Conventional dendritic cells (cDC) control adaptive immunity by sensing damage- and pathogen-associated molecular patterns and then inducing defined differentiation programs in T cells. Nevertheless, in the absence of specific proimmunogenic innate signals, generally referred to as the steady state, cDC also activate T cells to induce specific functional fates. Consistent with the maintenance of homeostasis, such specific outcomes of T cell activation in the steady state include T cell clonal anergy, deletion, and conversion of peripheral regulatory T cells (pTregs). However, the robust induction of protolerogenic mechanisms must be reconciled with the initiation of autoimmune responses and cancer immunosurveillance that are also observed under homeostatic conditions. Here we review the diversity of fates and functions of T cells involved in the opposing immunogenic and tolerogenic processes induced in the steady state by the relevant mechanisms of systemic cDC present in murine peripheral lymphoid organs.

Dendritic cells (DC) consist of two main populations: conventional (cDC) and plasmacytoid (pDC) (1, 2). cDC are further divided into the type 1 (cDC1) and type 2 (cDC2) subsets based on their development and direct characterization of their expression of specific surface markers (3, 4). Human and murine cDC develop from progenitors in the bone marrow that disseminate through the blood to secondary lymphoid organs such as the spleen and lymph nodes (LNs) to form a network of systemic cDC that constantly interact with T cells (35). In addition, some systemic cDC whose progenitors originally seed peripheral nonlymphoid tissues, then migrate to lymphoid organs (6, 7). The systemic cDC are not directly exposed to extrinsic signals, in contrast to the cDC found at anatomical barriers and the associated lymphoid tissues that are exposed to microbiota and other environmental factors. Instead, systemic cDC, the focus of this review, constantly survey local and circulating Ags derived from parenchymal, interstitial, and other nonbarrier tissues. Furthermore, the systemic cDC can present Ags from pathogens during infections as well as those introduced by i.m. vaccinations (810) (Fig. 1). It is important to note that migratory cDC that originate at anatomical barriers also drain to certain specialized LNs and, in the case of some respiratory infections, to the spleen (7, 11). Such migratory cDC have key roles in the initiation of effector responses under proinflammatory conditions (1116).

Initially, cDC were proposed to function in vivo as “nature’s adjuvant” due to their potent abilities to prime T cells that were first identified in vitro (1719). However, such efficient priming of effector T cells by cDC in vivo requires sensing of additional signals relayed through specific pattern recognition receptors (2024). These signals result in a proimmunogenic process that enhances specific functions of cDC and is generally referred to as “maturation,” which results in altered expression of costimulatory molecules, MHCs, and cytokines, as well as other changes in cDC that facilitate enhanced effector T cell priming under proinflammatory conditions (22, 25, 26). Overall, the functional partnership between cDC and T cells is the sine qua non to effective adaptive immune responses.

In contrast to proinflammatory conditions, the term “steady state” refers to the absence of a specific inflammatory process (27, 28). The maintenance of the steady state further depends on undisturbed microanatomical architecture supported by stromal cells of the secondary lymphoid tissues that provide a framework for the interactions between cDC and T cells (8, 9, 27, 29, 30). The functions of cDC in the steady state were unclear at first, and an apparent lack of T cell priming in the absence of proinflammatory stimuli originally led to the designation of such cDC as “immature” immune bystanders (18, 31, 32). However, these conclusions were often based on the results of in vitro experiments using bone marrow–derived DC (BMDC). In contrast to BMDC, which only express appreciable amounts of MHC class II (MHC-II) and other costimulatory molecules upon specific stimulation, endogenous cDC in vivo constitutively express at least intermediate amounts of these molecules (6, 21, 3336). Subsequent experimental results established that in the steady state, endogenous cDC that are present in both the spleen and LNs can very efficiently activate Ag-specific T cells in vivo (3740). This functional competency may depend on specific mechanisms that operate intrinsically in cDC under homeostatic conditions and may also develop in response to not yet completely identified molecular ligands that are constitutively present in the steady state (26, 32, 38, 4146). Such emerging concepts of “homeostatic maturation” are also consistent with observations of multiple specific gene expression changes in some cDC under steady state conditions that are comparable in scope to those observed during TLR ligand-mediated maturation (26, 4144, 46). Although the specific mechanisms governing cDC functions in vivo under steady-state conditions following their initial development from bone marrow precursors are still being uncovered, it is clear that these processes result in phenotypically heterogeneous cDC populations present within lymphoid organs (41, 42, 46).

Instead of being primed for effector differentiation, T cells activated by cDC in the steady state rather acquire various tolerogenic properties, including a conversion into peripheral regulatory T cells (pTregs) (8, 22, 31, 32). Overall, the current model established by the cumulative experimental evidence collected by multiple investigators specifies tolerogenic roles of cDC in the steady state and contrasts these functions with the effector-priming capacity of cDC under proinflammatory conditions (6, 21, 23, 32, 36, 47) (Fig. 2). Nevertheless, such protolerogenic roles of systemic cDC need to be reconciled with the initiation of autoimmune T cell responses and cancer immunosurveillance also observed in the steady state (4855). The diversity of T cell responses is further accentuated by the recently uncovered induction of specific effector programs in T cells in the steady state, possibly reflecting functional heterogeneity of homeostatically matured cDC (56). Whereas the exact impacts on T cell responses made by different subpopulations of homeostatically matured cDC remain to be uncovered, such various roles of cDC may, at least to some extent, be orchestrated by the specific immunomodulatory molecules that are differentially expressed in distinct subpopulations of cDC (57). Overall, in the steady state, cDC are emerging as versatile sculptors of the functional repertoires of Ag-specific T cells that are directly relevant to multiple homeostatic and pathologic conditions.

In contrast to some in vitro experimental approaches such as those based on BMDC, the functional analysis of endogenous cDC in vivo is more difficult. Such specific challenges stem from the relatively small numbers of endogenous cDC found at the relevant anatomical locations in situ and are further compounded by the short lifespans of cDC and their high sensitivity to external stimulation (6, 27, 32). Some early attempts to bypass these challenges relied on injecting in vitro prepared BMDC back into the animals, but it is unclear to what extent the in vitro derived and manipulated cells correspond to the bona fide endogenous cDC (21, 58). However, these in vitro systems still have utility for translational research, especially in the development of DC-based immunotherapies for cancer (59, 60). Fortunately, the roles of cDC in the steady state could be established directly by multiple other lines of independent experimental evidence. These approaches, which have been specifically designed to observe the impact of endogenous cDC in situ on T cell responses in vivo, have fallen into two main categories: (1) a depletion of the cDC normally present under steady-state conditions and (2) an introduction of defined T cell Ags into such endogenous cDC (32, 61). Although the experimental scope of these approaches differed substantially, the results of these independent studies pointed to the role of cDC in maintaining immune homeostasis and tolerance (8, 32), as discussed below .

The experimental deletion resulting in the absence of cDC in vivo was achieved by specifically expressing diphtheria toxin receptor followed by diphtheria toxin treatment, expressing diphtheria toxin A subunit directly, or by nonspecific chemical depletion (6266). Such induced absence of cDC resulted in spontaneous autoimmune T cell activation and increased formation of effector T cells (65, 66), defects in induction of tolerance to systemic Ags (64), disturbance of oral tolerance after exposure to specific Ags (62), and increased susceptibility to colitis (63). As indicated above, these results found key roles for cDC in the maintenance of homeostasis, in agreement with other studies that employed transgenic expression of cytosolic ectopic Ags in cDC as well as earlier studies employing the direct delivery of defined Ags to cDC in vivo for measuring the impact of specific antigenic presentation by endogenous cDC in the steady state (37, 6770). Although the results of the studies using expression of ectopic Ags in the cytosol of all cDC showed the crucial role of cDC in inducing tolerance of self-reactive CD8+ T cells in the steady state, such models are limited to defined MHC-I restricted T cell epitopes (6870). In contrast, the introduction of defined exogenous Ags to cDC in vivo allowed for controlling the availability of specific Ags to both CD4+ and CD8+ T cells and have served as a versatile tool for studying T cell responses elicited by cDC in settings that mimic the uptake by cDC of apoptotic and other endogenous materials (32, 37, 40, 67, 7177).

To obtain their specific presentation by cDC, cognate T cell epitopes can be delivered to cDC directly by using modified mAbs specific for molecules expressed on the surfaces of cDC, and such methods have been broadly adopted over the past two decades, as reviewed elsewhere (78, 79). Particularly, the recombinant Abs that include T cell Ags as fusion proteins within their constant regions have proved to have multiple advantages. In contrast to a chemical conjugation of Ags to native Igs, fusion proteins avoid unintentional stoichiometric differences in the amounts of antigenic molecules and also minimize unintended contamination with endotoxin and other undesired molecules, rendering these recombinant Ag-delivery reagents ideal for basic science and clinical applications (32, 7880). Furthermore, in chimeric Abs, the original constant regions are replaced with the species-specific constant regions. In the case of the recombinant chimeric Abs used in experimental mice, the engineered murine IgG1 also had additional mutations minimizing their nonspecific binding to Fc receptors (37). In addition to full Igs, single-chain fragment V region and single-domain Ab have also been used for Ag delivery under multiple immune conditions (32, 8188).

The endocytic receptor DEC-205 (DEC205, CD205, LY75) remains as one of the key molecules targeted by recombinant Abs to deliver Ags to murine cDC (37, 67, 78). DEC-205 is expressed at high levels on cDC1 and mediates an efficient endocytic pathway allowing for robust processing and presentation of Ags from proteins binding to this lectin receptor (31, 78, 89, 90). Ags delivered through DEC-205 are presented in the context of both MHC-I and MHC-II without causing other perceivable changes to cDC in the steady state (37, 71, 78). Although DEC-205 also can be expressed in other immune cells, such as germinal center B cells, the expression of DEC-205 in B cells in vivo is considerably increased only after immunization using adjuvants or in vitro after activation with LPS, anti-CD40, and IL-4 (31, 91, 92). Overall, Ag delivery through DEC-205 and other cDC surface proteins has allowed a comprehensive analysis of the outcomes of T cell activation by cognate Ags presented by cDC in the steady state (3840, 78, 93). Such experimental strategies that combined Ag delivery methods with various genetic models of cDC have additionally broadened our understanding of the roles of specific cDC populations, particularly clarifying the tolerogenic functions of cDC1 in the steady state (32). Furthermore, the specific methods of targeted Ag delivery to cDC are promising avenues for various immunotherapeutic applications (7881).

