Immune Netw. 2014 Jun;14(3):128-137. English.
Published online Jun 19, 2014.
Copyright © 2014 The Korean Association of Immunologists
Review

Differential Roles of Lung Dendritic Cell Subsets Against Respiratory Virus Infection

Tae Hoon Kim and Heung Kyu Lee
    • Laboratory of Host Defenses, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea.
Received April 29, 2014; Revised May 22, 2014; Accepted May 27, 2014.

This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Respiratory viruses can induce acute respiratory disease. Clinical symptoms and manifestations are dependent on interactions between the virus and host immune system. Dendritic cells (DCs), along with alveolar macrophages, constitute the first line of sentinel cells in the innate immune response against respiratory viral infection. DCs play an essential role in regulating the immune response by bridging innate and adaptive immunity. In the steady state, lung DCs can be subdivided into CD103+ conventional DCs (cDCs), CD11b+ cDCs, and plasmacytoid DCs (pDCs). In the inflammatory state, like a respiratory viral infection, monocyte-derived DCs (moDCs) are recruited to the lung. In inflammatory lung, discrimination between moDCs and CD11b+ DCs in the inflamed lung has been a critical challenge in understanding their role in the antiviral response. In particular, CD103+ cDCs migrate from the intraepithelial base to the draining mediastinal lymph nodes to primarily induce the CD8+ T cell response against the invading virus. Lymphoid CD8α+ cDCs, which have a developmental relationship with CD103+ cDCs, also play an important role in viral antigen presentation. Moreover, pDCs have been reported to promote an antiviral response by inducing type I interferon production rather than adaptive immunity. However, the role of these cells in respiratory infections remains unclear. These different DC subsets have functional specialization against respiratory viral infection. Under certain viral infection, contextually controlling the balance of these specialized DC subsets is important for an effective immune response and maintenance of homeostasis.

Keywords
Dendritic cells; Influenza; Respiratory syncytial virus; Lung; Infection

INTRODUCTION

The lung is the essential organ for respiration. Because the lung mucosal area contacts air for gas exchange, it can be infected easily by various microbes, such as influenza, respiratory syncytial virus (RSV), pneumococcus, and Aspergillus. Nevertheless, the lung possesses a sentinel system that identifies these threats and elicits an anti-microbial response. In this review, we focus on the immune response to respiratory viral infection, which can induce acute respiratory disease.

Dendritic cells (DCs) participate in the first line of defense in the innate immune response against respiratory viral infection. DCs are distributed throughout the entire lung, with each subset localized to a specific compartment of the organ (1). In the absence of inflammation, lung DCs can be subdivided into three distinct subsets based on the combined expression of cell surface markers: CD103+ conventional DCs (cDCs), CD11b+ cDCs, and plasmacytoid DCs (pDCs). During inflammation, monocyte-derived DCs (moDCs) are generated in the lung (2, 3) (Table I).

Table I
Established phenotype of mouse dendritic cells in the respiratory tract

In a respiratory virus infection, one virus can induce different types of immune responses depending on the type of DC subset activated (4, 5). In this process, cell type-specific pattern recognition receptors (PRRs) may also be involved (6). Each DC subset expresses different pattern recognition receptors, thereby enabling the cells to react differently depending on the type of virus infection (7). In particular, neither a vaccine nor an effective antiviral therapy is currently available against RSV infection (8). To development vaccine for RSV infection, understanding the role of the lung DC subsets is important. Determining the specialized functions of the various lung DC subsets is challenging. This review focuses on the distinctive features and antiviral functions exhibited by the various lung DC subsets during respiratory virus infection in mice.

CD103+ conventional dendritic cells

The CD103+CD11b- cDC subset shares its origin and function with lymphoid tissue CD8α+ cDCs (9, 10). CD103+ cDCs are primarily distributed to connective tissues. The proportion of CD103+ cDCs among total conventional DCs rarely exceeds 20~30%. These cells express higher fms-like tyrosine kinase 3 (Flt3) levels compared to CD11b+ cDCs and therefore proliferate in response to Flt3 ligand (11). CD103 expression is dependent on the tissue microenvironment and regulated by local production of the cytokine Csf-2 (GM-CSF) (12, 13, 14, 15). However, CD103-deficient mice do not exhibit major defects in DC development (16). CD103+ cDCs lack the macrophage-related markers CD11b, CD115, CD172a, F4/80, and CX3CR1. With the exception of intestinal and pancreatic CD103+ cDCs, these cells express the C-type lectin receptor langerin (11, 17).

Besides connective tissues, CD103+ cDCs are located in nonlymphoid tissues at the interface with the environment. Lung CD103+ cDCs can be found in the mucosa and vascular wall (18) (Fig. 1). Following antigen uptake, CD103+ cDCs migrate to the T cell zone of draining lymph nodes (10). In the airways and gut, DCs extend their processes between epithelial cells to contact the airway lumen directly. These airway mucosal DCs can conduct continuous immune surveillance of the airway luminal surface, thereby acting like a periscope (19, 20, 21). In mouse lungs, intraepithelial CD103+ cDCs express the tight-junction proteins claudin-1, claudin-7, and zonula-2, which form tight junctions with airway epithelial cells (18). As a result, CD103+ cDCs can sample contents within the airway lumen without disturbing the function of the epithelium barrier.

