suppressive or whether certain DC-derived signals may further polarize them into another phenotype such as Tregs.

Our lab addressed these topics mainly by using BM-DCs generated with GM-CSF (23) (Fig. 1) where immature and mature stages can be easily generated in contrast to ex vivo isolated spleen DCs or the in vivo-counterpart of BM-DCs which are inflammatory monocyte-derived DCs (24, 25). Since the impact of DC maturity, expression of costimulatory molecules and of IL-10 production on the induction of CD4+ Treg has been reviewed recently (3), we will not further elucidate on this topic here.

Figure 1
Induction of CD4+ T cell anergy, Treg subsets and polarized Th1/Th2 responses by DC can be directed by their maturation stages and cytokines. Immature DCs induce antigen-specific T cell anergy in naive T cells in the absence of TGF-β, but induce Foxp3+ iTregs when TGF-β is present. A second stimulation of non-regulatory anergized T cells by immature DCs generates regulatory IL-10+ Tr1 cells. A similar T cell phenotype of regulatory IL-10+ Tr1 is generated by repetitive stimulation with semi-mature BM-DCs generated by maturation with TNF or T. brucei VSG antigens characterized by a lack of cytokine production. A single stimulation with these DCs in the absence of TGF-β and IL-12 induces a Th2 phenotype that is lost upon repetitive stimulation. In vivo steady state migratory DCs (ssmDCs) resemble TNF-matured BM-DCs but capture TGF-β on their surface, thereby inducing naive T cell conversion into Foxp3+ iTreg specific for self-antigens. DCs matured with high doses of LPS or CgG oligonucleotides reach a full maturation stage characterized by cytokine release including IL-12p70 that leads to Th1 induction.

CONVERSION OF NAIVE INTO ANERGIC AND FURTHER INTO Foxp3 Tr1 CELLS BY IMMATURE DCs

T cell tolerance mechanisms include intrinsic and extrinsic mechanisms. Intrinsic control of T cells includes the induction of T cell anergy and T cell deletion, while extrinsic control is mediated by the activity of regulatory T cells (Tregs). The active role of Tregs for extrinsic T cell tolerance has been widely studied and also the result of T cell deletion appears obvious. In contrast, although the molecular details, how anergy is induced and maintained is increasingly understood (26, 27), an active functional role for anergic T cells for tolerance or any other usefulness for maintaining such cells in the immune system had not been described.

The term clonal anergy was used to define a specific functional unresponsive state of CD4+ T lymphocytes (initially characterized in Th1 T cell clones, i.e. previously activated T cells) achieved by a strong TCR/CD3 signal 1 in the absence of CD28 costimulation as a signal 2 (28, 29). Although a variety of experimental approaches have been used to induce T cell anergy in vitro, the most consistent hallmarks are defective IL-2 production and lack of proliferation upon TCR/CD3-mediated restimulation even in the presence of costimulation (29, 30, 31).

The phenotype of clonal T cell anergy can be reversed by addition of exogenous IL-2 and CD4+ T cell clones express high-affinity IL-2 receptors (IL-2R) (29). Reversal of CD4+ T cell anergy can also be performed by exposure to polyclonal stimuli circumventing TCR signaling by phorbol 12-myristate 13-acetate (PMA) and the calcium ionophores. However, in contrast to antigen-experienced T cell clones, naive CD4+ T cells have been reported to be resistant to anergy induction in vivo as well as in vitro upon TCR/CD3 stimulation alone in the absence of any secondary signals or costimulation (32).

Indeed, naive CD4+ T cells appear to be dependent on B7 costimulation-driven CTLA-4 engagement for anergy induction (33). CTLA-4 expression is obligatory for tolerance induction in vivo, also termed adaptive tolerance (29). This was observed in studies using T cells derived from CTLA-4−/− mice or antibody-mediated blocking experiments (34, 35). The exact role and signaling mechanisms of CTLA-4 for the induction of T cell anergy is still a matter of debate (36). Earlier reports suggested that CTLA-4 signaling prevents cell cycle progression through regulation of the cyclin-dependent kinase (cdk) inhibitors p27Kip1 and p21Cip1 (34, 37), although this may not be a strict requirement (38). Engagement of CTLA-4 has also been shown to block IL-2 production and IL-2R expression at least in part through decreased NFAT translocation to the nucleus (37, 39, 40).

Conversely, anergy induction in antigen-experienced T cells can be induced by Ca2+/calcineurin-dependent signaling through ionomycin only, thereby triggering downstream NFATc2 but not its transcriptional binding partner AP-1 (31, 41, 42). An NFAT-dependent transcriptional program inducing various anergy-associated genes was identified, which crucially contribute to anergy induction in vitro and in vivo (31, 41, 43, 44, 45). Several analyses revealed a dominant role for the early growth response genes 2 (Egr2) and Egr-3 as markers of clonal T cell anergy in vitro and murine anergy models (46, 47, 48, 49, 50). Interestingly, Egr-2 has been reported also to support tolerogenic functions in DCs (51).

After anergy induction the question remains which functional role anergic T cells might have since they appear to persist in vivo for relatively long periods of time and can potentially reverse their functional unresponsive state (49, 52). It was found that anergic T cells acquire the ability to produce anti-inflammatory cytokines such as IL-10 and suppress naive T cell responses (53, 54, 55). However, which signaling pathways or DC-derived instructive molecules are needed to induce IL-10 and regulatory function in anergic T cells remains unknown so far.

T cell anergy induction can be achieved by antigen presentation from immature DCs (12), similar to the generation of Foxp3+ Tregs (3) (Fig. 1). Using human monocyte-derived DCs we could show previously that naive allogeneic T cells required two rounds of stimulation in vitro to become anergic but without the capacity to release IL-10 or suppressing other T cell responses (18). This may be explained by the source of T cells used for the experiments, i.e. human peripheral blood versus murine lymphoid organs. Thus, while murine T cells receive tonic TCR signals in lymhoid organs increasing their responsiveness to secondary TCR engagements, this tonic activation state is shut off in human blood T cells an may require restoration before these T cells are fully functional (56). A single intravenous injection of soluble antigens induced T cell anergy as observed initially by neo-self antigen expression or superantigen injection (57, 58, 59, 60) or more recently in peptide-specific systems (61). Anergy induction and subsequent IL-10 production have been observed by these anergic T cells after repetitive intravenous injections of peptides, most likely captured and presented by immature DCs in this setting (62). However, the molecular requirements for an anergy-to-Tr1 switch had not been described.

