Seminars in Immunology - BioMedSearch

2 downloads 0 Views 2MB Size Report
progressive multifocal leukoencephalopathy; TCI, transcutaneous immunization. Keywords. Skin DCs; Homeostasis; Regulatory DCs; Chronic inflammation; ...
Sponsored document from

Seminars in Immunology Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Sponsored Document

Harnessing dendritic cells in inflammatory skin diseases Chung-Ching Chu, Paola Di Meglio, and Frank O. Nestle⁎ St. John's Institute of Dermatology, King's College London and NIHR Biomedical Research Centre, Guy's and St. Thomas’ Hospitals, 9th floor Tower Wing, Guy's Hospital, London SE1 9RT, United Kingdom

Abstract

Sponsored Document

The skin immune system harbors a complex network of dendritic cells (DCs). Recent studies highlight a diverse functional specialization of skin DC subsets. In addition to generating cellular and humoral immunity against pathogens, skin DCs are involved in tolerogenic mechanisms to ensure the maintenance of immune homeostasis, as well as in pathogenesis of chronic inflammation in the skin when excessive immune responses are initiated and unrestrained. Harnessing DCs by directly targeting DC-derived molecules or selectively modulate DC subsets is a convincing strategy to tackle inflammatory skin diseases. In this review we discuss recent advances underlining the functional specialization of skin DCs and discuss the potential implication for future DC-based therapeutic strategies.

Abbreviations SALT, skin-associated lymphoid tissue; DCs, dendritic cells; AD, atopic dermatitis; CHS, contact hypersensitivity; DDCs, dermal DCs; MHC, major histocompatibility complex; pDCs, plasmacytoid DCs; TLR, toll-like receptor; SLE, systemic lupus erythematousus; HEL, hen egg lysozyme; iNOS, inducible nitric oxide synthase; TIP-DCs, TNF- and iNOS-producing DCs; NO, Nitric oxide; IDECs, inflammatory dendritic epidermal cells; NR-UVB, narrow-band UVB; PML, progressive multifocal leukoencephalopathy; TCI, transcutaneous immunization

Keywords

Sponsored Document

Skin DCs; Homeostasis; Regulatory DCs; Chronic inflammation; Psoriasis

1

Introduction The skin is an organ rich in easily assessable immune cells that are ready to provide the first line immune defense mechanisms against a multitude of internal and external pathogenderived and environmental challenges. As the primary immunological barrier to the external environment, the skin is equipped with a complex network of immune cells, originally described as “skin-associated lymphoid tissue (SALT)” [1] and later as “skin immune system” [2]. Skin immune sentinels consist of both innate and adaptive immune cells [3]. Dendritic cells (DCs), as professional antigen presenting cells, are the main gate-keepers of

© 2011 Elsevier Ltd. ⁎

Corresponding author. Tel.: +44 20 7188 8086; fax: +44 20 7188 8050. [email protected]. This document was posted here by permission of the publisher. At the time of deposit, it included all changes made during peer review, copyediting, and publishing. The U.S. National Library of Medicine is responsible for all links within the document and for incorporating any publisher-supplied amendments or retractions issued subsequently. The published journal article, guaranteed to be such by Elsevier, is available for free, on ScienceDirect.

Chu et al.

Page 2

Sponsored Document

the immune system, bridging innate and adaptive immunity. It is now becoming clear that DCs with their inherent plasticity can initiate a diverse range of immune responses in addition to conventional functions in the generation of immunity against pathogens [4,5]. In the steady-state, DCs have an essential role in maintaining tissue homeostasis. In pathology, DCs are able to mature into inflammatory DCs at sites of inflammation in both autoimmune and allergic disease, thereby sustaining a continuous activation of the adaptive immune system at sites of inflammation [6]. DCs have a central role in many aspects of skin immunity. Understanding the skin DC network is critically important to tackle skin immune pathology. Over the years much effort has been devoted to characterize skin DC subsets and their functions. Many studies have taken advantage of genetically engineered animal models that allow researchers to selectively examine or target DC subsets under defined conditions. Although breakthrough findings in the murine system have brought valuable insights into our understanding of the skin DC network, the discrepancy between human and murine skin immune systems, in particular the heterogeneity of DCs, donor variability, and diverse markers for DC subsets identification, poses a challenge for translational findings from the murine to the human system (Fig. 1). Different experimental approaches (i.e. in situ or ex vivo examination; enzymatic digestion or naturally crawl-out methods to isolate skin DCs) applied in various studies have increased the complexity of data analysis. Thus, the functional characteristics of human skin DC subsets are only partially understood.

Sponsored Document

In this review we discuss the current understanding of the skin DC network, emphasizing the functional specialization of skin DC subsets in the steady-state conditions (Fig. 2) and pathology, using the common inflammatory skin diseases psoriasis and atopic dermatitis (AD) as examples. This review also highlights current therapeutic approaches as well as new concepts related to harnessing DCs as a convincing immunological intervention for the treatment of chronic inflammatory immune-mediated skin diseases.

2 2.1

Skin DC subsets and functions Epidermis (Langerhans cells)

Sponsored Document

Langerhans cells (LCs) are the primary DC subset in the epidermis of healthy skin. They are radio-resistant and are phenotypically characterized by the expression of langerin (CD207), CD1a, E-cadherin and epithelial-cell adhesion molecule (EpCAM) [7]. Morphologically, they are typified by the presence of Birbeck granules, a tennis racket-shaped cytoplasmic organelle mainly composed by langerin [8]. In the steady state, LCs are situated in the basal and suprabasal layers of epidermis, where they interact with keratinocytes through Ecadherin [9]. E-cadherin ligation may act to maintain LCs in an immature state [10]. Keratinocytes are believed to be an important source of mediators that help to support the development of LCs [11]. In adult quiescent skin, LCs are maintained at a stable density in the epidermis potentially through self-renewal or by skin-resident radio-resistant LC precursor cells [12]. During injury or inflammation when the skin is depleted of resident LCs as in the case of UV-B irradiation, circulating progenitors such as monocytes may enter the inflamed skin and replenish LCs in the epidermis [12–14]. In humans, CD34+ hematopoietic progenitor cells [15,16], monocytes [17], as well as dermal CD14+ DCs [18] were shown to give rise to LC-like cells in vitro. TGF-β is a critical factor for LC development both in vitro and in vivo, as LCs are absent in the epidermis of TGF-β deficient mice [19]. Despite the abundance of studies in the past decades aiming to uncover the functional role of LCs, the in vivo function of LCs is still not fully understood. As one of the first DCs coming into contact with invading pathogens, LCs were believed to have the capacity to sense

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 3

Sponsored Document

infection, capture antigens and acquire a strong immunogenic function [20,21]. The classical LC paradigm states that LCs are maintained in an immature state in quiescent skin. Upon encountering pathogens, LCs can capture antigens and undergo a maturation process which involves up-regulation of major histocompatibility complex (MHC) class I and class II molecules, costimulatory molecules including CD40, CD80 and CD86, and chemokine receptors such as CCR7, as well as down-regulation of E-cadherin that allows them to migrate out of the skin to draining lymph nodes, where they present antigens to T cells [22]. Both ex vivo isolated LCs or in vitro generated LC-like DCs display a strong T-cell stimulatory capacity [23]. In addition, LCs present exogenous antigens to CD8+ T cells via the MHC class I pathway, a process referred to as “cross-presentation” and promote a strong cytotoxic T cells responses [23,24].

Sponsored Document

Early work addressing in vivo function of LCs has focused on a mouse model of contact hypersensitivity (CHS) and demonstrated the potential role of LCs in CHS reactions [25]. This however has been challenged as removal of LCs by topical application of steroids [26] or using an inducible or constitutive LC ablation model (LC-deficient mice) showed no difference [27] or even an enhancement in the magnitude of CHS responses [28]. These reports suggested that LCs are dispensable in CHS reactions and raises the possibility that LCs may have an immune inhibitory role. The redundant role of LCs in CHS reaction is proposed to be compensated by a recently characterized subset of dermal resident CD207+ DCs. Notably, the concentration of sensitising antigens may be essential for dermal CD207+ DC-mediated CHS responses. When only a low concentration of antigens was applied to LC-deficient mice, a diminished CHS reaction was observed potentially due to limited access of antigens by dermal DCs (DDCs) [29,30]. The role of LCs in antimicrobial immunity has also been questioned by the finding that LCs were unable to generate CD8+ T cell immunity when the skin was infected with herpes simplex virus [31]. One explanation was that HSV can induce LC apoptosis and therefore diminish their function. It was proposed that apoptotic DCs can be taken up by splenic CD8α+ DCs that can efficiently cross-present antigens and initiate anti-viral immunity (Fig. 2). Taken together, the role of LCs in promoting or inhibiting immune responses remains to be fully clarified. It is likely that the nature of antigen as well as the cross-talk involving accessory epidermal cells (i.e. keratinocytes), LCs and DDCs will determined the outcome of the immune responses. 2.2

Dermis

Sponsored Document

2.2.1 Myeloid DCs—Interest on DDCs has significantly increased since a number of early studies functionally characterized resident DCs in the dermis of both murine and human skin [32,33]. Thus far several subpopulations of DDCs have been described under both normal and pathologic conditions. In healthy human skin the majority of DDCs are of myeloid origin and express CD1c (also known as BDCA1) [33], a surface marker widely used to identify the main subset of myeloid DCs in peripheral blood [34]. Zaba et al. extended previous findings and suggested that co-expression of CD1c and CD11c is a useful marker in situ to distinguish DDCs from macrophages [35]. These authors further proposed to differentiate between steady-state CD11c+CD1c+ DDCs and “inflammatory DCs” found in inflamed skin [36]. CD1c+ DDCs can be classified into at least three discrete subsets base on their surface expression of CD1a and CD14 [33]. It is now becoming more recognized that distinct subsets of DDCs display differential functional specialization. CD1a+CD14− DDCs can be distinguished from CD1a+ LCs based on their morphology, phenotype, and transcriptomic profiles [37]. Ex vivo isolated CD1a+CD14− DDCs have a mature phenotype and are potent inducers of allogeneic naïve CD4+ and CD8+ T-cell proliferation [23,33,38,39]. In contrast, CD14+ DDCs are less mature then CD1a+ DDCs and display a reduced capacity to prime naïve T-cell proliferation [40]. Interestingly, IL-23 treatment enhanced the T cell stimulatory capacity of CD14+ DDCs. Likewise, IL-23-neutralizing

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 4

Sponsored Document

antibody inhibited T cell proliferation stimulated by CD14− DDCs [41]. Although the molecular consequences of IL-23 action on DDCs remain to be investigated, given that DDCs produce IL-23 in skin pathology and the importance of IL-23 in Th17 cell immunity [42], this result may explain at least in part the clinical benefit seen in psoriatic patients treated with anti-IL-12/23p40 (see detail discussion in Section 4.1). While CD1a+ DDCs have strong T cell stimulatory capacity, other studies suggest that CD14+ DDCs are efficient at antigen up-take, potentially through C-type lectins such as mannose receptor (CD206) and DC-specific ICAM-3-grabbing non-integrin (DC-SIGN/CD209) expressed on the cells surface [18,40]. In addition, Klechevsky et al. demonstrated that CD14+ DDCs are able to polarize naïve CD4+ T cells into follicular helper T cells, which then promote naïve B cells differentiation [23] (Fig. 2). Although the significance of this finding needs to be further evaluated in more physiological experimental settings, these data underline a previously unappreciated function of CD14+ DDCs in the activation of humoral immunity.