Immune tolerance is indispensable for protecting self-tissues from autoimmune attack, and specific tolerogenic mechanisms also help to regulate the immune responses against infectious agents and cancer cells (32, 94, 95). The immune tolerance of T cells depends on thymic mechanisms that include clonal selection as well as the functions of thymically produced regulatory T cells (tTregs) (96). However, such tTregs can be overwhelmed by specific proinflammatory activation in the periphery. This underscores the risks conferred by autoreactive T cells that survive thymic negative selection yet remain responsive to Ags in the periphery because of the cross-reactive TCRs and the differing efficiencies of antigenic presentation in the thymus and in the periphery (32, 97106).

The peripherally induced mechanisms of tolerance that prevent the unwarranted priming of such autoreactive T cells include T cell anergy, T cell deletion, and a conversion of pTregs (8, 32, 107). The pTregs, which are de novo converted in response to specific self-antigens, bestow a dominant and long-lasting tolerance to peripheral Ags that can ameliorate various autoimmune responses, therefore crucially complementing the immunological tolerance first initiated in the thymus (8, 32, 102). The induction of systemic pTregs is efficiently mediated in the steady state by cDC with tolerogenic functions that belong to the Batf3-dependent cDC1, corresponding to previously defined CD8α+DEC-205+ cDC present both in LNs and in the spleen (1, 8, 31, 32, 38). These tolerogenic cDC1 are further distinguished by their high expression of B and T lymphocyte associated (BTLA) and constitute a majority of splenic cDC1 that are mostly considered to be “resident” cDC in the steady state (6, 9, 38). BTLA engages herpesvirus entry mediator (HVEM) in CD4+ T cells to modulate a CD5-dependent resistance of developing pTregs to effector-differentiating cytokines such as IL-4, IL-6, and IFN-γ (38, 102, 108, 109). Therefore, functions of the BTLA–HVEM–CD5 axis stabilize and promote the process of pTreg conversion that is initiated and further facilitated by other key molecules, such as TGF-β, retinoic acid (RA), IL-10, CD39, and others (42, 47, 57, 109116). In particular, RA, together with TGF-β, helps to promote induction of Foxp3 expression in pTregs, and β-catenin governs production of RA and TGF-β in some cDC (111, 117). Such cDC-mediated functions of TGF-β depend on the expression of TGF-β–activating integrin αvβ8 (118). In addition to these soluble mediators, other pathways dependent on the immunomodulatory properties of PD-L1/PD-1, CD80/CD86/CTLA-4, and B7h/ICOS signaling axes induce Foxp3 expression in developing pTregs as well as facilitate some other mechanisms of tolerance (57, 68, 119124).

Because pTregs are Ag specific, the availability of peripheral Ags is necessary for the generation of these cells. Apoptotic materials are a particularly abundant source of tissue-derived self-antigens, and cDC1 efficiently mediate the uptake, processing, and presentation of Ags from apoptotic materials (69, 72, 74, 75, 125127). BTLAhi cDC1 reside in the spleen and LNs and are ideally positioned to capture apoptotic and other materials that serve as a source of various systemic self-antigens for induction of tolerance (8, 9, 31, 69, 72, 73). Therefore, the de novo pTreg conversion can take place both in the spleen and in LNs (38, 56, 93). Furthermore, the induction of Ag-specific pTregs does not depend on specific receptors expressed in these cDC to mediate Ag uptake (32, 38, 93). However, the efficiency of such pTreg induction is diminished by the presence of high amounts of the specific Ag, especially if such Ags are presented to T cells also by other cDC that lack pTreg-inducing mechanisms (38, 93, 128, 129). Overall, in addition to the functions of the specific immunomodulatory mechanisms discussed above, the efficient conversion of pTregs requires only a moderate initial activation of T cells, also consistent with the diminished pTreg induction upon activation of cDC by specific proinflammatory stimuli (32, 93, 129).

In addition, some cDC2 can induce pTregs, especially in the intestines, expand the existing tTregs, and promote other forms of tolerance, such as T cell anergy and deletion (130132). However, in contrast to cDC1, cDC2 are generally less efficient at inducing mechanisms of tolerance. By using a diphtheria toxin receptor expression system in the intestines, the deletion of cDC2 did not exacerbate sensitivity to an inflammatory process, but a deletion of cDC1 exacerbated such inflammation (133). Furthermore, various experimental systems, including those based on Ag targeting in vivo, also revealed a superior ability of cDC1 to induce pTregs in comparison with cDC2 (32, 78).

In addition to the inherent tolerogenic functions of some systemic cDC in the steady state as outlined above and referred to as “natural” tolerogenic cDC (8), some microbiota-associated stimuli as well as certain key endogenous metabolites and vitamins can also induce tolerogenic functions in cDC (8, 134, 135). Conversely, a dysregulation of certain metabolic pathways such as adenosine deaminase may lead to autoimmune stimulation by cDC (136). Various types of cDC with induced tolerogenic functions, especially those that migrate to draining LNs, have important roles for maintaining homeostasis at multiple anatomical barriers exposed to the commensal microbiota and other environmental cues (8, 137143). Overall, induced tolerogenic functions complement the constitutive mechanisms of tolerance mediated by cDC with inherent tolerogenic roles (8).

Migratory cDC transport Ags from parenchymal tissues to the lymphoid organs in the steady state (144). However, systemic soluble Ags can be delivered to lymphoid organs directly (such as to the spleen via the bloodstream) for presentation by resident cDC (8, 9). Some migratory cDC isolated ex vivo can convert Tregs, but it remains unclear if such cDC induce tolerance in the steady state directly in vivo or if these cells ferry Ags from the peripheral tissues and pass them on to other tolerogenic cDC present in the lymphoid organs (8, 12, 42, 44, 128, 140, 145149). Furthermore, the Ags that trigger pTreg conversion are readily available in vivo to both migratory and resident cDC in the lymphoid organs (32, 38, 40). However, the de novo induction of such Ag-specific pTregs in the steady state is compromised in vivo in the absence of specific tolerogenic functions of BTLAhi cDC1 (32, 38). In addition, in the case of some tumors, certain cDC acquire regulatory functions within the tumor microenvironment and may migrate to lymphoid tissues to dampen antitumor responses (150153).

The functions of pTregs initially arising in response to systemically available self-antigens in the steady state confer dominant tolerance that prevents subsequently triggered organ-specific autoimmunity such as in experimental autoimmune encephalomyelitis, a model of multiple sclerosis, as well as other models of autoimmune diseases (32, 102). Such de novo induced tolerance is specifically perturbed in the absence of Hopx (homeodomain-only protein), a transcription cofactor required for the survival of pTregs under proinflammatory conditions (101, 102, 154). Therefore, the presence of Hopx or other factors that control either the initial conversion or the functions of pTregs is required for the ability of Ag-specific pTregs to restrain the subsequently triggered autoimmune process (101, 102, 155).

The induction of peripheral tolerance is not limited only to induction of mechanisms of tolerance in CD4+ T cells. By presenting endogenous Ags to both CD4+ and CD8+ T cells, cross-presenting cDC1 also help to maintain tolerance to self by directly deleting autoreactive CD8+ T cells (31, 156). Various recently identified mechanisms that govern the pathways of cross-presentation in cDC1 may therefore also contribute to these functions of cDC1 (157159). Overall, in addition to being considered as one of the key safeguards against autoimmune responses, the tolerogenic skewing of T cells in the periphery also provides a means for preventing transplant rejection (160).

The general model of immune system function postulates an immunogenic priming mediated by proinflammatory signals (21, 23, 28, 32, 36, 161163). Therefore, an induction of T cell tolerance in the steady state is critical for the prevention of subsequent autoimmune responses initiated when self-antigens are presented under proinflammatory conditions. However, such a preferential induction of tolerance in the steady state is not consistent with other experimental evidence indicating the induction of effector characteristics in CD4+ T cells under otherwise tolerizing conditions (4850, 164, 165). In addition, in the absence of sufficient immunoregulation, such T cells with effector characteristics may contribute to autoimmune responses (8, 50, 57, 166, 167). Furthermore, the mechanisms of cancer immunosurveillance postulated to constantly remove cancerous cells arising under homeostatic conditions are inconsistent with the predominantly tolerogenic outcomes of Ag-specific T cell activation under homeostatic conditions (51, 168). Similarly, productive antitumor responses can develop in the absence of major perturbations of homeostasis (5255, 169).

Overall, such an emerging diversity of T cell fates points to an underappreciated complexity of the biological processes mediated by cDC in the steady state. Recent results identified a process resulting in active programming by cDC of naive CD4+ T cells with specific epigenetic and transcriptional instructions leading to an acquisition of specific Th effector functions. Therefore, such “pre-effectors” activated in the steady state become poised for subsequent effector differentiation and, upon restimulation under nonskewing conditions in vitro or in vivo, readily express key factors such as IFN-γ that then can trigger expression of T-bet and possibly other effector master regulators (56). Pre-effectors were shown to contribute to initiation of the autoimmune responses and might possibly contribute to cancer immunosurveillance (56, 170).

In contrast to a binary model of effector versus regulatory fate determination, effector programming in the steady state occurs concurrently with the conversion into pTregs of T cells with the same Ag specificity (56). This dichotomous differentiation also fits a broader paradigm of how Th cell fates may diverge, resulting in different functional outcomes (171173). Overall, the functionally dichotomous effector and regulatory outcomes of CD4+ T cell activation in the steady state may increase the range, plasticity, and regulation of the subsequent immune responses.

The proliferative potential and gain of effector functions are also separable events in the differentiation of CD8+ T cells that depends on CD4+ T cell help (174). In the absence of such CD4+ T cell help, the initial proliferation of naive CD8+ T cells may lead to their deletional tolerance (175, 176). Therefore, divergent outcomes of CD4+ T cell activation in the steady state are also expected to directly affect CD8+ T cell responses. Overall, a regulation of CD4+ Th cells whose functions in the steady state can be kept in check by the correspondingly arising Ag-specific pTregs may help to better account for the observed complexities of T cell responses initiated in the steady state (56, 177).

