COMMENTARY Human Vaccines & Immunotherapeutics 12:1, 104--116; January 2016; © 2016 Taylor & Francis Group, LLC

Duality at the gate: Skin dendritic cells as mediators of vaccine immunity and tolerance Christopher J Nirschl and Niroshana Anandasabapathy* Department of Dermatology; Harvard Skin Disease Research Center; Brigham and Women’s Hospital; Boston, MA USA

S

Keywords: dendritic cells, DC targeted vaccines, immune tolerance, migratory DCs Abbreviations: DCs, Dendritic Cells; APCs, Antigen Presenting Cells; LCs, Langerhans Cells; migDC, migratory DCs; FLT-3, Fms-like Tyrosine Kinase 3; STAT3, Signal Transducer and Activator of Transcription 3; LN, Lymph Node. *Correspondence to: Niroshana Anandasabapathy. Email: [email protected] Submitted: 06/04/2015 Accepted: 06/20/2015 http://dx.doi.org/10.1080/21645515.2015.1066050 104

ince Edward Jenner’s discovery that intentional exposure to cowpox could provide lifelong protection from smallpox, vaccinations have been a major focus of medical research. However, while the protective benefits of many vaccines have been successfully translated into the clinic, the cellular and molecular mechanisms that differentiate effective vaccines from suboptimal ones are not well understood. Dendritic cells (DCs) are the gatekeepers of the immune system, and are ultimately responsible for the generation of adaptive immunity and lifelong protective memory through interactions with T cells. In addition to lymph node and spleen resident DCs, a number of tissue resident DC populations have been identified at barrier tissues, such as the skin, which migrate to the local lymph node (migDC). These populations have unique characteristics, and play a key role in the function of cutaneous vaccinations by shuttling antigen from the vaccination site to the draining lymph node, rapidly capturing freely draining antigens in the lymph node, and providing key stimuli to T cells. However, while migDCs are responsible for the generation of immunity following exposure to certain pathogens and vaccines, recent work has identified a tolerogenic role for migDCs in the steady state as well as during protein immunization. Here, we examine the roles and functions of skin DC populations in the generation of protective immunity, as well as their role as regulators of the immune system.

Introduction The immune system is responsible for defending its host against a wide variety of Human Vaccines & Immunotherapeutics

pathogens on a daily basis. Two key characteristics of this protection are the generation of specific, sterilizing immunity, and the formation of long lived immunological memory, both of which require an adaptive immune response. Clinically, the protective benefits of the immune system have been successfully harnessed through the development of vaccines. However, the cellular mechanisms underlying longterm protective immunity remain incompletely characterized. The Dendritic Cell (DC), was discovered in the 1970s by Ralph Steinman and Zanvil Cohn.1 As the major antigen presenting cells (APC) responsible for driving the generation of the adaptive immune response, DCs are central to establishing long-term protective immunity. Classically, DCs are dependent on the transcription factors ID22 and PU.1,3,4 as well as FMS-like Tyrosine Kinase 3 (Flt3)5,6 and Signal Transducer and Activator of Transcription 3 (STAT3)7 signaling. In vivo, DCs generate potent, cytotoxic CD8C T cells,8 as well as regulate the polarization of helper CD4C T cells toward the appropriate subtype (TH1, TH2, TH17, etc).9 Two specific populations of DC, which will not be discussed further but are mentioned here briefly, are plasmacytoid DCs and monocyte-derived DCs. Specialized for innate anti-viral immunity, plasmacytoid DCs express TLR7 and TLR9, and are the primary producers of type I IFNs in response to viral infections.10,11 DCs may also arise from monocytes in the setting of inflammation, taking on antigen presentation function to both CD4C and CD8C T cells,12 as well as an anti-microbial role through the production of TNFa and iNOS.13 Volume 12 Issue 1

In addition to plasmacytoid and monocyte derived DCs, there are several unique populations of DCs,14 which can be broadly grouped into 2 categories based on their anatomical location in the steadystate: lymphoid resident DCs ( also know as classical DC), or migratory DCs (migDC). Lymphoid resident DCs are found in the spleen and LNs of both mice and humans, originate from blood precursors, and include CD8aC and CD11bC subsets.15 In contrast, migDCs are comprised of several different populations (as discussed in detail16), which constitutively traffic to the local draining LN from a non-lymphoid organ, such as the skin, lungs, or gut, while being notably absent from the spleen.17 MigDC in the skin are perhaps the best characterized of these tissue DC populations, though their role in the generation of protective immunity is not fully understood. The skin is a unique immune environment, hosting both a diverse network of immune cells,18 as well as a significant microbiome made up of both fungal19 and bacterial20 populations. Furthermore, when directly compared to other routes of vaccine administration, such as intraperitoneal or intramuscular, cutaneous immunization offers superior cellular and humoral immune responses to both influenza21 and vaccinia,22 suggesting that the skin might be a specialized site for immune priming. As one such example, intradermal influenza is required at just one fifth of the dose needed via intramuscular injection, to achieve comparable vaccine titers and serum conversion.21 However the cellular mechanisms underlying enhanced immunization via the skin are incompletely understood and afford a key opportunity to improve rational vaccine design. In addition to hosting numerous commensal species, the skin is a site of high cellular turnover, between hair follicles and regular keratinocyte replacement. These features contribute to the unique nature of the skin, and play a significant role in the regulation of the immune system, as it must discriminate between normal cellular turnover and infection induced cell death. Hair follicles are considered to be an immune privileged site,23 yet when mechanically stimulated,

www.tandfonline.com

produce CCL2 and CCL20, recruiting Langerhans Cell precursors and allowing the entry of immune cells into the epidermis.24 However, despite high cellular turnover, healthy skin is not constantly inflamed, as the cell death of keratinocytes is closely regulated. Indeed, in mice with keratinocytes deficient in either FADD25 or caspase-8,26 skin lesions and constitutive inflammation develop. The skin must maintain tolerance to self, as well as to commensal bacteria, while responding appropriately to dangerous pathogens. This delicate balance is maintained through a specialized network of dendritic cells. In the skin of both humans and mice, there are a number of individual DC populations (4 or 5 respectively) as defined by both extracellular markers27 as well as dependence upon specific transcription factors.14,28-31 Langerhans Cells (LCs) are the only DC located in the epidermis, where they maintain surveillance of tight junctions,32-34 while binding broadly with keratinocytes through E-cadherin binding.35 In the dermis, an interstitial network of DCs is comprised of at least 3 individual populations, in addition to LC migrating through the dermis en route to the skin draining LN.31,36-39 These populations include CD103C DC (BDCA3C in humans), CD11bC DC (BDCA1C in humans), and CD11b¡ Langerin¡ DC (no human counterpart identified as of yet, although a BDCA3CBDCA1C human DC subset exists which has yet to be correlated to a mouse counterpart). Together with LCs, the dermal skin DCs form a complex system responsible for maintaining the equilibrium between barrier immunity and self-tolerance. Unique Properties of Skin DC Skin DCs survey the skin for pathogens and are capable of migrating to skin draining LNs bearing antigens to present to T cells (Fig. 1). This migration is CCR7 dependent, as mice genetically deficient for CCR7 mice do not have any migDCs in their cutaneous LNs.17 Unexpectedly, skin DC constitutively migrate in both germ free and MyD88/TRIF knockout mice,40 but fail to migrate when NF-kB signaling is inhibited in DCs,41 suggesting that the migratory program

Human Vaccines & Immunotherapeutics

occurs independently of TLR signaling, but requires NF-kB. However, irritant induced inflammation,42 TLR ligation,43 and infection44 result in increased trafficking, demonstrating that skin DC migration is responsive to immune stimuli. In addition, recent studies have demonstrated that skin DC will also increase their migration in response to low molecular weight hyaluronan through a TLR4 dependent mechanism, suggesting that skin DC may respond to a wide variety of stimuli.45 We and others have observed that skin DCs (in the steady state) are transcriptionally far more closely related to each other than to their LN resident counterparts.28,46,47 This finding was initially surprising, as some skin and LN populations share both common precursors, as well as a capacity for functionally similar properties, such as cross-presentation. Furthermore, even LCs are more closely related to the other skin DC populations,28 despite arising from a unique developmental pathway, more akin to monocytes and microglia than the remaining skin resident DCs.48,49 Moreover, skin DC in the steady-state and during Flt3L treatment maintain a transcriptional signature that is both mature as well as tolerogenic,28,46 while lymphoid resident DC tend to selectively express these markers during maturation, highlighting broad programming differences between these 2 groups. Phenotypically, skin (and other tissuederived) migDCs can be distinguished from LN resident DCs based on the expression level of classical DC identifiers; MHC II (or HLA-DR in human) and CD11c. While migDCs are identified as MHCIIhiCD11cint, LN resident DCs can be distinguished as MCHIIintCD11chi cells in the steady state. However, during an immune response to an infection, this distinction blurs- LN resident DCs upregulate MHC II and therefore resemble migDCs. Interestingly, Flt3L treatment can expand the numbers of both LN resident DCs, as well as migDCs in the skin and skin draining LN, with the notable exception of Langerhans cells.6,39 However, in contrast to LN resident DCs, migDCs constitutively express high levels of the classic DC activation markers

105

CD80 and CD86, despite having an immune dampening effect in some models, as will be discussed later. These findings, among others, have stimulated a discussion concerning the definition of a “mature” or even “semi mature” DC based on phenotypic markers, instead opting for functional studies about the licensing state of the DCs in question.50 Nevertheless, skin migDCs are a unique network of several DC populations with distinct functional and phenotypic characteristics.

