Natural and Induced Tolerogenic Dendritic Cells Courtney A. Iberg and Daniel Hawiger This information is current as J Immunol 2020; 204:733-744; ; of September 28, 2021. doi: 10.4049/jimmunol.1901121 http://www.jimmunol.org/content/204/4/733 Downloaded from References This article cites 203 articles, 76 of which you can access for free at: http://www.jimmunol.org/content/204/4/733.full#ref-list-1

Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

*average by guest on September 28, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2020 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Natural and Induced Tolerogenic Dendritic Cells Courtney A. Iberg and Daniel Hawiger Dendritic cells (DCs) are highly susceptible to extrinsic increasing the risk of autoimmune responses against self- signals that modify the functions of these crucial APCs. antigens (2, 4–7). Maturation of DCs induced by diverse proinflamma- The priming of self-reactive peripheral T cells is controlled tory conditions promotes immune responses, but cer- by tTreg cells (8). However, the functions of tTreg cells tain signals also induce tolerogenic functions in DCs. may be overwhelmed by specific proinflammatory auto- These “induced tolerogenic DCs” help to moderate immune activation; also, in some individuals, the devel- immune responses such as those to commensals present opment of tTreg cells may be compromised (2, 4, 9, 10). at specific anatomical locations. However, also under Various animal models of autoimmune diseases initiated in healthy animals after immunization with specific self-antigens steady-state conditions, some DCs are characterized Downloaded from by inherent tolerogenic properties. The immunomod- either in the presence of adjuvants or in the context of an ulatory mechanisms constitutively present in such introduced infectious agent have demonstrated that specific priming of preexisting self-reactive T cells mediates an auto- “natural tolerogenic DCs” help to promote tolerance immune process (2, 4–7). Therefore, pathways of thymic to peripheral Ags. By extending tolerance initially tolerance need to be extended by the specific mechanisms established in the thymus, these functions of DCs operating in the peripheral immune system. Particularly, au- help to regulate autoimmune and other immune re- toimmune responses can be ameliorated or even completely http://www.jimmunol.org/ sponses. In this review we will discuss the mechanisms prevented by the Ag-specific, peripherally formed Treg (pTreg) and functions of natural and induced tolerogenic DCs cells that are induced extrathymically by DCs (7, 11). and offer further insight into how their possible ma- nipulations may ultimately lead to more precise treat- Dendritic cells in peripheral tolerance ments for various immune-mediated conditions and diseases. The Journal of Immunology, 2020, 204: The roles of DCs in peripheral tolerance have been established 733–744. by multiple lines of independent experimental evidence. The

disturbance of tolerance and immune homeostasis caused by by guest on September 28, 2021 the absence of DCs and their subsets was observed in various endritic cells (DCs) are APCs critical for the initi- experimental systems that relied on a specific in vivo killing of ation of T cell immune responses and for the gov- DCs expressing diphtheria toxin receptor or diphtheria toxoid D erning of immune tolerance. Together with other A subunit or by other means, such as a chemical depletion of types of cells in the thymus, DCs have important roles in DCs (12–16). These results are in agreement with other early mediating central tolerance, combining thymic deletion studies that identified the roles of DCs in the induction of of self-reactive T cells and a production of thymically de- peripheral tolerance by employing methods of specific deliv- rived regulatory T (tTreg) cells in a cumulative process aimed ery of defined Ags to DCs in vivo, tracking the uptake of at preventing overt anti-self responses (1). However, because to DCs, and the transgenic expression of ectopic Ags of differing efficiencies of antigenic presentation in the as cytosolic proteins in DCs (17–20). The combination of thymus and the periphery, and cross-reactivity of TCRs, specific Ag-targeting methods with various genetic models the mature peripheral T cell repertoire still contains T cells of DCs has allowed for further advances in our understanding that may be reactive to self (2, 3). These self-reactive pe- of the importance of DCs in governing peripheral tolerance, ripheral T cells can then be primed in the periphery, even as we also recently discussed in Ref. 11. by low-affinity peptides that are below their original thresh- The specific functions of DCs depend, in part, on the de- olds for negative selection in the thymus (2, 4), ultimately velopmentally determined diversity of DC subsets reviewed

Department of Molecular Microbiology and Immunology, Saint Louis University conventional DC; DC, dendritic cell; DC-SIGN, DC-specific ICAM-3–grabbing School of Medicine, St. Louis, MO 63104 nonintegrin; EAE, experimental autoimmune encephalomyelitis; HVEM, herpes- virus entry mediator; itDC, induced tolerogenic DC; LN, lymph node; MHCII, Received for publication September 16, 2019. Accepted for publication October 4, MHC class II; moDC, monocyte-derived DC; MS, multiple sclerosis; mTOR, 2019. mammalian target of rapamycin; ntDC, natural tolerogenic DC; PD-1, pro- This work was supported in part by National Institute of Allergy and Infectious Diseases/ grammed death-1; pDC, plasmacytoid DC; PD-L1, programmed death -1; National Institutes of Health Grant R01AI113903 (to D.H.). PRR, pattern recognition receptor; pTreg,peripherallyformedTreg;RA,retinoic acid;Treg,regulatoryT;tTreg,thymicallyderivedregulatoryT;VDR,vitaminD Address correspondence and reprint requests to Dr. Daniel Hawiger, Department of receptor. Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Doisy Research Center, 1100 South Grand Boulevard, Room 715, St. Louis, MO Ó 63104. E-mail address: [email protected] Copyright 2020 by The American Association of Immunologists, Inc. 0022-1767/20/$37.50 Abbreviations used in this article: AHR, aryl hydrocarbon receptor; BMDC, bone marrow–derived DC; BTLA, B and T lymphocyte associated; cDC, www.jimmunol.org/cgi/doi/10.4049/jimmunol.1901121 734 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS extensively by Murphy and colleagues (21) and Merad et al. promote in T cells various mechanisms of tolerance, including (22). Both human and murine DCs consist of two main T cell anergy, T cell deletion, and a conversion of pTreg cells populations, conventional DC (cDC) and plasmacytoid DC (11). We therefore propose to refer to such DCs as “natural (pDC), both of which develop from progenitors in the bone tolerogenic DCs” (ntDCs) (Fig. 1). marrow and then differentiate into various subsets present throughout multiple tissues (23, 24). The cDC population Establishing peripheral tolerance by natural tolerogenic functions can be further divided into the DC1 and DC2 subsets, as of DCs defined by the transcription factors required for their devel- The physiological steady state can be defined by the undis- opment. The DC1 subset, which requires the transcription turbed expression of and other molecules contrib- factors Irf8, Id2, and Batf3 for development, is distinguished uting to the baseline conditions that, together with stromal by the expression of XCR1 and further characterized by the cells of the secondary lymphoid tissues, provide a framework expression of additional cell surface molecules, including B for the interactions between DCs and T cells (35, 36). Initially, and T lymphocyte associated (BTLA), CD8a, and DEC-205. it was postulated that, in the steady state, DCs remain im- In contrast, the development of DC2s is governed by the mature akin to BMDCs or moDCs that are characterized by transcription factors Irf4 and Notch2, and these DCs are lower expression of MHC and costimulatory molecules when distinguished by cell surface expression of CD172a (SIRPa) cultured in the absence of maturation signals (35). However, as well as DCIR2 and CD11b (11, 23, 24). Although not a the available experimental evidence has clearly shown that, main focus of this review, pDCs, characterized by the ex- even in the steady state, DCs can constitutively initiate ac- Downloaded from pression of cell surface molecules (including B220, bone tive mechanisms of tolerance in T cells, such as the conversion marrow stromal Ag 2 [BST2], and Siglec-H), are primarily of pTreg cells (7, 11). These general contradictions were involved in antiviral responses but also have some roles in recognized early by Lutz and Schuler, who proposed that + + + tolerance (11, 25). Importantly, CD141 (BDCA-3 ) XCR1 the division between “immature DCs” and “mature DCs” + + + + BTLA human DC1s and CD1c CD172a CD11b human (as defined by DC phenotypes) did not necessarily correspond DC2s share many developmental, phenotypical, and func- with “tolerogenic DCs” and “immunogenic DCs,” respec- http://www.jimmunol.org/ tional similarities with their murine counterparts (21, 26). tively. Instead, divisive tolerogenic and immunogenic matu- In addition to their roles in tolerance, DCs have crucial ration processes were proposed (31), further supported by the functions in the initiation of immune responses. The efficient identification of transcriptional determinants of certain tol- priming of immune responses by specific DC subsets requires erogenic and immunogenic maturation states in DCs as well additional signals from the proinflammatory environment that as by the emerging concept of “homeostatic maturation” of can be sensed through specific pattern recognition receptors DCs under steady-state conditions (37–40). For example, a (PRRs) (27–29). These signals lead to the DC acquisition decrease in E-cadherin–mediated cell–to–cell contact results of enhanced properties to induce immune responses in a pro- in specific increases in expression of MHC class II (MHCII) by guest on September 28, 2021 cess referred to as maturation. Overall, the model of such and costimulatory molecules (39). The concept of DC mat- proimmunogenic DC maturation postulates increased proin- uration in the steady state was additionally defined by other flammatory production and increased cell surface observations of multiple specific expression changes expression of costimulatory and MHC molecules and che- comparable, in scope, to those observed under TLR agonist– mokine ligands or receptors in response to microbial and mediated maturation (30). This process of maturation under other proinflammatory stimulation (28, 30). In addition to homeostatic conditions has also been proposed to result in this maturation process resulting in increased immune re- functions of DCs necessary to induce active mechanisms of sponses, specific extrinsic signals were also proposed to induce tolerance (38). Although the specific mechanisms governing tolerogenic differentiation of DCs. The experiments using DC functions in vivo under steady-state conditions following bone marrow–derived DCs (BMDCs), monocyte-derived their initial development from the bone marrow precursors are DCs (moDCs), and DCs obtained ex vivo showed that still being uncovered, it is clear that these processes are con- some PRR agonists, as well as various other physiological and tinuous, and some possibly cell autonomous, resulting in the pharmaceutical agents, can allow for the induction of DCs stable numbers and phenotypes of DCs expressing crucial with tolerogenic functions. Further, the experiments in vivo molecules involved in tolerance (Fig. 2 and as discussed below). revealed that, in response to certain signals in specific ana- In the steady state, DCs can induce multiple mechanisms of tomical sites (including the intestines and airways), some tolerance in T cells including anergy, but a de novo conversion DCs help to maintain immune homeostasis toward com- of pTreg cells bestows a dominant and long-lasting tolerance to mensal organisms and other Ags, even under partially peripheral Ags (7, 11, 41–44). Although in the steady state proimmunogenic conditions (28, 31–33). We propose to Ags initially acquired by all cDCs can induce tolerance, DC1s refer to such DCs that acquire tolerogenic properties ei- are more prone to induce tolerogenic effects as compared with ther in vitro or in vivo as “induced tolerogenic DCs” (itDCs), DC2s (7, 11, 42). The specialization among DCs can be at- as partially based on the terminology first introduced by tributed to different localization of DCs within a local ar- Maldonado and von Andrian (34). chitecture of immune organs, differences in the efficiencies However, even in the absence of specific extrinsic signals, of processing and presentation of Ags to T cells, and the generally referred to as steady-state conditions, many DCs that specific immunomodulatory mechanisms in DC1s and DC2s are present in the spleen and other lymphoid organs do not (11, 45–48). The immunomodulatory pathways are of par- necessarily remain as “immature” immunological bystanders ticular importance in the mediation of a tolerogenic part- but instead have important roles in initiating and maintaining nership of DCs and T cells (49). Importantly, the engagement tolerance to available peripheral Ags (11). These DCs inherently of immunomodulatory axes (including programmed death The Journal of Immunology 735