In contrast to the well-defined mechanisms employed by cDC1 for inducing pTregs, the specific processes resulting in the dichotomous tolerogenic and effector outcomes in the steady state remain unclear. Both cDC1 and cDC2 can induce effector programming in the steady state, although such outcomes are enhanced by cDC2 that lack specific mechanisms needed for the efficient conversion of pTregs (8, 47, 56). Therefore, it is likely that a pre-effector T cell fate determination is independent of the specific type of T cell–activating cDC, whereas conversion into pTregs is facilitated by specific mechanisms present only in certain cDC1, as discussed above and as shown in (Fig. 3. Furthermore, one might speculate that the BTLAlo cDC1 excel in inducing pre-effectors in contrast to BTLAhi cDC1 that are known to efficiently convert pTregs as discussed above. In addition, the induction of pre-effectors may rely on other specific characteristics of some cDC, such as their expression of effector T cell master regulatory genes and specific metabolic reprogramming (157, 178180). In addition to the differences in processing and presentation of Ags to T cells, as well as the expression of specific immunomodulatory mechanisms within various cDC populations, the specific functions of such cDC subsets and even individual cDC may be determined by their exact anatomical localization within immune organs (9, 32, 39, 57, 181). In this regard, it is important to recognize that the specific anatomical organization of gut-draining LNs plays an important role in balancing the local tolerogenic and immunogenic responses (141). Other recent results also showed the role of the commensal microbiota in licensing the functions of cDC to directly prime some CD8+ T cell responses in the steady state (182).

Furthermore, various extrinsic factors may help determine the balance of tolerogenic and immunogenic T cell responses in the steady state. In addition to serving as a source of Ags, apoptotic materials that engage receptors such as CD36 may mitigate effector-inducing properties, whereas necrotic materials derived from injured cells and recognized by Clec9a (DNGR-1) promote proimmunogenic effects (183187). As already discussed earlier in the text and as recently reviewed (8), other extrinsic factors can modulate functions of specialized cDC present at specific anatomical locations by activating key pathways, including NF-κB, Wnt/β-catenin, and mammalian target of rapamycin. Future research will continue to clarify how such signaling pathways may contribute to orchestrating a balance between proimmunogenic and protolerogenic functions.

The plethora of diverse T cell fates initiated in the steady state underscores the complexities underlying the orchestration of immune responses. An improved understanding of such intricate mechanisms is also likely to open the door to designing more effective immunotherapeutic approaches against different types of autoimmunity and cancer. In this regard, it should be noted that various tools currently used in basic research to probe the crucial outcomes of interactions between cDC and T cells also have a potential translational value. Overall, the emerging understanding of the specific functions in the steady state of cDC and T cells along with their corresponding mechanisms is expanding the current scope of research in immunology.

Figures were created with Biorender.com.

This work was supported by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (R01AI113903).