To answer the latter question, we used immature BM-DCs to induce T cell anergy in vitro. We stimulated naive TCR-transgenic CD4+ CD25 OT-II T cells with immature DCs in the presence of OVA peptide antigen in vitro. In the absence of TGF-β the induction of Foxp3+ iTregs could not be observed. Instead a single stimulation with immature DCs resulted in anergic, non-regulatory Egr2+ IL-10 CTLA-4+ CD25low T cells. Of note, immature DCs were superior in CTLA-4 induction as compared to mature DCs. A second stimulation of the anergized T cells with immature DCs thus triggered CD28 and CTLA-4 and by using blocking antibody Fab fragments our data indicate that both simultaneous signals are required to convert the anergic cells into regulatory T cells. The resulting population resembled Tr1 cells since they appeared as proliferating Egr2+ IL-10+ CTLA-4+ CD25high cells with regulatory capacity. Suppressor activity was facilitated by highly effective IL-2 deprivation, enabled through CD28-mediated CD25 upregulation and the simultaneous CTLA4-mediated inhibition of nuclear translocation of NFATc1 and block of IL-2 production. Thus, Tr1 cells proliferated at the expense of IL-2 produced by effector T cells without producing IL-2 themselves. Together, two rounds of antigen-specific stimulation of naive T cells by immature DCs providing moderate CD80/CD86 signals in the absence of TGF-β induce IL-10+ Tr1 cells (63). This notion adds to the current opinion that Tr1 cells can be generated from repetitively stimulated Th1 or Th2 cells (1, 22) as mentioned above but also from anergic T cells, however by different types of DCs (Fig. 1).

CONVERSION OF NAIVE CD4+ T CELLS INTO Foxp3+ iTregs BY RelB+ ssm DCs

In addition to their role in pathogen defense, migratory dendritic cells (DCs) are also critical for maintaining tolerance to self-antigens (64). Although the etiology of most autoimmune diseases remains obscure, abundant progress in this field has been made. It is now clear that immature DCs have tolerogenic properties by inducing T cell anergy or Tregs in vitro and in vivo (3). Especially the identification and characterization of Tregs has opened a new area of research, which promises to acquire sufficient knowledge for the development of new strategies against autoimmunity (65). To promote antigen-specific tolerance, DCs must capture, process and present self-antigens in a "steady state" phenotype within lymphoid tissues. This can occur either by lymph node resident immature DCs that capture soluble antigens through the lymph node reticular conduit system (66) and thus may induce T cell deletion, anergy or Tregs. In addition, also ssmDC display a partially mature (semi-mature) phenotype (Fig. 1, ssmDCs). They express CCR7 to migrate and transport self-antigens from peripheral organs such as the skin to the draining lymph nodes under homeostatic conditions (12). A murine transgenic model expressing OVA as a neo-self-antigen in the epidermis (K5-mOVA) showed that this antigen is carried by ssmDCs displaying a semi-mature phenotype and is then cross-presented for CD8+ T cell deletion or de novo conversion of naive CD4+ T cells into Foxp3+ Tregs (19, 67).

The question remained which transcription factor in DCs would force them to induce iTregs in vivo when self-antigens are presented by ssmDCs under physiological conditions. Members of the NF-kB family such as RelA, RelB and c-Rel have been associated with inflammation or immunogenicity for many cell types (68). However, the functional role of these molecules in tolerogenic DCs is not fully understood. Tolerance to self-antigens expressed in peripheral organs is maintained by CD4+ CD25+ Foxp3+ Tregs, which are generated as a result of thymic selection (nTregs) or peripheral induction (iTregs). We demonstrated that ssmDCs from the skin mediated iTreg conversion in draining lymph nodes of mice. These DCs displayed a partially mature MHC IIint CD86int CD40hi CCR7+ phenotype, used endogenous TGF-β/latency-associated peptide (LAP) complexes for conversion of naive T cells into Foxp3+ iTregs and showed nuclear RelB translocation. Heterozygous deficiency of the alternative NF-κB signaling pathway (RelB/p52) reduced steady state migration of DCs. These DCs transported and directly presented soluble OVA provided by subcutaneously implanted osmotic mini-pumps, as well as cell-associated epidermal OVA in transgenic K5-mOVA mice to CD4+ OVA-specific TCR-transgenic OT-II T cells. The Langerin+ dermal DCs subset, but not epidermal Langerhans cells, mediated conversion of naive OT-II x RAG-1−/− T cells into proliferating CD4+ CD25+ Foxp3+ Tregs. Thus, these data showed that Langerin+ dermal ssmDCs mediate peripheral iTreg conversion for epidermal self-antigen in skin-draining lymph nodes via RelB and TGF-β (67, 69, 70). Of note, homozygous RelB−/− mice lack peripheral lymph nodes (71), which does not allow to study ssmDCs in these mice and the results we obtained from heterozygous RelB+/− (67) mice may still involve indirect effects from other cell types. However, the efficient conversion of CD4+ T cells to Foxp3+ iTreg by Langerin+ ssmDCs was corroborated by antibody-mediated specific targeting of the self-antigen myelin oligodendrocyte glycoprotein (MOG) to different ssmDC subtypes (72). More recently, also a critical role for the classical NFκB pathway for ssmDCs and tolerance has been found as demonstrated by spontaneous development of autoimmunity in mice with specific IKKβ deficiency in CD11c+ cells (73).

The phenotype and transcriptional signature of TNF-matured BM-DCs generated with GM-CSF in vitro is highly similar as compared to ssmDCs from peripheral skin-draining lymph nodes. Both types show only a moderate to high expression of MHC II and costimulatory with the complete absence of detectable cytokine production, but with the exception to express LAP on their cell surface, which captures TGF-β to store it in its inactive form. Whether ssmDCs produce TGF-β by themselves, is unclear. However, the release of active TGF-β from ssmDCs by αVβ8 integrins is critical to maintain self-tolerance in mice (74). Thus, despite a similar semi-mature phenotype of TNF-matured BM-DC and ssmDCs these cells may be functionally distinguished by the capacity to provide TGF-β or not (Fig. 1).

INFLAMMATORY GENE SIGNATURES IN DCs MARK THEIR Th2 PROGRAMMING CAPACITIES

Helminths and other parasites represent prototype Th2-inducing pathogens. The way DCs sense type-2 pathogens ranges from pattern recognition receptors to tissue damage and to metabolic changes (75). Key factors released by DCs upon recognition of type-2 pathogens to instruct Th2 cells have not been identified so far. Although IL-4 represents a key factor for development and maintenance of Th2 responses, this cytokine is not produced by DCs. Since the absence of IL-12p70 production alone may allow Th2 polarization, a default pathway would induce Th2 cells. However, the failure of DCs to induce Th2 immunity in the absence of exogenous IL-4 produced by basophils does not support the concept of a default mechanism (76). In any case a common feature of Th2-inducing DCs is their semi-mature state, characterized by the up-regulation of antigen presenting molecules (MHC I, MHC II, CD1d) and costimulatory markers (CD80, CD86, CD40) on their surface but their lack to release polarizing cytokines such as IL-12p70 (Th1) or IL-6 and IL-23 (Th17) (77).

Is there a factor that could block full DC maturation while allowing partial maturation? The production of IL-10 by DCs has been associated with their propagation of Th2 immunity (78), however a clear and direct effect of IL-10 on naive T cell conversion to a Th2 phenotype is missing. IL-10 has been shown to fully block DC maturation and induce anergic or regulatory T cells mostly of the Tr1 type (79, 80). Thus, although not fully clear, it seems most likely that a partial activity of IL-10 would suppress the production of polarizing cytokines such as IL-12p70 in an autocrine and paracrine fashion (81, 82) and promote Th2 development indirectly via modulation of DCs. Since Th1 immunity can eradicate helminths and other parasitic infections but Th2 polarization allows for their persistence, many helminths developed mechanisms to promote IL-10 release from macrophages, DCs and other cells such as regulatory T cells (83) as mechanisms of immune evasion.