Approaches to Studying Skin DC A number of different approaches have been utilized to Figure 1. Graphical Schematic of Skin DC Migration to the Cutaneous Lymph Nodes. Epidermal (Langerhans study the function of skin DC. Cells) and dermal skin DC colonize their respective layers of the skin. In both the inflamed and steady state Among these, preclinical models setting, LC and dermal DC enter the lymphatic vessels, before migrating in a CCR7 dependent manner to the allowing for the restriction or local cutaneous draining lymph node bearing either pathogen derived or self antigens. Upon arrival at the ablation of individual DC poplymph node, skin migratory DC enter the paracortical area and present skin derived antigens to T cells. Additionally, post migration, migratory DC can capture and present antigens that have drained freely, such as ulations have been indispensable DEC205 targeted vaccinations. Images were provided by Matthew Woodruff (Wikimedia commons). for understanding how these populations function both individually and as a network.51 These models drive the human diphthe- left intact from the chimerization.55 model may be used in selective contexts ria toxin receptor under a promoter of Another method to selectively delete (such as excluding a role for CCR7 depenspecific marker genes, such as Lan- LCs is the use of Langerin-DTA model, dence when immune responses are not gerin,52,53 CD11c,8 or Zbtb46.54 Each where diphtheria toxin is driven under lost), but cannot pinpoint a role for of these models results in the ablation the human Langerin promoter, resulting migDC exclusive of other CCR7-depenof a unique set of cells, sometimes with in constitutive deletion of LCs.56 How- dent populations. Thus it can be very overlapping results (Table 1). For ever, only the LCs, both in the epider- useful for broad definitions, but is subopexample, use of the CD11c-DTR model mis and migrating through the dermis, timal for more detailed study of migDC results in ablation of classical DCs pref- are affected in this model, leaving the function.57,58 erentially but also ablates some macro- other dermal resident LangerinC In addition to diphtheria toxin driven phages, monocytes, and pDC,8 while CD103C DCs intact.55 ablation strategies, several models have the Zbtb46 DTR model ablates only While several models of inducible abla- been established where DC subsets fail to classical Flt3 dependent DC in both tion have been established, there is no develop due to the loss of requisite tranlymphoid and non-lymphoid organs.54 known model that can specifically ablate scription factors. Batf3 knockout mice are The use of a Langerin-DTR can deplete all skin migDCs while leaving cDCs deficient in LN resident CD8aC DCs as both LCs and a dermal DC population intact. One model that has been used to well as CD103C dermal DC (discussed in that expresses both Langerin and study the effects of migDCs as a whole is detail below), while leaving other cell popCD103 (but is quite functionally and the CCR7 knockout mouse. However, ulations intact.59 In Batf3 deficient mice, developmentally distinct from LC).52,53 while migration of migDCs to the skin exogenous or inflammation derived IL-12 However, when bone marrow from draining LN is blocked in these mice results in the recovery of the CD8aC DC Langerin-DTR mice is used to reconsti- (thereby removing them from LN during subset, but not the CD103C DCs.60,61 tute a wildtype animal, only the dermal immune priming), CCR7 also plays a crit- Another model, CD11c-Cre IRF4 fl/fl, has LangerinC CD103C DC population is ical role in central memory T cells, T reg- been found to prevent the accumulation sensitive to the administration of diph- ulatory cells, and B cells, and its loss can of CD11bC dermal DC in the skin draintheria toxin, as LCs are radioresistant, alter LN architecture, while na€ıve and ing LN, despite their continued presence and host LCs (no DTR expression) are effector T cell are left largely intact. This in the skin.62

106

Human Vaccines & Immunotherapeutics

Volume 12 Issue 1

Table 1. Approaches to study skin DC Approach Acute Ablation

Experimental Model

Summary

CD11c-DTR

Deletion of classical DC, PDC, and some monocytes, macrophages Zbtb46-DTR Deletion of classical DC, demonstrated loss of CD103C, CD11bC, and CD11b¡, unclear for LC Langerin-DTR Deletion of LC and dermal CD103C DC. When used in bone marrow chimera models, only CD103C dermal DC are deleted. Constitutive Deficit Langerin-DTA Diptheria toxin expressed under the human langerin promoter, resulting in constitutive deletion of Langerhans Cells Batf3 ¡/¡ Selective loss of CD103C dermal DC and CD8aC LN resident DC CD11c-Cre IRF4 fl/fl Restriction of CD11bC dermal DC to the skin CCR7 ¡/¡ Restriction of migDC to the skin, additional defects in Treg, T central memory, and B cell localization Topical Application FITC Painting Dibutyl phthalate acetone sensitization of the skin along with application of FITC Dinitrochlorobenzene Sensitization Regular application of sensitizing agent models contact hypersensitivity Imiquimod/Resiquimod Regular application of TLR7/8 agonist models human psoriasis Tape Stripping Mechanical removal of the stratum corneum, stimulates LC migration Skin Trophic Infections Vaccinia Virus Virally derived antigens are presented to T cells by CD103C dermal DC Herpes Simplex Virus CD103C dermal DC are involved in transporting antigen to the draining LN Leishmania Leishmania derived antigens are presented by LN resident DC, while Langerhans Cells dampen the response Candida Albicans Langerhans Cells are required for the generation of a TH17 response, while CD103C dermal DCs generate a protective TH1 response. Staphlycoccus Aureus Derived Exfoliative Toxin Langerhans Cells are required for the production of IgG1 to neutralize exofoliative toxin Imaging CD11c-mCherry Expression of mCherry under the CD11c promoter Langerin-EGFP Expression of EGFP under the langerin promoter CD11c-EYFP Expression of YFP under the CD11c (ITGAX) promoter Isolation Protocols Skin and Skin Draining LN Digestion Digestion of skin using a combination of dispase, DNAse, and collagenase to isolate single cell suspensions Skin Crawlout Culture of whole skin explant, with or without the addition of external chemokines, allowing DC to leave the skin In vivo DC Targeting DEC205 Targeting Vaccine fusion protein targeted to both migDC and LN resident DC. CD8aC LN resident DC stimulate T cell immunity, while migDC inhibit it DEC207 (Langerin) Targeting Vaccine fusion protein targeted to CD103C Dermal DC, Langerhans Cells, and CD8aC LN resident DC DEC209 Vaccine fusion protein targeted to several DC like populations, including monocytes, LN resident DC, and plasmacytoid DC DNGR-1 (Clec9a) Vaccine fusion protein targeted to several DC like populations, including CD8a LN resident DC and CD103C dermal DC

The application of topical agents may help define the role of skin DC in inflammatory contexts. “FITC painting,” involves the application of the fluorescent molecule FITC along with an irritant, such as dibutyl phthalate acetone.63 After painting, migDC rapidly transport FITC from the skin to the skin draining LN. Similarly, application of DNCB results in contact hypersensitivity, though unlike the FITC model, inflammation from this model

www.tandfonline.com

seems to by driven by TH1 cells.64 Repeated application of TLR 7/8 agonists (Imiquimod or Resiqiumod) can model human psoriasis in mice, including increased keratinocyte proliferation, recruitment of multiple immune subsets, and dependence on increased IL-17 signaling.65 Mild injury and wounding is modeled through “tape stripping,” which removes part of the stratum corneum while leaving the hair follicle intact.66 This

Human Vaccines & Immunotherapeutics

References 8

54

52,53

55

59 62 17

63

64

65 66

74,75

71,72

77–79

67-70

34

80,81 52 141 29,42,83,84

39,82

46,86,90,91,93–95,142

87

88

89

mechanical disruption of the epidermis induces inflammation and increases migration of LCs to the skin draining LN, while increasing recruitment of new LC precursors to the affected area.66 This model has often been used in conjunction with the topical administration of a target antigen,34 such as ovalbumin, to study T cell responses to topical treatments. In addition to these topical treatments, a number of skin-tropic infections have been studied in

107

the context of skin DC, including Candida increased activation of CD4C and CD8C immune tolerance generated by the tumor. Albicans,67-70 HSV,71-73 vaccinia virus,74-76 T cells compared to untargeted protein Intriguingly, 75% of patients receiving Staphlycoccus,34 and Leishmania.77-79 As vaccines,86,90,91 and increased anti-tumor this vaccine who subsequently enrolled in these pathogens are predisposed to infect immunity in the preclinical setting of met- checkpoint blockade studies had objective the skin, skin DCs have a unique role in astatic melanoma.92 Recently, we demon- tumor regression, suggesting the remarkthe generation of the immune response to strated that combining DEC205-targeted able potential to combine this vaccine them, as will be discussed later on. antigen with adjuvant and Flt3L treat- with combinatorial immune checkpoint Imaging studies of the skin have shed ment (to expand DC) can further amplify blockade therapies in the future.98 light on the anatomical structure and loca- the resulting immune response.46 HowDEC205 targeting has been largely tion of skin DCs, highlighting their loca- ever, in the absence of an adjuvant to successful in the generation of adaptive tion both in the skin as well as in the LN mature or license DC, DEC205 targeting immunity in both preclinical as well as (Fig. 2A). In addition to conventional generates immune tolerance. In a murine clinical studies. However, we and others immunofluorescence, imaging studies model of multiple sclerosis (Experimental have observed high DEC205 expression may utilize mouse models where fluores- Autoimmune Encephalitis), DEC205 on CD8aC lymphoid resident and all subcent reporters are expressed under DC fused to Myelin Oligodendrocyte Gylco- sets of migDCs.39 As such, DEC205-tarspecific promoters, such as CD11c80 and protein (MOG) was able to prevent and geted antigens are directed to both populations.46 The identification of a Langerin,52 allowing for either live, in ameliorate the disease phenotype.93-95 vivo imaging or immunoflourescent stainClinically, DEC205 targeting has been series of individual DC subsets with ing. Reporter mice have been invaluable found to be safe and effective in generat- unique functional properties offers an in demonstrating the increased motion of ing cellular and humoral responses in opportunity to fine-tune this vaccination LCs following tape stripping,52 localiza- healthy volunteers.96 Additionally, the strategy by directing targeted vaccination tion of DCs at the tumor border,81 or results of a phase I trial using a DEC205 to a specific DC population based on the visualizing the interactions between T cells targeted NY-ESO-1 vaccine (given in desired cellular or humoral outcome. and DCs during the immune response.80 combination with resiquimod or Hiltonol) In addition to visualizing skin DC, spe- were recently reported.97 In this study, Skin Dendritic Cells in Immune cialized isolation protocols have been patients with NY-ESO-1 expressing developed to isolate skin DC, either by tumors saw an increase in both cellular Activation Skin based responses to cytolytic directly digesting the skin29 or skin and humoral immunity to the target antidraining LNs,42 or by allowing DC to gen following vaccination, despite the viral infection are highly dependent on “crawlout” in culture.39,82 Furthermore, enzymatic digestion allows for a physical separation of epidermis from dermis, in both murine83 and human84 skin, thereby isolating anatomically distinct cell populations prior to further processing. In addition to in vitro studies, a number of methods have been explored for targeting DCs in vivo, specifically utilizing antigens fused to DC specific antibodies,85 such as DEC205 (CD205),86 Langerin (CD207),87 DC-SIGN (CD209)88 or Clec9a (DNGR).89 Of these potential DC targets, DEC205 targeting is the most widely used and first established strategy. By fusing antigens to DEC205, the vaccine is targeted to DEC205C DCs and specifically delivered into the cross-presentaFigure 2. Migratory DC in the Skin and Cutaneous Draining Lymph Node. (A) Murine skin was stained for tion pathway of antigen processCD11c (purple), Langerin (green), CD11b (red), and DAPI (blue) in the steady-state. Images were taken at 20 £ and are courtesy of Sze-Wah Tse. (B) Proportions of individual migratory DC populations among the total ing- the major pathway by which migratory DC subset in the cutaneous draining lymph node in the steady state. Data are adapted from Henri viral and tumor antigens are preet al.106 and Mollah et al.39 sented. This approach facilitates

