Skip to main content

REVIEW article

Front. Immunol., 06 March 2013
Sec. Antigen Presenting Cell Biology
This article is part of the Research Topic The nature of activatory and tolerogenic dendritic cell-derived signal 2 View all 13 articles

Tolerogenic and Activatory Plasmacytoid Dendritic Cells in Autoimmunity

  • Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland

Plasmacytoid dendritic cells (pDCs) are a particular subset of DCs that link innate and adaptive immunity. They are responsible for the substantial production of type 1 interferon (IFN-I) in response to viral RNA or DNA through activation of TLR7 and 9. Furthermore, pDCs present antigens (Ag) and induce naïve T cell differentiation. It has been demonstrated that pDCs can induce immunogenic T cell responses through differentiation of cytotoxic CD8+ T cells and effector CD4+ T cells. Conversely, pDCs exhibit strong tolerogenic functions by inducing CD8+ T cell deletion, CD4+ T cell anergy, and Treg differentiation. However, since IFN-I produced by pDCs efficiently activates and recruits conventional DCs, B cells, T cells, and NK cells, pDCs also indirectly affect the nature and the amplitude of adaptive immune responses. As a consequence, the precise role of Ag-presenting functions of pDCs in adaptive immunity has been difficult to dissect in vivo. Additionally, different experimental procedures led to conflicting results regarding the outcome of T cell responses induced by pDCs. During the development of autoimmunity, pDCs have been shown to play both immunogenic and tolerogenic functions depending on disease, disease progression, and the experimental conditions. In this review, we will discuss the relative contribution of innate and adaptive pDC functions in modulating T cell responses, particularly during the development of autoimmunity.

Plasmacytoid Dendritic Cell Characteristics

Plasmacytoid dendritic cells (pDCs) were first described as either interferon (IFN) producing cells (Ronnblom et al., 1983; Chehimi et al., 1989; Fitzgerald-Bocarsly, 1993) or as plasmacytoid monocytes or plasmacytoid T cells in reference to their plasma-like morphology in secondary lymphoid organs (SLOs) (Facchetti et al., 1988). They were further defined as pre-DC2, as during activation they can differentiate into conventional DCs (cDCs)-like cells. These cells exhibit a DC morphology, with increased MHC class II (MHCII) and costimulatory molecule expression, and the ability to induce naïve CD4+ T cell proliferation (Grouard et al., 1997). In 1999, the groups of M. Colonna and Y. J. Liu formally demonstrated that all these cell subtypes were actually the same entity, the pDCs (Cella et al., 1999; Siegal et al., 1999).

The development of pDCs occurs in the bone marrow (BM), after which they circulate through the blood stream, and reside in steady-state in the thymus and in the SLOs. Upon challenge via infection or inflammation, pDCs migrate and accumulate in inflamed tissues and draining lymph nodes (dLNs) (Reizis et al., 2011). The pDCs derive from the common dendritic progenitors (CDP) which express FLT3-R (CD135), CSF1-R (CD115), and low levels of c-kit (CD117) (Onai et al., 2007). Alternatively, pDCs may derive from lymphoid progenitors (Shigematsu et al., 2004; Luo and Lei, 2012; Sathe et al., 2013). Differentiation of pDCs relies essentially on Flt3-L which allows the expansion of cDC/pDC common progenitors and contributes to peripheral DC homeostasis (Waskow et al., 2008; Eidenschenk et al., 2010). The importance of E2-2 has been demonstrated for pDC differentiation (Cisse et al., 2008). It was further described that E2-2 drives the expression of other transcription factors involved in pDC fate, such as IRF-8 or Spi-B, while it inhibits other factors that are important for cDC differentiation, including Id-2 (Ghosh et al., 2010). Thus, a balance between the transcription factors E2-2 and Id-2 appears to control the differentiation toward the pDC lineage. Accordingly, in E2-2 deficient mice, pDCs exhibit an increased expression of Id-2 that correlates with a conversion into cDCs.

At the phenotypic level, pDCs are characterized by having intermediate (mouse) or no (human) expression of the DC marker CD11c. They are positive for the B cell marker CD45RA/B220, and express high levels of PDCA1, BST-2, Ly6C, and Ly49Q in mouse and BDCA-2, ILT-7, IL3Ra (CD123), and BDCA-4 in human (Reizis et al., 2011).

Innate Plasmacytoid DC Functions

Features of pDC Innate Functions

Plasmacytoid DCs are strong sensors of non-self nucleic acids derived from bacteria or viruses through binding to Toll-like receptors (TLR). Nucleic acids come from either viruses internalized by endocytosis, cytoplasmic viral RNA by autophagy (Lee et al., 2007), or other infected cells via exosome transport (Dreux et al., 2012). The receptors TLR7 and TLR9 are selectively expressed by pDCs. Interestingly, pDCs are the only DC subset expressing TLR9 in humans (Jarrossay et al., 2001; Kadowaki et al., 2001; Hornung et al., 2002) but not in mice (Chen et al., 2006). TLR7 senses guanosine or uridine rich single-stranded RNA from viruses or synthetic compounds such as imidazoquinoline and guanosine analogs (Diebold et al., 2004; Heil et al., 2004; Lund et al., 2004), while TLR9 recognizes single-stranded DNA containing unmethylated CpG motifs commonly found in viral and bacterial genomes (Hemmi et al., 2000; Bauer et al., 2001). The activation of TLR9 can also be through synthetic oligonucleotides (ODN) that mimic viral ssDNA responses (Kadowaki et al., 2001). TLR binding signals through MyD88, an adaptor protein that forms a signaling scaffold with IRAK-4, TRAF-6, and Btk, and induces the formation of the TRAF-3/IRAK-1/IKK-α/OPN/PI3K complex. As a consequence, IRF-7 is phosphorylated and subsequently translocated in the nucleus where it induces IFN-I gene transcription. Signaling through TRAF-6 also induces the NF-κB and MAPK pathways, leading to the secretion of inflammatory cytokines and chemokines, and the up-regulation of costimulatory molecules (Gilliet et al., 2008). Unlike other cells, where expression is dependent on IFNAR signaling, IRF-7 is constitutively expressed in pDCs (Honda et al., 2005b; Ito et al., 2006), possibly due to the low expression of the negative translational repressors 4EBPs (Colina et al., 2008).

The TLR ligands CpG-ODN have been classified as CpG-A, -B, and -C based on the different immune responses induced. In pDCs, CpG-A gives rise to robust IFN-I production, whereas CpG-B induces the production of inflammatory cytokines such as TNF or IL-6, as well as the up-regulation of MHCII and costimulatory molecules at the pDC surface (Kerkmann et al., 2005). These distinct effects of CpG-A and -B are pDC-specific and rely on different intracellular localizations after internalization. CpG-A possesses a poly-G tail, leading to the formation of a large multimeric complex that is retained in early endosomes and signals through MyD88 and IRF-7, thus inducing a strong IFN-I response. Conversely, monomeric CpG-B fails to be retained in early endosomes and rapidly travels to late endosomes/lysosomes to induce TNF and IL-6 production, and costimulatory molecule expression (Honda et al., 2005a; Guiducci et al., 2006).

Production of IFN-I by pDCs is much stronger (200–1000 times more effective) than any other cell type (Siegal et al., 1999). Multiple subtypes of IFN-I are secreted by pDCs, including IFNα, β, κ, ω, λ, and τ (Ito et al., 2006). During viral infections, pDC ablation selectively abrogates the early peak of IFN-I and leads to an increased viral burden (Swiecki et al., 2010), suggesting that IFN-I secretion is particularly crucial at the beginning of the antiviral response. By itself, IFN-I promotes the expression of IFN-stimulated genes that inhibit viral spreading through different mechanisms: (i) inhibition of viral replication by RNA degradation (Malathi et al., 2005) or by decreased protein synthesis (Barber et al., 1993); (ii) establishment of an antiviral state in uninfected cells; and (iii) induction of infected cell apoptosis (Pestka et al., 2004). Importantly, IFN-I links innate and adaptive immunity as it induces the differentiation, maturation, and activation of myeloid DCs that in turn promote antiviral T cell immunity (Paquette et al., 1998; Santini et al., 2000; Hibbert et al., 2003; Le Bon et al., 2003; Fonteneau et al., 2004; Yoneyama et al., 2005). Furthermore, IFN-I activates the antiviral functions of NK cells (Gerosa et al., 2005) and B cells (Jego et al., 2003). Using conditional targeting of the pDC-specific transcription factor E2-2, an elegant study has shown that pDC-deficient mice fail to clear chronic LCMV infection (Cervantes-Barragan et al., 2012). This defect correlates with impaired LCMV-specific CD4+ and CD8+ T cell numbers and functions, and relies on the lack of IFN-I production by pDCs, independently of their antigen (Ag) presenting capabilities.

