SlideShare a Scribd company logo
1 of 13
Download to read offline
REVIEWS




                                   Transcriptional programming of the
                                   dendritic cell network
                                   Gabrielle T. Belz and Stephen L. Nutt
                                   Abstract | Specialized subsets of dendritic cells (DCs) provide a crucial link between the
                                   innate and adaptive immune responses. The genetic programme that coordinates these
                                   distinct DC subsets is controlled by both cytokines and transcription factors. The initial steps
                                   in DC specification occur in the bone marrow and result in the generation of precursors
                                   committed to either the plasmacytoid or conventional DC pathways. DCs undergo further
                                   differentiation and lineage diversification in peripheral organs in response to local
                                   environmental cues. In this Review, we discuss new evidence regarding the coordination
                                   of the specification and commitment of precursor cells to different DC subsets and
                                   highlight the ensemble of transcription factors that control these processes.

                                  Dendritic cells (DCs) are essential for antigen presen-             Unravelling the complexity of the DC network
                                  tation and the initiation of protective T cell responses            DCs are a heterogeneous group of cells that have been
                                  and, thus, constitute a front-line defence against invad-           divided into different subsets. This segregation was
                                  ing pathogens. DCs are located throughout the body                  initially based on their distinct patterns of cell-surface
                                  and form a sophisticated and complex network that                   molecule expression. The four major categories of DCs
                                  allows them to communicate with different popula-                   are conventional DCs, which predominate in the steady
                                  tions of lymphocytes, thereby forming an interface                  state; Langerhans cells; plasmacytoid DCs (pDCs);
                                  between the external environment and the adaptive                   and monocyte-derived DCs, which are induced in
                                  immune system. To provide this protection, different                response to inflammation.
                                  subsets of DCs have evolved, and these DC subsets
                                  are specialized to exist in distinct locations, where               Conventional DCs. Conventional DCs are special-
                                  they acquire antigens and transport them to draining                ized for antigen processing and presentation. They
                                  lymph nodes for T cell priming. The DC network is                   can be grouped into two main classes based on their
                                  programmed by a group of transcription factors that                 localization in tissues and their migratory pathways
                                  determine the specification and differentiation of the              as they circulate in the body (FIG. 1a; TABLE 1). The first
                                  different subsets of DCs. Recently, it has been shown               category of conventional DCs is generally referred to
                                  that defects in transcription factor expression under-              as the migratory DCs. These DCs develop from early
                                  pin developmental defects in DCs and other immune                   precursors in the peripheral tissues, where they act
                                  cells, and these defects result in severe immuno­                   as antigen-sampling sentinels. From the peripheral
                                  deficiencies and enhanced susceptibility to bacterial,              tissues, they migrate to the regional lymph nodes via
                                  fungal and viral infections in humans1–3. Thus, disrup-             afferent lymphatics, a process that is accelerated in
                                  tion of transcription factor expression and selective loss          response to danger signals, such as those that occur
                                  of DC subsets is likely to have important implications              during pathogen infection. Migratory DCs are not
                                  for human disease.                                                  found in the spleen and are restricted to the lymph
                                      In this Review, we focus on the role of transcription           nodes4, where they constitute a variable proportion of
Division of Molecular
Immunology, Walter and            factors in generating different DC subsets and highlight            the steady-state DC population; this proportion
Eliza Hall Institute of Medical   the synergistic functions of cytokines in shaping DC                depends on the specific tissues that are drained by the
Research, 1G Royal Parade,        fate decisions. Furthermore, we discuss the molecular               lymph node5 (FIG. 1). Migratory DCs can be broadly
Melbourne, Victoria 3052,         pathways that may allow plasticity in DC fate decisions             divided into CD11b+ DCs (also known as dermal or
Australia.
e-mails: belz@wehi.edu.au;
                                  and that enable the rapid recruitment and differen-                 interstitial DCs) and CD11b– DCs6, which have more
nutt@wehi.edu.au                  tiation of DCs in response to diverse environmental                 recently been shown to express CD103 (also known as
doi:10.1038/nri3149               stimuli.                                                            integrin αE)4,7.


NATURE REVIEWS | IMMUNOLOGY	                                                                                                VOLUME 12 | FEBRUARY 2012 | 101

                                                       © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                          a                                                               Lymph nodes
                                                                Spleen
                                 Blood-derived DCs                     Lymphoid tissue-resident DCs                              Migratory DCs




                          Monocyte-           pDCs                CD4–CD8α–        CD4+ DCs       CD8α+ DCs           CD103+      CD11b+              Langerhans
                          derived DCs                             (DN) DCs                                            DCs         (interstitial or    cells
                                                                                                                                  dermal) DCs
                                      Inflammation                                                          Steady state

                          b

                                                                                        pDC       DN DC CD8α+ DC

                              Bone marrow
                                                                                       CD103+      CD11b+ Langerhans
                                                Non-lymphoid tissue                    DC          DC     cell            Skin
                                                 CD11b+ DC-SIGN+                                     Lymph node                          Langerhans cell
                                                 monocyte-derived DC
                                                                                                                                                       Epidermis
                                                             CD103+ CD11b+
                                                             DC     DC                                                                                   Dermis


                              HSC
                                                                                                                                 CD103+ DC           CD11b+ DC
                              FLT3+
                              CMP


                              MDP
                                                                   Pre-DC                      pDC
                                                    Monocyte                                                                                              Blood

                              CDP
                                                                          Pre-DC




                                                                                                           DN DC

                                                                                  CD4+ DC                  CD8α+ DC
                                          CD11b+ DC-SIGN+
                                          monocyte-derived DC                    Spleen

                         Figure 1 | Differentiation and trafficking of DC subsets. a | The figure shows the organization of the dendritic cell (DC)
                         network, and includes the key surface phenotype markers of different DC subsets, which are delineated on the basis of their
                         localization in secondary lymphoid tissues. Gut-associated DCs that express both CD103 and CD11b have been included in
                                                                                                                         Nature Reviews | Immunology
                         the CD11b+ DC subset. Inflammatory monocyte-derived DCs are rapidly recruited to sites of inflammation, whereas other
                         DC subsets are normally present in the steady state. The relationship between inflammatory and steady-state DCs remains
                         an open issue. Moreover, it is unclear whether monocyte-derived DCs can arise through in situ proliferation in addition to
                         arriving at tissues via the circulation. b | In the mouse bone marrow, haematopoietic stem cells (HSCs) differentiate into
                         common myeloid progenitors (CMPs), a fraction of which express FMS-related tyrosine kinase 3 (FLT3) and differentiate
                         into more-restricted macrophage and DC progenitors (MDPs). MDPs appear to be the direct precursor to common DC
                         progenitors (CDPs), which give rise to the DC lineages. CDPs produce precursor DCs (pre-DCs) and plasmacytoid DCs
                         (pDCs) that exit the bone marrow and travel through the blood to secondary lymphoid organs and non-haematopoietic
                         tissues. A small proportion of DCs may also be derived from CLPs in the bone marrow and from early T cell progenitors in the
                         thymus. Under steady-state conditions, lymphoid tissue-resident DCs that arise from pre-DCs are the only subsets found in
                         the spleen. This population is comprised of three conventional DC subsets, namely CD4+ DCs, CD8α+ DCs and CD8α–CD4–
                         double-negative (DN) DCs. Peripheral lymph nodes contain CD8α+ and CD8α– DC populations but are also populated by
                         two groups of migratory DCs. Langerhans cells develop in the epidermis and migrate through the basement membrane to
                         the draining lymph nodes via terminal lymphatic vessels that arise in the dermis. The dermal DC population is broadly
                         composed of CD11b+ and CD103+ DCs, and these cells migrate through the lymphatics to the lymph node. Monocytes
                         arrive at tissues from the blood. In response to inflammation, they can develop into monocyte-derived DCs, which adopt
                         many of the characteristics of conventional DCs. DC-SIGN, DC-specific ICAM3‑grabbing non-integrin.


102 | FEBRUARY 2012 | VOLUME 12	                                                                                           www.nature.com/reviews/immunol

                                                    © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Phenotypic markers of DC subsets
DC subset      DC type          CD8α CD103 CD205 EPCAM CD11b B220 or DC-SIGN Langerin Antigen                                                        Major
                                                 (CD326)     CD45RA          (CD207) presentation                                                    cytokine
                                                                                                                                                     produced

pDCs           Lymphoid-    +/–          –          –            –             –           +            ++          –            Poor                IFNα
               resident DCs
CD8α+ DCs      Lymphoid-    +            low        +            –             +           –            –           +/–          Cross-              IL‑12p70,
               resident DCs                                                                                                      presentation        IFNλ
                                                                                                                                 on MHC class I;
                                                                                                                                 expression of
                                                                                                                                 cystatin C
CD4+ DCs       Lymphoid-    –            –          –            –             +           –            –           –            Presentation on
               resident DCs                                                                                                      MHC class II
DN DCs         Lymphoid-    –            –          –            –             +           –            –           –            Presentation on
               resident DCs                                                                                                      MHC class II
CD11b+         Migratory        –        +/–        +            –             +           –            ND          –            Presentation on
DCs            DCs                                                                                                               MHC class II
CD103+ DCs Migratory                                                                                                             Cross-
•	Lung      DCs                 –        +          ++           +/–           –           –            –           +            presentation on
•	Intestine                     –        +          –            –             +           –            –           –            MHC class I
Langerhans     Migratory        –        –          ++           +             +           –            –           ++           Presentation        IL‑10
cells          DCs                                                                                                               of self antigens
                                                                                                                                 for tolerance
                                                                                                                                 induction
Monocyte-      Induced by   –            –          –            –             +           –            +           –            Cross-              TNF
derived        inflammation                                                                                                      presentation
DCs
DC, dendritic cell; DC-SIGN, DC-specific ICAM3-grabbing non-integrin; DN, double-negative; EPCAM, epithelial cell adhesion molecule; IFN, interferon;
IL, interleukin; ND, not determined; pDC, plasmacytoid DC.


                                  The second major category of conventional DCs                        that are present early in embryonic development and
                              is the lymphoid tissue-resident DCs that are found in                    that undergo a proliferative burst in the epidermis in
                              the major lymphoid organs, such as the lymph nodes,                      the first few days after birth24.
                              spleen and thymus. These DCs can be further classified
                              by their expression of the surface markers CD4 and                       pDCs. pDCs are quiescent cells that are broadly distrib-
                              CD8α into CD4+ DCs, CD8α+ DCs and CD4–CD8α–                              uted in the body. They are characterized by their ability
                              DCs (typically referred to as double-negative DCs)8,9                    to rapidly produce large amounts of type I interferons
                              (TABLE 1). CD8α + DCs are noted for their capacity to                    (IFNs)25,26, a feature most evident during viral infection.
                              cross-present antigens10 and for their major role in                     pDCs express several characteristic markers, includ-
                              priming cytotoxic CD8+ T cell responses11–16 (BOX 1).                    ing sialic acid-binding immunoglobulin-like lectin H
                              CD4 + DCs and CD4 –CD8α – DCs can also present                           (SIGLEC‑H) and bone marrow stromal antigen 2 (BST2)
                              MHC class I‑restricted antigens in some settings 15,17,                  in mice and blood DC antigen 2 (BDCA2; also known
                              but appear to be more efficient at presenting MHC                        as CLEC4C) and leukocyte immunoglobulin-like recep-
                              class  II-associated antigens to CD4 + T  cells 18–20 .                  tor, subfamily A, member 4 (LILRA4; also known as
                              Lymphoid tissue-resident DCs do not traffic from                         ILT7) in humans. In addition, both mouse and human
                              other tissues but develop from precursor DCs found                       pDCs express CD45RA27. pDCs have poor antigen-
                              in the lymphoid tissues themselves21. In the absence of                  presenting capacity, and their precise contribution to
                              infection, they exist in an immature state (which is char-               immune responses is still unclear 28.
                              acterized by a high endocytic capacity and lower MHC
                              class II expression compared with activated DCs), and                    Monocyte-derived DCs. Under inflammatory con-
                              their residency in lymphoid tissues makes them ideally                   ditions, circulating blood monocytes can be rapidly
                              placed to sense antigens or pathogens that are transported               mobilized and can differentiate into cells that possess
                              in the blood12,22,23.                                                    many prototypical features of DCs21,29–32 (FIG. 1). In the
                                                                                                       steady state, monocytes express the macrophage colony-
                              Langerhans cells. Langerhans cells are resident in the                   stimulating factor receptor (M-CSFR; also known as
                              skin and, like migratory DCs, migrate to the lymph                       CD115), which is essential for their development, as well
                              nodes to present antigens (FIG. 1). However, unlike con-                 as other markers, such as LY6C and CX3C-chemokine
                              ventional DCs, which arise from a bone marrow precur-                    receptor 1 (CX3CR1). In response to growth factors such as
                              sor cell, Langerhans cells are derived from a local LY6C+                granulocyte–macrophage colony-stimulating factor
                              myelomonocytic precursor cell population in the skin.                    (GM-CSF) in vitro or to Toll-like receptor 4 (TLR4) ligands
                              This precursor population originates from macrophages                    or bacteria in vivo, fully differentiated monocyte-derived


NATURE REVIEWS | IMMUNOLOGY	                                                                                                 VOLUME 12 | FEBRUARY 2012 | 103