Abbreviations used in this article

BMDC

bone marrow–derived dendritic cell

BTLA

B and T lymphocyte associated

cDC

conventional DC

cDC1

type 1 cDC

cDC2

type 2 cDC

DC

dendritic cell

LN

lymph node

MHC-II

MHC class II

pDC

plasmacytoid DC

pTreg

peripheral regulatory T cell

RA

retinoic acid

tTreg

thymically produced regulatory T cell

1.
Guilliams
M.
,
F.
Ginhoux
,
C.
Jakubzick
,
S. H.
Naik
,
N.
Onai
,
B. U.
Schraml
,
E.
Segura
,
R.
Tussiwand
,
S.
Yona
.
2014
.
Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny.
Nat. Rev. Immunol.
14
:
571
578
.
2.
Swiecki
M.
,
M.
Colonna
.
2015
.
The multifaceted biology of plasmacytoid dendritic cells.
Nat. Rev. Immunol.
15
:
471
485
.
3.
Durai
V.
,
K. M.
Murphy
.
2016
.
Functions of murine dendritic cells.
Immunity
45
:
719
736
.
4.
Guilliams
M.
,
C. A.
Dutertre
,
C. L.
Scott
,
N.
McGovern
,
D.
Sichien
,
S.
Chakarov
,
S.
Van Gassen
,
J.
Chen
,
M.
Poidinger
,
S.
De Prijck
, et al
2016
.
Unsupervised high-dimensional analysis aligns dendritic cells across tissues and species.
Immunity
45
:
669
684
.
5.
Collin
M.
,
V.
Bigley
.
2018
.
Human dendritic cell subsets: an update.
Immunology
154
:
3
20
.
6.
Cabeza-Cabrerizo
M.
,
A.
Cardoso
,
C. M.
Minutti
,
M.
Pereira da Costa
,
C.
Reis E Sousa
.
2021
.
Dendritic cells revisited.
Annu. Rev. Immunol.
39
:
131
166
.
7.
Sichien
D.
,
B. N.
Lambrecht
,
M.
Guilliams
,
C. L.
Scott
.
2017
.
Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues.
Mucosal Immunol.
10
:
831
844
.
8.
Iberg
C. A.
,
D.
Hawiger
.
2020
.
Natural and induced tolerogenic dendritic cells.
J. Immunol.
204
:
733
744
.
9.
Eisenbarth
S. C.
2019
.
Dendritic cell subsets in T cell programming: location dictates function.
Nat. Rev. Immunol.
19
:
89
103
.
10.
Langlet
C.
,
S.
Tamoutounour
,
S.
Henri
,
H.
Luche
,
L.
Ardouin
,
C.
Grégoire
,
B.
Malissen
,
M.
Guilliams
.
2012
.
CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization.
J. Immunol.
188
:
1751
1760
.
11.
Jenkins
M. M.
,
H.
Bachus
,
D.
Botta
,
M. D.
Schultz
,
A. F.
Rosenberg
,
B.
León
,
A.
Ballesteros-Tato
.
2021
.
Lung dendritic cells migrate to the spleen to prime long-lived TCF1hi memory CD8+ T cell precursors after influenza infection.
Sci. Immunol.
6
:
eabg6895
.
12.
Scheinecker
C.
,
R.
McHugh
,
E. M.
Shevach
,
R. N.
Germain
.
2002
.
Constitutive presentation of a natural tissue autoantigen exclusively by dendritic cells in the draining lymph node.
J. Exp. Med.
196
:
1079
1090
.
13.
Krishnaswamy
J. K.
,
S.
Alsén
,
U.
Yrlid
,
S. C.
Eisenbarth
,
A.
Williams
.
2018
.
Determination of T follicular helper cell fate by dendritic cells.
Front. Immunol.
9
:
2169
.
14.
Bajaña
S.
,
K.
Roach
,
S.
Turner
,
J.
Paul
,
S.
Kovats
.
2012
.
IRF4 promotes cutaneous dendritic cell migration to lymph nodes during homeostasis and inflammation.
J. Immunol.
189
:
3368
3377
.
15.
GeurtsvanKessel
C. H.
,
M. A.
Willart
,
L. S.
van Rijt
,
F.
Muskens
,
M.
Kool
,
C.
Baas
,
K.
Thielemans
,
C.
Bennett
,
B. E.
Clausen
,
H. C.
Hoogsteden
, et al
2008
.
Clearance of influenza virus from the lung depends on migratory langerin+CD11b but not plasmacytoid dendritic cells.
J. Exp. Med.
205
:
1621
1634
.
16.
Kim
T. S.
,
M. M.
Hufford
,
J.
Sun
,
Y.-X.
Fu
,
T. J.
Braciale
.
2010
.
Antigen persistence and the control of local T cell memory by migrant respiratory dendritic cells after acute virus infection.
J. Exp. Med.
207
:
1161
1172
.
17.
Steinman
R. M.
2007
.
Dendritic cells: understanding immunogenicity.
Eur. J. Immunol.
37
(
S1
,
Suppl 1
)
S53
S60
.
18.
Steinman
R. M.
1996
.
Dendritic cells and immune-based therapies.
Exp. Hematol.
24
:
859
862
.
19.
Steinman
R. M.
,
M. D.
Witmer
.
1978
.
Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice.
Proc. Natl. Acad. Sci. USA
75
:
5132
5136
.
20.
Coquerelle
C.
,
M.
Moser
.
2010
.
DC subsets in positive and negative regulation of immunity.
Immunol. Rev.
234
:
317
334
.
21.
Steinman
R. M.
2012
.
Decisions about dendritic cells: past, present, and future.
Annu. Rev. Immunol.
30
:
1
22
.
22.
Mellman
I.
2013
.
Dendritic cells: master regulators of the immune response.
Cancer Immunol. Res.
1
:
145
149
.
23.
Iwasaki
A.
,
R.
Medzhitov
.
2015
.
Control of adaptive immunity by the innate immune system.
Nat. Immunol.
16
:
343
353
.
24.
Janeway
C. A.
 Jr.
,
R.
Medzhitov
.
2002
.
Innate immune recognition.
Annu. Rev. Immunol.
20
:
197
216
.
25.
Schuler
G.
,
R. M.
Steinman
.
1985
.
Murine epidermal Langerhans cells mature into potent immunostimulatory dendritic cells in vitro.
J. Exp. Med.
161
:
526
546
.
26.
Dalod
M.
,
R.
Chelbi
,
B.
Malissen
,
T.
Lawrence
.
2014
.
Dendritic cell maturation: functional specialization through signaling specificity and transcriptional programming.
EMBO J.
33
:
1104
1116
.
27.
Steinman
R. M.
,
D.
Hawiger
,
K.
Liu
,
L.
Bonifaz
,
D.
Bonnyay
,
K.
Mahnke
,
T.
Iyoda
,
J.
Ravetch
,
M.
Dhodapkar
,
K.
Inaba
,
M.
Nussenzweig
.
2003
.
Dendritic cell function in vivo during the steady state: a role in peripheral tolerance.
Ann. N. Y. Acad. Sci.
987
:
15
25
.
28.
Pulendran
B.
2015
.
The varieties of immunological experience: of pathogens, stress, and dendritic cells.
Annu. Rev. Immunol.
33
:
563
606
.
29.
Zhang
M.
,
H.
Tang
,
Z.
Guo
,
H.
An
,
X.
Zhu
,
W.
Song
,
J.
Guo
,
X.
Huang
,
T.
Chen
,
J.
Wang
,
X.
Cao
.
2004
.
Splenic stroma drives mature dendritic cells to differentiate into regulatory dendritic cells. [Published erratum appears in 2014 Nat. Immunol. 15: 1090.]
Nat. Immunol.
5
:
1124
1133
.
30.
Saxena
V.
,
L.
Li
,
C.
Paluskievicz
,
V.
Kasinath
,
A.
Bean
,
R.
Abdi
,
C. M.
Jewell
,
J. S.
Bromberg
.
2019
.
Role of lymph node stroma and microenvironment in T cell tolerance.
Immunol. Rev.
292
:
9
23
.
31.
Steinman
R. M.
,
D.
Hawiger
,
M. C.
Nussenzweig
.
2003
.
Tolerogenic dendritic cells.
Annu. Rev. Immunol.
21
:
685
711
.
32.
Iberg
C. A.
,
A.
Jones
,
D.
Hawiger
.
2017
.
Dendritic cells as inducers of peripheral tolerance.
Trends Immunol.
38
:
793
804
.
33.
Sallusto
F.
,
A.
Lanzavecchia
.
1994
.
Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha.
J. Exp. Med.
179
:
1109
1118
.
34.
Winzler
C.
,
P.
Rovere
,
M.
Rescigno
,
F.
Granucci
,
G.
Penna
,
L.
Adorini
,
V. S.
Zimmermann
,
J.
Davoust
,
P.
Ricciardi-Castagnoli
.
1997
.
Maturation stages of mouse dendritic cells in growth factor-dependent long-term cultures.
J. Exp. Med.
185
:
317
328
.
35.
De Smedt
T.
,
B.
Pajak
,
E.
Muraille
,
L.
Lespagnard
,
E.
Heinen
,
P.
De Baetselier
,
J.
Urbain
,
O.
Leo
,
M.
Moser
.
1996
.
Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo.
J. Exp. Med.
184
:
1413
1424
.
36.
Merad
M.
,
P.
Sathe
,
J.
Helft
,
J.
Miller
,
A.
Mortha
.
2013
.
The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting.
Annu. Rev. Immunol.
31
:
563
604
.
37.
Hawiger
D.
,
K.
Inaba
,
Y.
Dorsett
,
M.
Guo
,
K.
Mahnke
,
M.
Rivera
,
J. V.
Ravetch
,
R. M.
Steinman
,
M. C.
Nussenzweig
.
2001
.
Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo.
J. Exp. Med.
194
:
769
779
.
38.
Jones
A.
,
J.
Bourque
,
L.
Kuehm
,
A.
Opejin
,
R. M.
Teague
,
C.
Gross
,
D.
Hawiger
.
2016
.
Immunomodulatory functions of BTLA and HVEM govern induction of extrathymic regulatory T cells and tolerance by dendritic cells.
Immunity
45
:
1066
1077
.
39.
Dudziak
D.
,
A. O.
Kamphorst
,
G. F.
Heidkamp
,
V. R.
Buchholz
,
C.
Trumpfheller
,
S.
Yamazaki
,
C.
Cheong
,
K.
Liu
,
H. W.
Lee
,
C. G.
Park
, et al
2007
.
Differential antigen processing by dendritic cell subsets in vivo.
Science
315
:
107
111
.
40.
Idoyaga
J.
,
C.
Fiorese
,
L.
Zbytnuik
,
A.
Lubkin
,
J.
Miller
,
B.
Malissen
,
D.
Mucida
,
M.
Merad
,
R. M.
Steinman
.
2013
.
Specialized role of migratory dendritic cells in peripheral tolerance induction.
J. Clin. Invest.
123
:
844
854
.
41.
Vander Lugt
B.
,
J.
Riddell
,
A. A.
Khan
,
J. A.
Hackney
,
J.
Lesch
,
J.
DeVoss
,
M. T.
Weirauch
,
H.
Singh
,
I.
Mellman
.
2017
.
Transcriptional determinants of tolerogenic and immunogenic states during dendritic cell maturation.
J. Cell Biol.
216
:
779
792
.
42.
Ardouin
L.
,
H.
Luche
,
R.
Chelbi
,
S.
Carpentier
,
A.
Shawket
,
F.
Montanana Sanchis
,
C.
Santa Maria
,
P.
Grenot
,
Y.
Alexandre
,
C.
Grégoire
, et al
2016
.
Broad and largely concordant molecular changes characterize tolerogenic and immunogenic dendritic cell maturation in thymus and periphery.