Innate pDC Functions and Autoimmunity

Self-DNA has been demonstrated to activate pDCs in autoimmune diseases. Under normal conditions, self-DNA is not recognized by pDCs since, when released by necrotic and apoptotic cells (Pisetsky and Fairhurst, 2007), it remains in the extracellular environment (Barton et al., 2006) and is rapidly degraded by DNAses. Following skin injury in psoriasis, self-DNA is released in the extracellular milieu (Lande et al., 2007) and sensed by pDCs (Nestle et al., 2005). In the wounded skin, the cationic anti-microbial cathelicidin LL37 is produced by keratinocytes and neutrophils (Zasloff, 2002). LL37 production is increased in psoriatic skin, binds the DNA released by dying cells, and forms aggregates which are resistant to extracellular nucleases (Lande et al., 2007). These complexes enter pDCs by endocytosis via lipid rafts and interactions with proteoglycans (Sandgren et al., 2004), and localize in early endosomes (similarly to CpG-A) to induce a strong IFN-I response. Consistently, an Imiquimod (TLR7 ligand)-containing topical cream was shown to exacerbate psoriatic lesions (Gilliet et al., 2004).

It has also been shown that IFN-I also plays a major role in the development of systemic lupus erythematosus (SLE) (Chan et al., 2012), by inducing the differentiation of both auto-antibody producing plasma cells (Jego et al., 2003; Thibault et al., 2009; Mathian et al., 2011) and cDC-driven effector T cells (Blanco et al., 2001). In SLE, apoptotic and necrotic cell derived self-DNA are complexed with (i) the peptide LL37, released by apoptotic neutrophil extracellular traps (NETs) in skin lesions (Bennett et al., 2003; Garcia-Romo et al., 2011; Lande et al., 2011), (ii) the protein HMGB1, also released in NETs, which binds aggregated nucleic acids (like CpG-A) (Tian et al., 2007), and (iii) by auto-antibodies directed against nucleic acids or nucleoproteins (Lovgren et al., 2004). These immune complexes allow the delivery of self-DNA into pDCs through the interaction of auto-Ab with FcγRII (CD32), and LC3-associated phagocytosis, a process described as a convergence between phagocytosis and non-conventional autophagy (Henault et al., 2012). In endosomes, self-DNA activates TLR signaling and subsequent IFN-I production (Lovgren et al., 2004; Barrat et al., 2005). Moreover, HMGB1 interacts with RAGE at the pDC surface, facilitating TLR9/DNA-binding in endosomes without inducing endocytosis (Tian et al., 2007). Finally, the resulting robust production of IFN-I, together with the presence of anti-LL37 auto-Abs in sera of SLE patients, induces the release of NETs and the continuous release of immune complexes (Garcia-Romo et al., 2011; Lande et al., 2011).

In multiple sclerosis (MS), many studies have suggested a protective role of pDCs and IFN-I. Indeed, relapsing patients treated with IFNβ-1a exhibit both a reduction in disease severity and a delay in relapses (Goodkin, 1996). In experimental autoimmune encephalomyelitis (EAE), a deficiency of IFNAR in central nervous system (CNS) myeloid cells exacerbates disease development (Prinz et al., 2008). In addition, pDCs have been shown to inhibit cDC functions and, consequently, dampen the development of encephalitogenic Th17 cells (Bailey et al., 2007), whereas anti-PDCA1 mediated pDC depletion increases EAE severity (Bailey-Bucktrout et al., 2008). As discussed by the authors, and consistent with the IFN-I dependent inhibition of Th17 inflammation in the CNS (Guo et al., 2008), pDC-mediated protection may indeed depend on IFN-I. However, the molecular mechanisms accounting for the local suppression of pathogenic T cells in the CNS by pro-inflammatory IFN-I remain unclear.

Until recently, the contribution of pDCs and IFN-I to pathogenesis in type 1 diabetes (T1D) remained controversial. The role of IFN-I, first believed to be protective (Sobel and Ahvazi, 1998; Sobel et al., 1998; Tanaka-Kataoka et al., 1999; Brod, 2002) has been revisited as IFN-I expression in insulin-producing β cells exacerbates T1D progression (Stewart et al., 1993; Alba et al., 2004). Furthermore, whereas pDCs were demonstrated to secrete IFN-I in dLNs from 3-week-old Non-Obese Diabetic (NOD) mice, pDC depletion in this strain dampened IFN-I production and diabetes progression. This protective effect is seemingly IFN-I dependent, since IFNAR blockade equally delayed diabetes onset (Li et al., 2008). Finally, Diana et al. recently formally demonstrated that IFN-α-producing pDCs are required for the initiation of diabetogenic T cell responses and T1D development. The study found that spontaneous β-cell death in young NOD mice induces the recruitment of B-1a cells, neutrophils, and pDCs in the pancreas. B-1a cells secrete anti-dsDNA IgGs which activate neutrophils that in turn release DNA-binding CRAMP (cathelicidin related anti-microbial peptide) in NETs. These immune complexes activate pDCs through TLR9, leading to local IFN-α production. Using depleting antibodies, the authors further demonstrated that IFN-α producing pDCs are essential to initiate T1D in NOD mice (Diana et al., 2013).

Adaptive Plasmacytoid DC Functions

pDCs Function as Bona Fide Ag Presenting Cells

Accumulating evidence has revealed that pDCs can function as Ag presenting cells (APCs). In steady-state, pDCs can be easily detected in the blood, the thymus, and all SLOs (Bendriss-Vermare et al., 2001; Nakano et al., 2001; Summers et al., 2001; Asselin-Paturel et al., 2003; Seth et al., 2011). Upon inflammation, pDCs are recruited on a massive scale to infected or inflamed tissues, as well as to associated dLNs, and, importantly, in the LN T cell area, supporting a role for pDCs in activating naïve T cells (Cella et al., 1999; Krug et al., 2002; Vanbervliet et al., 2003; Irla et al., 2010). In vitro, pDCs exhibit the ability to capture, process, and present Ags through MHCI and MHCII molecules (Villadangos and Young, 2008; Tel et al., 2012). Steady-state pDCs were mainly described to be tolerogenic. Moreover, following TLR activation, pDCs upregulate MHCII and costimulatory molecules, which allow the direct modulation of the adaptive immune response. However, distinct mechanisms regulate Ag capture and processing in cDCs and pDCs, as well as resulting T cell outcome, suggesting complementary and, for the most part, non-overlapping functions of these two DC subsets.

Intracellularly Ag derived peptides either expressed by the cell itself (Krug et al., 2003; Young et al., 2008) or derived from intracellular-virus (Fonteneau et al., 2003; Salio et al., 2004; Schlecht et al., 2004; McGill et al., 2008; Young et al., 2008) are efficiently presented by pDCs through MHCI. Whether pDCs can phagocytose bacteria is still unclear (Villadangos and Young, 2008), while they have been shown to endocytose virions and exogenous proteins. pDCs express various endocytic receptors: Siglec-H (Zhang et al., 2006); the tetherin BST-2 (CD317) (Neil et al., 2008; Viswanathan et al., 2011); DCIR, which mediates clathrin-dependent endocytosis (Meyer-Wentrup et al., 2008); FcgRII (CD32), which allows the uptake of opsonized-Ags (Benitez-Ribas et al., 2006; Bjorck et al., 2008); and, specific to human pDCs, BDCA-2 which allows Ag delivery in the Ag processing compartment (Dzionek et al., 2001). Other receptors, such as ILT-7 (Cao et al., 2006) or NKp44 (Brown et al., 2004) may mediate endocytosis by pDCs, although this hypothesis needs further experimental confirmation (Villadangos and Young, 2008). Interestingly, ligation of these endocytic receptors potently suppresses IFN-I production by pDCs after TLR9 triggering (Dzionek et al., 2001; Blasius et al., 2004; Cao et al., 2007; Meyer-Wentrup et al., 2008). Finally, pDCs also acquire Ags from exosomes or apoptotic bodies (Hoeffel et al., 2007; Bastos-Amador et al., 2012).

Recent data suggests that mouse and human pDCs efficiently cross-present exogenous Ags to CD8+ T cells. In vitro, TLR-activated pDCs capture, process, and cross-present exogenous proteins to CD8+ T cells to induce proliferation, IFN-γ production, and cytolytic activity (Mouries et al., 2008; Kool et al., 2011). Accordingly, in vivo OVA delivery to CpG-activated pDCs via Siglec-H induces the generation of Ag-specific CD8+ T cells (Zhang et al., 2006), although T cell effector functions were not investigated in this study. More recently, it was shown that pDC-depleted Siglec-H-DTR mice immunized with OVA protein in the presence of TLR ligands exhibit impaired OVA-specific CD8+ T cell proliferation. Furthermore, the generation of MHC-I-OVA tetramer+ CD44hi CD8+ T cells, as well as OVA-specific effector CTL functions, are impaired after pDC depletion (Takagi et al., 2011). However, this study cannot exclude that decreased CD8+ T cell activation and differentiation observed following pDC depletion does not reflect the abrogation of pDC cross-presenting functions, rather than simply the absence of pDC-mediated licensing of cDCs (Yoneyama et al., 2005). Accordingly, the depletion of cDCs in mice further co-immunized with OVA protein and CpG completely abrogates OVA-specific CD8+ T cell responses, suggesting that pDCs do not cross-present exogenous proteins to CD8+ T cells (Sapoznikov et al., 2007). Thus, the ability of murine pDCs to cross-present Ags to CD8+ T cells remains controversial. In humans however, it is accepted that blood pDCs efficiently cross-present viral Ags and initiate Ag-specific antiviral CD8+ T cell responses after being exposed to either influenza virus (Di Pucchio et al., 2008) or HIV-1 infected apoptotic cells (Hoeffel et al., 2007; Lui et al., 2009). The ability of pDCs to cross-present viral Ags does not seem to require IFN-I production but this function is strongly enhanced after TLR activation with synthetic compounds or influenza virus infection (Hoeffel et al., 2007).