Figure 1
Different type of DC subsets in the respiratory virus infected lung. In steady state, the lung contains multiple subsets of DCs, such as CD103+ cDCs, CD11b+ cDCs, CD8α+ cDCs, and pDCs. CD103+ cDCs are mainly located in mucosal walls, and extend their process to alveolar space for capture viral antigen. CD11b+ cDCs are distributed in lamina propria, which is below the basement membrane. pDCs are place in conducting airway, parenchyma and alveolar septa. After viral infection, inflammatory lung was induced the recruitment of moDCs. And viral antigen uptake migratory DCs translocate to draining mediastinal lymph nodes via afferent lymphatics. Migrated DCs can present to naïve T cells. Lymph node resident CD8α+ DCs can receive antigen from migratory DCs, and present T cells.

Current reports have shown that, following influenza or RSV infection, CD103+ cDCs migrate from the intraepithelial base to the draining mediastinal lymph nodes (22, 23), where they mainly present antigen to naïve CD4+ and CD8+ T cells. However, previous studies demonstrated that antigens are transferred from migratory DCs to CD8α+ resident cDCs, and presented to T cells by antigen-bearing CD8α+ resident cDCs (24).

As previously mentioned, studies have established that CD103+ cDCs belong to the CD8α+ subset of cDCs (2). Like lymphoid-derived CD8α+ cDCs, CD103+ cDCs originate exclusively from pre-DCs under the control of Flt3 ligand, inhibitor of DNA protein 2 (Id2), and interferon regulatory protein 8 (Irf8) (11, 25). Murphy et al. reported a developmental relationship between lymphoid organ-resident CD8α+ cDCs and nonlymphoid CD103+ cDCs using Batf3-deficient mice (26). A recent study used heat maps to demonstrate that the expression of pattern recognition receptors, cytokines, and chemokine receptors is similar between CD103+ cDCs and CD8α+ cDCs (27). In particular, both subsets express TLR3, TLR11, the scavenger receptor CD36, and C-type lectin Clec9A (28, 29, 30, 31). Desch, et al. showed that mouse lung CD103+ cDCs selectively express TLR3, while CD11b+ cDCs express TLR2 and TLR7 (32).

CD103+ cDCs play a nonredundant role in stimulating CD8+ T cell-mediated immunity. Influenza virus infection following depletion of CD103+ cDCs in langerin-diphtheria toxin receptor (langerin-DTR) mice results in severe illness, defective viral clearance, and abrogated antiviral response due to impaired development of influenza virus-specific CD8+ T cells (22). In the Batf3 knockout mouse model, the CD103+ cDC-deficient mice cannot produce CD8+ T cell priming in response to influenza infection (33). CD103+ cDCs play an important role in cross-presentation of apoptotic cell-associated antigen to CD8+ T cells (25, 32). However, whether CD103+ cDCs can induce a cytotoxic T cell response against RSV infection (similar to other viruses) remains to be investigated.

The role of lung CD103+ cDCs in the activation of CD4+ T cells is unclear. In cutaneous skin infection with Candida albicans, dermal CD103+ cDCs control the induction of pathogen-specific CD4+ IFN-γ+ T cells (34). A recent study using langerin-DTR mice demonstrated that ablation of CD103+ cDCs inhibited induction of the encephalitogenic CD4+ Th1 response and autoimmune encephalomyelitis (EAE) (35). However, some studies showed that the CD4+ T cell response was independent of CD103+ cDCs. Batf3 knockout mice that are deficient in CD103+ cDCs can mount an efficient CD4+ T cell response to West Nile virus or autoimmune EAE (14, 25). Moreover, ablation of CD103+ cDCs in langerin-DTR mice did not affect the CD4+ T cell response against Leishmania major infection (36).

CD8α+ cDCs and CD103+ cDCs are thought to participate in deletional tolerance of self-reactive T cells and the induction of antigen-specific regulatory T cells (Treg) (16). Splenic DCs captured dying cells and processed, then induced specific tolerance (37, 38). A report showed that the CD103+CD207+ subset of splenic CD8α+ cDCs is responsible for tolerance induction to cell-associated antigens (39). However, an autoimmune response was not observed in Batf3 knockout mice that lack CD8α+ cDCs and CD103+ cDCs. Thus, the tolerogenic function of lung CD103+ cDCs remains to be determined.

CD11b+ conventional dendritic cells

In the lung, CD11b+ cDCs reside mainly in the lamina propria, which is located below the basement membrane (Fig. 1). CD11b+ cDCs are heterogeneous and their development depends on both Flt3 and M-CSFR (11). Dependency on M-CSFR is suggestive of a monocytic origin, and some non-lymphoid CD11b+ cDCs can be reconstituted by pre-DC. CD11b+ cDCs frequently lack CD103 but express CD11b. Despite this, markers to distinguish the two ontogenically distinct subsets differ between tissues. For instance, expression of CD64 (FcγRI) helps distinguish between these two subpopulations in muscle, whereas expression of CD103 helps discriminate between the two CD11b+ DC subsets in the intestinal lamina propria (40, 41). Lambrecht et al. recommended detection of CD64 and MAR-1 expression as the most reliable method to discriminate between monocyte-derived DCs and CD11b+ cDCs in the lung and mediastinal lymph nodes (42).