Immunity
45
:
305
318
.
43.
Jiang
A.
,
O.
Bloom
,
S.
Ono
,
W.
Cui
,
J.
Unternaehrer
,
S.
Jiang
,
J. A.
Whitney
,
J.
Connolly
,
J.
Banchereau
,
I.
Mellman
.
2007
.
Disruption of E-cadherin-mediated adhesion induces a functionally distinct pathway of dendritic cell maturation.
Immunity
27
:
610
624
.
44.
Baratin
M.
,
C.
Foray
,
O.
Demaria
,
M.
Habbeddine
,
E.
Pollet
,
J.
Maurizio
,
C.
Verthuy
,
S.
Davanture
,
H.
Azukizawa
,
A.
Flores-Langarica
, et al
2015
.
Homeostatic NF-κB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance.
Immunity
42
:
627
639
.
45.
Hammer
G. E.
,
A.
Ma
.
2013
.
Molecular control of steady-state dendritic cell maturation and immune homeostasis.
Annu. Rev. Immunol.
31
:
743
791
.
46.
Manh
T. P.
,
Y.
Alexandre
,
T.
Baranek
,
K.
Crozat
,
M.
Dalod
.
2013
.
Plasmacytoid, conventional, and monocyte-derived dendritic cells undergo a profound and convergent genetic reprogramming during their maturation.
Eur. J. Immunol.
43
:
1706
1715
.
47.
Yin
X.
,
S.
Chen
,
S. C.
Eisenbarth
.
2021
.
Dendritic cell regulation of T helper cells.
Annu. Rev. Immunol.
39
:
759
790
.
48.
Huang
C. T.
,
D. L.
Huso
,
Z.
Lu
,
T.
Wang
,
G.
Zhou
,
E. P.
Kennedy
,
C. G.
Drake
,
D. J.
Morgan
,
L. A.
Sherman
,
A. D.
Higgins
, et al
2003
.
CD4+ T cells pass through an effector phase during the process of in vivo tolerance induction.
J. Immunol.
170
:
3945
3953
.
49.
Long
M.
,
A. M.
Slaiby
,
S.
Wu
,
A. T.
Hagymasi
,
M. A.
Mihalyo
,
S.
Bandyopadhyay
,
A. T.
Vella
,
A. J.
Adler
.
2007
.
Histone acetylation at the Ifng promoter in tolerized CD4 cells is associated with increased IFN-γ expression during subsequent immunization to the same antigen.
J. Immunol.
179
:
5669
5677
.
50.
Kawabe
T.
,
D.
Jankovic
,
S.
Kawabe
,
Y.
Huang
,
P. H.
Lee
,
H.
Yamane
,
J.
Zhu
,
A.
Sher
,
R. N.
Germain
,
W. E.
Paul
.
2017
.
Memory-phenotype CD4+ T cells spontaneously generated under steady-state conditions exert innate TH1-like effector function.
Sci. Immunol.
2
:
eaam9304
.
51.
Dunn
G. P.
,
A. T.
Bruce
,
H.
Ikeda
,
L. J.
Old
,
R. D.
Schreiber
.
2002
.
Cancer immunoediting: from immunosurveillance to tumor escape.
Nat. Immunol.
3
:
991
998
.
52.
Alspach
E.
,
D. M.
Lussier
,
A. P.
Miceli
,
I.
Kizhvatov
,
M.
DuPage
,
A. M.
Luoma
,
W.
Meng
,
C. F.
Lichti
,
E.
Esaulova
,
A. N.
Vomund
, et al
2019
.
MHC-II neoantigens shape tumour immunity and response to immunotherapy.
Nature
574
:
696
701
.
53.
Ferris
S. T.
,
V.
Durai
,
R.
Wu
,
D. J.
Theisen
,
J. P.
Ward
,
M. D.
Bern
,
J. T.
Davidson
IV
,
P.
Bagadia
,
T.
Liu
,
C. G.
Briseño
, et al
2020
.
cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity.
Nature
584
:
624
629
.
54.
Mattiuz
R.
,
C.
Brousse
,
M.
Ambrosini
,
J.-C.
Cancel
,
G.
Bessou
,
J.
Mussard
,
A.
Sanlaville
,
C.
Caux
,
N.
Bendriss-Vermare
,
J.
Valladeau-Guilemond
, et al
2021
.
Type 1 conventional dendritic cells and interferons are required for spontaneous CD4+ and CD8+ T-cell protective responses to breast cancer.
Clin. Transl. Immunology
10
:
e1305
.
55.
Fuertes
M. B.
,
A. K.
Kacha
,
J.
Kline
,
S.-R.
Woo
,
D. M.
Kranz
,
K. M.
Murphy
,
T. F.
Gajewski
.
2011
.
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells.
J. Exp. Med.
208
:
2005
2016
.
56.
Opejin
A.
,
A.
Surnov
,
Z.
Misulovin
,
M.
Pherson
,
C.
Gross
,
C. A.
Iberg
,
I.
Fallahee
,
J.
Bourque
,
D.
Dorsett
,
D.
Hawiger
.
2020
.
A two-step process of effector programming governs CD4+ T cell fate determination induced by antigenic activation in the steady state.
Cell Rep.
33
:
108424
.
57.
Bourque
J.
,
D.
Hawiger
.
2018
.
Immunomodulatory bonds of the partnership between dendritic cells and T cells.
Crit. Rev. Immunol.
38
:
379
401
.
58.
Helft
J.
,
J.
Böttcher
,
P.
Chakravarty
,
S.
Zelenay
,
J.
Huotari
,
B. U.
Schraml
,
D.
Goubau
,
C.
Reis e Sousa
.
2015
.
GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells.
Immunity
42
:
1197
1211
.
59.
Sabado
R. L.
,
S.
Balan
,
N.
Bhardwaj
.
2017
.
Dendritic cell-based immunotherapy.
Cell Res.
27
:
74
95
.
60.
Patente
T. A.
,
M. P.
Pinho
,
A. A.
Oliveira
,
G. C. M.
Evangelista
,
P. C.
Bergami-Santos
,
J. A. M.
Barbuto
.
2019
.
Human dendritic cells: their heterogeneity and clinical application potential in cancer immunotherapy.
Front. Immunol.
9
:
3176
.
61.
Anderson
III
D. A.
,
C.-A.
Dutertre
,
F.
Ginhoux
,
K. M.
Murphy
.
2021
.
Genetic models of human and mouse dendritic cell development and function.
Nat. Rev. Immunol.
21
:
101
115
.
62.
Esterházy
D.
,
J.
Loschko
,
M.
London
,
V.
Jove
,
T. Y.
Oliveira
,
D.
Mucida
.
2016
.
Classical dendritic cells are required for dietary antigen-mediated induction of peripheral Treg cells and tolerance.
Nat. Immunol.
17
:
545
555
.
63.
Ohta
T.
,
M.
Sugiyama
,
H.
Hemmi
,
C.
Yamazaki
,
S.
Okura
,
I.
Sasaki
,
Y.
Fukuda
,
T.
Orimo
,
K. J.
Ishii
,
K.
Hoshino
, et al
2016
.
Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis.
Sci. Rep.
6
:
23505
.
64.
Wang
L.
,
Z.
Li
,
B.
Ciric
,
F.
Safavi
,
G. X.
Zhang
,
A.
Rostami
.
2016
.
Selective depletion of CD11c+ CD11b+ dendritic cells partially abrogates tolerogenic effects of intravenous MOG in murine EAE.
Eur. J. Immunol.
46
:
2454
2466
.
65.
Ohnmacht
C.
,
A.
Pullner
,
S. B.
King
,
I.
Drexler
,
S.
Meier
,
T.
Brocker
,
D.
Voehringer
.
2009
.
Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity.
J. Exp. Med.
206
:
549
559
.
66.
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+ conventional dendritic cells impacts the regulation of T-cell immunity and homeostasis in vivo.
Proc. Natl. Acad. Sci. USA
109
:
11288
11293
.
67.
Hawiger
D.
,
R. F.
Masilamani
,
E.
Bettelli
,
V. K.
Kuchroo
,
M. C.
Nussenzweig
.
2004
.
Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo.
Immunity
20
:
695
705
.
68.
Probst
H. C.
,
K.
McCoy
,
T.
Okazaki
,
T.
Honjo
,
M.
van den Broek
.
2005
.
Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4.
Nat. Immunol.
6
:
280
286
.
69.
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
,
W. R.
Heath
.
2002
.
The CD8α+ dendritic cell is responsible for inducing peripheral self-tolerance to tissue-associated antigens.
J. Exp. Med.
196
:
1099
1104
.
70.
Probst
H. C.
,
J.
Lagnel
,
G.
Kollias
,
M.
van den Broek
.
2003
.
Inducible transgenic mice reveal resting dendritic cells as potent inducers of CD8+ T cell tolerance.
Immunity
18
:
713
720
.
71.
Bonifaz
L.
,
D.
Bonnyay
,
K.
Mahnke
,
M.
Rivera
,
M. C.
Nussenzweig
,
R. M.
Steinman
.
2002
.
Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance.
J. Exp. Med.
196
:
1627
1638
.
72.
Iyoda
T.
,
S.
Shimoyama
,
K.
Liu
,
Y.
Omatsu
,
Y.
Akiyama
,
Y.
Maeda
,
K.
Takahara
,
R. M.
Steinman
,
K.
Inaba
.
2002
.
The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo.
J. Exp. Med.
195
:
1289
1302
.
73.
Albert
M. L.
,
S. F.
Pearce
,
L. M.
Francisco
,
B.
Sauter
,
P.
Roy
,
R. L.
Silverstein
,
N.
Bhardwaj
.
1998
.
Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes.
J. Exp. Med.
188
:
1359
1368
.
74.
Kurts
C.
,
M.
Cannarile
,
I.
Klebba
,
T.
Brocker
.
2001
.
Dendritic cells are sufficient to cross-present self-antigens to CD8 T cells in vivo.
J. Immunol.
166
:
1439
1442
.
75.
Luckashenak
N.
,
S.
Schroeder
,
K.
Endt
,
D.
Schmidt
,
K.
Mahnke
,
M. F.
Bachmann
,
P.
Marconi
,
C. A.
Deeg
,
T.
Brocker
.
2008
.
Constitutive crosspresentation of tissue antigens by dendritic cells controls CD8+ T cell tolerance in vivo.
Immunity
28
:
521
532
.
76.
Cao
L.
,
X.
Shi
,
H.
Chang
,
Q.
Zhang
,
Y.
He
.
2015
.
pH-Dependent recognition of apoptotic and necrotic cells by the human dendritic cell receptor DEC205.
Proc. Natl. Acad. Sci. USA
112
:
7237
7242
.
77.
Shrimpton
R. E.
,
M.
Butler
,
A.-S.
Morel
,
E.
Eren
,
S. S.
Hue
,
M. A.
Ritter
.
2009
.
CD205 (DEC-205): a recognition receptor for apoptotic and necrotic self.
Mol. Immunol.
46
:
1229
1239
.
78.
Iberg
C. A.
,
D.
Hawiger
.
2019
.
Advancing immunomodulation by in vivo antigen delivery to DEC-205 and other cell surface molecules using recombinant chimeric antibodies.
Int. Immunopharmacol.
73
:
575
580
.
79.
Iberg
C. A.
,
D.
Hawiger
.
2020
.
Targeting dendritic cells with antigen-delivering antibodies for amelioration of autoimmunity in animal models of multiple sclerosis and other autoimmune diseases.
Antibodies (Basel)
9
:
23
.
80.
Bourque
J.
,