Although some studies cited above suggested that the efficiency of Ag cross-presentation by pDCs to CD8+ T cells was comparable to that of cDCs, pDCs appear to be much less potent APCs compared to cDCs in stimulating CD4+ T cells. For instance, both in vitro and in vivo, CpG-activated pDCs were found not to be as efficient as cDCs at presenting exogenous Ag through MHCII to CD4+ T cells (Young et al., 2008; Kool et al., 2011). One possible explanation is that mouse cDCs and pDCs exhibit major differences in their Ag presentation machinery. Notably, MHCII molecules are differentially regulated in pDCs and cDCs. MHCII expression is regulated by the class II master transactivator gene CIITA, itself expressed under the control of distinct cell specific promoters (Reith et al., 2005). In particular, pDCs rely strictly on the B cell promoter pIII, whereas macrophages and cDCs depend on pI (LeibundGut-Landmann et al., 2004). In contrast to cDCs, activated pDCs fail to shut down MHCII synthesis (LeibundGut-Landmann et al., 2004) and turnover (Young et al., 2008), thereby allowing continued Ag presentation after activation. These features may indeed render pDCs better equipped for the continuous presentation of Ags, and allow them to present newly formed MHCII-peptide complexes even after activation (Sadaka et al., 2009), a feature with obvious advantages at sites of infection (Villadangos and Young, 2008). On the other hand, since pDCs lack the ability to accumulate long-lived MHCII-peptide complexes generated shortly after activation, it may also render these cells less efficient compared to cDCs at promoting effector CD4+ T cell responses, due to the low the exposure time of the peptide on cell surface.

Impact on T Helper Functions

As discussed above, activated pDCs present Ags to naïve CD4+ T cells and thus directly contribute to T cell responses as APCs. In addition, pDCs can also indirectly impact T cells by producing inflammatory cytokines. As most studies eliminate pDCs genetically (Swiecki et al., 2010; Takagi et al., 2011) or through depleting mAbs (Bailey-Bucktrout et al., 2008; Jongbloed et al., 2009), it has been difficult so far to decipher the relative contribution of innate and adaptive pDC functions. However, accumulating evidence, mainly supported by in vitro studies, in vivo Ag targeting and abrogation of MHCII expression on pDCs, suggest a direct role of pDCs in impacting T cell responses. It is largely accepted that steady-state cDCs constantly present self- and non-self-Ags in a fashion that promotes T cell tolerance. Conversely, signals derived from pathogens or tissue damage generally boost cDC maturation, which promotes their capacity to induce effector T cell responses (Steinman, 2007). For pDCs however, their ability to promote either tolerance or immunity seems not to rely entirely on their activation state. Indeed, whereas immature pDCs exclusively promote tolerance, activated pDCs, depending on the anatomical localization and the cytokine milieu, may have both immunogenic and tolerogenic functions, although the exact nature of these functions remains to be established.

Immunogenic pDCs

Previous studies performed in vitro showed that both human and mouse activated pDCs, given an antigenic peptide together with appropriate activating signals, activate naïve CD4+ T cells, and promote Th1 differentiation (Cella et al., 2000; Krug et al., 2001; Boonstra et al., 2003). In mice devoid of cDCs, CpG-treated LN pDCs promote Th1 development and memory differentiation in vivo (Sapoznikov et al., 2007). Similarly, Ag-specific delivery to TLR-activated pDCs via BST-2 induces Ag-specific Th1 development in vivo, as demonstrated by the production of IFN-γ by CD4+ T cells and subsequent immunoglobulin production (Loschko et al., 2011b) (Figure 1). pDCs have also been shown to induce Th17 responses in different experimental models. Human TLR7-triggered pDCs promote Th17 differentiation from either naïve or memory CD4+ T cells (Yu et al., 2010). In patients with GVHD, increased pDCs and Th17 cell numbers in the intestinal mucosa correlate with disease severity, suggesting a role of pDCs in driving Th17 responses during disease (Bossard et al., 2012). Furthermore, in tumor bearing mice, pDCs activation through CpG correlates with an increase of tumor-specific Th17 cells and an inhibition of the tumor growth (Xu et al., 2012) (Figure 1). The ability of pDCs to promote Th17 differentiation seems to be enhanced in the presence of TGF-β. Indeed, the transfer of TGF-β treated pDCs to collagen-induced arthritic mice leads to increased Th17 responses in LNs leading to increased disease severity (Bonnefoy et al., 2011). In EAE, anti-PDCA1 mediated pDC depletion resulted in impaired encephalitogenic Th17 responses significantly reducing early clinical scores (Isaksson et al., 2009). Moreover, pDCs may be able to convert Tregs into Th17 cells. Indeed, in rats, Foxp3+ T cells start to produce IL-17 when activated by mature pDCs (Gautreau et al., 2011). Using genetically modified mice selectively lacking MHCII expression by pDCs (LeibundGut-Landmann et al., 2004), we have found that CpG-activated pDCs function as APCs to induce Th17 responses in vivo (Guery et al., manuscript in preparation) (Figure 1). Our unpublished data further suggest that the ability of activated pDCs to promote Th17 cells may be used as a vaccination strategy against tumors.

FIGURE 1
www.frontiersin.org

Figure 1. Activated pDCs induce dual tolerogenic and immunogenic T cell responses. The most relevant in vivo experimental animal systems are represented in this figure.

Tolerogenic pDCs

Plasmacytoid DCs have also been demonstrated to be involved in the induction of central and peripheral tolerance. It has been suggested that the function of pDCs within the thymus, as in other tissues, might simply be to protect the tissue from viral infections (Wu and Shortman, 2005). However, it was recently suggested that recirculating pDCs might present self-Ags in the thymus and contribute to the inactivation, or deletion, of autoreactive T cells. pDCs were detected in human thymus, colocalize with Foxp3+ Tregs, and, when activated with CD40L plus IL-3, efficiently promote the development of Foxp3+ natural Tregs (nTregs) from autologous thymocytes (Martin-Gayo et al., 2010). Similarly, human thymic pDCs activated with CpG and TSLP induce nTreg generation (Hanabuchi et al., 2010). In these two studies, Tregs generated by pDCs produce more IL-10 and less TGF-β compared to nTregs primed by cDCs under the same conditions, suggesting a complementary effect of the two DC subsets in the development of central tolerance. However, whether pDCs actually promote thymocyte differentiation into nTreg in vivo remains to be firmly demonstrated. In contrast to human pDCs, murine thymic pDCs do not efficiently induce Treg differentiation from thymocytes in vitro (Proietto et al., 2009). In vivo, a recent study illustrated the importance of CCR9 in targeting peripheral immature pDCs to the thymus. This indicates a role for pDCs in presenting extrathymically acquired Ags to further induce the deletion of Ag-specific CD4+ thymocytes (Hadeiba et al., 2012). No role for pDCs in the generation of nTregs was observed in this study, suggesting that mouse pDCs are intrinsically inefficient at inducing Tregs in the thymic environment in vivo.

Although it was suggested that pDCs are poor presenters of Ag in the absence of microbial stimulation (Colonna et al., 2004), immature pDCs promote T cell anergy, T cell deletion, as well as Treg differentiation. In humans, freshly isolated pDCs induce anergy in human CD4+ T cell clones, but it is reversed upon the addition of exogenous IL-2 (Kuwana, 2002). In mice, pDCs prevent oral T cell priming and are responsible for systemic tolerance to dietary Ags including proteins and haptens (Goubier et al., 2008). Indeed, pDC depletion induces hypersensitivity and CD8+ T cell responses toward oral Ags. In contrast, the transfer of Ag-loaded immature pDCs in naive mice suppresses both Ag-specific CD4+ and CD8+ T cell responses by inducing either anergy or deletion, suggesting that oral tolerance relies on Ag presentation by pDCs (Goubier et al., 2008) (Figure 2). Immature pDCs expressing CCR9 were defined as inducers of Foxp3+ Tregs that suppress Ag-specific immune response in a GVHD model. Importantly, transferred CCR9+ pDCs efficiently suppressed allogenic GVDH (Hadeiba et al., 2008). pDCs were also identified as phagocytic APCs essential for tolerance to vascularized cardiac allografts (Ochando et al., 2006) (Figure 2). In this model, alloantigen-presenting pDCs home to the LNs under tolerogenic conditions, where they mediate alloantigen-specific Treg development and allograft tolerance. In an other model of cardiac allograft transplantation, the absence of pDCs in LNs from CCR7−/− mice impairs Treg induction, and results in graft rejection, whereas pDC transfer restores both Treg frequencies and tolerance to the cardiac allograft (Liu et al., 2011) (Figure 2). In rats, pDCs also induce tolerance to allografts by inhibiting CD4+ T cells either directly through an indoleamine 2,3-dioxygenase (IDO)-dependent mechanism, or indirectly through the induction of CD8+ Tregs (Li et al., 2010) (Figure 2). Furthermore, pDC depletion in mice induces classical features of asthma after inhalation of an inert Ag, including IgE sensitization, airway eosinophilia, goblet cell hyperplasia, and Th2 cytokine production. In contrast, the adoptive transfer of pDCs before sensitization prevents asthma, possibly through the induction of Tregs (de Heer et al., 2004). Using Siglec-H inducible deficient mice, a recent study nicely showed that Siglec-H controls the ability of steady-state pDCs to induce the conversion of naïve CD4+ T cells in inducible Tregs (iTregs) in vivo (Takagi et al., 2011). In the contexts of tumors, the microenvironment maintains a resting pDC phenotype, characterized by low expression of costimulatory molecules and low IFN-I production (Hartmann et al., 2003; Conrad et al., 2012; Sisirak et al., 2012). Consequently, pDCs induce mainly tolerogenic tumor CD4+ T cell responses, through IDO-dependent Treg generation (Sharma et al., 2007) (Figure 2). Accordingly, IDO inhibition in pDCs promotes the conversion of Tregs into Th17 cells that efficiently inhibit tumor growth (Sharma et al., 2009). ICOS-L expression by pDCs in the tumor context has also been implicated in their ability to generate Tregs (Conrad et al., 2012; Faget et al., 2012) (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2. Immature pDCs induce tolergenic T cell responses. The most relevant in vivo experimental animal systems are represented in this figure.