Because CD11b+ cDCs are not a homogenous subset, the exact PRR profile of CD11b+ cDCs is complex. Nevertheless, these receptors are expressed differentially in CD103+ cDCs and CD8α+ cDCs (27). Quantitative proteomics has revealed that splenic CD11b+ cDCs express high levels of cytoplasmic viral sensors and are potent cytokine producers in the steady state and upon stimulation (43). Lung CD11b+ cDCs are major producers of proinflammatory chemokines, including MCP-1, MIP-1α, MIP-1β, RANTES, and MCP5, attracting inflammatory cells and effector T lymphocytes to the lung (44).

CD11b+ cDCs can capture antigens and migrate from nonlymphoid tissues to regional draining lymph nodes (23). Research has established that CD8α+ cDCs and CD103+ cDCs play crucial roles in cross-presentation. However, during influenza infection, CD103+ cDCs and CD11b+ cDCs are the primary mediators of antigen presentation to naïve CD8+ T cells in the draining lymph nodes (45).

During severe influenza infection, CD11b+ cDCs, but not CD103+ cDCs or CD8α+ resident cDCs, accumulate in the draining lymph nodes to become the predominant DC subset responsible for stimulating CD8+ T cells via the costimulatory molecule CD70 (46). These contradictory findings could be attributed to the different viral doses used for infection and the differential effects of direct DC infection by influenza virus. Severe viral infection induced CD11b+ cDCs that were incapable of antigen presentation to CD8+ T cells. However, low viral doses enabled directly infected CD11b+ cDCs to arrive at the draining lymph nodes ready to prime the CD8+ T cell response (47). In addition, CD11b+ cDCs are thought to play a predominant role in MHC class II presentation, including acting as the predominant presenters of viral antigens to CD4+ T cells in response to influenza virus infection (45).

CD11b+ cDCs constantly escape from the blood to the thymus to induce central tolerance, such as clonal deletion of autoreactive T cells or differentiation of Treg (48, 49). CD103+ CD11b+ cDCs purified from the lamina propria of the small intestine were found to promote a high level of Treg differentiation relative to lymphoid organ-derived DCs (50, 51). However, the contribution of lung CD11b+ cDCs in tolerance has not been established.

In addition to CD103+ cDC-mediated uptake in the airways, CD11b+ cDCs utilize another pathway to acquire inhaled antigens. TLR4 triggering of epithelial cells caused production of innate proallergic cytokines, including thymic stromal lymphopoietin (TSLP), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-25, and interleukin-33. In the absence of TLR4 on structural, but not hematopoietic cells, CD11b+ cDCs were not recruited or activated in a chimeric mouse model (52). It is unclear whether lung CD11b+ cDCs require epithelial activation as well.

CD11b+ cDCs are essential for the maintenance of inducible bronchus-associated lymphoid tissue (iBALT), a tertiary lymphoid organ (TLO) induced in the lungs after influenza infection (53). After viral clearance, CD11b+ cDCs isolated from the lungs of mice with iBALT no longer presented viral antigens to T cells but produced lymphotoxin (LT) β and homeostatic chemokines (CXCL-12, CXCL-13, CCL-19, and CCL-21) known to contribute to TLO organization. Using the replication-deficient modified vaccinia virus model, Halle, et al. described iBALT as a tertiary lymphoid structure that supports the efficient priming of T cells against unrelated inhaled antigens with DCs required for its maintenance (54).

Plasmacytoid dendritic cells

pDCs are distributed to conducting airways as well as parenchyma and alveolar septa in the lung (Fig. 1). These cells represent a small subset of DCs, which share a common origin with cDCs. pDCs develop in the bone marrow from a continuum of Flt3+c-Kitlow progenitors, including lymphoid progenitors and common DC progenitors (CDPs). Their development proceeds through the putative committed pDC progenitor and immature pDCs in the bone marrow toward the mature peripheral pDCs (55). Upregulation of the basic helix-loop-helix transcription factor (E protein) E2-2 serves as a key lineage commitment event in pDC development (56, 57). Because E proteins are essential regulators of lymphocyte development, E2-2 activity may underlie the distinct lymphoid features of pDCs. These cells express low levels of MHC class II and costimulatory molecules, as well as low levels of CD11c in the steady state (16). They also express a narrow range of PRRs, including TLR7 and TLR9.

Generally, pDCs function during the antiviral response to produce type I IFNs that induce the adaptive immune response. Some studies have shown that pDCs can trigger an influenza-specific CD8+ T cell response in vitro (58, 59, 60). However, RSV-stimulated pDCs cannot enhance the proliferation and maturation of antigen-specific T cells, but rather promote direct antiviral activity by secreting type I IFNs (61).

Following influenza infection in vivo, 120G8+CD11cint pDCs accumulate in the lung and lymph nodes carrying viral nucleoprotein (NP). Depletion of pDCs using 120G8 antibodies did not affect viral clearance or clinical severity during influenza infection (22). Instead, pDC depletion led to a reduction in antiviral antibody production after clearance of influenza from the lung. However, depletion of pDCs resulted in decreases viral clearance of RSV infection and exacerbation of all facets of immune-mediated pathology, including increase of airway hyper-responsiveness, pulmonary inflammation, and mucus production (62, 63).