D.
Hawiger
.
2021
.
Current and future immunotherapies for multiple sclerosis.
Mo. Med.
118
:
334
339
.
81.
Lehmann
C. H.
,
L.
Heger
,
G. F.
Heidkamp
,
A.
Baranska
,
J. J.
Lühr
,
A.
Hoffmann
,
D.
Dudziak
.
2016
.
Direct delivery of antigens to dendritic cells via antibodies specific for endocytic receptors as a promising strategy for future therapies.
Vaccines (Basel)
4
:
8
.
82.
Woodham
A. W.
,
R. W.
Cheloha
,
J.
Ling
,
M.
Rashidian
,
S. C.
Kolifrath
,
M.
Mesyngier
,
J. N.
Duarte
,
J. M.
Bader
,
J. G.
Skeate
,
D. M.
Da Silva
, et al
2018
.
Nanobody-antigen conjugates elicit HPV-specific antitumor immune responses.
Cancer Immunol. Res.
6
:
870
880
.
83.
Fang
T.
,
C. H. M. J.
Van Elssen
,
J. N.
Duarte
,
J. S.
Guzman
,
J. S.
Chahal
,
J.
Ling
,
H. L.
Ploegh
.
2017
.
Targeted antigen delivery by an anti-class II MHC VHH elicits focused αMUC1(Tn) immunity.
Chem. Sci. (Camb.)
8
:
5591
5597
.
84.
Duarte
J. N.
,
J. J.
Cragnolini
,
L. K.
Swee
,
A. M.
Bilate
,
J.
Bader
,
J. R.
Ingram
,
A.
Rashidfarrokhi
,
T.
Fang
,
A.
Schiepers
,
L.
Hanke
,
H. L.
Ploegh
.
2016
.
Generation of immunity against pathogens via single-domain antibody-antigen constructs.
J. Immunol.
197
:
4838
4847
.
85.
Pishesha
N.
,
T.
Harmand
,
L. Y.
Smeding
,
W.
Ma
,
L. S.
Ludwig
,
R.
Janssen
,
A.
Islam
,
Y. J.
Xie
,
T.
Fang
,
N.
McCaul
, et al
2021
.
Induction of antigen-specific tolerance by nanobody-antigen adducts that target class-II major histocompatibility complexes.
Nat. Biomed. Eng.
5
:
1389
1401
.
86.
Ring
S.
,
M.
Maas
,
D. M.
Nettelbeck
,
A. H.
Enk
,
K.
Mahnke
.
2013
.
Targeting of autoantigens to DEC205+ dendritic cells in vivo suppresses experimental allergic encephalomyelitis in mice.
J. Immunol.
191
:
2938
2947
.
87.
Johnson
T. S.
,
K.
Mahnke
,
V.
Storn
,
K.
Schönfeld
,
S.
Ring
,
D. M.
Nettelbeck
,
H. J.
Haisma
,
F.
Le Gall
,
R. E.
Kontermann
,
A. H.
Enk
.
2008
.
Inhibition of melanoma growth by targeting of antigen to dendritic cells via an anti-DEC-205 single-chain fragment variable molecule.
Clin. Cancer Res.
14
:
8169
8177
.
88.
Ahmad
Z. A.
,
S. K.
Yeap
,
A. M.
Ali
,
W. Y.
Ho
,
N. B.
Alitheen
,
M.
Hamid
.
2012
.
scFv antibody: principles and clinical application.
Clin. Dev. Immunol.
2012
:
980250
.
89.
Jiang
W.
,
W. J.
Swiggard
,
C.
Heufler
,
M.
Peng
,
A.
Mirza
,
R. M.
Steinman
,
M. C.
Nussenzweig
.
1995
.
The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing.
Nature
375
:
151
155
.
90.
Mahnke
K.
,
M.
Guo
,
S.
Lee
,
H.
Sepulveda
,
S. L.
Swain
,
M.
Nussenzweig
,
R. M.
Steinman
.
2000
.
The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments.
J. Cell Biol.
151
:
673
684
.
91.
Pasqual
G.
,
A.
Angelini
,
G. D.
Victora
.
2015
.
Triggering positive selection of germinal center B cells by antigen targeting to DEC-205.
Methods Mol. Biol.
1291
:
125
134
.
92.
Kamphorst
A. O.
,
P.
Guermonprez
,
D.
Dudziak
,
M. C.
Nussenzweig
.
2010
.
Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes.
J. Immunol.
185
:
3426
3435
.
93.
Joffre
O. P.
,
D.
Sancho
,
S.
Zelenay
,
A. M.
Keller
,
C.
Reis e Sousa
.
2010
.
Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A.
Eur. J. Immunol.
40
:
1255
1265
.
94.
Veiga-Parga
T.
,
S.
Sehrawat
,
B. T.
Rouse
.
2013
.
Role of regulatory T cells during virus infection.
Immunol. Rev.
255
:
182
196
.
95.
Plitas
G.
,
A. Y.
Rudensky
.
2020
.
Regulatory T cells in cancer.
Annu. Rev. Cancer Biol.
4
:
459
477
.
96.
Klein
L.
,
E. A.
Robey
,
C. S.
Hsieh
.
2019
.
Central CD4+ T cell tolerance: deletion versus regulatory T cell differentiation.
Nat. Rev. Immunol.
19
:
7
18
.
97.
Ooi
J. D.
,
J.
Petersen
,
Y. H.
Tan
,
M.
Huynh
,
Z. J.
Willett
,
S. H.
Ramarathinam
,
P. J.
Eggenhuizen
,
K. L.
Loh
,
K. A.
Watson
,
P. Y.
Gan
, et al
2017
.
Dominant protection from HLA-linked autoimmunity by antigen-specific regulatory T cells.
Nature
545
:
243
247
.
98.
Enouz
S.
,
L.
Carrié
,
D.
Merkler
,
M. J.
Bevan
,
D.
Zehn
.
2012
.
Autoreactive T cells bypass negative selection and respond to self-antigen stimulation during infection.
J. Exp. Med.
209
:
1769
1779
.
99.
Zehn
D.
,
M. J.
Bevan
.
2006
.
T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and cause autoimmunity.
Immunity
25
:
261
270
.
100.
Koehli
S.
,
D.
Naeher
,
V.
Galati-Fournier
,
D.
Zehn
,
E.
Palmer
.
2014
.
Optimal T-cell receptor affinity for inducing autoimmunity.
Proc. Natl. Acad. Sci. USA
111
:
17248
17253
.
101.
Jones
A.
,
A.
Opejin
,
J. G.
Henderson
,
C.
Gross
,
R.
Jain
,
J. A.
Epstein
,
R. A.
Flavell
,
D.
Hawiger
.
2015
.
Peripherally induced tolerance depends on peripheral regulatory T cells that require Hopx to inhibit intrinsic IL-2 expression.
J. Immunol.
195
:
1489
1497
.
102.
Jones
A.
,
D.
Hawiger
.
2017
.
Peripherally induced regulatory T cells: recruited protectors of the central nervous system against autoimmune neuroinflammation.
Front. Immunol.
8
:
532
.
103.
Anderson
A. C.
,
L. B.
Nicholson
,
K. L.
Legge
,
V.
Turchin
,
H.
Zaghouani
,
V. K.
Kuchroo
.
2000
.
High frequency of autoreactive myelin proteolipid protein-specific T cells in the periphery of naive mice: mechanisms of selection of the self-reactive repertoire.
J. Exp. Med.
191
:
761
770
.
104.
Bouneaud
C.
,
P.
Kourilsky
,
P.
Bousso
.
2000
.
Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion.
Immunity
13
:
829
840
.
105.
Nelson
R. W.
,
D.
Beisang
,
N. J.
Tubo
,
T.
Dileepan
,
D. L.
Wiesner
,
K.
Nielsen
,
M.
Wüthrich
,
B. S.
Klein
,
D. I.
Kotov
,
J. A.
Spanier
, et al
2015
.
T cell receptor cross-reactivity between similar foreign and self peptides influences naive cell population size and autoimmunity. [Published erratum appears in 2015 Immunity 42: 1212–1213.]
Immunity
42
:
95
107
.
106.
Richards
D. M.
,
B.
Kyewski
,
M.
Feuerer
.
2016
.
Re-examining the nature and function of self-reactive T cells.
Trends Immunol.
37
:
114
125
.
107.
Kalekar
L. A.
,
D. L.
Mueller
.
2017
.
Relationship between CD4 regulatory T cells and anergy in vivo.
J. Immunol.
198
:
2527
2533
.
108.
Henderson
J. G.
,
A.
Opejin
,
A.
Jones
,
C.
Gross
,
D.
Hawiger
.
2015
.
CD5 instructs extrathymic regulatory T cell development in response to self and tolerizing antigens.
Immunity
42
:
471
483
.
109.
Bourque
J.
,
D.
Hawiger
.
2019
.
The BTLA–HVEM–CD5 immunoregulatory axis – an instructive mechanism governing pTreg cell differentiation.
Front. Immunol.
10
:
1163
.
110.
Munn
D. H.
,
M. D.
Sharma
,
J. R.
Lee
,
K. G.
Jhaver
,
T. S.
Johnson
,
D. B.
Keskin
,
B.
Marshall
,
P.
Chandler
,
S. J.
Antonia
,
R.
Burgess
, et al
2002
.
Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase.
Science
297
:
1867
1870
.
111.
Mucida
D.
,
Y.
Park
,
G.
Kim
,
O.
Turovskaya
,
I.
Scott
,
M.
Kronenberg
,
H.
Cheroutre
.
2007
.
Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid.
Science
317
:
256
260
.
112.
Coombes
J. L.
,
K. R.
Siddiqui
,
C. V.
Arancibia-Cárcamo
,
J.
Hall
,
C. M.
Sun
,
Y.
Belkaid
,
F.
Powrie
.
2007
.
A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β and retinoic acid-dependent mechanism.
J. Exp. Med.
204
:
1757
1764
.
113.
Sun
C. M.
,
J. A.
Hall
,
R. B.
Blank
,
N.
Bouladoux
,
M.
Oukka
,
J. R.
Mora
,
Y.
Belkaid
.
2007
.
Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid.
J. Exp. Med.
204
:
1775
1785
.
114.
Manicassamy
S.
,
R.
Ravindran
,
J.
Deng
,
H.
Oluoch
,
T. L.
Denning
,
S. P.
Kasturi
,
K. M.
Rosenthal
,
B. D.
Evavold
,
B.
Pulendran
.
2009
.
Toll-like receptor 2-dependent induction of vitamin A-metabolizing enzymes in dendritic cells promotes T regulatory responses and inhibits autoimmunity.
Nat. Med.
15
:
401
409
.
115.
Li
M. O.
,
R. A.
Flavell
.
2008
.
Contextual regulation of inflammation: a duet by transforming growth factor-β and interleukin-10.
Immunity
28
:
468
476
.
116.
Mascanfroni
I. D.
,
A.
Yeste
,
S. M.
Vieira
,
E. J.
Burns
,
B.
Patel
,
I.
Sloma
,
Y.
Wu
,
L.
Mayo
,
R.
Ben-Hamo
,
S.
Efroni
, et al
2013
.
IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39.
Nat. Immunol.
14
:
1054
1063
.
117.
Manicassamy
S.
,
B.
Reizis
,
R.
Ravindran
,
H.
Nakaya
,
R. M.
Salazar-Gonzalez
,
Y. C.
Wang
,
B.
Pulendran
.
2010
.
Activation of β-catenin in dendritic cells regulates immunity versus tolerance in the intestine.
Science
329
:
849
853
.
118.
Travis
M. A.
,
B.
Reizis
,
A. C.
Melton
,
E.
Masteller
,
Q.
Tang
,
J. M.
Proctor