108

Human Vaccines & Immunotherapeutics

Volume 12 Issue 1

cross-presentation of vaccine antigens, suggesting that cell death of locally infected cells may be a licensing cue for optimal skin DC subset activation. In a series of elegant murine experiments, Shen et. al. added the human CMV gene product US11 to the vaccinia virus genome, generating a virus that prevents direct presentation by interfering with MHCI:peptide expression.99 In this model, CD8C T cell responses were severely impaired when vaccinia was administered by intravenous, intraperitonial, or intramuscular injection, indicating a role for direct presentation in these vaccination routes. In contrast, when vaccinia was administered by either intradermal injection or skin scarification, the resultant CD8C T cell response was unaffected by the loss of direct presentation, highlighting the unique importance of cross-presentation for these routes of vaccination. In agreement with these findings, the loss of DNGR-1 (a DC receptor that recognizes dead cells and mediates crosspresentation of scavenged antigens) results in significantly decreased CD8C T cell immunity to vaccinia virus (when given by skin scarification).75 This requirement for cross-presentation may also translate into superior immunity, as shown by Liu and Kupper et al., who found that skin based vaccinations with vaccinia were superior to other routes in terms of generating prophylactic protective immunity to either viral or tumor challenge through the generation of tissue resident memory T cells.22 However, within skin based vaccination routes, skin scarification is superior to intradermal vaccination in the generation of protection against either viral rechallenge22 or tumor growth,74 though the molecular and cellular mechanisms defining these differences in vaccine efficacy are not well understood. Yet, the presence of a unique combination of DC populations (Fig. 2B) suggests that the interaction between the skin and its resident DC network are of major significance for the generation of protective immunity.

Langerhans Cells In both the murine and human epidermis, Langerhans Cells (LCs) are the only

www.tandfonline.com

migDC population.14 Although long considered the prototype DC, LCs have unique characteristics that align them more closely with monocytes than other DC populations, including other skin DC. For example, during neonatal development, LC precursor cells are originally recruited to the skin in a CCR2 and CCR6 dependent manner24 from the yolk sac, but are then replaced by precursors from the fetal liver,84 while the other skin DC populations are derived from the bone marrow. LCs require TGFb, but not Flt3L signaling for their development, while LangerinC CD103C dermal DC are independent of TGFb, but, like other classical DC, are Flt3L dependent.39,100 Additionally, LCs demonstrate radioresistance when compared to dermal skin DC, which are sensitive to radiation.55 LCs are largely reconstituted from skin resident, rather than blood derived progenitors in the steady-state,14 relying on IL-34 signaling through the CSF-1 receptor.101 However, during inflammatory situations, both skin resident precursors102 as well as blood derived monocytes contribute to the repopulation of LCs in skin.103,104 Indeed, following infection with Herpes Simplex Virus, LCs are rapidly depleted from the infected area. Once the infection has been cleared, the subsequent LC repopulation of the previously infected skin is derived about half from monocyte derived precursors and half from long lived skin resident progenitor cells.73 One of the major functions of LCs is to monitor the integrity of the skin as a barrier tissue. In order to thoroughly survey this unique environment, LCs extend dendrites through keratinocyte tight junctions to monitor the external barrier of the skin,33,105 and respond to mechanical injury to the epidermis (in the form of tape stripping) by increasing motility and projection of dendrites.24,52 By extending dendrites through the tight junctions without compromising the epithelial barrier, LCs are able to generate immunity to antigens that have not breached the barrier yet, thereby generating protection without true infection. This was demonstrated in a model where patch immunization against the Staphylococcus aureus derived exfoliative toxin (ET) protected against blistering

Human Vaccines & Immunotherapeutics

during toxin rechallenge, despite the ET not penetrating the epithelial barrier.34 This is advantageous to the host because it allows the immune system to generate adaptive immunity prior to a true pathogenic challenge, similar to the ideology behind prophylactic vaccination. In the LN, LCs can be distinguished from dermal migDCs by their co-expression of Langerin and EpCam,83 while lacking expression of CD103.106 In the steady state, LCs in the LN express high levels of CD80, CD86, CD40, PDL1, and PDL2, though this expression can be further upregulated during exposure to a contact allergen, 2,4,6-trinitro-1-chlorobenzene (TNCB).42 In addition to these markers, LCs express high levels of IL15107 as well as the IL-15 receptor, IL15R-a, allowing them to present IL-15 to responding T cells. This co-stimulatory cytokine results in increased expansion of the T cell response through STAT5 signaling, even when presenting a normally tolerizing antigen, such as the tumor antigen Wilm’s Tumor-1.108 LCs may also be uniquely prone to tailoring the humoral response to antigens toward an IgG1 response, as the loss of LCs resulted specifically in the decrease of this antibody isotype in a gene gun vaccination model.83 In the setting of infectious disease, LCs have a demonstrated role in adaptive immunity to C. Albicans. In response to skin infection with this pathogen, LCs increase production of IL-1b and IL-6 while presenting pathogen derived antigens to CD4C T cells, resulting in TH17 differentiation.69,70 Interestingly, this production of IL-6 is dependent upon the engagement of a specific C type lectin receptor, Dectin-1, whose ligand is only expressed by the yeast form of C. Albicans, when it is located on the epidermis.67 Surprisingly, MyD88 signaling is not involved in LC migration in response to C. Albicans, and the generation of both the CD4C TH1 and CD8C T cell response is instead dependent on a separate, dermal resident DC population, CD103C migDCs, which will be discussed later. However, the TH17 cells generated by LC are important in generating protective immune memory in the skin, while TH1 cells provide systemic protection67 in this model, thereby highlighting the role of

109

LCs in maintaining and protecting the skin as a barrier tissue. Langerin¡CD11b¡ DCs In addition to the epidermal resident LC, several populations of migDCs coexist in the dermis of both mice and humans. The first of these populations was uncharacterized until very recently, and is identified as lacking either Langerin or CD11b expression.39,106 This population shares a common progenitor with other classical DC,106 is dependent upon FLT3 signaling, and requires the classical DC transcription factor ZBTB46 for development.39 We demonstrated that these cells are skin resident and actively transport FITC from the skin to the draining LN like other migratory DC subsets, present antigen with equal efficiency to other skin and LN DC subsets and, by hierarchy transcriptome analysis, we observed that Langerin¡ CD11b¡ DCs likely derive from a precursor (pre-DC) that also gives rise to Flt3L dependent CD11bC migDC, to which they are most closely related.39 These cells are equally capable of presenting antigens with an efficiency that parallels both other migDC populations and lymphoid resident DCs ex vivo.39 Quite recently this population has gained increasing recognition and was found to depend on KLF4 (Kruppel-like factor 4 transcription factor) by an independent group (though KLF4 loss impacted both this subset and CD11bC migratory DC). This study demonstrated a shared role for these populations in response to house dust mite allergen and Schistosoma mansoni infection.109 However, it is currently unknown if this population has a human counterpart, if phenotypic labeling of BDCA3C dermal DC in humans incorporates its counterpart, or if this population is related to in BDCA1CBDCA3C intermediary cells. Further research will explore the in vivo relevance of this population in vaccination and infection, and determine whether these cells have a unique function when compared to other dermal DCs, or functional redundancy with other skin DC subsets. This may also represent a subset that can be further specialized, differentiated, or licensed in an appropriate infectious context.

110

Langerin¡CD11bC DCs In addition to the CD11b¡ DCs, a separate, Langerin¡CD11bC population of DCs was identified in mice. The human counterpart of this subset is identified based on expression of BDCA-1.110,111 Up regulation of CCR7 and the subsequent migration of Langerin¡CD11bC DCs depends on IRF4 signaling, as tested by modeling contact hypersensitivity in mice lacking IRF4.112 In terms of antigen presentation, in vivo Langerin¡CD11bC DCs were observed to be uniquely capable of presenting soluble antigens, such as house dust mite antigen,113 ovalbumin,114-116 or part of the class II MHC Ea chain,117 to CD4C T cells. In the context of intramuscular vaccination with alum, CD11bC migDC were capable of cross-presentation of soluble OVA to CD8C T cells, though in these studies, CD64C monocyte derived DCs, were equally capable of presenting antigen.116 CD11bC migDC also have a role in CD4C T cell priming during infection. Langerin¡CD11bC DCs present antigen derived from Herpes Simplex Virus to CD4C T cells71 while also maintaining a role as the major cytokine producing population of skin DCs. In response to influenza vaccination, Langerin¡CD11bC DCs produced higher levels of the chemokines MIP1a, MIP1b, and RANTES than CD103C DCs, although the CD103C DCs (to be discussed next) were identified as key presenters of influenza derived antigens.118 In accordance with their role as both cytokine producers and antigen presenters, CD11bC migDC can also skew CD4C T cells toward a TH2 phenotype, either in the context of a TH2 inducing adjuvant (papain) or a TH2 driving infection (Nippostrongylus brasiliensis).62 Additionally, CD11bC migDC are required for the generation of a protective TH17 response to Aspergillus fumigatus challenge,119 demonstrating their highly versatile capacity to shape the resulting T cell response. LangerinCCD103C DCs This dermal migDC population is associated with a cross-presentation function and identified by co-expression of Langerin and the integrin CD103. These cells depend on the transcription

Human Vaccines & Immunotherapeutics

factor BATF3,120 as well as the transcriptional regulator IRF8.121 Although CD103C DC, like other DC, depend on Flt3L and are significantly reduced in numbers in the Flt3L knockout mouse, upon Flt3L treatment they do not expand as significantly as CD11bC and CD11b¡ dermal DC, suggesting that once they have differentiated from pre-DC in the tissue, they either lose Flt3L dependence or become dependent on or susceptible to pathways opposing Flt signaling.39 CD103C DCs have been ascribed a unique capacity for cross-presentation, as facilitated by high expression of the cell death receptor, DNGR-1 (CLEC9A).75 Indeed, C CD103 DCs play a key role in crosspresentation of antigens derived from self,106,122 as well as those derived from Herpes Simplex Virus-1122, influenza,123,124 vaccinia virus,74 and C. Albicans.70 Concordant with this specialized function, CD103C DCs have decreased endosomal acidification when compared to Langerin¡CD11bC DCs, allowing CD103C DCs to preserve the integrity of acquired antigens for a longer period of time.81,123 In the case of influenza, 2 studies have independently demonstrated that CD103C DCs are the primary APC responsible for the generation of the adaptive immune responses. However, in one study, CD103C DCs were not productively infected by the virus, and thus were utilizing cross-presentation,123 while in the other study, CD103C DC were found to be infected with the virus, and thereby directly presenting antigens.125 In both studies, type I IFN signaling was capable of preventing further viral infection, though further studies will be necessary to determine which presentation pathway (direct or cross) is the primary route of antigen presentation in this infection. Additionally, a recent study has also identified the potential of CD103C DC to skew the T cell response to Leishmania, despite being dispensable for antigen presentation.126 In humans, a similar population of DCs has been identified, based on expression of BDCA-3 or CD141.29 In agreement with the murine studies, human BDCA-3C DCs are uniquely

