key: cord-338092-barmkkwx authors: Geginat, Jens; Nizzoli, Giulia; Paroni, Moira; Maglie, Stefano; Larghi, Paola; Pascolo, Steve; Abrignani, Sergio title: Immunity to Pathogens Taught by Specialized Human Dendritic Cell Subsets date: 2015-10-13 journal: Front Immunol DOI: 10.3389/fimmu.2015.00527 sha: doc_id: 338092 cord_uid: barmkkwx Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that have a key role in immune responses because they bridge the innate and adaptive arms of the immune system. They mature upon recognition of pathogens and upregulate MHC molecules and costimulatory receptors to activate antigen-specific CD4(+) and CD8(+) T cells. It is now well established that DCs are not a homogeneous population but are composed of different subsets with specialized functions in immune responses to specific pathogens. Upon viral infections, plasmacytoid DCs (pDCs) rapidly produce large amounts of IFN-α, which has potent antiviral functions and activates several other immune cells. However, pDCs are not particularly potent APCs and induce the tolerogenic cytokine IL-10 in CD4(+) T cells. In contrast, myeloid DCs (mDCs) are very potent APCs and possess the unique capacity to prime naive T cells and consequently to initiate a primary adaptive immune response. Different subsets of mDCs with specialized functions have been identified. In mice, CD8α(+) mDCs capture antigenic material from necrotic cells, secrete high levels of IL-12, and prime Th1 and cytotoxic T-cell responses to control intracellular pathogens. Conversely, CD8α(−) mDCs preferentially prime CD4(+) T cells and promote Th2 or Th17 differentiation. BDCA-3(+) mDC2 are the human homologue of CD8α(+) mDCs, since they share the expression of several key molecules, the capacity to cross-present antigens to CD8(+) T-cells and to produce IFN-λ. However, although several features of the DC network are conserved between humans and mice, the expression of several toll-like receptors as well as the production of cytokines that regulate T-cell differentiation are different. Intriguingly, recent data suggest specific roles for human DC subsets in immune responses against individual pathogens. The biology of human DC subsets holds the promise to be exploitable in translational medicine, in particular for the development of vaccines against persistent infections or cancer. iNTRODUCTiON Human beings are constantly exposed to a myriad of pathogens, including bacteria, fungi, and viruses. These foreign invaders or cohabitants contain molecular structures that are sensed by the innate immune system, which mounts a first-line defense and also activates a pathogen-specific, adaptive immune response. The adaptive immune system is composed of B cells that produce specific antibodies, CD8 + T cells that can kill pathogen-infected cells, and CD4 + T cells that produce effector cytokines and coordinate the immune response. T cells express antigen receptors (T-cell antigen receptors, TCR) that recognize specific peptides presented on MHC molecules. CD8 + T cells recognize peptides presented by MHC class-I molecules that are ubiquitously expressed, whereas CD4 + T cells are activated by peptide-MHC class-II complexes, which are largely restricted to antigen-presenting cells (APCs). Dendritic cells (DCs) can express very high levels of MHC and costimulatory molecules, and it is generally accepted that they are the relevant cells to induce the activation ("priming") of antigen-specific "naive" T cells (1, 2) and induce their differentiation into various types of effector T cells. The elimination or containment of different types of pathogens requires dedicated classes of adaptive immune responses (3) . Thus, pathogens like viruses or intracellular bacteria require CD4 + and CD8 + T cells that produce IFN-γ and kill infected cells (Th1 and CTL, respectively). IL-12 is the critical cytokine that induces this type of response, but IL-12 production by DC is tightly controlled and requires several stimuli derived from pathogens and from CD4 + helper T cells (4) (5) (6) (7) (8) (9) . Conversely, extracellular bacteria and fungi require a different type of response that can be mediated by Th17 cells (10) (11) (12) . These effector cells are induced by proinflammatory cytokines produced by DC and macrophages (13) and attract neutrophils that in turn phagocytose extracellular bacteria (14) . A third type of effector response is the Th2 response, which is required to expel extracellular parasites such as helminths by activating eosinophils and basophils and by inducing antibodies of the IgE class (15) . IL-4 is the critical cytokine that induces this response (16) , but IL-4 is normally not produced by DC (17, 18) . Finally, these different effector responses have to be controlled by specialized regulatory T cells, in particular by IL-10-producing T cells ("Tr1 cells"), which are generated from effector cells and are important to avoid excessive tissue damage by adaptive immune responses (19) (20) (21) (22) . Cytokines that promote this type of regulatory T-cell response are IFN-α, IL-27, and IL-10 (23) (24) (25) , and all these cytokines can be produced by DCs (26, 27) . Professional APCs have to present pathogen-derived peptides on MHC molecules to activate antigen-specific T cells. DCs are phagocytic in the immature state, i.e., under steady-state conditions and upon initial pathogen encounter, and can take up antigenic material by pinocytosis or by surface receptor-mediated internalization (28) . Proteins from pathogens are then shuttled to lysosomes where they are chopped to peptides and loaded on MHC class-II molecules (29, 30) . These peptide-MHC complexes are then transported to the plasma membrane to activate specific CD4 + T cells. The presentation of peptides derived from exogenous proteins on MHC class-I, a process called cross-presentation (31, 32) , is a largely unique feature of DCs and is particularly important to activate CD8 + T cells in viral infections. Virus-infected cells express viral proteins in the cytosol where they are degraded to peptides by the proteasome, translocated to the endoplasmic reticulum by TAP proteins, and loaded on MHC class-I molecules (31) . However, since DCs are not necessarily infected by viruses, they must be able to process virus-derived proteins also from external sources, such as virus-infected cells, to activate CD8 + T cells. The