Sponsored Document

Another level of functional specialization for DDCs is highlighted by the detailed characterization of dermal-resident CD207+CD103+ DCs in mice. This population of DDCs has previously been overlooked, mainly due to its low frequency in the dermis that has led to the assumption that all CD207+ DCs found in the dermis are LCs en route to draining lymph nodes [43]. A series of animal studies provided clear evidence suggesting that dermal CD207+ DCs are distinct from LCs in both their origin and function [44–46]. Recent studies further revealed a unique functional characteristic of CD207+CD103+ DDCs to crosspresent epidermal-derived viral and self antigens to CD8+ T cells [47,48]. It appears that CD207+CD103+ DDCs can capture keratinocyte-derived antigens and cross-present them in skin draining lymph nodes (Fig. 2). Further studies are required to understand how dermal resident CD207+CD103+ DDCs gain access to epidermal-derived antigens since LCs seem to be dispensable in this process [48].

Sponsored Document

The unique functional characteristics of CD207+CD103+ DDCs make this DC subset an attractive target for therapy. This also raises an important question as to the presence of their human DDC counterparts. Dermal CD207+ DCs represent less than 5% of DDCs in the human skin (unpublished observation). Whether human CD207+ DDCs correspond to the CD207+CD103+ DDCs described in mice remains to be investigated. Human CD141+ (also known as BDCA3) DCs has recently been proposed as putative equivalents of mouse CD8α+ DCs, a primary subset of splenic DCs responsible for cross-presentation [49–51]. Despite CD141+ DCs representing a minor DC population in peripheral blood, they are found in various non-lymphoid tissues including the skin [34,35,52–55]. The function of CD141+ DCs in the human skin is currently unidentified. Future studies are required to address whether skin-resident CD141+ DCs have similar functional specialization to those found in the peripheral blood. 2.2.2 Plasmacytoid DCs—Human plasmacytoid DCs (pDCs) represent a minor population of DCs in peripheral blood and are identified in both primary and secondary lymphoid organs including thymus, bone marrow, spleen, and lymph nodes [56,57]. pDCs play a major role in anti-viral immunity because of their extraordinary capacity to rapidly produce large amounts of type I interferons (IFNs) upon viral infection [58,59]. The unique characteristic of pDCs in sensing viral infection is linked to their selective endosomal expression of toll-like receptor (TLR) 7 and TLR9 that, respectively, recognizes singlestranded RNA and DNA derived from pathogens invading cells via the endocytotic route [60]. pDCs are normally absent in the skin and other non-lymphoid tissues in the steady state. However, pDCs can infiltrate the dermis of inflamed skin under several pathogenic conditions, such as psoriasis [61], systemic lupus erythematousus (SLE) [62,63], and certain

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 5

Sponsored Document

skin tumors [64]. Several chemotactic factors may be involved in the recruitment of pDCs into peripheral tissues. Chemokine receptor CXCR3 expressed by pDCs can mediate their migration in response to inflammatory chemokines such as CXCL9, CXCL10 and CXCL11 [64]. More recently, chemerin has been shown as a unique inflammatory chemotactic factor that can directly promote pDC migration through interaction with its cognate receptor ChemR23 [65]. Chemerin expression is strongly associated with pDC infiltration in both SLE and psoriasis and therefore may serve as a crucial factor for early pDC recruitment into inflamed skin [65]. During homeostasis, pDCs can sense pathogen-derived nucleic acids but are tolerant to self DNA or RNA released from cells under going necrosis or apoptosis. Breaking tolerance of pDCs to self DNA/RNA can lead to autoimmunity [60]. This process involves the formation of immune complexes between self DNA with DNA-specific antibodies as in the case of SLE [66], or the generation of self DNA and RNA aggregates with the antimicrobial peptide LL-37 as described in psoriasis [67,68]. The pathogenic roles of pDCs in chronic skin inflammatory disorders are discussed in Section 3.1. 2.3

Immune regulatory DCs

Sponsored Document

Several studies have highlighted the critical roles of DCs in regulatory control of immune responses. In vivo evidence suggests that DCs can induce “deletional tolerance”, essentially through the presentation of soluble and tissue-associated self antigens to self-reactive T cells in the steady-state [69]. Delivery of hen egg lysozyme (HEL) to DCs in the steady state conditions induces CD4+ T cell unresponsiveness to subsequent systemic challenge of HEL. It was proposed that depletion of HEL-specific T cells was a potential tolerance mechanism involved in this model system [70]. Liu et al. further showed that in vivo delivery of dying cells to DCs led to immune tolerance. In this model, dying cells were captured and crosspresented to CD8+ T cells by the CD8α+ DC subset. Following cross-presentation, antigenreactive CD8+ T cells were initially driven into cell cycle but were then deleted [71]. Later work also suggested that the induction of regulatory T cells may be a potential mechanism involved in CD8α+ DC-mediated tolerance [72]. These experiments supported the concept that DCs can play a major role in peripheral tolerance by continually capturing and presenting cell-associated antigens or environmental proteins under steady state conditions.

Sponsored Document

Skin DCs have been shown capable of mediating T cell tolerance. The notion that LCs have immunoregulatory property came from the observation that LC-deficient mice develop enhanced CHS reaction [28]. When transplanting skin graft of a LC-deficient donor to genetically mismatched recipient, efficient graft rejection was routinely observed suggesting that donor LCs are not necessary for graft rejection. Under certain experimental conditions, the role of LCs in supporting long-term skin engraftment was also described [73]. The mechanism of LC-mediated immune regulation is not fully characterized. In the inflamed skin, LCs may produce IL-10 and promote the expansion of regulatory T cells through interaction with receptor activator of NF-κB ligand (RANKL) expressed by keratinocytes [74,75]. In the steady state, DCs of both dermal and epidermal origin were able to transport keratinocyte-associated antigens to skin-draining lymph nodes as demonstrated in transgenic mice expressing membrane-bound form of OVA under the control of the keratin 5 promoter. When adoptively transferring OVA-specific TCR transgenic CD8+ T cells (OT-I) into these mice, deletional tolerance was observed as OT-I cells were initially proliferating in skindraining lymph nodes and then were found disappeared six weeks after transfer. An impaired response of the transferred OT-I cells to subsequent antigenic challenge was also reported in this model [76]. More recently, CD207+CD103+ DDCs were proposed to be the major skin DC subset involved in antigen transfer and presentation. Future studies are required to further dissect the differential immunoregulatory roles of skin DC subsets in the maintenance of skin homeostasis. Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 6

Sponsored Document

In humans much less is known about regulatory DCs and their in vivo functions. Jonuleit et al. and others demonstrated that allogeneic T cells co-cultured with immature DCs became unresponsive to antigenic re-stimulation. In addition, immature DCs induce IL-10 producing regulatory T cells that suppress the function of other effector T cells in a partially IL-10 dependent manner [77,78]. Other work indicated that pro-inflammatory cytokine-matured DCs can efficiently induce regulatory T cells with immune suppressive function [79,80]. The physiological significance of these findings remains to be explored. Thus far, it is not known whether certain subsets of human DCs may have specialized function in immune regulation under tissue homeostasis as described in the murine system.

3

Pathogenic role of DCs in inflammatory skin disease DCs have been recognized as key players in several inflammatory skin diseases. Given their capacity to initiate a cascade of immune responses, DCs of both plasmacytoid and myeloid lineage are thought to display a pathogenic role. Here we focus our discussion on psoriasis and AD, two common types of inflammatory skin pathologies, highlighting the essential role of several DCs subtypes in their respective immunopathogenesis.

3.1

Psoriasis

Sponsored Document

Psoriasis is a common, chronic inflammatory skin disease which affects 2–3% of the Caucasian population and is characterized by sharply demarcated, scaly, red plaques [81]. Psoriasis is believed to result from the combination of environmental triggers and genetic factors, conferring disease susceptibility. Environmental factors include β-haemolytic streptococcal pharyngitis, stress, skin trauma and various drugs (such as β blockers and lithium), while at least nine chromosomal loci with statistically significant linkage to psoriasis have been identified. The major genetic determinant of psoriasis is PSORS1 located within the major histocompatibility complex (MHC) on chromosome 6p [82], which accounts for up to 50% of the heritability of the disease. Within PSORS1 is the human leukocyte antigen-C (HLA-C) gene which is the strongest candidate gene for psoriasis identified to date, with its allele HLA-Cw*0602 shown to be the predominant risk allele [83,84]. In addition, genome-wide association studies have implicated several other psoriasis associated genes, involved in various biological processes, including IL-23/IL-17 pathway [83,85–89], MHC class I processing [89,90], epidermal cell differentiation [90,91], NF-κB signalling [83,89,92], ubiquitin pathway [90,93], Th2-type response [83] as well as genes of yet unknown function [94].

Sponsored Document

Effector cells of both the innate and the adaptive immune system, including keratinocytes, DCs and T cells have all been shown to take part in a dysregulated immune response responsible for the typical histological features of psoriasis (Fig. 3). Excessive keratinocyte proliferation and reduced differentiation lead to thickening of the skin (acanthosis), elongation of the epidermis into the papillary dermis (papillomatosis) and retention of the nucleus in the stratum corneum (parakeratosis). Moreover, dermal blood vessels become dilated and increase in number, giving raise to the red colour of psoriasis lesions and are filled with leukocytes that accumulate in both dermis and epidermis. In particular, neutrophils and CD8+ T cells are found in the epidermis, while pDCs, myeloid DDCs, and CD4+ T cells, mainly Th1 and Th17, are in the dermis. The initial clinical observation of a case of psoriasis exacerbated by topical treatment with the TLR7 agonist imiquimod [95], the occurrence of an evident IFN-α signature in psoriasis [96] and the presence in elevate number of pDCs in psoriatic skin [95,97] lead to further investigations about the role of pDCs in psoriasis.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 7

Sponsored Document Sponsored Document

pDCs have been identified not only in lesional psoriatic skin but also in uninvolved, healthylooking psoriatic skin and shown to be the principal IFN-α-producing cells in early and developing psoriasis [61]. This early recruitment of pDCs to the psoriatic skin could be mediated by chemerin abundantly expressed in pre-psoriatic skin adjacent to fully established lesions [65]. Blocking of type I IFN signalling or inhibiting of IFN-α release by pDCs prevented the activation and expansion of pathogenic T cells and the development of psoriasis in a xenograft mouse model of human psoriasis. Moreover, temporal analysis of psoriasis development in this model showed that an IFN-α signalling signature is indeed present in developing psoriasis and precedes the characteristic psoriatic phenotype, including the epidermal changes [61]. Finally, the molecular mechanisms of pDCs activation have been also elucidated. Proteomic analysis of psoriatic skin revealed the pDC activating substance in psoriatic skin. The cathelicidin LL-37, an endogenous antimicrobial peptide produced by keratinocytes and neutrophils and overexpressed in psoriatic skin, has been shown to bind to self-DNA/RNA fragments released by stressed or dying skin cells and to trigger TLR9/TLR7-mediated pDC activation and type I IFN production [67,68]. Additionally, self-RNA-LL-37 complexes signalling through TLR8 have been shown to promote myeloid DC maturation [68]. Therefore, LL-37-mediated breakdown of innate tolerance to self DNA/RNA and a pathogenic cross talk involving stressed epidermal cells and recruited pDC represent one of the first events in psoriasis development, followed by IFN-α-dependent maturation and activation of bystander DDCs and subsequent adaptive immune responses (Fig. 3). Association between self-RNA-LL-37 complexes and mature myeloid DCs observed in psoriatic lesions further suggests a potential pathogenic role of LL-37 in the maintenance phase of psoriasis [68]. While pDCs are believed to be early key players in the upstream events leading to skin lesion formation, myeloid DDCs are thought to be instrumental in sustaining and amplifying the T-cells mediated inflammatory reaction underlying the epidermal hyperplasia. In the nineties, pioneering early work led to the isolation of DDCs from psoriatic lesions and suggested for the first time that these previously overlooked cells might exert a pivotal role in disease pathogenesis [98]. CD1c and HLA-DR expressing DDCs were found in large number in psoriatic dermis, positioned immediately beneath the hyperplastic epidermis, and surrounded by T cells. DDCs derived from psoriatic lesions were found to be much more effective stimulators of spontaneous autologous T cell proliferation compared with either psoriatic blood-derived or normal healthy skin-derived DC. Moreover, cytokine profiles studies showed that DDCs obtained from psoriatic lesions mediated a T cell response with high levels of IL-2 and IFN-γ, thus contributing to the definition of psoriasis as Th1-type disease [98–100].