FIGURE 1. The role of ntDCs and itDCs in extrathymic tolerance. Peripheral tolerance extends tolerance initiated centrally in thymus. The ntDCs and itDCs function to promote the deletion or anergy of self-reactive T cells as well as to induce pTreg cells. ntDCs are generally tissue-resident DCs found in multiple immune organs, including the spleen and LNs (highlighted in blue). itDCs are generally found

in various tissues and organs, including the lungs, the Downloaded from intestines, and the skin (highlighted in yellow). In these locations, itDCs are in constant contact with a variety of environmental stimuli provided by com- mensal and pathogenic organisms. http://www.jimmunol.org/ by guest on September 28, 2021 ligand-1 [PD-L1]/programmed death-1 [PD-1], CD80/CD86/ protolerogenic cytokine (34, 49, 59, 60). By acting directly CTLA-4, and B7h/ICOS) can promote Foxp3 expression, on T cells, TGF-b promotes differentiation of both Treg pTreg cell induction, and tolerance (7, 11, 49). Specifically, the and some effector T cells (43, 61–65). Metabolites, such as PD-L1/PD-1 axis promotes immune tolerance via PD-L1’s retinoic acid (RA), enhance tolerogenic properties of cy- competition with costimulatory CD28 for binding with tokines including TGF-b, further promoting the induction B7-1 as well as by the recruitment of SHP-2 by PD-1, of Foxp3 expression and the amelioration of autoimmune which negatively impacts TCR signaling (50). Moreover, disease (66–70). CTLA-4 (CD152) expressed on the T cell surface competes In addition to the immunomodulatory pathways described with the costimulatory molecule CD28 for binding with above, which can directly contribute to inducing Foxp3 ex- CD80/CD86 (B7-1 and B7-2) present on the surface of pression in pTreg cells, recent results identified the roles of the DC. Although CTLA-4 can remove CD80/CD86 from BTLA and CD5 in promoting pTreg cell homeostasis via the the DC surface, it also directly negatively regulates CD4+ modulation of the sensitivity of the developing pTreg cells to and CD8+ T cell activation by dampening TCR signaling, effector-differentiating cytokines (7, 42, 71, 72). Among the hindering IL-2 production, and preventing cell cycle pro- DC populations, BTLA is expressed specifically in DC1s (72). gression (51–53). Finally, B7h (ICOS-L or B7RP-1) is During the interactions between the BTLAhi DC1s and constitutively expressed on DCs, and expression of its re- T cells in the steady state, BTLA signals through herpesvirus ceptor, ICOS, is induced upon T cell activation (49). Re- entry mediator (HVEM) in naive CD4+ T cells to activate lated to other members of the CD28 Ig superfamily, ICOS MEK and, subsequently, the transcription factor ETS1 to (together with its binding partner B7h) has proven impor- increase expression of Cd5 (42, 72). High CD5 expression tant in the development of Treg cells, and a deficiency in then allows for a conversion of these T cells to Foxp3+ pTreg ICOS signaling has rendered mice more susceptible to cells by interfering with mammalian target of rapamycin various autoimmune pathologies, although ICOS functions (mTOR) activation in response to effector-differentiating are required for the progression of some autoimmune processes cytokines, such as IL-4, IL-6, and IFN-g (7, 42, 71). (54–58). BTLAhi DC1s reside in lymphoid organs, including the Among various cytokines governing immune responses, the spleen, and these resident lymphoid tissue DCs are ideally presence of TGF-b is often correlated with maintaining im- positioned to capture systemic self-antigens, including those mune homeostasis. The absence of TGF-b leads to sponta- derived from apoptotic cells (20, 73, 74). Although the neous lymphoproliferation and inflammatory disease, and specific roles of BTLA in governing tolerance among CD8+ TGF-b also influences the expression of IL-10, an important T cells remain unclear, DC1s can maintain tolerance to 736 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS Downloaded from http://www.jimmunol.org/ FIGURE 2. ntDCs promote tolerance in the steady state. Using multiple receptors (examples of which are shown), ntDCs constitutively uptake and process tissue-derived Ags from apoptotic cells for subsequent presentation to T cells, thereby further enhancing tolerance toward self-antigens. Similarly, ntDCs uptake, process, and present soluble Ag. ntDCs, which are predominantly DC1s, inherently express immunomodulatory molecules (including BTLA, PD-L1, B7h, and CD80/CD86) as well as cytokines (such as TGF-b and IL-10). Interaction of BTLA with the T cell–expressed receptor HVEM is necessary for the efficient induction of pTreg cells and long-lasting tolerance. self-antigens by presenting endogenous Ags to both CD4+ (87, 88). However, under steady-state conditions, the resident + hi and CD8 T cells (73, 75). DC1s also have important roles lymphoid tissue BTLA DC1s (as well as some migratory by guest on September 28, 2021 in eliciting Th1 responses as well as in cross-priming CD8+ DCs) exhibit inherent tolerogenic properties (11). The ability cytotoxic T cells (23). Such versatile functions of some of DC1s to uptake, process, and present Ags to T cells in the DC1s may reflect complex mechanisms mediated by BTLA steady state has been demonstrated by using multiple meth- and HVEM (76–79), yet it is also interesting to speculate ods, including a direct targeting of Ags to such DCs by using that at least some of these proimmune functions may be chimeric Abs specific for DEC-205 and other surface recep- performed by DC1s characterized by low expression of tors (11, 17, 89). Importantly, the combination of genetic BTLA. models including a DC-specific deletion of Irf4 (resulting in Nevertheless, it is clear that apoptotic materials are an an increased DC1/DC2 ratio) or a deletion of Batf3 (resulting abundant source of tissue self-antigens crucial for the main- in decreased numbers of DC1s) and targeted Ag delivery to tenance of immune tolerance (20, 80, 81). The relationship DC-specific molecules has helped to further clarify the between the uptake, processing, and presentation of apoptotic specific functions of DC subsets (11, 17, 42, 49). materials and the functional characteristics of DCs is complex. In addition to the tolerance promoted by resident lymphoid The phagocytic scavenger receptor CD36 has long been rec- tissue DCs, tolerance to self-antigens is also promoted by ognized for its role in facilitating the uptake of apoptotic migratory DCs that transport Ags from the nonlymphoid materials via recognition of phosphatidylserine found on the tissue to the lymph nodes (LNs). Such migratory DC1s, es- outer leaflets of the membranes of apoptotic cells (74, 82). pecially those found in skin and parenchymal organs, undergo Consistent with the notion that DC1s are primarily respon- homeostatic maturation in the steady state and have tolero- sible for the constitutive uptake of apoptotic materials, CD36 genic functions (40, 90–92). However, the tolerogenic func- is expressed highly on DC1s compared with DC2s and pDCs tions of many migratory DCs are also induced at certain (83, 84). In addition to being a source of Ags, an exposure to anatomical locations (such as the intestines) upon exposure to apoptotic materials may also enhance the tolerogenic prop- specific extrinsic stimuli (as discussed below). erties of some DCs. An engagement of CD36 can inhibit maturation of moDCs induced by proimmunogenic stimuli Inducing tolerogenic DCs for the maintenance of homeostasis and can induce tolerogenic BMDCs that resist LPS stimula- In contrast to resident lymphoid tissue DCs and DCs mi- tion and induce Foxp3+ pTreg cell development (85, 86), also grating from the parenchymal organs in the steady state, many discussed later in the text. DCs that are exposed to various environmental stimuli present In contrast, necrotic-type materials derived from injured cells in the intestines, airways, and skin are constantly at risk for can be recognized by Clec9a (DNGR-1), a C-type lectin expressed undergoing immunogenic maturation. However, as discussed by some DC1s, resulting in inflammation and maturation of DCs earlier in the text, these DCs do not induce detrimental The Journal of Immunology 737 immune responses and instead induce tolerogenic functions. production, whereas stimulation of TLR-4 promotes Th17- The specific tolerogenic mechanisms employed by itDCs re- and IL-17–driven pathology (119). Various TLRs may also main an area of active investigation, but it is clear that, despite physically associate as heterodimers to promote contrasting the presence of proinflammatory mediators, these DCs are responses depending on the specific composition of each typically characterized by an elevated production of various heterodimer. Studies using the Yersinia pestis virulence factor anti-inflammatory cytokines and other regulatory molecules LcrV showed that recognition of LcrV by a TLR-2/TLR-6 (11, 93–99). heterodimer could lead to itDC induction, complete with Among such specific anatomical sites, the skin represents a IL-10 production and Tr1 induction, whereas recognition crucial barrier that is in constant contact with foreign Ags and of LcrV by a TLR-2/TLR-1 heterodimer resulted in IL-12 commensal microbes and that requires intricately regulated production and induction of effector Th1 cells (120). immune responses orchestrated by DCs (93, 100). The lungs A particular agonist may also lead to divergent immune are another crucial anatomical site that is continually exposed responses by stimulating different PRRs. Early reports sug- to commensals and pathogens, therefore also requiring active gested that zymosan, a glucan derived from yeast cell walls, immunoregulation. Correspondingly, DCs obtained from promotes IL-10 production and tolerance by concomitant patients with chronic obstructive pulmonary disease were engagement of TLR-2 and dectin-1 (121). However, later shown to produce IL-10, leading to the induction of Tr1 reports from ex vivo and in vivo experimental systems sug- regulatory cells (95, 101, 102). The intestines are yet an- gested that divergent responses may arise from zymosan’s other key organ that remains in constant contact with a large stimulation of these PRRs. It was found that TLR-2 ligation Downloaded from number of commensal bacteria and that can also be exposed by zymosan increases the expression of Raldh2 by DCs, to potentially pathogenic microbes. Oral administration of leading to the production of RA and the eventual promotion Ags is well established to lead to the induction of Foxp3+ Treg of Treg cells via the suppression of effector differentiation. In cells, thereby helping to maintain homeostasis (103). contrast, ligation of dectin-1 increases Th1 and Th17 differ- It has recently been suggested that the specific anatomical entiation and exacerbates autoimmunity (116). In some in- organization of gut-draining LNs may play a role in the balance stances, the concomitant signaling of specific combinations of http://www.jimmunol.org/ of tolerance and immunity, as proximal and distal gut-draining other PRRs (such as TLR-2 and TLR-4 or TLR-3, -4, or -5 LNs supported primarily tolerogenic and immunogenic re- and DC-SIGN) may result in tolerogenic profiles, including sponses, respectively (47). In addition to such anatomic spe- increased IL-10 production and lower costimulatory molecule cialization, other studies have demonstrated that the presence expression by DCs (122, 123). of commensal microbes from the human gut, dietary me- In addition to TLRs, signaling via G-–coupled tabolites, and some other biologically active molecules results receptors (such as GPR109a and GPR81) extends DC- in the formation of itDCs that can differentiate Foxp3+ Treg mediated tolerance in the gut. The G-protein–coupled

cells and regulatory Tr1 cells and decrease the numbers of receptor GPR109a is a receptor for commensal bacteria– by guest on September 28, 2021 effector T cells (33, 104–107). Accordingly, the pTreg cell– produced butyrate and niacin that induce production of IL-10 2 inducing functions of Irf8/Batf3-dependent CD103+CD11b and Aldh1a1 in DCs (124). The deficiency of GPR109a 2 2 DC1s help maintain a local immune homeostasis within the (genetically modeled in Niacr1 / mice) results in an in- gut-associated lymphoid tissues as well as at other mucosal creased susceptibility to colonic inflammation and colon surfaces and also some immune-privileged sites, such as the cancer in azoxymethane- and dextran sulfate sodium–treated eye (12, 13, 32, 108–110). Although DC2s are generally less mice, respectively (124). Similarly, the G-protein–coupled efficient at inducing pTreg cells, some itDC2s in the intes- receptor GPR81 is expressed in intestinal DCs and macro- tines still promote both Treg cell–independent and Treg cell– phages and has been shown to be activated by lactate, a dependent tolerance (111, 112). product of microbial fermentation that is present in abun- Recent work identified that stimulation through certain danceinthecolon(125,126).Thegeneticdeletionof PRRs and the presence of specific cytokines and metabolites GPR81 results in a decrease in tolerance protecting from can actively divert DCs toward tolerogenic functions [(34, 47, colitis, as evidenced by an increase in proinflammatory cy- 93, 101, 113) and (Fig. 3)]. In contrast to their proimmu- tokine production, a decrease in regulatory factors such as nogenic roles, some PRRs may contribute to a tolerogenic IL-10, and a decrease in IDO1 expression; correspondingly, sensitization (29, 33, 114–117). However, the impact of in- pharmacological activation of GPR81 has been shown to dividual PRRs on the induction of tolerogenic DCs is likely to lessen murine colitis severity (126). Also, other molecules, be context dependent. For example, BMDCs differentiated in such as DC-SIGN, play a specific role in itDC induction, as the presence of splenic stroma were found to produce high well as in the induction of tolerogenic functions in macro- amounts of IL-10 and dampen naive CD4+ T cell responses in phages, by promoting specific mechanisms of tolerance (such culture (36). Paradoxically, an additional stimulation with as the production of IL-10) in response to various microor- TLR-2, -3, -4, and -9 agonists of DCs cocultured with splenic ganisms (117, 127, 128). However, the binding of DC-SIGN stromal cells further enhances their tolerogenic state, resulting (possibly in concert with the binding of TLRs) to various in heightened production and secretion of CXCR3 chemo- ligands, including cell wall components and modified oligo- kine IFN-g–inducible protein 10 (IP-10) and a correspond- saccharides derived from bacterial LPS, results in divergent ing decrease in Th1 proliferation (118). Nevertheless, certain tolerogenic or immunogenic DC–mediated immune re- TLRs such as TLR-2 appear to be more tolerogenic than sponses and may also lead to Th1 responses (122, 129–131). other TLRs. In a murine disease model of arthritis, the mi- Certain cytokines and metabolites have crucial roles in in- crobial commensals’ stimulation of TLR-2 on APCs pro- ducing and governing the functions of itDCs as well as in motes Treg cell suppressive functions and dampens IFN-g shaping the responses of DCs to PRR ligands. For example, the 738 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS Downloaded from