Activated pDCs were demonstrated to favor Treg development. Treg induction has been correlated with low peptide-MHCII densities on APCs (Kang et al., 2007; Turner et al., 2009). Thus, the ability of activated pDCs to induce Tregs might be explained by the fact that they do not stabilize peptide-MHCII complexes at their cell surface, and, thus, provide a weak TCR engagement promoting Treg development. In humans, TLR-activated pDCs induce the development of IL-10 producing Tregs in an ICOS dependent manner (Ito et al., 2007; Ogata et al., 2013). Furthermore, HIV-stimulated IDO expressing pDCs induce the differentiation of naive CD4+ T cells into Tregs (Manches et al., 2008). CpG-activated human pDCs can also promote Treg differentiation (Moseman et al., 2004), possibly in an IDO-dependent fashion (Chen et al., 2008). Finally, TLR-activated rat pDCs induce Treg proliferation in vitro (Ouabed et al., 2008).

The severity of several autoimmune diseases has been demonstrated to be regulated by pDCs. In rheumatoid arthritis, mature pDCs from patients express high levels of IDO and are necessary for the differentiation of allogeneic naïve CD4+ CD25 T cells into IL-10 producing Treg (Tr1) cells (Kavousanaki et al., 2010). Moreover, in an in vivo mouse model of arthritis, pDC depletion correlates with enhanced articular pathology and increased T and B cell autoimmune responses to type II collagen (Jongbloed et al., 2009). In lupus, low dose Ag therapy induces the production of TGF-β by pDCs and dampens their ability to respond to TLR stimulation. The transfer of these tolerogenic pDCs promotes Treg expansion while simultaneously suppressing inflammatory Th17 infiltrating the kidney of lupus-prone mice (Kang et al., 2007). Ag targeting in pDCs has also been shown to inhibit T helper cell dependent autoimmunity. In EAE, Siglec-H mediated MOG35–55 delivery to pDCs dampens EAE, by inducing MOG-specific CD4+ T cell hyporesponsiveness resulting in the impaired induction of Th1 and Th17 cells, without promoting Treg differentiation (Loschko et al., 2011a) (Figure 1). Using a genetic mouse model in which MHCII is specifically abrogated in pDCs, we also identified a tolerogenic role for Ag presenting pDC functions during EAE. We demonstrated that pDCs, by presenting myelin Ags to naïve CD4+ T cells, induce the expansion of nTregs. As a consequence, mice carrying a selective abrogation of MHCII on pDCs exhibit impaired nTreg expansion, increased encephalitogenic Th1 and Th17 responses and exacerbated EAE (Irla et al., 2010) (Figure 1).

As discussed before, pDCs contribute to the pathology of T1D through the production of IFN-I. However, pDCs are also implicated in the inhibition of diabetogenic T cells during infections. In RIP-LCMV mice, OX40-OX40L dependent pDC–iNKT cell interactions control viral replication in pancreatic islets of LCMV infected mice by inducing IFN-I production by pDCs (Diana et al., 2009). In addition, this pDC–iNKT cooperation has been reported to promote TGF-β production by pDCs, that in turn, acquire the ability to convert naive anti-islet CD4+ T cells into Foxp3+ Tregs in pancreatic dLNs. These Tregs are then recruited in the pancreatic islets where they produce TGF-β that inhibits islet-specific CD8+ T cells and dampens T1D severity (Diana et al., 2011). In NOD mice, pDC depletion leads to accelerated insulitis and disease onset, and correlates with a local loss of IDO, suggesting that during T1D, pDCs may exert IDO-dependent tolerogenic functions by regulating islet-specific CD4+ T cell responses (Saxena et al., 2007).

In summary, pDCs in SLO were shown to contribute to T cell tolerance in several experimental systems, regardless of whether they exhibit a steady-state or activated phenotype (Figures 1 and 2). Interestingly, tolerogenic pDC functions were demonstrated to be dependent on IDO in several models. Furthermore, IDO contribution does not seem to rely on a pDC inflammatory environment. Why and how IDO is involved in the ability of pDCs to dampen some, but not all, Ag-specific T cell responses is still a matter of debate. This phenomenon is more complex due to recent findings demonstrating a second function for pDC-derived IDO. Together with the tryptophan catalytic activity inhibiting effector T cell function, IDO has been postulated to act as a signaling protein in response to TGF-β inducing the conversion of naïve CD4+ T cells into Tregs (Pallotta et al., 2011). Thus, the relative contribution of the dual IDO functions needs to be addressed in the different models where an IDO-dependent inhibition of T cell responses by pDCs has been described.

Concluding Remarks

Thus, steady-state pDCs exclusively promote T cell tolerance. However, the emerging picture of pDC functions during the development of inflammatory autoimmune disorders is that they contribute to disease pathogenesis by the production of IFN-I, while promoting self-Ag-specific CD4+ T cell tolerance though their ability to present auto-Ag. Altogether, studies show that innate and adaptive pDC functions may have opposite effects on T cell tolerance toward self-tissues. Interestingly, the engagement of endocytic receptors favors Ag presenting pDC functions, while it dampens their ability to produce IFN-I, suggesting that Ag targeting in pDCs would represent an attractive therapeutical strategy to control autoimmunity and graft rejection.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Walter G. Ferlin and Boris Lee for critical reading of the review. Work in the laboratory is supported by the Swiss National Science Foundation (grant PP00P3_128394), the European Research Council (pROsPeCT 281365), the Leenaards Foundation, and the MS Swiss Society.

References

Alba, A., Puertas, M. C., Carrillo, J., Planas, R., Ampudia, R., Pastor, X., et al. (2004). IFN beta accelerates autoimmune type 1 diabetes in nonobese diabetic mice and breaks the tolerance to beta cells in nondiabetes-prone mice. J. Immunol. 173, 6667–6675.

Pubmed Abstract | Pubmed Full Text

Asselin-Paturel, C., Brizard, G., Pin, J. J., Briere, F., and Trinchieri, G. (2003). Mouse strain differences in plasmacytoid dendritic cell frequency and function revealed by a novel monoclonal antibody. J. Immunol. 171, 6466–6477.

Pubmed Abstract | Pubmed Full Text

Bailey, S. L., Schreiner, B., McMahon, E. J., and Miller, S. D. (2007). CNS myeloid DCs presenting endogenous myelin peptides “preferentially” polarize CD4+ T(H)-17 cells in relapsing EAE. Nat. Immunol. 8, 172–180.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bailey-Bucktrout, S. L., Caulkins, S. C., Goings, G., Fischer, J. A., Dzionek, A., and Miller, S. D. (2008). Cutting edge: central nervous system plasmacytoid dendritic cells regulate the severity of relapsing experimental autoimmune encephalomyelitis. J. Immunol. 180, 6457–6461.

Pubmed Abstract | Pubmed Full Text

Barber, G. N., Wambach, M., Wong, M. L., Dever, T. E., Hinnebusch, A. G., and Katze, M. G. (1993). Translational regulation by the interferon-induced double-stranded-RNA-activated 68-kDa protein kinase. Proc. Natl. Acad. Sci. U.S.A. 90, 4621–4625.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Barrat, F. J., Meeker, T., Gregorio, J., Chan, J. H., Uematsu, S., Akira, S., et al. (2005). Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus erythematosus. J. Exp. Med. 202, 1131–1139.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Barton, G. M., Kagan, J. C., and Medzhitov, R. (2006). Intracellular localization of Toll-like receptor 9 prevents recognition of self DNA but facilitates access to viral DNA. Nat. Immunol. 7, 49–56.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bastos-Amador, P., Perez-Cabezas, B., Izquierdo-Useros, N., Puertas, M. C., Martinez-Picado, J., Pujol-Borrell, R., et al. (2012). Capture of cell-derived microvesicles (exosomes and apoptotic bodies) by human plasmacytoid dendritic cells. J. Leukoc. Biol. 91, 751–758.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bauer, S., Kirschning, C. J., Hacker, H., Redecke, V., Hausmann, S., Akira, S., et al. (2001). Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proc. Natl. Acad. Sci. U.S.A. 98, 9237–9242.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bendriss-Vermare, N., Barthelemy, C., Durand, I., Bruand, C., Dezutter-Dambuyant, C., Moulian, N., et al. (2001). Human thymus contains IFN-alpha-producing CD11c(−), myeloid CD11c(+), and mature interdigitating dendritic cells. J. Clin. Invest. 107, 835–844.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Benitez-Ribas, D., Adema, G. J., Winkels, G., Klasen, I. S., Punt, C. J., Figdor, C. G., et al. (2006). Plasmacytoid dendritic cells of melanoma patients present exogenous proteins to CD4+ T cells after Fc gamma RII-mediated uptake. J. Exp. Med. 203, 1629–1635.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bennett, L., Palucka, A. K., Arce, E., Cantrell, V., Borvak, J., Banchereau, J., et al. (2003). Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J. Exp. Med. 197, 711–723.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bjorck, P., Beilhack, A., Herman, E. I., Negrin, R. S., and Engleman, E. G. (2008). Plasmacytoid dendritic cells take up opsonized antigen leading to CD4+ and CD8+ T cell activation in vivo. J. Immunol. 181, 3811–3817.