Volume 12 Issue 1

proficient in cross-presentation of antigens.29 In the setting of cancer, CD103C DCs were just reported to infiltrate tumors in 2, separate murine models; ectopically implanted B16 melanoma, as well as spontaneously arising mammary tumors driven by expression of the mouse mammary tumor virus promoter.81 In agreement with these findings, CD103C DC also infiltrated human metastatic melanoma. In this important study, the authors generated a signature associated with either CD103C tumor-infiltrating DCs or CD11bC tumor-infiltrating DCs, and found that a high CD103 to CD11b signature ratio correlated with a better prognosis in a clinical retrospective analysis of breast cancer, head and neck squamous cell carcinoma, or lung adenocarcinoma.81 However, it was not clear whether this stratification was driven more by the CD103C DC or by the CD11bC side of this ratio, as in human tumor analysis, PDL1C tolerizing interstitial myeloid cells (represented here within the denominator) correlate with response to immunotherapy.127,128 Also, while a mechanism for DC recruitment was not demonstrated in this study, tumors secrete high levels of CCL19 and CCL21 (the ligands for CCR7), which could potentially attract migratory DCs as well as other CCR7 dependent populations such as regulatory T cells (as discussed below).129 Also it is unclear as to whether intra-tumoral DCs were originally skinderived and infiltrated the tumor, or were recruited from the bone marrow derived pre-DC circulating through blood, which then entered the tumor and there differentiated into a tissue-resident DC. Many questions remain concerning the interactions between these DCs and tumors, but it is clear that CD103C DCs are critical for the generation of cytolytic CD8C T cell responses to a variety of pathogens, and their unique cross-presentation capability offers distinct potential in the presentation of tumor-derived antigens. Together, skin migDC form a network of sentinel cells, with each population playing a unique role in shaping the response to a variety of pathogens. Understanding the unique functional properties associated with individual DC subsets will

www.tandfonline.com

allow for further fine tuning of the immune response being generated, either by specifically targeting one population using antibody therapies as discussed previously or by understanding key pathways utilized by these individual DC populations during their licensing. However, in order to fully utilize the potential of the skin DC network, future studies are necessary to understand the coordination of these subsets, since, in vivo, each population plays a distinct role in shaping pathogen specific responses, as seen by the coordination of CD103C migDC and LC during infection with C. Albicans, or the compensatory roles of CD103C migDC and CD11bC migDC in response to influenza. While individual DC populations are capable of generating an adaptive immune response, the interactions between the members of the skin DC network tailors the response specifically toward an individual pathogen, and thereby shapes the resultant immunological memory. The power to fine tune the immune response by harnessing these DC interactions will be indispensable for future vaccines. Skin Resident Dendritic Cells in Immune Regulation As early as the late 1990s, it had been hypothesized that DCs played an active role in the generation and maintenance of self tolerance, and that migratory DC in particular may be especially well suited for this function.130 Indeed, DCs are capable of engulfing both apoptotic and necrotic cells, and generating MHC:peptide complexes from them.131 However, only exposure to necrotic cells results in increased expression of classical DC maturation markers, such as MHC II, CD40, and CD86, as well as triggers DCs potential to activate na€ıve T cells.132,133 MigDC are regularly exposed to normal cellular turnover (apoptotic), as mentioned above, and constitutively migrate to the cutaneous lymph node, making them an ideal candidate for DC induced tolerance to self antigens. Indeed, recent data as demonstrated presentation of murine skin-derived antigens in the non-inflamed state is restricted to migDCs, including both LCs and dermal DCs, and this presentation enforces deletional tolerance of self reactive T

Human Vaccines & Immunotherapeutics

cells.134 In the case of Leishmania infection, deletion of LCs results in a greater production of IFNg by CD4C T cells, as well as decreased parasite loads at the site of infection,77 though it is difficult to discern if this is due to LCs playing a tolerizing role, or due to the fact that Leishmania infects directly propogates in LCs.77 Additionally, murine LCs,95 CD103C DCs,95 and Langerin¡CD11bC DCs117,135 are all capable of generating CD4C FoxP3C Tregs, and a similar phenotype has been identified in human LCs.84 Furthermore, in some settings, murine CD4C T cells activated by LCs do not fully differentiate into cytokine producing effector cells or persist in vivo, despite initially dividing upon antigen encounter.136 In accordance with a tolerizing role in immunity, human BDCA-3C DCs, the counterpart to murine CD103C DCs, have been found to produce large amounts of IL-10, present self-antigens, and induce regulatory T cells.30 Recently, we found that murine migratory DC collectively and individually play a tolerogenic role during the setting of DEC205 protein vaccination.39 Surprisingly, this effect occurred despite the inclusion of an adjuvant targeting TLR4 in our vaccine design, (GLA).137 In these studies, LN resident DCs were responsible for the generation of adaptive immunity in response to vaccination, either by subcutaneous or intradermal administration. Despite enhanced antigen capture by migDC that had reached the LN (as compared to LN resident DC), immune responses were enhanced in CCR7 knockout animals, in which migDC could not traffic to the LN, demonstrating that migDCs were not required for productive immunity. By using a DTR model where LC and CD103C DCs (or just CD103C alone in bone marrow chimera) could be transiently ablated (Table 1), we observed deletion of individual migratory DC subsets also enhanced immunity. Loss of either LC and CD103C migDC or CD103C migDCs alone resulted in increased IFNg production by effector CD4C T cells, revealing that CD103C DCs can inhibit the immune response to a DEC205 targeted vaccination, despite the inclusion of adjuvant. In our model,

111

an increased CD4C T cell response also translated into increased vaccine specific IgG titers, further extending the immune regulatory function of CD103C DCs to the humoral arm of the adaptive immune system. Furthermore, upon examination of the transcriptional signature of migDCs as compared to LN resident DCs, we found that migDCs expressed high levels of a variety of transcripts associated with immune tolerance. Parallel findings were found when comparing murine lung migDC to LN resident DCs.28 Of translational importance, this tolerance signature was also present when we compared human peripheral blood DCs to skin resident DCs, suggesting that this program was conserved between mice and humans. We identified NF-kB signaling by ingenuity pathway analysis as central to this program. Another recent study has also identified a number of these tolerance associated genes when comparing migratory DC in the skin to those in the lymph node, suggesting that at least a large part of the program we identified may be amplified following migration to the cutaneous lymph node.41 Collectively, overlapping features of the tolerance signature and migration signature are programmed through NFkB signaling in DCs. DC specific loss of NF-kB signaling in mice resulted in loss of tolerance to self antigens in the steadystate, resulting in a lupus-like autoimmune phenotype marked by significantly increased lymph node size, as well as increased germinal centers and antinuclear antibody deposits in the kidney.41 In addition to these studies, one study using gene gun based vaccination also noted higher numbers of CD4C and CD8C IFNg producing cells upon ablation of dermal langerinC DC.83 However, despite having a regulatory role in the T cell response, CD103C dermal DC and LC had a coordinated stimulatory role in the development of the antibody response in this study. The antigen amount and delivery methods may underscore the differences between these 2 studies. By subcutaneous injection we observe antigen rapidly accumulating and captured in LN within 3 hours by both resident and already migrated DC, while a gene gun based approach results in

112

only very small doses of antigen over a longer period of time, which presumably must be actively transported by migratory DC for priming to occur. Some recent studies have also reported that migDC are not needed to generate adaptive immunity to viral vaccines or particulate antigens. In one study, using UV-inactivated influenza as a model vaccine, UV inactivated influenza was found to drain directly to the LN following footpad or intradermal ear injection.138 Additionally, removing the vaccinated ear 30 minutes after vaccination (thereby preventing new skin DC migration) had no affect on the generation of protective immunity against viral rechallenge. Another study used particulate antigen and found that a LN resident DC population (enriched in LN resident CD11bC DC) was positioned to filter particulate antigens that drain directly from the vaccine site. Again, in this model, migDC were unnecessary for generating a T cell response.139 In both these studies, as well as in our study, vaccine antigens were capable of draining freely to the local LN, without active transport from migDC. Therefore, in order to effectively target migDC, it may be important to prevent this type of antigen draining. These observations are important to note for future vaccine design as they suggest that targeting vaccinations to migDCs may not be ideal in all contexts, and that a unique set of licensing requirements exist for migDCs. Recent work has also begun to address the role of migDC in the generation of anti-tumor immunity, as discussed earlier. In a murine melanoma model, B16, it has been demonstrated that expression of CCL21 increases tumor growth, while a tumor line where CCL21 was genetically ablated resulted in decreased tumor growth in a T cell dependent manner.129 However, while the authors attributed this to increased infiltration of the tumor by regulatory T cells, skin migDC also express high levels of the receptor for CCL21, namely CCR7, which could be driving the increase in Treg formation. Indeed, when CCR7 signaling was decreased either through the use of a knockdown tumor cell line, or CCR7 knockout host mice, significantly less

Human Vaccines & Immunotherapeutics

CD11cC cells were found within the tumor, despite increased tumor specific CD8C T cell infiltration. Furthermore, both CD103C and CD11bC migrate into skin tumors in mice, and are capable of transporting antigen to the draining LN,81 though their status (stimulatory vs. tolerizing) following interaction with a neoplasm is just beginning to be addressed.81 How skin cancer affects migDC programming and how in turn these populations contribute to cancer surveillance and therapy is an important area of future research, and could be a contributor to immune priming vs. tolerance to tumors by the immune system.

Conclusions The skin is a unique microenvironment that interacts with pathogens and self-antigens regularly. In order to maintain the appropriate balance between activation to pathogens and tolerance to self, a network of migDCs surveys the skin and mediates the interactions between the immune system and foreign invaders. The individual characteristics of these populations make them attractive for targeted therapy, and have the potential to further amplify or tune a vaccine to the response desired depending upon the utilized DC populations. For example, targeting a protein vaccine to CD103C DCs could be more effective in generating a cytolytic T cells response when compared to broader DC targeting, while targeting Langerin¡CD11bC could lead to enhanced production of neutralizing antibodies. In addition, understanding how individual migDC populations coordinate responses to pathogens will allow for greater fine tuning of the response to vaccinations. However, as seen in the setting of DEC205 vaccinations, it will be important to fully understand the licensing cues that drive each of these subsets to activate adaptive immunity, so as to stimulate an optimal immune response. Despite the need to perform more human skin based immunology, additional pre-clinical models will be invaluable to add mechanistic insight toward

Volume 12 Issue 1

understanding the specific roles of individual migDC populations, as well as the function of migDCs as a whole. As a vaccination route, the skin is unique, depending predominantly on cross-presentation of antigens from infected and dying cells, instead of on direct presentation though infection of DCs. When utilized correctly, skin based vaccinations are capable of generating sterilizing and protective immunity, as seen by the overwhelming success of the smallpox vaccine. However, work in our lab and others has recently revealed that the DCs involved in generating this protection are also responsible for tightly regulating the immune system, likely to prevent horror-autotoxicus,140 and may be hampering the immune response in certain settings. Disclosure of Potential Conflicts of Interest

9.