Sponsored Document

Further studies have expanded these findings and DCs have emerged as key cellular players in psoriasis. A 30-fold increase in CD11c+ DCs has been shown in the dermis of psoriatic skin lesions compared to uninvolved psoriatic or normal skin [101]. Detailed in situ phenotypic analysis of DCs present in psoriatic skin suggested that psoriatic dermis harbors two distinct populations of DCs [36]: a classical CD11c+CD1c+ population resides normally in the quiescent skin as well as a population of “inflammatory DCs” identified by their CD11c+CD1c− phenotype [102]. The CD11c+CD1c− inflammatory DCs are relatively immature DCs, with little dendritic cell-lysosomal-membrane-associated protein (DC-LAMP) and DEC-205/CD205 coexpression and decreased HLA-DR expression while showing some expression of CD14+, DC-SIGN, and CD163 [102]. Inflammatory DCs are also quite heterogeneous in their function, as they possibly include TNF- and inducible nitric oxide synthase (iNOS)producing DCs (named TIP-DCs), IL-20-producing DCs and IL-23-producing DCs.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 8

Sponsored Document

TIP-DCs were first described in the spleen during a murine model of Listeria monocytogenes infection [103] and have also been found in murine Escherichia coli [104], Brucella melitensis [105] and influenza infection [106] where they are important for pathogens clearance. In humans, iNOS and TNF positive CD11+ DCs are abundant in psoriatic skin lesions and are reduced following successful psoriasis treatment [107]. Both TNF and iNOS are well established pro-inflammatory mediators in psoriasis. TNF induces expression of intracellular adhesion molecule-1 (ICAM-1), neutrophils-chemoattractant IL-8 as well as pro-inflammatory cytokines, IL-6 and IL-1 in keratinocytes. Nitric oxide (NO) generated from iNOS in inflamed tissues induces vasodilation and inflammation, thus possibly accounting for the vasodilation of dermal blood vessels in psoriatic skin. Other proinflammatory mediator produced by “inflammatory DCs” included IL-20 [108], which enhances keratinocytes activation and proliferation, and, very importantly, IL-23 [101,109].

Sponsored Document

In just ten years from its discovery [110] IL-23 has quickly emerged as a key proinflammatory molecule in psoriasis. IL-23 is highly expressed in psoriatic skin lesions and is down regulated following successful conventional and biologics systemic therapies [111,112]. We and others have shown by genome-wide association studies that several genes of the IL-23 pathway, including IL23R, IL12B and IL23A are associated with psoriasis [83,85,86]. Intradermal injection of IL-23 in mice leads to erythema, induration and prominent dermal papillary blood vessels with histopathological features resembling psoriasis [113]. We have recently shown that selectively targeting IL-23 prevents psoriasis development in a clinically relevant psoriasis mouse model [114]. Moreover, IL-23 plays a role in terminal differentiation and peripheral pathogenicity of effector Th17 cells that infiltrate psoriatic lesion [115] where high level of pro-inflammatory Th17 cytokines, including IL-17A, have been detected [116]. DDCs are the main producers of IL-23 in psoriasis [101,109,117,118]. The crucial link between DDCs and the IL-23/Th17 axis has been further strengthen by ex vivo studies showing that DDCs obtained from psoriatic lesions, but not those obtained from normal skin, activates T cells to produce both IL-17 and IFN-γ [102]. Finally, DDCs are also a potential source of the Th1 polarizing cytokine IL-12, although there are conflicting reports in the literature about weather or not there are higher level of IL-12 mRNA and protein in psoriatic vs. normal skin [109,119,120].

Sponsored Document

Taken together, IL-23 and IL-12 secreted by DDCs induce activation of skin resident T cells with release of pro-inflammatory Th1 and Th17 cytokines, that act on keratinocytes, which in turn sustain and amplify the chronic inflammatory disease process by producing proinflammatory cytokines, chemokines, members of the S100 family and antimicrobial peptides (e.g. TNF, IL-8, CCL-20, S100 molecules, defensins, cathelicidins) (Fig. 3). Therefore, a critical role for skin DCs as both direct and indirect sources of proinflammatory mediators, complementing their classic role as antigen-presenting cells, is being recognized in psoriasis. However it remains to be established weather CD11c+CD1c+ “resident DCs” and CD11c+CD1c− “inflammatory DCs” are effectively two distinct populations of different origin and function or “inflammatory DCs” are rather “resident DCs” in an activated status. A very recent study has started to shed some light on this topic, showing that the transcriptome of psoriatic CD11c+CD1c− inflammatory DCs is closely related to that of CD11c+CD1c+ resident DCs [121]. Nevertheless, psoriatic CD11c+CD1c− inflammatory DCs expressed a wide range of inflammatory molecules, including TNFrelated apoptosis-inducing ligand (TRAIL) and several other inflammatory mediators, compared to skin-resident CD11c+CD1c+ DCs. It has been suggested that CD11c+ myeloid DDCs are capable of remarkable plasticity and may differentiate into inflammatory DCs,

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 9

which encompass both TIP-DC in psoriasis and IDECs present in AD (see below), depending on environmental triggers [122,123]. Further studies, perhaps involving humanized mouse models of psoriasis, will be needed to clarify the exact nature of inflammatory DCs in skin inflammation.

Sponsored Document

3.2

Atopic dermatitis

Sponsored Document

Atopic dermatitis (AD), also named atopic eczema, is a chronic relapsing, inflammatory skin disease often associated with other systemic atopic disorders such as asthma, food allergy, and allergic rhinitis. Acute AD is characterized by eczematous patches and plaques, epidermal edema (spongiosis) and prominent cellular infiltrate in the dermis. In subacute and chronic phases plaques are lichenified, the epidermis is thickened and its upper layer becomes hypertrophied [124]. Recent insights into the phatophysiology of AD point to an important role for genetically determined structural abnormalities in the epidermis combined with immune dysregulation. Genetic variants of filaggrin, which contribute to the keratin cytoskeleton, have been identified in AD [125,126]. Such epidermal-barrier dysfunction allows the penetration in the skin of allergens such as pollens, house-dust-mite products, microbes and food allergens that trigger an overwhelming inflammatory response mediated by cells of both innate and adaptive immunity. Although AD has been considered a paradigmatic Th2-mediated disease, sequential biopsies in AD patients after exposure to aeroallergens have demonstrated a biphasic immunologic response characterized by a switch toward a Th1 phenotype in later and more chronic phases of the disease [127] (Fig. 4). Two different populations of skin myeloid DCs seem to play a pivotal role in these two phases of AD: LCs and inflammatory dendritic epidermal cells (IDECs) [128].

Sponsored Document

LCs in AD lesions have been shown to display a high-density of IgE high-affinity receptor (FcɛRI) [129]. This allows them to bind to, internalize and present allergens specific for the IgE molecules, ultimately inducing a typical Th2 type response, characterized by increased IgE production and induction of Th2 cells producing IL-4, IL-10, and IL-13 [130] either in the local skin-draining lymph nodes or possibly in situ in the skin. Moreover, in vitro studies suggest that on ligation of FcɛRI by IgE, LCs produce pro-inflammatory cytokines IL-8, MCP-1/CCL-2 and IL-16 [130] thus favoring the recruitment to the skin of eosinophils, monocytes and T cells, respectively. Additionally, LCs express high levels of the receptor for thymic stromal lymphopoietin (TSLP), an IL-7-like cytokine, which is abundantly produced by AD keratinocytes [122,131]. Interestingly, high levels of TSLP in AD lesions correlates with increased CD207+ DCs redistribution from epidermis to the dermis where they display a mature phenotype (DC-LAMP+), thus suggesting that TSLP receptor+ DCs in the dermis may be mature LCs [131]. TSLP is known to induce Ag-independent maturation of DC, to maintain their survival and to induce production of CCL-24/Eotaxin2, CCL-17/ TARC and CCL-22/MDC as well as IL-8 and IL-15, which favor recruitment of Th2 cells toward inflammatory sites [132]. Finally, TSLP conditions DCs to promote Th2 cells, which produce typical Th2 cytokines, IL-4, IL-5, IL-13, as well as TNF [133]. IDECs were first described using flow cytometric analysis of epidermal single-cell suspensions of AD lesions [128]. Following their discovery IDECs were defined by the surface phenotype of CD11c+, HLA-DR+, Lin−, CD1a+, CD206+, CD36+, FcɛRIhigh, IgE+, CD1b/c+, CD11b+, and DC-SIGN+ [128,134]. In contrast to what is described for LCs, in vitro FcɛRI ligation of IDECs results in production of IL-1, MIP-1α, IL-16, and Th1-polarizing cytokines IL-12p70 and IL-18 leading to IFN-γ production [130]. Therefore, stimulation of these DCs by allergens via FcɛRI may be responsible for the chronic maintenance phase of AD sustained by a pronounced Th1 cytokine profile. Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 10

Sponsored Document

Recently, a discrete population bearing IDEC markers has been found in the dermis of AD lesions [122] and it has been suggested to re-define IDECs as part of the myeloid inflammatory DC populations spanning both dermal and epidermal compartments of inflamed tissues [122,123]. In contrast to inflammatory DCs present in psoriasis, those found in AD do not express iNOS but are possibly responsible for the high level of CCL17 and CCL18 found in AD [122].

4 Clinical significance of DCs as therapeutic target during skin inflammation Harnessing DCs is a sensible therapeutic intervention due to their diverse pathogenic roles in inflammatory skin diseases. Clinical applications are being developed to directly target DCderived molecules or manipulate DC functions. Current approaches and new concepts related to targeting DCs in skin pathology is summarized in Fig. 5. 4.1

Anti-cytokine therapy

Sponsored Document Sponsored Document

Given the fundamental role of DCs in shaping immune inflammatory responses in skin it is not surprising that the clinical benefit of several conventional anti-psoriasis therapies, including psoralen and ultraviolet A (PUVA) [135], narrow-band ultraviolet B (NR-UVB) [111,136,137] as well as cyclosporine therapy [22] correlates with a significant decrease in DDC numbers and/or the expression of DC-derived proinflammatory mediators iNOS, TNF, IL12/23p40 and IL-23p19 in psoriatic skin. Similarly, the effective targeting of the psoriatic cytokine network [138] with either neutralizing antibodies or soluble receptors has also been shown to impact on DC biology. Currently regarded as the gold standard treatment for patients with moderate-to-severe psoriasis, anti-TNF strategy is achieved by using either a human p75 TNF-receptor Fc fusion protein (etanercept), a humanized chimeric anti-TNF monoclonal antibody (infliximab), or a fully human monoclonal anti-TNF antibody (adalimumab). Etanercept treatment has been shown to reduce DDC numbers and downstream effector molecules such as iNOS, IL-20, IL-12/23p40 and IL-23p19 [112,139]. Moreover, DDCs rapidly down-regulated co-stimulatory and maturation markers CD83, DC-LAMP, CD86, and HLA-DR after 2 weeks of etanercept treatment [139]. Consistently, etanercept blocked in vitro–derived DC maturation, IL-23 production and immunostimulatory capacity, and shifted differentiation toward a macrophage-like phenotype [139]. Recently, analysis of genomic profiles of etanercept-treated patients showed that myeloid cells-expressed genes were the most rapidly down-modulated by etanercept [140], suggesting that TNF inhibition directly modulates myeloid-lineage gene products, which in turn affect T cells activation. Interestingly expression of CD1c and CD207 was on the contrary unregulated by etanercept treatment, suggesting that either resident CD1c+ DDCs or LCs do not play an active role in TNF-driven inflammation or they have a rather beneficial role [140]. Although much less investigated, similar effects in terms of DDC numbers and downregulated maturation markers have been reported for adalimimab [141], while infliximab has been shown to affect the in vitro differentiation of monocytederived DCs and their antigen-presenting capacity [142]. Collectively these data suggest that the clinical benefit of TNF neutralization in psoriasis might derive from the attenuation of inflammatory DC activity in the lesions. This is not surprising given the dual role of TNF as critical inducer of DC maturation and activation as well as proinflammatory mediator for T cells activation. Another therapeutic strategy that interferes with DC activity involves targeting IL-23 [42]. Two anti IL-12/IL-23p40 monoclonal antibodies (mAbs) have been developed, namely ustekinumab (formerly CNTO-1275) and briakinumab (formerly ABT-874). Both ustekinumab and briakinumab are human IgG1 mAbs that bind to the p40 subunit of human