In this respect, deviation into Th2 immunity seems to represent a mild form of pathogen-directed immune evasion. When testing different developmental stages of the fox tape-worm Echinococcus multilocularis on BM-DCs, the early vulnerable stages of the larvae such as oncospheres and metacestodes induced more drastic forms of immune evasion, namely DC apoptosis or inhibition of DC maturation. Thereby the induction of immunity at the DC level is completely blocked, while the successor protoscolex stage that is protected by its laminated layer "allowed" partial maturation of DCs (84) for induction of Th2 immunity, similar to what has been observed for Echinococcus granulosus(85).

Such a partial maturation profile can be observed also in BM-DCs after treatment with the pro-inflammatory cytokine TNF or products of Trypanosoma brucei (77). The question remained whether DC maturation by inflammatory TNF is qualitatively or quantitatively different for maturation markers and T cell programming as compared with maturation by different types of variant surface glycoproteins (VSGs) of Trypanosoma brucei. Microarray analyses revealed a common inflammatory signature of 24 genes regulated by all stimuli. Only few differences were observed between TNF and two types of VSGs, although being endogenous and inflammatory as opposed to foreign and pathogen-derived. TNF and the VSGs induced similar partial DC maturation as marked by high MHC II, costimulation and Jagged-2 expression but low cytokine profiles and their functional instruction of Th2 responses. In contrast, DC stimulation by LPS regulated almost 5000 genes, resulted in Delta-4 and IL-12p70 induction and polarization towards Th1. All partially matured DC showed comparable effects after injection on Th2-mediated asthma in mice and therapeutic influences on Th1/Th17-mediated experimental autoimmune encephalomyelitis (EAE) with respect to antibody isotype switching or regulatory IL-10 producing Tr1 induction, respectively. Full maturation by LPS regulated the same 24 inflammatory genes as observed after treatment with TNF or VSGs; but in addition numerous other genes, which led to Th1 skewing. In sum, few genes regulated in DCs allow their Th2 polarization, while additional (the same plus more) genes are regulated in DCs inducing Th1 responses. Thereby these data add to the concept of quantitative determination of Th polarization at the DC level (9, 77). Thus, after years of searching for specific DC factors directing Th2 responses, quantitative effects and the default concept in determining Th2 polarization have come back into focus (2, 9).

Th1 INDUCTION BY DCs AND THE SOURCES OF IL-12p70

The textbook knowledge suggests that IL-12 production by DCs directs optimal activation of polarized T helper type-1 cell (Th1) responses (Fig. 1). The following will encourage to have a closer look on the published results that leave decisive questions open. Several unexpected findings about IL-12 production reward a more systematic analysis. For example, the typical Th2 cytokine IL-4 could enhance bacteria-induced IL-12 production by DCs (86, 87). Also IL-10, well established as an immunosuppressive cytokine, could enhance the IL-18-mediated IFN-γ release by CD8+ T cells (88). While the three-signal-theory for differentiated T cell responses has been first established for CD4+ Th1 cells (1/TCR, 2/costimulation, 3/IL-12), this could be clearly adapted also to CTLs (89) and IL-12 as signal 3 is considered as one of the most critical adjuvants for tumor cell killing (90).

The production of IL-12p70 is accepted as the critical event for Th1 polarization (91, 92). The cytokine is encoded by the two genes Il12a (p35) and Il12b (p40) resulting in the synthesis of a IL-12p70 disulfide-linked heterodimer. Major inducers of IL-12 in DCs are pathogens triggering Toll-like receptors (TLRs) (93, 94). TLRs can also cooperate with each other to enhance IL-12 release by human (95) and mouse DCs (96). Although the IL-12-independent Th1 induction has also been described via IL-18, type-I interferons or CD70 (97, 98, 99), IL-12 is considered the most potent Th1 inducer (100). Reversely, human DCs treated with PGE2 were blocked for IL-12 production and failed to generate Th1 responses (101). For murine DCs we found that dose and timing of the mast cell-derived PGD2 exposure of DCs are critical to exert this effect (102). Depending on the infection IL-12 was necessary for microbial elimination as in murine Leishmania major models (103) or dispensable such as in Chagas' disease or mouse hepatitis virus infection (97, 104). Vaccination strategies against Leishmania infection using DCs from IL-12-deficient mice indicate that Th1 responses may rely mainly on IL-12 by sensitizing Langerhans cells (105). The same group also showed that using BM-DCs as vaccines, bystander IL-12 production from the recipient mice could compensate for the lack of DC-vaccine-derived IL-12 (106). It is possible that DCs transmit cytokine production signals to other DCs, such as from migratory to lymph node-resident DC subtypes (107). In addition, two waves of DC migration into lymph nodes have been observed and are necessary to fully prime T cell responses (108). These date indicate that different migratory and lymph node-resident DC subsets may directly interact or coordinately work to generate polarized Th1 responses.

While CD8α+ splenic DCs did not require other signals than pathogen to produce IL-12p40, macrophages needed an IFN-γ priming prior to the pathogen signal (109). This IFN-γ priming to enable IL-12p70 release has been proposed also for some DCs (110). Recently, monocyte-derived DCs were found to release IL-12p40 upon entry to an infection site, which was dependent on NK cell-derived IFN-γ(111). Of note, in this study the IL-12 production by DCs at the infection site appeared not to depend on phagocytosis of pathogens, pointing to bystander effects.

It also became apparent the last years that early during Th1 development NK cell cooperation with DCs plays a critical role (112, 113, 114). IL-12 production by DCs stimulates the IFN-γ production by NK cells and in a feedback loop represents a major cofactor for further IL-12 production by DCs and macrophages. In addition, IFN-γ signaling via STAT1 into developing Th1 cells enhances subsequent IL-12 signals by inducing STAT1 and STAT4 binding to the promoter region of Tbx21, the gene encoding T-bet, which is the master transcription factor of Th1 cells (115). Skin infection of Leishmania major led to the recruitment of NK cells from the blood into lymph nodes and their IFN-γ production starts 9h after infection (113). Whether soluble IL-12 can be transported via the afferent lymphatics and lymph node conduit system to high endothelial venules (HEV) to meet T cells (116) or whether it is provided by the lymph node migratory DCs, is not known.

The widely accepted view about CD4+ T cell activation and differentiation into functional subsets proposes that three signals from DCs are critical to induce Th1 cells. However, some more recent data on DCs and Th cell differentiation indicate that this model may not be complete, pointing out major problems with this all-three-signals-from-one-DC concept.

Time points of IL-12 secretion by DCs

DCs reside at relatively high densities in epithelial tissues such as the skin at an immature state. Typically, microbial infection after skin injury will lead to pathogen uptake and DC maturation. One consequence of microbe-induced DC activation/maturation at the infection site is their subsequent migration to the draining lymph nodes where they process and present the transported antigens to T cells. This migration capacity to T cell areas of secondary lymphoid organs is one hallmark of DC biology as opposed to macrophages, which remain at the infection site to clear the microbes from the local environment. The chemokine receptor CCR7 is a strict requirement for this migration, which is up-regulated continuously after DC activation reaching half-maximal levels only after 24 hours in vitro (117), whereas DC immigration in draining lymph nodes of mice can be observed after 8h (118). In vitro and in vivo data indicate that also IL-12 secretion by human monocyte-derived DCs and murine DCs within the spleen can be detected after 10h or 4h, respectively (109, 119). Others showed IL-12 secretion by DCs into the synapse formed with T cells 5h after DC activation (120). Thus, after pathogen recognition, DCs produce IL-12 with a fast kinetics but up-regulate CCR7 to migrate to lymph nodes later at 24-48h, a time period when IL-12 release is already terminated (118). How can DCs then coordinate the two functions of migration and IL-12 secretion for Th1 induction?