Pubmed Abstract | Pubmed Full Text

Blanco, P., Palucka, A. K., Gill, M., Pascual, V., and Banchereau, J. (2001). Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science 294, 1540–1543.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Blasius, A., Vermi, W., Krug, A., Facchetti, F., Cella, M., and Colonna, M. (2004). A cell-surface molecule selectively expressed on murine natural interferon-producing cells that blocks secretion of interferon-alpha. Blood 103, 4201–4206.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bonnefoy, F., Couturier, M., Clauzon, A., Remy-Martin, J. P., Gaugler, B., Tiberghien, P., et al. (2011). TGF-beta-exposed plasmacytoid dendritic cells participate in Th17 commitment. J. Immunol. 186, 6157–6164.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Boonstra, A., Asselin-Paturel, C., Gilliet, M., Crain, C., Trinchieri, G., Liu, Y. J., et al. (2003). Flexibility of mouse classical and plasmacytoid-derived dendritic cells in directing T helper type 1 and 2 cell development: dependency on antigen dose and differential toll-like receptor ligation. J. Exp. Med. 197, 101–109.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bossard, C., Malard, F., Arbez, J., Chevallier, P., Guillaume, T., Delaunay, J., et al. (2012). Plasmacytoid dendritic cells and Th17 immune response contribution in gastrointestinal acute graft-versus-host disease. Leukemia 26, 1471–1474.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Brod, S. A. (2002). Ingested type I interferon: a potential treatment for autoimmunity. J. Interferon Cytokine Res. 22, 1153–1166.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Brown, D., Trowsdale, J., and Allen, R. (2004). The LILR family: modulators of innate and adaptive immune pathways in health and disease. Tissue Antigens 64, 215–225.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cao, W., Rosen, D. B., Ito, T., Bover, L., Bao, M., Watanabe, G., et al. (2006). Plasmacytoid dendritic cell-specific receptor ILT7-Fc epsilonRI gamma inhibits Toll-like receptor-induced interferon production. J. Exp. Med. 203, 1399–1405.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cao, W., Zhang, L., Rosen, D. B., Bover, L., Watanabe, G., Bao, M., et al. (2007). BDCA2/Fc epsilon RI gamma complex signals through a novel BCR-like pathway in human plasmacytoid dendritic cells. PLoS Biol. 5:e248. doi:10.1371/journal.pbio.0050248

CrossRef Full Text

Cella, M., Facchetti, F., Lanzavecchia, A., and Colonna, M. (2000). Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization. Nat. Immunol. 1, 305–310.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cella, M., Jarrossay, D., Facchetti, F., Alebardi, O., Nakajima, H., Lanzavecchia, A., et al. (1999). Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat. Med. 5, 919–923.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cervantes-Barragan, L., Lewis, K. L., Firner, S., Thiel, V., Hugues, S., Reith, W., et al. (2012). Plasmacytoid dendritic cells control T-cell response to chronic viral infection. Proc. Natl. Acad. Sci. U.S.A. 109, 3012–3017.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chan, V. S., Nie, Y. J., Shen, N., Yan, S., Mok, M. Y., and Lau, C. S. (2012). Distinct roles of myeloid and plasmacytoid dendritic cells in systemic lupus erythematosus. Autoimmun. Rev. 11, 890–897.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chehimi, J., Starr, S. E., Kawashima, H., Miller, D. S., Trinchieri, G., Perussia, B., et al. (1989). Dendritic cells and IFN-alpha-producing cells are two functionally distinct non-B, non-monocytic HLA-DR+ cell subsets in human peripheral blood. Immunology 68, 486–490.

Pubmed Abstract | Pubmed Full Text

Chen, L., Arora, M., Yarlagadda, M., Oriss, T. B., Krishnamoorthy, N., Ray, A., et al. (2006). Distinct responses of lung and spleen dendritic cells to the TLR9 agonist CpG oligodeoxynucleotide. J. Immunol. 177, 2373–2383.

Pubmed Abstract | Pubmed Full Text

Chen, W., Liang, X., Peterson, A. J., Munn, D. H., and Blazar, B. R. (2008). The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J. Immunol. 181, 5396–5404.

Pubmed Abstract | Pubmed Full Text

Cisse, B., Caton, M. L., Lehner, M., Maeda, T., Scheu, S., Locksley, R., et al. (2008). Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell 135, 37–48.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Colina, R., Costa-Mattioli, M., Dowling, R. J., Jaramillo, M., Tai, L. H., Breitbach, C. J., et al. (2008). Translational control of the innate immune response through IRF-7. Nature 452, 323–328.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Colonna, M., Trinchieri, G., and Liu, Y. J. (2004). Plasmacytoid dendritic cells in immunity. Nat. Immunol. 5, 1219–1226.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Conrad, C., Gregorio, J., Wang, Y. H., Ito, T., Meller, S., Hanabuchi, S., et al. (2012). Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. 72, 5240–5249.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

de Heer, H. J., Hammad, H., Soullie, T., Hijdra, D., Vos, N., Willart, M. A., et al. (2004). Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen. J. Exp. Med. 200, 89–98.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Di Pucchio, T., Chatterjee, B., Smed-Sorensen, A., Clayton, S., Palazzo, A., Montes, M., et al. (2008). Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class I. Nat. Immunol. 9, 551–557.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Diana, J., Brezar, V., Beaudoin, L., Dalod, M., Mellor, A., Tafuri, A., et al. (2011). Viral infection prevents diabetes by inducing regulatory T cells through NKT cell-plasmacytoid dendritic cell interplay. J. Exp. Med. 208, 729–745.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Diana, J., Griseri, T., Lagaye, S., Beaudoin, L., Autrusseau, E., Gautron, A. S., et al. (2009). NKT cell-plasmacytoid dendritic cell cooperation via OX40 controls viral infection in a tissue-specific manner. Immunity 30, 289–299.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Diana, J., Simoni, Y., Furio, L., Beaudoin, L., Agerberth, B., Barrat, F., et al. (2013). Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat. Med. 19, 65–73.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S., and Reis e Sousa, C. (2004). Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303, 1529–1531.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Dreux, M., Garaigorta, U., Boyd, B., Decembre, E., Chung, J., Whitten-Bauer, C., et al. (2012). Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host Microbe 12, 558–570.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Dzionek, A., Sohma, Y., Nagafune, J., Cella, M., Colonna, M., Facchetti, F., et al. (2001). BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction. J. Exp. Med. 194, 1823–1834.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Eidenschenk, C., Crozat, K., Krebs, P., Arens, R., Popkin, D., Arnold, C. N., et al. (2010). Flt3 permits survival during infection by rendering dendritic cells competent to activate NK cells. Proc. Natl. Acad. Sci. U.S.A. 107, 9759–9764.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Facchetti, F., de Wolf-Peeters, C., Mason, D. Y., Pulford, K., van den Oord, J. J., and Desmet, V. J. (1988). Plasmacytoid T cells. Immunohistochemical evidence for their monocyte/macrophage origin. Am. J. Path. 133, 15–21.

Faget, J., Bendriss-Vermare, N., Gobert, M., Durand, I., Olive, D., Biota, C., et al. (2012). ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells. Cancer Res. 72, 6130–6141.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Fitzgerald-Bocarsly, P. (1993). Human natural interferon-alpha producing cells. Pharmacol. Ther. 60, 39–62.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Fonteneau, J. F., Gilliet, M., Larsson, M., Dasilva, I., Munz, C., Liu, Y. J., et al. (2003). Activation of influenza virus-specific CD4+ and CD8+ T cells: a new role for plasmacytoid dendritic cells in adaptive immunity. Blood 101, 3520–3526.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Fonteneau, J. F., Larsson, M., Beignon, A. S., McKenna, K., Dasilva, I., Amara, A., et al. (2004). Human immunodeficiency virus type 1 activates plasmacytoid dendritic cells and concomitantly induces the bystander maturation of myeloid dendritic cells. J. Virol. 78, 5223–5232.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Garcia-Romo, G. S., Caielli, S., Vega, B., Connolly, J., Allantaz, F., Xu, Z., et al. (2011). Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra20.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gautreau, L., Chabannes, D., Heslan, M., and Josien, R. (2011). Modulation of regulatory T cell-Th17 balance by plasmacytoid dendritic cells. J. Leukoc. Biol. 90, 521–527.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gerosa, F., Gobbi, A., Zorzi, P., Burg, S., Briere, F., Carra, G., et al. (2005). The reciprocal interaction of NK cells with plasmacytoid or myeloid dendritic cells profoundly affects innate resistance functions. J. Immunol. 174, 727–734.