In IkarosL/L mice, expressing low levels of the transcription factor Ikaros (Ik(L/L)) lack peripheral pDCs, pDCs regulate T cell accumulation in the bronchoalveolar space during early influenza virus infection, but are not essential for controlling this disease (64). These data demonstrated that the antiviral CD8+ T cell response was independent of pDCs. However, in BDCA2-DTR mice, pDC depletion reduced early type I IFN production, enhanced early viral replication, and impaired the survival and accumulation of virus-specific cytotoxic T lymphocytes in systemic MCMV or VSV infection (65).

According to a recent report, pDCs do not appear to influence viral burden, survival, or virus-specific CD8+ T cell response during local HSV infection. In contrast, pDCs were important for early type I IFN production, NK cell activation, and CD8+ T cell response during systemic HSV infection (66). These results help elucidate the antiviral role of pDCs in respiratory virus infection. However, whether pDCs can differentially respond under different conditions between host and virus remains to be determined.

Monocyte-derived dendritic cells

Inflammatory moDCs differentiate from circulating Ly6Chi monocytes (67) (Fig. 1). Recent studies have established that, under conditions of stress, such as TLR stimulation, early hematopoietic precursors can differentiate into DCs, bypassing normal growth and differentiation requirements (68, 69). However, the contribution of monocytes and DC-related precursors to the differentiation of lung moDCs in response to respiratory virus infection remains unclear.

Most inflammatory DCs are characterized by the expression of Ly6C, CD11b, MHC class II, and intermediate levels of CD11c (67). Ly6C is a distinct marker of monocytes, but that is downregulated rapidly in the presence of moDCs (42, 70, 71). Therefore, distinguishing inflammatory moDCs from nonlymphoid CD11b+ DCs is challenging. As mentioned in the preceding section, one report demonstrated that staining with the MAR-1 antibody directed against the high affinity immunoglobulin E (IgE) α chain receptor (FcεRI) is better than staining for Ly6C (2). A recent study showed that inflammatory moDCs are recruited to draining lymph nodes following lipopolysaccharide (LPS) stimulation, and that these moDCs express the lectin DC-SIGN/CD209, the mannose receptor CD206, and CD14 (71).

Monocytes were originally considered the immediate upstream precursors of cDCs. This hypothesis originated from studies showing that DCs could be differentiated in vitro from human blood mononuclear cells using GM-CSF and IL-4 (72). When monocytes were transferred into mice with an inflammatory milieu dependent on GM-CSF, monocytes produced a distinct type of splenic DC (73). Nowadays, the concept that monocytes are a precursor of inflammatory DCs is widely accepted. More recent studies have shown that monocytes contribute to cDC development in the steady state (41, 74, 75, 76). However, because this review focuses on DC subsets that act against respiratory virus infection, we refer to mononuclear cell-derived DCs as moDCs in inflammation.

CD11b+ DCs can produce TNF and iNOS-derived NO during L. monocytogenes infection. These Tip-DCs are dependent on CCR2 and mediate innate immunity against this intracellular bacterial pathogen (77), suggesting that Tip-DCs may contribute to the elimination of intracellular pathogens.

A recent study identified an uncharacterized zinc finger transcription factor named zDC (Zbtb46, Btbd4) that is expressed specifically by cDCs and committed cDC precursors but not by monocytes, pDCs, or other immune cell populations (78, 79). zDC-DTR mice treated with diphtheria toxin eliminated LPS-induced inflammatory moDCs, suggesting that LPS induced inflammatory moDCs that belong to a real DC population. However, L. monocytogenes infection-induced Tip-DCs were not ablated by DT treatment in these mice. Given this result, Tip-DCs most likely resemble monocytes more than DCs.

CD11b+ moDCs are recruited to inflammatory sites in the lungs following exposure to respiratory antigen or virus. During influenza infection, moDCs also differentiate from monocytes in the lung. These trafficking and differentiation process are dependent on type I IFN signaling and CCR2 during influenza infection (80, 81). Some in vitro studies suggested that type I IFN-producing moDCs can regulate viral replication (82, 83); however, whether moDCs participate directly in the antiviral response remains unclear. Interestingly, CCR2-deficient mice did not exhibit increased influenza viral titer.

Whether moDCs can migrate to draining lymph nodes and induce the T cell response has not been determined (45, 46). Monocyte-derived CD11c+ DCs, which express CX3CR1, can patrol the vessel wall of the pulmonary arterial vasculature and capture embolic materials. Thus, these cells are essential and sufficient for priming of naïve T cells in lung draining mediastinal lymph nodes (84). Some studies have shown that moDCs may be important for the interaction of effector T cells present in the infection site instead of the lymph nodes (85, 86).

CD8α+ dendritic cells

Generally, CD8α+ cDCs do not exist in the lung because these cells are non-migrating, lymphoid-organ resident DCs. However, CD8α+ cDCs are involved in respiratory virus infection. They can induce the T cell response in mediastinal lymph nodes. CD8α+ cDCs constitute 20~40% of spleen and lymph node cDCs. Similar to CD103+ cDCs, CD8α+ cDCs lack expression of CD11b and other macrophage markers. However, they express high levels of Flt3 and proliferate in response to Flt3 ligand (87).

Lymphoid resident CD8α+ cDCs are immature in the steady state, but microbial products can induce maturation of CD8α+ cDCs. Lymph node CD8α+ cDCs are located in the subcapsular sinus, the site of afferent lymphatic vessel entry (39, 88). After antigen uptake, these CD8α+ cDCs migrate to the T cell zone where they present antigens.