                                                        © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

 Box 1 | Direct presentation, cross-presentation and cross-dressing
                                                                                                     Cytokines regulate DC development
                                                                                                     The differentiation of DCs from haematopoietic
 Efficient presentation of antigens to CD8+ T cells depends on the generation of                     progenitor cells relies on the activity of cytokines,
 peptides for loading into MHC class I complexes. Several pathways have been                         in particular FMS-related tyrosine kinase 3 ligand
 uncovered to achieve this activation of CD8+ T cells. Direct antigen presentation by                (FLT3L), M‑CSF and GM‑CSF. These cytokines con-
 infected or malignant cells ensures the destruction of these cells by cytotoxic CD8+
                                                                                                     trol the initial production and lineage diversification
 T cells (CTLs), without harming adjacent healthy cells. The restriction of antigen
 presentation to directly infected cells, however, is not sufficient to ensure the activation
                                                                                                     of DCs, although the factors that regulate the expres-
 of CD8+ T cells, particularly when the pathogen does not infect professional                        sion of the receptors for these key cytokines and the
 antigen-presenting cells, such as dendritic cells (DCs). In this case, CD8+ T cells can be          downstream transcriptional programmes instigated by
 activated by DCs that present extracellular antigens on their MHC class I molecules via             FLT3L, M‑CSF and GM‑CSF are only now emerging.
 the process of cross-presentation. This pathway can result in the generation of CTLs                The ability of these cytokines to stimulate the differ-
 that are reactive to foreign antigens, or in the induction of tolerance through the                 entiation of DCs in vitro (BOX 2) provides a tractable
 deletion of autoreactive CD8+ T cells following the cross-presentation of self antigens.            model system to address the influence of extrinsic
 In mice, most focus has been on the cross-presenting capacities of CD8α+ DCs, and                   factors on the DC transcriptional network.
 more recently CD103+ DCs6,7,10,138, although other populations also possess this ability.               FLT3L and FLT3 constitute the best-characterized
 For example, fully differentiated monocyte-derived DCs that express DC-specific
                                                                                                     growth factor–receptor axis for DCs, as mouse haemato­
 ICAM3‑grabbing non-integrin (DC‑SIGN) are potent cross-presenting cells33.
   An alternative pathway for the acquisition of peptide–MHC complexes is known as
                                                                                                     poietic progenitor cells cultured with FLT3L generate a
 ‘cross-dressing’139,140 (originally known as trogocytosis141). Through this pathway, DCs            diverse array of conventional DC subsets and pDCs36,37.
 acquire preformed peptide–MHC class I complexes from infected cells. Complexes                      In agreement with the important role of FLT3L in DC
 acquired in this way can drive the activation of memory, but not naive, CD8+ T cells                differentiation, DCs can be generated from essentially
 during viral infection, perhaps owing to the lower activation threshold of memory cells.            any FLT3+ progenitor cell either in vitro or following
 The molecular mechanisms involved in cross-presentation and cross-dressing are only                 adoptive transfer in vivo38–40 (FIG. 2). In addition, enforced
 beginning to be unravelled.                                                                         expression of FLT3 in megakaryocyte–erythrocyte
                                                                                                     progenitors (MEPs), which are normally FLT3–, results
                                                                                                     in the acquisition of DC potential41. An instructive role
                               DCs emerge. Similarly to conventional DCs, monocyte-                  for FLT3 in DC development is further supported by the
                               derived DCs express CD11c, MHC class II molecules,                    finding that in all cell lineages except the DC lineage,
                               CD24 and SIRPα (also known as CD172a), and they upreg-                FLT3 is downregulated following differentiation and,
                               ulate their expression of DC-specific ICAM3‑grabbing                  at least in the case of B cells, this repression of FLT3 is
                               non-integrin (DC‑SIGN; also known as CD209a) but                      essential for further development 42.
                               lose expression of both M‑CSFR and LY6C33 (TABLE 1).                      Mice deficient for FLT3L or signal transducer and
                               Monocyte-derived DCs also express the macrophage                      activator of transcription 3 (STAT3; a signalling mol-
                               marker MAC3 (also known as CD107b and LAMP2)21,32.                    ecule downstream of FLT3) have markedly reduced
                               In addition, these cells acquire potent antigen-presenting            numbers of lymphoid-resident conventional DCs and
                               capacity, including the ability to cross-present antigens33–35        pDCs, whereas mice lacking FLT3 have a milder pheno-
                               (BOX 1). Thus, it is emerging that monocyte-derived DCs               type, which suggests the presence of a second ligand for
                               are a crucial reservoir of professional antigen-presenting            this receptor 43–46. FLT3 signalling is also crucial for the
                               cells (APCs) that are recruited into immune responses                 development of migratory DCs, as the numbers of both
                               to certain microorganisms and potentially have an                     pre-DCs and CD103+ DCs were found to be reduced in
                               emergency back-up role in cases of acute inflammation.                a range of tissues from Flt3l–/– mice compared with the


                                Box 2 | In vitro models for investigating DC development and behaviour
                                The establishment of well-defined cell culture systems that allow the generation of large numbers of dendritic cells (DCs)
                                from bone marrow has been instrumental for understanding DC biology. Recent refinement of the tools and surface
                                markers used to analyse cultures now allows the resolution of DC precursors equivalent to those found in vivo, together
                                with fully differentiated DC subsets, in the tissue culture flask.
                                Generation of steady-state DC subsets in vitro
                                The development of steady-state DCs depends on signalling through FMS-related tyrosine kinase 3 (FLT3), which is
                                expressed on the surface of DC precursors. Bone marrow precursors cultured with FLT3 ligand (FLT3L) give rise to
                                plasmacytoid DCs (pDCs) and multiple lymphoid tissue-resident conventional DC subsets36,37,89. Intriguingly,
                                in vitro-generated DCs do not express CD4 or CD8, but their patterns of expression of the markers CD103, CD11b,
                                CD172a and CD24 indicate the presence of conventional DC subsets in addition to DC precursors.
                                Generation of monocyte-derived DCs in vitro
                                Perhaps the most commonly used approach to generate DCs involves the culture of bone marrow precursors in a medium
                                supplemented with granulocyte–macrophage colony-stimulating factor (GM-CSF). As these DCs can also be produced by
                                culturing monocytes in GM‑CSF and interleukin‑4 (IL‑4), they are referred to as monocyte-derived DCs and correspond
                                to the dominant inflammatory DC type that is mobilized during some bacterial infections33.
                                  These highly refined culture systems allow comparative studies between different DC subsets. Furthermore, the 	
                                ability to generate large numbers of DCs in vitro should greatly facilitate molecular exploration of the genomic and
                                transcriptional machinery that leads to the generation of different DC subsets.



104 | FEBRUARY 2012 | VOLUME 12	                                                                                              www.nature.com/reviews/immunol

                                                      © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                                                                                      BATF3
                                                                                                           FLT3L


                                                                          Transitional                                        CD103+ DC
                         Lymphocytes                                      pre-DC
                                                                                         ID2                E4BP4 BATF3

              CLP
                                                                                                                   FLT3L
                                                               IRF8                            Pre-CD8α+ DC                                            PU.1hi
                                                                                           IRF2                                CD8α+ DC                ID2hi
                      PU.1          GFI1            PU.1                       RELB        IRF4    Ikaros (null)
         ?    FLT3+
  LMPP                       MDP              CDP           Pre-DC
              CMP                                                                                             GM-CSF
                                                                                                              M-CSF
                                                    FLT3L                                                                     CD11b+ DC


                                   Monocyte

                                                                         Immature pDC                                                        Loss
                M-CSF                  GM-CSF                                                                                                of E2-2

                                                                                          E2-2       Ikaros (L)      IRF8
                                                                                                                                                       PU.1low
                                                                                                                                                       ID2–
                                                                                                                                                       E2-2hi
                 Macrophage      Monocyte-                                                                                    Mature pDC
                                 derived DC
                         Figure 2 | Growth factors and transcription factors that regulate DC differentiation. The developmental pathways
                         from myeloid and lymphoid progenitors to precursor dendritic cells (pre-DCs) in the bone marrow and Reviews | Immunology
                                                                                                                          Nature the peripheral
                         diversification of DC subsets are shown (see FIG. 1 legend for details). The approximate points at which key transcription
                         factors are first required for DC development are indicated by vertical lines. Stages at which key growth factors have been
                         determined to be essential are indicated. The development of both DCs and monocytes depends on high concentrations
                         of PU.1, which regulates the expression of the cytokine receptors FMS-related tyrosine kinase 3 (FLT3), macrophage
                         colony-stimulating factor receptor (M-CSFR) and granulocyte–macrophage colony-stimulating factor receptor (GM-CSFR).
                         The development of CD8α+ and CD103+ DCs relies on the stepwise activity of interferon-regulatory factor 8 (IRF8), inhibitor
                         of DNA binding 2 (ID2), E4 promoter-binding protein 4 (E4BP4) and basic leucine zipper transcription factor, ATF-like 3
                         (BATF3), as well as on FLT3 signalling. CD11b+ DCs depend on a unique set of transcription factors, including RELB, IRF2, IRF4
                         and Ikaros, and to some extent on the cytokines M‑CSF and GM‑CSF. The plasmacytoid DC (pDC) lineage requires IRF8, a low
                         level of PU.1 and the absence of ID2. The differentiation of pDCs from an immature precursor requires E2‑2 and Ikaros, with
                         induced loss of E2‑2 converting pDCs into cells that closely resemble CD8α+ conventional DCs. CDP, common DC progenitor;
                         CLP, common lymphoid progenitor; CMP, common myeloid progenitor; FLT3L, FLT3 ligand; GFI1, growth factor
                         independent 1; LMPP, lymphoid-primed multipotent progenitor; MDP, macrophage and DC progenitor.



                         numbers in wild-type mice. By contrast, the development                 that resemble monocyte-derived DCs, while repress-
                         of CD11b+ DCs and Langerhans cells is largely independ-                 ing the development of pDCs in a STAT5‑dependent
                         ent of FLT3L47. Thus, FLT3L has two distinct roles in DC                manner 46,51. This finding has generally been interpreted
                         biology: it is required for the early development of DCs                to show that GM‑CSF has a greater role in the produc-
                         from haematopoietic progenitors; and later it functions                 tion of monocyte-derived DCs than in the generation
                         to maintain DC homeostasis by promoting limited levels                  of other DC subsets21,52, although the importance of this
                         of proliferation of DCs in peripheral tissues43.                        process in vivo is still to be established.
                             In addition to FLT3L, GM‑CSF has long been known                        M-CSF is the major cytokine involved in the produc-
                         to stimulate DC differentiation in culture (BOX  2) .                   tion of monocytes and macrophages53. A role for this
                         However, GM‑CSF is not essential for DC differentia-                    cytokine in DC biology was suggested by the expression
                         tion in the steady state, as mice that lack the GM‑CSF                  of M‑CSFR by DCs54,55 but came to prominence with the
                         receptor (GM-CSFR) have only mildly reduced num-                        finding that M‑CSFR-deficient mice lack Langerhans
                         bers of DCs48. Nonetheless, GM‑CSF is not completely                    cells56. Surprisingly, mice lacking M‑CSF (op/op mice)
                         redundant in DC production, as mice deficient in both                   have normal numbers of Langerhans cells, a quandary
                         GM‑CSF and FLT3L have a greater loss of DCs than either                 that was resolved by the identification of interleukin‑34
                         single-knockout strain44. Moreover, other recent studies                (IL‑34) as a second ligand for M‑CSFR57. The relatively
                         have demonstrated that GM‑CSF is necessary for there                    normal DC numbers in mice lacking M‑CSF, despite
                         to be normal numbers of CD103+CD11b+ DCs in the                         the profound reduction in monocytes, demonstrates
                         lamina propria49,50. The addition of GM‑CSF to cultures                 that the monocytic system is not the major source of
                         of bone marrow cells promotes the development of cells                  steady-state DCs. M‑CSF is, however, required for the


NATURE REVIEWS | IMMUNOLOGY	                                                                                                VOLUME 12 | FEBRUARY 2012 | 105

                                                © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                         normal development of CD103–CD11b+ DCs in non-                      that specifies the DC lineage in more-immature
                         lymphoid tissues49 and is able to support conventional              pro­ enitors and then drives differentiation into the DC
                                                                                                 g
                         DC and pDC differentiation in cell culture in the                   subsets. Three transcription factors — PU.1 (encoded by
                         absence of FLT3 (REF. 58).                                          Sfpi1), Ikaros and growth factor independent 1 (GFI1)
                                                                                             — appear to be prime candidates for DC-specifying fac-
                         Stages of programming DC identity                                   tors. In addition, the signalling and transcription factors
                         DC ontogeny. Although it is well established that all               STAT3 and STAT5 are known to have a role in DC differ-
                         DCs, with the exception of Langerhans cells, are derived            entiation, as they mediate the signals transduced through
                         from bone marrow-resident haematopoietic stem cells                 FLT3 and GM‑CSFR, respectively (TABLE 2).
                         (HSCs), mapping the origins of the DC lineages has                      PU.1 belongs to the ETS family of transcription
                         proven to be both difficult and controversial. Early                factors, which has multiple context-specific roles in
                         transfer experiments led to the surprising conclusion               haemato­poiesis. PU.1 is an attractive candidate for being
                         that DCs can develop with approximately equal effi-                 a crucial regulator of the DC lineages, as it is expressed by
                         ciency from both lymphoid and myeloid pro­genitors59,60,            all DCs and by CDPs66–68. A role for PU.1 in DC develop-
                         whereas in vitro cultures with GM‑CSF show that DCs                 ment was initially suggested by the analysis of mice with
                         can arise from monocytic precursors, as mentioned                   a germline deficiency of PU.1. Indeed, one such study
                         above. However, monocytes are not likely to be a major              concluded that PU.1 was necessary for all embryonic DC
                         source of steady-state DCs in lymphoid organs, as                   development 69, although a second study reported that
                         lineage-tracing experiments have shown that monocytic               Sfpi1–/– fetal thymi could generate DCs70. However, these
                         cells give rise to neutrophils and macrophages but not              approaches could not distinguish between the require-
                         DCs5. More recently, the adoption of a ‘FLT3‑centric’               ments for PU.1 in multipotent progenitors and the role
                         view of haematopoiesis38–40,61 has established that most            of PU.1 specifically in the DC lineages. Moreover, the
                         steady-state DCs arise from FLT3+ progenitors (FIG. 2).             impact of enforced expression of PU.1 in haematopoietic
                             The pathway of DC differentiation from primitive                progenitors suggests an instructive and concentration-
                         bone marrow progenitors has been extensively reviewed               dependent role for PU.1 in promoting macrophage and
                         elsewhere4,53,62 and is only briefly summarized here. FLT3          DC development71–73.
                         expression is first induced in a subset of the HSC com-                 A recent study used conditional gene deletion in
                         partment that has only short-term pan-haematopoietic                defined haematopoietic progenitors and CDPs to show
                         repopulating activity, and the expression of this recep-            that PU.1 is absolutely essential for the generation of
                         tor is then maintained in lymphoid-primed multipotent               all conventional DCs and pDCs both in vivo and in
                         progenitors (LMPPs)63 and in a subpopulation of com-                FLT3L‑containing cultures in vitro 66. Moreover, in
                         mon myeloid progenitors (CMPs)38. CMPs are thought                  line with its established role in regulating GM‑CSFR
                         to differentiate into macrophage and DC progenitors                 expression, PU.1 is required for GM‑CSF-induced DC
                         (MDPs)64, which appear to be the direct precursors of               differentiation from early haematopoietic progenitors.
                         common DC progenitors (CDPs)40,61. Both MDPs and                    Among the many genes that are potentially regulated
                         CDPs are proliferating cells that reside in the bone mar-           by PU.1, Flt3 was demonstrated by molecular studies
                         row and express FLT3 and M‑CSFR. CDPs differentiate                 to be directly regulated by PU.1 in DCs and haemato­
                         directly into pDCs and into the precursors of conven-               poietic progenitors66. This regulation occurred in a
                         tional DC subsets, termed pre-DCs, but they lack the                concentration-dependent manner, as Sfpi1+/– cells had
                         potential to give rise to macrophages21,65. Pre-DCs then            reduced FLT3 expression and an impaired ability to
                         leave the bone marrow and are found in blood, second-               generate conventional DCs. Interestingly, previous stud-
                         ary lymphoid organs and some tissues21,49,52,65, where              ies have shown that FLT3 signalling is able to activate
                         they mature into the conventional DC subsets (FIG. 1).              PU.1 expression in MEPs, suggesting a self-reinforcing
                         Differentiation into different conventional DC subsets              loop between PU.1 and FLT3 in DCs41. Whether PU.1
                         appears to be a late step in DC development that is per-            is required for monocyte-derived DC formation in vivo
                         haps important in maintaining the stability or plasticity           remains to be determined.
                         of the peripheral DC compartment. The key features and                  Ikaros is a zinc-finger transcription factor that has
                         mechanisms involved in the plasticity of the DC network             important roles in haematopoiesis74. Expression of a
                         are likely to include the short lifespan of mature conven-          dominant-negative form of Ikaros that also impairs the
                         tional DCs (5–7 days, although up to 25 days in some cir-           function of other Ikaros family members, such as Aiolos,
                         cumstances4,47); the rapid recruitment and proliferation of         resulted in a complete loss of all conventional DC sub-
                         pre-DCs and their capacity to respond to extrinsic signals          sets. By contrast, a null mutation in the gene encoding
                         (such as TLR ligands and pro-inflammatory cytokines);               Ikaros led to the selective loss of CD11b+ DCs, with some
                         and the active expression by DCs of transcription factors           CD8α+ DCs being retained75. Whether Ikaros directly
                         such as E2‑2 (also known as TCF4).                                  regulates DC differentiation, as opposed to having a role
                                                                                             in early myeloid progenitors, is at present unclear 76, as
                         Initiating the DC programme in haematopoietic progeni-              mice homozygous for a severely hypomorphic allele of
                         tors. The information outlined above demonstrates the               the Ikaros gene (IkL/L mice) lack mature pDCs but con-
                         rapid progress that is being made in understanding the              tain relatively normal numbers of conventional DCs77.
                         developmental stages and cell biology of the DC lineages.           DC‑specific conditional mutagenesis is now required to
                         Much less is known about the transcriptional programme              decipher the exact function of Ikaros in DCs (FIG. 2).