,
Y.
Wang
,
X.
Bernstein
,
X.
Huang
,
L. F.
Reichardt
, et al
2007
.
Loss of integrin αvβ8 on dendritic cells causes autoimmunity and colitis in mice.
Nature
449
:
361
365
.
119.
Wang
L.
,
K.
Pino-Lagos
,
V. C.
de Vries
,
I.
Guleria
,
M. H.
Sayegh
,
R. J.
Noelle
.
2008
.
Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3+CD4+ regulatory T cells.
Proc. Natl. Acad. Sci. USA
105
:
9331
9336
.
120.
Wing
K.
,
Y.
Onishi
,
P.
Prieto-Martin
,
T.
Yamaguchi
,
M.
Miyara
,
Z.
Fehervari
,
T.
Nomura
,
S.
Sakaguchi
.
2008
.
CTLA-4 control over Foxp3+ regulatory T cell function.
Science
322
:
271
275
.
121.
Francisco
L. M.
,
V. H.
Salinas
,
K. E.
Brown
,
V. K.
Vanguri
,
G. J.
Freeman
,
V. K.
Kuchroo
,
A. H.
Sharpe
.
2009
.
PD-L1 regulates the development, maintenance, and function of induced regulatory T cells.
J. Exp. Med.
206
:
3015
3029
.
122.
Fife
B. T.
,
K. E.
Pauken
,
T. N.
Eagar
,
T.
Obu
,
J.
Wu
,
Q.
Tang
,
M.
Azuma
,
M. F.
Krummel
,
J. A.
Bluestone
.
2009
.
Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal.
Nat. Immunol.
10
:
1185
1192
.
123.
Akbari
O.
,
G. J.
Freeman
,
E. H.
Meyer
,
E. A.
Greenfield
,
T. T.
Chang
,
A. H.
Sharpe
,
G.
Berry
,
R. H.
DeKruyff
,
D. T.
Umetsu
.
2002
.
Antigen-specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit allergen-induced airway hyperreactivity.
Nat. Med.
8
:
1024
1032
.
124.
Yogev
N.
,
F.
Frommer
,
D.
Lukas
,
K.
Kautz-Neu
,
K.
Karram
,
D.
Ielo
,
E.
von Stebut
,
H. C.
Probst
,
M.
van den Broek
,
D.
Riethmacher
, et al
2012
.
Dendritic cells ameliorate autoimmunity in the CNS by controlling the homeostasis of PD-1 receptor+ regulatory T cells.
Immunity
37
:
264
275
.
125.
den Haan
J. M.
,
S. M.
Lehar
,
M. J.
Bevan
.
2000
.
CD8+ but not CD8 dendritic cells cross-prime cytotoxic T cells in vivo.
J. Exp. Med.
192
:
1685
1696
.
126.
Huang
F. P.
,
N.
Platt
,
M.
Wykes
,
J. R.
Major
,
T. J.
Powell
,
C. D.
Jenkins
,
G. G.
MacPherson
.
2000
.
A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes.
J. Exp. Med.
191
:
435
444
.
127.
Perry
J. S. A.
,
E. V.
Russler-Germain
,
Y. W.
Zhou
,
W.
Purtha
,
M. L.
Cooper
,
J.
Choi
,
M. A.
Schroeder
,
V.
Salazar
,
T.
Egawa
,
B. C.
Lee
, et al
2018
.
Transfer of cell-surface antigens by scavenger receptor CD36 promotes thymic regulatory T cell receptor repertoire development and allo-tolerance. [Published erratum appears in 2018 Immunity 48: 1271.]
Immunity
48
:
923
936.e4
.
128.
Vitali
C.
,
F.
Mingozzi
,
A.
Broggi
,
S.
Barresi
,
F.
Zolezzi
,
J.
Bayry
,
G.
Raimondi
,
I.
Zanoni
,
F.
Granucci
.
2012
.
Migratory, and not lymphoid-resident, dendritic cells maintain peripheral self-tolerance and prevent autoimmunity via induction of iTreg cells.
Blood
120
:
1237
1245
.
129.
Kretschmer
K.
,
I.
Apostolou
,
D.
Hawiger
,
K.
Khazaie
,
M. C.
Nussenzweig
,
H.
von Boehmer
.
2005
.
Inducing and expanding regulatory T cell populations by foreign antigen.
Nat. Immunol.
6
:
1219
1227
.
130.
Nutsch
K.
,
J. N.
Chai
,
T. L.
Ai
,
E.
Russler-Germain
,
T.
Feehley
,
C. R.
Nagler
,
C. S.
Hsieh
.
2016
.
Rapid and efficient generation of regulatory T cells to commensal antigens in the periphery.
Cell Rep.
17
:
206
220
.
131.
Price
J. D.
,
C.
Hotta-Iwamura
,
Y.
Zhao
,
N. M.
Beauchamp
,
K. V.
Tarbell
.
2015
.
DCIR2+ cDC2 DCs and Zbtb32 restore CD4+ T-cell tolerance and inhibit diabetes.
Diabetes
64
:
3521
3531
.
132.
Yamazaki
S.
,
D.
Dudziak
,
G. F.
Heidkamp
,
C.
Fiorese
,
A. J.
Bonito
,
K.
Inaba
,
M. C.
Nussenzweig
,
R. M.
Steinman
.
2008
.
CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells.
J. Immunol.
181
:
6923
6933
.
133.
Muzaki
A. R.
,
P.
Tetlak
,
J.
Sheng
,
S. C.
Loh
,
Y. A.
Setiagani
,
M.
Poidinger
,
F.
Zolezzi
,
K.
Karjalainen
,
C.
Ruedl
.
2016
.
Intestinal CD103+CD11b dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells.
Mucosal Immunol.
9
:
336
351
.
134.
Anderson
A. E.
,
D. J.
Swan
,
B. L.
Sayers
,
R. A.
Harry
,
A. M.
Patterson
,
A.
von Delwig
,
J. H.
Robinson
,
J. D.
Isaacs
,
C. M.
Hilkens
.
2009
.
LPS activation is required for migratory activity and antigen presentation by tolerogenic dendritic cells.
J. Leukoc. Biol.
85
:
243
250
.
135.
Zhou
F.
,
B.
Ciric
,
G. X.
Zhang
,
A.
Rostami
.
2014
.
Immunotherapy using lipopolysaccharide-stimulated bone marrow-derived dendritic cells to treat experimental autoimmune encephalomyelitis.
Clin. Exp. Immunol.
178
:
447
458
.
136.
Ghaemi Oskouie
F.
,
A.
Shameli
,
A.
Yang
,
M. D.
Desrosiers
,
A. D.
Mucsi
,
M. R.
Blackburn
,
Y.
Yang
,
P.
Santamaria
,
Y.
Shi
.
2011
.
High levels of adenosine deaminase on dendritic cells promote autoreactive T cell activation and diabetes in nonobese diabetic mice.
J. Immunol.
186
:
6798
6806
.
137.
Manicassamy
S.
,
B.
Pulendran
.
2011
.
Dendritic cell control of tolerogenic responses.
Immunol. Rev.
241
:
206
227
.
138.
Arpaia
N.
,
C.
Campbell
,
X.
Fan
,
S.
Dikiy
,
J.
van der Veeken
,
P.
deRoos
,
H.
Liu
,
J. R.
Cross
,
K.
Pfeffer
,
P. J.
Coffer
,
A. Y.
Rudensky
.
2013
.
Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation.
Nature
504
:
451
455
.
139.
Cummings
R. J.
,
G.
Barbet
,
G.
Bongers
,
B. M.
Hartmann
,
K.
Gettler
,
L.
Muniz
,
G. C.
Furtado
,
J.
Cho
,
S. A.
Lira
,
J. M.
Blander
.
2016
.
Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs.
Nature
539
:
565
569
.
140.
Guilliams
M.
,
K.
Crozat
,
S.
Henri
,
S.
Tamoutounour
,
P.
Grenot
,
E.
Devilard
,
B.
de Bovis
,
L.
Alexopoulou
,
M.
Dalod
,
B.
Malissen
.
2010
.
Skin-draining lymph nodes contain dermis-derived CD103 dendritic cells that constitutively produce retinoic acid and induce Foxp3+ regulatory T cells.
Blood
115
:
1958
1968
.
141.
Esterházy
D.
,
M. C. C.
Canesso
,
L.
Mesin
,
P. A.
Muller
,
T. B. R.
de Castro
,
A.
Lockhart
,
M.
ElJalby
,
A. M. C.
Faria
,
D.
Mucida
.
2019
.
Compartmentalized gut lymph node drainage dictates adaptive immune responses.
Nature
569
:
126
130
.
142.
Russler-Germain
E. V.
,
J.
Yi
,
S.
Young
,
K.
Nutsch
,
H. S.
Wong
,
T. L.
Ai
,
J. N.
Chai
,
V.
Durai
,
D. H.
Kaplan
,
R. N.
Germain
, et al
2021
.
Gut Helicobacter presentation by multiple dendritic cell subsets enables context-specific regulatory T cell generation.
eLife
10
:
e54792
.
143.
Kashem
S. W.
,
M.
Haniffa
,
D. H.
Kaplan
.
2017
.
Antigen-presenting cells in the skin.
Annu. Rev. Immunol.
35
:
469
499
.
144.
Randolph
G. J.
,
J.
Ochando
,
S.
Partida-Sánchez
.
2008
.
Migration of dendritic cell subsets and their precursors.
Annu. Rev. Immunol.
26
:
293
316
.
145.
Azukizawa
H.
,
A.
Döhler
,
N.
Kanazawa
,
A.
Nayak
,
M.
Lipp
,
B.
Malissen
,
I.
Autenrieth
,
I.
Katayama
,
M.
Riemann
,
F.
Weih
, et al
2011
.
Steady state migratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction of antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells.
Eur. J. Immunol.
41
:
1420
1434
.
146.
Gurevich
I.
,
T.
Feferman
,
I.
Milo
,
O.
Tal
,
O.
Golani
,
I.
Drexler
,
G.
Shakhar
.
2017
.
Active dissemination of cellular antigens by DCs facilitates CD8+ T-cell priming in lymph nodes.
Eur. J. Immunol.
47
:
1802
1818
.
147.
Allan
R. S.
,
J.
Waithman
,
S.
Bedoui
,
C. M.
Jones
,
J. A.
Villadangos
,
Y.
Zhan
,
A. M.
Lew
,
K.
Shortman
,
W. R.
Heath
,
F. R.
Carbone
.
2006
.
Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming.
Immunity
25
:
153
162
.
148.
Inaba
K.
,
S.
Turley
,
F.
Yamaide
,
T.
Iyoda
,
K.
Mahnke
,
M.
Inaba
,
M.
Pack
,
M.
Subklewe
,
B.
Sauter
,
D.
Sheff
, et al
1998
.
Efficient presentation of phagocytosed cellular fragments on the major histocompatibility complex class II products of dendritic cells.
J. Exp. Med.
188
:
2163
2173
.
149.
Ruhland
M. K.
,
E. W.
Roberts
,
E.
Cai
,
A. M.
Mujal
,
K.
Marchuk
,
C.
Beppler
,
D.
Nam
,
N. K.
Serwas
,
M.
Binnewies
,
M. F.
Krummel
.
2020
.
Visualizing synaptic transfer of tumor antigens among dendritic cells.
Cancer Cell
37
:
786
799.e5
.
150.
Harimoto
H.
,
M.
Shimizu
,
Y.
Nakagawa
,
K.
Nakatsuka
,
A.
Wakabayashi
,
C.
Sakamoto
,
H.
Takahashi
.
2013
.
Inactivation of tumor-specific CD8+ CTLs by tumor-infiltrating tolerogenic dendritic cells.
Immunol. Cell Biol.
91
:
545
555
.
151.
Wang
Y.
,
X.
Du
,
J.
Wei
,
L.
Long
,
H.
Tan
,
C.
Guy
,
Y.
Dhungana
,
C.
Qian
,
G.
Neale
,
Y. X.
Fu
, et al
2019
.
LKB1 orchestrates dendritic cell metabolic quiescence and anti-tumor immunity.
Cell Res.
29
:
391
405
.
152.
Maier
B.
,