Pubmed Abstract | Pubmed Full Text

Ghosh, H. S., Cisse, B., Bunin, A., Lewis, K. L., and Reizis, B. (2010). Continuous expression of the transcription factor e2-2 maintains the cell fate of mature plasmacytoid dendritic cells. Immunity 33, 905–916.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gilliet, M., Cao, W., and Liu, Y. J. (2008). Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nat. Rev. Immunol. 8, 594–606.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gilliet, M., Conrad, C., Geiges, M., Cozzio, A., Thurlimann, W., Burg, G., et al. (2004). Psoriasis triggered by toll-like receptor 7 agonist imiquimod in the presence of dermal plasmacytoid dendritic cell precursors. Arch. Dermatol. 140, 1490–1495.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Goodkin, D. E. (1996). Interferon beta treatment for multiple sclerosis: persisting questions. Mult. Scler. 1, 321–324.

Pubmed Abstract | Pubmed Full Text

Goubier, A., Dubois, B., Gheit, H., Joubert, G., Villard-Truc, F., Asselin-Paturel, C., et al. (2008). Plasmacytoid dendritic cells mediate oral tolerance. Immunity 29, 464–475.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Grouard, G., Rissoan, M. C., Filgueira, L., Durand, I., Banchereau, J., and Liu, Y. J. (1997). The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 185, 1101–1111. doi:10.1371/journal.pone.0007111

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Guiducci, C., Ott, G., Chan, J. H., Damon, E., Calacsan, C., Matray, T., et al. (2006). Properties regulating the nature of the plasmacytoid dendritic cell response to Toll-like receptor 9 activation. J. Exp. Med. 203, 1999–2008.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Guo, B., Chang, E. Y., and Cheng, G. (2008). The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J. Clin. Invest. 118, 1680–1690.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hadeiba, H., Lahl, K., Edalati, A., Oderup, C., Habtezion, A., Pachynski, R., et al. (2012). Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity 36, 438–450.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hadeiba, H., Sato, T., Habtezion, A., Oderup, C., Pan, J., and Butcher, E. C. (2008). CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nat. Immunol. 9, 1253–1260.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hanabuchi, S., Ito, T., Park, W. R., Watanabe, N., Shaw, J. L., Roman, E., et al. (2010). Thymic stromal lymphopoietin-activated plasmacytoid dendritic cells induce the generation of FOXP3+ regulatory T cells in human thymus. J. Immunol. 184, 2999–3007.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hartmann, E., Wollenberg, B., Rothenfusser, S., Wagner, M., Wellisch, D., Mack, B., et al. (2003). Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer. Cancer Res. 63, 6478–6487.

Pubmed Abstract | Pubmed Full Text

Heil, F., Hemmi, H., Hochrein, H., Ampenberger, F., Kirschning, C., Akira, S., et al. (2004). Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303, 1526–1529.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hemmi, H., Takeuchi, O., Kawai, T., Kaisho, T., Sato, S., Sanjo, H., et al. (2000). A Toll-like receptor recognizes bacterial DNA. Nature 408, 740–745.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Henault, J., Martinez, J., Riggs, J. M., Tian, J., Mehta, P., Clarke, L., et al. (2012). Noncanonical autophagy is required for type I interferon secretion in response to DNA-immune complexes. Immunity 37, 986–997.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hibbert, L., Pflanz, S., De Waal Malefyt, R., and Kastelein, R. A. (2003). IL-27 and IFN-alpha signal via Stat1 and Stat3 and induce T-Bet and IL-12Rbeta2 in naive T cells. J. Interferon Cytokine Res. 23, 513–522.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hoeffel, G., Ripoche, A. C., Matheoud, D., Nascimbeni, M., Escriou, N., Lebon, P., et al. (2007). Antigen cross presentation by human plasmacytoid dendritic cells. Immunity 27, 481–492.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Honda, K., Ohba, Y., Yanai, H., Negishi, H., Mizutani, T., Takaoka, A., et al. (2005a). Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature 434, 1035–1040.

CrossRef Full Text

Honda, K., Yanai, H., Negishi, H., Asagiri, M., Sato, M., Mizutani, T., et al. (2005b). IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434, 772–777.

CrossRef Full Text

Hornung, V., Rothenfusser, S., Britsch, S., Krug, A., Jahrsdorfer, B., Giese, T., et al. (2002). Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J. Immunol. 168, 4531–4537.

Pubmed Abstract | Pubmed Full Text

Irla, M., Kupfer, N., Suter, T., Lissilaa, R., Benkhoucha, M., Skupsky, J., et al. (2010). MHC class II-restricted antigen presentation by plasmacytoid dendritic cells inhibits T cell-mediated autoimmunity. J. Exp. Med. 207, 1891–1905.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Isaksson, M., Ardesjo, B., Ronnblom, L., Kampe, O., Lassmann, H., Eloranta, M. L., et al. (2009). Plasmacytoid DC promote priming of autoimmune Th17 cells and EAE. Eur. J. Immunol. 39, 2925–2935.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ito, T., Kanzler, H., Duramad, O., Cao, W., and Liu, Y. J. (2006). Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells. Blood 107, 2423–2431.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ito, T., Yang, M., Wang, Y. H., Lande, R., Gregorio, J., Perng, O. A., et al. (2007). Plasmacytoid dendritic cells prime IL-10-producing T regulatory cells by inducible costimulator ligand. J. Exp. Med. 204, 105–115.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jarrossay, D., Napolitani, G., Colonna, M., Sallusto, F., and Lanzavecchia, A. (2001). Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells. Eur. J. Immunol. 31, 3388–3393.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jego, G., Palucka, A. K., Blanck, J. P., Chalouni, C., Pascual, V., and Banchereau, J. (2003). Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity 19, 225–234.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jongbloed, S. L., Benson, R. A., Nickdel, M. B., Garside, P., McInnes, I. B., and Brewer, J. M. (2009). Plasmacytoid dendritic cells regulate breach of self-tolerance in autoimmune arthritis. J. Immunol. 182, 963–968.

Pubmed Abstract | Pubmed Full Text

Kadowaki, N., Ho, S., Antonenko, S., Malefyt, R. W., Kastelein, R. A., Bazan, F., et al. (2001). Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J. Exp. Med. 194, 863–869.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kang, H. K., Liu, M., and Datta, S. K. (2007). Low-dose peptide tolerance therapy of lupus generates plasmacytoid dendritic cells that cause expansion of autoantigen-specific regulatory T cells and contraction of inflammatory Th17 cells. J. Immunol. 178, 7849–7858.

Pubmed Abstract | Pubmed Full Text

Kavousanaki, M., Makrigiannakis, A., Boumpas, D., and Verginis, P. (2010). Novel role of plasmacytoid dendritic cells in humans: induction of interleukin-10-producing Treg cells by plasmacytoid dendritic cells in patients with rheumatoid arthritis responding to therapy. Arthritis Rheum. 62, 53–63.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kerkmann, M., Costa, L. T., Richter, C., Rothenfusser, S., Battiany, J., Hornung, V., et al. (2005). Spontaneous formation of nucleic acid-based nanoparticles is responsible for high interferon-alpha induction by CpG-A in plasmacytoid dendritic cells. J. Biol. Chem. 280, 8086–8093.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kool, M., Geurtsvankessel, C., Muskens, F., Madeira, F. B., van Nimwegen, M., Kuipers, H., et al. (2011). Facilitated antigen uptake and timed exposure to TLR ligands dictate the antigen-presenting potential of plasmacytoid DCs. J. Leukoc. Biol. 90, 1177–1190.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Krug, A., Towarowski, A., Britsch, S., Rothenfusser, S., Hornung, V., Bals, R., et al. (2001). Toll-like receptor expression reveals CpG DNA as a unique microbial stimulus for plasmacytoid dendritic cells which synergizes with CD40 ligand to induce high amounts of IL-12. Eur. J. Immunol. 31, 3026–3037.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Krug, A., Uppaluri, R., Facchetti, F., Dorner, B. G., Sheehan, K. C., Schreiber, R. D., et al. (2002). IFN-producing cells respond to CXCR3 ligands in the presence of CXCL12 and secrete inflammatory chemokines upon activation. J. Immunol. 169, 6079–6083.