106 | FEBRUARY 2012 | VOLUME 12	                                                                                     www.nature.com/reviews/immunol

                                              © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                    Table 2 | Transcription factors guiding steady-state DC subset development
                                    Transcription Transcription factor family                        Function                                                        Refs
                                    factor
                                    PU.1 (SFPI1,       ETS-domain transcription factor;              Required for the development of all DC subsets                   66,
                                    SPI1)              binds to PU box sequences                                                                                    69,70
                                    IRF2               Interferon-regulatory factor; inhibits        Alters pDC ratios; in its absence the numbers of                 122
                                                       the IRF1-mediated transcription of            CD8α– DCs and Langerhans cells are reduced
                                                       type I IFNs
                                    IRF4               Interferon-regulatory factor                  Required for non-CD8α+ DC lineage development               126,127
                                    IRF8 (ICSBP)       Interferon-regulatory factor                  Required for the development of pDCs and most                  90,92,
                                                                                                     conventional DCs                                              93,104
                                    GFI1               Zinc-finger protein; transcriptional          GFI1 deficiency results in a 50% reduction in the                 79
                                                       repressor                                     numbers of conventional DCs and pDCs and
                                                                                                     increased numbers of Langerhans cells
                                    ID2                Inhibitor of DNA binding family               Required for the development of CD103+ DCs and                   47,
                                                       protein containing HLH domains                CD8α+ DCs in PLNs and spleen; not required for                 87,89
                                                                                                     DCs in MLNs
                                    E4BP4 (NFIL3)      PAR-related bZIP transcription factor Required for the development of CD8α+ DCs                                108
                                    E2‑2 (TCF4)        E protein containing bHLH domains             Required for the development and maintenance                   80,84
                                                                                                     of pDCs
                                    STAT3              Signal transducer and activator of            STAT3 deficiency results in a substantial reduction               46
                                                       transcription                                 in conventional DC numbers
                                    STAT5A and         Signal transducer and activator of            Inhibit pDC development by interacting with IRF8;                 51
                                    STAT5B             transcription                                 deficiency results in reduced conventional DC and
                                                                                                     increased pDC numbers
                                    Ikaros (IKZF1)     Zinc-finger DNA-binding protein               Ikaros deficiency results in the absence of most               75,77
                                                                                                     DCs; a hypomorphic mutation leads a specific loss
                                                                                                     of pDCs
                                    BATF3              bZIP family; heterodimerizes with             BATF3-deficient mice fail to develop CD103+ DCs                   13,
                                                       JUN                                           and show impaired survival of precursor CD8α+ DCs             89,111
                                    RELB               REL-homology domain family;                   RELB deficiency results in the loss of CD8α– DCs            129,130
                                                       interacts with NF‑κB family members
                                    SPIB               ETS-domain transcription factor               Required for human pDC differentiation                            91
                                    BATF3, basic leucine zipper transcription factor, ATF-like 3; bHLH, basic HLH; bZIP, basic leucine zipper; DC, dendritic cell; E4BP4,
                                    E4 promoter-binding protein 4; GFI1, growth factor independent 1; HLH, helix-loop-helix; ID2, inhibitor of DNA binding 2; IFN,
                                    interferon; IRF, interferon-regulatory factor; MLN, mesenteric lymph node; NF‑κB, nuclear factor‑κB; PAR, proline- and acidic-rich
                                    region; pDC, plasmacytoid DC; PLN, peripheral lymph node; STAT, signal transducer and activator of transcription.



                                       GFI1 is a small, zinc-finger-containing transcriptional            it is surprising how little we actually understand about
                                   repressor that is important for early haemato­ oiesis78.
                                                                                     p                    this process. Two developmental systems appear to be
                                   GFI1 is expressed in DC precursors, and Gfi1–/– mice                   in place to separate pDCs and conventional DCs (FIG. 2).
                                   have reduced numbers of all lymphoid-resident DC                       First, pDCs absolutely rely on the expression of the
                                   subsets, whereas Langerhans cell numbers were actu-                    E protein E2‑2 (REF. 80) and the absence of the E protein
                                   ally increased79. Interestingly, GFI1‑deficient haemato­               antagonist inhibitor of DNA binding 2 (ID2)81. Second,
                                   poietic progenitor cells were unable to develop into DCs               pDCs have a uniquely low level of PU.1 (REF. 68) and an
                                   in vitro in the presence of either FLT3L or GM‑CSF and                 extremely high concentration of interferon-regulatory
                                   instead differentiated into macrophages, suggesting that               factor 8 (IRF8), a transcription factor that can form
                                   GFI1 is a crucial modulator of DC versus macrophage                    a complex with PU.1 on a class of ‘composite’ DNA
                                   development (FIG. 2).                                                  elements82.
                                                                                                              One model to explain the pDC versus conventional
                                   Establishing pDC and conventional DC identity.                         DC lineage split is to assume that the conventional DC
E protein
                                   Restriction of the developmental programme of DC                       is the default setting and that progenitors have to be
The E proteins (including E12,
E47, HEB and E2‑2) have            progenitors to the conventional DC and pDC lineages                    diverted to the pDC lineage83. E2‑2 fits the bill for a fac-
emerged as key regulators of       occurs at the CDP stage40,61 (FIG. 2). Conventional DCs                tor that could control this diversion, as it is abundantly
the immune system. They are a      and pDCs differ markedly in their appearance, functions                expressed by pDCs and is required for pDC lineage
family of basic helix-loop-helix   and transcriptional programmes, and thus how a CDP                     specification80. Once progenitors have committed to a
factors that work together with
their antagonists, the ID
                                   is influenced to develop into either a pDC or a conven-                pDC fate, it appears that continuous expression of E2‑2
proteins (ID1–ID4), to regulate    tional DC is a question of major importance for under-                 is essential to maintain the mature pDC phenotype84.
lymphocyte development.            standing and manipulating DC biology. In this context,                 In mice, a specific deletion of the gene encoding E2‑2


NATURE REVIEWS | IMMUNOLOGY	                                                                                                       VOLUME 12 | FEBRUARY 2012 | 107

                                                           © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                   in pDCs led to the expansion of a population of DCs                 they have been shown to share a few targets, such as
                                   that exhibit many characteristics of conventional DCs.              Ciita94, Tlr9 (REF. 95) and Ifna96). However, BXH2 mice,
                                   In the in vivo setting, however, it is particularly dif-            which harbour a spontaneous point mutation in Irf8,
                                   ficult to dissect whether E2‑2‑deficient pDCs undergo               have defects in CD8α + DC development but not in
                                   phenotypic conversion to conventional DCs owing to a                pDC generation. This mutation is thought to ablate
                                   reduction in E2‑2‑mediated repression of ID2, as pro-               the interaction of PU.1 and IRF8, raising the possibil-
                                   posed, or whether normal conventional DC numbers                    ity that the PU.1–IRF8 complex is not as crucial for
                                   are increased in the absence of a full pDC compart-                 the differentiation of pDCs as for the development
                                   ment, as occurs in other settings. Nevertheless, pDCs               of conventional DCs93. There are, as yet, no genome-
                                   are particularly sensitive to E2‑2 concentration, as both           wide DNA-binding datasets for IRF8 and PU.1 in
                                   E2‑2‑deficient mice and patients with a rare mono-                  DCs, although similar data from macrophages sug-
                                   allelic loss of E2‑2 (Pitt–Hopkins syndrome) show                   gest that PU.1 might occupy most of the active regula-
                                   impaired pDC formation and function80. E2‑2 binds                   tory regions in DCs97,98 and that IRF8 might bind to
                                   directly to the promoters of several pDC-expressed                  a subset of these sites99. Importantly, in macrophages
                                   genes, including BDCA2, LILRA4, IRF7, the pre-TCR                   and myeloid progenitors, PU.1 is directly involved in
                                   α-chain gene, IRF8 and SPIB (which encodes a close                  nucleosome remodelling, and this leads to the generation
                                   relative of PU.1 that is expressed by human and mouse               of an open chromatin conformation and histone modi‑
                                   pDCs)80. The reliance of pDCs on an E protein such as               fications, suggesting that PU.1 can directly programme
                                   E2‑2 may explain the observation that pDCs express                  the fate of myeloid cells97,98.
                                   many lymphocyte-associated transcripts (including                       The ETS-family transcription factor SPIB — the
                                   SPIB, RAG1, IL7R and TDT), as E proteins are central                closest homologue of PU.1 in the mammalian genome
                                   to many aspects of lymphopoiesis27.                                 — is expressed, within the DC lineages, specifically by
                                       ID proteins are direct inhibitors of DNA bind-                  pDCs100. Knockdown of the expression of either SPIB
                                   ing by E proteins. ID2 is the predominant ID protein                or PU.1 in human haematopoietic progenitors strongly
                                   expressed in the DC lineage and is also involved in                 inhibits pDC formation, suggesting that both factors
                                   the development of multiple lineages during haemato­                function in human pDCs91. The extent of any functional
                                   poiesis, particularly that of lymphoid tissue-inducer               redundancy between PU.1 and SPIB in pDCs has so far
                                   cells (LTi cells) and natural killer (NK) cells85–88. ID2           not been addressed in mice.
                                   expression is extremely low in CDPs, pre-DCs and                        In summary, the separation of pDC and conven-
                                   pDCs, whereas all conventional DC populations                       tional DC lineages represents the first major division
Lymphoid tissue-inducer            express high levels of ID2 (REF. 89) (FIG. 3). In line with         in the DC pathway and requires the concerted action of
cells
(LTi cells). A cell type that is
                                   this expression pattern, ID2‑deficient mice have a                  both E proteins and the PU.1–IRF8 complex (FIGS 2,3).
present in developing lymph        profoundly altered conventional DC compartment                      Although the activation of STAT5 inhibits pDC for-
nodes, Peyer’s patches and         but still produce pDCs (see below)87. This leads to a               mation in vitro 51, the exact signals that initiate this
nasopharynx-associated             model whereby the acquisition of high levels of ID2                 process are not known. In addition, the ways in which
lymphoid tissue (NALT).
                                   and subsequent suppression of E2‑2 activity blocks                  the E2‑2–ID2 and PU.1–IRF8 axes interact are still
LTi cells are required for
the development of these           pDC development and allows progression along the                    unclear, as are the identities of most of the genes tar-
lymphoid organs. The inductive     conventional DC pathway. This model is clearly an                   geted by these transcription factors. pDCs represent the
capacity of these cells for the    oversimplification, as E2‑2‑deficient mice still produce            end point of their lineage; however, the production of
generation of Peyer’s patches      some pDC progenitors and ID2 is not essential for the               conventional DCs is only the first step in their further
and NALT has been shown by
adoptive transfer, and it is
                                   differentiation of all conventional DCs.                            diversification, which is outlined in the next section.
generally assumed that they            The development of pDCs also depends on PU.1,
have a similar function in the     IRF8 and potentially SPIB 66,90,91. pDCs are absent                 Genetic programming of conventional DC subsets
formation of lymph nodes.          in PU.1- or IRF8‑deficient mice 66,90,92, as well as in             Although it is fairly clear that conventional DCs, pDCs,
                                   humans with a mutation in IRF8 (REF. 3), although it                Langerhans cells and monocyte-derived DCs represent
Nucleosome remodelling
Changes in the nucleosome          remains to be proven whether these factors function                 developmentally distinct lineages, the relationships
structure are mediated by          specifically in the pDC lineage or have a role in CDPs              between the various anatomically, phenotypically and
dedicated nuclear enzymes          (FIG. 2) . However, circumstantial evidence favours a               functionally distinct conventional DC populations
(for example, ATP-dependent        specific function for these factors in determining pDC              remain to be fully elucidated. Conventional DCs are
nucleosome-remodelling
enzymes) that change the
                                   versus conventional DC fate. PU.1 is expressed at a                 thought to be derived from circulating pre-DCs21,65.
accessibility of DNA and           uniformly high level in CDPs, but at a lower level in               Although it remains possible that multiple develop-
the expression of genes.           pDCs, with the timing of downregulation coinciding                  mentally distinct types of pre-DC exist and act as the
                                   with pDC formation66, whereas IRF8 is expressed at                  precursors for individual conventional DC lineages,
Histone modifications
                                   very high levels in both CDPs and pDCs (FIG. 3). The                we favour a model that considers all the populations
Histones are essential to
maintain DNA organization          stoichiometric relationship between PU.1 and IRF8 is                of conventional DCs as related subsets derived from a
and may be modified by             likely to be important for the pDC versus conventional              single pre-DC population. In this model, the conven-
methylation and acetylation —      DC branch point, as both of these factors are known to              tional DC subsets are induced by the environmental
changes that are thought to        function in a dose-dependent manner 66,93 and can bind              milieu that they reside in, and thus it is the interaction
keep genes active or silent,
respectively — thereby altering
                                   to distinct DNA sequences both individually and in a                of these extrinsic signals with the core transcriptional
the genetic code read by           complex 82. The extent to which PU.1 and IRF8 share                 programme of conventional DCs that dictates the
transcriptional regulators.        target genes in pDCs is currently unknown (although                 outcome of DC terminal differentiation.