mechanism of cross-presentation is still incompletely understood, but two distinct pathways via vacuoles and peptide translocation from phagolysosomes to the cytosol have been described (32) . It is believed that cross-presentation is the most important pathway leading to the induction of cytotoxic T-cell responses, and excellent reviews have been published on this relevant topic (31) (32) (33) . Naive T cells have a very high activation threshold (34) , and only professional APCs that express high levels of MHC and costimulatory molecules such as DCs are able to induce proliferation of naive T cells (35) . Several receptor-ligand interactions contribute to naive T-cell activation (36) (37) (38) , but CD28 costimulation is particularly important to amplify the signal transduced by the TCR (39) . Monocytes efficiently present peptides derived from extracellular proteins on MHC class-II to activate antigen-experienced CD4 + T cells (34) , and this capacity can be exploited to selectively expand antigen-specific memory T cells (40) . However, monocytes have an approximately 1000-fold lower capacity to prime naive CD4 + T cells as compared to DCs (Nizzoli et al., under review) and home to non-lymphoid tissues in the steady state. However, upon inflammation, they can differentiate to inflammatory DCs (41) and home to lymph nodes where they can activate T cells (42, 43) . In addition, there is some evidence that CD16 + subsets of human blood monocytes might contain DCs (27, 44, 45) . Naive T cells constantly recirculate in the blood and migrate through secondary lymphoid organs (46) , but are largely excluded from non-lymphoid tissues. In secondary lymphoid tissues, they migrate to the T-cell zone, where they encounter DCs (47) . B cells are also present in secondary lymphoid organs and can potently present antigen to T cells when they internalize and process antigens that have specifically bound to their B-cell receptor (48) . However, B cells are physically separated from naive T cells in lymph nodes and only following TCR activation naive T cells migrate to the B-cell zone where they interact with antigen-specific B cells to induce antibody production (49, 50) . Thus, antigen presentation by B cells appears to be important for the activation of antigen-experienced T cells rather than for naive T-cell priming. Dendritic cells are generated from committed precursors in the bone marrow that are released into the circulation to seed peripheral organs (51) (52) (53) (54) (55) . Both monocytes and DCs can be derived from common myeloid progenitors (CMPs), but committed precursors that selectively give rise to monocytes or DCs (51) or even selected DC subsets (53, 54) have been identified in humans and mice. DCs are poorly stimulatory in the immature state and can induce a partial T-cell activation, leading to deletion of autoreactive CD8 + T cells (56) (57) (58) (59) . In addition, they promote self-tolerance by inducing Foxp3 + regulatory CD4 + T cells that suppress autoreactive T cells (60) . Pathogens induce the maturation of DCs that consequently acquire the capacity to produce polarizing cytokines and to prime pathogen-specific effector T-cell responses. Pathogen-derived molecular patterns [PAMPs (61, 62) ] are recognized by DCs and lead to the efficient presentation of antigens to T cells (63) . There are different classes of pathogen-sensing receptors, including Toll-like receptors (62, 64) , nucleotide-binding oligomerization domain (NOD)like receptors (65) , retinoic acid-inducible gene 1 (RIG-I)-like receptors (66) , and C-type lectins (67) . TLRs recognize different PAMPs, including nucleic acids or cell wall components such as proteins and lipoproteins (68, 69) . In the case of viruses, nucleic acids are sensed not only by different TLRs in endosomes but also by cytosolic receptors like RIG-I (66, 70) and induce a potent activation of DCs. Importantly, subsets of DCs express different patterns of pathogen-sensing receptors and might thus preferentially respond to individual pathogens (71, 72) . DNA viruses such as cytomegalovirus (CMV) and herpes simplex virus (HSV) and also bacteria can activate DCs via unmethylated CpG-containing DNA (69) , which is sensed by TLR9. Double-and single-stranded RNAs, which are generated by both DNA and RNA viruses, are sensed by DCs via TLR3 (73) and TLR7/8 (74, 75) , respectively. Of note, TLR3 is restricted to mDCs (71) and induces cross-presentation capacities (76) . Viruses such as respiratory syncytial virus (RSV) and hepatitis C virus (HCV) can also activate DCs via TLR2 or TLR4, which are expressed on the plasma membrane and recognize viral proteins (77) . TLR2 is also involved in immune responses to fungi (78) and Gram-positive bacteria (79, 80) while TLR4 recognizes lipopolysaccharide (LPS) (81), a cell membrane compound of Gram-negative bacteria. Many pathogens like viruses activate DCs via multiple TLRs (77) . Moreover, other immune cells, including T cells themselves, feed-back on DCs to regulate the ongoing response. In particular, CD40 stimulation by CD4 + helper T cells is crucial for CD8 + T-cell stimulation and IL-12 production (4, 5). Moreover, IFN-γ (6) and paradoxically also IL-4 (7, 8) that can be provided by T cells further enhance IL-12 production (9). Surface TLRs such as TLR2 and TLR4 signal via the adaptor protein Myd88 (82) to induce the activation of Map kinases and the nuclear translocation of the transcription factor NF-κB, which in turn induces the transcription of proinflammatory cytokines (62) . Endosomal TLRs 7, 8, and 9 also signal via Myd88 but activate IRF7, which in turn induces type-1 interferon production (83, 84) . TLR3 is an exception since it does not signal via Myd88 but utilizes TRIF (85) to activate IRF3 (86, 87) or IRF7 (88) . How all these complex signaling pathways are integrated by DCs to induce the appropriate T-cell response is still incompletely understood (88) (89) (90) . Dendritic cells in mice can be subdivided into distinct subsets with specific functions. Some DCs are stably resident in lymph nodes while others are positioned in non-lymphoid tissues to sense tissue-invading pathogens, but are migratory and are recruited via the lymph following pathogen encounter in a CCR7-dependent manner (91, 92) . In secondary lymphoid tissues, two major DC subsets are myeloid DCs (mDCs) and plasmacytoid DCs (pDCs; Table 