FIGURE 3. itDCs promote T cell tolerance in response to exogenous signals. itDCs can be induced by multiple types of molecules, including microbial components (which bind to various PRRs, such as TLR-2), pharmacological and dietary agents (such as corticosteroids and probiotics) or certain physiological agents that bind to the AHR or the VDR, and also by other specific metabolites, cytokines, and growth factors. The downstream diverse immunoregulatory effects are mediated by several different signaling pathways, including NF-kB, Wnt/b-catenin, and mTOR, ultimately resulting in active mechanisms of tolerance that induce the expression and production of multiple immunomodulatory mediators, including IL-10, RA, and kynurenine (Kyn) (a metabolic product of tryptophan http://www.jimmunol.org/ catabolism that is mediated by IDO1). addition of IL-10 to human DC cultures may lead to decreased signaling directly affected DCs and subsequent T cell re- expression of MHCII (HLA-DR) and costimulatory mole- sponses. A more recent report highlighted the role of IL-27 cules, resulting in T cell anergy (132, 133). In the small in- and the immunoregulatory molecule CD39 expressed by testine, RA is locally found at high concentrations due to the DCs. Specifically, CD39, whose expression is induced by metabolism of dietary vitamin A (109, 134). This localized IL-27, reduced NLRP3 inflammasome activation in DCs, presence of RA then promotes the expression of Raldh2 and thereby reducing subsequent Th1 and Th17 effector T cell by guest on September 28, 2021 production of additional RA by CD103+ DCs in the lamina differentiation (146). propria, ultimately resulting in the increased induction of Other extensively studied physiological factors that function IL-10–producing Foxp3+ Treg cells (as well as in the inhibi- in itDC promotion are ligands for the aryl hydrocarbon re- tion of TGF-b–mediated Th17 cell induction and in the ceptor (AHR), which is important in the regulation of the imprinting of gut-homing receptors on T cells) (66, 108, 109, balance between the formation of regulatory and effector 135–137). Consistent with its protolerogenic roles, IL-10 can T cells (147). The endogenous ligands of AHR, such as then downregulate DC expression of MHCII and costimu- 2-(19H-indole-39-carbonyl)-thiazole-4-carboxylic acid methyl latory molecules and reverse the effects of proinflammatory ester (ITE), also act directly on DCs by skewing them toward cytokines, such as IL-6 and TNF-a (60). Further, IL-10 a tolerogenic profile characterized by decreased production treatment of human DCs upregulates TLR-2 expression in of inflammatory cytokines like IL-6 and IL-12 and in- response to LPS administration, consistent with the pro- creased production of suppressive TGF-b and IL-10 (104, tolerogenic functions observed following TLR-2 activation 148). In DCs, AHR can induce expression of IDO1, an (as discussed above). Such treated DCs also decrease ex- immunosuppressive enzyme that catabolizes tryptophan into pression of IL-12–related cytokines, possibly further indi- kynurenine (Kyn) and other metabolites (148). Further, the cating collaborative roles of IL-10 and TLR-2 in the upregulation of IDO1 and Socs2 expression and increased RA dampening of the immune response (138). production by DCs contribute to a decrease in the proin- In addition to IL-10, the IL-12 cytokine family member flammatory cytokine milieu and the subsequent induction IL-35, predominantly produced by Treg cells, was proposed to of Foxp3+ Treg cells (149). Together, these findings indicate dampen T cell responses (139). Similarly, IL-37 decreases the a crucial role of ITE and AHR signaling in the induction of production of proinflammatory cytokines induced by LPS itDCs that could have therapeutic roles in multiple types of stimulation, and the forced expression of IL-37b in murine autoimmune responses (150). skin DCs promotes tolerogenic DC induction, thereby af- Vitamin D3 represents another key physiological factor fecting contact hypersensitivity challenge (140, 141). Among that may induce tolerogenic DCs (151–153). The active form other cytokines, IL-27 was initially considered to be proim- of vitamin D3 induces immunoregulatory properties upon munogenic (142). However, other studies indicated the role binding to the vitamin D receptor (VDR), which is selec- of IL-27 in suppressing the differentiation of effector T cells tively expressed by various cell types, including intestinal and in autoimmune models, such as experimental autoimmune skin epithelial cells, osteoblasts, CD4+ and CD8+ T cells, encephalomyelitis (EAE), an animal model of multiple scle- and also multiple APCs, including macrophages, monocytes, rosis (MS) (143–145). Still, it remained unknown how IL-27 and DCs (151, 154). VDR agonists increase IL-10 production The Journal of Immunology 739 but decrease expression of IL-12 and CD80/CD86 and CD40 relevant for transplantations as well as tumor immunology by DCs (155). Additionally, itDCs induced by VDR agonists, (11). Both ntDCs and itDCs are relevant for such therapeutic such as calcitriol and paricalcitol, are poor inducers of Ag- manipulations, and the targeted delivery of Ags to DEC-205+ specific effector T cells but are potent inducers of Treg cells and other DCs has proven to be a powerful way to reinforce (151–153, 156). It was further demonstrated that DCs may tolerance to self-antigens implicated in the autoimmune also synthesize the active form of vitamin D3, therefore process (89). For example, the spontaneous induction of pe- providing a local source of this crucial immunomodulant, ripheral tolerance in response to Ags derived from organs altering immune cell trafficking, and further increasing the insulated from the immune system (such as the CNS), is DC secretion of the chemokine CCL22, which attracts Treg likely less efficient (7, 11). This results in an increased po- cells (157–161). Overall, VDR agonists are promising ther- tential for autoimmune diseases such as in animal models of apeutics for autoimmune diseases, transplantation tolerance, MS, which can be readily provoked after an immunization of and allergies (151–153, 156). healthy animals with CNS Ags (7, 178, 179). However, such Other physiological factors may also play a role in itDC autoimmune responses can be blocked by tolerance induced promotion. Vasoactive intestinal peptide decreases production by targeted delivery of various tissue-specific Ags to DCs, of proinflammatory TNF-a and IL-6 and increases produc- allowing for efficient Ag presentation to self-reactive T cells tion of IL-10 in human DCs despite a pre-exposure to LPS, (11, 17, 18, 89). thereby mediating T cell anergy (162). Also, an addition of In these initial experiments, DCs were targeted in vivo with seminal plasma to differentiating moDCs yielded high regu- anti–DEC-205 chimeric Ab to deliver a potentially enceph- Downloaded from latory cytokine and low proinflammatory cytokine production alitogenic peptide Ag derived from myelin oligodendrocyte profiles in such moDCs (163). glycoprotein, which was genetically fused to the Ab molecule, In addition to these physiological factors, 14-dehydroergosterol to prevent subsequently induced EAE (18). These early results (14-DHE), an ergosterol analogue–based compound derived were then extended to other EAE models and various en- from fermented wheat bran, induces tolerogenic properties in cephalitogenic Ags that were delivered through DC-specific

DCs, although the exact mechanisms are unclear (164, 165). molecules, as recently reviewed in Refs. 11 and 89. The targeting http://www.jimmunol.org/ Similarly, multiple pharmacological agents, including chem- of Ags to DCs has also been successful in mediating tolerance ically modified TLR ligands, JAK inhibitors, corticosteroids, in multiple different models of autoimmunity, including dia- cisplatin, antibiotics, probiotics (including bacterial species of betes, colitis, and arthritis, as well as in a model of graft-versus- the Lactobacillus, Bifidobacterium, and Streptococcus genera), host disease (180–183). Overall, this specific delivery of Ags and dietary supplements (such as zinc) have been shown to results in tolerogenic mechanisms that prevent autoimmunity. promote tolerogenic profiles in BMDCs and moDCs. These In addition to the delivery of Ags specifically targeted to profiles are characterized by changes at both the transcrip- DCs, other studies found that certain formulations of Ags

tional and translational levels, ultimately leading to decreased (such as nanoparticles or specifically modified cellular material) by guest on September 28, 2021 production of the inflammatory cytokines TNF-a and IL-6, could lead to their in vivo acquisition by DCs and to the decreased expression of costimulatory molecules, increased amelioration of autoimmune processes including EAE and expression of coinhibitory molecules like PD-L1, and the diabetes (184–187). Despite generally lacking cell target production of the anti-inflammatory cytokines IL-10 and specificity, these methods showed promise in the treatment of TGF-b (166–169). ongoing autoimmune processes, particularly when addition- Among the intrinsic signaling pathways involved in sensing ally coupled with agents known to induce the formation of specific extrinsic factors that induce tolerogenic properties in itDCs under proinflammatory conditions (149). Some of DCs, Wnt/b-catenin is a major molecular pathway involved those methods also incorporated DC-specific Abs to en- in the promotion of itDCs and the increased production of hance the specificity of the delivery system (188), although anti-inflammatory cytokines (such as TGF-b, RA, and IL-10) most of such Ab-coupled immunogenic particles have been and, conversely, in blocking the production of NF-kB– tested for new vaccine approaches (189). In addition to induced proinflammatory cytokines (32, 33, 67, 170). De- controlling autoimmunity, DC-mediated tolerance holds ficiencies in Wnt receptors LRP5/6 and b-catenin–signaling promise in mitigating transplant rejection (as recently mechanisms enhance proinflammatory cytokine production reviewed by Thomson et al. in Ref. 190). Importantly, DCs and Th1/Th17 effector responses and increase disease severity can cooperate with other types of immune cells, such as in mouse models including colitis and MS (67, 171–174). NKTcells,topreventgraftrejections(191). Noncanonical Wnt signaling mediated by Wnt5a is also in- In human systems, the most available options thus far have volved in the promotion of tolerance by modifying DC been to induce itDCs ex vivo, analogous to the induction of maturation and IL-10 production in response to TLR agonists itDCs from murine BMDCs, and to subsequently treat them (175). In addition to Wnt/b-catenin, mTOR emerges as an with agents that can potentially further promote such itDC important regulator of DC functions that may also affect differentiation in vivo. Either murine BMDCs or moDCs tolerogenic mechanisms (176, 177). derived from humans afflicted with an autoimmune disease were treated in vitro with the pharmacologic agents PEGylated–TLR- Harnessing the tolerogenic functions of DCs for 7 ligand, dexamethasone plus monophosphoryl lipid A, therapeutic applications tofacitinib, or prednisolone and were then used to delay dis- ThefunctionsofDCsasinducersoftolerancerepresent ease onset or ameliorate disease severity in diabetes, rheuma- important therapeutic opportunities. Modulation of such DC- toid arthritis, EAE, and myasthenia gravis, respectively (166, induced tolerance can help block different forms of autoim- 167, 169, 192–194). Also, treatment with IL-10 leads to the munity and also impact other types of immune responses formation of itDCs that likely possess clinical relevance (195). 740 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS

Further, the treatment of BMDCs with the antitumor drug 14. Wang, L., Z. Li, B. Ciric, F. Safavi, G. X. Zhang, and A. Rostami. 2016. Selective depletion of CD11c+ CD11b+ dendritic cells partially abrogates tolerogenic effects cisplatin in conjunction with various TLR agonists results in of intravenous MOG in murine EAE. Eur. J. Immunol. 46: 2454–2466. increased IL-10 production by the BMDCs as well as the 15. Ohnmacht, C., A. Pullner, S. B. King, I. Drexler, S. Meier, T. Brocker, and D. Voehringer. 2009. Constitutive ablation of dendritic cells breaks self-tolerance prevention of Th1 and Th17 responses (196). of CD4 T cells and results in spontaneous fatal autoimmunity. J. Exp. Med. 206: In contrast to the generally beneficial functions of tolero- 549–559. genic DCs in the prevention of autoimmunities as discussed 16. Fukaya, T., R. Murakami, H. Takagi, K. Sato, Y. Sato, H. Otsuka, M. Ohno, A. Hijikata, O. Ohara, M. Hikida, et al. 2012. Conditional ablation of CD205+ above, the tumor microenvironment can skew DCs toward conventional dendritic cells impacts the regulation of T-cell immunity and ho- tolerogenic functions, thereby diminishing tumor rejection meostasis in vivo. Proc. Natl. Acad. Sci. USA 109: 11288–11293. 17. Hawiger, D., K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. Rivera, J. V. Ravetch, (177, 197–202). In an effort to induce antitumor immunity, R. M. Steinman, and M. C. Nussenzweig. 2001. Dendritic cells induce peripheral multiple DC-based immunotherapies against cancer have T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194: 769–779. been gaining importance, as recently reviewed by Sancho and 18. Hawiger, D., R. F. Masilamani, E. Bettelli, V. K. Kuchroo, and M. C. Nussenzweig. colleagues (203). Even more breakthroughs that will deter- 2004. Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo. Immunity 20: 695–705. mine the contributions of various DCs to tumor-immune 19. Probst, H. C., K. McCoy, T. Okazaki, T. Honjo, and M. van den Broek. 2005. evasion and allow for the harnessing of such DCs for anti- Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and tumor therapies are expected. CTLA-4. Nat. Immunol. 6: 280–286. 20. Belz, G. T., G. M. Behrens, C. M. Smith, J. F. Miller, C. Jones, K. Lejon, C.G.Fathman,S.N.Mueller,K.Shortman,F.R.Carbone,andW.R.Heath. 2002. The CD8alpha(+) dendritic cell is responsible for inducing peripheral self- Conclusions tolerance to tissue-associated antigens. J. Exp. Med. 196: 1099–1104. The recent years have seen a growing understanding of DC 21. Murphy, T. L., G. E. Grajales-Reyes, X. Wu, R. Tussiwand, C. G. Brisen˜o, Downloaded from functions in both the initiation and the regulation of immune A. Iwata, N. M. Kretzer, V. Durai, and K. M. Murphy. 2016. Transcriptional control of dendritic cell development. Annu. Rev. Immunol. 34: 93–119. responses. Overall, whereas functions of itDCs contribute to 22. Merad, M., P. Sathe, J. Helft, J. Miller, and A. Mortha. 2013. The dendritic cell the maintenance of homeostasis under potentially proin- lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31: 563–604. flammatory conditions, ntDCs help to establish tolerance 23. Durai, V., and K. M. Murphy. 2016. Functions of murine dendritic cells. under steady-state conditions. Importantly, the new insights Immunity 45: 719–736.

24. Guilliams, M., C. A. Dutertre, C. L. Scott, N. McGovern, D. Sichien, http://www.jimmunol.org/ are providing us with a framework for exploiting the DC- S. Chakarov, S. Van Gassen, J. Chen, M. Poidinger, S. De Prijck, et al. 2016. mediated mechanisms of tolerance for more effective im- Unsupervised high-dimensional analysis aligns dendritic cells across tissues and munotherapies, which will, hopefully, be burdened by fewer species. Immunity 45: 669–684. 25. Swiecki, M., and M. Colonna. 2015. The multifaceted biology of plasmacytoid side effects. dendritic cells. Nat. Rev. Immunol. 15: 471–485. 26. Ziegler-Heitbrock, L., P. Ancuta, S. Crowe, M. Dalod, V. Grau, D. N. Hart, P. J. Leenen, Y. J. Liu, G. MacPherson, G. J. Randolph, et al. 2010. Nomenclature Disclosures of monocytes and dendritic cells in blood. Blood 116: e74–e80. The authors have no financial conflicts of interest. 27. Steinman, R. M. 2012. Decisions about dendritic cells: past, present, and future. Annu. Rev. Immunol. 30: 1–22. 28. Mellman, I. 2013. Dendritic cells: master regulators of the immune response.

Cancer Immunol. Res. 1: 145–149. by guest on September 28, 2021 References 29. Iwasaki, A., and R. Medzhitov. 2015. Control of adaptive immunity by the innate + 1. Klein, L., E. A. Robey, and C. S. Hsieh. 2019. Central CD4 T cell tolerance: immune system. Nat. Immunol. 16: 343–353. deletion versus regulatory T cell differentiation. Nat. Rev. Immunol. 19: 7–18. 30. Dalod, M., R. Chelbi, B. Malissen, and T. Lawrence. 2014. Dendritic cell mat- 2. Enouz, S., L. Carrie´, D. Merkler, M. J. Bevan, and D. Zehn. 2012. Autoreactive uration: functional specialization through signaling specificity and transcriptional T cells bypass negative selection and respond to self-antigen stimulation during programming. EMBO J. 33: 1104–1116. infection. J. Exp. Med. 209: 1769–1779. 31. Lutz, M. B., and G. Schuler. 2002. Immature, semi-mature and fully mature 3. Nelson, R. W., D. Beisang, N. J. Tubo, T. Dileepan, D. L. Wiesner, K. Nielsen, dendritic cells: which signals induce tolerance or immunity? Trends Immunol. 23: M. Wuthrich,€ B. S. Klein, D. I. Kotov, J. A. Spanier, et al. 2015. T cell receptor 445–449. cross-reactivity between similar foreign and self peptides influences naive cell 32. Manicassamy, S., and B. Pulendran. 2011. Dendritic cell control of tolerogenic population size and autoimmunity. [Published erratum appears in 2015 Immunity responses. Immunol. Rev. 241: 206–227. 42: 1212–1213.] Immunity 42: 95–107. 33. Pulendran, B. 2015. The varieties of immunological experience: of pathogens, 4. Zehn, D., and M. J. Bevan. 2006. T cells with low avidity for a tissue-restricted stress, and dendritic cells. Annu. Rev. Immunol. 33: 563–606. antigen routinely evade central and peripheral tolerance and cause autoimmunity. 34. Maldonado, R. A., and U. H. von Andrian. 2010. How tolerogenic dendritic cells Immunity 25: 261–270. induce regulatory T cells. Adv. Immunol. 108: 111–165. 5. Anderson, A. C., L. B. Nicholson, K. L. Legge, V. Turchin, H. Zaghouani, and 35. Steinman, R. M., D. Hawiger, K. Liu, L. Bonifaz, D. Bonnyay, K. Mahnke, V. K. Kuchroo. 2000. High frequency of autoreactive myelin proteolipid protein- T. Iyoda, J. Ravetch, M. Dhodapkar, K. Inaba, and M. Nussenzweig. 2003. specific T cells in the periphery of naive mice: mechanisms of selection of the self- Dendritic cell function in vivo during the steady state: a role in peripheral toler- reactive repertoire. J. Exp. Med. 191: 761–770. ance. Ann. N. Y. Acad. Sci. 987: 15–25. 6. Bouneaud, C., P. Kourilsky, and P. Bousso. 2000. Impact of negative selection on 36. Zhang, M., H. Tang, Z. Guo, H. An, X. Zhu, W. Song, J. Guo, X. Huang, the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones T. Chen, J. Wang, and X. Cao. 2004. Splenic stroma drives mature dendritic cells escapes clonal deletion. Immunity 13: 829–840. to differentiate into regulatory dendritic cells. [Published erratum appears in 2014 7. Jones, A., and D. Hawiger. 2017. Peripherally induced regulatory T cells: recruited Nat. Immunol. 15: 1090.] Nat. Immunol. 5: 1124–1133. protectors of the central nervous system against autoimmune neuroinflammation. 37. Vander Lugt, B., J. Riddell, A. A. Khan, J. A. Hackney, J. Lesch, J. DeVoss, Front. Immunol. 8: 532. M. T. Weirauch, H. Singh, and I. Mellman. 2017. Transcriptional determinants 8. Josefowicz, S. Z., L. F. Lu, and A. Y. Rudensky. 2012. Regulatory T cells: of tolerogenic and immunogenic states during dendritic cell maturation. J. Cell mechanisms of differentiation and function. Annu. Rev. Immunol. 30: 531–564. Biol. 216: 779–792. 9. Ooi, J. D., J. Petersen, Y. H. Tan, M. Huynh, Z. J. Willett, S. H. Ramarathinam, 38. Ardouin, L., H. Luche, R. Chelbi, S. Carpentier, A. Shawket, F. Montanana P. J. Eggenhuizen, K. L. Loh, K. A. Watson, P. Y. Gan, et al. 2017. Dominant Sanchis, C. Santa Maria, P. Grenot, Y. Alexandre, C. Gre´goire, et al. 2016. Broad protection from HLA-linked autoimmunity by antigen-specific regulatory T cells. and largely concordant molecular changes characterize tolerogenic and immuno- Nature 545: 243–247. genic dendritic cell maturation in thymus and periphery. Immunity 45: 305–318. 10. Koehli, S., D. Naeher, V. Galati-Fournier, D. Zehn, and E. Palmer. 2014. Op- 39. Jiang, A., O. Bloom, S. Ono, W. Cui, J. Unternaehrer, S. Jiang, J. A. Whitney, timal T-cell receptor affinity for inducing autoimmunity. Proc. Natl. Acad. Sci. J. Connolly, J. Banchereau, and I. Mellman. 2007. Disruption of E-cadherin- USA 111: 17248–17253. mediated adhesion induces a functionally distinct pathway of dendritic cell mat- 11. Iberg, C. A., A. Jones, and D. Hawiger. 2017. Dendritic cells as inducers of pe- uration. Immunity 27: 610–624. ripheral tolerance. Trends Immunol. 38: 793–804. 40. Baratin, M., C. Foray, O. Demaria, M. Habbeddine, E. Pollet, J. Maurizio, 12. Esterha´zy, D., J. Loschko, M. London, V. Jove, T. Y. Oliveira, and D. Mucida. C. Verthuy, S. Davanture, H. Azukizawa, A. Flores-Langarica, et al. 2015. Ho- 2016. Classical dendritic cells are required for dietary antigen-mediated induction meostatic NF-kB signaling in steady-state migratory dendritic cells regulates im- of peripheral T(reg) cells and tolerance. Nat. Immunol. 17: 545–555. mune homeostasis and tolerance. Immunity 42: 627–639. 13. Ohta, T., M. Sugiyama, H. Hemmi, C. Yamazaki, S. Okura, I. Sasaki, Y. Fukuda, 41.Jones,A.,A.Opejin,J.G.Henderson,C.Gross,R.Jain,J.A.Epstein, T. Orimo, K. J. Ishii, K. Hoshino, et al. 2016. Crucial roles of XCR1-expressing R. A. Flavell, and D. Hawiger. 2015. Peripherally induced tolerance depends on dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune ho- peripheral regulatory T cells that require Hopx to inhibit intrinsic IL-2 expression. meostasis. Sci. Rep. 6: 23505. J. Immunol. 195: 1489–1497. The Journal of Immunology 741