108 | FEBRUARY 2012 | VOLUME 12	                                                                                              www.nature.com/reviews/immunol

                                                        © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                                                      BATF3 +++       clearly has a key role in the function of conventional DCs,
                                                                                      IRF8 +++        Irf8–/– mice develop a myeloproliferative syndrome that is
                                                                                      ID2 +++         characterized by the overproduction of granulocytes105.
                                                                                      IRF4 ++
                                                                                      E4BP4 ++        This implies that IRF8 may also be required for the gen-
                                                                     CD103+ DC
                                                                                      PU.1 +++        eration or maintenance of MDPs, which can give rise
                                                                                      E2-2 +          to monocytes, conventional DCs and pDCs. The exact
                                  Transitional                                                        developmental stage at which IRF8 exerts its activity on
                                  pre-DC         Pre-CD8α+ DC                         BATF3 ++        conventional DCs remains to be fully determined.
                                                                                      IRF8 +++
                                                                                      ID2 +++             E4BP4 is a mammalian basic leucine zipper (bZIP)
                                                                                      IRF4 +          transcription factor that is required for the development
                                                                                      E4BP4 ++        of NK cells but not of other lymphocyte lineages106,107.
                                    IRF8 +++       IRF8 +++                           PU.1 +++
                                    ID2 +/–        ID2 ++             CD8α+ DC        E2-2 +          In NK cells, E4BP4 acts in a dose-dependent man-
                                    PU.1 +++       PU.1 +++                                           ner downstream of the IL‑15 receptor to regulate ID2
   CDP              Pre-DC                                                            BATF3 ++        expression106,107. More recently, the induction of E4BP4
                                                                                      IRF8 +          has been shown to be important for the development of
                                                                                      ID2 ++          CD8α+ DCs108. E4BP4‑deficient CDPs had lower levels
                                                                                      IRF4 +++
                                                                                      E4BP4 ++        of BATF3 expression than control CDPs, and enforced
IRF8 +++           IRF8 +++
                                                                        CD11b+
                                                                                      PU.1 +++        expression of BATF3 in the mutant cells rescued CD8α+
ID2 +/–            ID2 +/–                                                            E2-2 +          DC development in vitro108. Thus, E4BP4 is emerging as
PU.1 +++           PU.1 +++                                             DC
                                                                                                      a key regulator of conventional DC diversity. Whether
                                  Immature pDC                                        BATF3 +
                                                                                      IRF8 +++        E4BP4 also acts through ID2 to mediate these effects
                                                                                      ID2 +/–         in DCs has not yet been addressed.
                                                                                      IRF4 ++             ID2 is expressed by all conventional DC subsets,
                                                                                      E4BP4 ++
                                    IRF8 +++                            Mature        PU.1 +          with the highest levels of expression in CD8α + and
                                    ID2 +/–                             pDC           E2-2 +++        CD103+CD11b– DCs47,89. Loss of ID2 prevents the devel-
                                    PU.1 +                                                            opment of these two subsets in the skin-draining lymph
                                    E2-2 +
                                                                                                      nodes and spleen. However, CD103+CD11b+ DCs in the
Figure 3 | Differential expression of transcription factors regulating DC                             mesenteric lymph nodes, together with CD4+ and CD4–
differentiation. The stages of differentiation of conventional dendritic cells Immunology
                                                               Nature Reviews | (DCs) and             CD8α– DCs in lymphoid tissues, appear to develop nor-
plasmacytoid DCs (pDCs) from the common DC progenitor (CDP) are shown, together                       mally in the absence of ID2 (REFS 13,47,87). Although it
with the relative levels of expression of key transcription factors in each cell type indicated
                                                                                                      is at present unclear which E protein (or E proteins) —
on an arbitrary scale. The expression pattern of some of the factors has not been
determined at the earliest stages of DC ontogeny. –, no expression; +, low expression;                E2A (also known as TCF3), E2‑2 or HEB (also known as
++, intermediate expression; +++, maximal expression; BATF3, basic leucine zipper                     TCF12) — is the crucial target of ID2 in conventional DCs,
transcription factor, ATF-like 3; E4BP4, E4 promoter-binding protein 4; ID2, inhibitor                selective deletion of the gene encoding E2‑2 in mature
of DNA binding 2; IRF, interferon-regulatory factor; pre-DC, precursor DC. Figure is                  pDCs results in the spontaneous differentiation of pDCs
modified, with permission, from REF. 89 © Macmillan Publishers Ltd. All rights reserved.              into cells that exhibit conventional DC properties, perhaps
                                                                                                      through the induction of ID2 (REF. 84). Progress in dis-
                                                                                                      cerning the key target genes of E proteins in conventional
                                  Transcriptional regulators of CD8α + and CD103 +                    DCs will be required to understand why ID2 has such
                                  DCs. CD8α+ and CD103+ DCs have gained consider-                     an important role in CD8α+ and CD103+CD11b– DCs.
NFAT
(Nuclear factor of activated
                                  able attention owing to their specialized roles as induc-               BATF3, which is also known as JUN-dimerization
T cells). A family of             ers of MHC class I‑restricted immune responses to                   protein p21SNFT, is a bZIP transcription factor that
transcription factors that are    pathogens. Moreover, a human BDCA3+ DC subset has                   acts to repress the activity of NFAT –AP1 complexes
regulated by calcium signalling   recently been identified that shares features with both             by competing with FOS for JUN dimerization 109,110.
and expressed by a variety of
                                  of these mouse DC subsets; such features include cross-             BATF3 was the first transcription factor that appeared
immune cells.
                                  presenting capacity and the expression of XC-chemokine              to have an exclusive role in the development of the
AP1                               receptor 1 (XCR1)101–103. The differentiation of DC pre-            CD8α+ DCs111, although it has since been shown to
(Activator protein 1).            cursors into the CD8α+ and CD103+ DC lineages appears               be involved in the development of CD103+CD11b–
A heterodimeric transcription     to depend on the integration of four key transcription              DCs in peripheral lymphoid tissues13, but not that of
factor that is composed of
proteins belonging to the FOS,
                                  factors — namely, IRF8, E4BP4 (E4 promoter-binding                  CD103+CD11b+ DCs isolated from gut lymphoid tis-
JUN and JUN-dimerization          protein 4; also known as NFIL3), ID2 and BATF3 (basic               sues112. More recently, detailed analyses of Batf3–/– mice
protein families. AP1 controls    leucine zipper transcription factor, ATF-like 3).                   have shown that despite the reduction in the frequency
various cellular processes,           IRF8 is highly expressed in CDPs, pDCs (dis-                    of CD8α+ DCs, particularly in the spleen, CD8α+ DCs
including differentiation,
                                  cussed in detail above) and conventional DCs, par-                  are still present in the absence of BATF3 (REFS 89,113).
proliferation and apoptosis.
                                  ticularly the CD8α+ and CD103+ DC subsets (FIG. 3).                 Nevertheless, Batf3–/– mice exhibit severe defects in their
Cross-priming                     IRF8‑deficient mice lack many mature DC subsets,                    capacity to respond to pathogen infections — includ-
A mechanism by which              including Langerhans cells90,104. In addition to regulating         ing West Nile virus, influenza virus and Toxoplasma
immunogenic CD8+ T cells are      the generation of pDCs and conventional DC subsets,                 gondii infections111,114–116 — and an impaired ability to
activated by the presentation
of an antigen that was not
                                  IRF8 controls various functional features of DCs, such              mediate cross-priming. This suggests that the main role
synthesized by the                as the expression of TLR9 and IFNα by pDCs and the                  of BATF3 may be in regulating the cross-presentation of
antigen-presenting cell itself.   production of IL‑12 by CD8α+ DCs93. Although IRF8                   exogenous antigens to CD8+ T cells (BOX 1).


NATURE REVIEWS | IMMUNOLOGY	                                                                                                VOLUME 12 | FEBRUARY 2012 | 109

                                                       © 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

                         The transcriptional network in CD8α+ and CD103+                         IRF2 acts as a transcriptional repressor of genes
                         DCs. As outlined above, a deficiency of IRF8, E4BP4,                encoding type I IFNs (IFNα and IFNβ) and thus limits
                         ID2 or BATF3 results in a lack of both the CD8α+ and                inflammation120,121. Mice lacking IRF2 exhibit a selective,
                         CD103+ DC subsets. This dependency on the same                      cell-autonomous loss of CD4+ DCs in the spleen and epi-
                         transcriptional regulators, as well as similar func-                dermis, and these subsets are restored when type I IFN
                         tional and localization characteristics13,47,111, suggests          signalling is eliminated122,123. Precisely how IRF2 is regu-
                         that CD8α+ and CD103+ DCs represent a single sub-                   lated during DC development is not clear, but one role of
                         set. However, similar developmental requirements                    IRF2 may be to protect developing DCs from maturation
                         do not necessarily imply close lineage relationships;               arrest when the levels of type I IFNs are elevated. IRF2
                         for example, PU.1 is required for the development of                has also been reported to form complexes with IRF8
                         multiple distinct lineages, including macrophages,                  and to act cooperatively with this factor in regulating
                         granulo­ ytes and DCs, but very different mechanisms
                                   c                                                         the expression of IL12 (REF. 124).
                         are involved66,117.                                                     IRF4, a crucial regulator of many aspects of lympho-
                             One approach used to investigate the relative con-              cyte differentiation125, is most highly expressed in CD4+
                         tributions of IRF8, ID2 and BATF3 to conventional                   DCs (in which IRF8 expression is lowest). In line with
                         DC differentiation has been to engineer a fluorescent               this expression profile, CD4+ DCs are largely absent from
                         reporter into the Id2 locus (to generate Id2gfp mice)89.            mice lacking IRF4, but these mice also show defects in
                         Analyses of Id2gfp mice showed that neither CDPs nor                pDCs in the spleen126,127. Moreover, DCs generated in vitro
                         pre-DCs expressed ID2. Thus, it is more likely that ID2             through the stimulation of bone marrow precursors with
                         drives the terminal differentiation of different DC sub-            FLT3L appear to rely on IRF8 rather than IRF4, but those
                         sets rather than influencing early lineage-commitment               generated in the presence of GM‑CSF depend on IRF4
                         decisions. Using Irf8–/– or Batf3–/– mice crossed with              (REFS 126,127). Thus, it has been proposed that the main
                         Id2gfp mice, it became clear that IRF8 is required for              action of IRF4 is to coordinate signals from GM‑CSF
                         the generation of DC precursors from a very early                   stimulation through the nuclear factor-κB (NF‑κB) path-
                         time point, whereas BATF3 has a role later in conven-               way 128. The extent to which IRF4 and IRF8 regulate simi-
                         tional DC development, downstream of ID2 (REF. 89).                 lar or distinct sets of target genes, with or without PU.1,
                         BATF3‑deficient progenitors gave rise to precursors of              remains an open question that needs to be addressed.
                         SIRPα– DCs (which are the precursors of CD8α+ and                       RELB is a member of the NF-κB family and can func-
                         CD103+ DCs) in FLT3L‑containing cultures and to a                   tion either as an activator or as a repressor of transcrip-
                         lesser extent in vivo, although both the frequency of               tion by forming heterodimers with the p50 and p52
                         these DCs and their expression of CD8α were reduced                 NF‑κB family members. RELB is most highly expressed
                         compared with wild-type CD8α+ DCs89 (FIG. 2). Thus,                 in the CD8α– and CD11b+ DC subsets, and these DC
                         it is clear that IRF8, ID2 and BATF3 each functions at              subsets are absent in RELB-deficient mice129,130. To date,
                         a distinct point in the differentiation of CD103+ and               there have been no studies addressing the mechanism
                         CD8α+ DCs. What is lacking, however, is an under-                   by which RELB controls DC differentiation. It is pos-
                         standing of how the programmes that are activated by                sible that IRF4 — the deletion of which also results
                         each of these transcription factors interact to give rise           in a substantial loss of CD4+ DCs127 — may be a key
                         to the diversity of DC subsets with unique functions. At            target of the RELB pathway. RELB has a crucial role in
                         present, the major approaches for dissecting this net-              the upregulation of the signalling molecule CD40, which
                         work are the deletion of individual transcription factor            is required to induce immunogenic DCs and for the
                         genes from the entire haematopoietic compartment,                   induction of IFNα131,132. Loss of IFNα results in impaired
                         and/or limited sampling of tissues and inference with the           cross-priming of exogenous antigens132,133. CD40 acts
                         on/off regulatory switches. But these approaches provide            through TNF receptor-associated factor 6 (TRAF6) to
                         only ‘black and white snapshots’ of transcription factor            activate the NF‑κB cascade134. Strikingly, Traf6–/– mice
                         involvement in DC development. The establishment                    lack CD8α– DCs and have an impairment in DC matu-
                         of the identity of DC subsets, their differentiation and            ration similar to that observed in Relb–/– mice. It seems
                         their maintenance are unlikely to be so simple and may              likely that TRAF6 regulates CD4+ DC development in a
                         require combinatorial interactions between transcrip-               RELB-dependent manner.
                         tion factors to guide fate decisions118,119. Genome-wide                Much attention has been focused on delineating the
                         DNA-binding data will be crucial for the precise eluci-             development of CD8α+ and CD103+ DCs. However,
                         dation of the manner in which different transcription               despite the crucial role of CD8α – DCs in immune
                         factors work together to define different DC subsets.               responses — particularly in the activation of CD4 +
                                                                                             T cells18,19 and possibly of follicular helper T cells135–137
                         Transcriptional regulation of CD8α– DCs. Much less is               — a detailed understanding of the transcription factor
                         known about the transcription factors that regulate the             networks that drive CD8α– DC development is lacking.
                         differentiation decisions of CD8α– conventional DC lin-             Furthermore, transcription factors normally associ-
                         eages, despite the dominant role of these cells in present-         ated with CD8α+ and CD103+ DC lineages, such as ID2
                         ing antigens to CD4+ T cells. The transcription factors             (REF. 89) and IRF8, are differentially expressed among
                         IRF2, IRF4 and RELB have been shown to be important                 CD8α– DCs, suggesting that they may have additional
                         in the development of these subsets and their subsequent            roles in the generation of fully matured DCs that do not
                         maturation (FIG. 2; TABLE 2).                                       express CD8α.