10.

11.

12.

13.

14.

No potential conflicts of interest were disclosed. 15.

References 1. Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med 1973; 137:1142-62; PMID:4573839; http://dx.doi. org/10.1084/jem.137.5.1142 2. Hacker C, Kirsch RD, Ju XS, Hieronymus T, Gust TC, Kuhl C, Jorgas T, Kurz SM, Rose-John S, Yokota Y, et al. Transcriptional profiling identifies Id2 function in dendritic cell development. Nat Immunol 2003; 4:380-6; PMID:12598895; http://dx.doi.org/ 10.1038/ni903 3. Anderson KL, Perkin H, Surh CD, Venturini S, Maki RA, Torbett BE. Transcription factor PU.1 is necessary for development of thymic and myeloid progenitor-derived dendritic cells. J Immunol 2000; 164:1855-61; PMID:10657634; http://dx.doi.org/ 10.4049/jimmunol.164.4.1855 4. Guerriero A, Langmuir PB, Spain LM, Scott EW. PU.1 is required for myeloid-derived but not lymphoid-derived dendritic cells. Blood 2000; 95:879-85; PMID:10648399 5. McKenna HJ, Stocking KL, Miller RE, Brasel K, De Smedt T, Maraskovsky E, Maliszewski CR, Lynch DH, Smith J, Pulendran B, et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood 2000; 95:3489-97; PMID:10828034 6. Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K, McKenna HJ. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med 1996; 184:195362; PMID:8920882; http://dx.doi.org/10.1084/ jem.184.5.1953 7. Laouar Y, Welte T, Fu X-Y, Flavell RA. STAT3 is required for Flt3L-dependent dendritic cell differentiation. Immunity 2003; 19:903-12; PMID:14670306; http://dx.doi.org/10.1016/S1074-7613(03)00332-7 8. Jung S, Unutmaz D, Wong P, Sano G, De los Santos K, Sparwasser T, Wu S, Vuthoori S, Ko K, Zavala F,

www.tandfonline.com

16.

17.

18.

19.

20.

21.

22.

23.

et al. In vivo depletion of CD11cC dendritic cells abrogates priming of CD8C T cells by exogenous cellassociated antigens. Immunity 2002; 17:211-20; PMID:12196292; http://dx.doi.org/10.1016/S10747613(02)00365-5 Kapsenberg ML. Dendritic-cell control of pathogendriven T-cell polarization. Nat Rev Immunol 2003; 3:984-93; PMID:14647480; http://dx.doi.org/ 10.1038/nri1246 Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 2011; 29:163-83; PMID:21219184; http://dx.doi.org/10.1146/ annurev-immunol-031210-101345 Colonna M, Trinchieri G, Liu Y-J. Plasmacytoid dendritic cells in immunity. Nat Immunol 2004; 5:121926; PMID:15549123; http://dx.doi.org/10.1038/ ni1141 Domınguez PM, Ardavın C. Differentiation and function of mouse monocyte-derived dendritic cells in steady state and inflammation. Immunol Rev 2010; 234:90-104; PMID:20193014; http://dx.doi.org/ 10.1111/j.0105-2896.2009.00876.x Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA, Pamer EG. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003; 19:59-70; PMID:12871639; http://dx.doi.org/10.1016/S10747613(03)00171-7 Helft J, Ginhoux F, Bogunovic M, Merad M. Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice. Immunol Rev 2010; 234:5575; PMID:20193012; http://dx.doi.org/10.1111/ j.0105-2896.2009.00885.x Shortman K, Heath WR. The CD8C dendritic cell subset. Immunol Rev 2010; 234:18-31; PMID:20193009; http://dx.doi.org/10.1111/j.01052896.2009.00870.x Levin C, Perrin H, Combadiere B. Tailored immunity by skin antigen-presenting cells. Hum Vaccin Immunother 2015; 11:27-36; PMID:25483512; http://dx. doi.org/10.4161/hv.34299 Ohl L, Mohaupt M, Czeloth N, Hintzen G, Kiafard Z, Zwirner J, Blankenstein T, Henning G, F€orster R. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 2004; 21:279-88; PMID:15308107; http://dx.doi. org/10.1016/j.immuni.2004.06.014 Pasparakis M, Haase I, Nestle FO. Mechanisms regulating skin immunity and inflammation. Nat Rev Immunol 2014; 14:289-301; PMID:24722477 Findley K, Oh J, Yang J, Conlan S, Deming C, Meyer JA, Schoenfeld D, Nomicos E, Park M; NIH Intramural Sequencing Center Comparative Sequencing Program, et al. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013; 498:367-70; PMID:23698366; http://dx.doi.org/ 10.1038/nature12171 Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC; NISC Comparative Sequencing Program, Bouffard GG, Blakesley RW, Murray PR, Green ED, et al. Topographical and temporal diversity of the human skin microbiome. Science 2009; 324:1190-2; PMID:19478181; http://dx.doi.org/10.1126/science. 1171700 Kenney RT, Frech SA, Muenz LR, Villar CP, Glenn GM. Dose sparing with intradermal injection of influenza vaccine. N Engl J Med 2004; 351:2295-301; PMID:15525714; http://dx.doi.org/10.1056/ NEJMoa043540 Liu L, Zhong Q, Tian T, Dubin K, Athale SK, Kupper TS. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat Med 2010; 16:224-7; PMID:20081864; http://dx.doi.org/ 10.1038/nm.2078 Meyer KC, Klatte JE, Dinh HV, Harries MJ, Reithmayer K, Meyer W, Sinclair R, Paus R. Evidence that

Human Vaccines & Immunotherapeutics

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

the bulge region is a site of relative immune privilege in human hair follicles. Br J Dermatol 2008; 159:1077-85; PMID:18795933 Nagao K, Kobayashi T, Moro K, Ohyama M, Adachi T, Kitashima DY, Ueha S, Horiuchi K, Tanizaki H, Kabashima K, et al. Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin. Nat Immunol 2012; 13:74452; PMID:22729248; http://dx.doi.org/10.1038/ ni.2353 Bonnet MC, Preukschat D, Welz PS, van Loo G, Ermolaeva MA, Bloch W, Haase I, Pasparakis M. The adaptor protein FADD protects epidermal keratinocytes from necroptosis in vivo and prevents skin inflammation. Immunity 2011; 35:572-82; PMID:22000287; http://dx.doi.org/10.1016/j. immuni.2011.08.014 Kovalenko A, Kim JC, Kang TB, Rajput A, Bogdanov K, Dittrich-Breiholz O, Kracht M, Brenner O, Wallach D. Caspase-8 deficiency in epidermal keratinocytes triggers an inflammatory skin disease. J Exp Med 2009; 206:2161-77; PMID:19720838; http://dx.doi. org/10.1084/jem.20090616 Malissen B, Tamoutounour S, Henri S. The origins and functions of dendritic cells and macrophages in the skin. Nat Rev Immunol 2014; 14:417-28; PMID:24854591; http://dx.doi.org/10.1038/nri3683 Miller JC, Brown BD, Shay T, Gautier EL, Jojic V, Cohain A, Pandey G, Leboeuf M, Elpek KG, Helft J, et al. Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 2012; 13:888-99; PMID:22797772; http://dx.doi.org/10.1038/ni.2370 Haniffa M, Shin A, Bigley V, McGovern N, Teo P, See P, Wasan PS, Wang XN, Malinarich F, Malleret B, et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103C nonlymphoid dendritic cells. Immunity 2012; 37:60-73; PMID:22795876; http://dx.doi. org/10.1016/j.immuni.2012.04.012 Chu C-C, Ali N, Karagiannis P, Di Meglio P, Skowera A, Napolitano L, Barinaga G, Grys K, SharifPaghaleh E, Karagiannis SN, et al. Resident CD141 (BDCA3)C dendritic cells in human skin produce IL10 and induce regulatory T cells that suppress skin inflammation. J Exp Med 2012; 209:935-945; PMID:22547651; http://dx.doi.org/10.1084/jem. 20112583 Zaba LC, Fuentes-Duculan J, Steinman RM, Krueger JG, Lowes MA. Normal human dermis contains distinct populations of CD11c CBDCA-1C dendritic cells and CD163CFXIIIA C macrophages. J Clin Invest 2007; 117:2517-25; PMID:17786242; http:// dx.doi.org/10.1172/JCI32282 Yoshida K, Kubo A, Fujita H, Yokouchi M, Ishii K, Kawasaki H, Nomura T, Shimizu H, Kouyama K, Ebihara T, et al. Distinct behavior of human Langerhans cells and inflammatory dendritic epidermal cells at tight junctions in patients with atopic dermatitis. J Allergy Clin Immunol 2014; 134:856-64; PMID:25282566; http://dx.doi.org/10.1016/j.jaci. 2014.08.001 Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med 2009; 206:2937-46; PMID:19995951; http:// dx.doi.org/10.1084/jem.20091527 Ouchi T, Kubo A, Yokouchi M, Adachi T, Kobayashi T, Kitashima DY, Fujii H, Clausen BE, Koyasu S, Amagai M, et al. Langerhans cell antigen capture through tight junctions confers preemptive immunity in experimental staphylococcal scalded skin syndrome. J Exp Med 2011; 208:2607-13; PMID:22143886; http://dx.doi.org/10.1084/jem.20111718 Tang A, Amagai M, Granger LG, Stanley JR, Udey MC. Adhesion of epidermal Langerhans cells to keratinocytes mediated by E-cadherin. Nature 1993; 361:82-5; PMID:8421498; http://dx.doi.org/ 10.1038/361082a0