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 11

Sponsored Document

IL-12 and IL-23, and prevent its interaction with IL-12Rb1. While briakinumab is currently in phase III clinical trials [143], ustekinumab has been recently approved for moderate-tosevere psoriasis on the basis of its efficacy in 70–80% of cases [144,145]. Designed in the late 1990s with the aim to block IL-12 (IL-23 was still unknown at that time), ustekinumab targets the common IL-12/23p40 subunit, thus blocking both IL-12 and IL-23 signalling. Early study evaluating the effect of ustekinumab at cellular and molecular level showed a decrease in lesional CD11c+ DCs at 2 wk, although this decrease was not statistically significant [146]. Of importance, mRNA levels of IL-12/IL-23p40, IL-23p19 and TNF, were all significantly decreased in the responder populations [146], suggesting that neutralization of IL-12/IL-23 reduces the production of DC-derived pro-inflammatory cytokines. Although preliminary, these results open the possibility that the beneficial effect of targeting IL-12/ IL-23 might derive from both an impaired Th17 response but also from the interruption of an autocrine feedback of IL-12/IL-23 on DCs. Further studies, aiming to dissect the molecular and cellular events following anti-IL-12/ IL-23 mAbs administration to large cohort of patients are needed to fully understand the mechanisms of action of this promising new therapeutic agent. Although referred as anti-T cells therapy, two other biologics drugs for the treatment of moderate-to-severe psoriasis interfere with the interactions between antigen-presenting cells and T-cells at the level of the immunological synapse and are therefore briefly mentioned here.

Sponsored Document

Alefacept, a CD2-binding leukocyte-function associated antigen (LFA)-3 Ig fusion protein has been proposed to clear psoriasis lesions through the depletion of CD2-expressing cells [147], which includes T cells but also DCs, although to low level. Interestingly, DC-derived pro-inflammatory molecules iNOS and IL-23 are also reduced suggesting that although T cells are the primary target, alefacept also impairs DC activation [147]. Efalizumab, a human mAb directed against LFA-1 which blocks the interaction between LFA-1 on T cells and ICAM-1 on DCs, has been also shown to be effective in clearing psoriatic lesions as well as in reducing CD11c+ DC numbers [107]. However, efalizumab was recently withdrawn from the Europe and US market after 5 years of use because of 3 cases of progressive multifocal leukoencephalopathy (PML), a serious life-threatening infection [148].

Sponsored Document

Taken together data coming from successful psoriasis treatment not only provides further support for the key role of DDCs in the pathogenesis of psoriasis and AD but also suggests that DC can be a useful therapeutic target for the treatment of other immune-mediated dermatological conditions. 4.2

Generation of immune regulatory DCs It is now becoming clear that differentially modulated DCs can become tolerogenic and acquire an immunoregulatory capacity. Several studies reported that anti-inflammatory or immunosuppressive biological and pharmacological agents are able to induce regulatory DCs [149]. Notable examples are gucocorticoids, mycophenolate mofetail, rapamycin (sirolimus), and acetylsalicylic acid (Aspirin) [150]. DCs interacting with regulatory T cells may also acquire immune regulatory properties [151]. Common features of DCs that are affected by immunosuppressive agents include decreased antigen uptake and processing [152], reduced maturation phenotype and function [153], enhanced IL-10 production [154,155], and enhanced DC apoptosis [154,156,157]. In addition, several studies have shown that DCs manipulated to acquire regulatory properties are able to promote the expansion of regulatory T cells [153,158]. Upregulated expression of inhibitory receptors immunoglobulin-like transcript (ILT) 3 and ILT4 is observed in almost all selectively Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 12

Sponsored Document

conditioned DCs [159] and is proposed to be involved in the function of regulatory DCs [151,160]. Signalling through ILT3 inhibits NF-κB activation and the expression of costimulatory molecules in antigen presenting cells, which then lead to reduced T-cell priming capacity [151]. When acting on T cells, ILT3 is able to promote CD4+ T cell anergy and prevent the generation of cytotoxic activity of CD8+ T cells [160]. Expression of ILT3 and ILT4 may serve as markers to identify regulatory DCs, although their expression has been shown dispensable for their regulatory T cell-inducing capacity [161].

Sponsored Document

The use of immunomodulatory agents has been reported in the treatment of chronic inflammatory skin diseases. Among several agents investigated, the active form of vitamin D3 (1α,25-dihydroxy D3) and its analogues, alone or in combination with corticosteroids, have been shown to be useful drugs for the treatment of mild to moderate form of psoriasis due to both their effectiveness and high safety profiles [162,163]. Traditionally, the beneficial effects of vitamin D3 and its analogues were believed to be linked to their capacity to inhibit differentiation and induce apoptosis on keratinocytes. Later studies revealing the profound regulatory effects of vitamin D3 and its analogues on the phenotype and function of DCs have brought new insights into potential mechanisms of vitamin D3 treatment in psoriasis. Several studies demonstrated that vitamin D3-treated DCs were resistant to maturation and produced diminished amounts of IL-12 and IL-23 as well as reduced T cell-stimulatory capacity [164]. In the epidermis, vitamin D3 was also reported to impair LC distribution and antigen-presentation property [165,166]. Vitamin D3-induced RANKL expression on keratinocytes may promote IL-10 production by LCs and the induction of regulatory T cells [74]. Psoriasis lesions treated with calcipotriol, a vitamin D analogue, showed up-regulated ILT3 expression on DCs [161]. Overall, generation of regulatory DCs could be a useful approach for DC-mediated inflammatory skin diseases. 4.3

Antigen delivery to skin DCs

Sponsored Document

The skin has long been appreciated as a potential site for antigen delivery since it is rich in easily assessable DCs. Topical application of both peptides and full proteins following barrier disruption (e.g. tape stripping) and with the aid of appropriate adjuvants (e.g. CpG or cholera toxin), an approach known as transcutaneous immunization (TCI), has been shown able to promote systemic cellular and humoral immune responses [167]. Both LCs and DDCs are believed to be involved although the relative contribution of each skin DC subset is not yet fully understood. A more specific approach to selectively deliver antigens to DCs has been proposed using antigens conjugated with antibodies against endocytic receptors expressed by DCs [168]. Pioneer studies using OVA antigen conjugated with antibody against DEC-205, an endocytic receptor abundantly expressed by lymphoid DCs, has demonstrated an efficient antigen uptake by DCs comparing to the un-conjugated OVA protein [169]. Co-administration of DEC-205-conjugated antigens with potent DC maturation stimuli has led to a strong CD4+ and CD8+ T cells immunity [170]. These findings have recently been extended to target different DC subsets via a variety of DC receptors with the aim to generate potent anti-tumor immunity [168]. Notably, when antigen delivery to DCs was conducted in the steady state (without DC maturation), antigen-specific tolerance instead of immunity was induced potentially through the depletion of effector T cells [70,169] as well as the induction of regulatory T cells [72,171]. This approach have also been shown to prevent the onset and progression of type I diabetics in mouse models [172,173], as well as to induce long-term allogeneic skin graft survival in a transplantation model [174], where immune tolerance induction is required. Although antigen delivery to DCs has great therapeutic potential, several questions remain to be addressed before applying this to the treatment of inflammatory skin diseases. For example, in the context of chronic skin inflammation, inadvertent DC maturation during antigen targeting could be avoided by co-administration of immune regulatory agents such Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 13

Sponsored Document

as vitamin D3 and steroids. Indeed, UV-B irradiation or vitamin D3 analogue treatment prior to TCI has been shown to induce regulatory T cells and immune tolerance even in the presence of strong DC maturation stimuli [175,176]. Exposure to vitamin D3 during tolerance induction may also “imprint” a skin homing phenotype on regulatory T cells [177] and thus help to retain their immune suppressive function in the skin. Another obstacle is the lack of defined autoantigens in many inflammatory skin diseases including psoriasis. The discovery of putative autoantigens would dramatically enhance the specificity of current available therapies. Finally, it is essential to further improve our knowledge of skin DCs subsets and their functional characteristics, in particular translating the current understanding from the murine system to the humans. This will provide the foundation for future therapeutic studies to selectively target a certain subset of skin DCs aiming to generate a specific type of immune response.

5

Conclusions

Sponsored Document

DCs with their inherent plasticity play crucial roles in initiating and modulating immune responses. Recent advance in the knowledge of skin DCs network in health and pathology has opened the avenues for the development of therapies that harness skin DCs with specialized properties to control immunity. Thus far, several therapeutic interventions targeting skin DCs have been proven beneficial to psoriatic patients. Future studies are required to further understand the contribution of skin DC subsets in immunity and tolerance, as well as the cross-talk among the complex network of DCs subsets, skinresident innate and adaptive immune sentinels, and accessory epidermal and dermal components during homeostasis and pathology. This will provide significant insights in future translational research studies and ultimately lead to the development of novel therapies.

References

Sponsored Document

1. Streilein J.W. Skin-associated lymphoid tissues (SALT): origins and functions. J Invest Dermatol. 1983; 80(Suppl):12s–16s. [PubMed: 6602189] 2. Bos J.D. Kapsenberg M.L. The skin immune-system – its cellular-constituents and their interactions. Immunol Today. 1986; 7:235–240. 3. Nestle F.O. Di Meglio P. Qin J.Z. Nickoloff B.J. Skin immune sentinels in health and disease. Nat Rev Immunol. 2009; 9:679–691. [PubMed: 19763149] 4. Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006; 6:476–483. [PubMed: 16691244] 5. Steinman R.M. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991; 9:271–296. [PubMed: 1910679] 6. Coquerelle C. Moser M. DC subsets in positive and negative regulation of immunity. Immunol Rev. 2010; 234:317–334. [PubMed: 20193028] 7. Merad M. Ginhoux F. Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol. 2008; 8:935–947. [PubMed: 19029989] 8. Valladeau J. Ravel O. Dezutter-Dambuyant C. Moore K. Kleijmeer M. Liu Y. Langerin, a novel Ctype lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules. Immunity. 2000; 12:71–81. [PubMed: 10661407] 9. Tang A. Amagai M. Granger L.G. Stanley J.R. Udey M.C. Adhesion of epidermal Langerhans cells to keratinocytes mediated by E-cadherin. Nature. 1993; 361:82–85. [PubMed: 8421498] 10. Riedl E. Stockl J. Majdic O. Scheinecker C. Knapp W. Strobl H. Ligation of E-cadherin on in vitro-generated immature Langerhans-type dendritic cells inhibits their maturation. Blood. 2000; 96:4276–4284. [PubMed: 11110702] 11. Uchi H. Terao H. Koga T. Furue M. Cytokines and chemokines in the epidermis. J Dermatol Sci. 2000; 24(Suppl 1):S29–S38. [PubMed: 11137393]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 14