As mentioned above, CD8α+ cDCs share their origin and function with nonlymphoid CD103+ cDCs. However, the function of CD8α+ cDCs themselves is still unclear. Generation of conditional or knockout mouse models for specific depletion of CD8α+ cDCs will aid in our understanding of the function of these cells. Additional studies are required to determine whether these cells have a common immediate precursor and to investigate which cell is the precursor and progeny for CD8α+ cDC (89).

CONCLUSION

Respiratory viruses can induce acute respiratory disease. In the lung, DCs are the first line of sentinel cells in the innate immune response against respiratory viral infection, similar to alveolar macrophages. DCs are crucial in regulating the immune response by bridging innate and adaptive immunity. These cells can produce inflammatory cytokines and chemokines, as well as migrate to the draining lymph nodes to initiate the adaptive immune response through antigen presentation. Lung DCs associated with viral infection can be subdivided into CD103+ cDCs, CD11b+ cDCs, pDCs, and moDCs. Lymphoid CD8α+ cDCs also play an important role in the antiviral response. These different DC subsets have functional specialization against respiratory viral infection. One virus can induce different immune responses depending on the type of DC subset activated. Moreover, one subset can react differently depending on the type of virus encountered. Contextually controlling the balance between these specialized DC subsets is important for an effective antiviral response and maintaining immune homeostasis. Moreover, understanding the differential roles of lung dendritic cell subsets against respiratory virus infection is a key point to develop a vaccine.

Notes

The authors have no financial conflict of interest.

Abbreviations

DC dendritic cell
cDC conventional dendritic cell
pDC plasmacytoid dendritic cell
moDC monocyte-derived dendritic cell
Treg regulatory T cells
PRRs pattern recognition receptors
TLR toll-like receptor
DTR diphtheria toxin receptor
IFN interferon
Tip-DC TNF and iNOS derived NO producing dendritic cell

ACKNOWLEDGEMENTS

We thank Sang Eun Oh for her help with the figure. This work was supported by the National Research Foundation (NRF-2013R1A1A2063347, NRF-2012R1A1A2046001, NRF-2012M3A9B4028274) and the Converging Research Center Program (2011K000864) funded by the Ministry of Science, ICT and Future Planning of Korea.