113

36. Henri S, Guilliams M, Poulin LF, Tamoutounour S, Ardouin L, Dalod M, Malissen B. Disentangling the complexity of the skin dendritic cell network. Immunol Cell Biol 2010; 88:366-75; PMID:20231850; http://dx.doi.org/10.1038/icb.2010.34 37. Haniffa M, Gunawan M, Jardine L. Human skin dendritic cells in health and disease. J Dermatol Sci 2015; 77:85-92; PMID:25301671; http://dx.doi.org/ 10.1016/j.jdermsci.2014.08.012 38. Nestle FO, Zheng XG, Thompson CB, Turka LA, Nickoloff BJ. Characterization of dermal dendritic cells obtained from normal human skin reveals phenotypic and functionally distinctive subsets. J Immunol 1993; 151:6535-45; PMID:7504023 39. Mollah SA, Dobrin JS, Feder RE, Tse SW, Matos IG, Cheong C, Steinman RM, Anandasabapathy N. Flt3L dependence helps define an uncharacterized subset of murine cutaneous dendritic cells. J Invest Dermatol 2014; 134:1265-75; PMID:24288007; http://dx.doi. org/10.1038/jid.2013.515 40. Wilson NS, Young LJ, Kupresanin F, Naik SH, Vremec D, Heath WR, Akira S, Shortman K, Boyle J, Maraskovsky E, et al. Normal proportion and expression of maturation markers in migratory dendritic cells in the absence of germs or Toll-like receptor signaling. Immunol Cell Biol 2008; 86:200-5; PMID:18026177; http://dx.doi. org/10.1038/sj.icb.7100125 41. Baratin M, Foray C, Demaria O, Habbeddine M, Pollet E, Maurizio J, Verthuy C, Davanture S, Azukizawa H, Flores-Langarica A, et al. Homeostatic NF-kB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance. Immunity 2015; 42:627-39; PMID:25862089; http://dx.doi. org/10.1016/j.immuni.2015.03.003 42. Stoitzner P, Tripp CH, Douillard P, Saeland S, Romani N. Migratory Langerhans cells in mouse lymph nodes in steady state and inflammation. J Invest Dermatol 2005; 125:116-25; PMID:15982311; http://dx.doi.org/10.1111/ j.0022-202X.2005.23757.x 43. Kastenm€ uller K, Wille-Reece U, Lindsay RW, Trager LR, Darrah PA, Flynn BJ, Becker MR, Udey MC, Clausen BE, Igyarto BZ, et al. Protective T cell immunity in mice following protein-TLR7/8 agonist-conjugate immunization requires aggregation, type I IFN, and multiple DC subsets. J Clin Invest 2011; 121:1782-96; PMID:21540549; http://dx.doi.org/ 10.1172/JCI45416 44. Guermonprez P, Helft J, Claser C, Deroubaix S, Karanje H, Gazumyan A, Darasse-Jeze G, Telerman SB, Breton G, Schreiber HA, et al. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nat Med 2013; 19:730-8; PMID:23685841; http://dx.doi.org/ 10.1038/nm.3197 45. Muto J, Morioka Y, Yamasaki K, Kim M, Garcia A, Carlin AF, Varki A, Gallo RL. Hyaluronan digestion controls DC migration from the skin. J Clin Invest 2014; 124:1309-19; PMID:24487587; http://dx.doi. org/10.1172/JCI67947 46. Anandasabapathy N, Feder R, Mollah S, Tse SW, Longhi MP, Mehandru S, Matos I, Cheong C, Ruane D, Brane L, et al. Classical Flt3L-dependent dendritic cells control immunity to protein vaccine. J Exp Med 2014; 211:1875-91; PMID:25135299; http://dx.doi. org/10.1084/jem.20131397 47. Harman AN, Bye CR, Nasr N, Sandgren KJ, Kim M, Mercier SK, Botting RA, Lewin SR, Cunningham AL, Cameron PU. Identification of lineage relationships and novel markers of blood and skin human dendritic cells. J Immunol 2013; 190:6679; PMID:23183897; http://dx.doi.org/10.4049/ jimmunol.1200779 48. Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, Leboeuf M, Low D, Oller G, Almeida F, et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med

114

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

2012; 209:1167-81; PMID:22565823; http://dx.doi. org/10.1084/jem.20120340 Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010; 330:841-5; PMID:20966214; http://dx.doi. org/10.1126/science.1194637 Heath WR, Villadangos JA. No driving without a license. Nat Immunol 2005; 6:125-6; PMID:15662437; http://dx.doi.org/10.1038/ni0205-125 Chow A, Brown BD, Merad M. Studying the mononuclear phagocyte system in the molecular age. Nat Rev Immunol 2011; 11:788-98; PMID:22025056; http://dx.doi.org/10.1038/nri3087 Kissenpfennig A, Henri S, Dubois B, Laplace-Builhe C, Perrin P, Romani N, Tripp CH, Douillard P, Leserman L, Kaiserlian D, et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 2005; 22:64354; PMID:15894281; http://dx.doi.org/10.1016/j. immuni.2005.04.004 Bennett CL, van Rijn E, Jung S, Inaba K, Steinman RM, Kapsenberg ML, Clausen BE. Inducible ablation of mouse Langerhans cells diminishes but fails to abrogate contact hypersensitivity. J Cell Biol 2005; 169:569-76; PMID:15897263; http://dx.doi.org/ 10.1083/jcb.200501071 Meredith MM, Liu K, Darrasse-Jeze G, Kamphorst AO, Schreiber HA, Guermonprez P, Idoyaga J, Cheong C, Yao KH, Niec RE, et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med 2012; 209:1153-65; PMID:22615130; http://dx.doi. org/10.1084/jem.20112675 Bursch LS, Wang L, Igyarto B, Kissenpfennig A, Malissen B, Kaplan DH, Hogquist KA. Identification of a novel population of LangerinC dendritic cells. J Exp Med 2007; 204:3147-56; PMID:18086865; http://dx.doi.org/10.1084/jem.20071966 Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, Shlomchik MJ. Epidermal Langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity 2005; 23:611-20; PMID:16356859; http://dx.doi.org/10.1016/j.immuni.2005.10.008 F€orster R, Davalos-Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity and tolerance. Nat Rev Immunol 2008; 8:362-71; PMID:18379575; http:// dx.doi.org/10.1038/nri2297 Schneider MA, Meingassner JG, Lipp M, Moore HD, Rot A. CCR7 is required for the in vivo function of CD4C CD25C regulatory T cells. J Exp Med 2007; 204:735-45; PMID:17371928; http://dx.doi.org/ 10.1084/jem.20061405 Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama M, Calderon B, Schraml BU, Unanue ER, Diamond MS, et al. Batf3 deficiency reveals a critical role for CD8alphaC dendritic cells in cytotoxic T cell immunity. Science 2008; 322:1097-100; PMID:19008445; http://dx.doi.org/ 10.1126/science.1164206 Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, Goldszmid RS, Sher A, Ploegh HL, Murphy TL, Sibley LD, et al. CD8a(C) dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 2011; 35:249-59; PMID:21867928; http://dx. doi.org/10.1016/j.immuni.2011.08.008 Tussiwand R, Lee WL, Murphy TL, Mashayekhi M, KC W, Albring JC, Satpathy AT, Rotondo JA, Edelson BT, Kretzer NM, et al. Compensatory dendritic cell development mediated by BATF-IRF interactions. Nature 2012; 490:502-7; PMID:22992524; http:// dx.doi.org/10.1038/nature11531 Gao Y, Nish SA, Jiang R, Hou L, Licona-Limon P, Weinstein JS, Zhao H, Medzhitov R. Control of T helper 2 responses by transcription factor IRF4-

Human Vaccines & Immunotherapeutics

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

dependent dendritic cells. Immunity 2013; 39:72232; PMID:24076050; http://dx.doi.org/10.1016/j. immuni.2013.08.028 Macatonia SE. Localization of antigen on lymph node dendritic cells after exposure to the contact sensitizer fluorescein isothiocyanate. Functional and morphological studies. J Exp Med 1987; 166:1654-67; PMID:3119761; http://dx.doi.org/10.1084/jem.166. 6.1654 Hayashi M, Higashi K, Kato H, Kaneko H. Assessment of preferential Th1 or Th2 induction by lowmolecular-weight compounds using a reverse transcription-polymerase chain reaction method: comparison of two mouse strains, C57BL/6 and BALB/c. Toxicol Appl Pharmacol 2001; 177:38-45; PMID:11708898; http://dx.doi.org/10.1006/taap. 2001.9286 Van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, Cornelissen F, Mus AM, Florencia E, Prens EP, et al. Imiquimod-induced psoriasislike skin inflammation in mice is mediated via the IL23/IL-17 axis. J Immunol 2009; 182:5836-45; PMID:19380832; http://dx.doi.org/10.4049/ jimmunol.0802999 Holzmann S, Tripp CH, Schmuth M, Janke K, Koch F, Saeland S, Stoitzner P, Romani N. A Model System Using Tape Stripping for Characterization of Langerhans Cell-Precursors In Vivo. J Invest Dermatol 2004; 122:1165-74; PMID:15140219; http://dx.doi.org/ 10.1111/j.0022-202X.2004.22520.x Kashem SW, Igyarto BZ, Gerami-Nejad M, Kumamoto Y, Mohammed J, Jarrett E, Drummond RA, Zurawski SM, Zurawski G, Berman J, et al. Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation. Immunity 2015; 42:356-66; PMID:25680275; http://dx.doi.org/ 10.1016/j.immuni.2015.01.008 Bonifazi P, Zelante T, D’Angelo C, De Luca A, Moretti S, Bozza S, Perruccio K, Iannitti RG, Giovannini G, Volpi C, et al. Balancing inflammation and tolerance in vivo through dendritic cells by the commensal Candida albicans. Mucosal Immunol 2009; 2:362-74; PMID:19421183; http://dx.doi.org/10.1038/ mi.2009.17 Haley K, Igyarto BZ, Ortner D, Bobr A, Kashem S, Schenten D, Kaplan DH. Langerhans cells require MyD88-dependent signals for Candida albicans response but not for contact hypersensitivity or migration. J Immunol 2012; 188:4334-9; PMID:22442445; http://dx.doi.org/10.4049/ jimmunol.1102759 Igyarto BZ, Haley K, Ortner D, Bobr A, GeramiNejad M, Edelson BT, Zurawski SM, Malissen B, Zurawski G, Berman J, et al. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity 2011; 35:260-72; PMID:21782478; http://dx.doi. org/10.1016/j.immuni.2011.06.005 Zhao X, Deak E, Soderberg K, Linehan M, Spezzano D, Zhu J, Knipe DM, Iwasaki A. Vaginal submucosal dendritic cells, but not Langerhans cells, induce protective Th1 responses to herpes simplex virus-2. J Exp Med 2003; 197:153-62; PMID:12538655; http://dx. doi.org/10.1084/jem.20021109 Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, Zhan Y, Lew AM, Shortman K, Heath WR, Carbone FR. Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 2006; 25:153-62; PMID: 16860764; http://dx.doi.org/10.1016/j. immuni.2006.04.017 Eidsmo L, Allan R, Caminschi I, van Rooijen N, Heath WR, Carbone FR. Differential migration of epidermal and dermal dendritic cells during skin infection. J Immunol 2009; 182:3165-72; PMID:19234214; http://dx.doi.org/10.4049/ jimmunol.0802950