110 | FEBRUARY 2012 | VOLUME 12	                                                                                     www.nature.com/reviews/immunol

                                              © 2012 Macmillan Publishers Limited. All rights reserved
1 células dendríticas - primeira apresentação
1 células dendríticas - primeira apresentação
1 células dendríticas - primeira apresentação

More Related Content

Similar to 1 células dendríticas - primeira apresentação

Chapter 29 dendritic cells
Chapter 29 dendritic cellsChapter 29 dendritic cells
Chapter 29 dendritic cellsNilesh Kucha
 
Cells of the immune system
Cells of the immune system Cells of the immune system
Cells of the immune system Afra Fathima
 
Dc 11.11.14 amity
Dc 11.11.14 amityDc 11.11.14 amity
Dc 11.11.14 amityJaved Khan
 
dendritic cells/cosmetic dentistry courses
dendritic cells/cosmetic dentistry coursesdendritic cells/cosmetic dentistry courses
dendritic cells/cosmetic dentistry coursesIndian dental academy
 
tri insti poster
tri insti postertri insti poster
tri insti postermtchin08
 
Lecture 1 molecular tech. rdt 11 50-2020
Lecture 1 molecular tech. rdt 11 50-2020 Lecture 1 molecular tech. rdt 11 50-2020
Lecture 1 molecular tech. rdt 11 50-2020 Dr Vishnu Kumar
 
Lecture 1 molecular tech. RDT By Dr Vishnu Kumar Professor, Biochemistry
Lecture 1 molecular tech. RDT  By Dr Vishnu Kumar Professor, BiochemistryLecture 1 molecular tech. RDT  By Dr Vishnu Kumar Professor, Biochemistry
Lecture 1 molecular tech. RDT By Dr Vishnu Kumar Professor, BiochemistryDr Vishnu Kumar
 
Immunoreactivity and Immunotolerance
Immunoreactivity and ImmunotoleranceImmunoreactivity and Immunotolerance
Immunoreactivity and ImmunotoleranceDrAisheePal
 
MICA Antibodies in Renal Transplantation
MICA Antibodies in Renal TransplantationMICA Antibodies in Renal Transplantation
MICA Antibodies in Renal TransplantationChristos Argyropoulos
 
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdf
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdfCell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdf
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdfahntagencies
 
Recombinant dna technology introduction
Recombinant dna technology introductionRecombinant dna technology introduction
Recombinant dna technology introductionAmeena Kadar
 
Celulas tronco hematopoieticas
Celulas tronco hematopoieticasCelulas tronco hematopoieticas
Celulas tronco hematopoieticasdervoz2012
 
Basics of gene therapy
Basics of gene therapyBasics of gene therapy
Basics of gene therapyDr.reena singh
 
Metastasis- Cancer cell migration
Metastasis- Cancer cell migrationMetastasis- Cancer cell migration
Metastasis- Cancer cell migrationQussai Abbas
 

Similar to 1 células dendríticas - primeira apresentação (20)

Dendritic Cell in Oncology
Dendritic Cell in OncologyDendritic Cell in Oncology
Dendritic Cell in Oncology
 
Chapter 29 dendritic cells
Chapter 29 dendritic cellsChapter 29 dendritic cells
Chapter 29 dendritic cells
 
Cells of the immune system
Cells of the immune system Cells of the immune system
Cells of the immune system
 
Dc 11.11.14 amity
Dc 11.11.14 amityDc 11.11.14 amity
Dc 11.11.14 amity
 
Fimmu 05-00055
Fimmu 05-00055Fimmu 05-00055
Fimmu 05-00055
 
dendritic cells/cosmetic dentistry courses
dendritic cells/cosmetic dentistry coursesdendritic cells/cosmetic dentistry courses
dendritic cells/cosmetic dentistry courses
 
tri insti poster
tri insti postertri insti poster
tri insti poster
 
Lecture 1 molecular tech. rdt 11 50-2020
Lecture 1 molecular tech. rdt 11 50-2020 Lecture 1 molecular tech. rdt 11 50-2020
Lecture 1 molecular tech. rdt 11 50-2020
 
Lecture 1 molecular tech. RDT By Dr Vishnu Kumar Professor, Biochemistry
Lecture 1 molecular tech. RDT  By Dr Vishnu Kumar Professor, BiochemistryLecture 1 molecular tech. RDT  By Dr Vishnu Kumar Professor, Biochemistry
Lecture 1 molecular tech. RDT By Dr Vishnu Kumar Professor, Biochemistry
 
Immunoreactivity and Immunotolerance
Immunoreactivity and ImmunotoleranceImmunoreactivity and Immunotolerance
Immunoreactivity and Immunotolerance
 
PlOSone paper
PlOSone paperPlOSone paper
PlOSone paper
 
MICA Antibodies in Renal Transplantation
MICA Antibodies in Renal TransplantationMICA Antibodies in Renal Transplantation
MICA Antibodies in Renal Transplantation
 
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdf
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdfCell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdf
Cell adhesion, both cell-cell and cell-ECM, can influence whether a c.pdf
 
Recombinant dna technology introduction
Recombinant dna technology introductionRecombinant dna technology introduction
Recombinant dna technology introduction
 
Dendritic cells
Dendritic cellsDendritic cells
Dendritic cells
 
Dna vaccine
Dna vaccineDna vaccine
Dna vaccine
 
Celulas tronco hematopoieticas
Celulas tronco hematopoieticasCelulas tronco hematopoieticas
Celulas tronco hematopoieticas
 
Basics of gene therapy
Basics of gene therapyBasics of gene therapy
Basics of gene therapy
 
Dendritic cells biology of the skin
Dendritic cells biology of the skinDendritic cells biology of the skin
Dendritic cells biology of the skin
 
Metastasis- Cancer cell migration
Metastasis- Cancer cell migrationMetastasis- Cancer cell migration
Metastasis- Cancer cell migration
 

More from ufamimunologia

5 linfócitos b e receptores do tipo toll
5 linfócitos b e receptores do tipo toll5 linfócitos b e receptores do tipo toll
5 linfócitos b e receptores do tipo tollufamimunologia
 
5 seleção de linfócitos t reg
5 seleção de linfócitos t reg5 seleção de linfócitos t reg
5 seleção de linfócitos t regufamimunologia
 
3 moléculas acessórias de toll
3 moléculas acessórias de toll3 moléculas acessórias de toll
3 moléculas acessórias de tollufamimunologia
 
2 complemento -segunda apresentação
2 complemento -segunda apresentação2 complemento -segunda apresentação
2 complemento -segunda apresentaçãoufamimunologia
 
2 complemento receptores - segunda apresentação
2  complemento receptores - segunda apresentação2  complemento receptores - segunda apresentação
2 complemento receptores - segunda apresentaçãoufamimunologia
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentaçãoufamimunologia
 
2 complemento e imunidades -segunda apresentação
2 complemento e imunidades -segunda apresentação2 complemento e imunidades -segunda apresentação
2 complemento e imunidades -segunda apresentaçãoufamimunologia
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentaçãoufamimunologia
 
1 neutrófilos - primeira apresentação.pdf
1  neutrófilos - primeira apresentação.pdf1  neutrófilos - primeira apresentação.pdf
1 neutrófilos - primeira apresentação.pdfufamimunologia
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentaçãoufamimunologia
 

More from ufamimunologia (18)

5 n fkapa b
5 n fkapa b5 n fkapa b
5 n fkapa b
 
5 linfócitos t
5 linfócitos t5 linfócitos t
5 linfócitos t
 
5 linfócitos b
5 linfócitos b5 linfócitos b
5 linfócitos b
 
5 linfócitos b e receptores do tipo toll
5 linfócitos b e receptores do tipo toll5 linfócitos b e receptores do tipo toll
5 linfócitos b e receptores do tipo toll
 
5 seleção de linfócitos t reg
5 seleção de linfócitos t reg5 seleção de linfócitos t reg
5 seleção de linfócitos t reg
 
4 cd1a reciclagem
4 cd1a reciclagem4 cd1a reciclagem
4 cd1a reciclagem
 
4 cd1
4 cd14 cd1
4 cd1
 
4 cd1 nature
4 cd1 nature4 cd1 nature
4 cd1 nature
 
3 prr
3 prr3 prr
3 prr
 
3 nod
3 nod3 nod
3 nod
 
3 moléculas acessórias de toll
3 moléculas acessórias de toll3 moléculas acessórias de toll
3 moléculas acessórias de toll
 
2 complemento -segunda apresentação
2 complemento -segunda apresentação2 complemento -segunda apresentação
2 complemento -segunda apresentação
 
2 complemento receptores - segunda apresentação
2  complemento receptores - segunda apresentação2  complemento receptores - segunda apresentação
2 complemento receptores - segunda apresentação
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentação
 
2 complemento e imunidades -segunda apresentação
2 complemento e imunidades -segunda apresentação2 complemento e imunidades -segunda apresentação
2 complemento e imunidades -segunda apresentação
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentação
 
1 neutrófilos - primeira apresentação.pdf
1  neutrófilos - primeira apresentação.pdf1  neutrófilos - primeira apresentação.pdf
1 neutrófilos - primeira apresentação.pdf
 
1 basófilo e mastócitos - primeira apresentação
1  basófilo e mastócitos - primeira apresentação1  basófilo e mastócitos - primeira apresentação
1 basófilo e mastócitos - primeira apresentação
 

Recently uploaded

College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...Miss joya
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.MiadAlsulami
 
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call girls in Ahmedabad High profile
 
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoybabeytanya
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Serviceparulsinha
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Miss joya
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escortsaditipandeya
 
Bangalore Call Girls Hebbal Kempapura Number 7001035870 Meetin With Bangalor...
Bangalore Call Girls Hebbal Kempapura Number 7001035870  Meetin With Bangalor...Bangalore Call Girls Hebbal Kempapura Number 7001035870  Meetin With Bangalor...
Bangalore Call Girls Hebbal Kempapura Number 7001035870 Meetin With Bangalor...narwatsonia7
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...narwatsonia7
 
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service CoimbatoreCall Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatorenarwatsonia7
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...narwatsonia7
 
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safenarwatsonia7
 
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoybabeytanya
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girlsnehamumbai
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalorenarwatsonia7
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escortsvidya singh
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Miss joya
 

Recently uploaded (20)

College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
 
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
Call Girls Service Navi Mumbai Samaira 8617697112 Independent Escort Service ...
 
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Vashi Mumbai📲 9833363713 💞 Full Night Enjoy
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
 
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore EscortsVIP Call Girls Indore Kirti 💚😋  9256729539 🚀 Indore Escorts
VIP Call Girls Indore Kirti 💚😋 9256729539 🚀 Indore Escorts
 
Bangalore Call Girls Hebbal Kempapura Number 7001035870 Meetin With Bangalor...
Bangalore Call Girls Hebbal Kempapura Number 7001035870  Meetin With Bangalor...Bangalore Call Girls Hebbal Kempapura Number 7001035870  Meetin With Bangalor...
Bangalore Call Girls Hebbal Kempapura Number 7001035870 Meetin With Bangalor...
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
 
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service CoimbatoreCall Girl Coimbatore Prisha☎️  8250192130 Independent Escort Service Coimbatore
Call Girl Coimbatore Prisha☎️ 8250192130 Independent Escort Service Coimbatore
 
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
Russian Call Girls in Delhi Tanvi ➡️ 9711199012 💋📞 Independent Escort Service...
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
 
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCREscort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
 
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Marathahalli 📞 9907093804 High Profile Service 100% Safe
 
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night EnjoyCall Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoy
Call Girl Number in Panvel Mumbai📲 9833363713 💞 Full Night Enjoy
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
 
sauth delhi call girls in Bhajanpura 🔝 9953056974 🔝 escort Service
sauth delhi call girls in Bhajanpura 🔝 9953056974 🔝 escort Servicesauth delhi call girls in Bhajanpura 🔝 9953056974 🔝 escort Service
sauth delhi call girls in Bhajanpura 🔝 9953056974 🔝 escort Service
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
 