Mutual exclusiveness of migration and cytokine release

Analyses of human monocyte-derived DC populations indicate that mutually exclusive subsets of migratory and IL-12 cytokine producing cells exist in these bulk cultures (121). This finding was supported by our in vivo data. When murine in vitro generated BM-DCs were matured, fluorescence-labeled with CFSE and injected s.c. into mice, the CFSE+ migratory DC which arrived in the lymph nodes were completely negative for intracellular cytokine detection, while endogenous CFSE DCs appeared positive for various cytokines (107). Thus migration and cytokine production might not occur simultaneously in individual DCs. Experiments using IL-12p40-YFP+ reporter mice indicated that YFP+ BM-DCs were still able to migrate to the draining lymph nodes after s.c. injection (116). This would indicate IL-12 production at the infection site followed by DC migration to the lymph node. Such DCs have been reported as "exhausted" DCs that were unable to prime Th1 responses but induced Th2 cells (119). The reverse situation, that DCs become IL-12+ after migration to the lymph node has not been demonstrated so far. To achieve this, genetic manipulation leading to the expression of lymph node homing receptors in DC was necessary to demonstrate their cytokine-producing and lymph node-homing potential (122, 123). Although these genetic manipulations improved T cell responses, they do not contribute to understand the physiological series of events needed for Th1 cell priming.

Time window of IL-12 susceptibility of CD4+ T cells

During T cell priming an early wave of IL-12 production may not be detectable for T cells. After arrival of the DCs in the lymph nodes, T cell priming is initiated with subsequent polarization for CD4+ T cells. However, at the early time points human and mouse CD4+ T cells do not express a functional IL-12R and its upregulation requires TCR signals (124, 125, 126, 127, 128). After intravenous Plasmodium injection into mice IL-12Rβ was upregulated only at days 3~4 after infection (129). Thus, early IL-12 production may occur but may not elicit polarization signals in T cells at these early time points in the lymph node. These investigations on the polarization of Th1 cells further revealed that after initial T cell priming via TCR and CD28, TCR down-regulation is required to allow IL-12R expression for IL-12-mediated Th1 polarization. Thus, after initial antigen-specific T cell priming the subsequent polarization signal may occur ideally in the absence of antigen presentation.

In addition, IL-12 may act on the DCs itself. IL-12 has been shown to induce NF-κB in DCs, DC maturation as well as IL-12 production (130, 131). A second wave of late IL-12 production potentially occurs in a bystander fashion and may allow full differentiation of Th1 cells.

Bystander activation

Although DCs release cytokines after pathogen contact, also endogenous signals can initiate cytokine production. The interaction of 4-1BBL on mature DC with 4-1BB on co-cultured immature DCs has been shown to induce IL-12 production by the immature DCs (132, 133, 134).

Mature CFSE-labeled BM-DCs injected s.c. into mice remained negative for intracellular cytokines when re-analyzed from the draining lymph nodes 24h later, whereas the endogenous lymph node-resident CD11c+ DC population showed the production of various cytokines (107). It requires clarification, whether migratory DCs transmit bystander activation signals (e.g. via 4-1BB-L to 4-1BB) or secrete factors that contribute to the bystander cytokine production (e.g. tissue injury after DC injection). In the case of a bystander activation signal 4-1BB/4-1BB ligand/receptor trimerization is required (135) that cannot be detected by simple surface staining. The latter possibility on the role of extrinsic factors is based on the findings that TNF-matured DCs are neither able to produce IL-12 themselves (22, 77) nor to stimulate IL-12 via bystander activation in vivo(136). It is also conceivable that mature migratory DCs simply transport and then "present" the pathogens itself to lymph node resident immature DCs via DC-DC interactions, similar as reported for presentation of intact antigen by migratory DCs to B cells (137, 138).

Together, the available data indicate that the common model on IL-12 production by DCs is not sufficient to explain all the published findings mentioned above. Despite a plethora of publications on IL-12 production by DCs, the precise mechanisms and time points of IL-12 production still remain elusive. A recent report may point to a unifying concept. While the conventional DC subsets were found to promote proliferation but poor polarization, secondarily generated monocyte-derived DCs during allogeneic immune responses showed the reverse functions by promoting Th1 and Th17 polarization and suppressing T cell proliferation, partially by release of nitric oxide (139). Although the IL-12 production by DCs in this setting has not been tested, it is tempting to speculate that the first wave of T cell stimulation by conventional DCs may occur largely in the absence of their IL-12 production to stimulate T cell proliferation whereas the second wave of inflammation-induced monocyte-derived DCs may release IL-12 to polarize T cells into Th1.

Notes

CONFLICTS OF INTEREST:The authors have no financial conflict of interest.

Abbreviations

BM bone marrow
DCs dendritic cells
HEV high endothelial venules
iTregs induced regulatory T cells
ssmDCs steady state migratory DCs
Treg regulatory T cells
VSGs variant surface glycoproteins

ACKNOWLEDGEMENTS

Research projects of the author that are published or mentioned in this article, were funded by the Wilhelm-Sander-Foundation and the German Research Council, DFG (LU851/8-1, TR52 TP-B7, IRTG1522).