Sponsored Document Sponsored Document Sponsored Document

12. Merad M. Manz M.G. Karsunky H. Wagers A. Peters W. Charo I. Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol. 2002; 3:1135–1141. [PubMed: 12415265] 13. Ginhoux F. Tacke F. Angeli V. Bogunovic M. Loubeau M. Dai X.M. Langerhans cells arise from monocytes in vivo. Nat Immunol. 2006; 7:265–273. [PubMed: 16444257] 14. Merad M. Hoffmann P. Ranheim E. Slaymaker S. Manz M.G. Lira S.A. Depletion of host Langerhans cells before transplantation of donor alloreactive T cells prevents skin graft-versushost disease. Nat Med. 2004; 10:510–517. [PubMed: 15098028] 15. Caux C. Dezutter-Dambuyant C. Schmitt D. Banchereau J. GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells. Nature. 1992; 360:258–261. [PubMed: 1279441] 16. Schaerli P. Willimann K. Ebert L.M. Walz A. Moser B. Cutaneous CXCL14 targets blood precursors to epidermal niches for Langerhans cell differentiation. Immunity. 2005; 23:331–342. [PubMed: 16169505] 17. Geissmann F. Prost C. Monnet J.P. Dy M. Brousse N. Hermine O. Transforming growth factor beta1, in the presence of granulocyte/macrophage colony-stimulating factor and interleukin 4, induces differentiation of human peripheral blood monocytes into dendritic Langerhans cells. J Exp Med. 1998; 187:961–966. [PubMed: 9500798] 18. Larregina A.T. Morelli A.E. Spencer L.A. Logar A.J. Watkins S.C. Thomson A.W. Dermalresident CD14+ cells differentiate into Langerhans cells. Nat Immunol. 2001; 2:1151–1158. [PubMed: 11702065] 19. Borkowski T.A. Letterio J.J. Farr A.G. Udey M.C. A role for endogenous transforming growth factor beta 1 in Langerhans cell biology: the skin of transforming growth factor beta 1 null mice is devoid of epidermal Langerhans cells. J Exp Med. 1996; 184:2417–2422. [PubMed: 8976197] 20. Schuler G. Steinman R.M. Murine epidermal Langerhans cells mature into potent immunostimulatory dendritic cells in vitro. J Exp Med. 1985; 161:526–546. [PubMed: 3871837] 21. Romani N. Koide S. Crowley M. Witmer-Pack M. Livingstone A.M. Fathman C.G. Presentation of exogenous protein antigens by dendritic cells to T cell clones. Intact protein is presented best by immature, epidermal Langerhans cells. J Exp Med. 1989; 169:1169–1178. [PubMed: 2522497] 22. Wilson N.S. Villadangos J.A. Lymphoid organ dendritic cells: beyond the Langerhans cells paradigm. Immunol Cell Biol. 2004; 82:91–98. [PubMed: 14984600] 23. Klechevsky E. Morita R. Liu M. Cao Y. Coquery S. Thompson-Snipes L. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008; 29:497–510. [PubMed: 18789730] 24. Stoitzner P. Tripp C.H. Eberhart A. Price K.M. Jung J.Y. Bursch L. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci USA. 2006; 103:7783–7788. [PubMed: 16672373] 25. Sullivan S. Bergstresser P.R. Tigelaar R.E. Streilein J.W. Induction and regulation of contact hypersensitivity by resident, bone marrow-derived, dendritic epidermal cells: Langerhans cells and Thy-1+ epidermal cells. J Immunol. 1986; 137:2460–2467. [PubMed: 2876041] 26. Grabbe S. Steinbrink K. Steinert M. Luger T.A. Schwarz T. Removal of the majority of epidermal Langerhans cells by topical or systemic steroid application enhances the effector phase of murine contact hypersensitivity. J Immunol. 1995; 155:4207–4217. [PubMed: 7594576] 27. Kissenpfennig A. Henri S. Dubois B. Laplace-Builhe C. Perrin P. Romani N. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity. 2005; 22:643–654. [PubMed: 15894281] 28. Kaplan D.H. Jenison M.C. Saeland S. Shlomchik W.D. Shlomchik M.J. Epidermal Langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity. 2005; 23:611–620. [PubMed: 16356859] 29. Bacci S. Alard P. Dai R. Nakamura T. Streilein J.W. High and low doses of haptens dictate whether dermal or epidermal antigen-presenting cells promote contact hypersensitivity. Eur J Immunol. 1997; 27:442–448. [PubMed: 9045915] 30. Bennett C.L. Noordegraaf M. Martina C.A. Clausen B.E. Langerhans cells are required for efficient presentation of topically applied hapten to T cells. J Immunol. 2007; 179:6830–6835. [PubMed: 17982073]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 15

Sponsored Document Sponsored Document Sponsored Document

31. Allan R.S. Smith C.M. Belz G.T. van Lint A.L. Wakim L.M. Heath W.R. Epidermal viral immunity induced by CD8alpha+ dendritic cells but not by Langerhans cells. Science. 2003; 301:1925–1928. [PubMed: 14512632] 32. Lenz A. Heine M. Schuler G. Romani N. Human and murine dermis contain dendritic cells. Isolation by means of a novel method and phenotypical and functional characterization. J Clin Invest. 1993; 92:2587–2596. [PubMed: 8254016] 33. Nestle F.O. Zheng X.G. Thompson C.B. Turka L.A. Nickoloff B.J. Characterization of dermal dendritic cells obtained from normal human skin reveals phenotypic and functionally distinctive subsets. J Immunol. 1993; 151:6535–6545. [PubMed: 7504023] 34. MacDonald K.P. Munster D.J. Clark G.J. Dzionek A. Schmitz J. Hart D.N. Characterization of human blood dendritic cell subsets. Blood. 2002; 100:4512–4520. [PubMed: 12393628] 35. Zaba L.C. Fuentes-Duculan J. Steinman R.M. Krueger J.G. Lowes M.A. Normal human dermis contains distinct populations of CD11c+ BDCA-1+ dendritic cells and CD163+ FXIIIA+ macrophages. J Clin Invest. 2007; 117:2517–2525. [PubMed: 17786242] 36. Zaba L.C. Krueger J.G. Lowes M.A. Resident and “inflammatory” dendritic cells in human skin. J Invest Dermatol. 2009; 129:302–308. [PubMed: 18685620] 37. Santegoets S.J. Gibbs S. Kroeze K. van de Ven R. Scheper R.J. Borrebaeck C.A. Transcriptional profiling of human skin-resident Langerhans cells and CD1a+ dermal dendritic cells: differential activation states suggest distinct functions. J Leukoc Biol. 2008; 84:143–151. [PubMed: 18436579] 38. Angel C.E. George E. Brooks A.E. Ostrovsky L.L. Brown T.L. Dunbar P.R. Cutting edge: CD1a+ antigen-presenting cells in human dermis respond rapidly to CCR7 ligands. J Immunol. 2006; 176:5730–5734. [PubMed: 16670277] 39. Haniffa M. Ginhoux F. Wang X.N. Bigley V. Abel M. Dimmick I. Differential rates of replacement of human dermal dendritic cells and macrophages during hematopoietic stem cell transplantation. J Exp Med. 2009; 206:371–385. [PubMed: 19171766] 40. Angel C.E. Lala A. Chen C.J. Edgar S.G. Ostrovsky L.L. Dunbar P.R. CD14+ antigen-presenting cells in human dermis are less mature than their CD1a+ counterparts. Int Immunol. 2007; 19:1271–1279. [PubMed: 17804688] 41. Morelli A.E. Rubin J.P. Erdos G. Tkacheva O.A. Mathers A.R. Zahorchak A.F. CD4+ T cell responses elicited by different subsets of human skin migratory dendritic cells. J Immunol. 2005; 175:7905–7915. [PubMed: 16339526] 42. Di Cesare A. Di Meglio P. Nestle F.O. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009; 129:1339–1350. [PubMed: 19322214] 43. Romani N. Ratzinger G. Pfaller K. Salvenmoser W. Stossel H. Koch F. Migration of dendritic cells into lymphatics-the Langerhans cell example: routes, regulation, and relevance. Int Rev Cytol. 2001; 207:237–270. [PubMed: 11352268] 44. Poulin L.F. Henri S. de Bovis B. Devilard E. Kissenpfennig A. Malissen B. The dermis contains langerin+ dendritic cells that develop and function independently of epidermal Langerhans cells. J Exp Med. 2007; 204:3119–3131. [PubMed: 18086861] 45. Ginhoux F. Collin M.P. Bogunovic M. Abel M. Leboeuf M. Helft J. Blood-derived dermal langerin+ dendritic cells survey the skin in the steady state. J Exp Med. 2007; 204:3133–3146. [PubMed: 18086862] 46. Bursch L.S. Wang L. Igyarto B. Kissenpfennig A. Malissen B. Kaplan D.H. Identification of a novel population of Langerin+ dendritic cells. J Exp Med. 2007; 204:3147–3156. [PubMed: 18086865] 47. Bedoui S. Whitney P.G. Waithman J. Eidsmo L. Wakim L. Caminschi I. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009; 10:488–495. [PubMed: 19349986] 48. Henri S. Poulin L.F. Tamoutounour S. Ardouin L. Guilliams M. de Bovis B. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J Exp Med. 2010; 207:189–206. S181–S186. [PubMed: 20038600]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 16

Sponsored Document Sponsored Document Sponsored Document

49. Bachem A. Guttler S. Hartung E. Ebstein F. Schaefer M. Tannert A. Superior antigen crosspresentation and XCR1 expression define human CD11c+ CD141+ cells as homologues of mouse CD8+ dendritic cells. J Exp Med. 2010 50. Jongbloed S.L. Kassianos A.J. McDonald K.J. Clark G.J. Ju X. Angel C.E. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010 51. Poulin L.F. Salio M. Griessinger E. Anjos-Afonso F. Craciun L. Chen J.L. Characterization of human DNGR-1+DCA3+ leukocytes as putative equivalents of mouse CD8{alpha}+ dendritic cells. J Exp Med. 2010 52. Demedts I.K. Brusselle G.G. Vermaelen K.Y. Pauwels R.A. Identification and characterization of human pulmonary dendritic cells. Am J Respir Cell Mol Biol. 2005; 32:177–184. [PubMed: 15576669] 53. Fiore N. Castellano G. Blasi A. Capobianco C. Loverre A. Montinaro V. Immature myeloid and plasmacytoid dendritic cells infiltrate renal tubulointerstitium in patients with lupus nephritis. Mol Immunol. 2008; 45:259–265. [PubMed: 17570528] 54. Narbutt J. Lesiak A. Sysa-Jedrzejowska A. Smolewski P. Robak T. Zalewska A. The number and distribution of blood dendritic cells in the epidermis and dermis of healthy human subjects. Folia Histochem Cytobiol. 2006; 44:61–63. [PubMed: 16584094] 55. Tsoumakidou M. Tzanakis N. Papadaki H.A. Koutala H. Siafakas N.M. Isolation of myeloid and plasmacytoid dendritic cells from human bronchoalveolar lavage fluid. Immunol Cell Biol. 2006; 84:267–273. [PubMed: 16509829] 56. Colonna M. Trinchieri G. Liu Y.J. Plasmacytoid dendritic cells in immunity. Nat Immunol. 2004; 5:1219–1226. [PubMed: 15549123] 57. Velasquez-Lopera M.M. Correa L.A. Garcia L.F. Human spleen contains different subsets of dendritic cells and regulatory T lymphocytes. Clin Exp Immunol. 2008; 154:107–114. [PubMed: 18727627] 58. Cella M. Jarrossay D. Facchetti F. Alebardi O. Nakajima H. Lanzavecchia A. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat Med. 1999; 5:919–923. [PubMed: 10426316] 59. Siegal F.P. Kadowaki N. Shodell M. Fitzgerald-Bocarsly P.A. Shah K. Ho S. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999; 284:1835–1837. [PubMed: 10364556] 60. Gilliet M. Cao W. Liu Y.J. Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nat Rev Immunol. 2008; 8:594–606. [PubMed: 18641647] 61. Nestle F.O. Conrad C. Tun-Kyi A. Homey B. Gombert M. Boyman O. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med. 2005; 202:135–143. [PubMed: 15998792] 62. Farkas L. Beiske K. Lund-Johansen F. Brandtzaeg P. Jahnsen F.L. Plasmacytoid dendritic cells (natural interferon- alpha/beta-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am J Pathol. 2001; 159:237–243. [PubMed: 11438470] 63. Meller S. Winterberg F. Gilliet M. Muller A. Lauceviciute I. Rieker J. Ultraviolet radiationinduced injury, chemokines, and leukocyte recruitment: an amplification cycle triggering cutaneous lupus erythematosus. Arthritis Rheum. 2005; 52:1504–1516. [PubMed: 15880822] 64. Vermi W. Bonecchi R. Facchetti F. Bianchi D. Sozzani S. Festa S. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas. J Pathol. 2003; 200:255–268. [PubMed: 12754747] 65. Albanesi C. Scarponi C. Pallotta S. Daniele R. Bosisio D. Madonna S. Chemerin expression marks early psoriatic skin lesions and correlates with plasmacytoid dendritic cell recruitment. J Exp Med. 2009; 206:249–258. [PubMed: 19114666] 66. Lovgren T. Eloranta M.L. Bave U. Alm G.V. Ronnblom L. 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. 2004; 50:1861–1872. [PubMed: 15188363]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 17