A. M.
Leader
,
S. T.
Chen
,
N.
Tung
,
C.
Chang
,
J.
LeBerichel
,
A.
Chudnovskiy
,
S.
Maskey
,
L.
Walker
,
J. P.
Finnigan
, et al
2020
.
A conserved dendritic-cell regulatory program limits antitumour immunity. [Published erratum appears in 2020 Nature 582: E17.]
Nature
580
:
257
262
.
153.
Ghiringhelli
F.
,
P. E.
Puig
,
S.
Roux
,
A.
Parcellier
,
E.
Schmitt
,
E.
Solary
,
G.
Kroemer
,
F.
Martin
,
B.
Chauffert
,
L.
Zitvogel
.
2005
.
Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation.
J. Exp. Med.
202
:
919
929
.
154.
Hawiger
D.
,
Y. Y.
Wan
,
E. E.
Eynon
,
R. A.
Flavell
.
2010
.
The transcription cofactor Hopx is required for regulatory T cell function in dendritic cell-mediated peripheral T cell unresponsiveness.
Nat. Immunol.
11
:
962
968
.
155.
Josefowicz
S. Z.
,
R. E.
Niec
,
H. Y.
Kim
,
P.
Treuting
,
T.
Chinen
,
Y.
Zheng
,
D. T.
Umetsu
,
A. Y.
Rudensky
.
2012
.
Extrathymically generated regulatory T cells control mucosal TH2 inflammation.
Nature
482
:
395
399
.
156.
Kurts
C.
,
J. F.
Miller
,
R. M.
Subramaniam
,
F. R.
Carbone
,
W. R.
Heath
.
1998
.
Major histocompatibility complex class I-restricted cross-presentation is biased towards high dose antigens and those released during cellular destruction.
J. Exp. Med.
188
:
409
414
.
157.
Du
X.
,
J.
Wen
,
Y.
Wang
,
P. W. F.
Karmaus
,
A.
Khatamian
,
H.
Tan
,
Y.
Li
,
C.
Guy
,
T. M.
Nguyen
,
Y.
Dhungana
, et al
2018
.
Hippo/Mst signalling couples metabolic state and immune function of CD8α+ dendritic cells.
Nature
558
:
141
145
.
158.
Theisen
D. J.
,
J. T.
Davidson
IV
,
C. G.
Briseño
,
M.
Gargaro
,
E. J.
Lauron
,
Q.
Wang
,
P.
Desai
,
V.
Durai
,
P.
Bagadia
,
J. R.
Brickner
, et al
2018
.
WDFY4 is required for cross-presentation in response to viral and tumor antigens.
Science
362
:
694
699
.
159.
Kretzer
N. M.
,
D. J.
Theisen
,
R.
Tussiwand
,
C. G.
Briseño
,
G. E.
Grajales-Reyes
,
X.
Wu
,
V.
Durai
,
J.
Albring
,
P.
Bagadia
,
T. L.
Murphy
,
K. M.
Murphy
.
2016
.
RAB43 facilitates cross-presentation of cell-associated antigens by CD8α+ dendritic cells.
J. Exp. Med.
213
:
2871
2883
.
160.
Thomson
A. W.
,
D. M.
Metes
,
M. B.
Ezzelarab
,
D.
Raïch-Regué
.
2019
.
Regulatory dendritic cells for human organ transplantation.
Transplant. Rev. (Orlando)
33
:
130
136
.
161.
Segura
E.
,
S.
Amigorena
.
2013
.
Inflammatory dendritic cells in mice and humans.
Trends Immunol.
34
:
440
445
.
162.
Zelenay
S.
,
C.
Reis e Sousa
.
2013
.
Adaptive immunity after cell death.
Trends Immunol.
34
:
329
335
.
163.
Yatim
N.
,
S.
Cullen
,
M. L.
Albert
.
2017
.
Dying cells actively regulate adaptive immune responses.
Nat. Rev. Immunol.
17
:
262
275
.
164.
Gossel
G.
,
T.
Hogan
,
D.
Cownden
,
B.
Seddon
,
A. J.
Yates
.
2017
.
Memory CD4 T cell subsets are kinetically heterogeneous and replenished from naive T cells at high levels.
eLife
6
:
e23013
.
165.
Vokali
E.
,
S. S.
Yu
,
S.
Hirosue
,
M.
Rinçon-Restrepo
,
F.
V Duraes
,
S.
Scherer
,
P.
Corthésy-Henrioud
,
W. W.
Kilarski
,
A.
Mondino
,
D.
Zehn
, et al
2020
.
Lymphatic endothelial cells prime naïve CD8+ T cells into memory cells under steady-state conditions.
Nat. Commun.
11
:
538
.
166.
ElTanbouly
M. A.
,
Y.
Zhao
,
E.
Nowak
,
J.
Li
,
E.
Schaafsma
,
I.
Le Mercier
,
S.
Ceeraz
,
J. L.
Lines
,
C.
Peng
,
C.
Carriere
, et al
2020
.
VISTA is a checkpoint regulator for naïve T cell quiescence and peripheral tolerance.
Science
367
:
eaay0524
.
167.
Sharpe
A. H.
,
K. E.
Pauken
.
2018
.
The diverse functions of the PD1 inhibitory pathway.
Nat. Rev. Immunol.
18
:
153
167
.
168.
Dunn
G. P.
,
L. J.
Old
,
R. D.
Schreiber
.
2004
.
The three Es of cancer immunoediting.
Annu. Rev. Immunol.
22
:
329
360
.
169.
Sedlacek
A. L.
,
T. P.
Younker
,
Y. J.
Zhou
,
L.
Borghesi
,
T.
Shcheglova
,
I. I.
Mandoiu
,
R. J.
Binder
.
2019
.
CD91 on dendritic cells governs immunosurveillance of nascent, emerging tumors.
JCI Insight
4
:
e127239
.
170.
Surnov
A.
,
D.
Hawiger
.
2021
.
The formation of pre-effectors in the steady state opens a new perspective for cancer immunosurveillance.
Oncotarget
12
:
1318
1320
.
171.
Wu
C.
,
N.
Yosef
,
T.
Thalhamer
,
C.
Zhu
,
S.
Xiao
,
Y.
Kishi
,
A.
Regev
,
V. K.
Kuchroo
.
2013
.
Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1.
Nature
496
:
513
517
.
172.
Wu
C.
,
Z.
Chen
,
S.
Xiao
,
T.
Thalhamer
,
A.
Madi
,
T.
Han
,
V.
Kuchroo
.
2018
.
SGK1 governs the reciprocal development of Th17 and regulatory T cells.
Cell Rep.
22
:
653
665
.
173.
DiToro
D.
,
C. J.
Winstead
,
D.
Pham
,
S.
Witte
,
R.
Andargachew
,
J. R.
Singer
,
C. G.
Wilson
,
C. L.
Zindl
,
R. J.
Luther
,
D. J.
Silberger
, et al
2018
.
Differential IL-2 expression defines developmental fates of follicular versus nonfollicular helper T cells.
Science
361
:
eaao2933
.
174.
Hernández
J.
,
S.
Aung
,
K.
Marquardt
,
L. A.
Sherman
.
2002
.
Uncoupling of proliferative potential and gain of effector function by CD8+ T cells responding to self-antigens.
J. Exp. Med.
196
:
323
333
.
175.
Hernandez
J.
,
S.
Aung
,
W. L.
Redmond
,
L. A.
Sherman
.
2001
.
Phenotypic and functional analysis of CD8+ T cells undergoing peripheral deletion in response to cross-presentation of self-antigen.
J. Exp. Med.
194
:
707
717
.
176.
Parish
I. A.
,
J.
Waithman
,
G. M.
Davey
,
G. T.
Belz
,
J. D.
Mintern
,
C.
Kurts
,
R. M.
Sutherland
,
F. R.
Carbone
,
W. R.
Heath
.
2009
.
Tissue destruction caused by cytotoxic T lymphocytes induces deletional tolerance.
Proc. Natl. Acad. Sci. USA
106
:
3901
3906
.
177.
Wong
H. S.
,
K.
Park
,
A.
Gola
,
A. P.
Baptista
,
C. H.
Miller
,
D.
Deep
,
M.
Lou
,
L. F.
Boyd
,
A. Y.
Rudensky
,
P. A.
Savage
, et al
2021
.
A local regulatory T cell feedback circuit maintains immune homeostasis by pruning self-activated T cells.
Cell
184
:
3981
3997.e22
.
178.
Lugo-Villarino
G.
,
R.
Maldonado-López
,
R.
Possemato
,
C.
Peñaranda
,
L. H.
Glimcher
.
2003
.
T-bet is required for optimal production of IFN-γ and antigen-specific T cell activation by dendritic cells.
Proc. Natl. Acad. Sci. USA
100
:
7749
7754
.
179.
Bachus
H.
,
K.
Kaur
,
A. M.
Papillion
,
T. T.
Marquez-Lago
,
Z.
Yu
,
A.
Ballesteros-Tato
,
S.
Matalon
,
B.
León
.
2019
.
Impaired tumor-necrosis-factor-α-driven dendritic cell activation limits lipopolysaccharide-induced protection from allergic inflammation in infants.
Immunity
50
:
225
240.e4
.
180.
Brown
C. C.
,
H.
Gudjonson
,
Y.
Pritykin
,
D.
Deep
,
V.-P.
Lavallée
,
A.
Mendoza
,
R.
Fromme
,
L.
Mazutis
,
C.
Ariyan
,
C.
Leslie
, et al
2019
.
Transcriptional basis of mouse and human dendritic cell heterogeneity.
Cell
179
:
846
863.e24
.
181.
Baptista
A. P.
,
A.
Gola
,
Y.
Huang
,
P.
Milanez-Almeida
,
P.
Torabi-Parizi
,
J. F.
Urban
Jr.
,
V. S.
Shapiro
,
M. Y.
Gerner
,
R. N.
Germain
.
2019
.
The chemoattractant receptor Ebi2 drives intranodal naive CD4+ T cell peripheralization to promote effective adaptive immunity.
Immunity
50
:
1188
1201.e6
.
182.
Schaupp
L.
,
S.
Muth
,
L.
Rogell
,
M.
Kofoed-Branzk
,
F.
Melchior
,
S.
Lienenklaus
,
S. C.
Ganal-Vonarburg
,
M.
Klein
,
F.
Guendel
,
T.
Hain
, et al
2020
.
Microbiota-induced type I interferons instruct a poised basal state of dendritic cells.
Cell
181
:
1080
1096.e19
.
183.
Sachet
M.
,
Y. Y.
Liang
,
R.
Oehler
.
2017
.
The immune response to secondary necrotic cells.
Apoptosis
22
:
1189
1204
.
184.
Sancho
D.
,
O. P.
Joffre
,
A. M.
Keller
,
N. C.
Rogers
,
D.
Martínez
,
P.
Hernanz-Falcón
,
I.
Rosewell
,
C.
Reis e Sousa
.
2009
.
Identification of a dendritic cell receptor that couples sensing of necrosis to immunity.
Nature
458
:
899
903
.
185.
Kushwah
R.
,
J.
Wu
,
J. R.
Oliver
,
G.
Jiang
,
J.
Zhang
,
K. A.
Siminovitch
,
J.
Hu
.
2010
.
Uptake of apoptotic DC converts immature DC into tolerogenic DC that induce differentiation of Foxp3+ Treg.
Eur. J. Immunol.
40
:
1022
1035
.
186.
Urban
B. C.
,
N.
Willcox
,
D. J.
Roberts
.
2001
.
A role for CD36 in the regulation of dendritic cell function.
Proc. Natl. Acad. Sci. USA
98
:
8750
8755
.
187.
Canton
J.
,
H.
Blees
,
C. M.
Henry
,
M. D.
Buck
,
O.
Schulz
,
N. C.
Rogers
,
E.
Childs
,
S.
Zelenay
,
H.
Rhys
,
M.-C.
Domart
, et al
2021
.
The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. [Published erratum appears in 2021 Nat. Immunol. 22: 391.]
Nat. Immunol.
22
:
140
153
.

The authors have no financial conflicts of interest.