References

    1. O'Garra A, Vieira PL, Vieira P, Goldfeld AE. IL-10-producing and naturally occurring CD4+ Tregs: limiting collateral damage. J Clin Invest 2004;114:1372–1378.
    1. O'Garra A, Vieira P. T(H)1 cells control themselves by producing interleukin-10. Nat Rev Immunol 2007;7:425–428.
    1. Pletinckx K, Dohler A, Pavlovic V, Lutz MB. Role of dendritic cell maturity/costimulation for generation, homeostasis, and suppressive activity of regulatory T cells. Front Immunol 2011;2:39.
    1. Chakraborty NG, Li L, Sporn JR, Kurtzman SH, Ergin MT, Mukherji B. Emergence of regulatory CD4+ T cell response to repetitive stimulation with antigen-presenting cells in vitro: implications in designing antigen-presenting cell-based tumor vaccines. J Immunol 1999;162:5576–5583.
    1. Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell 2010;140:845–858.
    1. Joo H, Upchurch K, Zhang W, Ni L, Li D, Xue Y, Li XH, Hori T, Zurawski S, Liu YJ, Zurawski G, Oh S. Opposing roles of Dectin-1 expressed on human plasmacytoid dendritic cells and myeloid dendritic cells in Th2 polarization. J Immunol 2015;195:1723–1731.
    1. Tjota MY, Sperling AI. Distinct dendritic cell subsets actively induce Th2 polarization. Curr Opin Immunol 2014;31:44–50.
    1. Walsh KP, Mills KH. Dendritic cells and other innate determinants of T helper cell polarisation. Trends Immunol 2013;34:521–530.
    1. O'Garra A, Gabrysova L, Spits H. Quantitative events determine the differentiation and function of helper T cells. Nat Immunol 2011;12:288–294.
    1. van Panhuys N, Klauschen F, Germain RN. T-cell-receptor-dependent signal intensity dominantly controls CD4(+) T cell polarization in vivo. Immunity 2014;41:63–74.
      Ref ID: 10.
    1. Lutz MB. How quantitative differences in dendritic cell maturation can direct T1/T2-cell polarization. Oncoimmunology 2013;2:e22796.
    1. Lutz MB, Schuler G. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity? Trends Immunol 2002;23:445–449.
    1. Langenkamp A, Casorati G, Garavaglia C, Dellabona P, Lanzavecchia A, Sallusto F. T cell priming by dendritic cells: thresholds for proliferation, differentiation and death and intraclonal functional diversification. Eur J Immunol 2002;32:2046–2054.
    1. Ohl L, Mohaupt M, Czeloth N, Hintzen G, Kiafard Z, Zwirner J, Blankenstein T, Henning G, Forster R. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 2004;21:279–288.
    1. Schumann K, Lammermann T, Bruckner M, Legler DF, Polleux J, Spatz JP, Schuler G, Forster R, Lutz MB, Sorokin L, Sixt M. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity 2010;32:703–713.
    1. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, Leenen PJ, Liu YJ, MacPherson G, Randolph GJ, Scherberich J, Schmitz J, Shortman K, Sozzani S, Strobl H, Zembala M, Austyn JM, Lutz MB. Nomenclature of monocytes and dendritic cells in blood. Blood 2010;116:e74–e80.
    1. Weber C, Meiler S, Doring Y, KochM, Drechsler M, Megens RT, Rowinska Z, Bidzhekov K, Fecher C, Ribechini E, van Zandvoort MA, Binder CJ, Jelinek I, Hristov M, Boon L, Jung S, Korn T, Lutz MB, Forster I, Zenke M, Hieronymus T, Junt T, Zernecke A. CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice. J Clin Invest 2011;121:2898–2910.
    1. Berger TG, Schulze-Koops H, Schafer M, Muller E, Lutz MB. Immature and maturation-resistant human dendritic cells generated from bone marrow require two stimulations to induce T cell anergy in vitro. PLoS One 2009;4:e6645.
    1. Waithman J, Allan RS, Kosaka H, Azukizawa H, Shortman K, Lutz MB, Heath WR, Carbone FR, Belz GT. Skin-derived dendritic cells can mediate deletional tolerance of class I-restricted self-reactive T cells. J Immunol 2007;179:4535–4541.
    1. Apostolou I, von Boehmer H. In vivo instruction of suppressor commitment in naive T cells. J Exp Med 2004;199:1401–1408.
    1. Akbari O, DeKruyff RH, Umetsu DT. Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol 2001;2:725–731.
    1. Menges M, Rossner S, Voigtlander C, Schindler H, Kukutsch NA, Bogdan C, Erb K, Schuler G, Lutz MB. Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce antigen-specific protection of mice from autoimmunity. J Exp Med 2002;195:15–21.
    1. Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, Schuler G. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods 1999;223:77–92.
    1. Naik SH. Demystifying the development of dendritic cell subtypes, a little. Immunol Cell Biol 2008;86:439–452.
    1. Cheong C, Matos I, Choi JH, Dandamudi DB, Shrestha E, Longhi MP, Jeffrey KL, Anthony RM, Kluger C, Nchinda G, Koh H, Rodriguez A, Idoyaga J, Pack M, Velinzon K, Park CG, Steinman RM. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas. Cell 2010;143:416–429.
    1. Valdor R, Macian F. Induction and stability of the anergic phenotype in T cells. Semin Immunol 2013;25:313–320.
    1. Huynh A, Zhang R, Turka LA. Signals and pathways controlling regulatory T cells. Immunol Rev 2014;258:117–131.
    1. Jenkins MK, Chen CA, Jung G, Mueller DL, Schwartz RH. Inhibition of antigen-specific proliferation of type 1 murine T cell clones after stimulation with immobilized anti-CD3 monoclonal antibody. J Immunol 1990;144:16–22.
    1. Schwartz RH. T cell anergy. Annu Rev Immunol 2003;21:305–334.
    1. Sloan-Lancaster J, Evavold BD, Allen PM. Induction of T-cell anergy by altered T-cell-receptor ligand on live antigen-presenting cells. Nature 1993;363:156–159.
    1. Macian F, Garcia-Cozar F, Im SH, Horton HF, Byrne MC, Rao A. Transcriptional mechanisms underlying lymphocyte tolerance. Cell 2002;109:719–731.
    1. Andris F, Denanglaire S, de Mattia F, Urbain J, Leo O. Naive T cells are resistant to anergy induction by anti-CD3 antibodies. J Immunol 2004;173:3201–3208.
    1. Wells AD, Walsh MC, Bluestone JA, Turka LA. Signaling through CD28 and CTLA-4 controls two distinct forms of T cell anergy. J Clin Invest 2001;108:895–903.
    1. Greenwald RJ, Boussiotis VA, Lorsbach RB, Abbas AK, Sharpe AH. CTLA-4 regulates induction of anergy in vivo. Immunity 2001;14:145–155.
    1. Perez VL, Van Parijs L, Biuckians A, Zheng XX, Strom TB, Abbas AK. Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement. Immunity 1997;6:411–417.
    1. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev 2009;229:12–26.
    1. Brunner MC, Chambers CA, Chan FK, Hanke J, Winoto A, Allison JP. CTLA-4-Mediated inhibition of early events of T cell proliferation. J Immunol 1999;162:5813–5820.
    1. Verdoodt B, Blazek T, Rauch P, Schuler G, Steinkasserer A, Lutz MB, Funk JO. The cyclin-dependent kinase inhibitors p27Kip1 and p21Cip1 are not essential in T cell anergy. Eur J Immunol 2003;33:3154–3163.