1 ) (72, 93, 94) . Both pDCs and mDCs upregulate MHC and costimulatory molecules like CD80 and CD86 upon maturation (72) that bind to CD28 and are required to induce full T-cell stimulation (39) . However, pDCs are poorly phagocytic and have a different regulation of MHC class-II turnover upon maturation as compared to mDCs (95) . Thus, mDCs stop phagocytosis and peptide loading on MHC upon pathogen recognition and stably present peptides from antigenic material they had acquired upon pathogen encounter (30, 96, 97) . This maturation-induced stabilization of peptide-MHC complexes enhances the priming of pathogenspecific T cells by mDCs. In contrast, pDCs continue to present new peptides on MHC complexes even in the mature stage (95) . On the one hand, this limits their capacity to stimulate pathogen-specific T cells; on the other hand, this enables them to present also late-expressed viral antigens when they are actively infected. This diverse regulation of MHC-peptide stability in mDCs and pDCs suggests that they present different antigens to T cells. n/a n/a n/a Plasmacytoid DCs are present in lymph nodes and are largely absent from non-lymphoid organs, but they can be recruited upon inflammation (98) . The role of pDC in T-cell priming is still debated (99) . There is consensus that they are poorly stimulatory in their resting state (100, 101), but while some groups proposed that they become potent APCs following TLR stimulation and prime CD4 + and cross-prime CD8 + T-cell responses (102) (103) (104) (105) , others concluded that also mature pDCs have only low priming and cross-priming capacities and might rather be tolerogenic (101) . The rapid and abundant production of type-1 interferon by pDC suggests a pivotal role in viral infections, even if their capacity to prime virus-specific T cells directly appears to be limited. IFN-α can also be produced by other immune cells and by virus-infected cells, but the early and systemic IFN-α response is believed to depend on pDCs (101) . Consistently, in the case of HSV infections, it was shown that pDCs were important for systemic but not local protection (106) . However, in several other viral infections in mice, including vesicular stomatitis virus (VSV), lymphocytic choriomeningitis virus (LCMV), RSV, and mouse cytomegalovirus (MCMV), pDCs do not seem to play a major role (100) . In marked contrast, in mouse hepatitis virus (MHV) infection, the antiviral response against this coronavirus was largely pDC dependent (107) (Figure 1) . Finally, pDCs have been found by several groups to induce the production of the anti-inflammatory cytokine IL-10 by CD4 + T cells, suggesting that they might be important to inhibit excessive T-cell responses. Several proteins expressed by pDCs were found to promote IL-10 induction in T cells, including the Notch ligand Delta-like 4 (108), ICOSL (109, 110) , as well as IFN-α (23, Nizzoli et al., under review). Myeloid DCs are a heterogeneous population, and different mDC subsets can be identified that preferentially initiate different types of adaptive immune responses (Figure 1 ). In the spleen of mice, mDCs can be subdivided into CD8α + and CD8α − subsets ( Table 1) . CD8α + mDCs produce high levels of bioactive IL-12p70 and efficiently cross-prime CD8 + T-cell responses (111) . They express CLEC9A, a C-type lectin, that enables them to take up antigenic material from dying cells, and their generation was shown to rely on the transcription factors BATF3 and IRF8 (112, 113) . Moreover, they express the chemokine receptor XCR1 that favors their colocalization with CD8 + T cells (114) . Altogether the present evidence indicates that CD8α + DCs are specialized to induce Th1 and CTL responses in response to intracellular pathogens (115, 116) . Notably, DCs in the gut that express CD103 have similar characteristics and are closely related to CD8α + DC (117, 118) . CD8α − DCs express CD11b and can be further subdivided into CD4 + and CD4 − CD8 − subsets. They preferentially prime CD4 + T-cell responses (119) and promote Th17 responses, but they can also induce Th2 cells (113, 120) . Interestingly, CD11b + DCs produce IL-23 in the gut and are required for protection against Citrobacter rodentium (121) . Their generation depends on the transcription factor IRF4, while KLF4 expression is required for Th2, but not for Th17 induction (122) . Notably, however, CD8α − DCs and also pDCs can cross-prime CD8 + T-cell responses under certain conditions (102) (103) (104) 123) . Moreover, it was shown that upon appropriate microbial stimulation all mDC subsets have the potential to promote either Th1 or Th2 responses (124) . Thus, although the proposed functional specialization of DC subsets is an intriguing and helpful concept, it might also be an oversimplification, since DC subsets have considerable plasticity and the induction of a specific type of immune response critically depends on the stimuli they receive from pathogens as well as from other immune cells (125) . High numbers of human DCs can be generated in vitro by culturing monocytes with cytokines (41) , and the large majority of studies on human DCs have been done with these monocytederived DCs. They are primary cells and show many behaviors of in vivo occurring DCs, including cytokine production as well as stable and potent antigen presentation upon maturation with TLR ligands (125) . However, monocyte-derived DCs are not the appropriate model to study the role of specialized DC subsets in human immune responses. Dendritic cells circulating at low frequency in human peripheral blood share several features with murine splenic DC subsets (126) ( Table 1) . Human pDCs have been identified more than 15 years ago as the natural IFN-α-producing cells (127, 128) . They express TLR7 and TLR9 and produce large amounts of IFN-α in response to CpG DNA or influenza virus. Similar to their murine counterparts, they are poorly stimulatory (94) , express the C-type lectin BDCA-2 (93), and induce IL-10 production in CD4 + T cells (129) . In addition, subsets of mDCs can also be found in human blood and in tissues (130) (131) (132) (133) . As their murine homologues, they express CD11c and potently prime CD4 + and CD8 + T-cell responses. The expression of CD1c/BDCA-1 and CD141/BDCA-3 identifies two subsets among human mDCs in peripheral blood (93) and also in secondary lymphoid organs (105, 