References

    1. De Heer HJ, Hammad H, Kool M, Lambrecht BN. Dendritic cell subsets and immune regulation in the lung. Semin Immunol 2005;17:295–303.
    1. Neyt K, Lambrecht BN. The role of lung dendritic cell subsets in immunity to respiratory viruses. Immunol Rev 2013;255:57–67.
    1. Guilliams M, Lambrecht BN, Hammad H. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal Immunol 2013;6:464–473.
    1. Johnson TR, Johnson CN, Corbett KS, Edwards GC, Graham BS. Primary human mDC1, mDC2, and pDC dendritic cells are differentially infected and activated by respiratory syncytial virus. PLoS One 2011;6:e16458.
    1. Kim TH, Lee HK. Innate immune recognition of respiratory syncytial virus infection. BMB Rep 2014;47:184–191.
    1. Iwasaki A, Pillai PS. Innate immunity to influenza virus infection. Nat Rev Immunol 2014;14:315–328.
    1. Guerrero-Plata A, Casola A, Suarez G, Yu X, Spetch L, Peeples ME, Garofalo RP. Differential response of dendritic cells to human metapneumovirus and respiratory syncytial virus. Am J Respir Cell Mol Biol 2006;34:320–329.
    1. Chang J. Current progress on development of respiratory syncytial virus vaccine. BMB Rep 2011;44:232–237.
    1. del Rio ML, Bernhardt G, Rodriguez-Barbosa JI, Forster R. Development and functional specialization of CD103+ dendritic cells. Immunol Rev 2010;234:268–281.
    1. Helft J, Ginhoux F, Bogunovic M, Merad M. Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice. Immunol Rev 2010;234:55–75.
    1. Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, Hashimoto D, Price J, Yin N, Bromberg J, Lira SA, Stanley ER, Nussenzweig M, Merad M. The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med 2009;206:3115–3130.
    1. Zhan Y, Carrington EM, van Nieuwenhuijze A, Bedoui S, Seah S, Xu Y, Wang N, Mintern JD, Villadangos JA, Wicks IP, Lew AM. GM-CSF increases cross-presentation and CD103 expression by mouse CD8+ spleen dendritic cells. Eur J Immunol 2011;41:2585–2595.
    1. Sathe P, Pooley J, Vremec D, Mintern J, Jin JO, Wu L, Kwak JY, Villadangos JA, Shortman K. The acquisition of antigen cross-presentation function by newly formed dendritic cells. J Immunol 2011;186:5184–5192.
    1. Edelson BT, Bradstreet TR, KC W, Hildner K, Herzog JW, Sim J, Russell JH, Murphy TL, Unanue ER, Murphy KM. Batf3-dependent CD11b(low/-) peripheral dendritic cells are GM-CSF-independent and are not required for Th cell priming after subcutaneous immunization. PLoS One 2011;6:e25660.
    1. Greter M, Helft J, Chow A, Hashimoto D, Mortha A, Agudo-Cantero J, Bogunovic M, Gautier EL, Miller J, Leboeuf M, Lu G, Aloman C, Brown BD, Pollard JW, Xiong H, Randolph GJ, Chipuk JE, Frenette PS, Merad M. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity 2012;36:1031–1046.
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: Ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 2013;31:563–604.
    1. Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 2008;8:935–947.
    1. Sung SS, Fu SM, Rose CE Jr, Gaskin F, Ju ST, Beaty SR. A major lung CD103 (αE)-β7 integrin-positive epithelial dendritic cell population expressing Langerin and tight junction proteins. J Immunol 2006;176:2161–2172.
    1. Jahnsen FL, Strickland DH, Thomas JA, Tobagus IT, Napoli S, Zosky GR, Turner DJ, Sly PD, Stumbles PA, Holt PG. Accelerated antigen sampling and transport by airway mucosal dendritic cells following inhalation of a bacterial stimulus. J Immunol 2006;177:5861–5867.
    1. Chieppa M, Rescigno M, Huang AY, Germain RN. Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement. J Exp Med 2006;203:2841–2852.
    1. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol 2008;8:193–204.
    1. GeurtsvanKessel CH, Willart MA, van Rijt LS, Muskens F, Kool M, Baas C, Thielemans K, Bennett C, Clausen BE, Hoogsteden HC, Osterhaus AD, Rimmelzwaan GF, Lambrecht BN. Clearance of influenza virus from the lung depends on migratory langerin+ CD11b- but not plasmacytoid dendritic cells. J Exp Med 2008;205:1621–1634.
    1. Lukens MV, Kruijsen D, Coenjaerts FE, Kimpen JL, van Bleek GM. Respiratory syncytial virus-induced activation and migration of respiratory dendritic cells and subsequent antigen presentation in the lung-draining lymph node. J Virol 2009;83:7235–7243.
    1. Belz GT, Smith CM, Kleinert L, Reading P, Brooks A, Shortman K, Carbone FR, Heath WR. Distinct migrating and nonmigrating dendritic cell populations are involved in MHC class I-restricted antigen presentation after lung infection with virus. Proc Natl Acad Sci U S A 2004;101:8670–8675.
    1. Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama M, Calderon B, Schraml BU, Unanue ER, Diamond MS, Schreiber RD, Murphy TL, Murphy KM. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 2008;322:1097–1100.
    1. Edelson BT, KC W, Juang R, Kohyama M, Benoit LA, Klekotka PA, Moon C, Albring JC, Ise W, Michael DG, Bhattacharya D, Stappenbeck TS, Holtzman MJ, Sung SS, Murphy TL, Hildner K, Murphy KM. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8α+ conventional dendritic cells. J Exp Med 2010;207:823–836.
    1. Miller JC, Brown BD, Shay T, Gautier EL, Jojic V, Cohain A, Pandey G, Leboeuf M, Elpek KG, Helft J, Hashimoto D, Chow A, Price J, Greter M, Bogunovic M, Bellemare-Pelletier A, Frenette PS, Randolph GJ, Turley SJ, Merad M. The Immunological Genome Consortium. Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 2012;13:888–899.
    1. Edwards AD, Diebold SS, Slack EM, Tomizawa H, Hemmi H, Kaisho T, Akira S, Reis e Sousa C. Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8α+ DC correlates with unresponsiveness to imidazoquinolines. Eur J Immunol 2003;33:827–833.