    1. Walunas TL, Bakker CY, Bluestone JA. CTLA-4 ligation blocks CD28-dependent T cell activation. J Exp Med 1996;183:2541–2550.
    1. Krummel MF, Allison JP. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med 1996;183:2533–2540.
    1. Baine I, Abe BT, Macian F. Regulation of T-cell tolerance by calcium/NFAT signaling. Immunol Rev 2009;231:225–240.
    1. Macian F, Im SH, Garcia-Cozar FJ, Rao A. T-cell anergy. Curr Opin Immunol 2004;16:209–216.
    1. Fathman CG, Lineberry NB. Molecular mechanisms of CD4+ T-cell anergy. Nat Rev Immunol 2007;7:599–609.
    1. Kriegel MA, Rathinam C, Flavell RA. E3 ubiquitin ligase GRAIL controls primary T cell activation and oral tolerance. Proc Natl Acad Sci U S A 2009;106:16770–16775.
    1. Jeon MS, Atfield A, Venuprasad K, Krawczyk C, Sarao R, Elly C, Yang C, Arya S, Bachmaier K, Su L, Bouchard D, Jones R, Gronski M, Ohashi P, Wada T, Bloom D, Fathman CG, Liu YC, Penninger JM. Essential role of the E3 ubiquitin ligase Cbl-b in T cell anergy induction. Immunity 2004;21:167–177.
    1. Lechner O, Lauber J, Franzke A, Sarukhan A, von Boehmer H, Buer J. Fingerprints of anergic T cells. Curr Biol 2001;11:587–595.
    1. Safford M, Collins S, Lutz MA, Allen A, Huang CT, Kowalski J, Blackford A, Horton MR, Drake C, Schwartz RH, Powell JD. Egr-2 and Egr-3 are negative regulators of T cell activation. Nat Immunol 2005;6:472–480.
    1. Harris JE, Bishop KD, Phillips NE, Mordes JP, Greiner DL, Rossini AA, Czech MP. Early growth response gene-2, a zinc-finger transcription factor, is required for full induction of clonal anergy in CD4+ T cells. J Immunol 2004;173:7331–7338.
    1. Knoechel B, Lohr J, Zhu S, Wong L, Hu D, Ausubel L, Abbas AK. Functional and molecular comparison of anergic and regulatory T lymphocytes. J Immunol 2006;176:6473–6483.
    1. Zheng Y, Zha Y, Driessens G, Locke F, Gajewski TF. Transcriptional regulator early growth response gene 2 (Egr2) is required for T cell anergy in vitro and in vivo. J Exp Med 2012;209:2157–2163.
    1. Miah MA, Byeon SE, Ahmed MS, Yoon CH, Ha SJ, Bae YS. Egr2 induced during DC development acts as an intrinsic negative regulator of DC immunogenicity. Eur J Immunol 2013;43:2484–2496.
    1. Pape KA, Merica R, Mondino A, Khoruts A, Jenkins MK. Direct evidence that functionally impaired CD4+ T cells persist in vivo following induction of peripheral tolerance. J Immunol 1998;160:4719–4729.
    1. Groux H, O'Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 1997;389:737–742.
    1. Buer J, Lanoue A, Franzke A, Garcia C, von Boehmer H, Sarukhan A. Interleukin 10 secretion and impaired effector function of major histocompatibility complex class II-restricted T cells anergized in vivo. J Exp Med 1998;187:177–183.
    1. Jooss K, Gjata B, Danos O, von Boehmer H, Sarukhan A. Regulatory function of in vivo anergized CD4(+) T cells. Proc Natl Acad Sci U S A 2001;98:8738–8743.
    1. Wegner J, Hackenberg S, Scholz CJ, Chuvpilo S, Tyrsin D, Matskevich AA, Grigoleit GU, Stevanovic S, Hunig T. High-density preculture of PBMCs restores defective sensitivity of circulating CD8 T cells to virus- and tumor-derived antigens. Blood 2015;126:185–194.
    1. Burkly LC, Lo D, Kanagawa O, Brinster RL, Flavell RA. T-cell tolerance by clonal anergy in transgenic mice with nonlymphoid expression of MHC class II I-E. Nature 1989;342:564–566.
    1. Rammensee HG, Kroschewski R, Frangoulis B. Clonal anergy induced in mature V beta 6+ T lymphocytes on immunizing Mls-1b mice with Mls-1a expressing cells. Nature 1989;339:541–544.
    1. Kawabe Y, Ochi A. Selective anergy of V beta 8+, CD4+ T cells in Staphylococcus enterotoxin B-primed mice. J Exp Med 1990;172:1065–1070.
    1. Rellahan BL, Jones LA, Kruisbeek AM, Fry AM, Matis LA. In vivo induction of anergy in peripheral V beta 8+ T cells by staphylococcal enterotoxin B. J Exp Med 1990;172:1091–1100.
    1. Gabrysova L, Wraith DC. Antigenic strength controls the generation of antigen-specific IL-10-secreting T regulatory cells. Eur J Immunol 2010;40:1386–1395.
    1. Gabrysova L, Nicolson KS, Streeter HB, Verhagen J, Sabatos-Peyton CA, Morgan DJ, Wraith DC. Negative feedback control of the autoimmune response through antigen-induced differentiation of IL-10-secreting Th1 cells. J Exp Med 2009;206:1755–1767.
    1. Pletinckx K, Vaeth M, Schneider T, Beyersdorf N, Hunig T, Berberich-Siebelt F, Lutz MB. Immature dendritic cells convert anergic nonregulatory T cells into Foxp3 IL-10+ regulatory T cells by engaging CD28 and CTLA-4. Eur J Immunol 2015;45:480–491.
    1. Steinman RM, Nussenzweig MC. Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci U S A 2002;99:351–358.
    1. Sakaguchi S, Powrie F, Ransohoff RM. Re-establishing immunological self-tolerance in autoimmune disease. Nat Med 2012;18:54–58.
    1. Sixt M, Kanazawa N, Selg M, Samson T, Roos G, Reinhardt DP, Pabst R, Lutz MB, Sorokin L. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 2005;22:19–29.
    1. Azukizawa H, Dohler A, Kanazawa N, Nayak A, Lipp M, Malissen B, Autenrieth I, Katayama I, Riemann M, Weih F, Berberich-Siebelt F, Lutz MB. Steady state migratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction of antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells. Eur J Immunol 2011;41:1420–1434.
    1. Hayden MS, Ghosh S. NF-kappaB, the first quarter-century: remarkable progress and outstanding questions. Genes Dev 2012;26:203–234.
    1. Lutz MB, Dohler A, Azukizawa H. Revisiting the tolerogenicity of epidermal Langerhans cells. Immunol Cell Biol 2010;88:381–386.
    1. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 2005;6:1142–1151.
    1. Weih F, Caamano J. Regulation of secondary lymphoid organ development by the nuclear factor-kappaB signal transduction pathway. Immunol Rev 2003;195:91–105.
    1. Idoyaga J, Fiorese C, Zbytnuik L, Lubkin A, Miller J, Malissen B, Mucida D, Merad M, Steinman RM. Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Invest 2013;123:844–854.
    1. Baratin M, Foray C, Demaria O, Habbeddine M, Pollet E, Maurizio J, Verthuy C, Davanture S, Azukizawa H, Flores-Langarica A, Dalod M, Lawrence T. Homeostatic NF-kappaB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance. Immunity 2015;42:627–639.
    1. Travis MA, Reizis B, Melton AC, Masteller E, Tang Q, Proctor JM, Wang Y, Bernstein X, Huang X, Reichardt LF, Bluestone JA, Sheppard D. Loss of integrin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice. Nature 2007;449:361–365.
    1. Pulendran B, Artis D. New paradigms in type 2 immunity. Science 2012;337:431–435.