1 células dendríticas - primeira apresentação

  • 1. REVIEWS Transcriptional programming of the dendritic cell network Gabrielle T. Belz and Stephen L. Nutt Abstract | Specialized subsets of dendritic cells (DCs) provide a crucial link between the innate and adaptive immune responses. The genetic programme that coordinates these distinct DC subsets is controlled by both cytokines and transcription factors. The initial steps in DC specification occur in the bone marrow and result in the generation of precursors committed to either the plasmacytoid or conventional DC pathways. DCs undergo further differentiation and lineage diversification in peripheral organs in response to local environmental cues. In this Review, we discuss new evidence regarding the coordination of the specification and commitment of precursor cells to different DC subsets and highlight the ensemble of transcription factors that control these processes. Dendritic cells (DCs) are essential for antigen presen- Unravelling the complexity of the DC network tation and the initiation of protective T cell responses DCs are a heterogeneous group of cells that have been and, thus, constitute a front-line defence against invad- divided into different subsets. This segregation was ing pathogens. DCs are located throughout the body initially based on their distinct patterns of cell-surface and form a sophisticated and complex network that molecule expression. The four major categories of DCs allows them to communicate with different popula- are conventional DCs, which predominate in the steady tions of lymphocytes, thereby forming an interface state; Langerhans cells; plasmacytoid DCs (pDCs); between the external environment and the adaptive and monocyte-derived DCs, which are induced in immune system. To provide this protection, different response to inflammation. subsets of DCs have evolved, and these DC subsets are specialized to exist in distinct locations, where Conventional DCs. Conventional DCs are special- they acquire antigens and transport them to draining ized for antigen processing and presentation. They lymph nodes for T cell priming. The DC network is can be grouped into two main classes based on their programmed by a group of transcription factors that localization in tissues and their migratory pathways determine the specification and differentiation of the as they circulate in the body (FIG. 1a; TABLE 1). The first different subsets of DCs. Recently, it has been shown category of conventional DCs is generally referred to that defects in transcription factor expression under- as the migratory DCs. These DCs develop from early pin developmental defects in DCs and other immune precursors in the peripheral tissues, where they act cells, and these defects result in severe immuno­ as antigen-sampling sentinels. From the peripheral deficiencies and enhanced susceptibility to bacterial, tissues, they migrate to the regional lymph nodes via fungal and viral infections in humans1–3. Thus, disrup- afferent lymphatics, a process that is accelerated in tion of transcription factor expression and selective loss response to danger signals, such as those that occur of DC subsets is likely to have important implications during pathogen infection. Migratory DCs are not for human disease. found in the spleen and are restricted to the lymph In this Review, we focus on the role of transcription nodes4, where they constitute a variable proportion of Division of Molecular Immunology, Walter and factors in generating different DC subsets and highlight the steady-state DC population; this proportion Eliza Hall Institute of Medical the synergistic functions of cytokines in shaping DC depends on the specific tissues that are drained by the Research, 1G Royal Parade, fate decisions. Furthermore, we discuss the molecular lymph node5 (FIG. 1). Migratory DCs can be broadly Melbourne, Victoria 3052, pathways that may allow plasticity in DC fate decisions divided into CD11b+ DCs (also known as dermal or Australia. e-mails: belz@wehi.edu.au; and that enable the rapid recruitment and differen- interstitial DCs) and CD11b– DCs6, which have more nutt@wehi.edu.au tiation of DCs in response to diverse environmental recently been shown to express CD103 (also known as doi:10.1038/nri3149 stimuli. integrin αE)4,7. NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | FEBRUARY 2012 | 101 © 2012 Macmillan Publishers Limited. All rights reserved
  • 2. REVIEWS a Lymph nodes Spleen Blood-derived DCs Lymphoid tissue-resident DCs Migratory DCs Monocyte- pDCs CD4–CD8α– CD4+ DCs CD8α+ DCs CD103+ CD11b+ Langerhans derived DCs (DN) DCs DCs (interstitial or cells dermal) DCs Inflammation Steady state b pDC DN DC CD8α+ DC Bone marrow CD103+ CD11b+ Langerhans Non-lymphoid tissue DC DC cell Skin CD11b+ DC-SIGN+ Lymph node Langerhans cell monocyte-derived DC Epidermis CD103+ CD11b+ DC DC Dermis HSC CD103+ DC CD11b+ DC FLT3+ CMP MDP Pre-DC pDC Monocyte Blood CDP Pre-DC DN DC CD4+ DC CD8α+ DC CD11b+ DC-SIGN+ monocyte-derived DC Spleen Figure 1 | Differentiation and trafficking of DC subsets. a | The figure shows the organization of the dendritic cell (DC) network, and includes the key surface phenotype markers of different DC subsets, which are delineated on the basis of their localization in secondary lymphoid tissues. Gut-associated DCs that express both CD103 and CD11b have been included in Nature Reviews | Immunology the CD11b+ DC subset. Inflammatory monocyte-derived DCs are rapidly recruited to sites of inflammation, whereas other DC subsets are normally present in the steady state. The relationship between inflammatory and steady-state DCs remains an open issue. Moreover, it is unclear whether monocyte-derived DCs can arise through in situ proliferation in addition to arriving at tissues via the circulation. b | In the mouse bone marrow, haematopoietic stem cells (HSCs) differentiate into common myeloid progenitors (CMPs), a fraction of which express FMS-related tyrosine kinase 3 (FLT3) and differentiate into more-restricted macrophage and DC progenitors (MDPs). MDPs appear to be the direct precursor to common DC progenitors (CDPs), which give rise to the DC lineages. CDPs produce precursor DCs (pre-DCs) and plasmacytoid DCs (pDCs) that exit the bone marrow and travel through the blood to secondary lymphoid organs and non-haematopoietic tissues. A small proportion of DCs may also be derived from CLPs in the bone marrow and from early T cell progenitors in the thymus. Under steady-state conditions, lymphoid tissue-resident DCs that arise from pre-DCs are the only subsets found in the spleen. This population is comprised of three conventional DC subsets, namely CD4+ DCs, CD8α+ DCs and CD8α–CD4– double-negative (DN) DCs. Peripheral lymph nodes contain CD8α+ and CD8α– DC populations but are also populated by two groups of migratory DCs. Langerhans cells develop in the epidermis and migrate through the basement membrane to the draining lymph nodes via terminal lymphatic vessels that arise in the dermis. The dermal DC population is broadly composed of CD11b+ and CD103+ DCs, and these cells migrate through the lymphatics to the lymph node. Monocytes arrive at tissues from the blood. In response to inflammation, they can develop into monocyte-derived DCs, which adopt many of the characteristics of conventional DCs. DC-SIGN, DC-specific ICAM3‑grabbing non-integrin. 102 | FEBRUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol © 2012 Macmillan Publishers Limited. All rights reserved
  • 3. REVIEWS Table 1 | Phenotypic markers of DC subsets DC subset DC type CD8α CD103 CD205 EPCAM CD11b B220 or DC-SIGN Langerin Antigen Major (CD326) CD45RA (CD207) presentation cytokine produced pDCs Lymphoid- +/– – – – – + ++ – Poor IFNα resident DCs CD8α+ DCs Lymphoid- + low + – + – – +/– Cross- IL‑12p70, resident DCs presentation IFNλ on MHC class I; expression of cystatin C CD4+ DCs Lymphoid- – – – – + – – – Presentation on resident DCs MHC class II DN DCs Lymphoid- – – – – + – – – Presentation on resident DCs MHC class II CD11b+ Migratory – +/– + – + – ND – Presentation on DCs DCs MHC class II CD103+ DCs Migratory Cross- • Lung DCs – + ++ +/– – – – + presentation on • Intestine – + – – + – – – MHC class I Langerhans Migratory – – ++ + + – – ++ Presentation IL‑10 cells DCs of self antigens for tolerance induction Monocyte- Induced by – – – – + – + – Cross- TNF derived inflammation presentation DCs DC, dendritic cell; DC-SIGN, DC-specific ICAM3-grabbing non-integrin; DN, double-negative; EPCAM, epithelial cell adhesion molecule; IFN, interferon; IL, interleukin; ND, not determined; pDC, plasmacytoid DC. The second major category of conventional DCs that are present early in embryonic development and is the lymphoid tissue-resident DCs that are found in that undergo a proliferative burst in the epidermis in the major lymphoid organs, such as the lymph nodes, the first few days after birth24. spleen and thymus. These DCs can be further classified by their expression of the surface markers CD4 and pDCs. pDCs are quiescent cells that are broadly distrib- CD8α into CD4+ DCs, CD8α+ DCs and CD4–CD8α– uted in the body. They are characterized by their ability DCs (typically referred to as double-negative DCs)8,9 to rapidly produce large amounts of type I interferons (TABLE 1). CD8α + DCs are noted for their capacity to (IFNs)25,26, a feature most evident during viral infection. cross-present antigens10 and for their major role in pDCs express several characteristic markers, includ- priming cytotoxic CD8+ T cell responses11–16 (BOX 1). ing sialic acid-binding immunoglobulin-like lectin H CD4 + DCs and CD4 –CD8α – DCs can also present (SIGLEC‑H) and bone marrow stromal antigen 2 (BST2) MHC class I‑restricted antigens in some settings 15,17, in mice and blood DC antigen 2 (BDCA2; also known but appear to be more efficient at presenting MHC as CLEC4C) and leukocyte immunoglobulin-like recep- class  II-associated antigens to CD4 + T  cells 18–20 . tor, subfamily A, member 4 (LILRA4; also known as Lymphoid tissue-resident DCs do not traffic from ILT7) in humans. In addition, both mouse and human other tissues but develop from precursor DCs found pDCs express CD45RA27. pDCs have poor antigen- in the lymphoid tissues themselves21. In the absence of presenting capacity, and their precise contribution to infection, they exist in an immature state (which is char- immune responses is still unclear 28. acterized by a high endocytic capacity and lower MHC class II expression compared with activated DCs), and Monocyte-derived DCs. Under inflammatory con- their residency in lymphoid tissues makes them ideally ditions, circulating blood monocytes can be rapidly placed to sense antigens or pathogens that are transported mobilized and can differentiate into cells that possess in the blood12,22,23. many prototypical features of DCs21,29–32 (FIG. 1). In the steady state, monocytes express the macrophage colony- Langerhans cells. Langerhans cells are resident in the stimulating factor receptor (M-CSFR; also known as skin and, like migratory DCs, migrate to the lymph CD115), which is essential for their development, as well nodes to present antigens (FIG. 1). However, unlike con- as other markers, such as LY6C and CX3C-chemokine ventional DCs, which arise from a bone marrow precur- receptor 1 (CX3CR1). In response to growth factors such as sor cell, Langerhans cells are derived from a local LY6C+ granulocyte–macrophage colony-stimulating factor myelomonocytic precursor cell population in the skin. (GM-CSF) in vitro or to Toll-like receptor 4 (TLR4) ligands This precursor population originates from macrophages or bacteria in vivo, fully differentiated monocyte-derived NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | FEBRUARY 2012 | 103 © 2012 Macmillan Publishers Limited. All rights reserved
  • 4. REVIEWS Box 1 | Direct presentation, cross-presentation and cross-dressing Cytokines regulate DC development The differentiation of DCs from haematopoietic Efficient presentation of antigens to CD8+ T cells depends on the generation of progenitor cells relies on the activity of cytokines, peptides for loading into MHC class I complexes. Several pathways have been in particular FMS-related tyrosine kinase 3 ligand uncovered to achieve this activation of CD8+ T cells. Direct antigen presentation by (FLT3L), M‑CSF and GM‑CSF. These cytokines con- infected or malignant cells ensures the destruction of these cells by cytotoxic CD8+ trol the initial production and lineage diversification T cells (CTLs), without harming adjacent healthy cells. The restriction of antigen presentation to directly infected cells, however, is not sufficient to ensure the activation of DCs, although the factors that regulate the expres- of CD8+ T cells, particularly when the pathogen does not infect professional sion of the receptors for these key cytokines and the antigen-presenting cells, such as dendritic cells (DCs). In this case, CD8+ T cells can be downstream transcriptional programmes instigated by activated by DCs that present extracellular antigens on their MHC class I molecules via FLT3L, M‑CSF and GM‑CSF are only now emerging. the process of cross-presentation. This pathway can result in the generation of CTLs The ability of these cytokines to stimulate the differ- that are reactive to foreign antigens, or in the induction of tolerance through the entiation of DCs in vitro (BOX 2) provides a tractable deletion of autoreactive CD8+ T cells following the cross-presentation of self antigens. model system to address the influence of extrinsic In mice, most focus has been on the cross-presenting capacities of CD8α+ DCs, and factors on the DC transcriptional network. more recently CD103+ DCs6,7,10,138, although other populations also possess this ability. FLT3L and FLT3 constitute the best-characterized For example, fully differentiated monocyte-derived DCs that express DC-specific growth factor–receptor axis for DCs, as mouse haemato­ ICAM3‑grabbing non-integrin (DC‑SIGN) are potent cross-presenting cells33. An alternative pathway for the acquisition of peptide–MHC complexes is known as poietic progenitor cells cultured with FLT3L generate a ‘cross-dressing’139,140 (originally known as trogocytosis141). Through this pathway, DCs diverse array of conventional DC subsets and pDCs36,37. acquire preformed peptide–MHC class I complexes from infected cells. Complexes In agreement with the important role of FLT3L in DC acquired in this way can drive the activation of memory, but not naive, CD8+ T cells differentiation, DCs can be generated from essentially during viral infection, perhaps owing to the lower activation threshold of memory cells. any FLT3+ progenitor cell either in vitro or following The molecular mechanisms involved in cross-presentation and cross-dressing are only adoptive transfer in vivo38–40 (FIG. 2). In addition, enforced beginning to be unravelled. expression of FLT3 in megakaryocyte–erythrocyte progenitors (MEPs), which are normally FLT3–, results in the acquisition of DC potential41. An instructive role DCs emerge. Similarly to conventional DCs, monocyte- for FLT3 in DC development is further supported by the derived DCs express CD11c, MHC class II molecules, finding that in all cell lineages except the DC lineage, CD24 and SIRPα (also known as CD172a), and they upreg- FLT3 is downregulated following differentiation and, ulate their expression of DC-specific ICAM3‑grabbing at least in the case of B cells, this repression of FLT3 is non-integrin (DC‑SIGN; also known as CD209a) but essential for further development 42. lose expression of both M‑CSFR and LY6C33 (TABLE 1). Mice deficient for FLT3L or signal transducer and Monocyte-derived DCs also express the macrophage activator of transcription 3 (STAT3; a signalling mol- marker MAC3 (also known as CD107b and LAMP2)21,32. ecule downstream of FLT3) have markedly reduced In addition, these cells acquire potent antigen-presenting numbers of lymphoid-resident conventional DCs and capacity, including the ability to cross-present antigens33–35 pDCs, whereas mice lacking FLT3 have a milder pheno- (BOX 1). Thus, it is emerging that monocyte-derived DCs type, which suggests the presence of a second ligand for are a crucial reservoir of professional antigen-presenting this receptor 43–46. FLT3 signalling is also crucial for the cells (APCs) that are recruited into immune responses development of migratory DCs, as the numbers of both to certain microorganisms and potentially have an pre-DCs and CD103+ DCs were found to be reduced in emergency back-up role in cases of acute inflammation. a range of tissues from Flt3l–/– mice compared with the Box 2 | In vitro models for investigating DC development and behaviour The establishment of well-defined cell culture systems that allow the generation of large numbers of dendritic cells (DCs) from bone marrow has been instrumental for understanding DC biology. Recent refinement of the tools and surface markers used to analyse cultures now allows the resolution of DC precursors equivalent to those found in vivo, together with fully differentiated DC subsets, in the tissue culture flask. Generation of steady-state DC subsets in vitro The development of steady-state DCs depends on signalling through FMS-related tyrosine kinase 3 (FLT3), which is expressed on the surface of DC precursors. Bone marrow precursors cultured with FLT3 ligand (FLT3L) give rise to plasmacytoid DCs (pDCs) and multiple lymphoid tissue-resident conventional DC subsets36,37,89. Intriguingly, in vitro-generated DCs do not express CD4 or CD8, but their patterns of expression of the markers CD103, CD11b, CD172a and CD24 indicate the presence of conventional DC subsets in addition to DC precursors. Generation of monocyte-derived DCs in vitro Perhaps the most commonly used approach to generate DCs involves the culture of bone marrow precursors in a medium supplemented with granulocyte–macrophage colony-stimulating factor (GM-CSF). As these DCs can also be produced by culturing monocytes in GM‑CSF and interleukin‑4 (IL‑4), they are referred to as monocyte-derived DCs and correspond to the dominant inflammatory DC type that is mobilized during some bacterial infections33. These highly refined culture systems allow comparative studies between different DC subsets. Furthermore, the ability to generate large numbers of DCs in vitro should greatly facilitate molecular exploration of the genomic and transcriptional machinery that leads to the generation of different DC subsets. 104 | FEBRUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol © 2012 Macmillan Publishers Limited. All rights reserved