Pubmed Abstract | Pubmed Full Text

Krug, A., Veeraswamy, R., Pekosz, A., Kanagawa, O., Unanue, E. R., Colonna, M., et al. (2003). Interferon-producing cells fail to induce proliferation of naive T cells but can promote expansion and T helper 1 differentiation of antigen-experienced unpolarized T cells. J. Exp. Med. 197, 899–906.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kuwana, M. (2002). Induction of anergic and regulatory T cells by plasmacytoid dendritic cells and other dendritic cell subsets. Hum. Immunol. 63, 1156–1163.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lande, R., Ganguly, D., Facchinetti, V., Frasca, L., Conrad, C., Gregorio, J., et al. (2011). Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra19.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lande, R., Gregorio, J., Facchinetti, V., Chatterjee, B., Wang, Y. H., Homey, B., et al. (2007). Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 449, 564–569.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Le Bon, A., Etchart, N., Rossmann, C., Ashton, M., Hou, S., Gewert, D., et al. (2003). Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat. Immunol. 4, 1009–1015.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lee, H. K., Lund, J. M., Ramanathan, B., Mizushima, N., and Iwasaki, A. (2007). Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science 315, 1398–1401.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

LeibundGut-Landmann, S., Waldburger, J. M., Reis e Sousa, C., Acha-Orbea, H., and Reith, W. (2004). MHC class II expression is differentially regulated in plasmacytoid and conventional dendritic cells. Nat. Immunol. 5, 899–908.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Li, Q., Xu, B., Michie, S. A., Rubins, K. H., Schreriber, R. D., and McDevitt, H. O. (2008). Interferon-alpha initiates type 1 diabetes in nonobese diabetic mice. Proc. Natl. Acad. Sci. U.S.A. 105, 12439–12444.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Li, X. L., Menoret, S., Bezie, S., Caron, L., Chabannes, D., Hill, M., et al. (2010). Mechanism and localization of CD8 regulatory T cells in a heart transplant model of tolerance. J. Immunol. 185, 823–833.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Liu, X., Mishra, P., Yu, S., Beckmann, J., Wendland, M., Kocks, J., et al. (2011). Tolerance induction towards cardiac allografts under costimulation blockade is impaired in CCR7-deficient animals but can be restored by adoptive transfer of syngeneic plasmacytoid dendritic cells. Eur. J. Immunol. 41, 611–623.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Loschko, J., Heink, S., Hackl, D., Dudziak, D., Reindl, W., Korn, T., et al. (2011a). Antigen targeting to plasmacytoid dendritic cells via Siglec-H inhibits Th cell-dependent autoimmunity. J. Immunol. 187, 6346–6356.

CrossRef Full Text

Loschko, J., Schlitzer, A., Dudziak, D., Drexler, I., Sandholzer, N., Bourquin, C., et al. (2011b). Antigen delivery to plasmacytoid dendritic cells via BST2 induces protective T cell-mediated immunity. J. Immunol. 186, 6718–6725.

CrossRef Full Text

Lovgren, T., Eloranta, M. L., Bave, U., Alm, G. V., and Ronnblom, L. (2004). Induction of interferon-alpha production in plasmacytoid dendritic cells by immune complexes containing nucleic acid released by necrotic or late apoptotic cells and lupus IgG. Arthritis Rheum. 50, 1861–1872.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lui, G., Manches, O., Angel, J., Molens, J. P., Chaperot, L., and Plumas, J. (2009). Plasmacytoid dendritic cells capture and cross-present viral antigens from influenza-virus exposed cells. PLoS ONE 4:e7111. doi:10.1371/journal.pone.0007111

CrossRef Full Text

Lund, J. M., Alexopoulou, L., Sato, A., Karow, M., Adams, N. C., Gale, N. W., et al. (2004). Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc. Natl. Acad. Sci. U.S.A. 101, 5598–5603.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Luo, X. M., and Lei, M. Y. (2012). Recombination activating gene-2 regulates cpg-mediated interferon-alpha production in mouse bone marrow-derived plasmacytoid dendritic cells. PLoS ONE 7:e47952. doi:10.1371/journal.pone.0047952

CrossRef Full Text

Malathi, K., Paranjape, J. M., Bulanova, E., Shim, M., Guenther-Johnson, J. M., Faber, P. W., et al. (2005). A transcriptional signaling pathway in the IFN system mediated by 2’-5’-oligoadenylate activation of RNase L. Proc. Natl. Acad. Sci. U.S.A. 102, 14533–14538.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Manches, O., Munn, D., Fallahi, A., Lifson, J., Chaperot, L., Plumas, J., et al. (2008). HIV-activated human plasmacytoid DCs induce Tregs through an indoleamine 2,3-dioxygenase-dependent mechanism. J. Clin. Invest. 118, 3431–3439.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Martin-Gayo, E., Sierra-Filardi, E., Corbi, A. L., and Toribio, M. L. (2010). Plasmacytoid dendritic cells resident in human thymus drive natural Treg cell development. Blood 115, 5366–5375.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Mathian, A., Gallegos, M., Pascual, V., Banchereau, J., and Koutouzov, S. (2011). Interferon-alpha induces unabated production of short-lived plasma cells in pre-autoimmune lupus-prone (NZBxNZW)F1 mice but not in BALB/c mice. Eur. J. Immunol. 41, 863–872.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

McGill, J., Van Rooijen, N., and Legge, K. L. (2008). Protective influenza-specific CD8 T cell responses require interactions with dendritic cells in the lungs. J. Exp. Med. 205, 1635–1646.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Meyer-Wentrup, F., Benitez-Ribas, D., Tacken, P. J., Punt, C. J., Figdor, C. G., de Vries, I. J., et al. (2008). Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood 111, 4245–4253.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Moseman, E. A., Liang, X., Dawson, A. J., Panoskaltsis-Mortari, A., Krieg, A. M., Liu, Y. J., et al. (2004). Human plasmacytoid dendritic cells activated by CpG oligodeoxynucleotides induce the generation of CD4+CD25+ regulatory T cells. J. Immunol. 173, 4433–4442.

Pubmed Abstract | Pubmed Full Text

Mouries, J., Moron, G., Schlecht, G., Escriou, N., Dadaglio, G., and Leclerc, C. (2008). Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation. Blood 112, 3713–3722.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nakano, H., Yanagita, M., and Gunn, M. D. (2001). CD11c(+)B220(+)Gr-1(+) cells in mouse lymph nodes and spleen display characteristics of plasmacytoid dendritic cells. J. Exp. Med. 194, 1171–1178.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Neil, S. J., Zang, T., and Bieniasz, P. D. (2008). Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 451, 425–430.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nestle, F. O., Conrad, C., Tun-Kyi, A., Homey, B., Gombert, M., Boyman, O., et al. (2005). Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J. Exp. Med. 202, 135–143.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ochando, J. C., Homma, C., Yang, Y., Hidalgo, A., Garin, A., Tacke, F., et al. (2006). Alloantigen-presenting plasmacytoid dendritic cells mediate tolerance to vascularized grafts. Nat. Immunol. 7, 652–662.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ogata, M., Ito, T., Shimamoto, K., Nakanishi, T., Satsutani, N., Miyamoto, R., et al. (2013). Plasmacytoid dendritic cells have a cytokine-producing capacity to enhance ICOS ligand-mediated IL-10 production during T-cell priming. Int. Immunol. 25, 171–182.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Onai, N., Obata-Onai, A., Schmid, M. A., Ohteki, T., Jarrossay, D., and Manz, M. G. (2007). Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8, 1207–1216.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ouabed, A., Hubert, F. X., Chabannes, D., Gautreau, L., Heslan, M., and Josien, R. (2008). Differential control of T regulatory cell proliferation and suppressive activity by mature plasmacytoid versus conventional spleen dendritic cells. J. Immunol. 180, 5862–5870.

Pubmed Abstract | Pubmed Full Text

Pallotta, M. T., Orabona, C., Volpi, C., Vacca, C., Belladonna, M. L., Bianchi, R., et al. (2011). Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat. Immunol. 12, 870–878.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Paquette, R. L., Hsu, N. C., Kiertscher, S. M., Park, A. N., Tran, L., Roth, M. D., et al. (1998). Interferon-alpha and granulocyte-macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells. J. Leukoc. Biol. 64, 358–367.

Pubmed Abstract | Pubmed Full Text

Pestka, S., Krause, C. D., and Walter, M. R. (2004). Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 202, 8–32.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pisetsky, D. S., and Fairhurst, A. M. (2007). The origin of extracellular DNA during the clearance of dead and dying cells. Autoimmunity 40, 281–284.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Prinz, M., Schmidt, H., Mildner, A., Knobeloch, K. P., Hanisch, U. K., Raasch, J., et al. (2008). Distinct and nonredundant in vivo functions of IFNAR on myeloid cells limit autoimmunity in the central nervous system. Immunity 28, 675–686.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Proietto, A. I., van Dommelen, S., and Wu, L. (2009). The impact of circulating dendritic cells on the development and differentiation of thymocytes. Immunol. Cell Biol. 87, 39–45.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Reith, W., LeibundGut-Landmann, S., and Waldburger, J. M. (2005). Regulation of MHC class II gene expression by the class II transactivator. Nat. Rev. Immunol. 5, 793–806.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Reizis, B., Bunin, A., Ghosh, H. S., Lewis, K. L., and Sisirak, V. (2011). Plasmacytoid dendritic cells: recent progress and open questions. Annu. Rev. Immunol. 29, 163–183.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ronnblom, L., Ramstedt, U., and Alm, G. V. (1983). Properties of human natural interferon-producing cells stimulated by tumor cell lines. Eur. J. Immunol. 13, 471–476.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sadaka, C., Marloie-Provost, M. A., Soumelis, V., and Benaroch, P. (2009). Developmental regulation of MHC II expression and transport in human plasmacytoid-derived dendritic cells. Blood 113, 2127–2135.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Salio, M., Palmowski, M. J., Atzberger, A., Hermans, I. F., and Cerundolo, V. (2004). CpG-matured murine plasmacytoid dendritic cells are capable of in vivo priming of functional CD8 T cell responses to endogenous but not exogenous antigens. J. Exp. Med. 199, 567–579.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sandgren, S., Wittrup, A., Cheng, F., Jonsson, M., Eklund, E., Busch, S., et al. (2004). The human antimicrobial peptide LL-37 transfers extracellular DNA plasmid to the nuclear compartment of mammalian cells via lipid rafts and proteoglycan-dependent endocytosis. J. Biol. Chem. 279, 17951–17956.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Santini, S. M., Lapenta, C., Logozzi, M., Parlato, S., Spada, M., Di Pucchio, T., et al. (2000). Type I interferon as a powerful adjuvant for monocyte-derived dendritic cell development and activity in vitro and in Hu-PBL-SCID mice. J. Exp. Med. 191, 1777–1788.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sapoznikov, A., Fischer, J. A., Zaft, T., Krauthgamer, R., Dzionek, A., and Jung, S. (2007). Organ-dependent in vivo priming of naive CD4+, but not CD8+, T cells by plasmacytoid dendritic cells. J. Exp. Med. 204, 1923–1933.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sathe, P., Vremec, D., Wu, L., Corcoran, L., and Shortman, K. (2013). Convergent differentiation: myeloid and lymphoid pathways to murine plasmacytoid dendritic cells. Blood 121, 11–19.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Saxena, V., Ondr, J. K., Magnusen, A. F., Munn, D. H., and Katz, J. D. (2007). The countervailing actions of myeloid and plasmacytoid dendritic cells control autoimmune diabetes in the nonobese diabetic mouse. J. Immunol. 179, 5041–5053.