    1. Tang H, Cao W, Kasturi SP, Ravindran R, Nakaya HI, Kundu K, Murthy N, Kepler TB, Malissen B, Pulendran B. The T helper type 2 response to cysteine proteases requires dendritic cell-basophil cooperation via ROS-mediated signaling. Nat Immunol 2010;11:608–617.
    1. Pletinckx K, Stijlemans B, Pavlovic V, Laube R, Brandl C, Kneitz S, Beschin A, De Baetselier P, Lutz MB. Similar inflammatory DC maturation signatures induced by TNF or Trypanosoma brucei antigens instruct default Th2-cell responses. Eur J Immunol 2011;41:3479–3494.
    1. De Smedt T, Van Mechelen M, De Becker G, Urbain J, Leo O, Moser M. Effect of interleukin-10 on dendritic cell maturation and function. Eur J Immunol 1997;27:1229–1235.
    1. Raker VK, Domogalla MP, Steinbrink K. Tolerogenic dendritic cells for regulatory T cell induction in man. Front Immunol 2015;6:569.
    1. Amodio G, Gregori S. Human tolerogenic DC-10: perspectives for clinical applications. Transplant Res 2012;1:14.
    1. Maldonado-Lopez R, Maliszewski C, Urbain J, Moser M. Cytokines regulate the capacity of CD8alpha(+) and CD8alpha(−) dendritic cells to prime Th1/Th2 cells in vivo. J Immunol 2001;167:4345–4350.
    1. Perona-Wright G, Jenkins SJ, Crawford A, Gray D, Pearce EJ, MacDonald AS. Distinct sources and targets of IL-10 during dendritic cell-driven Th1 and Th2 responses in vivo. Eur J Immunol 2006;36:2367–2375.
    1. Redpath SA, van der Werf N, Cervera AM, MacDonald AS, Gray D, Maizels RM, Taylor MD. ICOS controls Foxp3(+) regulatory T-cell expansion, maintenance and IL-10 production during helminth infection. Eur J Immunol 2013;43:705–715.
    1. Nono JK, Pletinckx K, Lutz MB, Brehm K. Excretory/secretory-products of Echinococcus multilocularis larvae induce apoptosis and tolerogenic properties in dendritic cells in vitro. PLoS Negl Trop Dis 2012;6:e1516.
    1. Casaravilla C, Pittini A, Ruckerl D, Seoane PI, Jenkins SJ, MacDonald AS, Ferreira AM, Allen JE, Diaz A. Unconventional maturation of dendritic cells induced by particles from the laminated layer of larval Echinococcus granulosus. Infect Immun 2014;82:3164–3176.
    1. Hochrein H, O'Keeffe M, Luft T, Vandenabeele S, Grumont RJ, Maraskovsky E, Shortman K. Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 production by mouse and human dendritic cells. J Exp Med 2000;192:823–833.
    1. Lutz MB, Schnare M, Menges M, Rossner S, Rollinghoff M, Schuler G, Gessner A. Differential functions of IL-4 receptor types I and II for dendritic cell maturation and IL-12 production and their dependency on GM-CSF. J Immunol 2002;169:3574–3580.
    1. Freeman BE, Hammarlund E, Raue HP, Slifka MK. Regulation of innate CD8+ T-cell activation mediated by cytokines. Proc Natl Acad Sci U S A 2012;109:9971–9976.
    1. Mescher MF, Curtsinger JM, Agarwal P, Casey KA, Gerner M, Hammerbeck CD, Popescu F, Xiao Z. Signals required for programming effector and memory development by CD8+ T cells. Immunol Rev 2006;211:81–92.
    1. Curtsinger JM, Gerner MY, Lins DC, Mescher MF. Signal 3 availability limits the CD8 T cell response to a solid tumor. J Immunol 2007;178:6752–6760.
    1. Heufler C, Koch F, Stanzl U, Topar G, Wysocka M, Trinchieri G, Enk A, Steinman RM, Romani N, Schuler G. Interleukin-12 is produced by dendritic cells and mediates T helper 1 development as well as interferon-gamma production by T helper 1 cells. Eur J Immunol 1996;26:659–668.
    1. Cella M, Scheidegger D, Palmer-Lehmann K, Lane P, Lanzavecchia A, Alber G. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med 1996;184:747–752.
    1. Steinman RM, Hemmi H. Dendritic cells: translating innate to adaptive immunity. Curr Top Microbiol Immunol 2006;311:17–58.
    1. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol 2003;3:133–146.
    1. Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol 2005;6:769–776.
    1. Theiner G, Rossner S, Dalpke A, Bode K, Berger T, Gessner A, Lutz MB. TLR9 cooperates with TLR4 to increase IL-12 release by murine dendritic cells. Mol Immunol 2008;45:244–252.
    1. Muller U, Kohler G, Mossmann H, Schaub GA, Alber G, Di Santo JP, Brombacher F, Holscher C. IL-12-independent IFN-gamma production by T cells in experimental Chagas' disease is mediated by IL-18. J Immunol 2001;167:3346–3353.
    1. Longhi MP, Trumpfheller C, Idoyaga J, Caskey M, Matos I, Kluger C, Salazar AM, Colonna M, Steinman RM. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J Exp Med 2009;206:1589–1602.
    1. Soares H, Waechter H, Glaichenhaus N, Mougneau E, Yagita H, Mizenina O, Dudziak D, Nussenzweig MC, Steinman RM. A subset of dendritic cells induces CD4+ T cells to produce IFN-gamma by an IL-12-independent but CD70-dependent mechanism in vivo. J Exp Med 2007;204:1095–1106.
    1. de Jong R, Altare F, Haagen IA, Elferink DG, Boer T, van Breda Vriesman PJ, Kabel PJ, Draaisma JM, van Dissel JT, Kroon FP, Casanova JL, Ottenhoff TH. Severe mycobacterial and Salmonella infections in interleukin-12 receptor-deficient patients. Science 1998;280:1435–1438.
    1. Kalinski P, Hilkens CM, Snijders A, Snijdewint FG, Kapsenberg ML. IL-12-deficient dendritic cells, generated in the presence of prostaglandin E2, promote type 2 cytokine production in maturing human naive T helper cells. J Immunol 1997;159:28–35.
    1. Theiner G, Gessner A, Lutz MB. The mast cell mediator PGD2 suppresses IL-12 release by dendritic cells leading to Th2 polarized immune responses in vivo. Immunobiology 2006;211:463–472.
    1. Mattner F, Di Padova K, Alber G. Interleukin-12 is indispensable for protective immunity against Leishmania major. Infect Immun 1997;65:4378–4383.
    1. Schijns VE, Haagmans BL, Wierda CM, Kruithof B, Heijnen IA, Alber G, Horzinek MC. Mice lacking IL-12 develop polarized Th1 cells during viral infection. J Immunol 1998;160:3958–3964.
    1. Berberich C, Ramirez-Pineda JR, Hambrecht C, Alber G, Skeiky YA, Moll H. Dendritic cell (DC)-based protection against an intracellular pathogen is dependent upon DC-derived IL-12 and can be induced by molecularly defined antigens. J Immunol 2003;170:3171–3179.
    1. Ramirez-Pineda JR, Frohlich A, Berberich C, Moll H. Dendritic cells (DC) activated by CpG DNA ex vivo are potent inducers of host resistance to an intracellular pathogen that is independent of IL-12 derived from the immunizing DC. J Immunol 2004;172:6281–6289.
    1. Voigtlander C, Rossner S, Cierpka E, Theiner G, Wiethe C, Menges M, Schuler G, Lutz MB. Dendritic cells matured with TNF can be further activated in vitro and after subcutaneous injection in vivo which converts their tolerogenicity into immunogenicity. J Immunother 2006;29:407–415.