Volume 12 Issue 1

74. Seneschal J, Jiang X, Kupper TS. LangerinC dermal DC, but not Langerhans cells, are required for effective CD8-mediated immune responses after skin scarification with vaccinia virus. J Invest Dermatol 2014; 134:686-94; PMID:24126845; http://dx.doi.org/ 10.1038/jid.2013.418 75. Iborra S, Izquierdo HM, Martınez-Lopez M, BlancoMenendez N, Reis e Sousa C, Sancho D. The DC receptor DNGR-1 mediates cross-priming of CTLs during vaccinia virus infection in mice. J Clin Invest 2012; 122:1628-43; PMID:22505455; http://dx.doi. org/10.1172/JCI60660 76. Frenz T, Waibler Z, Hofmann J, Hamdorf M, Lantermann M, Reizis B, Tovey MG, Aichele P, Sutter G, Kalinke U. Concomitant type I IFN receptor-triggering of T cells and of DC is required to promote maximal modified vaccinia virus Ankara-induced T-cell expansion. Eur J Immunol 2010; 40:2769-77; PMID:20821729; http://dx.doi.org/10.1002/ eji.201040453 77. Kautz-Neu K, Noordegraaf M, Dinges S, Bennett CL, John D, Clausen BE, von Stebut E. Langerhans cells are negative regulators of the anti-Leishmania response. J Exp Med 2011; 208:885-91; PMID:21536741; http:// dx.doi.org/10.1084/jem.20102318 78. Ritter U, Meissner A, Scheidig C, K€orner H. CD8 alpha- and Langerin-negative dendritic cells, but not Langerhans cells, act as principal antigen-presenting cells in leishmaniasis. Eur J Immunol 2004; 34:154250; PMID:15162423; http://dx.doi.org/10.1002/ eji.200324586 79. Iezzi G, Fr€ohlich A, Ernst B, Ampenberger F, Saeland S, Glaichenhaus N, Kopf M. Lymph node resident rather than skin-derived dendritic cells initiate specific T cell responses after Leishmania major infection. J Immunol 2006; 177:1250-6; PMID:16818784; http://dx.doi.org/10.4049/ jimmunol.177.2.1250 80. Khanna KM, Blair DA, Vella AT, McSorley SJ, Datta SK, Lefran¸c ois L. T cell and APC dynamics in situ control the outcome of vaccination. J Immunol 2010; 185:239-52; PMID:20530268; http://dx.doi.org/ 10.4049/jimmunol.0901047 81. Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, Barczak A, Rosenblum MD, Daud A, Barber DL, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014; 26:638-52; PMID:25446897; http://dx.doi. org/10.1016/j.ccell.2014.09.007 82. Stoitzner P, Pfaller K, St€ossel H, Romani N. A closeup view of migrating Langerhans cells in the skin. J Invest Dermatol 2002; 118, 117-25; PMID:11851884; http://dx.doi.org/10.1046/j.0022202x.2001.01631.x 83. Nagao K, Ginhoux F, Leitner WW, Motegi S, Bennett CL, Clausen BE, Merad M, Udey MC. Murine epidermal Langerhans cells and langerin-expressing dermal dendritic cells are unrelated and exhibit distinct functions. Proc Natl Acad Sci U S A 2009; 106:3312-7; PMID:19218433; http://dx.doi.org/ 10.1073/pnas.0807126106 84. Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper TS. Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity 2012; 36:87384; PMID:22560445; http://dx.doi.org/10.1016/j. immuni.2012.03.018 85. Trumpfheller C, Longhi MP, Caskey M, Idoyaga J, Bozzacco L, Keler T, Schlesinger SJ, Steinman RM. Dendritic cell-targeted protein vaccines: a novel approach to induce T-cell immunity. J Intern Med 2012; 271:183-92; PMID:22126373; http://dx.doi. org/10.1111/j.1365-2796.2011.02496.x 86. Bonifaz LC, Bonnyay DP, Charalambous A, Darguste DI, Fujii S, Soares H, Brimnes MK, Moltedo B, Moran TM, Steinman RM. In vivo targeting of antigens to maturing dendritic cells via the DEC-205

www.tandfonline.com

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

receptor improves T cell vaccination. J Exp Med 2004; 199:815-24; PMID:15024047; http://dx.doi. org/10.1084/jem.20032220 Idoyaga J, Cheong C, Suda K, Suda N, Kim JY, Lee H, Park CG, Steinman RM. Cutting edge: langerin/ CD207 receptor on dendritic cells mediates efficient antigen presentation on MHC I and II products in vivo. J Immunol 2008; 180:3647-50; PMID:18322168; http://dx.doi.org/10.4049/ jimmunol.180.6.3647 Tacken PJ, de Vries IJ, Gijzen K, Joosten B, Wu D, Rother RP, Faas SJ, Punt CJ, Torensma R, Adema GJ, et al. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood 2005; 106:1278-85; PMID:15878980; http://dx.doi. org/10.1182/blood-2005-01-0318 Sancho D, Mour~ao-Sa D, Joffre OP, Schulz O, Rogers NC, Pennington DJ, Carlyle JR, Reis e Sousa C. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J Clin Invest 2008; 118:2098-110; PMID:18497879; http://dx.doi.org/ 10.1172/JCI34584 Charalambous A, Oks M, Nchinda G, Yamazaki S, Steinman RM. Dendritic cell targeting of survivin protein in a xenogeneic form elicits strong CD4C T cell immunity to mouse survivin. J Immunol 2006; 177:8410-21; PMID:17142738; http://dx.doi.org/ 10.4049/jimmunol.177.12.8410 Cheong C, Choi JH, Vitale L, He LZ, Trumpfheller C, Bozzacco L, Do Y, Nchinda G, Park SH, Dandamudi DB, et al. Improved cellular and humoral immune responses in vivo following targeting of HIV Gag to dendritic cells within human anti-human DEC205 monoclonal antibody. Blood 2010; 116:3828-38; PMID:20668230; http://dx.doi.org/ 10.1182/blood-2010-06-288068 Mahnke K, Qian Y, Fondel S, Brueck J, Becker C, Enk AH. Targeting of antigens to activated dendritic cells in vivo cures metastatic melanoma in mice. Cancer Res 2005; 65:7007-12; PMID:16061687; http:// dx.doi.org/10.1158/0008-5472.CAN-05-0938 Ring S, Maas M, Nettelbeck DM, Enk AH, Mahnke K. Targeting of autoantigens to DEC205C dendritic cells in vivo suppresses experimental allergic encephalomyelitis in mice. J Immunol 2013; 191:2938-47; PMID:23945139; http://dx.doi.org/10.4049/ jimmunol.1202592 Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC. Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo. Immunity 2004; 20:695-705; PMID:15189735; http://dx.doi.org/10.1016/j.immuni.2004.05.002 Idoyaga J, Fiorese C, Zbytnuik L, Lubkin A, Miller J, Malissen B, Mucida D, Merad M, Steinman RM. Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Invest 2013; 123:844-54; PMID:23298832 Caskey M, Trumpfheller C, Pollak S, Sinnenberg L, Hurley A, Pring J, Shimeliovich I, Yipp B, Anandasabapathy N, Mehandru S, et al. In vivo targeting of HIV gag to dendritic cells in combination with poly ICLC is safe and immunogenic in healthy volunteers. Retrovirology 2012; 9:O51; http://dx.doi.org/10.1186/1742-4690-9-S2-O51 Dhodapkar MV, Sznol M, Zhao B, Wang D, Carvajal RD, Keohan ML, Chuang E, Sanborn RE, Lutzky J, Powderly J, et al. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci Transl Med 2014; 6:232ra51; PMID:24739759; http://dx.doi.org/ 10.1126/scitranslmed.3008068 Nirschl CJ, Drake CG. Molecular pathways: coexpression of immune checkpoint molecules: signaling pathways and implications for cancer immunotherapy. Clin Cancer Res 2013 19:4917-24; PMID:23868869; http://dx.doi.org/10.1158/10780432.CCR-12-1972

Human Vaccines & Immunotherapeutics

99. Shen X, Wong SBJ, Buck CB, Zhang J, Siliciano RF. Direct priming and cross-priming contribute differentially to the induction of CD8C CTL following exposure to vaccinia virus via different routes. J Immunol 2002; 169:4222-9; PMID:12370352; http://dx.doi. org/10.4049/jimmunol.169.8.4222 100. Zahner SP, Kel JM, Martina CA, Brouwers-Haspels I, van Roon MA, Clausen BE. Conditional deletion of TGF-bR1 using Langerin-Cre mice results in Langerhans cell deficiency and reduced contact hypersensitivity. J Immunol 2011; 187:5069-76; PMID:21998450; http://dx.doi.org/10.4049/ jimmunol.1101880 101. Greter M, Lelios I, Pelczar P, Hoeffel G, Price J, Leboeuf M, K€undig TM, Frei K, Ginhoux F, Merad M, et al. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity 2012; 37:1050-60; PMID:23177320; http://dx.doi.org/ 10.1016/j.immuni.2012.11.001 102. Chorro L, Sarde A, Li M, Woollard KJ, Chambon P, Malissen B, Kissenpfennig A, Barbaroux JB, Groves R, Geissmann F. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J Exp Med 2009; 206:3089-100; PMID:19995948; http://dx.doi.org/10.1084/ jem.20091586 103. Sere K, Baek JH, Ober-Bl€obaum J, M€uller-Newen G, Tacke F, Yokota Y, Zenke M, Hieronymus T. Two distinct types of langerhans cells populate the skin during steady state and inflammation. Immunity 2012; 37:905-16; PMID:23159228; http://dx.doi.org/ 10.1016/j.immuni.2012.07.019 104. Ginhoux F, Tacke F, Angeli V, Bogunovic M, Loubeau M, Dai XM, Stanley ER, Randolph GJ, Merad M. Langerhans cells arise from monocytes in vivo. Nat Immunol 2006; 7:265-73; PMID:16444257; http:// dx.doi.org/10.1038/ni1307 105. Nishibu A, Ward BR, Jester JV, Ploegh HL, Boes M, Takashima A. Behavioral responses of epidermal Langerhans cells in situ to local pathological stimuli. J Invest Dermatol 2006; 126:787-96; PMID:16439974; http:// dx.doi.org/10.1038/sj.jid.5700107 106. Henri S, Poulin LF, Tamoutounour S, Ardouin L, Guilliams M, de Bovis B, Devilard E, Viret C, Azukizawa H, Kissenpfennig A, et al. CD207C CD103C dermal dendritic cells cross-present keratinocytederived antigens irrespective of the presence of Langerhans cells. J Exp Med 2010; 207:189-206; PMID:20038600; http://dx.doi.org/10.1084/jem. 20091964 107. Banchereau J, Thompson-Snipes L, Zurawski S, Blanck JP, Cao Y, Clayton S, Gorvel JP, Zurawski G, Klechevsky E. The differential production of cytokines by human Langerhans cells and dermal CD14(C) DCs controls CTL priming. Blood 2012; 119:57429; PMID:22535664; http://dx.doi.org/10.1182/ blood-2011-08-371245 108. Romano E, Cotari JW, Barreira da Silva R, Betts BC, Chung DJ, Avogadri F, Fink MJ, St Angelo ET, Mehrara B, Heller G, et al. Human Langerhans cells use an IL-15R-a/IL-15/pSTAT5-dependent mechanism to break T-cell tolerance against the self-differentiation tumor antigen WT1. Blood 2012; 119:5182-90; PMID:22510877; http://dx.doi.org/10.1182/blood2011-09-382200 109. Tussiwand R, Everts B, Grajales-Reyes GE, Kretzer NM, Iwata A, Bagaitkar J, Wu X, Wong R, Anderson DA, Murphy TL, et al. Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 2015; 42:916-28; PMID:25992862; http://dx.doi.org/10.1016/j. immuni.2015.04.017 110. Klechevsky E. Human dendritic cells - stars in the skin. Eur J Immunol 2013; 43:3147-55; PMID:24222336; http://dx.doi.org/10.1002/ eji.201343790