132, 134, 135) . BDCA-3 + "mDC2" ( Table 1 ) are rare, but it could recently be demonstrated that they represent the human counterpart of murine CD8α + DCs (136) (137) (138) (139) (140) . Thus, as CD8α + DCs, they selectively express CLEC9A and XCR1 and are dependent on the transcription factor BATF3 (112, 136, 138, 140, 141) . Importantly, they can cross-present exogenous antigens on MHC class-I to CD8 + T cells and produce IL-12 (134) (135) (136) . CD1c + "mDC1" ( Table 1 ) are more frequent and share some features with CD8α − DC, including CD11b expression and IL-23 production (121, 142, Nizzoli et al., under review). Also TLR3 expression in DC subsets appears to be similar in humans and mice, since it is expressed at high levels on CD8α + DCs and mDC2, at lower levels on CD8α − DCs and mDC1, and absent on pDC. Surprisingly, TLR3 in mice is not required for immune responses against several viruses, including LCMV, VSV, MCMV, and Reovirus, suggesting that TLR3 has not a pivotal role in antiviral immune defense (143) . Consistently, TLR3 deficiency in humans selectively leads to uncontrolled HSV1 infections in the central nervous system (CNS) (144) . Different subsets of DC have also been identified in human non-lymphoid tissues where they are strategically positioned to recognize invading pathogens, in particular at barrier surfaces. These migratory DC subsets play a crucial role to transport antigenic material of pathogens that invade specific tissues to draining lymph nodes and thus to initiate a tissue-specific T-cell response (130, 145, 146) . Human Langerhans cells were first described more than a century ago and reside in the epidermis and are thus the first DCs that encounter skin-invading pathogens. Upon activation, they mature and migrate to draining lymph nodes to activate CD4 + and CD8 + T cells. In the dermis, different subsets of interstitial DCs are present and can be classified according to CD14, CD1a, and CD141 expression. Dermal CD14 + cells might represent monocyte-derived macrophages rather then DCs (147) , but CD1a + and CD141 + DCs, respectively, resemble the CD1c + and CD141 + DC subsets in peripheral blood (148) . Also in the lung and the liver, DC subsets that are related to CD1c + and CD141 + DCs could be identified (133) . Finally, in the human intestine, DC subsets that express CD11b and CD103 are similar to CD1c + and CD141 + DCs, respectively, and these intestinal DC subsets are also largely conserved between humans and mice (149) . Although the similarities between human and mouse DC subsets are often emphasized, there are also some important differences in pathogen sensing by DCs in humans and mice (150) . Importantly, the expression of several relevant TLRs is not conserved (Figure 1) , presumably because humans and mice have evolved under the selective pressure of different pathogens. Thus, in mice, TLR7 and TLR9 are expressed by both pDC and mDC subsets (71) , whereas in humans, they are restricted to pDCs (72) . Also TLR4 expression is more restricted in human DCs, since it is expressed by mDC1 but not by mDC2 (136) . Moreover, TLR8 is not expressed by human pDCs (72) , and some agonists of human TLR8 such as the resiquimod R848 do not activate murine TLR8 (75, 151) . Another relevant difference seems to be the role of the adaptor protein Myd88, which transduces signals from all TLRs with the notable exception of TLR3. Thus, mice deficient for Myd88 are highly susceptible to several infections by bacteria, viruses, parasites, and fungi. Conversely, Myd88-deficient patients are selectively affected by infections with pyogenic bacteria in childhood (152) . Finally, human CD1c + DCs and also Langerhans cells seem to have superior capacities to cross-present antigens and to induce CTL responses as compared to their murine homologues (105, 134, 153, 154) . Overall, these differences in pathogen sensing and T-cell activation between human and murine DCs are likely to have an important impact on their role in immune responses against specific pathogens. Dendritic cell subsets in humans and mice express not only different patterns of toll-like receptors, but they have also partially distinct cytokine profiles (Figure 1) . In particular, human mDC1 have a complex and quite unique regulation of cytokine production. Thus, while LPS triggers only low levels of cytokine production by mDC1, dual TLR stimulation with LPS or Poly-I:C (TLR3 ligand) in combination with R848 induces very high levels of a broad panel of cytokines, including TNF, IL-6, IL-10, IL-12, and IL-23 (Nizzoli et al., under review) . The very potent cytokine-producing capacity of mDC1 has been missed in several studies where mDC1 were activated with single TLR ligands (45, 155, 156) . Of note, single TLR stimulation is sufficient to induce antiviral cytokines by mDC2 and pDCs (see below) and proinflammatory cytokines by monocytes. Although mDC1 can secrete several proinflammatory cytokines that promote Th17 cell generation including IL-23 (142) , it is unclear if they are the physiological inducers of Th17 cells or if monocyte-derived, inflammatory DCs do the job (12, 157) . Also the identity of the DC subset that induces human Th2 responses is still enigmatic. It was originally proposed that mDCs induce Th1 polarization and pDCs Th2, but later it was shown that also pDCs can drive Th1 responses (158, 159) . More recently, mDC2 but not mDC1 were found to induce Th2 cells in an aberrant response to influenza virus (160) . In apparent contrast to CD8α − DCs, mDC1 can produce high levels of IL-12 (134, 135) , suggesting a relevant role in immune responses against intracellular pathogens. Moreover, the production of the anti-inflammatory cytokine IL-10, which can be produced by all mDCs in mice, is largely restricted to mDC1 in humans (Nizzoli et al., under review) . Stimulation of mDC1 with the intestinal bacterium Escherichia coli or with LPS alone induces IL-10 and was proposed to induce a tolerogenic state in mDC1 (155) . Although IL-10 is indeed a tolerogenic cytokine and a well-established negative regulator of DC maturation and cytokine production (161) , it can paradoxically also have positive effects, in particular on CD8 + T-cell responses (162, 163) . Consistently, we found that IL-10 produced by mDC1 completely blocked the cross-priming of low-affinity CTL and enhanced the responsiveness of CD8 + memory