    1. Itano AA, McSorley SJ, Reinhardt RL, Ehst BD, Ingulli E, Rudensky AY, Jenkins MK. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity 2003;19:47–57.
    1. Reis e Sousa C, Hieny S, Scharton-Kersten T, Jankovic D, Charest H, Germain RN, Sher A. In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas. J Exp Med 1997;186:1819–1829.
    1. Abdi K, Singh N, Matzinger P. T-cell control of IL-12p75 production. Scand J Immunol 2006;64:83–92.
    1. Goldszmid RS, Caspar P, Rivollier A, White S, Dzutsev A, Hieny S, Kelsall B, Trinchieri G, Sher A. NK cell-derived interferon-gamma orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection. Immunity 2012;36:1047–1059.
    1. Moretta L, Ferlazzo G, Bottino C, Vitale M, Pende D, Mingari MC, Moretta A. Effector and regulatory events during natural killer-dendritic cell interactions. Immunol Rev 2006;214:219–228.
    1. Bajenoff M, Breart B, Huang AY, Qi H, Cazareth J, Braud VM, Germain RN, Glaichenhaus N. Natural killer cell behavior in lymph nodes revealed by static and real-time imaging. J Exp Med 2006;203:619–631.
    1. Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A, Sallusto F. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat Immunol 2004;5:1260–1265.
    1. Oestreich KJ, Weinmann AS. Transcriptional mechanisms that regulate T helper 1 cell differentiation. Curr Opin Immunol 2012;24:191–195.
    1. Reinhardt RL, Hong S, Kang SJ, Wang ZE, Locksley RM. Visualization of IL-12/23p40 in vivo reveals immunostimulatory dendritic cell migrants that promote Th1 differentiation. J Immunol 2006;177:1618–1627.
    1. Sallusto F, Palermo B, Lenig D, Miettinen M, Matikainen S, Julkunen I, Forster R, Burgstahler R, Lipp M, Lanzavecchia A. Distinct patterns and kinetics of chemokine production regulate dendritic cell function. Eur J Immunol 1999;29:1617–1625.
    1. Ingulli E, Mondino A, Khoruts A, Jenkins MK. In vivo detection of dendritic cell antigen presentation to CD4(+) T cells. J Exp Med 1997;185:2133–2141.
    1. Langenkamp A, Messi M, Lanzavecchia A, Sallusto F. Kinetics of dendritic cell activation: impact on priming of TH1, TH2 and nonpolarized T cells. Nat Immunol 2000;1:311–316.
    1. Pulecio J, Petrovic J, Prete F, Chiaruttini G, Lennon-Dumenil AM, Desdouets C, Gasman S, Burrone OR, Benvenuti F. Cdc42-mediated MTOC polarization in dendritic cells controls targeted delivery of cytokines at the immune synapse. J Exp Med 2010;207:2719–2732.
    1. Luft T, Jefford M, Luetjens P, Toy T, Hochrein H, Masterman KA, Maliszewski C, Shortman K, Cebon J, Maraskovsky E. Functionally distinct dendritic cell (DC) populations induced by physiologic stimuli: prostaglandin E(2) regulates the migratory capacity of specific DC subsets. Blood 2002;100:1362–1372.
    1. Knippertz I, Hesse A, Schunder T, Kampgen E, Brenner MK, Schuler G, Steinkasserer A, Nettelbeck DM. Generation of human dendritic cells that simultaneously secrete IL-12 and have migratory capacity by adenoviral gene transfer of hCD40L in combination with IFN-gamma. J Immunother 2009;32:524–538.
    1. Dorrie J, Schaft N, Muller I, Wellner V, Schunder T, Hanig J, Oostingh GJ, Schon MP, Robert C, Kampgen E, Schuler G. Introduction of functional chimeric E/L-selectin by RNA electroporation to target dendritic cells from blood to lymph nodes. Cancer Immunol Immunother 2008;57:467–477.
    1. Schulz EG, Mariani L, Radbruch A, Hofer T. Sequential polarization and imprinting of type 1 T helper lymphocytes by interferon-gamma and interleukin-12. Immunity 2009;30:673–683.
    1. Elloso MM, Scott P. Differential requirement of CD28 for IL-12 receptor expression and function in CD4(+) and CD8(+) T cells. Eur J Immunol 2001;31:384–395.
    1. Ahlers JD, Belyakov IM, Matsui S, Berzofsky JA. Signals delivered through TCR instruct IL-12 receptor (IL-12R) expression: IL-12 and tumor necrosis factor-alpha synergize for IL-12R expression at low antigen dose. Int Immunol 2001;13:1433–1442.
    1. Szabo SJ, Dighe AS, Gubler U, Murphy KM. Regulation of the interleukin (IL)-12R beta 2 subunit expression in developing T helper 1 (Th1) and Th2 cells. J Exp Med 1997;185:817–824.
    1. Rogge L, Barberis-Maino L, Biffi M, Passini N, Presky DH, Gubler U, Sinigaglia F. Selective expression of an interleukin-12 receptor component by human T helper 1 cells. J Exp Med 1997;185:825–831.
    1. Sam H, Stevenson MM. In vivo IL-12 production and IL-12 receptors beta1 and beta2 mRNA expression in the spleen are differentially up-regulated in resistant B6 and susceptible A/J mice during early blood-stage Plasmodium chabaudi AS malaria. J Immunol 1999;162:1582–1589.
    1. Grohmann U, Belladonna ML, Bianchi R, Orabona C, Ayroldi E, Fioretti MC, Puccetti P. IL-12 acts directly on DC to promote nuclear localization of NF-kappaB and primes DC for IL-12 production. Immunity 1998;9:315–323.
    1. Puccetti P, Belladonna ML, Grohmann U. Effects of IL-12 and IL-23 on antigen-presenting cells at the interface between innate and adaptive immunity. Crit Rev Immunol 2002;22:373–390.
    1. Laderach D, Wesa A, Galy A. 4-1BB-ligand is regulated on human dendritic cells and induces the production of IL-12. Cell Immunol 2003;226:37–44.
    1. Futagawa T, Akiba H, Kodama T, Takeda K, Hosoda Y, Yagita H, Okumura K. Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells. Int Immunol 2002;14:275–286.
    1. Wilcox RA, Chapoval AI, Gorski KS, Otsuji M, Shin T, Flies DB, Tamada K, Mittler RS, Tsuchiya H, Pardoll DM, Chen L. Cutting edge: Expression of functional CD137 receptor by dendritic cells. J Immunol 2002;168:4262–4267.
    1. Wyzgol A, Muller N, Fick A, Munkel S, Grigoleit GU, Pfizenmaier K, Wajant H. Trimer stabilization, oligomerization, and antibody-mediated cell surface immobilization improve the activity of soluble trimers of CD27L, CD40L, 41BBL, and glucocorticoid-induced TNF receptor ligand. J Immunol 2009;183:1851–1861.
    1. Sporri R, Reis e Sousa C. Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol 2005;6:163–170.
    1. Wykes M, Pombo A, Jenkins C, MacPherson GG. Dendritic cells interact directly with naive B lymphocytes to transfer antigen and initiate class switching in a primary T-dependent response. J Immunol 1998;161:1313–1319.
    1. Qi H, Egen JG, Huang AY, Germain RN. Extrafollicular activation of lymph node B cells by antigen-bearing dendritic cells. Science 2006;312:1672–1676.
    1. Chow KV, Lew AM, Sutherland RM, Zhan Y. Monocyte-derived dendritic cells promote Th polarization, whereas conventional dendritic cells promote Th proliferation. J Immunol 2016;196:624–636.

Metrics
Share
Figures

1 / 1

Funding Information
PERMALINK