  • 5. REVIEWS BATF3 FLT3L Transitional CD103+ DC Lymphocytes pre-DC ID2 E4BP4 BATF3 CLP FLT3L IRF8 Pre-CD8α+ DC PU.1hi IRF2 CD8α+ DC ID2hi PU.1 GFI1 PU.1 RELB IRF4 Ikaros (null) ? FLT3+ LMPP MDP CDP Pre-DC CMP GM-CSF M-CSF FLT3L CD11b+ DC Monocyte Immature pDC Loss M-CSF GM-CSF of E2-2 E2-2 Ikaros (L) IRF8 PU.1low ID2– E2-2hi Macrophage Monocyte- Mature pDC derived DC Figure 2 | Growth factors and transcription factors that regulate DC differentiation. The developmental pathways from myeloid and lymphoid progenitors to precursor dendritic cells (pre-DCs) in the bone marrow and Reviews | Immunology Nature the peripheral diversification of DC subsets are shown (see FIG. 1 legend for details). The approximate points at which key transcription factors are first required for DC development are indicated by vertical lines. Stages at which key growth factors have been determined to be essential are indicated. The development of both DCs and monocytes depends on high concentrations of PU.1, which regulates the expression of the cytokine receptors FMS-related tyrosine kinase 3 (FLT3), macrophage colony-stimulating factor receptor (M-CSFR) and granulocyte–macrophage colony-stimulating factor receptor (GM-CSFR). The development of CD8α+ and CD103+ DCs relies on the stepwise activity of interferon-regulatory factor 8 (IRF8), inhibitor of DNA binding 2 (ID2), E4 promoter-binding protein 4 (E4BP4) and basic leucine zipper transcription factor, ATF-like 3 (BATF3), as well as on FLT3 signalling. CD11b+ DCs depend on a unique set of transcription factors, including RELB, IRF2, IRF4 and Ikaros, and to some extent on the cytokines M‑CSF and GM‑CSF. The plasmacytoid DC (pDC) lineage requires IRF8, a low level of PU.1 and the absence of ID2. The differentiation of pDCs from an immature precursor requires E2‑2 and Ikaros, with induced loss of E2‑2 converting pDCs into cells that closely resemble CD8α+ conventional DCs. CDP, common DC progenitor; CLP, common lymphoid progenitor; CMP, common myeloid progenitor; FLT3L, FLT3 ligand; GFI1, growth factor independent 1; LMPP, lymphoid-primed multipotent progenitor; MDP, macrophage and DC progenitor. numbers in wild-type mice. By contrast, the development that resemble monocyte-derived DCs, while repress- of CD11b+ DCs and Langerhans cells is largely independ- ing the development of pDCs in a STAT5‑dependent ent of FLT3L47. Thus, FLT3L has two distinct roles in DC manner 46,51. This finding has generally been interpreted biology: it is required for the early development of DCs to show that GM‑CSF has a greater role in the produc- from haematopoietic progenitors; and later it functions tion of monocyte-derived DCs than in the generation to maintain DC homeostasis by promoting limited levels of other DC subsets21,52, although the importance of this of proliferation of DCs in peripheral tissues43. process in vivo is still to be established. In addition to FLT3L, GM‑CSF has long been known M-CSF is the major cytokine involved in the produc- to stimulate DC differentiation in culture (BOX  2) . tion of monocytes and macrophages53. A role for this However, GM‑CSF is not essential for DC differentia- cytokine in DC biology was suggested by the expression tion in the steady state, as mice that lack the GM‑CSF of M‑CSFR by DCs54,55 but came to prominence with the receptor (GM-CSFR) have only mildly reduced num- finding that M‑CSFR-deficient mice lack Langerhans bers of DCs48. Nonetheless, GM‑CSF is not completely cells56. Surprisingly, mice lacking M‑CSF (op/op mice) redundant in DC production, as mice deficient in both have normal numbers of Langerhans cells, a quandary GM‑CSF and FLT3L have a greater loss of DCs than either that was resolved by the identification of interleukin‑34 single-knockout strain44. Moreover, other recent studies (IL‑34) as a second ligand for M‑CSFR57. The relatively have demonstrated that GM‑CSF is necessary for there normal DC numbers in mice lacking M‑CSF, despite to be normal numbers of CD103+CD11b+ DCs in the the profound reduction in monocytes, demonstrates lamina propria49,50. The addition of GM‑CSF to cultures that the monocytic system is not the major source of of bone marrow cells promotes the development of cells steady-state DCs. M‑CSF is, however, required for the NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | FEBRUARY 2012 | 105 © 2012 Macmillan Publishers Limited. All rights reserved
  • 6. REVIEWS normal development of CD103–CD11b+ DCs in non- that specifies the DC lineage in more-immature lymphoid tissues49 and is able to support conventional pro­ enitors and then drives differentiation into the DC g DC and pDC differentiation in cell culture in the subsets. Three transcription factors — PU.1 (encoded by absence of FLT3 (REF. 58). Sfpi1), Ikaros and growth factor independent 1 (GFI1) — appear to be prime candidates for DC-specifying fac- Stages of programming DC identity tors. In addition, the signalling and transcription factors DC ontogeny. Although it is well established that all STAT3 and STAT5 are known to have a role in DC differ- DCs, with the exception of Langerhans cells, are derived entiation, as they mediate the signals transduced through from bone marrow-resident haematopoietic stem cells FLT3 and GM‑CSFR, respectively (TABLE 2). (HSCs), mapping the origins of the DC lineages has PU.1 belongs to the ETS family of transcription proven to be both difficult and controversial. Early factors, which has multiple context-specific roles in transfer experiments led to the surprising conclusion haemato­poiesis. PU.1 is an attractive candidate for being that DCs can develop with approximately equal effi- a crucial regulator of the DC lineages, as it is expressed by ciency from both lymphoid and myeloid pro­genitors59,60, all DCs and by CDPs66–68. A role for PU.1 in DC develop- whereas in vitro cultures with GM‑CSF show that DCs ment was initially suggested by the analysis of mice with can arise from monocytic precursors, as mentioned a germline deficiency of PU.1. Indeed, one such study above. However, monocytes are not likely to be a major concluded that PU.1 was necessary for all embryonic DC source of steady-state DCs in lymphoid organs, as development 69, although a second study reported that lineage-tracing experiments have shown that monocytic Sfpi1–/– fetal thymi could generate DCs70. However, these cells give rise to neutrophils and macrophages but not approaches could not distinguish between the require- DCs5. More recently, the adoption of a ‘FLT3‑centric’ ments for PU.1 in multipotent progenitors and the role view of haematopoiesis38–40,61 has established that most of PU.1 specifically in the DC lineages. Moreover, the steady-state DCs arise from FLT3+ progenitors (FIG. 2). impact of enforced expression of PU.1 in haematopoietic The pathway of DC differentiation from primitive progenitors suggests an instructive and concentration- bone marrow progenitors has been extensively reviewed dependent role for PU.1 in promoting macrophage and elsewhere4,53,62 and is only briefly summarized here. FLT3 DC development71–73. expression is first induced in a subset of the HSC com- A recent study used conditional gene deletion in partment that has only short-term pan-haematopoietic defined haematopoietic progenitors and CDPs to show repopulating activity, and the expression of this recep- that PU.1 is absolutely essential for the generation of tor is then maintained in lymphoid-primed multipotent all conventional DCs and pDCs both in vivo and in progenitors (LMPPs)63 and in a subpopulation of com- FLT3L‑containing cultures in vitro 66. Moreover, in mon myeloid progenitors (CMPs)38. CMPs are thought line with its established role in regulating GM‑CSFR to differentiate into macrophage and DC progenitors expression, PU.1 is required for GM‑CSF-induced DC (MDPs)64, which appear to be the direct precursors of differentiation from early haematopoietic progenitors. common DC progenitors (CDPs)40,61. Both MDPs and Among the many genes that are potentially regulated CDPs are proliferating cells that reside in the bone mar- by PU.1, Flt3 was demonstrated by molecular studies row and express FLT3 and M‑CSFR. CDPs differentiate to be directly regulated by PU.1 in DCs and haemato­ directly into pDCs and into the precursors of conven- poietic progenitors66. This regulation occurred in a tional DC subsets, termed pre-DCs, but they lack the concentration-dependent manner, as Sfpi1+/– cells had potential to give rise to macrophages21,65. Pre-DCs then reduced FLT3 expression and an impaired ability to leave the bone marrow and are found in blood, second- generate conventional DCs. Interestingly, previous stud- ary lymphoid organs and some tissues21,49,52,65, where ies have shown that FLT3 signalling is able to activate they mature into the conventional DC subsets (FIG. 1). PU.1 expression in MEPs, suggesting a self-reinforcing Differentiation into different conventional DC subsets loop between PU.1 and FLT3 in DCs41. Whether PU.1 appears to be a late step in DC development that is per- is required for monocyte-derived DC formation in vivo haps important in maintaining the stability or plasticity remains to be determined. of the peripheral DC compartment. The key features and Ikaros is a zinc-finger transcription factor that has mechanisms involved in the plasticity of the DC network important roles in haematopoiesis74. Expression of a are likely to include the short lifespan of mature conven- dominant-negative form of Ikaros that also impairs the tional DCs (5–7 days, although up to 25 days in some cir- function of other Ikaros family members, such as Aiolos, cumstances4,47); the rapid recruitment and proliferation of resulted in a complete loss of all conventional DC sub- pre-DCs and their capacity to respond to extrinsic signals sets. By contrast, a null mutation in the gene encoding (such as TLR ligands and pro-inflammatory cytokines); Ikaros led to the selective loss of CD11b+ DCs, with some and the active expression by DCs of transcription factors CD8α+ DCs being retained75. Whether Ikaros directly such as E2‑2 (also known as TCF4). regulates DC differentiation, as opposed to having a role in early myeloid progenitors, is at present unclear 76, as Initiating the DC programme in haematopoietic progeni- mice homozygous for a severely hypomorphic allele of tors. The information outlined above demonstrates the the Ikaros gene (IkL/L mice) lack mature pDCs but con- rapid progress that is being made in understanding the tain relatively normal numbers of conventional DCs77. developmental stages and cell biology of the DC lineages. DC‑specific conditional mutagenesis is now required to Much less is known about the transcriptional programme decipher the exact function of Ikaros in DCs (FIG. 2). 106 | FEBRUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol © 2012 Macmillan Publishers Limited. All rights reserved
  • 7. REVIEWS Table 2 | Transcription factors guiding steady-state DC subset development Transcription Transcription factor family Function Refs factor PU.1 (SFPI1, ETS-domain transcription factor; Required for the development of all DC subsets 66, SPI1) binds to PU box sequences 69,70 IRF2 Interferon-regulatory factor; inhibits Alters pDC ratios; in its absence the numbers of 122 the IRF1-mediated transcription of CD8α– DCs and Langerhans cells are reduced type I IFNs IRF4 Interferon-regulatory factor Required for non-CD8α+ DC lineage development 126,127 IRF8 (ICSBP) Interferon-regulatory factor Required for the development of pDCs and most 90,92, conventional DCs 93,104 GFI1 Zinc-finger protein; transcriptional GFI1 deficiency results in a 50% reduction in the 79 repressor numbers of conventional DCs and pDCs and increased numbers of Langerhans cells ID2 Inhibitor of DNA binding family Required for the development of CD103+ DCs and 47, protein containing HLH domains CD8α+ DCs in PLNs and spleen; not required for 87,89 DCs in MLNs E4BP4 (NFIL3) PAR-related bZIP transcription factor Required for the development of CD8α+ DCs 108 E2‑2 (TCF4) E protein containing bHLH domains Required for the development and maintenance 80,84 of pDCs STAT3 Signal transducer and activator of STAT3 deficiency results in a substantial reduction 46 transcription in conventional DC numbers STAT5A and Signal transducer and activator of Inhibit pDC development by interacting with IRF8; 51 STAT5B transcription deficiency results in reduced conventional DC and increased pDC numbers Ikaros (IKZF1) Zinc-finger DNA-binding protein Ikaros deficiency results in the absence of most 75,77 DCs; a hypomorphic mutation leads a specific loss of pDCs BATF3 bZIP family; heterodimerizes with BATF3-deficient mice fail to develop CD103+ DCs 13, JUN and show impaired survival of precursor CD8α+ DCs 89,111 RELB REL-homology domain family; RELB deficiency results in the loss of CD8α– DCs 129,130 interacts with NF‑κB family members SPIB ETS-domain transcription factor Required for human pDC differentiation 91 BATF3, basic leucine zipper transcription factor, ATF-like 3; bHLH, basic HLH; bZIP, basic leucine zipper; DC, dendritic cell; E4BP4, E4 promoter-binding protein 4; GFI1, growth factor independent 1; HLH, helix-loop-helix; ID2, inhibitor of DNA binding 2; IFN, interferon; IRF, interferon-regulatory factor; MLN, mesenteric lymph node; NF‑κB, nuclear factor‑κB; PAR, proline- and acidic-rich region; pDC, plasmacytoid DC; PLN, peripheral lymph node; STAT, signal transducer and activator of transcription. GFI1 is a small, zinc-finger-containing transcriptional it is surprising how little we actually understand about repressor that is important for early haemato­ oiesis78. p this process. Two developmental systems appear to be GFI1 is expressed in DC precursors, and Gfi1–/– mice in place to separate pDCs and conventional DCs (FIG. 2). have reduced numbers of all lymphoid-resident DC First, pDCs absolutely rely on the expression of the subsets, whereas Langerhans cell numbers were actu- E protein E2‑2 (REF. 80) and the absence of the E protein ally increased79. Interestingly, GFI1‑deficient haemato­ antagonist inhibitor of DNA binding 2 (ID2)81. Second, poietic progenitor cells were unable to develop into DCs pDCs have a uniquely low level of PU.1 (REF. 68) and an in vitro in the presence of either FLT3L or GM‑CSF and extremely high concentration of interferon-regulatory instead differentiated into macrophages, suggesting that factor 8 (IRF8), a transcription factor that can form GFI1 is a crucial modulator of DC versus macrophage a complex with PU.1 on a class of ‘composite’ DNA development (FIG. 2). elements82. One model to explain the pDC versus conventional Establishing pDC and conventional DC identity. DC lineage split is to assume that the conventional DC E protein Restriction of the developmental programme of DC is the default setting and that progenitors have to be The E proteins (including E12, E47, HEB and E2‑2) have progenitors to the conventional DC and pDC lineages diverted to the pDC lineage83. E2‑2 fits the bill for a fac- emerged as key regulators of occurs at the CDP stage40,61 (FIG. 2). Conventional DCs tor that could control this diversion, as it is abundantly the immune system. They are a and pDCs differ markedly in their appearance, functions expressed by pDCs and is required for pDC lineage family of basic helix-loop-helix and transcriptional programmes, and thus how a CDP specification80. Once progenitors have committed to a factors that work together with their antagonists, the ID is influenced to develop into either a pDC or a conven- pDC fate, it appears that continuous expression of E2‑2 proteins (ID1–ID4), to regulate tional DC is a question of major importance for under- is essential to maintain the mature pDC phenotype84. lymphocyte development. standing and manipulating DC biology. In this context, In mice, a specific deletion of the gene encoding E2‑2 NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | FEBRUARY 2012 | 107 © 2012 Macmillan Publishers Limited. All rights reserved