42. Jones, A., J. Bourque, L. Kuehm, A. Opejin, R. M. Teague, C. Gross, and 69. Xiao, S., H. Jin, T. Korn, S. M. Liu, M. Oukka, B. Lim, and V. K. Kuchroo. D. Hawiger. 2016. Immunomodulatory functions of BTLA and HVEM govern 2008. Retinoic acid increases Foxp3+ regulatory T cells and inhibits development induction of extrathymic regulatory T cells and tolerance by dendritic cells. of Th17 cells by enhancing TGF-beta-driven Smad3 signaling and inhibiting IL-6 Immunity 45: 1066–1077. and IL-23 receptor expression. J. Immunol. 181: 2277–2284. 43. Kretschmer, K., I. Apostolou, D. Hawiger, K. Khazaie, M. C. Nussenzweig, and 70. Massacesi, L., E. Castigli, M. Vergelli, J. Olivotto, A. L. Abbamondi, F. Sarlo, and H. von Boehmer. 2005. Inducing and expanding regulatory T cell populations by L. Amaducci. 1991. Immunosuppressive activity of 13-cis-retinoic acid and pre- foreign antigen. Nat. Immunol. 6: 1219–1227. vention of experimental autoimmune encephalomyelitis in rats. J. Clin. Invest. 88: 44. Kalekar, L. A., and D. L. Mueller. 2017. Relationship between CD4 regulatory 1331–1337. T cells and anergy in vivo. J. Immunol. 198: 2527–2533. 71. Henderson, J. G., A. Opejin, A. Jones, C. Gross, and D. Hawiger. 2015. CD5 45. Eisenbarth, S. C. 2019. Dendritic cell subsets in T cell programming: location instructs extrathymic regulatory T cell development in response to self and toler- dictates function. Nat. Rev. Immunol. 19: 89–103. izing antigens. Immunity 42: 471–483. 46. Baptista, A. P., A. Gola, Y. Huang, P. Milanez-Almeida, P. Torabi-Parizi, 72. Bourque, J., and D. Hawiger. 2019. The BTLA-HVEM-CD5 immunoregulatory J. F. Urban, Jr., V. S. Shapiro, M. Y. Gerner, and R. N. Germain. 2019. The axis-an instructive mechanism governing pTreg cell differentiation. Front. Immu- chemoattractant receptor Ebi2 drives intranodal naive CD4+ T cell peripherali- nol. 10: 1163. zation to promote effective adaptive immunity. Immunity 50: 1188–1201.e6. 73. Steinman, R. M., D. Hawiger, and M. C. Nussenzweig. 2003. Tolerogenic den- 47. Esterha´zy, D., M. C. C. Canesso, L. Mesin, P. A. Muller, T. B. R. de Castro, dritic cells. Annu. Rev. Immunol. 21: 685–711. A. Lockhart, M. ElJalby, A. M. C. Faria, and D. Mucida. 2019. Compartmen- 74. Albert, M. L., S. F. Pearce, L. M. Francisco, B. Sauter, P. Roy, R. L. Silverstein, talized gut lymph node drainage dictates adaptive immune responses. Nature 569: and N. Bhardwaj. 1998. Immature dendritic cells phagocytose apoptotic cells via 126–130. alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. 48. Dudziak, D., A. O. Kamphorst, G. F. Heidkamp, V. R. Buchholz, C. Trumpfheller, J. Exp. Med. 188: 1359–1368. S. Yamazaki, C. Cheong, K. Liu, H. W. Lee, C. G. Park, et al. 2007. Differential 75. Kurts, C., J. F. Miller, R. M. Subramaniam, F. R. Carbone, and W. R. Heath. antigen processing by dendritic cell subsets in vivo. Science 315: 107–111. 1998. Major histocompatibility complex class I-restricted cross-presentation is 49. Bourque, J., and D. Hawiger. 2018. Immunomodulatory bonds of the partnership biased towards high dose antigens and those released during cellular destruction. between dendritic cells and T cells. Crit. Rev. Immunol. 38: 379–401. J. Exp. Med. 188: 409–414. 50. Sharpe, A. H., and K. E. Pauken. 2018. The diverse functions of the PD1 in- 76. Steinberg, M. W., Y. Huang, Y. Wang-Zhu, C. F. Ware, H. Cheroutre, and hibitory pathway. Nat. Rev. Immunol. 18: 153–167. M. Kronenberg. 2013. BTLA interaction with HVEM expressed on CD8(+) Downloaded from 51. Walunas, T. L., C. Y. Bakker, and J. A. Bluestone. 1996. CTLA-4 ligation blocks T cells promotes survival and memory generation in response to a bacterial in- CD28-dependent T cell activation. [Published erratum appears in 1996 J. Exp. fection. PLoS One 8: e77992. Med. 184: 301.] J. Exp. Med. 183: 2541–2550. 77. Shui, J. W., M. W. Steinberg, and M. Kronenberg. 2011. Regulation of inflam- 52. Qureshi, O. S., Y. Zheng, K. Nakamura, K. Attridge, C. Manzotti, E. M. Schmidt, mation, autoimmunity, and infection immunity by HVEM-BTLA signaling. J. Baker, L. E. Jeffery, S. Kaur, Z. Briggs, et al. 2011. Trans-endocytosis of CD80 J. Leukoc. Biol. 89: 517–523. and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 78. Murphy, T. L., and K. M. Murphy. 2010. Slow down and survive: enigmatic 332: 600–603. immunoregulation by BTLA and HVEM. Annu. Rev. Immunol. 28: 389–411.

53. Bucktrout, S. L., J. A. Bluestone, and F. Ramsdell. 2018. Recent advances in 79. So, T., and M. Croft. 2013. Regulation of PI-3-kinase and Akt signaling in http://www.jimmunol.org/ immunotherapies: from infection and autoimmunity, to cancer, and back again. T lymphocytes and other cells by TNFR family molecules. Front. Immunol. 4: 139. Genome Med. 10: 79. 80. Iyoda, T., S. Shimoyama, K. Liu, Y. Omatsu, Y. Akiyama, Y. Maeda, K. Takahara, 54. Akbari, O., G. J. Freeman, E. H. Meyer, E. A. Greenfield, T. T. Chang, R. M. Steinman, and K. Inaba. 2002. The CD8+ dendritic cell subset selectively A. H. Sharpe, G. Berry, R. H. DeKruyff, and D. T. Umetsu. 2002. Antigen- endocytoses dying cells in culture and in vivo. J. Exp. Med. 195: 1289–1302. specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit 81. Luckashenak, N., S. Schroeder, K. Endt, D. Schmidt, K. Mahnke, M. F. Bachmann, allergen-induced airway hyperreactivity. Nat. Med. 8: 1024–1032. P. Marconi, C. A. Deeg, and T. Brocker. 2008. Constitutive crosspresentation of 55.Ito,T.,M.Yang,Y.-H.Wang,R.Lande,J.Gregorio,O.A.Perng,X.-F.Qin, tissue antigens by dendritic cells controls CD8+ T cell tolerance in vivo. Immunity 28: Y.-J. Liu, and M. Gilliet. 2007. Plasmacytoid dendritic cells prime IL-10- 521–532. producing T regulatory cells by inducible costimulator ligand. J. Exp. Med. 82. Rigotti, A., S. L. Acton, and M. Krieger. 1995. The class B scavenger receptors SR- 204: 105–115. BI and CD36 are receptors for anionic phospholipids. J. Biol. Chem. 270: 16221– 56. Dong, C., A. E. Juedes, U. A. Temann, S. Shresta, J. P. Allison, N. H. Ruddle, and 16224.

R. A. Flavell. 2001. ICOS co-stimulatory receptor is essential for T-cell activation 83. Immunological Genome Project Consortium. 2008. The Immunological Genome by guest on September 28, 2021 and function. Nature 409: 97–101. Project: networks of in immune cells. Nat. Immunol. 9: 1091– 57. Iwai, H., M. Abe, S. Hirose, F. Tsushima, K. Tezuka, H. Akiba, H. Yagita, 1094. K. Okumura, H. Kohsaka, N. Miyasaka, and M. Azuma. 2003. Involvement of 84. Perry, J. S. A., E. V. Russler-Germain, Y. W. Zhou, W. Purtha, M. L. Cooper, inducible costimulator-B7 homologous protein costimulatory pathway in murine J. Choi, M. A. Schroeder, V. Salazar, T. Egawa, B. C. Lee, et al. 2018. Transfer of lupus nephritis. J. Immunol. 171: 2848–2854. cell-surface antigens by scavenger receptor CD36 promotes thymic regulatory 58. Hawiger, D., E. Tran, W. Du, C. J. Booth, L. Wen, C. Dong, and R. A. Flavell. T cell receptor repertoire development and allo-tolerance. [Published erratum 2008. ICOS mediates the development of insulin-dependent diabetes mellitus in appears in 2018 Immunity 48: 1271.] Immunity 48: 923–936.e4. nonobese diabetic mice. J. Immunol. 180: 3140–3147. 85. Kushwah, R., J. Wu, J. R. Oliver, G. Jiang, J. Zhang, K. A. Siminovitch, and 59. Zhang, X., H. Huang, J. Yuan, D. Sun, W. S. Hou, J. Gordon, and J. Xiang. J. Hu. 2010. Uptake of apoptotic DC converts immature DC into tolerogenic DC 2005. CD4-8- dendritic cells prime CD4+ T regulatory 1 cells to suppress anti- that induce differentiation of Foxp3+ Treg. Eur. J. Immunol. 40: 1022–1035. tumor immunity. J. Immunol. 175: 2931–2937. 86. Urban, B. C., N. Willcox, and D. J. Roberts. 2001. A role for CD36 in the 60. Li, M. O., and R. A. Flavell. 2008. Contextual regulation of inflammation: a regulation of dendritic cell function. Proc. Natl. Acad. Sci. USA 98: 8750–8755. duet by transforming growth factor-beta and interleukin-10. Immunity 28: 468– 87. Sachet, M., Y. Y. Liang, and R. Oehler. 2017. The immune response to secondary 476. necrotic cells. Apoptosis 22: 1189–1204. 61. Chen, W., W. Jin, N. Hardegen, K. J. Lei, L. Li, N. Marinos, G. McGrady, and 88.Sancho,D.,O.P.Joffre,A.M.Keller,N.C.Rogers,D.Martı´nez, S. M. Wahl. 2003. Conversion of peripheral CD4+CD25- naive T cells to P. Hernanz-Falco´n, I. Rosewell, and C. Reis e Sousa. 2009. Identification of a CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor dendritic cell receptor that couples sensing of necrosis to immunity. Nature Foxp3. J. Exp. Med. 198: 1875–1886. 458: 899–903. 62. Fantini, M. C., C. Becker, G. Monteleone, F. Pallone, P. R. Galle, and 89. Iberg, C. A., and D. Hawiger. 2019. Advancing immunomodulation by in vivo M. F. Neurath. 2004. Cutting edge: TGF-beta induces a regulatory phenotype in antigen delivery to DEC-205 and other cell surface molecules using recombinant CD4+CD25- T cells through Foxp3 induction and down-regulation of Smad7. chimeric antibodies. Int. Immunopharmacol. 73: 575–580. J. Immunol. 172: 5149–5153. 90. Azukizawa, H., A. Do¨hler, N. Kanazawa, A. Nayak, M. Lipp, B. Malissen, 63. Liu, Y., P. Zhang, J. Li, A. B. Kulkarni, S. Perruche, and W. Chen. 2008. I. Autenrieth, I. Katayama, M. Riemann, F. Weih, et al. 2011. Steady state mi- A critical function for TGF-beta signaling in the development of natural gratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction of CD4+CD25+Foxp3+ regulatory T cells. Nat. Immunol. 9: 632–640. antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells. Eur. J. Immunol. 41: 64. Mangan, P. R., L. E. Harrington, D. B. O’Quinn, W. S. Helms, D. C. Bullard, 1420–1434. C. O. Elson, R. D. Hatton, S. M. Wahl, T. R. Schoeb, and C. T. Weaver. 2006. 91. Guilliams, M., K. Crozat, S. Henri, S. Tamoutounour, P. Grenot, E. Devilard, Transforming growth factor-beta induces development of the T(H)17 lineage. B. de Bovis, L. Alexopoulou, M. Dalod, and B. Malissen. 2010. Skin-draining Nature 441: 231–234. lymph nodes contain dermis-derived CD103(-) dendritic cells that constitutively 65. Bettelli, E., Y. Carrier, W. Gao, T. Korn, T. B. Strom, M. Oukka, H. L. Weiner, produce retinoic acid and induce Foxp3(+) regulatory T cells. Blood 115: 1958– and V. K. Kuchroo. 2006. Reciprocal developmental pathways for the generation 1968. of pathogenic effector TH17 and regulatory T cells. Nature 441: 235–238. 92. Idoyaga, J., C. Fiorese, L. Zbytnuik, A. Lubkin, J. Miller, B. Malissen, D. Mucida, 66. Mucida, D., Y. Park, G. Kim, O. Turovskaya, I. Scott, M. Kronenberg, and M. Merad, and R. M. Steinman. 2013. Specialized role of migratory dendritic cells H. Cheroutre. 2007. Reciprocal TH17 and regulatory T cell differentiation me- in peripheral tolerance induction. J. Clin. Invest. 123: 844–854. diated by retinoic acid. Science 317: 256–260. 93. Volz, T., Y. Skabytska, E. Guenova, K. M. Chen, J. S. Frick, C. J. Kirschning, 67.Manicassamy,S.,B.Reizis,R.Ravindran,H.Nakaya,R.M.Salazar- S. Kaesler, M. Ro¨cken, and T. Biedermann. 2014. Nonpathogenic bacteria alle- Gonzalez, Y. C. Wang, and B. Pulendran. 2010. Activation of beta-catenin in viating atopic dermatitis inflammation induce IL-10-producing dendritic cells and dendritic cells regulates immunity versus tolerance in the intestine. Science regulatory Tr1 cells. J. Invest. Dermatol. 134: 96–104. 329: 849–853. 94. Krempski, J., L. Karyampudi, M. D. Behrens, C. L. Erskine, L. Hartmann, 68. Nozaki, Y., T. Yamagata, M. Sugiyama, S. Ikoma, K. Kinoshita, and M. Funauchi. H. Dong, E. L. Goode, K. R. Kalli, and K. L. Knutson. 2011. Tumor-infiltrating 2006. Anti-inflammatory effect of all-trans-retinoic acid in inflammatory arthritis. programmed death receptor-1+ dendritic cells mediate immune suppression in Clin. Immunol. 119: 272–279. ovarian cancer. J. Immunol. 186: 6905–6913. 742 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS

95. Weiner, H. L. 2001. The mucosal milieu creates tolerogenic dendritic cells and 121. Dillon, S., S. Agrawal, K. Banerjee, J. Letterio, T. L. Denning, K. Oswald-Richter, T(R)1 and T(H)3 regulatory cells. Nat. Immunol. 2: 671–672. D. J. Kasprowicz, K. Kellar, J. Pare, T. van Dyke, et al. 2006. Yeast zymosan, a 96. Owen, J. L., and M. Mohamadzadeh. 2013. Microbial activation of gut dendritic stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and cells and the control of mucosal immunity. J. Interferon Cytokine Res. 33: 619–631. immunological tolerance. J. Clin. Invest. 116: 916–928. 97. Lambrecht, B. N., J. B. Prins, and H. C. Hoogsteden. 2001. Lung dendritic cells 122. Gringhuis, S. I., J. den Dunnen, M. Litjens, B. van Het Hof, Y. van Kooyk, and host immunity to infection. Eur. Respir. J. 18: 692–704. and T. B. Geijtenbeek. 2007. C-type lectin DC-SIGN modulates toll-like 98. Naik, S., N. Bouladoux, C. Wilhelm, M. J. Molloy, R. Salcedo, W. Kastenmuller, receptor signaling via Raf-1 kinase-dependent acetylation of transcription C. Deming, M. Quinones, L. Koo, S. Conlan, et al. 2012. Compartmentalized factor NF-kappaB. Immunity 26: 605–616. control of skin immunity by resident commensals. Science 337: 1115–1119. 123. Cruz-Aguilar, M., A. I. Castillo-Rodal, A. Schcolnik-Cabrera, L. C. Bonifaz, 99. Menges, M., S. Ro¨ssner, C. Voigtla¨nder, H. Schindler, N. A. Kukutsch, G. Molina, and Y. Lo´pez-Vidal. 2016. TLR4 and DC-SIGN receptors recognized C. Bogdan, K. Erb, G. Schuler, and M. B. Lutz. 2002. Repetitive injections of Mycobacterium scrofulaceum promoting semi-activated phenotype on bone mar- dendritic cells matured with tumor necrosis factor alpha induce antigen-specific row dendritic cells. Tuberculosis (Edinb.) 99: 31–40. protection of mice from autoimmunity. J. Exp. Med. 195: 15–21. 124. Singh, N., A. Gurav, S. Sivaprakasam, E. Brady, R. Padia, H. Shi, M. Thangaraju, 100. Naik, S., N. Bouladoux, J. L. Linehan, S. J. Han, O. J. Harrison, C. Wilhelm, P. D. Prasad, S. Manicassamy, D. H. Munn, et al. 2014. Activation of Gpr109a, S. Conlan, S. Himmelfarb, A. L. Byrd, C. Deming, et al. 2015. Commensal- receptor for niacin and the commensal metabolite butyrate, suppresses colonic dendritic-cell interaction specifies a unique protective skin immune signature. inflammation and carcinogenesis. Immunity 40: 128–139. Nature 520: 104–108. 125. Liu, C., J. Wu, J. Zhu, C. Kuei, J. Yu, J. Shelton, S. W. Sutton, X. Li, S. J. Yun, 101. Akbari, O., R. H. DeKruyff, and D. T. Umetsu. 2001. Pulmonary dendritic cells T. Mirzadegan, et al. 2009. Lactate inhibits lipolysis in fat cells through activation producing IL-10 mediate tolerance induced by respiratory exposure to antigen. of an orphan G-protein-coupled receptor, GPR81. J. Biol. Chem. 284: 2811–2822. Nat. Immunol. 2: 725–731. 126. Ranganathan, P., A. Shanmugam, D. Swafford, A. Suryawanshi, P. Bhattacharjee, 102. Tsoumakidou, M., S. Tousa, M. Semitekolou, P. Panagiotou, A. Panagiotou, M. S. Hussein, P. A. Koni, P. D. Prasad, Z. B. Kurago, M. Thangaraju, et al. I. Morianos, E. Litsiou, A. I. Trochoutsou, M. Konstantinou, K. Potaris, et al. 2018. GPR81, a cell-surface receptor for lactate, regulates intestinal homeostasis 2014. Tolerogenic signaling by pulmonary CD1c+ dendritic cells induces reg- and protects mice from experimental colitis. J. Immunol. 200: 1781–1789. ulatory T cells in patients with chronic obstructive pulmonary disease by IL-27/ 127. Engering, A., T. B. Geijtenbeek, S. J. van Vliet, M. Wijers, E. van Liempt, IL-10/inducible costimulator ligand. J. Allergy Clin. Immunol. 134: 944–954.e8. N. Demaurex, A. Lanzavecchia, J. Fransen, C. G. Figdor, V. Piguet, and Y. van 103. Wawrzyniak, M., L. O’Mahony, and M. Akdis. 2017. Role of regulatory cells in Kooyk. 2002. The dendritic cell-specific adhesion receptor DC-SIGN internalizes Downloaded from oral tolerance. Allergy Asthma Immunol. Res. 9: 107–115. antigen for presentation to T cells. J. Immunol. 168: 2118–2126. 104. Quintana, F. J., G. Murugaiyan, M. F. Farez, M. Mitsdoerffer, A. M. Tukpah, 128. Conde, P., M. Rodriguez, W. van der Touw, A. Jimenez, M. Burns, J. Miller, E. J. Burns, and H. L. Weiner. 2010. An endogenous aryl hydrocarbon receptor M. Brahmachary, H. M. Chen, P. Boros, F. Rausell-Palamos, et al. 2015. DC- ligand acts on dendritic cells and T cells to suppress experimental autoimmune SIGN(+) macrophages control the induction of transplantation tolerance. Immu- encephalomyelitis. Proc. Natl. Acad. Sci. USA 107: 20768–20773. nity 42: 1143–1158. 105. Takenaka, M. C., L. P. Araujo, J. T. Maricato, V. M. Nascimento, 129. Geijtenbeek, T. B., J. den Dunnen, and S. I. Gringhuis. 2009. Pathogen recog- M. G. Guereschi, R. M. Rezende, F. J. Quintana, and A. S. Basso. 2016. Nor- nition by DC-SIGN shapes adaptive immunity. Future Microbiol. 4: 879–890.

epinephrine controls effector T cell differentiation through b2-adrenergic 130. Gringhuis, S. I., J. den Dunnen, M. Litjens, M. van der Vlist, and T. B. Geijtenbeek. http://www.jimmunol.org/ receptor-mediated inhibition of NF-kB and AP-1 in dendritic cells. J. Immunol. 2009. Carbohydrate-specific signaling through the DC-SIGN signalosome tai- 196: 637–644. lors immunity to Mycobacterium tuberculosis,HIV-1andHelicobacter pylori. 106. Atarashi, K., T. Tanoue, T. Shima, A. Imaoka, T. Kuwahara, Y. Momose, Nat. Immunol. 10: 1081–1088. G. Cheng, S. Yamasaki, T. Saito, Y. Ohba, et al. 2011. Induction of colonic 131. Steeghs, L., S. J. van Vliet, H. Uronen-Hansson, A. van Mourik, A. Engering, regulatory T cells by indigenous Clostridium species. Science 331: 337–341. M. Sanchez-Hernandez, N. Klein, R. Callard, J. P. van Putten, P. van der Ley, 107. Li, J., F. Ahmet, L. C. Sullivan, A. G. Brooks, S. J. Kent, R. De Rose, et al. 2006. Neisseria meningitidis expressing lgtB lipopolysaccharide targets DC- A. M. Salazar, C. Reis e Sousa, K. Shortman, M. H. Lahoud, et al. 2015. Anti- SIGN and modulates dendritic cell function. Cell. Microbiol. 8: 316–325. bodies targeting Clec9A promote strong humoral immunity without adjuvant in 132. Steinbrink, K., M. Wo¨lfl, H. Jonuleit, J. Knop, and A. H. Enk. 1997. Induction of mice and non-human primates. Eur. J. Immunol. 45: 854–864. tolerance by IL-10-treated dendritic cells. J. Immunol. 159: 4772–4780. 108. Coombes, J. L., K. R. Siddiqui, C. V. Arancibia-Ca´rcamo, J. Hall, C. M. Sun, 133. Torres-Aguilar, H., S. R. Aguilar-Ruiz, G. Gonza´lez-Pe´rez, R. Munguı´a, S. Bajan˜a, Y. Belkaid, and F. Powrie. 2007. A functionally specialized population of mucosal M. A. Meraz-Rı´os, and C. Sa´nchez-Torres. 2010. Tolerogenic dendritic cells

CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid- generated with different immunosuppressive cytokines induce antigen-specific by guest on September 28, 2021 dependent mechanism. J. Exp. Med. 204: 1757–1764. anergy and regulatory properties in memory CD4+ T cells. J. Immunol. 184: 109. Sun, C. M., J. A. Hall, R. B. Blank, N. Bouladoux, M. Oukka, J. R. Mora, and 1765–1775. Y. Belkaid. 2007. Small intestine lamina propria dendritic cells promote de novo 134. Mucida, D., and H. Cheroutre. 2007. TGFbeta and retinoic acid intersect in generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204: 1775–1785. immune-regulation. Cell Adhes. Migr. 1: 142–144. 110. Josefowicz, S. Z., R. E. Niec, H. Y. Kim, P. Treuting, T. Chinen, Y. Zheng, 135. Iliev, I. D., I. Spadoni, E. Mileti, G. Matteoli, A. Sonzogni, G. M. Sampietro, D. T. Umetsu, and A. Y. Rudensky. 2012. Extrathymically generated regulatory D. Foschi, F. Caprioli, G. Viale, and M. Rescigno. 2009. Human intestinal epi- T cells control mucosal TH2 inflammation. Nature 482: 395–399. thelial cells promote the differentiation of tolerogenic dendritic cells. Gut 58: 111. Nutsch, K., J. N. Chai, T. L. Ai, E. Russler-Germain, T. Feehley, C. R. Nagler, 1481–1489. and C. S. Hsieh. 2016. Rapid and efficient generation of regulatory T cells to 136. Iwata, M., A. Hirakiyama, Y. Eshima, H. Kagechika, C. Kato, and S. Y. Song. commensal antigens in the periphery. Cell Rep 17: 206–220. 2004. Retinoic acid imprints gut-homing specificity on T cells. Immunity 21: 527– 112. Price, J. D., C. Hotta-Iwamura, Y. Zhao, N. M. Beauchamp, and K. V. Tarbell. 538. 2015. DCIR2+ cDC2 DCs and Zbtb32 restore CD4+ T-cell tolerance and inhibit 137. Bakdash, G., L. T. Vogelpoel, T. M. van Capel, M. L. Kapsenberg, and E. C. de diabetes. Diabetes 64: 3521–3531. Jong. 2015. Retinoic acid primes human dendritic cells to induce gut-homing, 113. Weiner, H. L., A. P. da Cunha, F. Quintana, and H. Wu. 2011. Oral tolerance. IL-10-producing regulatory T cells. Mucosal Immunol. 8: 265–278. Immunol. Rev. 241: 241–259. 138. Chamorro, S., J. J. Garcı´a-Vallejo, W. W. Unger, R. J. Fernandes, S. C. Bruijns, 114. Medzhitov, R., and C. A. Janeway, Jr. 2002. Decoding the patterns of self and S. Laban, B. O. Roep, B. A. ’t Hart, and Y. van Kooyk. 2009. TLR triggering on nonself by the innate immune system. Science 296: 298–300. tolerogenic dendritic cells results in TLR2 up-regulation and a reduced proin- 115. Hacini-Rachinel, F., M. Gomez de Aguero,€ R. Kanjarawi, L. Moro-Sibilot, J. B. Le flammatory immune program. J. Immunol. 183: 2984–2994. Luduec, C. Macari, G. Boschetti, E. Bardel, P. Langella, B. Dubois, and 139. Collison, L. W., C. J. Workman, T. T. Kuo, K. Boyd, Y. Wang, D. Kaiserlian. 2018. Intestinal dendritic cell licensing through toll-like receptor 4 K. M. Vignali, R. Cross, D. Sehy, R. S. Blumberg, and D. A. Vignali. 2007. is required for oral tolerance in allergic contact dermatitis. J. Allergy Clin. Immunol. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. 141: 163–170. Nature 450: 566–569. 116. Manicassamy, S., R. Ravindran, J. Deng, H. Oluoch, T. L. Denning, S. P. Kasturi, 140. Nold, M. F., C. A. Nold-Petry, J. A. Zepp, B. E. Palmer, P. Bufler, and K. M. Rosenthal, B. D. Evavold, and B. Pulendran. 2009. Toll-like receptor C. A. Dinarello. 2010. IL-37 is a fundamental inhibitor of innate immunity. Nat. 2-dependent induction of vitamin A-metabolizing enzymes in dendritic cells pro- Immunol. 11: 1014–1022. motes T regulatory responses and inhibits autoimmunity. Nat. Med. 15: 401–409. 141. Luo, Y., X. Cai, S. Liu, S. Wang, C. A. Nold-Petry, M. F. Nold, P. Bufler, 117. Caparro´s, E., P. Munoz, E. Sierra-Filardi, D. Serrano-Go´mez, A. Puig-Kro¨ger, D. Norris, C. A. Dinarello, and M. Fujita. 2014. Suppression of antigen-specific J. L. Rodrı´guez-Ferna´ndez, M. Mellado, J. Sancho, M. Zubiaur, and A. L. Corbı´. adaptive immunity by IL-37 via induction of tolerogenic dendritic cells. Proc. Natl. 2006. DC-SIGN ligation on dendritic cells results in ERK and PI3K activation Acad. Sci. USA 111: 15178–15183. and modulates cytokine production. Blood 107: 3950–3958. 142. Hunter, C. A., and R. Kastelein. 2012. Interleukin-27: balancing protective and 118. Qian, C., H. An, Y. Yu, S. Liu, and X. Cao. 2007. TLR agonists induce regulatory pathological immunity. Immunity 37: 960–969. dendritic cells to recruit Th1 cells via preferential IP-10 secretion and inhibit Th1 143. Batten, M., J. Li, S. Yi, N. M. Kljavin, D. M. Danilenko, S. Lucas, J. Lee, F. J. de proliferation. Blood 109: 3308–3315. Sauvage, and N. Ghilardi. 2006. Interleukin 27 limits autoimmune encephalo- 119. Abdollahi-Roodsaz, S., L. A. Joosten, M. I. Koenders, I. Devesa, M. F. Roelofs, myelitis by suppressing the development of interleukin 17-producing T cells. Nat. T. R. Radstake, M. Heuvelmans-Jacobs, S. Akira, M. J. Nicklin, F. Ribeiro-Dias, Immunol. 7: 929–936. and W. B. van den Berg. 2008. Stimulation of TLR2 and TLR4 differentially 144. Fitzgerald, D. C., B. Ciric, T. Touil, H. Harle, J. Grammatikopolou, J. Das skews the balance of T cells in a mouse model of arthritis. J. Clin. Invest. 118: 205– Sarma, B. Gran, G. X. Zhang, and A. Rostami. 2007. Suppressive effect of IL-27 216. on encephalitogenic Th17 cells and the effector phase of experimental autoimmune 120. Depaolo, R. W., F. Tang, I. Kim, M. Han, N. Levin, N. Ciletti, A. Lin, D. Anderson, encephalomyelitis. J. Immunol. 179: 3268–3275. O. Schneewind, and B. Jabri. 2008. Toll-like receptor 6 drives differentiation of 145. Kastelein, R. A., C. A. Hunter, and D. J. Cua. 2007. Discovery and biology of tolerogenic dendritic cells and contributes to LcrV-mediated plague pathogenesis. IL-23 and IL-27: related but functionally distinct regulators of inflammation. Cell Host Microbe 4: 350–361. Annu. Rev. Immunol. 25: 221–242. The Journal of Immunology 743

146. Mascanfroni, I. D., A. Yeste, S. M. Vieira, E. J. Burns, B. Patel, I. Sloma, Y. Wu, 171. Suryawanshi, A., I. Manoharan, Y. Hong, D. Swafford, T. Majumdar, M. M. Taketo, L. Mayo, R. Ben-Hamo, S. Efroni, et al. 2013. IL-27 acts on DCs to suppress the B. Manicassamy, P. A. Koni, M. Thangaraju, Z. Sun, et al. 2015. Canonical wnt T cell response and autoimmunity by inducing expression of the immunoregula- signaling in dendritic cells regulates Th1/Th17 responses and suppresses autoimmune tory molecule CD39. Nat. Immunol. 14: 1054–1063. neuroinflammation. J. Immunol. 194: 3295–3304. 147. Quintana, F. J., A. S. Basso, A. H. Iglesias, T. Korn, M. F. Farez, E. Bettelli, 172. Reuter, S., H. Martin, H. Beckert, M. Bros, E. Montermann, C. Belz, A. Heinz, M. Caccamo, M. Oukka, and H. L. Weiner. 2008. Control of T(reg) and T(H)17 S. Ohngemach, U. Sahin, M. Stassen, et al. 2014. The Wnt/b-catenin pathway cell differentiation by the aryl hydrocarbon receptor. Nature 453: 65–71. attenuates experimental allergic airway disease. J. Immunol. 193: 485–495. 148. Nguyen, N. T., A. Kimura, T. Nakahama, I. Chinen, K. Masuda, K. Nohara, 173. Swafford, D., and S. Manicassamy. 2015. Wnt signaling in dendritic cells: its role Y. Fujii-Kuriyama, and T. Kishimoto. 2010. Aryl hydrocarbon receptor negatively in regulation of immunity and tolerance. Discov. Med. 19: 303–310. regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. 174. Swafford, D., A. Shanmugam, P. Ranganathan, M. S. Hussein, P. A. Koni, Proc. Natl. Acad. Sci. USA 107: 19961–19966. P. D. Prasad, M. Thangaraju, and S. Manicassamy. 2018. Canonical Wnt sig- 149. Yeste, A., M. C. Takenaka, I. D. Mascanfroni, M. Nadeau, J. E. Kenison, B. Patel, naling in CD11c+ APCs regulates microbiota-induced inflammation and immune A. M. Tukpah, J. A. Babon, M. DeNicola, S. C. Kent, et al. 2016. Tolerogenic cell homeostasis in the colon. J. Immunol. 200: 3259–3268. nanoparticles inhibit T cell-mediated autoimmunity through SOCS2. Sci. Signal. 175. Valencia, J., C. Herna´ndez-Lo´pez, V. G. Martı´nez, L. Hidalgo, A. G. Zapata, 9: ra61. A´. Vicente, A. Varas, and R. Sacedo´n. 2011. Wnt5a skews dendritic cell differ- 150. Takenaka, M. C., and F. J. Quintana. 2017. Tolerogenic dendritic cells. Semin. entiation to an unconventional phenotype with tolerogenic features. J. Immunol. Immunopathol. 39: 113–120. 187: 4129–4139. 151. Adorini, L., and G. Penna. 2009. Dendritic cell tolerogenicity: a key mechanism in 176. Hackstein, H., T. Taner, A. F. Zahorchak, A. E. Morelli, A. J. Logar, A. Gessner, immunomodulation by vitamin D receptor agonists. Hum. Immunol. 70: 345– and A. W. Thomson. 2003. Rapamycin inhibits IL-4--induced dendritic cell 352. maturation in vitro and dendritic cell mobilization and function in vivo. Blood 152. Barragan, M., M. Good, and J. K. Kolls. 2015. Regulation of dendritic cell 101: 4457–4463. function by vitamin D. Nutrients 7: 8127–8151. 177. Wang, Y., X. Du, J. Wei, L. Long, H. Tan, C. Guy, Y. Dhungana, C. Qian, 153. Bscheider, M., and E. C. Butcher. 2016. Vitamin D immunoregulation through G. Neale, Y. X. Fu, et al. 2019. LKB1 orchestrates dendritic cell metabolic qui- dendritic cells. Immunology 148: 227–236. escence and anti-tumor immunity. Cell Res. 29: 391–405. 154. Wang, Y., J. Zhu, and H. F. DeLuca. 2012. Where is the vitamin D receptor? 178. Kuchroo, V. K., A. C. Anderson, H. Waldner, M. Munder, E. Bettelli, and Arch. Biochem. Biophys. 523: 123–133. L. B. Nicholson. 2002. T cell response in experimental autoimmune encephalo- Downloaded from 155. Penna, G., S. Amuchastegui, N. Giarratana, K. C. Daniel, M. Vulcano, S. Sozzani, myelitis (EAE): role of self and cross-reactive antigens in shaping, tuning, and and L. Adorini. 2007. 1,25-Dihydroxyvitamin D3 selectively modulates tolero- regulating the autopathogenic T cell repertoire. Annu. Rev. Immunol. 20: 101–123. genic properties in myeloid but not plasmacytoid dendritic cells. J. Immunol. 178: 179. Simmons, S. B., E. R. Pierson, S. Y. Lee, and J. M. Goverman. 2013. Modeling 145–153. the heterogeneity of multiple sclerosis in animals. Trends Immunol. 34: 410–422. 156. Sochorova´, K., V. Budinsky´, D. Rozkova´, Z. Tobiasova´, S. Dusilova´-Sulkova´, 180. Mukherjee, G., A. Geliebter, J. Babad, P. Santamaria, D. V. Serreze, G. J. Freeman, R. Spı´sek, and J. Bart˚unkova´. 2009. Paricalcitol (19-nor-1,25-dihydroxyvitamin K. V. Tarbell, A. Sharpe, and T. P. DiLorenzo. 2013. DEC-205-mediated antigen D2) and calcitriol (1,25-dihydroxyvitamin D3) exert potent immunomodulatory targeting to steady-state dendritic cells induces deletion of diabetogenic CD8+ Tcells

effects on dendritic cells and inhibit induction of antigen-specific T cells. Clin. independently of PD-1 and PD-L1. Int. Immunol. 25: 651–660. http://www.jimmunol.org/ Immunol. 133: 69–77. 181. Wadwa, M., R. Klopfleisch, J. Buer, and A. M. Westendorf. 2016. Targeting 157. Sigmundsdottir, H., J. Pan, G. F. Debes, C. Alt, A. Habtezion, D. Soler, and antigens to Dec-205 on dendritic cells induces immune protection in experimental E. C. Butcher. 2007. DCs metabolize sunlight-induced vitamin D3 to ‘program’ colitis in mice. Eur. J. Microbiol. Immunol. (Bp.) 6: 1–8. T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 8: 285–293. 182. Spiering, R., B. Margry, C. Keijzer, C. Petzold, A. Hoek, J. Wagenaar-Hilbers, 158. Hewison, M., L. Freeman, S. V. Hughes, K. N. Evans, R. Bland, A. G. Eliopoulos, R. van der Zee, W. van Eden, K. Kretschmer, and F. Broere. 2015. DEC205+ M. D. Kilby, P. A. Moss, and R. Chakraverty. 2003. Differential regulation of dendritic cell-targeted tolerogenic vaccination promotes immune tolerance in ex- vitamin D receptor and its ligand in human monocyte-derived dendritic cells. perimental autoimmune arthritis. J. Immunol. 194: 4804–4813. J. Immunol. 170: 5382–5390. 183. Ettinger, M., I. K. Gratz, C. Gruber, C. Hauser-Kronberger, T. S. Johnson, 159. Enioutina, E. Y., D. Bareyan, and R. A. Daynes. 2009. TLR-induced local K. Mahnke, J. Thalhamer, H. Hintner, D. Peckl-Schmid, and J. W. Bauer. 2012. metabolism of vitamin D3 plays an important role in the diversification of adaptive Targeting of the hNC16A collagen domain to dendritic cells induces tolerance to immune responses. J. Immunol. 182: 4296–4305. human type XVII collagen. Exp. Dermatol. 21: 395–398.