    1. Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL, Serhan CN, Hayden MS, Hieny S, Sutterwala FS, Flavell RA, Ghosh S, Sher A. TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science 2005;308:1626–1629.
    1. Sancho D, Joffre OP, Keller AM, Rogers NC, Martinez D, Hernanz-Falcon P, Rosewell I, Reis e Sousa C. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 2009;458:899–903.
    1. Davey GM, Wojtasiak M, Proietto AI, Carbone FR, Heath WR, Bedoui S. Cutting edge: priming of CD8 T cell immunity to herpes simplex virus type 1 requires cognate TLR3 expression in vivo. J Immunol 2010;184:2243–2246.
    1. Desch AN, Randolph GJ, Murphy K, Gautier EL, Kedl RM, Lahoud MH, Caminschi I, Shortman K, Henson PM, Jakubzick CV. CD103+ pulmonary dendritic cells preferentially acquire and present apoptotic cell-associated antigen. J Exp Med 2011;208:1789–1797.
    1. Helft J, Manicassamy B, Guermonprez P, Hashimoto D, Silvin A, Agudo J, Brown BD, Schmolke M, Miller JC, Leboeuf M, Murphy KM, Garcia-Sastre A, Merad M. Cross-presenting CD103+ dendritic cells are protected from influenza virus infection. J Clin Invest 2012;122:4037–4047.
    1. Igyarto BZ, Haley K, Ortner D, Bobr A, Gerami-Nejad M, Edelson BT, Zurawski SM, Malissen B, Zurawski G, Berman J, Kaplan DH. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity 2011;35:260–272.
    1. King IL, Kroenke MA, Segal BM. GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization. J Exp Med 2010;207:953–961.
    1. Brewig N, Kissenpfennig A, Malissen B, Veit A, Bickert T, Fleischer B, Mostbock S, Ritter U. Priming of CD8+ and CD4+ T cells in experimental leishmaniasis is initiated by different dendritic cell subtypes. J Immunol 2009;182:774–783.
    1. Liu K, Idoyaga J, Charalambous A, Fujii S, Bonito A, Mordoh J, Wainstok R, Bai XF, Liu Y, Steinman RM. Innate NKT lymphocytes confer superior adaptive immunity via tumor-capturing dendritic cells. J Exp Med 2005;202:1507–1516.
    1. Liu K, Iyoda T, Saternus M, Kimura Y, Inaba K, Steinman RM. Immune tolerance after delivery of dying cells to dendritic cells in situ. J Exp Med 2002;196:1091–1097.
    1. Qiu CH, Miyake Y, Kaise H, Kitamura H, Ohara O, Tanaka M. Novel subset of CD8α+ dendritic cells localized in the marginal zone is responsible for tolerance to cell-associated antigens. J Immunol 2009;182:4127–4136.
    1. Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, Pabst O. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med 2009;206:3101–3114.
    1. Langlet C, Tamoutounour S, Henri S, Luche H, Ardouin L, Gregoire C, Malissen B, Guilliams M. CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization. J Immunol 2012;188:1751–1760.
    1. Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W, Vanhoutte L, Neyt K, Killeen N, Malissen B, Hammad H, Lambrecht BN. Conventional and monocyte-derived CD11b+ dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 2013;38:322–335.
    1. Luber CA, Cox J, Lauterbach H, Fancke B, Selbach M, Tschopp J, Akira S, Wiegand M, Hochrein H, O'Keeffe M, Mann M. Quantitative proteomics reveals subset-specific viral recognition in dendritic cells. Immunity 2010;32:279–289.
    1. Beaty SR, Rose CE Jr, Sung SS. Diverse and potent chemokine production by lung CD11bhigh dendritic cells in homeostasis and in allergic lung inflammation. J Immunol 2007;178:1882–1895.
    1. Kim TS, Braciale TJ. Respiratory dendritic cell subsets differ in their capacity to support the induction of virus-specific cytotoxic CD8+ T cell responses. PLoS One 2009;4:e4204.
    1. Ballesteros-Tato A, Leon B, Lund FE, Randall TD. Temporal changes in dendritic cell subsets, cross-priming and costimulation via CD70 control CD8+ T cell responses to influenza. Nat Immunol 2010;11:216–224.
    1. Plantinga M, Hammad H, Lambrecht BN. Origin and functional specializations of DC subsets in the lung. Eur J Immunol 2010;40:2112–2118.
    1. Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH. Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 2006;7:1092–1100.
    1. Proietto AI, van Dommelen S, Zhou P, Rizzitelli A, D'Amico A, Steptoe RJ, Naik SH, Lahoud MH, Liu Y, Zheng P, Shortman K, Wu L. Dendritic cells in the thymus contribute to T-regulatory cell induction. Proc Natl Acad Sci U S A 2008;105:19869–19874.
    1. Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med 2007;204:1775–1785.
    1. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 2007;204:1757–1764.
    1. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 2009;15:410–416.
    1. GeurtsvanKessel CH, Willart MA, Bergen IM, van Rijt LS, Muskens F, Elewaut D, Osterhaus AD, Hendriks R, Rimmelzwaan GF, Lambrecht BN. Dendritic cells are crucial for maintenance of tertiary lymphoid structures in the lung of influenza virus-infected mice. J Exp Med 2009;206:2339–2349.
    1. Halle S, Dujardin HC, Bakocevic N, Fleige H, Danzer H, Willenzon S, Suezer Y, Hammerling G, Garbi N, Sutter G, Worbs T, Forster R. Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. J Exp Med 2009;206:2593–2601.
    1. Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 2011;29:163–183.
    1. Cisse B, Caton ML, Lehner M, Maeda T, Scheu S, Locksley R, Holmberg D, Zweier C, den Hollander NS, Kant SG, Holter W, Rauch A, Zhuang Y, Reizis B. Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell 2008;135:37–48.
    1. Reizis B. Regulation of plasmacytoid dendritic cell development. Curr Opin Immunol 2010;22:206–211.
    1. Cella M, Facchetti F, Lanzavecchia A, Colonna M. Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization. Nat Immunol 2000;1:305–310.
    1. Fonteneau JF, Gilliet M, Larsson M, Dasilva I, Münz C, Liu YJ, Bhardwaj N. Activation of influenza virus-specific CD4+ and CD8+ T cells: a new role for plasmacytoid dendritic cells in adaptive immunity. Blood 2003;101:3520–3526.