Sponsored Document Sponsored Document Sponsored Document

67. Lande R. Gregorio J. Facchinetti V. Chatterjee B. Wang Y.H. Homey B. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature. 2007; 449:564–569. [PubMed: 17873860] 68. Ganguly D. Chamilos G. Lande R. Gregorio J. Meller S. Facchinetti V. Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. J Exp Med. 2009; 206:1983–1994. [PubMed: 19703986] 69. Steinman R.M. Hawiger D. Nussenzweig M.C. Tolerogenic dendritic cells. Annu Rev Immunol. 2003; 21:685–711. [PubMed: 12615891] 70. Hawiger D. Inaba K. Dorsett Y. Guo M. Mahnke K. Rivera M. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med. 2001; 194:769–779. [PubMed: 11560993] 71. Liu K. Iyoda T. Saternus M. Kimura Y. Inaba K. Steinman R.M. Immune tolerance after delivery of dying cells to dendritic cells in situ. J Exp Med. 2002; 196:1091–1097. [PubMed: 12391020] 72. Yamazaki S. Dudziak D. Heidkamp G.F. Fiorese C. Bonito A.J. Inaba K. CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells. J Immunol. 2008; 181:6923– 6933. [PubMed: 18981112] 73. Obhrai J.S. Oberbarnscheidt M. Zhang N. Mueller D.L. Shlomchik W.D. Lakkis F.G. Langerhans cells are not required for efficient skin graft rejection. J Invest Dermatol. 2008; 128:1950–1955. [PubMed: 18337832] 74. Loser K. Mehling A. Loeser S. Apelt J. Kuhn A. Grabbe S. Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat Med. 2006; 12:1372–1379. [PubMed: 17143276] 75. Yoshiki R. Kabashima K. Sugita K. Atarashi K. Shimauchi T. Tokura Y. IL-10-producing Langerhans cells and regulatory T cells are responsible for depressed contact hypersensitivity in grafted skin. J Invest Dermatol. 2009; 129:705–713. [PubMed: 18843293] 76. Waithman J. Allan R.S. Kosaka H. Azukizawa H. Shortman K. Lutz M.B. Skin-derived dendritic cells can mediate deletional tolerance of class I-restricted self-reactive T cells. J Immunol. 2007; 179:4535–4541. [PubMed: 17878350] 77. Dhodapkar M.V. Steinman R.M. Krasovsky J. Munz C. Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001; 193:233–238. [PubMed: 11208863] 78. Jonuleit H. Schmitt E. Schuler G. Knop J. Enk A.H. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med. 2000; 192:1213–1222. [PubMed: 11067871] 79. Banerjee D.K. Dhodapkar M.V. Matayeva E. Steinman R.M. Dhodapkar K.M. Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients. Blood. 2006; 108:2655–2661. [PubMed: 16763205] 80. Menges M. Rossner S. Voigtlander C. Schindler H. Kukutsch N.A. Bogdan C. 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. [PubMed: 11781361] 81. Nestle F.O. Kaplan D.H. Barker J. Psoriasis. N Engl J Med. 2009; 361:496–509. [PubMed: 19641206] 82. Trembath R.C. Clough R.L. Rosbotham J.L. Jones A.B. Camp R.D. Frodsham A. Identification of a major susceptibility locus on chromosome 6p and evidence for further disease loci revealed by a two stage genome-wide search in psoriasis. Hum Mol Genet. 1997; 6:813–820. [PubMed: 9158158] 83. Nair R.P. Duffin K.C. Helms C. Ding J. Stuart P.E. Goldgar D. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat Genet. 2009; 41:199–204. [PubMed: 19169254] 84. Nair R.P. Stuart P.E. Nistor I. Hiremagalore R. Chia N.V. Jenisch S. Sequence and haplotype analysis supports HLA-C as the psoriasis susceptibility 1 gene. Am J Hum Genet. 2006; 78:827– 851. [PubMed: 16642438]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 18

Sponsored Document Sponsored Document Sponsored Document

85. Capon F. Di Meglio P. Szaub J. Prescott N.J. Dunster C. Baumber L. Sequence variants in the genes for the interleukin-23 receptor (IL23R) and its ligand (IL12B) confer protection against psoriasis. Hum Genet. 2007; 122:201–206. [PubMed: 17587057] 86. Cargill M. Schrodi S.J. Chang M. Garcia V.E. Brandon R. Callis K.P. A large-scale genetic association study confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes. Am J Hum Genet. 2007; 80:273–290. [PubMed: 17236132] 87. Ellinghaus E. Ellinghaus D. Stuart P.E. Nair R.P. Debrus S. Raelson J.V. Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet. 2010; 42:991–995. [PubMed: 20953188] 88. Huffmeier U. Uebe S. Ekici A.B. Bowes J. Giardina E. Korendowych E. Common variants at TRAF3IP2 are associated with susceptibility to psoriatic arthritis and psoriasis. Nat Genet. 2010; 42:996–999. [PubMed: 20953186] 89. Strange A. Capon F. Spencer C.C. Knight J. Weale M.E. Allen M.H. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010; 42:985–990. [PubMed: 20953190] 90. Sun L.D. Cheng H. Wang Z.X. Zhang A.P. Wang P.G. Xu J.H. Association analyses identify six new psoriasis susceptibility loci in the Chinese population. Nat Genet. 2010; 42:1005–1009. [PubMed: 20953187] 91. de Cid R. Riveira-Munoz E. Zeeuwen P.L. Robarge J. Liao W. Dannhauser E.N. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nat Genet. 2009; 41:211–215. [PubMed: 19169253] 92. Stuart P.E. Nair R.P. Ellinghaus E. Ding J. Tejasvi T. Gudjonsson J.E. Genome-wide association analysis identifies three psoriasis susceptibility loci. Nat Genet. 2010; 42:1000–1004. [PubMed: 20953189] 93. Capon F. Bijlmakers M.J. Wolf N. Quaranta M. Huffmeier U. Allen M. Identification of ZNF313/ RNF114 as a novel psoriasis susceptibility gene. Hum Mol Genet. 2008; 17:1938–1945. [PubMed: 18364390] 94. Quaranta M. Burden A.D. Griffiths C.E. Worthington J. Barker J.N. Trembath R.C. Differential contribution of CDKAL1 variants to psoriasis, Crohn's disease and type II diabetes. Genes Immun. 2009; 10:654–658. [PubMed: 19587699] 95. Gilliet M. Conrad C. Geiges M. Cozzio A. Thurlimann W. Burg G. Psoriasis triggered by toll-like receptor 7 agonist imiquimod in the presence of dermal plasmacytoid dendritic cell precursors. Arch Dermatol. 2004; 140:1490–1495. [PubMed: 15611427] 96. Schmid P. Itin P. Cox D. McMaster G.K. Horisberger M.A. The type I interferon system is locally activated in psoriatic lesions. J Interferon Res. 1994; 14:229–234. [PubMed: 7861026] 97. Wollenberg A. Wagner M. Gunther S. Towarowski A. Tuma E. Moderer M. Plasmacytoid dendritic cells: a new cutaneous dendritic cell subset with distinct role in inflammatory skin diseases. J Invest Dermatol. 2002; 119:1096–1102. [PubMed: 12445198] 98. Nestle F.O. Turka L.A. Nickoloff B.J. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. J Clin Invest. 1994; 94:202–209. [PubMed: 8040262] 99. Austin L.M. Ozawa M. Kikuchi T. Walters I.B. Krueger J.G. The majority of epidermal T cells in Psoriasis vulgaris lesions can produce type 1 cytokines, interferon-gamma, interleukin-2, and tumor necrosis factor-alpha, defining TC1 (cytotoxic T lymphocyte) and TH1 effector populations: a type 1 differentiation bias is also measured in circulating blood T cells in psoriatic patients. J Invest Dermatol. 1999; 113:752–759. [PubMed: 10571730] 100. Nestle F.O. Conrad C. The IL-12 family member p40 chain as a master switch and novel therapeutic target in psoriasis. J Invest Dermatol. 2004; 123 xiv–xv. 101. Zaba L.C. Cardinale I. Gilleaudeau P. Sullivan-Whalen M. Suarez-Farinas M. Fuentes-Duculan J. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J Exp Med. 2007; 204:3183–3194. [PubMed: 18039949] 102. Zaba L.C. Fuentes-Duculan J. Eungdamrong N.J. Abello M.V. Novitskaya I. Pierson K.C. Psoriasis is characterized by accumulation of immunostimulatory and Th1/Th17 cell-polarizing myeloid dendritic cells. J Invest Dermatol. 2009; 129:79–88. [PubMed: 18633443]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 19