T cells to the homeostatic cytokine IL-15. Thus, mDC1-derived IL-10 appears to play an important positive role in CTL responses, since it selects high affinity cells upon priming and inhibits CTL memory attrition at the same time (Nizzoli et al., under review) . While mDC1 can secrete a broad panel of pro-and antiinflammatory cytokines, mDC2 and pDC are largely dedicated to secrete high levels of antiviral cytokines. The subset-specific production of IFN-α by pDC (128) and of IFN-λ by CD8α + and mDC2 (134, 137) appears to be largely conserved between humans and mice. The very potent IFN-λ-producing capacities of BDCA-3 + DC (134, 137) suggest that analogous to pDCs they might be the relevant source of early and systemic IFN-λ in viral infections. Notably, IFN-λ has antiproliferative and antiviral activities similar to type-I interferon, but the expression of the IFN-λ receptor is much more restricted and found mainly on epithelial cells at barrier surfaces and in the liver (164) . MDC2 can also secrete selected isoforms of IFN-α (165) and some IL-12 (134) (135) (136) 138) , consistent with the view that they play an important role in antiviral immune responses. As previously mentioned for murine pDCs, IFN-α is not only a powerful antiviral cytokine that activates several different types of immune cells, but it also induces IL-10 production in CD4 + T cells, suggesting that pDCs induce Tr1-like regulatory T cells also in humans (21, 23, 108, Nizzoli et al., under review). The more restricted expression of TLRs and the specific cytokineproducing capacities of human DC subset suggest that they play unique roles in immune responses against individual pathogens. The roles of human DC subsets in pathogen-specific immune response are however difficult to address directly because patients that selectively lack a DC subset of interest have not been identified so far. Nevertheless, some interesting findings were reported. In particular, mDC2 appear to be highly relevant in HCV infection. Single-nucleotide polymorphisms in the IFN-λ3 gene locus are strongly associated with spontaneous clearance and response to therapy in HCV patients (166) . All DC subsets can secrete some IFN-λ1 (134, 167) , but mDC2 produce much higher amounts. Moreover, IFN-λ2/3 are largely restricted to mDC2, and importantly HCV induces IFN-λ3 production by mDC2 (168, Nizzoli et al., under review) . Thus, mDC2 appear to be a highly relevant source for protective IFN-λ3 in HCV infection (169) . Interestingly, an important role for mDC1 rather than for mDC2 was recently proposed in tuberculosis (170, 171) . Thus, mDC1 were more efficiently infected with the Bacillus Calmette-Guérin (BCG) vaccine than other DCs and induced the activation of pDCs and CD8 + T cells. Notably, mDC1 could not be replaced by mDC2 in this system, suggesting that mDC1 could play a non-redundant role in the defense against selected intracellular pathogens. MDC1 and mDC2 have also been suggested to play different roles in RSV infection (172, 173) . Thus, mDC subsets produced different cytokines in response to RSV, consistent with their different cytokine profiles upon stimulation with purified TLR ligands (134, Nizzoli et al., under review) . Moreover, they induced different classes of T-cell responses, with mDC1 inducing preferentially Th1 cells and mDC2 inducing predominantly Th2 and T-regulatory cells. Similarly, mDC2, but not mDC1, were found to induce Th2 response to influenza virus (160) . Also the capacity of pDCs to induce IL-10-producing regulatory T cells has been documented with a relevant pathogen, since pDCs were shown to induce IFN-γ and IL-10 production in antigen-experienced CD4 + T cells specific for mumps virus (129) . Conversely, CD11c + mDCs, which contain both mDC1 and mDC2, induced IFN-γ and, surprisingly, IL-5. It is largely accepted that pDC-derived IFN-α is important to contain human viral infections. Thus, stabilized pegylated IFN-α is a widely used therapy for HCV patients. IFN-λ appears to be similar effective, but is less toxic presumably because of the more restricted expression of its receptor (164) . Interestingly, the HCV glycoprotein E2 is a ligand for BDCA-2, which is specifically expressed on pDCs ( Table 1 ) and inhibits IFN-α production (174, 175) . In this way, HCV might inhibit IFN-α production to establish chronic infection. Finally, pDCs are also targeted by human immunodeficiency virus (HIV), but whether they play a protective or detrimental role is still unclear (176) . Vaccines have been a major breakthrough for human health. Attenuated or killed pathogens are highly efficient to induce protective cellular and humoral immune responses, and the induced protective memory can last for a lifetime (177, 178) . However, since these pathogen-based vaccines also have considerable side effects, proteins in combination with adjuvants that activate APCs are more often used. Protein vaccines induce CD4 + T-cell responses and neutralizing antibodies, but they are poorly efficient in inducing cytotoxic T-cell responses and are also rather inefficient in inducing Th1 cells (179, 180) . Frequently used adjuvants are alum, oil-in-water emulsions like MF59, and more recently also monophosphoryl lipid A (MPL), a detoxified form of LPS. In mice, different adjuvants were shown to induce different proinflammatory cytokines. Thus, alum acts via uric acid on inflammatory DCs (181) , which leads to NOD-like receptor protein-3 (NALP3)-dependent IL-1β production (182) . Conversely, MPL does not induce IL-1β (183) but induces specific antibodies through an IL-6-dependent mechanism (184), while MF-59 and alum act independently of IL-6 (185). However, the different TLR expression and cytokine production by human APC subsets should be considered when translating this knowledge from animal models to patients. A recent interesting report analyzed the response of APC subsets to 13 different vaccines and concluded that different vaccines activate indeed different APC populations (186) . More direct information on the effect of DCs was obtained by vaccinations with peptide-pulsed monocyte-derived DCs in cancer patients, which can induce tumor-specific CD8 + T cells (187) , but the clinical responses were so far largely insufficient. MDCs might be more potent and are currently tested in clinical trials. Nucleic acid-sensing TLRs are particularly potent to induce