115

111. Collin M, McGovern N, Haniffa M. Human dendritic cell subsets. Immunology 2013; 140:22-30; PMID:23621371; http://dx.doi.org/10.1111/ imm.12117 112. Baja~ na S, Roach K, Turner S, Paul J, Kovats S. IRF4 promotes cutaneous dendritic cell migration to lymph nodes during homeostasis and inflammation. J Immunol 2012; 189:3368-77; PMID:22933627; http://dx. doi.org/10.4049/jimmunol.1102613 113. Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W, Vanhoutte L, Neyt K, Killeen N, Malissen B, et al. Conventional and monocyte-derived CD11bC dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 2013; 38:322-35; PMID:23352232; http://dx.doi. org/10.1016/j.immuni.2012.10.016 114. Nizza ST, Campbell JJ. CD11bC migratory dendritic cells mediate CD8 T cell cross-priming and cutaneous imprinting after topical immunization. PLoS One 2014; 9; PMID:24618819; http://dx.doi.org/ 10.1371/journal.pone.0091054 115. Del Rio M-L, Rodriguez-Barbosa J-I, Kremmer E, F€orster R. CD103- and CD103C bronchial lymph node dendritic cells are specialized in presenting and cross-presenting innocuous antigen to CD4C and CD8C T cells. J Immunol 2007; 178:6861-6; PMID:17513734; http://dx.doi.org/10.4049/ jimmunol.178.11.6861 116. Langlet C, Tamoutounour S, Henri S, Luche H, Ardouin L, Gregoire C, Malissen B, Guilliams M. CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization. J Immunol 2012; 188:1751-60; PMID:22262658; http://dx.doi. org/10.4049/jimmunol.1102744 117. McLachlan JB, Catron DM, Moon JJ, Jenkins MK. Dendritic cell antigen presentation drives simultaneous cytokine production by effector and regulatory T cells in inflamed skin. Immunity 2009; 30:277-88; PMID:19200757; http://dx.doi.org/10.1016/j. immuni.2008.11.013 118. Beaty SR, Rose CE, Sung S-S. J. Diverse and potent chemokine production by lung CD11bhigh dendritic cells in homeostasis and in allergic lung inflammation. J Immunol 2007; 178:1882-95; PMID:17237439; http://dx.doi.org/10.4049/jimmunol.178.3.1882 119. Schlitzer A, McGovern N, Teo P, Zelante T, Atarashi K, Low D, Ho AW, See P, Shin A, Wasan PS, et al. IRF4 transcription factor-dependent CD11bC dendritic cells in human and mouse control mucosal IL17 cytokine responses. Immunity 2013; 38:970-83; PMID:23706669; http://dx.doi.org/10.1016/j. immuni.2013.04.011 120. Edelson BT, Bradstreet TR, KC W, Hildner K, Herzog JW, Sim J, Russell JH, Murphy TL, Unanue ER, Murphy KM. Batf3-dependent cd11b low/- peripheral dendritic cells are gm-csf-independent and are not required for th cell priming after subcutaneous immunization. PLoS One 2011; 6; PMID:22065991; http://dx.doi.org/10.1371/journal.pone.0025660 121. Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, Hashimoto D, Price J, Yin N, Bromberg J, Lira SA, et al. The origin and development of nonlymphoid tissue CD103C DCs. J Exp Med 2009; 206:3115-30; PMID:20008528; http://dx.doi.org/10.1084/ jem.20091756

116

122. Bedoui S, Whitney PG, Waithman J, Eidsmo L, Wakim L, Caminschi I, Allan RS, Wojtasiak M, Shortman K, Carbone FR, et al. Cross-presentation of viral and self antigens by skin-derived CD103C dendritic cells. Nat Immunol 2009; 10:488-95; PMID:19349986; http://dx.doi.org/10.1038/ ni.1724 123. Helft J, Manicassamy B, Guermonprez P, Hashimoto D, Silvin A, Agudo J, Brown BD, Schmolke M, Miller JC, Leboeuf M, et al. Cross-presenting CD103C dendritic cells are protected from influenza virus infection. J Clin Invest 2012; 122:4037-47; PMID:23041628; http://dx.doi.org/10.1172/JCI60659 124. GeurtsvanKessel CH, Willart MA, van Rijt LS, Muskens F, Kool M, Baas C, Thielemans K, Bennett C, Clausen BE, Hoogsteden HC, et al. Clearance of influenza virus from the lung depends on migratory langerinCCD11bbut not plasmacytoid dendritic cells. J Exp Med 2008; 205:1621-34; PMID:18591406; http://dx.doi.org/ 10.1084/jem.20071365 125. Moltedo B, Li W, Yount JS, Moran TM. Unique type I interferon responses determine the functional fate of migratory lung dendritic cells during influenza virus infection. PLoS Pathog 2011; 7:e1002345; PMID:22072965; http://dx.doi.org/10.1371/journal. ppat.1002345 126. Martınez-Lopez M, Iborra S, Conde-Garrosa R, Sancho D. Batf3-dependent CD103C dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur J Immunol 2015; 45:119-29; PMID:25312824; http:// dx.doi.org/10.1002/eji.201444651 127. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders RA. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti-PD-1 Therapy. Clin Cancer Res 2014; 20:5064-74. doi:10.1158/1078-0432.CCR-133271; PMID:24714771; http://dx.doi.org/10.1158/ 1078-0432.CCR-13-3271 128. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014; 515:563-7; PMID:25428504; http://dx.doi.org/10.1038/nature14011 129. Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science (80) 2010; 328:749-52; PMID:20339029; http://dx.doi.org/10.1126/science.1185837 130. Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 2000; 191:411-6; PMID:10662786; http://dx.doi.org/10.1084/jem. 191.3.411 131. Inaba K, Turley S, Yamaide F, Iyoda T, Mahnke K, Inaba M, Pack M, Subklewe M, Sauter B, Sheff D, et al. Efficient presentation of phagocytosed cellular fragments on the major histocompatibility complex class II products of dendritic cells. J Exp Med 1998; 188:2163-73; PMID:9841929; http://dx.doi.org/ 10.1084/jem.188.11.2163 132. Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: expo-

Human Vaccines & Immunotherapeutics

133.

134.

135.

136.

137.

138.

139.

140.

141.

142.

sure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med 2000; 191:423-34; PMID:10662788; http://dx.doi.org/ 10.1084/jem.191.3.423 Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med 1999; 5:1249-55; PMID:10545990; http://dx. doi.org/10.1038/15200 Waithman J, Allan RS, Kosaka H, Azukizawa H, Shortman K, Lutz MB, Heath WR, Carbone FR, Belz GT. Skin-derived dendritic cells can mediate deletional tolerance of class I-restricted self-reactive T cells. J Immunol 2007; 179:4535-41; PMID:17878350; http://dx.doi.org/10.4049/jimmunol.179.7.4535 Guilliams M, Crozat K, Henri S, Tamoutounour S, Grenot P, Devilard E, de Bovis B, Alexopoulou L, Dalod M, Malissen B. Skin-draining lymph nodes contain dermis-derived CD103(¡) dendritic cells that constitutively produce retinoic acid and induce Foxp3 (C) regulatory T cells. Blood 2010; 115:1958-68; PMID:20068222; http://dx.doi.org/10.1182/blood2009-09-245274 Shklovskaya E, O’Sullivan BJ, Ng LG, Roediger B, Thomas R, Weninger W, Fazekas de St Groth B. Langerhans cells are precommitted to immune tolerance induction. Proc Natl Acad Sci U S A 2011; 108:18049-54; PMID:22006331; http://dx.doi.org/ 10.1073/pnas.1110076108 Pantel A, Cheong C, Dandamudi D, Shrestha E, Mehandru S, Brane L, Ruane D, Teixeira A, Bozzacco L, Steinman RM, et al. A new synthetic TLR4 agonist, GLA, allows dendritic cells targeted with antigen to elicit Th1 T-cell immunity in vivo. Eur J Immunol 2012; 42:101-9; PMID:22002164; http://dx.doi.org/ 10.1002/eji.201141855 Woodruff MC, Heesters BA, Herndon CN, Groom JR, Thomas PG, Luster AD, Turley SJ, Carroll MC. Trans-nodal migration of resident dendritic cells into medullary interfollicular regions initiates immunity to influenza vaccine. J Exp Med 2014; 211:1611-21; PMID:25049334; http://dx.doi.org/10.1084/jem. 20132327 Gerner MY, Torabi-Parizi P, Germain RN. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 2014; 42:172-85; PMID:25607462; http://dx. doi.org/10.1016/j.immuni.2014.12.024 Steinman RM, Nussenzweig MC. Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci U S A 2002; 99:351-8; PMID:11773639; http://dx.doi.org/ 10.1073/pnas.231606698 Caton ML, Smith-Raska MR, Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J Exp Med 2007; 204:1653-64; PMID:17591855 Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, Steinman RM. Efficient targeting of protein antigen to the dendritic cell receptor DEC205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8C T cell tolerance. J Exp Med 2002; 196:1627-38; PMID:12486105; http://dx.doi. org/10.1084/jem.20021598

Volume 12 Issue 1

Duality at the gate: Skin dendritic cells as mediators of vaccine immunity and tolerance.

Since Edward Jenner's discovery that intentional exposure to cowpox could provide lifelong protection from smallpox, vaccinations have been a major fo...
377KB Sizes 5 Downloads 9 Views