Pubmed Abstract | Pubmed Full Text

Schlecht, G., Garcia, S., Escriou, N., Freitas, A. A., Leclerc, C., and Dadaglio, G. (2004). Murine plasmacytoid dendritic cells induce effector/memory CD8+ T-cell responses in vivo after viral stimulation. Blood 104, 1808–1815.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Seth, S., Oberdorfer, L., Hyde, R., Hoff, K., Thies, V., Worbs, T., et al. (2011). CCR7 essentially contributes to the homing of plasmacytoid dendritic cells to lymph nodes under steady-state as well as inflammatory conditions. J. Immunol. 186, 3364–3372.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sharma, M. D., Baban, B., Chandler, P., Hou, D. Y., Singh, N., Yagita, H., et al. (2007). Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Invest. 117, 2570–2582.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sharma, M. D., Hou, D. Y., Liu, Y., Koni, P. A., Metz, R., Chandler, P., et al. (2009). Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 113, 6102–6111.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Shigematsu, H., Reizis, B., Iwasaki, H., Mizuno, S., Hu, D., Traver, D., et al. (2004). Plasmacytoid dendritic cells activate lymphoid-specific genetic programs irrespective of their cellular origin. Immunity 21, 43–53.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Siegal, F. P., Kadowaki, N., Shodell, M., Fitzgerald-Bocarsly, P. A., Shah, K., Ho, S., et al. (1999). The nature of the principal type 1 interferon-producing cells in human blood. Science 284, 1835–1837.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sisirak, V., Faget, J., Gobert, M., Goutagny, N., Vey, N., Treilleux, I., et al. (2012). Impaired IFN-alpha production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 72, 5188–5197.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sobel, D. O., and Ahvazi, B. (1998). Alpha-interferon inhibits the development of diabetes in NOD mice. Diabetes 47, 1867–1872.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sobel, D. O., Creswell, K., Yoon, J. W., and Holterman, D. (1998). Alpha interferon administration paradoxically inhibits the development of diabetes in BB rats. Life Sci. 62, 1293–1302.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Steinman, R. M. (2007). Dendritic cells: understanding immunogenicity. Eur. J. Immunol. 37(Suppl. 1), S53–S60.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Stewart, T. A., Hultgren, B., Huang, X., Pitts-Meek, S., Hully, J., and MacLachlan, N. J. (1993). Induction of type I diabetes by interferon-alpha in transgenic mice. Science 260, 1942–1946.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Summers, K. L., Hock, B. D., McKenzie, J. L., and Hart, D. N. (2001). Phenotypic characterization of five dendritic cell subsets in human tonsils. Am. J. Pathol. 159, 285–295.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Swiecki, M., Gilfillan, S., Vermi, W., Wang, Y., and Colonna, M. (2010). Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8(+) T cell accrual. Immunity 33, 955–966.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Takagi, H., Fukaya, T., Eizumi, K., Sato, Y., Sato, K., Shibazaki, A., et al. (2011). Plasmacytoid dendritic cells are crucial for the initiation of inflammation and T cell immunity in vivo. Immunity 35, 958–971.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tanaka-Kataoka, M., Kunikata, T., Takayama, S., Iwaki, K., Fujii, M., Ohashi, K., et al. (1999). Oral use of interferon-alpha delays the onset of insulin-dependent diabetes mellitus in nonobese diabetes mice. J. Interferon Cytokine Res. 19, 877–879.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tel, J., van der Leun, A. M., Figdor, C. G., Torensma, R., and de Vries, I. J. (2012). Harnessing human plasmacytoid dendritic cells as professional APCs. Cancer Immunol. Immunother. 61, 1279–1288.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Thibault, D. L., Graham, K. L., Lee, L. Y., Balboni, I., Hertzog, P. J., and Utz, P. J. (2009). Type I interferon receptor controls B-cell expression of nucleic acid-sensing Toll-like receptors and autoantibody production in a murine model of lupus. Arthritis Res. Ther. 11, R112.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tian, J., Avalos, A. M., Mao, S. Y., Chen, B., Senthil, K., Wu, H., et al. (2007). Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat. Immunol. 8, 487–496.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Turner, M. S., Kane, L. P., and Morel, P. A. (2009). Dominant role of antigen dose in CD4+Foxp3+ regulatory T cell induction and expansion. J. Immunol. 183, 4895–4903.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Vanbervliet, B., Bendriss-Vermare, N., Massacrier, C., Homey, B., de Bouteiller, O., Briere, F., et al. (2003). The inducible CXCR3 ligands control plasmacytoid dendritic cell responsiveness to the constitutive chemokine stromal cell-derived factor 1 (SDF-1)/CXCL12. J. Exp. Med. 198, 823–830.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Villadangos, J. A., and Young, L. (2008). Antigen-presentation properties of plasmacytoid dendritic cells. Immunity 29, 352–361.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Viswanathan, K., Smith, M. S., Malouli, D., Mansouri, M., Nelson, J. A., and Fruh, K. (2011). BST2/tetherin enhances entry of human cytomegalovirus. PLoS Pathog. 7:e1002332. doi:10.1371/journal.ppat.1002332

CrossRef Full Text

Waskow, C., Liu, K., Darrasse-Jeze, G., Guermonprez, P., Ginhoux, F., Merad, M., et al. (2008). The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat. Immunol. 9, 676–683.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wu, L., and Shortman, K. (2005). Heterogeneity of thymic dendritic cells. Semin. Immunol. 17, 304–312.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Xu, L., Wang, C., Zhou, Y., Ren, T., and Wen, Z. (2012). CpG oligonucleotides induce the differentiation of CD4(+)Th17 cells by triggering plasmacytoid dendritic cells in adoptively cell transfer immunotherapy. Immunol. Lett. 142, 55–63.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Yoneyama, H., Matsuno, K., Toda, E., Nishiwaki, T., Matsuo, N., Nakano, A., et al. (2005). Plasmacytoid DCs help lymph node DCs to induce anti-HSV CTLs. J. Exp. Med. 202, 425–435.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Young, L. J., Wilson, N. S., Schnorrer, P., Proietto, A., ten Broeke, T., Matsuki, Y., et al. (2008). Differential MHC class II synthesis and ubiquitination confers distinct antigen-presenting properties on conventional and plasmacytoid dendritic cells. Nat. Immunol. 9, 1244–1252.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Yu, C. F., Peng, W. M., Oldenburg, J., Hoch, J., Bieber, T., Limmer, A., et al. (2010). Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation. J. Immunol. 184, 1159–1167.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zasloff, M. (2002). Antimicrobial peptides of multicellular organisms. Nature 415, 389–395.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zhang, J., Raper, A., Sugita, N., Hingorani, R., Salio, M., Palmowski, M. J., et al. (2006). Characterization of Siglec-H as a novel endocytic receptor expressed on murine plasmacytoid dendritic cell precursors. Blood 107, 3600–3608.

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: plasmacytoid dendritic cells, type-I IFNs, antigen-presentation, tolerance, autoimmunity

Citation: Guéry L and Hugues S (2013) Tolerogenic and activatory plasmacytoid dendritic cells in autoimmunity. Front. Immunol. 4:59. doi: 10.3389/fimmu.2013.00059

Received: 03 January 2013; Paper pending published: 08 February 2013;
Accepted: 19 February 2013; Published online: 06 March 2013.

Edited by:

Francesca Granucci, University of Milano-Bicocca, Italy

Reviewed by:

Natalio Garbi, University of Bonn, Germany
Jean M. Davoust, Institut National de la Santé et la Recherche Médicale, France

Copyright: © 2013 Guéry and Hugues. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in other forums, provided the original authors and source are credited and subject to any copyright notices concerning any third-party graphics etc.

*Correspondence: Stéphanie Hugues, University of Geneva Medical School, Centre Medical Universitaire, 1 rue Michel Servet, CH-1211 Geneva, Switzerland. e-mail: stephanie.hugues@unige.ch

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.