160. Sze´les, L., G. Keresztes, D. To¨ro¨csik, Z. Balajthy, L. Krena´cs,S.Po´liska, 184. Maldonado, R. A., R. A. LaMothe, J. D. Ferrari, A. H. Zhang, R. J. Rossi, by guest on September 28, 2021 A. Steinmeyer, U. Zuegel, M. Pruenster, A. Rot, and L. Nagy. 2009. 1,25- P. N. Kolte, A. P. Griset, C. O’Neil, D. H. Altreuter, E. Browning, et al. 2015. dihydroxyvitamin D3 is an autonomous regulator of the transcriptional changes Polymeric synthetic nanoparticles for the induction of antigen-specific immuno- leading to a tolerogenic dendritic cell phenotype. J. Immunol. 182: 2074–2083. logical tolerance. Proc. Natl. Acad. Sci. USA 112: E156–E165. 161.Chu,C.C.,N.Ali,P.Karagiannis,P.DiMeglio,A.Skowera,L.Napolitano, 185. Clemente-Casares, X., J. Blanco, P. Ambalavanan, J. Yamanouchi, S. Singha, G. Barinaga, K. Grys, E. Sharif-Paghaleh, S. N. Karagiannis, et al. 2012. C. Fandos, S. Tsai, J. Wang, N. Garabatos, C. Izquierdo, et al. 2016. Expanding Resident CD141 (BDCA3)+ dendritic cells in human skin produce IL-10 and antigen-specific regulatory networks to treat autoimmunity. Nature 530: 434–440. induce regulatory T cells that suppress skin inflammation. J. Exp. Med. 209: 186. Getts, D. R., A. J. Martin, D. P. McCarthy, R. L. Terry, Z. N. Hunter, W. T. Yap, 935–945. M. T. Getts, M. Pleiss, X. Luo, N. J. King, et al. 2012. Microparticles bearing 162. Gonzalez-Rey, E., A. Chorny, A. Fernandez-Martin, D. Ganea, and M. Delgado. encephalitogenic peptides induce T-cell tolerance and ameliorate experimental 2006. Vasoactive intestinal peptide generates human tolerogenic dendritic cells autoimmune encephalomyelitis. Nat. Biotechnol. 30: 1217–1224. that induce CD4 and CD8 regulatory T cells. Blood 107: 3632–3638. 187. Miller, S. D., D. M. Turley, and J. R. Podojil. 2007. Antigen-specific tolerance 163. Remes Lenicov, F., C. Rodriguez Rodrigues, J. Sabatte´, M. Cabrini, C. Jancic, strategies for the prevention and treatment of autoimmune disease. Nat. Rev. M. Ostrowski, A. Merlotti, H. Gonzalez, A. Alonso, R. A. Pasqualini, et al. 2012. Immunol. 7: 665–677. Semen promotes the differentiation of tolerogenic dendritic cells. J. Immunol. 189: 188. Stead, S. O., S. J. P. McInnes, S. Kireta, P. D. Rose, S. Jesudason, D. Rojas- 4777–4786. Canales, D. Warther, F. Cunin, J. O. Durand, C. J. Drogemuller, et al. 2018. 164. Ano, Y., K. Ikado, K. Shindo, H. Koizumi, and D. Fujiwara. 2017. Identification Manipulating human dendritic cell phenotype and function with targeted porous of 14-dehydroergosterol as a novel anti-inflammatory compound inducing tol- silicon nanoparticles. Biomaterials 155: 92–102. erogenic dendritic cells. Sci. Rep. 7: 13903. 189. Sehgal, K., K. M. Dhodapkar, and M. V. Dhodapkar. 2014. Targeting human 165. Sugihara, Y., S. Ikushima, M. Miyake, T. Kirisako, Y. Yada, and D. Fujiwara. dendritic cells in situ to improve vaccines. Immunol. Lett. 162(1 Pt A): 59–67. 2018. Improvement of skin conditions by ingestion of Aspergillus kawachii (Koji) 190. Thomson, A. W., D. M. Metes, M. B. Ezzelarab, and D. Raı¨ch-Regue´. 2019. extract containing 14-dehydroergosterol in a randomized, double-blind, controlled Regulatory dendritic cells for human organ transplantation. Transplant. Rev. trial. Clin. Cosmet. Investig. Dermatol. 11: 115–124. (Orlando) 33: 130–136. 166. Hayashi, T., S. Yao, B. Crain, V. J. Promessi, L. Shyu, C. Sheng, M. Kang, 191. Hongo, D., X. Tang, X. Zhang, E. G. Engleman, and S. Strober. 2017. Tolero- H. B. Cottam, D. A. Carson, and M. Corr. 2015. Induction of tolerogenic den- genic interactions between CD8+ dendritic cells and NKT cells prevent rejection of dritic cells by a PEGylated TLR7 ligand for treatment of type 1 diabetes. PLoS One bone marrow and organ grafts. Blood 129: 1718–1728. 10: e0129867. 192. Luther, C., E. Adamopoulou, C. Stoeckle, V. Brucklacher-Waldert, D. Rosenkranz, 167. Zhou, Y., X. Leng, S. Luo, Z. Su, X. Luo, H. Guo, C. Mo, Q. Zou, Y. Liu, and L. Stoltze, S. Lauer, S. Poeschel, A. Melms, and E. Tolosa. 2009. Prednisolone Y. Wang. 2016. Tolerogenic dendritic cells generated with tofacitinib ameliorate treatment induces tolerogenic dendritic cells and a regulatory milieu in myasthenia experimental autoimmune encephalomyelitis through modulation of Th17/Treg gravis patients. J. Immunol. 183: 841–848. balance. J. Immunol. Res. 2016: 5021537. 193. Garcı´a-Gonza´lez, P. A., K. Schinnerling, A. Sepu´lveda-Gutie´rrez,J.Maggi,L.Hoyos, 168. Kwon, H. K., C. G. Lee, J. S. So, C. S. Chae, J. S. Hwang, A. Sahoo, J. H. Nam, R. A. Morales, A. M. Mehdi, H. J. Nel, L. Soto, B. Pesce, et al. 2016. Treatment with J. H. Rhee, K. C. Hwang, and S. H. Im. 2010. Generation of regulatory dendritic dexamethasone and monophosphoryl lipid A removes disease-associated transcrip- cells and CD4+Foxp3+ T cells by probiotics administration suppresses immune tional signatures in monocyte-derived dendritic cells from rheumatoid arthritis pa- disorders. Proc. Natl. Acad. Sci. USA 107: 2159–2164. tients and confers tolerogenic features. Front. Immunol. 7: 458. 169. Garcı´a-Gonza´lez, P., R. Morales, L. Hoyos, J. Maggi, J. Campos, B. Pesce, 194. Garcı´a-Gonza´lez, P., G. Ubilla-Olguı´n, D. Catala´n, K. Schinnerling, and D. Ga´rate, M. Larrondo, R. Gonza´lez, L. Soto, et al. 2013. A short protocol using J. C. Aguillo´n. 2016. Tolerogenic dendritic cells for reprogramming of lymphocyte dexamethasone and monophosphoryl lipid A generates tolerogenic dendritic cells responses in autoimmune diseases. Autoimmun. Rev. 15: 1071–1080. that display a potent migratory capacity to lymphoid chemokines. J. Transl. Med. 195. Kryczanowsky, F., V. Raker, E. Graulich, M. P. Domogalla, and K. Steinbrink. 2016. 11: 128. IL-10-modulated human dendritic cells for clinical use: identification of a stable and 170. Shan, M., M. Gentile, J. R. Yeiser, A. C. Walland, V. U. Bornstein, K. Chen, migratory subset with improved tolerogenic activity. J. Immunol. 197: 3607–3617. B. He, L. Cassis, A. Bigas, M. Cols, et al. 2013. Mucus enhances gut homeostasis 196. Kim, W. S., H. Kim, K. W. Kwon, S. H. Im, B. R. Lee, S. J. Ha, and S. J. Shin. and oral tolerance by delivering immunoregulatory signals. Science 342: 447–453. 2016. Cisplatin induces tolerogenic dendritic cells in response to TLR agonists via 744 BRIEF REVIEWS: NATURAL AND INDUCED TOLEROGENIC DENDRITIC CELLS

the abundant production of IL-10, thereby promoting Th2- and Tr1-biased T-cell in hypoxia down-modulate antigen uptake and change their chemokine expression immunity. Oncotarget 7: 33765–33782. profile. J. Leukoc. Biol. 84: 1472–1482. 197. Wu, A. A., V. Drake, H. S. Huang, S. Chiu, and L. Zheng. 2015. Reprogramming 201. Oussa, N. A., A. Dahmani, M. Gomis, M. Richaud, E. Andreev, A. R. Navab- the tumor microenvironment: tumor-induced immunosuppressive factors paralyze Daneshmand, J. Taillefer, C. Carli, S. Boulet, L. Sabbagh, et al. 2016. VEGF T cells. OncoImmunology 4: e1016700. requires the receptor NRP-1 to inhibit lipopolysaccharide-dependent dendritic cell 198. Jahchan, N. S., A. M. Mujal, J. L. Pollack, M. Binnewies, V. Sriram, L. Reyno, maturation. J. Immunol. 197: 3927–3935. and M. F. Krummel. 2019. Tuning the tumor myeloid microenvironment to fight 202. Oyama, T., S. Ran, T. Ishida, S. Nadaf, L. Kerr, D. P. Carbone, and cancer. Front. Immunol. 10: 1611. D. I. Gabrilovich. 1998. Vascular endothelial growth factor affects dendritic cell 199. Harimoto, H., M. Shimizu, Y. Nakagawa, K. Nakatsuka, A. Wakabayashi, maturation through the inhibition of nuclear factor-kappa B activation in hemo- C. Sakamoto, and H. Takahashi. 2013. Inactivation of tumor-specific CD8+ CTLs poietic progenitor cells. J. Immunol. 160: 1224–1232. by tumor-infiltrating tolerogenic dendritic cells. Immunol. Cell Biol. 91: 545–555. 203. Wculek, S. K., F. J. Cueto, A. M. Mujal, I. Melero, M. F. Krummel, and 200. Elia, A. R., P. Cappello, M. Puppo, T. Fraone, C. Vanni, A. Eva, T. Musso, D. Sancho. 2019. Dendritic cells in cancer immunology and immunotherapy. Nat. F. Novelli, L. Varesio, and M. Giovarelli. 2008. Human dendritic cells differentiated Rev. Immunol. 20: 7–24. Downloaded from http://www.jimmunol.org/ by guest on September 28, 2021