    1. Hoeffel G, Ripoche AC, Matheoud D, Nascimbeni M, Escriou N, Lebon P, Heshmati F, Guillet JG, Gannage M, Caillat-Zucman S, Casartelli N, Schwartz O, De la Salle H, Hanau D, Hosmalin A, Maranon C. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity 2007;27:481–492.
    1. Boogaard I, van Oosten M, van Rijt LS, Muskens F, Kimman TG, Lambrecht BN, Buisman AM. Respiratory syncytial virus differentially activates murine myeloid and plasmacytoid dendritic cells. Immunology 2007;122:65–72.
    1. Smit JJ, Rudd BD, Lukacs NW. Plasmacytoid dendritic cells inhibit pulmonary immunopathology and promote clearance of respiratory syncytial virus. J Exp Med 2006;203:1153–1159.
    1. Wang H, Peters N, Schwarze J. Plasmacytoid dendritic cells limit viral replication, pulmonary inflammation, and airway hyperresponsiveness in respiratory syncytial virus infection. J Immunol 2006;177:6263–6270.
    1. Wolf AI, Buehler D, Hensley SE, Cavanagh LL, Wherry EJ, Kastner P, Chan S, Weninger W. Plasmacytoid dendritic cells are dispensable during primary influenza virus infection. J Immunol 2009;182:871–879.
    1. Swiecki M, Gilfillan S, Vermi W, Wang Y, Colonna M. Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8+ T cell accrual. Immunity 2010;33:955–966.
    1. Swiecki M, Wang Y, Gilfillan S, Colonna M. Plasmacytoid dendritic cells contribute to systemic but not local antiviral responses to HSV infections. PLoS Pathog 2013;9:e1003728.
    1. Dominguez PM, Ardavin C. Differentiation and function of mouse monocyte-derived dendritic cells in steady state and inflammation. Immunol Rev 2010;234:90–104.
    1. Nagai Y, Garrett KP, Ohta S, Bahrun U, Kouro T, Akira S, Takatsu K, Kincade PW. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity 2006;24:801–812.
    1. Takizawa H, Boettcher S, Manz MG. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood 2012;119:2991–3002.
    1. Leon B, Lopez-Bravo M, Ardavin C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 2007;26:519–531.
    1. Cheong C, Matos I, Choi JH, Dandamudi DB, Shrestha E, Longhi MP, Jeffrey KL, Anthony RM, Kluger C, Nchinda G, Koh H, Rodriguez A, Idoyaga J, Pack M, Velinzon K, Park CG, Steinman RM. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209+ dendritic cells for immune T cell areas. Cell 2010;143:416–429.
    1. Sallusto F, Lanzavecchia A. 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 1994;179:1109–1118.
    1. Naik SH, Metcalf D, van Nieuwenhuijze A, Wicks I, Wu L, O'Keeffe M, Shortman K. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat Immunol 2006;7:663–671.
    1. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, Cheong C, Liu K, Lee HW, Park CG, Steinman RM, Nussenzweig MC. Differential antigen processing by dendritic cell subsets in vivo. Science 2007;315:107–111.
    1. Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M. Origin of the lamina propria dendritic cell network. Immunity 2009;31:513–525.
    1. Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity 2009;31:502–512.
    1. Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA, Pamer EG. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003;19:59–70.
    1. Satpathy AT, KC W, Albring JC, Edelson BT, Kretzer NM, Bhattacharya D, Murphy TL, Murphy KM. Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages. J Exp Med 2012;209:1135–1152.
    1. Meredith MM, Liu K, Darrasse-Jeze G, Kamphorst AO, Schreiber HA, Guermonprez P, Idoyaga J, Cheong C, Yao KH, Niec RE, Nussenzweig MC. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med 2012;209:1153–1165.
    1. Lin KL, Suzuki Y, Nakano H, Ramsburg E, Gunn MD. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J Immunol 2008;180:2562–2572.
    1. Seo SU, Kwon HJ, Ko HJ, Byun YH, Seong BL, Uematsu S, Akira S, Kweon MN. Type I interferon signaling regulates Ly6C(hi) monocytes and neutrophils during acute viral pneumonia in mice. PLoS Pathog 2011;7:e1001304.
    1. Cao W, Taylor AK, Biber RE, Davis WG, Kim JH, Reber AJ, Chirkova T, De La Cruz JA, Pandey A, Ranjan P, Katz JM, Gangappa S, Sambhara S. Rapid differentiation of monocytes into type I IFN-producing myeloid dendritic cells as an antiviral strategy against influenza virus infection. J Immunol 2012;189:2257–2265.
    1. Hou W, Gibbs JS, Lu X, Brooke CB, Roy D, Modlin RL, Bennink JR, Yewdell JW. Viral infection triggers rapid differentiation of human blood monocytes into dendritic cells. Blood 2012;119:3128–3131.
    1. Willart MA, Jan de Heer H, Hammad H, Soullie T, Deswarte K, Clausen BE, Boon L, Hoogsteden HC, Lambrecht BN. The lung vascular filter as a site of immune induction for T cell responses to large embolic antigen. J Exp Med 2009;206:2823–2835.
    1. Iijima N, Mattei LM, Iwasaki A. Recruited inflammatory monocytes stimulate antiviral Th1 immunity in infected tissue. Proc Natl Acad Sci U S A 2011;108:284–289.
    1. Soudja SM, Ruiz AL, Marie JC, Lauvau G. Inflammatory monocytes activate memory CD8+ T and innate NK lymphocytes independent of cognate antigen during microbial pathogen invasion. Immunity 2012;37:549–562.
    1. Waskow C, Liu K, Darrasse-Jeze G, Guermonprez P, Ginhoux F, Merad M, Shengelia T, Yao K, Nussenzweig M. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat Immunol 2008;9:676–683.
    1. Idoyaga J, Suda N, Suda K, Park CG, Steinman RM. Antibody to Langerin/CD207 localizes large numbers of CD8α+ dendritic cells to the marginal zone of mouse spleen. Proc Natl Acad Sci USA 2009;106:1524–1529.
    1. Kang SJ. The bloodline of CD8α+ dendritic cells. Mol Cells 2012;34:219–229.

Metrics
Share
Figures

1 / 1

Tables

1 / 1

Funding Information
PERMALINK