Sponsored Document Sponsored Document Sponsored Document

103. Serbina N.V. Salazar-Mather T.P. Biron C.A. Kuziel W.A. Pamer E.G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity. 2003; 19:59–70. [PubMed: 12871639] 104. Engel D. Dobrindt U. Tittel A. Peters P. Maurer J. Gutgemann I. Tumor necrosis factor alphaand inducible nitric oxide synthase-producing dendritic cells are rapidly recruited to the bladder in urinary tract infection but are dispensable for bacterial clearance. Infect Immun. 2006; 74:6100–6107. [PubMed: 16966414] 105. Copin R. De Baetselier P. Carlier Y. Letesson J.J. Muraille E. MyD88-dependent activation of B220-CD11b+ LY-6C+ dendritic cells during Brucella melitensis infection. J Immunol. 2007; 178:5182–5191. [PubMed: 17404301] 106. Aldridge J.R. Jr. Moseley C.E. Boltz D.A. Negovetich N.J. Reynolds C. Franks J. TNF/iNOSproducing dendritic cells are the necessary evil of lethal influenza virus infection. Proc Natl Acad Sci USA. 2009; 106:5306–5311. [PubMed: 19279209] 107. Lowes M.A. Chamian F. Abello M.V. Fuentes-Duculan J. Lin S.L. Nussbaum R. Increase in TNF-alpha and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proc Natl Acad Sci USA. 2005; 102:19057–19062. [PubMed: 16380428] 108. Wang F. Lee E. Lowes M.A. Haider A.S. Fuentes-Duculan J. Abello M.V. Prominent production of IL-20 by CD68+/CD11c+ myeloid-derived cells in psoriasis: gene regulation and cellular effects. J Invest Dermatol. 2006; 126:1590–1599. [PubMed: 16645593] 109. Lee E. Trepicchio W.L. Oestreicher J.L. Pittman D. Wang F. Chamian F. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med. 2004; 199:125–130. [PubMed: 14707118] 110. Oppmann B. Lesley R. Blom B. Timans J.C. Xu Y. Hunte B. Novel p19 protein engages IL-12p40 to form a cytokine. IL-23, with biological activities similar as well as distinct from IL-12. Immunity. 2000; 13:715–725. [PubMed: 11114383] 111. Piskin G. Tursen U. Sylva-Steenland R.M. Bos J.D. Teunissen M.B. Clinical improvement in chronic plaque-type psoriasis lesions after narrow-band UVB therapy is accompanied by a decrease in the expression of IFN-gamma inducers – IL-12, IL-18 and IL-23. Exp Dermatol. 2004; 13:764–772. [PubMed: 15560760] 112. Gottlieb A.B. Chamian F. Masud S. Cardinale I. Abello M.V. Lowes M.A. TNF inhibition rapidly down-regulates multiple proinflammatory pathways in psoriasis plaques. J Immunol. 2005; 175:2721–2729. [PubMed: 16081850] 113. Chan J.R. Blumenschein W. Murphy E. Diveu C. Wiekowski M. Abbondanzo S. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med. 2006; 203:2577–2587. [PubMed: 17074928] 114. Tonel G. Conrad C. Laggner U. Di Meglio P. Grys K. McClanahan T.K. Cutting edge: a critical functional role for IL-23 in psoriasis. J Immunol. 2010; 185:5688–5691. [PubMed: 20956338] 115. Lowes M.A. Kikuchi T. Fuentes-Duculan J. Cardinale I. Zaba L.C. Haider A.S. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. J Invest Dermatol. 2008; 128:1207– 1211. [PubMed: 18200064] 116. Wilson N.J. Boniface K. Chan J.R. McKenzie B.S. Blumenschein W.M. Mattson J.D. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol. 2007; 8:950–957. [PubMed: 17676044] 117. Guttman-Yassky E. Lowes M.A. Fuentes-Duculan J. Zaba L.C. Cardinale I. Nograles K.E. Low expression of the IL-23/Th17 pathway in atopic dermatitis compared to psoriasis. J Immunol. 2008; 181:7420–7427. [PubMed: 18981165] 118. Piskin G. Sylva-Steenland R.M.R. Bos J.D. Teunissen M.B.M. In vitro and in situ expression of IL-23 by keratinocytes in healthy skin and psoriasis lesions: enhanced expression in psoriatic skin. J Immunol. 2006; 176:1908–1915. [PubMed: 16424222] 119. Yawalkar N. Karlen S. Hunger R. Brand C.U. Braathen L.R. Expression of interleukin-12 is increased in psoriatic skin. J Invest Dermatol. 1998; 111:1053–1057. [PubMed: 9856816]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 20

Sponsored Document Sponsored Document Sponsored Document

120. Yawalkar N. Tscharner G.G. Hunger R.E. Hassan A.S. Increased expression of IL-12p70 and IL-23 by multiple dendritic cell and macrophage subsets in plaque psoriasis. J Dermatol Sci. 2009; 54:99–105. [PubMed: 19264456] 121. Zaba L.C. Fuentes-Duculan J. Eungdamrong N.J. Johnson-Huang L.M. Nograles K.E. White T.R. Identification of TNF-related apoptosis-inducing ligand and other molecules that distinguish inflammatory from resident dendritic cells in patients with psoriasis. J Allergy Clin Immunol. 2010; 125:1261–1268. e1269. [PubMed: 20471070] 122. Guttman-Yassky E. Lowes M.A. Fuentes-Duculan J. Whynot J. Novitskaya I. Cardinale I. Major differences in inflammatory dendritic cells and their products distinguish atopic dermatitis from psoriasis. J Allergy Clin Immunol. 2007; 119:1210–1217. [PubMed: 17472813] 123. Johnson-Huang L.M. McNutt N.S. Krueger J.G. Lowes M.A. Cytokine-producing dendritic cells in the pathogenesis of inflammatory skin diseases. J Clin Immunol. 2009; 29:247–256. [PubMed: 19252974] 124. Bieber T. Atopic dermatitis. N Engl J Med. 2008; 358:1483–1494. [PubMed: 18385500] 125. Palmer C.N. Irvine A.D. Terron-Kwiatkowski A. Zhao Y. Liao H. Lee S.P. Common loss-offunction variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet. 2006; 38:441–446. [PubMed: 16550169] 126. Smith F.J. Irvine A.D. Terron-Kwiatkowski A. Sandilands A. Campbell L.E. Zhao Y. Loss-offunction mutations in the gene encoding filaggrin cause ichthyosis vulgaris. Nat Genet. 2006; 38:337–342. [PubMed: 16444271] 127. Grewe M. Walther S. Gyufko K. Czech W. Schopf E. Krutmann J. Analysis of the cytokine pattern expressed in situ in inhalant allergen patch test reactions of atopic dermatitis patients. J Invest Dermatol. 1995; 105:407–410. [PubMed: 7665922] 128. Wollenberg A. Kraft S. Hanau D. Bieber T. Immunomorphological and ultrastructural characterization of Langerhans cells and a novel, inflammatory dendritic epidermal cell (IDEC) population in lesional skin of atopic eczema. J Invest Dermatol. 1996; 106:446–453. [PubMed: 8648175] 129. Bieber T. de la Salle H. Wollenberg A. Hakimi J. Chizzonite R. Ring J. Human epidermal Langerhans cells express the high affinity receptor for immunoglobulin E (Fc epsilon RI). J Exp Med. 1992; 175:1285–1290. [PubMed: 1533242] 130. Novak N. Valenta R. Bohle B. Laffer S. Haberstok J. Kraft S. Fc epsilon RI engagement of Langerhans cell-like dendritic cells and inflammatory dendritic epidermal cell-like dendritic cells induces chemotactic signals and different T-cell phenotypes in vitro. J Allergy Clin Immunol. 2004; 113:949–957. [PubMed: 15131579] 131. Soumelis V. Reche P.A. Kanzler H. Yuan W. Edward G. Homey B. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002; 3:673– 680. [PubMed: 12055625] 132. Liu Y.J. Soumelis V. Watanabe N. Ito T. Wang Y.H. Malefyt Rde W. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007; 25:193–219. [PubMed: 17129180] 133. Ebner S. Nguyen V.A. Forstner M. Wang Y.H. Wolfram D. Liu Y.J. Thymic stromal lymphopoietin converts human epidermal Langerhans cells into antigen-presenting cells that induce proallergic T cells. J Allergy Clin Immunol. 2007; 119:982–990. [PubMed: 17320941] 134. Wollenberg A. Mommaas M. Oppel T. Schottdorf E.M. Gunther S. Moderer M. Expression and function of the mannose receptor CD206 on epidermal dendritic cells in inflammatory skin diseases. J Invest Dermatol. 2002; 118:327–334. [PubMed: 11841552] 135. Erkin G. Ugur Y. Gurer C.K. Asan E. Korkusuz P. Sahin S. Effect of PUVA, narrow-band UVB and cyclosporin on inflammatory cells of the psoriatic plaque. J Cutan Pathol. 2007; 34:213–219. [PubMed: 17302604] 136. Johnson-Huang L.M. Suarez-Farinas M. Sullivan-Whalen M. Gilleaudeau P. Krueger J.G. Lowes M.A. Effective narrow-band UVB radiation therapy suppresses the IL-23/IL-17 axis in normalized psoriasis plaques. J Invest Dermatol. 2010 137. Haider A.S. Lowes M.A. Suarez-Farinas M. Zaba L.C. Cardinale I. Khatcherian A. Identification of cellular pathways of “Type 1,” Th17 T cells, and TNF- and inducible nitric oxide synthase-

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 21

Sponsored Document Sponsored Document Sponsored Document

producing dendritic cells in autoimmune inflammation through pharmacogenomic study of cyclosporine A in psoriasis. J Immunol. 2008; 180:1913–1920. [PubMed: 18209089] 138. Nickoloff B.J. Xin H. Nestle F.O. Qin J.Z. The cytokine and chemokine network in psoriasis. Clin Dermatol. 2007; 25:568–573. [PubMed: 18021894] 139. Zaba L.C. Cardinale I. Gilleaudeau P. Sullivan-Whalen M. Suarez Farinas M. Fuentes-Duculan J. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J Exp Med. 2007; 204:3183–3194. [PubMed: 18039949] 140. Zaba L.C. Suarez-Farinas M. Fuentes-Duculan J. Nograles K.E. Guttman-Yassky E. Cardinale I. Effective treatment of psoriasis with etanercept is linked to suppression of IL-17 signaling, not immediate response TNF genes. J Allergy Clin Immunol. 2009; 124:1022–1030. e1021–e1395. [PubMed: 19895991] 141. Marble D.J. Gordon K.B. Nickoloff B.J. Targeting TNFalpha rapidly reduces density of dendritic cells and macrophages in psoriatic plaques with restoration of epidermal keratinocyte differentiation. J Dermatol Sci. 2007; 48:87–101. [PubMed: 17689932] 142. Bedini C. Nasorri F. Girolomoni G. Pita O. Cavani A. Antitumour necrosis factor-alpha chimeric antibody (infliximab) inhibits activation of skin-homing CD4+ and CD8+ T lymphocytes and impairs dendritic cell function. Br J Dermatol. 2007; 157:249–258. [PubMed: 17489975] 143. Gandhi M. Alwawi E. Gordon K.B. Anti-p40 antibodies ustekinumab and briakinumab: blockade of interleukin-12 and interleukin-23 in the treatment of psoriasis. Semin Cutan Med Surg. 2010; 29:48–52. [PubMed: 20430307] 144. Leonardi C.L. Kimball A.B. Papp K.A. Yeilding N. Guzzo C. Wang Y. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet. 2008; 371:1665–1674. [PubMed: 18486739] 145. Papp K.A. Langley R.G. Lebwohl M. Krueger G.G. Szapary P. Yeilding N. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008; 371:1675–1684. [PubMed: 18486740] 146. Toichi E. Torres G. McCormick T.S. Chang T. Mascelli M.A. Kauffman C.L. An anti-IL-12p40 antibody down-regulates type 1 cytokines, chemokines, and IL-12/IL-23 in psoriasis. J Immunol. 2006; 177:4917–4926. [PubMed: 16982934] 147. Chamian F. Lowes M.A. Lin S.L. Lee E. Kikuchi T. Gilleaudeau P. Alefacept reduces infiltrating T cells, activated dendritic cells, and inflammatory genes in psoriasis vulgaris. Proc Natl Acad Sci USA. 2005; 102:2075–2080. [PubMed: 15671179] 148. Tan C.S. Koralnik I.J. Progressive multifocal leukoencephalopathy and other disorders caused by JC virus: clinical features and pathogenesis. Lancet Neurol. 2010; 9:425–437. [PubMed: 20298966] 149. Adorini L. Giarratana N. Penna G. Pharmacological induction of tolerogenic dendritic cells and regulatory T cells. Semin Immunol. 2004; 16:127–134. [PubMed: 15036236] 150. Hackstein H. Thomson A.W. Dendritic cells: emerging pharmacological targets of immunosuppressive drugs. Nat Rev Immunol. 2004; 4:24–34. [PubMed: 14704765] 151. Chang C.C. Ciubotariu R. Manavalan J.S. Yuan J. Colovai A.I. Piazza F. Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol. 2002; 3:237–243. [PubMed: 11875462] 152. Hackstein H. Taner T. Logar A.J. Thomson A.W. Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells. Blood. 2002; 100:1084–1087. [PubMed: 12130531] 153. Penna G. Giarratana N. Amuchastegui S. Mariani R. Daniel K.C. Adorini L. Manipulating dendritic cells to induce regulatory T cells. Microbes Infect. 2005; 7:1033–1039. [PubMed: 15919222] 154. Penna G. Adorini L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol. 2000; 164:2405–2411. [PubMed: 10679076]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 22