  • 8. REVIEWS in pDCs led to the expansion of a population of DCs they have been shown to share a few targets, such as that exhibit many characteristics of conventional DCs. Ciita94, Tlr9 (REF. 95) and Ifna96). However, BXH2 mice, In the in vivo setting, however, it is particularly dif- which harbour a spontaneous point mutation in Irf8, ficult to dissect whether E2‑2‑deficient pDCs undergo have defects in CD8α + DC development but not in phenotypic conversion to conventional DCs owing to a pDC generation. This mutation is thought to ablate reduction in E2‑2‑mediated repression of ID2, as pro- the interaction of PU.1 and IRF8, raising the possibil- posed, or whether normal conventional DC numbers ity that the PU.1–IRF8 complex is not as crucial for are increased in the absence of a full pDC compart- the differentiation of pDCs as for the development ment, as occurs in other settings. Nevertheless, pDCs of conventional DCs93. There are, as yet, no genome- are particularly sensitive to E2‑2 concentration, as both wide DNA-binding datasets for IRF8 and PU.1 in E2‑2‑deficient mice and patients with a rare mono- DCs, although similar data from macrophages sug- allelic loss of E2‑2 (Pitt–Hopkins syndrome) show gest that PU.1 might occupy most of the active regula- impaired pDC formation and function80. E2‑2 binds tory regions in DCs97,98 and that IRF8 might bind to directly to the promoters of several pDC-expressed a subset of these sites99. Importantly, in macrophages genes, including BDCA2, LILRA4, IRF7, the pre-TCR and myeloid progenitors, PU.1 is directly involved in α-chain gene, IRF8 and SPIB (which encodes a close nucleosome remodelling, and this leads to the generation relative of PU.1 that is expressed by human and mouse of an open chromatin conformation and histone modi‑ pDCs)80. The reliance of pDCs on an E protein such as fications, suggesting that PU.1 can directly programme E2‑2 may explain the observation that pDCs express the fate of myeloid cells97,98. many lymphocyte-associated transcripts (including The ETS-family transcription factor SPIB — the SPIB, RAG1, IL7R and TDT), as E proteins are central closest homologue of PU.1 in the mammalian genome to many aspects of lymphopoiesis27. — is expressed, within the DC lineages, specifically by ID proteins are direct inhibitors of DNA bind- pDCs100. Knockdown of the expression of either SPIB ing by E proteins. ID2 is the predominant ID protein or PU.1 in human haematopoietic progenitors strongly expressed in the DC lineage and is also involved in inhibits pDC formation, suggesting that both factors the development of multiple lineages during haemato­ function in human pDCs91. The extent of any functional poiesis, particularly that of lymphoid tissue-inducer redundancy between PU.1 and SPIB in pDCs has so far cells (LTi cells) and natural killer (NK) cells85–88. ID2 not been addressed in mice. expression is extremely low in CDPs, pre-DCs and In summary, the separation of pDC and conven- pDCs, whereas all conventional DC populations tional DC lineages represents the first major division Lymphoid tissue-inducer express high levels of ID2 (REF. 89) (FIG. 3). In line with in the DC pathway and requires the concerted action of cells (LTi cells). A cell type that is this expression pattern, ID2‑deficient mice have a both E proteins and the PU.1–IRF8 complex (FIGS 2,3). present in developing lymph profoundly altered conventional DC compartment Although the activation of STAT5 inhibits pDC for- nodes, Peyer’s patches and but still produce pDCs (see below)87. This leads to a mation in vitro 51, the exact signals that initiate this nasopharynx-associated model whereby the acquisition of high levels of ID2 process are not known. In addition, the ways in which lymphoid tissue (NALT). and subsequent suppression of E2‑2 activity blocks the E2‑2–ID2 and PU.1–IRF8 axes interact are still LTi cells are required for the development of these pDC development and allows progression along the unclear, as are the identities of most of the genes tar- lymphoid organs. The inductive conventional DC pathway. This model is clearly an geted by these transcription factors. pDCs represent the capacity of these cells for the oversimplification, as E2‑2‑deficient mice still produce end point of their lineage; however, the production of generation of Peyer’s patches some pDC progenitors and ID2 is not essential for the conventional DCs is only the first step in their further and NALT has been shown by adoptive transfer, and it is differentiation of all conventional DCs. diversification, which is outlined in the next section. generally assumed that they The development of pDCs also depends on PU.1, have a similar function in the IRF8 and potentially SPIB 66,90,91. pDCs are absent Genetic programming of conventional DC subsets formation of lymph nodes. in PU.1- or IRF8‑deficient mice 66,90,92, as well as in Although it is fairly clear that conventional DCs, pDCs, humans with a mutation in IRF8 (REF. 3), although it Langerhans cells and monocyte-derived DCs represent Nucleosome remodelling Changes in the nucleosome remains to be proven whether these factors function developmentally distinct lineages, the relationships structure are mediated by specifically in the pDC lineage or have a role in CDPs between the various anatomically, phenotypically and dedicated nuclear enzymes (FIG. 2) . However, circumstantial evidence favours a functionally distinct conventional DC populations (for example, ATP-dependent specific function for these factors in determining pDC remain to be fully elucidated. Conventional DCs are nucleosome-remodelling enzymes) that change the versus conventional DC fate. PU.1 is expressed at a thought to be derived from circulating pre-DCs21,65. accessibility of DNA and uniformly high level in CDPs, but at a lower level in Although it remains possible that multiple develop- the expression of genes. pDCs, with the timing of downregulation coinciding mentally distinct types of pre-DC exist and act as the with pDC formation66, whereas IRF8 is expressed at precursors for individual conventional DC lineages, Histone modifications very high levels in both CDPs and pDCs (FIG. 3). The we favour a model that considers all the populations Histones are essential to maintain DNA organization stoichiometric relationship between PU.1 and IRF8 is of conventional DCs as related subsets derived from a and may be modified by likely to be important for the pDC versus conventional single pre-DC population. In this model, the conven- methylation and acetylation — DC branch point, as both of these factors are known to tional DC subsets are induced by the environmental changes that are thought to function in a dose-dependent manner 66,93 and can bind milieu that they reside in, and thus it is the interaction keep genes active or silent, respectively — thereby altering to distinct DNA sequences both individually and in a of these extrinsic signals with the core transcriptional the genetic code read by complex 82. The extent to which PU.1 and IRF8 share programme of conventional DCs that dictates the transcriptional regulators. target genes in pDCs is currently unknown (although outcome of DC terminal differentiation. 108 | FEBRUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol © 2012 Macmillan Publishers Limited. All rights reserved
  • 9. REVIEWS BATF3 +++ clearly has a key role in the function of conventional DCs, IRF8 +++ Irf8–/– mice develop a myeloproliferative syndrome that is ID2 +++ characterized by the overproduction of granulocytes105. IRF4 ++ E4BP4 ++ This implies that IRF8 may also be required for the gen- CD103+ DC PU.1 +++ eration or maintenance of MDPs, which can give rise E2-2 + to monocytes, conventional DCs and pDCs. The exact Transitional developmental stage at which IRF8 exerts its activity on pre-DC Pre-CD8α+ DC BATF3 ++ conventional DCs remains to be fully determined. IRF8 +++ ID2 +++ E4BP4 is a mammalian basic leucine zipper (bZIP) IRF4 + transcription factor that is required for the development E4BP4 ++ of NK cells but not of other lymphocyte lineages106,107. IRF8 +++ IRF8 +++ PU.1 +++ ID2 +/– ID2 ++ CD8α+ DC E2-2 + In NK cells, E4BP4 acts in a dose-dependent man- PU.1 +++ PU.1 +++ ner downstream of the IL‑15 receptor to regulate ID2 CDP Pre-DC BATF3 ++ expression106,107. More recently, the induction of E4BP4 IRF8 + has been shown to be important for the development of ID2 ++ CD8α+ DCs108. E4BP4‑deficient CDPs had lower levels IRF4 +++ E4BP4 ++ of BATF3 expression than control CDPs, and enforced IRF8 +++ IRF8 +++ CD11b+ PU.1 +++ expression of BATF3 in the mutant cells rescued CD8α+ ID2 +/– ID2 +/– E2-2 + DC development in vitro108. Thus, E4BP4 is emerging as PU.1 +++ PU.1 +++ DC a key regulator of conventional DC diversity. Whether Immature pDC BATF3 + IRF8 +++ E4BP4 also acts through ID2 to mediate these effects ID2 +/– in DCs has not yet been addressed. IRF4 ++ ID2 is expressed by all conventional DC subsets, E4BP4 ++ IRF8 +++ Mature PU.1 + with the highest levels of expression in CD8α + and ID2 +/– pDC E2-2 +++ CD103+CD11b– DCs47,89. Loss of ID2 prevents the devel- PU.1 + opment of these two subsets in the skin-draining lymph E2-2 + nodes and spleen. However, CD103+CD11b+ DCs in the Figure 3 | Differential expression of transcription factors regulating DC mesenteric lymph nodes, together with CD4+ and CD4– differentiation. The stages of differentiation of conventional dendritic cells Immunology Nature Reviews | (DCs) and CD8α– DCs in lymphoid tissues, appear to develop nor- plasmacytoid DCs (pDCs) from the common DC progenitor (CDP) are shown, together mally in the absence of ID2 (REFS 13,47,87). Although it with the relative levels of expression of key transcription factors in each cell type indicated is at present unclear which E protein (or E proteins) — on an arbitrary scale. The expression pattern of some of the factors has not been determined at the earliest stages of DC ontogeny. –, no expression; +, low expression; E2A (also known as TCF3), E2‑2 or HEB (also known as ++, intermediate expression; +++, maximal expression; BATF3, basic leucine zipper TCF12) — is the crucial target of ID2 in conventional DCs, transcription factor, ATF-like 3; E4BP4, E4 promoter-binding protein 4; ID2, inhibitor selective deletion of the gene encoding E2‑2 in mature of DNA binding 2; IRF, interferon-regulatory factor; pre-DC, precursor DC. Figure is pDCs results in the spontaneous differentiation of pDCs modified, with permission, from REF. 89 © Macmillan Publishers Ltd. All rights reserved. into cells that exhibit conventional DC properties, perhaps through the induction of ID2 (REF. 84). Progress in dis- cerning the key target genes of E proteins in conventional Transcriptional regulators of CD8α + and CD103 + DCs will be required to understand why ID2 has such DCs. CD8α+ and CD103+ DCs have gained consider- an important role in CD8α+ and CD103+CD11b– DCs. NFAT (Nuclear factor of activated able attention owing to their specialized roles as induc- BATF3, which is also known as JUN-dimerization T cells). A family of ers of MHC class I‑restricted immune responses to protein p21SNFT, is a bZIP transcription factor that transcription factors that are pathogens. Moreover, a human BDCA3+ DC subset has acts to repress the activity of NFAT –AP1 complexes regulated by calcium signalling recently been identified that shares features with both by competing with FOS for JUN dimerization 109,110. and expressed by a variety of of these mouse DC subsets; such features include cross- BATF3 was the first transcription factor that appeared immune cells. presenting capacity and the expression of XC-chemokine to have an exclusive role in the development of the AP1 receptor 1 (XCR1)101–103. The differentiation of DC pre- CD8α+ DCs111, although it has since been shown to (Activator protein 1). cursors into the CD8α+ and CD103+ DC lineages appears be involved in the development of CD103+CD11b– A heterodimeric transcription to depend on the integration of four key transcription DCs in peripheral lymphoid tissues13, but not that of factor that is composed of proteins belonging to the FOS, factors — namely, IRF8, E4BP4 (E4 promoter-binding CD103+CD11b+ DCs isolated from gut lymphoid tis- JUN and JUN-dimerization protein 4; also known as NFIL3), ID2 and BATF3 (basic sues112. More recently, detailed analyses of Batf3–/– mice protein families. AP1 controls leucine zipper transcription factor, ATF-like 3). have shown that despite the reduction in the frequency various cellular processes, IRF8 is highly expressed in CDPs, pDCs (dis- of CD8α+ DCs, particularly in the spleen, CD8α+ DCs including differentiation, cussed in detail above) and conventional DCs, par- are still present in the absence of BATF3 (REFS 89,113). proliferation and apoptosis. ticularly the CD8α+ and CD103+ DC subsets (FIG. 3). Nevertheless, Batf3–/– mice exhibit severe defects in their Cross-priming IRF8‑deficient mice lack many mature DC subsets, capacity to respond to pathogen infections — includ- A mechanism by which including Langerhans cells90,104. In addition to regulating ing West Nile virus, influenza virus and Toxoplasma immunogenic CD8+ T cells are the generation of pDCs and conventional DC subsets, gondii infections111,114–116 — and an impaired ability to activated by the presentation of an antigen that was not IRF8 controls various functional features of DCs, such mediate cross-priming. This suggests that the main role synthesized by the as the expression of TLR9 and IFNα by pDCs and the of BATF3 may be in regulating the cross-presentation of antigen-presenting cell itself. production of IL‑12 by CD8α+ DCs93. Although IRF8 exogenous antigens to CD8+ T cells (BOX 1). NATURE REVIEWS | IMMUNOLOGY VOLUME 12 | FEBRUARY 2012 | 109 © 2012 Macmillan Publishers Limited. All rights reserved
  • 10. REVIEWS The transcriptional network in CD8α+ and CD103+ IRF2 acts as a transcriptional repressor of genes DCs. As outlined above, a deficiency of IRF8, E4BP4, encoding type I IFNs (IFNα and IFNβ) and thus limits ID2 or BATF3 results in a lack of both the CD8α+ and inflammation120,121. Mice lacking IRF2 exhibit a selective, CD103+ DC subsets. This dependency on the same cell-autonomous loss of CD4+ DCs in the spleen and epi- transcriptional regulators, as well as similar func- dermis, and these subsets are restored when type I IFN tional and localization characteristics13,47,111, suggests signalling is eliminated122,123. Precisely how IRF2 is regu- that CD8α+ and CD103+ DCs represent a single sub- lated during DC development is not clear, but one role of set. However, similar developmental requirements IRF2 may be to protect developing DCs from maturation do not necessarily imply close lineage relationships; arrest when the levels of type I IFNs are elevated. IRF2 for example, PU.1 is required for the development of has also been reported to form complexes with IRF8 multiple distinct lineages, including macrophages, and to act cooperatively with this factor in regulating granulo­ ytes and DCs, but very different mechanisms c the expression of IL12 (REF. 124). are involved66,117. IRF4, a crucial regulator of many aspects of lympho- One approach used to investigate the relative con- cyte differentiation125, is most highly expressed in CD4+ tributions of IRF8, ID2 and BATF3 to conventional DCs (in which IRF8 expression is lowest). In line with DC differentiation has been to engineer a fluorescent this expression profile, CD4+ DCs are largely absent from reporter into the Id2 locus (to generate Id2gfp mice)89. mice lacking IRF4, but these mice also show defects in Analyses of Id2gfp mice showed that neither CDPs nor pDCs in the spleen126,127. Moreover, DCs generated in vitro pre-DCs expressed ID2. Thus, it is more likely that ID2 through the stimulation of bone marrow precursors with drives the terminal differentiation of different DC sub- FLT3L appear to rely on IRF8 rather than IRF4, but those sets rather than influencing early lineage-commitment generated in the presence of GM‑CSF depend on IRF4 decisions. Using Irf8–/– or Batf3–/– mice crossed with (REFS 126,127). Thus, it has been proposed that the main Id2gfp mice, it became clear that IRF8 is required for action of IRF4 is to coordinate signals from GM‑CSF the generation of DC precursors from a very early stimulation through the nuclear factor-κB (NF‑κB) path- time point, whereas BATF3 has a role later in conven- way 128. The extent to which IRF4 and IRF8 regulate simi- tional DC development, downstream of ID2 (REF. 89). lar or distinct sets of target genes, with or without PU.1, BATF3‑deficient progenitors gave rise to precursors of remains an open question that needs to be addressed. SIRPα– DCs (which are the precursors of CD8α+ and RELB is a member of the NF-κB family and can func- CD103+ DCs) in FLT3L‑containing cultures and to a tion either as an activator or as a repressor of transcrip- lesser extent in vivo, although both the frequency of tion by forming heterodimers with the p50 and p52 these DCs and their expression of CD8α were reduced NF‑κB family members. RELB is most highly expressed compared with wild-type CD8α+ DCs89 (FIG. 2). Thus, in the CD8α– and CD11b+ DC subsets, and these DC it is clear that IRF8, ID2 and BATF3 each functions at subsets are absent in RELB-deficient mice129,130. To date, a distinct point in the differentiation of CD103+ and there have been no studies addressing the mechanism CD8α+ DCs. What is lacking, however, is an under- by which RELB controls DC differentiation. It is pos- standing of how the programmes that are activated by sible that IRF4 — the deletion of which also results each of these transcription factors interact to give rise in a substantial loss of CD4+ DCs127 — may be a key to the diversity of DC subsets with unique functions. At target of the RELB pathway. RELB has a crucial role in present, the major approaches for dissecting this net- the upregulation of the signalling molecule CD40, which work are the deletion of individual transcription factor is required to induce immunogenic DCs and for the genes from the entire haematopoietic compartment, induction of IFNα131,132. Loss of IFNα results in impaired and/or limited sampling of tissues and inference with the cross-priming of exogenous antigens132,133. CD40 acts on/off regulatory switches. But these approaches provide through TNF receptor-associated factor 6 (TRAF6) to only ‘black and white snapshots’ of transcription factor activate the NF‑κB cascade134. Strikingly, Traf6–/– mice involvement in DC development. The establishment lack CD8α– DCs and have an impairment in DC matu- of the identity of DC subsets, their differentiation and ration similar to that observed in Relb–/– mice. It seems their maintenance are unlikely to be so simple and may likely that TRAF6 regulates CD4+ DC development in a require combinatorial interactions between transcrip- RELB-dependent manner. tion factors to guide fate decisions118,119. Genome-wide Much attention has been focused on delineating the DNA-binding data will be crucial for the precise eluci- development of CD8α+ and CD103+ DCs. However, dation of the manner in which different transcription despite the crucial role of CD8α – DCs in immune factors work together to define different DC subsets. responses — particularly in the activation of CD4 + T cells18,19 and possibly of follicular helper T cells135–137 Transcriptional regulation of CD8α– DCs. Much less is — a detailed understanding of the transcription factor known about the transcription factors that regulate the networks that drive CD8α– DC development is lacking. differentiation decisions of CD8α– conventional DC lin- Furthermore, transcription factors normally associ- eages, despite the dominant role of these cells in present- ated with CD8α+ and CD103+ DC lineages, such as ID2 ing antigens to CD4+ T cells. The transcription factors (REF. 89) and IRF8, are differentially expressed among IRF2, IRF4 and RELB have been shown to be important CD8α– DCs, suggesting that they may have additional in the development of these subsets and their subsequent roles in the generation of fully matured DCs that do not maturation (FIG. 2; TABLE 2). express CD8α. 110 | FEBRUARY 2012 | VOLUME 12 www.nature.com/reviews/immunol © 2012 Macmillan Publishers Limited. All rights reserved