CD8 + T-cell responses in mice (188) and have recently been employed as adjuvants in vaccines. Examples are CpG-DNA that stimulates TLR9 (189) , and the TLR7 ligand imiquimod, which is used as a cream to stimulate DC locally in the skin, and was shown to induce CD8 + T-cell responses in situ (190) . Vaccines consisting of plasmid DNA coding for relevant protein antigens are a novel approach that efficiently induces humoral and cellular immune responses in animals. However, in humans, these DNA vaccines are often poorly immunogenic (191) , presumably because they have only low adjuvant activity and stimulate mainly cytosolic DNA sensors rather than TLR9 (192) , which in addition is restricted to pDCs and B cells in humans. An alternative promising approach is the vaccination with mRNA (193, 194) , which delivers not only the antigenic protein directly to the cytosol, thereby bypassing the requirements for cross-presentation (195) , but also induces mDC and pDC maturation and cytokine production via TLR7/8 at the same time (196) . Indeed, intradermal injection of naked mRNA results in local uptake and translation of the nucleic acid (197) followed by the development of an adaptive immunity in mice (198) and in humans (199, 200) . Since also lymph node-resident DCs are expected to be appropriate APCs to process antigens encoded by mRNA, direct injection of nucleic acid into lymph nodes has also been evaluated. In animal models, intra-lymph node injections of mRNA result in expression of the protein encoded by the mRNA in DCs. Furthermore, the injected mRNA activated lymph node-resident APCs and induced potent CD4 + and CD8 + T-cell responses as well as prophylactic and therapeutic antitumor immunity (201) . The approach is currently being evaluated through two clinical studies exploring the efficacy of intra-lymph node mRNA vaccination in advanced melanoma patients. As a further development, systemic administration of a liposomal formulation of mRNA that delivers the nucleic acids to APCs present in secondary lymphoid organs is also being evaluated. Using the functional diversity of DCs in vivo, and their specific capabilities in generating appropriate adaptive immune responses, those systemic synthetic vaccines might recapitulate the natural mechanisms of immunity developed during pathogen infection and guarantee the development of therapeutically efficacious immune responses. Dendritic cells continue to attract much interest of immunologists because they are the most potent APCs in the immune system and are the principal inducers of naive T-cell differentiation. Intensive research in the last years has established that different subsets of DC exist in mice that have specialized functions and preferentially induce different types of immune responses. In humans, much has been learned from in vitro differentiated monocytederived DCs, and more recently, also different subsets of DC populating human tissues have been analyzed at the molecular and functional levels. It is fundamental to further define the biology of these in vivo occurring human DC subsets to understand and cure pathogenic immune-mediated processes in so different settings as autoimmunity, infections, and cancer. In particular, appropriate targeting of DC subsets by vaccines holds the promise to induce cytotoxic T-cell responses to eradicate persistent intracellular pathogens or tumors. ACKNOwLeDGMeNTS JG, GN, and SA are supported by the CARIPLO foundation, SA is supported by the European Research Council, and the INGM is supported by the "Romeo ed Enrica Invernizzi" foundation. The dendritic cell system and its role in immunogenicity Immunobiology of dendritic cells The CD4-centered universe of human T cell subsets Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation High level IL-12 production by murine dendritic cells: upregulation via MHC class II and CD40 molecules and downregulation by IL-4 and IL-10 Differential regulation of human blood dendritic cell subsets by IFNs IL-4 is a mediator of IL-12p70 induction by human Th2 cells: reversal of polarized Th2 phenotype by dendritic cells Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 production by mouse and human dendritic cells Differential production of IL-12, IFN-α, and IFN-γ by mouse dendritic cell subsets Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3 Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells Induction, function and regulation of IL-17-producing T cells TH2 and 'TH2-like' cells in allergy and asthma: pharmacological perspectives Disruption of the murine IL-4 gene blocks Th2 cytokine responses Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation TH2, allergy and group 2 innate lymphoid cells Conventional T-bet(+)Foxp3(-) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis Identification and characterization of IL-10/IFN-gamma-producing effector-like T cells with regulatory function in human blood Th17 cells express interleukin-10 receptor and are controlled by Foxp3 and Foxp3+ regulatory CD4+ T cells in an interleukin-10-dependent manner IFN-alpha and IL-10 induce the differentiation of human type 1 T regulatory cells IL-27 is a key regulator of IL-10 and IL-17 production by human CD4+ T cells IFN-alpha and CD46 stimulation are associated with active lupus and skew natural T regulatory cell differentiation to type 1 regulatory T (Tr1) cells IL-10 and IL-27 producing dendritic cells capable of enhancing IL-10 production of T cells are induced in oral tolerance Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: downregulation by cytokines and bacterial products The formation of immunogenic major histocompatibility complex class II-peptide ligands in lysosomal compartments of dendritic cells is regulated by inflammatory stimuli Transport of peptide-MHC class II complexes in developing dendritic cells Cross-presentation in viral immunity and self-tolerance Cross-presentation by dendritic cells The biochemistry and cell biology of antigen processing and presentation Enhanced responsiveness of human memory T cells to CD2 and CD3 receptor-mediated activation Generation of antigen-specific CD4+ T cell lines from naive precursors CD28 and LFA-1 contribute to cyclosporin A-resistant T cell growth by stabilizing the IL-2 mRNA through distinct signaling pathways OX40 costimulation enhances interleukin-4 (IL-4) expression at priming and promotes the