Sponsored Document Sponsored Document Sponsored Document

155. Rea D. van Kooten C. van Meijgaarden K.E. Ottenhoff T.H. Melief C.J. Offringa R. Glucocorticoids transform CD40-triggering of dendritic cells into an alternative activation pathway resulting in antigen-presenting cells that secrete IL-10. Blood. 2000; 95:3162–3167. [PubMed: 10807783] 156. Woltman A.M. de Fijter J.W. Kamerling S.W. van Der Kooij S.W. Paul L.C. Daha M.R. Rapamycin induces apoptosis in monocyte- and CD34-derived dendritic cells but not in monocytes and macrophages. Blood. 2001; 98:174–180. [PubMed: 11418477] 157. Woltman A.M. Massacrier C. de Fijter J.W. Caux C. van Kooten C. Corticosteroids prevent generation of CD34+ -derived dermal dendritic cells but do not inhibit Langerhans cell development. J Immunol. 2002; 168:6181–6188. [PubMed: 12055231] 158. Adorini L. Penna G. Giarratana N. Uskokovic M. Tolerogenic dendritic cells induced by vitamin D receptor ligands enhance regulatory T cells inhibiting allograft rejection and autoimmune diseases. J Cell Biochem. 2003; 88:227–233. [PubMed: 12520519] 159. Manavalan J.S. Rossi P.C. Vlad G. Piazza F. Yarilina A. Cortesini R. High expression of ILT3 and ILT4 is a general feature of tolerogenic dendritic cells. Transpl Immunol. 2003; 11:245–258. [PubMed: 12967778] 160. Kim-Schulze S. Scotto L. Vlad G. Piazza F. Lin H. Liu Z. Recombinant Ig-like transcript 3-Fc modulates T cell responses via induction of Th anergy and differentiation of CD8+ T suppressor cells. J Immunol. 2006; 176:2790–2798. [PubMed: 16493035] 161. Penna G. Roncari A. Amuchastegui S. Daniel K.C. Berti E. Colonna M. Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4+ Foxp3+ regulatory T cells by 1,25-dihydroxyvitamin D3. Blood. 2005; 106:3490–3497. [PubMed: 16030186] 162. Fenton C. Plosker G.L. Calcipotriol/betamethasone dipropionate: a review of its use in the treatment of psoriasis vulgaris. Am J Clin Dermatol. 2004; 5:463–478. [PubMed: 15663344] 163. Scott L.J. Dunn C.J. Goa K.L. Calcipotriol ointment. A review of its use in the management of psoriasis. Am J Clin Dermatol. 2001; 2:95–120. [PubMed: 11705309] 164. Pedersen A.W. Holmstrom K. Jensen S.S. Fuchs D. Rasmussen S. Kvistborg P. Phenotypic and functional markers for 1alpha,25-dihydroxyvitamin D(3)-modified regulatory dendritic cells. Clin Exp Immunol. 2009; 157:48–59. [PubMed: 19659770] 165. Kowitz A. Greiner M. Thieroff-Ekerdt R. Inhibitory effect of 1alpha,25-dihydroxyvitamin D3 on allogeneic lymphocyte stimulation and Langerhans cell maturation. Arch Dermatol Res. 1998; 290:540–546. [PubMed: 9836504] 166. Dam T.N. Moller B. Hindkjaer J. Kragballe K. The vitamin D3 analog calcipotriol suppresses the number and antigen-presenting function of Langerhans cells in normal human skin. J Investig Dermatol Symp Proc. 1996; 1:72–77. 167. Warger T. Schild H. Rechtsteiner G. Initiation of adaptive immune responses by transcutaneous immunization. Immunol Lett. 2007; 109:13–20. [PubMed: 17320194] 168. Romani N. Thurnher M. Idoyaga J. Steinman R.M. Flacher V. Targeting of antigens to skin dendritic cells: possibilities to enhance vaccine efficacy. Immunol Cell Biol. 2010; 88:424–430. [PubMed: 20368713] 169. Bonifaz L. Bonnyay D. Mahnke K. Rivera M. Nussenzweig M.C. Steinman R.M. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J Exp Med. 2002; 196:1627–1638. [PubMed: 12486105] 170. Bonifaz L.C. Bonnyay D.P. Charalambous A. Darguste D.I. Fujii S. Soares H. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med. 2004; 199:815–824. [PubMed: 15024047] 171. Joffre O.P. Sancho D. Zelenay S. Keller A.M. Reis e Sousa C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/ CLEC9A. Eur J Immunol. 2010; 40:1255–1265. [PubMed: 20333625] 172. Bruder D. Westendorf A.M. Hansen W. Prettin S. Gruber A.D. Qian Y. On the edge of autoimmunity: T-cell stimulation by steady-state dendritic cells prevents autoimmune diabetes. Diabetes. 2005; 54:3395–3401. [PubMed: 16306354]

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 23

Sponsored Document

173. Mukhopadhaya A. Hanafusa T. Jarchum I. Chen Y.G. Iwai Y. Serreze D.V. Selective delivery of beta cell antigen to dendritic cells in vivo leads to deletion and tolerance of autoreactive CD8+ T cells in NOD mice. Proc Natl Acad Sci USA. 2008; 105:6374–6379. [PubMed: 18430797] 174. Tanriver Y. Ratnasothy K. Bucy R.P. Lombardi G. Lechler R. Targeting MHC class I monomers to dendritic cells inhibits the indirect pathway of allorecognition and the production of IgG alloantibodies leading to long-term allograft survival. J Immunol. 2010; 184:1757–1764. [PubMed: 20083658] 175. Ghoreishi M. Bach P. Obst J. Komba M. Fleet J.C. Dutz J.P. Expansion of antigen-specific regulatory T cells with the topical vitamin d analog calcipotriol. J Immunol. 2009; 182:6071– 6078. [PubMed: 19414758] 176. Ghoreishi M. Dutz J.P. Tolerance induction by transcutaneous immunization through ultravioletirradiated skin is transferable through CD4+ CD25+ T regulatory cells and is dependent on hostderived IL-10. J Immunol. 2006; 176:2635–2644. [PubMed: 16456026] 177. Sigmundsdottir H. Pan J. Debes G.F. Alt C. Habtezion A. Soler D. DCs metabolize sunlightinduced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol. 2007; 8:285–293. [PubMed: 17259988]

Acknowledgments

Sponsored Document

We apologize to all the authors whose work could not be discussed and cited due to space limitations. We acknowledge support by the following grant funding bodies: Wellcome Trust Programme GR078173MA, NIH RO1AR040065, NIHR Comprehensive Biomedical Research Centre Guy's and St. Thomas’ Hospital and King's College London, Medical Research Council UK Programme G0601387, Dunhill Medical Trust. C.C. Chu is supported by St. John's Institute of Dermatology Studentship and Overseas Research Students Awards Scheme (ORSAS).

Sponsored Document Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 24

Sponsored Document Sponsored Document Fig. 1.

Dendritic cell populations in the skin of human and mice. aCommon cellular markers associated with human and mice skin DC subsets.

Sponsored Document Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 25

Sponsored Document Sponsored Document

Fig. 2.

Sponsored Document

Skin DC subsets and function during tissue homeostasis. Healthy skin consists of a complex network of DCs that play important roles during tissue homeostasis. In the steady state, CD207+ DDCs (found in the dermis of mice) can capture dead cells or tissue antigens and promote T cell tolerance via cross-presentation and/or Treg induction in the draining lymph nodes. During tissue damage or infection, LCs and dermal DDCs may initiate cytotoxic T lymphocyte (Tc) responses via cross-presentation. Infected LCs that undergo apoptosis/ necrosis could potentially transfer antigens to other cell types capable of cross-presentation. LCs are re-populated by resident or circulating precursors. CD14+ DDCs polarize follicular T helper cells (Thf) which promote B cell development, while other DDCs (including CD1a+ DDCs) can initiate Th responses which may be sustained by pDCs via IFN-α production. Dotted lines refer to speculative relationships.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 26

Sponsored Document Sponsored Document

Fig. 3.

Sponsored Document

A DC perspective on psoriasis pathogenesis. A combination of environmental triggers and genetic factors leads to disease onset. In the initial phase, pDCs are recruited to pre-psoriatic skin through chemerin and activated by self DNA and/or RNA that is released by keratinocytes and forms complexes with LL-37. IFN-α released by activated pDCs, selfRNA-LL-37 complexes, as well as keratinocyte-derived pro-inflammatory cytokines, TNF, IL-6, and IL-1β, leads to activation of DDCs. Activated DDCs migrate to the skin-draining lymph nodes where they promote the differentiation of naïve T cells into Th1 and/or Th17 cells. In psoriatic lesions, DDCs and inflammatory DCs produce IL-12, TNF, IL-20, nitric oxide (NO) radicals and IL-23 which activate skin resident T cells to produce proinflammatory cytokines. Pro-inflammatory Th1 and Th17 cytokines then act on keratinocytes and feedback on DDCs, leading to a sustained and amplified inflammatory status which ultimately leads to the formation of a psoriatic plaque.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 27

Sponsored Document Sponsored Document Fig. 4.

Sponsored Document

Pathogenic roles of skin DC subsets in atopic dermatitis. In atopic dermatitis (AD), genetically determined epidermal-barrier dysfunction allows the penetration in the skin of allergens that trigger an inflammatory response sustained by cells of the innate and adaptive immunity. In the initial phase of the disease, FcɛRI ligation induces epidermal LCs to produce eosinophils and Th2 cells recruiting-cytokines IL-8 and IL-16 and to migrate to local skin-draining lymph nodes where they promote polarization of Th2 cells producing IL-4, IL-5, IL-10 and IL-13. In the acute phase of the disease, characterized by eczematous plaques and spongiosis, activated LC can induce Th2 response in situ in the skin, possibly both in the epidermis and in the dermis, the latter being favored by thymic stromal lymphopoietin (TSLP) produced by keratinocytes. TSLP also promotes DDC antigenindependent maturation and production of pro-inflammatory cytokines and chemokines (CCL-17, CCL-24, CCL-22, IL-8, IL-15) enhancing T cell recruitment and survival in the skin. In the chronic phase of AD, characterized by increased epidermal thickness and plaques lichenification, FcɛRI ligation of IDECs promotes production of IL-12 and IL-18, thus inducing a switch toward a Th1-polarizing environment with Th1 cells secreting IFN-γ. IDEC producing T cell recruiting-chemokines CCL-17 and CCL-18 have been also recently identified in the dermis, raising the possibility that they might migrate from/to the epidermis or are generated from immature and/or activated DDCs.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.

Chu et al.

Page 28

Sponsored Document Sponsored Document

Fig. 5.

Sponsored Document

Harnessing skin DCs for therapeutic intervention in psoriasis. Effective targeting of the psoriatic cytokine network with TNF neutralizing antibodies or soluble receptors reduces DDC numbers, interferes with DDC activation, and inhibits down-stream effector molecules. Targeting the common IL-12/IL-23p40 subunit reduces the production of DCderived pro-inflammatory cytokines and impairs Th1 and Th17 cell responses. Immunosuppressive agents may interfere with LC distribution, DDC activation, and promote the generation of regulatory T cells. Finally, targeting LFA-1 blocks T cell migration and interaction with DCs, while human LFA-3 Ig fusion protein binds to CD2expressing cells and interferes with DC-T cell interaction. *Efalizumab has recently been withdrawn from the market due to its side effects.

Published as: Semin Immunol. 2011 February ; 23(1): 28–41.