differentiation of naive human CD4(+) T cells into high IL-4-producing effectors Analysis of 4-1BB ligand (4-1BBL)-deficient mice and of mice lacking both 4-1BBL and CD28 reveals a role for 4-1BBL in skin allograft rejection and in the cytotoxic T cell response to influenza virus Accessory cell-derived signals required for T cell activation Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells 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 Microbial stimulation fully differentiates monocytes to DC-SIGN/ CD209(+) dendritic cells for immune T cell areas Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses Human 6-sulfo LacNAc-expressing dendritic cells are principal producers of early interleukin-12 and are controlled by erythrocytes Functional specialization of human circulating CD16 and CD1c myeloid dendritic-cell subsets Naive and memory T cells show distinct pathways of lymphocyte recirculation Dendritic cells in the T-cell areas of lymphoid organs Antigen-specific interaction between T and B cells Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production Shaping up adaptive immunity: the impact of CCR7 and CXCR5 on lymphocyte trafficking Development and function of dendritic cell subsets In vivo analysis of dendritic cell development and homeostasis Circulating precursors of human CD1c+ and CD141+ dendritic cells Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen T cell fitness determined by signal strength CD4+ regulatory T cells: mechanisms of induction and effector function The immune system evolved to discriminate infectious nonself from noninfectious self A human homologue of the Drosophila toll protein signals activation of adaptive immunity Origin, maturation and antigen presenting function of dendritic cells A family of human receptors structurally related to Drosophila toll NOD-like receptors: versatile cytosolic sentinels RIG-I: tri-ing to discriminate between self and non-self RNA Myeloid C-type lectin receptors in pathogen recognition and host defense Toll-like receptors: a growing family of immune receptors that are differentially expressed and regulated by different leukocytes A toll-like receptor recognizes bacterial DNA Deficient signaling in mice devoid of double-stranded RNA-dependent protein kinase Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8 alpha+ DC correlates with unresponsiveness to imidazoquinolines Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells Recognition of double-stranded RNA and activation of NF-kappaB by toll-like receptor 3 Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8 Toll-like receptor 3 promotes cross-priming to virus-infected cells Toll-like receptors and viruses: induction of innate antiviral immune responses Fungal recognition by TLR2 and dectin-1 Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components Toll-like receptor 2 (TLR2) mediates astrocyte activation in response to the Gram-positive bacterium Staphylococcus aureus Cutting edge: toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product MyD88 is an adaptor protein in the hToll/IL-1 receptor family signaling pathways IRF-7 is the master regulator of type-I interferon-dependent immune responses Interferon-alpha induction through toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6 Cutting edge: a novel Toll/IL-1 receptor domain-containing adapter that preferentially activates the IFN-beta promoter in the toll-like receptor signaling IRF3 mediates a TLR3/TLR4-specific antiviral gene program Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway How toll-like receptors signal: what we know and what we don't know Duration, combination and timing: the signal integration model of dendritic cell activation Cutting edge: synchronization of IRF1, JunB, and C/EBPbeta activities during TLR3-TLR7 cross-talk orchestrates timely cytokine synergy in the proinflammatory response Differential regulation of chemokine receptors during dendritic cell maturation: a model for their trafficking properties Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood Characterization of human blood dendritic cell subsets Developmental regulation of MHC II expression and transport in human plasmacytoid-derived dendritic cells Developmental regulation of MHC class II transport in mouse dendritic cells Capture and processing of exogenous antigens for presentation on MHC molecules Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules Antigen-presentation properties of plasmacytoid dendritic cells Plasmacytoid dendritic cells: recent progress and open questions Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation Antigen crosspresentation by human plasmacytoid dendritic cells Human plasmacytoid dendritic cells efficiently cross-present exogenous Ags to CD8+ T-cells, despite lower Ag uptake than myeloid dendritic cell subsets Similar antigen cross-presentation capacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells Plasmacytoid dendritic cells contribute to systemic but not local antiviral responses to HSV infections Control of coronavirus infection through plasmacytoid dendritic-cell-derived type I interferon Cutting edge: plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta-like-4/Notch axis ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28 Plasmacytoid dendritic cells prime IL-10-producing T regulatory cells by inducible costimulator ligand The CD8+ dendritic cell subset DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues Transcriptional control of dendritic cell development Selective expression of the chemokine receptor XCR1 on cross-presenting dendritic cells determines cooperation with CD8+ T cells Cytokines regulate the capacity of CD8alpha(+) and CD8alpha(-) dendritic cells to prime Th1/Th2 cells in vivo Cross-presenting dendritic cells are required for control of Leishmania major infection Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells Dendritic cells in intestinal homeostasis and disease Differential antigen processing by dendritic cell subsets in vivo CD8alpha+ and CD8alpha− subclasses of dendritic cells direct the development of distinct T helper cells in vivo Notch2-dependent classical dendritic cells orchestrate intestinal immunity to attaching-and-effacing bacterial pathogens Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses Constitutive versus activation-dependent cross-presentation of immune complexes by CD8(+) and CD8(-) dendritic cells in vivo The ability of murine dendritic cell subsets to direct T helper cell differentiation is dependent on microbial signals The instructive role of dendritic cells on T-cell responses Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature The nature of the principal type 1 interferon-producing cells in human blood Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon Plasmacytoid dendritic cells induce a distinct cytokine pattern in virus-specific CD4+ memory T cells that is modulated by CpG oligodeoxynucleotides Characterization of resident and migratory dendritic cells in human lymph nodes The number and distribution of blood dendritic cells in the epidermis and dermis of healthy human subjects CD1c+ and CD303+ dendritic cells in peripheral blood, lymph nodes and tumor tissue of patients with non-small cell lung cancer Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T cell responses Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens Mouse CD8alpha+ DCs and human BDCA3+ DCs are major producers of IFN-lambda in response to poly IC Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells Found in translation: the human equivalent of mouse CD8+ dendritic cells The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells Human intestinal lamina propria CD1c+ dendritic cells display an activated phenotype at steady state and produce IL-23 in response to TLR7/8 stimulation Does toll-like receptor 3 play a biological role in virus infections? TLR3 deficiency in patients with herpes simplex encephalitis Human dendritic cells -stars in the skin CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived Frontiers in Immunology | www.frontiersin.org antigens irrespective of the presence of Langerhans cells Human dermal CD14(+) cells are a transient population of monocyte-derived macrophages Regulation of dendritic cell function in inflammation Comparative transcriptional and functional profiling defines conserved programs of intestinal DC differentiation in humans and mice Experimental and natural infections in MyD88-and IRAK-4-deficient mice and humans Recognition of nucleic acid and nucleic acid analogs by toll-like receptors 7, 8 and 9 Pyogenic bacterial infections in humans with MyD88 deficiency Modular expression analysis reveals functional conservation between human Langerhans cells and mouse cross-priming dendritic cells Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation Human CD1c (BDCA-1)+ myeloid dendritic cells secrete IL-10 and display an immuno-regulatory phenotype and function in response to Escherichia coli Selected toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells Human inflammatory dendritic cells induce Th17 cell differentiation Reciprocal control of T helper cell and dendritic cell differentiation Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent Th1 polarization Human CD141+ dendritic cells induce CD4+ T cells to produce type 2 cytokines Biology of interleukin-10 Inhibitory and stimulatory effects of IL-10 on human CD8+ T cells An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells Interferon-lambda and therapy for chronic hepatitis C virus infection CD141+ dendritic cells produce prominent amounts of IFN-alpha after dsRNA recognition and can be targeted via DEC-205 in humanized mice Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance IL-4 enhances IFN-lambda1 (IL-29) production by plasmacytoid DCs via monocyte secretion of IL-1Ra Human type 2 myeloid dendritic cells produce interferon-lambda and amplify interferon-alpha in response to hepatitis C virus infection Human blood dendritic cell antigen 3 (BDCA3)(+) dendritic cells are a potent producer of interferon-lambda in response to hepatitis C virus Communication between human dendritic cell subsets in tuberculosis: requirements for naive CD4(+) T cell stimulation Crosstalk between human DC subsets promotes antibacterial activity and CD8+ T-cell stimulation in response to bacille Calmette-Guerin Differential response of BDCA-1+ and BDCA-3+ myeloid dendritic cells to respiratory syncytial virus infection Paramyxovirus infection regulates T cell responses by BDCA-1+ and BDCA-3+ myeloid dendritic cells HCV glycoprotein E2 is a novel BDCA-2 ligand and acts as an inhibitor of IFN production by plasmacytoid dendritic cells BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction Plasmacytoid dendritic cells in HIV infection Duration of antiviral immunity after smallpox vaccination Yellow fever vaccine Protein vaccines induce uncommitted IL-2-secreting human and mouse CD4 T cells, whereas infections induce more IFN-gamma-secreting cells The form of NY-ESO-1 antigen has an impact on the clinical efficacy of anti-tumor vaccination Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells Gout-associated uric acid crystals activate the NALP3 inflammasome A potent adjuvant monophosphoryl lipid A triggers various immune responses, but not secretion of IL-1beta or activation of caspase-1 Interleukin-6 has differential influence on the ability of adjuvant formulations to potentiate antibody responses to a Plasmodium falciparum blood-stage vaccine Neither interleukin-6 nor signalling via tumour necrosis factor receptor-1 contribute to the adjuvant activity of Alum and Freund's adjuvant Transcriptional specialization of human dendritic cell subsets in response to microbial vaccines Harnessing dendritic cells to generate cancer vaccines Differential ability of surface and endosomal TLRs to induce CD8 T cell responses in vivo Clinical evaluation of CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases and cancer Topical rather than intradermal application of the TLR7 ligand imiquimod leads to human dermal dendritic cell maturation and CD8+ T-cell cross-priming Vaccine adjuvant systems containing monophosphoryl lipid A and QS21 induce strong and persistent humoral and T cell responses against hepatitis B surface antigen in healthy adult volunteers DNA vaccines: a simple DNA sensing matter? RNA-based vaccines The messenger's great message for vaccination Human peripheral blood mononuclear cells transfected with messenger RNA stimulate antigen-specific cytotoxic T-lymphocytes in vitro Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies Results of the first phase I/II clinical vaccination trial with direct injection of mRNA Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity