Dendritic cells (DCs) represent a small and heterogeneous fraction of the hematopoietic system, specialized in antigen capture, processing, and presentation. The different DC subsets act as sentinels throughout the body and perform a key role in the induction of immunogenic as well as tolerogenic immune responses. Because of their limited lifespan, continuous replenishment of DC is required. Whereas the importance of GM-CSF in regulating DC homeostasis has long been underestimated, this cytokine is currently considered a critical factor for DC development under both steady-state and inflammatory conditions. Regulation of cellular actions by GM-CSF depends on the activation of intracellular signaling modules, including JAK/STAT, MAPK, PI3K, and canonical NF-κB. By directing the activity of transcription factors and other cellular effector proteins, these pathways influence differentiation, survival and/or proliferation of uncommitted hematopoietic progenitors, and DC subset–specific precursors, thereby contributing to specific aspects of DC subset development. The specific intracellular events resulting from GM-CSF–induced signaling provide a molecular explanation for GM-CSF–dependent subset distribution as well as clues to the specific characteristics and functions of GM-CSF–differentiated DCs compared with DCs generated by fms-related tyrosine kinase 3 ligand. This knowledge can be used to identify therapeutic targets to improve GM-CSF–dependent DC-based strategies to regulate immunity.

Dendritic cells (DCs) constitute a crucial and heterogeneous fraction of the hematopoietic system, with an essential role in the induction and regulation of immunity.1  Because DCs are relatively short-lived, they are continuously replenished from bone marrow-, blood-, or tissue-derived precursors that are different for the distinct DC subsets.2  The ontogeny of DC subsets belonging to one of the 3 main categories defined elsewhere,2,3  migratory DCs, lymphoid-resident DCs, and plasmacytoid DCs, is shown in Figure 1. Supplemental Tables 1 and 2 (available on the Blood Web site; see the Supplemental Materials link at the top of the online article) show the phenotype and assigned classification of different DC populations that have been identified in distinct tissues in vivo or can be generated through specific culture methods. Efficient DC development from hematopoietic stem cells (HSCs) involves proliferation and survival as well as phenotypic and functional differentiation of progenitors with gradually restricted developmental options.

Figure 1

The role of GM-CSF during DC subset development. DCs are derived from HSCs through gradually restricted precursors. DC subsets can be classified into 3 main categories: migratory DCs, lymphoid-resident DCs, and plasmacytoid DCs. Supplemental Tables 1 and 2 show the phenotype and assigned classification of different DC populations that have been identified in distinct tissues in vivo or can be generated through specific culture methods. GM-CSF supports development of the common DC progenitor (CDP) and of the granulocyte/macrophage progenitor (G/M). Commitment of the CDP toward the plasmacytoid DC lineage is inhibited by GM-CSF, but terminal differentiation of committed plasmacytoid DC precursors (interferon-producing cells [IPCs]) is probably supported by GM-CSF. In contrast to lymphoid-resident DCs, whose development is hardly influenced by GM-CSF, migratory DC development requires GM-CSF. CLP indicates common lymphoid progenitor; linFlt3+, lineageFlt3+ hematopoietic progenitor; CMP, common myeloid progenitor; MDP, macrophage and DC progenitor; CDP, common DC progenitor; pre-DC, precursor DC; mono, monocytes; pDC, plasmacytoid DC; cDC, conventional DC; intDC, interstitial DC; LC, Langerhans cell; and TIP-DC, TNF-α and inducible nitric-oxide synthase-producing DC.

Figure 1

The role of GM-CSF during DC subset development. DCs are derived from HSCs through gradually restricted precursors. DC subsets can be classified into 3 main categories: migratory DCs, lymphoid-resident DCs, and plasmacytoid DCs. Supplemental Tables 1 and 2 show the phenotype and assigned classification of different DC populations that have been identified in distinct tissues in vivo or can be generated through specific culture methods. GM-CSF supports development of the common DC progenitor (CDP) and of the granulocyte/macrophage progenitor (G/M). Commitment of the CDP toward the plasmacytoid DC lineage is inhibited by GM-CSF, but terminal differentiation of committed plasmacytoid DC precursors (interferon-producing cells [IPCs]) is probably supported by GM-CSF. In contrast to lymphoid-resident DCs, whose development is hardly influenced by GM-CSF, migratory DC development requires GM-CSF. CLP indicates common lymphoid progenitor; linFlt3+, lineageFlt3+ hematopoietic progenitor; CMP, common myeloid progenitor; MDP, macrophage and DC progenitor; CDP, common DC progenitor; pre-DC, precursor DC; mono, monocytes; pDC, plasmacytoid DC; cDC, conventional DC; intDC, interstitial DC; LC, Langerhans cell; and TIP-DC, TNF-α and inducible nitric-oxide synthase-producing DC.

Close modal

GM-CSF was the first cytokine shown to efficiently promote DC development in vitro4  and has been used to induce DC differentiation from human monocytes5  as well as human and mouse hematopoietic progenitor cells.6,8  Significantly increased DC numbers have been found in the spleen and thymus of mice injected with GM-CSF or transgenic mice overexpressing GM-CSF,9,10  suggesting that GM-CSF can promote DC expansion in vivo. However, only a marginal reduction in lymphoid organ DCs was found in mice lacking GM-CSF or the GM-CSF receptor (GM-CSFR).10  In later studies, mice lacking GM-CSF or the GM-CSFR were shown to have substantially reduced numbers of migratory DCs in skin and gut,11,13  indicating that development of these subsets requires GM-CSF under steady-state conditions. The 3-fold decrease in lymph node DCs observed earlier10  probably reflects the loss of these migratory DCs, without alterations in the lymphoid-resident DC subset. Besides subset-specific GM-CSF dependency, GM-CSF can also negatively affect the development of specific DC subsets, as demonstrated for plasmacytoid DCs14  (Figure 1). Interestingly, although GM-CSF inhibits commitment of bone marrow progenitors to the plasmacytoid DC lineage,14  it supports the survival and can even initiate terminal differentiation of circulating interferon-producing cells, the direct precursors of plasmacytoid DCs.15  Because the secretion of GM-CSF, which circulates at very low concentrations under steady-state conditions, is increased during infection and inflammation,16  its influence on the composition of the DC pool becomes more pronounced during inflammation. As an illustration, inflammation-driven monocyte conversion to murine splenic DCs3,17  and DCs generated during acute inflammatory arthritis or antigen-induced peritonitis18  was reported to depend on GM-CSF. Furthermore, besides skewing the development of specific DC subsets, GM-CSF induces development of DCs with a relatively immunogenic phenotype and functionality, which resemble TNF-α and inducible nitric-oxide synthase-producing DCs19  that develop during infection.3 

Thus, the role of GM-CSF in DC development appears situation- as well as subset-specific. Under steady-state conditions, GM-CSF supports migratory DC development, whereas its effect on other subtypes is either redundant or even detrimental (Figure 1). Increased GM-CSF production during the onset of inflammation contributes to rapid differentiation of precursor DCs (pre-DCs) but also unconventional DC precursors, such as monocytes. This influences the relative contributions of certain DC subsets as well as the immunogenicity of the DCs generated. These functions are very different from the actions of fms-related tyrosine kinase 3 ligand (Flt3L), a cytokine that supports the development of most DC subsets, with a relatively tolerogenic functionality.2,20  Although in the past Flt3L-induced DC development under steady-state conditions has attracted more attention than GM-CSF–induced development induced by inflammation, in the light of the continuous exposure to pathogens, inflammation-induced DC development appears highly relevant. Taken together, GM-CSF appears a significant factor in the maintenance of DC homeostasis under both steady-state and inflammatory conditions.

The GM-CSFR contains 2 distinct subunits, the GM-CSF–specific α-chain (GM-CSFRα; CD116) and the common β-receptor (βc; CD131), which is shared between the GM-CSFR, the IL-3 receptor, and the IL-5 receptor.21,22  Downstream signaling cascades are primarily induced through interaction of effector proteins with the βc subunit. Signaling is initiated by the cytoplasmic tyrosine kinase janus kinase 2 (JAK2), which then acts on various downstream proteins (Figures 2 and 3).21,,24  The principle signaling modules activated include the janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway, the mitogen-activated protein kinase (MAPK) pathway, and the phosphatidylinositol 3-kinase (PI3K) pathway (Figures 2 and 3). In addition, canonical NF-κB transcription factors are activated. Although NF-κB activation could occur secondary to the activation of signaling proteins, such as STAT5, PI3K, or PKB, or depend on GM-CSF–induced secretion of other factors initiating canonical NF-κB activation, activation independent of such mechanisms has been reported. βc-bound TRAF6 causes NF-κB translocation through induction of IκBα degradation25  and direct interaction between IκB kinase (IKK) and GM-CSFRα resulting in IκBα degradation has been described,26  demonstrating that canonical NF-κB activation can be a direct consequence of GM-CSFR engagement (Figures 2 and 3). To elucidate the role of these signaling cascades in DC development, these pathways have been manipulated in DC progenitors or precursors and their DC generating ability has been evaluated.

Figure 2

The GM-CSF receptor: initiation of signal transduction. In the absence of GM-CSF, preformed βc dimers as well as single GM-CSFRα chains are present on the plasma membrane. Low affinity binding of GM-CSF to GM-CSFRα causes association with a βc dimer. This leads to the formation of a hexamer of 2 GM-CSF molecules, 2 GM-CSFRα chains, and 2 βc chains, and induces high-affinity GM-CSF/GM-CSFRα binding. Two of these hexamers then dimerize. This brings the βc subunits in close enough proximity to enable JAK2 transactivation, which initiates downstream signaling. Activated JAK2 phosphorylates several tyrosine domains on βc (Y577, Y612, Y695, Y750, Y806, and Y866), which subsequently serve as docking sites for a variety of proteins. Phosphorylation of recruited STATs results in their activation and serves as start of signaling. Activation of the MAPK pathway is initiated by recruitment of SHC to Y577. Its subsequent phosphorylation allows interaction with GRB2 and mSOS, after which the activation of RAS is catalyzed. Recruitment and activation of PI3K has also been suggested to depend on βc residue phosphorylation, and its activation is promoted by JAK2-mediated phosphorylation of PI3K. Finally, GM-CSF activates canonical NF-κB transcription factors. Activation of the IKK complex as a direct consequence of GM-CSFR engagement has been reported, but the proteins involved and the complete chain of events remain to be elucidated.21,,,,26 

Figure 2

The GM-CSF receptor: initiation of signal transduction. In the absence of GM-CSF, preformed βc dimers as well as single GM-CSFRα chains are present on the plasma membrane. Low affinity binding of GM-CSF to GM-CSFRα causes association with a βc dimer. This leads to the formation of a hexamer of 2 GM-CSF molecules, 2 GM-CSFRα chains, and 2 βc chains, and induces high-affinity GM-CSF/GM-CSFRα binding. Two of these hexamers then dimerize. This brings the βc subunits in close enough proximity to enable JAK2 transactivation, which initiates downstream signaling. Activated JAK2 phosphorylates several tyrosine domains on βc (Y577, Y612, Y695, Y750, Y806, and Y866), which subsequently serve as docking sites for a variety of proteins. Phosphorylation of recruited STATs results in their activation and serves as start of signaling. Activation of the MAPK pathway is initiated by recruitment of SHC to Y577. Its subsequent phosphorylation allows interaction with GRB2 and mSOS, after which the activation of RAS is catalyzed. Recruitment and activation of PI3K has also been suggested to depend on βc residue phosphorylation, and its activation is promoted by JAK2-mediated phosphorylation of PI3K. Finally, GM-CSF activates canonical NF-κB transcription factors. Activation of the IKK complex as a direct consequence of GM-CSFR engagement has been reported, but the proteins involved and the complete chain of events remain to be elucidated.21,,,,26 

Close modal
Figure 3

GM-CSF–activated signaling modules. PI3K93,95 : GM-CSF induces activation of the class IA PI3Ks, which consist of 2 subunits: a catalytic subunit, p110, and a regulatory subunit, p85. Activity of PI3K is promoted by JAK2-mediated phosphorylation of p85. On activation, PI3K functions mainly through the generation of PtdIns(3,4,5)P3 (PIP3), an activity counteracted by phosphatases PTEN and SHIP. PIP3 acts as a second messenger, regulating a large variety of downstream targets, including protein kinase B (PKB; also called c-AKT). By recruiting PKB to the plasma membrane, PIP3 enables its activation through phosphorylation by PDK1 and mTORC2. Activated PKB regulates many targets, including the FOXO transcription factors, the TSC1/TSC2 complex, and the mTOR complex 1 (mTORC1). Like FOXO, mTORC1 acts through the regulation of transcription, but its main function is in the regulation of protein translation. JAK/STAT90 : STAT proteins as well as Src kinases are recruited to βc by their SH2 domains that interact with phosphorylated Y612, Y695, and Y750. The STATs are primarily phosphorylated by JAK2, but kinase activity of the Src kinases has also been reported. STAT phosphorylation at a conserved tyrosine residue alters their conformation, which allows the formation of homodimers or heterodimers with DNA-binding and transcription-regulating ability. These dimers then translocate to the nucleus where they act as functional transcription factors. MAPK59 : Although the human MAPK family includes at least 11 members subdivided into 6 groups, the principle MAPK pathway activated by the GM-CSF receptor is the MEK/ERK pathway. Recruitment of mSOS to the SHC/GRB2 complex enables mSOS to catalyze RAS activation. Formation of active GTP-bound RAS from inactive GDP-bound RAS leads to the successive activation of RAF, MEK, and ERK. On activation, ERK expresses kinase activity toward a variety of cytoplasmic molecules and nuclear proteins, which in turn regulate gene expression. NF-κB127 : In resting cells, canonical NF-κB dimers consisting of NF-κB/Rel family members RelA, c-Rel, p50, and/or p52 are retained in the cytoplasm by binding to inhibitor of NF-κB proteins (IκBs). Activation is achieved through the IKK complex, which phosphorylates IκB proteins. These are subsequently ubiquitinated and finally degraded, enabling nuclear translocation of canonical NF-κB dimers.

Figure 3

GM-CSF–activated signaling modules. PI3K93,95 : GM-CSF induces activation of the class IA PI3Ks, which consist of 2 subunits: a catalytic subunit, p110, and a regulatory subunit, p85. Activity of PI3K is promoted by JAK2-mediated phosphorylation of p85. On activation, PI3K functions mainly through the generation of PtdIns(3,4,5)P3 (PIP3), an activity counteracted by phosphatases PTEN and SHIP. PIP3 acts as a second messenger, regulating a large variety of downstream targets, including protein kinase B (PKB; also called c-AKT). By recruiting PKB to the plasma membrane, PIP3 enables its activation through phosphorylation by PDK1 and mTORC2. Activated PKB regulates many targets, including the FOXO transcription factors, the TSC1/TSC2 complex, and the mTOR complex 1 (mTORC1). Like FOXO, mTORC1 acts through the regulation of transcription, but its main function is in the regulation of protein translation. JAK/STAT90 : STAT proteins as well as Src kinases are recruited to βc by their SH2 domains that interact with phosphorylated Y612, Y695, and Y750. The STATs are primarily phosphorylated by JAK2, but kinase activity of the Src kinases has also been reported. STAT phosphorylation at a conserved tyrosine residue alters their conformation, which allows the formation of homodimers or heterodimers with DNA-binding and transcription-regulating ability. These dimers then translocate to the nucleus where they act as functional transcription factors. MAPK59 : Although the human MAPK family includes at least 11 members subdivided into 6 groups, the principle MAPK pathway activated by the GM-CSF receptor is the MEK/ERK pathway. Recruitment of mSOS to the SHC/GRB2 complex enables mSOS to catalyze RAS activation. Formation of active GTP-bound RAS from inactive GDP-bound RAS leads to the successive activation of RAF, MEK, and ERK. On activation, ERK expresses kinase activity toward a variety of cytoplasmic molecules and nuclear proteins, which in turn regulate gene expression. NF-κB127 : In resting cells, canonical NF-κB dimers consisting of NF-κB/Rel family members RelA, c-Rel, p50, and/or p52 are retained in the cytoplasm by binding to inhibitor of NF-κB proteins (IκBs). Activation is achieved through the IKK complex, which phosphorylates IκB proteins. These are subsequently ubiquitinated and finally degraded, enabling nuclear translocation of canonical NF-κB dimers.

Close modal

JAK/STAT

Conditional JAK2 deletion in adult mice significantly reduced their splenic DC numbers.27  In addition, murine JAK2−/− bone marrow progenitors generated decreased numbers of conventional DCs after culture in the presence of GM-CSF.27  Despite their status as direct JAK substrates, studies investigating the function of STAT proteins for DC development are relatively scarce. In DCs and their progenitors, GM-CSF–induced activation of STAT1, STAT3, STAT5, and STAT6 has been proposed, but only the roles of STAT3 and STAT5 have been further evaluated.28,,,32  STAT3 has proven to be both required and instructive in Flt3L-induced murine DC development but appeared dispensable in GM-CSF–driven DC differentiation cultures.30,33  With this in mind, the normal DC numbers found in the blood of hyper IgE-syndrome patients carrying STAT3 mutations34  could either result from residual STAT3 activity or from compensation by other STATs. For example, in GM-CSF–induced murine bone marrow-derived DC differentiation cultures, STAT5 has been shown to have the capacity to compensate for loss of STAT3 activity.28,30  Although STAT3 may also be able to compensate for STAT5 deficiency,29  STAT5 and not STAT3 has a key function in the induction of DC development driven by GM-CSF.

STAT5 has been suggested to support the differentiation of human migratory DCs and murine bone marrow–derived conventional DCs.28,29,32,35,36  In a recent study, we showed that GM-CSF–induced interstitial DC differentiation from human CD34+ hematopoietic progenitor cells (HPCs) requires STAT5 activity.32  However, inhibition of STAT5 promotes the development of CD34-derived Langerhans cells,32  demonstrating subset-specific regulation within the migratory lineage. Furthermore, ectopic STAT5 activation reduced the in vitro development of murine conventional DCs37  and abrogated commitment of human CD34+ HPCs to both the interstitial DCs and the Langerhans cell linage.32  In contrast, elevated STAT5 activity levels promoted terminal differentiation of already committed human CD34-derived pre-interstitial DCs and pre-Langerhans cells.32  These data show that the levels and timing of STAT5 activation need to be carefully regulated to ensure DC development. Whereas the activity levels induced by GM-CSF allow the development of migratory and conventional DCs, GM-CSF–induced STAT5 activity inhibits commitment of murine bone marrow progenitors to the plasmacytoid DC lineage.14,28,29  In contrast, GM-CSF treatment of interferon-producing cells promotes, rather than restrains, terminal differentiation,15  again indicating the importance of timing. Thus, STAT5 does not act in a simple on-off binary manner and the required STAT5 activity levels differ between DC subsets and differentiation stages.

PI3K/PKB

Spleens of mice lacking the p85α subunit of class IA PI3K contain DCs, but the numbers have not been compared with control mice.38  PI3K generates PtdIns(3,4,5)P3, which acts as a second messenger to induce PI3K-dependent signaling (Figure 3). The PtdIns(3,4,5)P3 phosphatases PTEN and SHIP prevent PI3K-induced activation of downstream signaling. Increased numbers of CD8α+ DCs were observed in spleens of irradiated mice reconstituted with PTEN−/− bone marrow progenitors, and spleens of mice with a DC-specific PTEN deletion also showed an amplified CD8α+ DC population.39  Together with the increased CD11c+ splenic DC numbers described for SHIP−/− mice,40  these data indicate that PI3K activity supports DC development. A major target of PI3K-dependent signaling is protein kinase B (PKB/c-AKT), a protein whose activation involves phosphorylation by PDK-1. DC populations of mice expressing approximately 10% of normal PDK-1 were unaffected,41  suggesting that either PKB is not required for DC development or the residual PDK1 activity maintained PKB activity at sufficient levels. Bone marrow of β2-microglobulin−/−NOD/SCID mice transplanted with human CD34+ HPCs expressing a constitutively active PKB mutant contain increased human BDCA-1+ DCs compared with mice transplanted with control HPCs,42  indicating a supportive role for PKB in DC development. Although the PI3K/PKB axis has many downstream effectors, impaired generation of DC after in vivo administration of rapamycin, a pharmacologic inhibitor of mammalian target of rapamycin (mTOR), to mice,43  indicates that activity of PKB target mTOR complex 1 (mTORC1) is required. Although it has been suggested that the unaffected CD11c+ DC numbers in the blood of kidney transplant patients treated with rapamycin triple therapy indicate mTOR-independent regulation of human blood DC, this study lacks the proper control group.44 

In vivo experiments clearly demonstrate the importance of PI3K, PKB, and mTORC1 activity in DC homeostasis, but in vitro experiments were required to specifically establish the role of PI3K-PKB-mTOR signaling during GM-CSF–induced DC development. Pharmacologic inhibition of PI3K or mTOR inhibits in vitro GM-CSF–driven human DC development from monocytes44,,,48  and hematopoietic progenitors42,46  because of reduced proliferation and survival of DC precursors. Increased signaling improves survival of DCs and their precursors, and these cells can even be rescued from GM-CSF deprivation-induced apoptosis by ectopic activation of this pathway.42,49,50  Although the survival of most DC lineages appears to be supported by PI3K-dependent signaling, important differences between distinct subsets and differentiation phases have been observed. For instance, human CD34-derived pre-interstitial DCs and pre-Langerhans cells require PI3K activity to survive, but the survival of terminally differentiated DCs of these lineages is independent of PI3K or mTOR activity.42,46  Conversely, whereas monocyte survival was found to be resistant to inhibition of PI3K or mTOR, the survival of human monocyte-derived DCs requires activity of this pathway.46,48  Contradictory experiments were shown in a recent study that reported apoptosis of monocytes 2 days after rapamycin administration.44  These discrepancies may further emphasize the very specific requirements of distinct subsets at specific differentiation stages.

Although DC numbers are strongly reduced in cultures where PI3K or mTOR is inhibited, the cells show a normal DC phenotype in most studies. However, despite their DC phenotype, these cells are functionally impaired,42,44,51,52  indicating that the acquisition of full DC function requires activation of this pathway. Besides mTORC1, other PKB-regulated effectors may be involved in DC differentiation. Activity of glycogen synthase kinase-3β (GSK-3β), which is negatively regulated by PKB-dependent phosphorylation, appears required to avoid human monocyte-to-macrophage differentiation in monocyte-derived DC differentiation cultures.53  However, GM-CSF–induced murine bone marrow-derived conventional DC differentiation was found to be independent of GSK-3β.54  Although PKB activity levels may affect phenotypic differentiation of some DC subsets, the greatest significance of the PI3K/PKB pathway lies in functional differentiation and promoting DC numbers by ensuring sufficient expansion and survival.

MAPK

Few studies have critically evaluated the role of MAPK signaling during DC development. Although there are several MAPK family members regulating distinct signaling events, the MEK/ERK pathway is the only one directly activated by the GM-CSFR (Figures 2 and 3). In a recent study, conventional DCs derived in vitro from murine ERK1−/− bone marrow progenitors were found to show increased surface expression of activation markers and enhanced T-cell stimulation,55  suggesting that ERK1 negatively influences functional differentiation. Effects on DC yields or typical DC markers were not detailed. Although these data could indicate that ERK is not required for DC development, compensation by the ERK2 isoform provides an alternative explanation. Pharmacologic inhibition of MEK or ERK abrogates both differentiation and survival during human monocyte-derived DC development.47,48  Although further direct evidence is limited, an association between loss of MEK/ERK activity and reduced DC development has been reported by several groups.56,58  Interestingly, activation of p38 MAPK59  by stimuli, such as tumor-secreted factors or Toll-like receptor triggering, can impair GM-CSF–driven DC differentiation, whereas differentiation is improved by pharmacologic inhibition of p38 MAPK.47,60,61  Thus, GM-CSF can activate the MEK/ERK signaling module to promote DC development, whereas other factors may use p38 MAPK to modulate this. However, the significance of MAPK signaling remains poorly defined, and additional research is required. Considering the key function MAPK proteins have in regulating survival, proliferation, and differentiation of a large variety of cell types, an important role of this pathway in DC development is to be expected.

NF-κB

Individual knockout of the canonical NF-κB proteins RelA, c-Rel, or p50 had no effect on DC populations present in mouse spleen, but spleens of mice with a combined RelA and p50 deficiency contained reduced CD11c+ DCs.62  Moreover, RelA−/−p50−/− hematopoietic progenitors were unable to generate DCs after adoptive transfer, and their DC differentiation ability in GM-CSF–driven cultures was strongly impaired.62  A requirement for canonical NF-κB transcription factor activity has also been demonstrated for human migratory DC differentiation from monocytes or CD34+ hematopoietic progenitors in GM-CSF–supplemented cultures.63,65  Besides supporting the conclusion that canonical NF-κB is required for mouse and human GM-CSF–driven DC development, these in vitro studies allowed separate evaluation of the different aspects of DC development.

Pharmacologic- or viral transduction-mediated NF-κB inhibition strongly reduces cell yields in GM-CSF–driven DC differentiation cultures from human monocytes64  and human or mouse hematopoietic progenitors.62,64  The survival of both DC precursors63,64  and differentiated DCs64,65  was shown to depend on intact canonical NF-κB activity. Cells derived from GM-CSF–supplemented murine RelA−/−p50−/− bone marrow progenitor cultures had a typical conventional DC morphology,62  suggesting that differentiation was unaffected. However, canonical NF-κB inhibition in human monocytes and hematopoietic progenitors impaired their acquisition of a DC phenotype,64,66  indicating that at least for some subsets canonical NF-κB activity is required for differentiation. In particular, human monocyte-derived and CD34-derived interstitial DC differentiation was inhibited in the presence of pharmacologic NF-κB inhibitors,64  whereas CD34-derived pre-Langerhans cells developed despite NF-κB inhibition.64  However, terminal differentiation of pre-Langerhans cells was hindered by introduction of an IκBα super-repressor.66 

GM-CSF–induced canonical NF-κB activation thus appears crucial to ensure differentiation and survival of DC precursors. Interestingly, despite their incomplete DC phenotype, antigen uptake, activation-induced costimulatory molecule expression, and allogeneic T-cell stimulation by human monocyte-derived DCs generated in the presence of NF-κB inhibitors was comparable with control monocyte-derived DCs.64  However, their cytokine production abilities might be affected.64  Similarly, conventional DCs generated in vitro from p50−/−c-Rel−/− murine bone marrow progenitors had a reduced ability to produce IL-12.62  Continued NF-κB inhibition after differentiation could be responsible for impaired functionality because DC activation induced by pathogenic stimulation, T-cell signals, or inflammatory cytokines other than GM-CSF is strongly dependent on canonical NF-κB activity.67  Although more detailed investigation is required, it is probable that the activation of canonical NF-κB transcription factors during DC differentiation affects the functionality of the cells generated.

GM-CSF regulates DC development through an integrated molecular network

Manipulation of the activity of central signaling proteins and the evaluation of the consequences for DC development has led to the identification of specific functions for the various GM-CSF–activated signaling pathways described above (Figure 4). However, although in these studies the distinct signaling cascades are often regarded as separate entities, this is mostly not the case because the different pathways share signaling proteins, upstream initiators of signaling, and proteins regulating negative feedback (Table 1). Therefore, manipulation of one signaling module will probably also affect others, meaning that observed effects on DC development may be accounted for by other pathways than the canonical pathway to which the targeted protein belongs. This realization stresses the need to carefully evaluate the available data. The difficulty this causes in the interpretation of some studies is highlighted by the finding that, although PI3K activates NF-κB (Table 1), canonical NF-κB but not PI3K regulates phenotypic human monocyte-derived DCs and CD34-derived interstitial DC differentiation42,46,64  (Figure 4). This discrepancy is probably explained by PI3K-independent NF-κB activation. However, it seems prudent to keep interpathway regulation in mind when trying to assemble a reliable picture of the molecular aspects of GM-CSF–driven DC development.

Figure 4

GM-CSF regulates DC development through an integrated molecular network. The signaling modules activated by GM-CSF from an integrated network with overlapping and separate functions that enable an adequate response to a wide range of situations. The currently known functions of the specific cascades in the regulation of GM-CSF–induced DC development are shown. Of the signaling proteins contributing to GM-CSF–driven DC development, JAK2-activated STAT5 is the clearest regulator of differentiation. The main role of the PI3K-PKB signaling module is in promoting expansion and survival of DC precursors rather than their differentiation, although its activity is also required to generate DCs with full functionality. Finally, activation of MEK/ERK and canonical NF-κB transcription factors is associated with differentiation and survival.

Figure 4

GM-CSF regulates DC development through an integrated molecular network. The signaling modules activated by GM-CSF from an integrated network with overlapping and separate functions that enable an adequate response to a wide range of situations. The currently known functions of the specific cascades in the regulation of GM-CSF–induced DC development are shown. Of the signaling proteins contributing to GM-CSF–driven DC development, JAK2-activated STAT5 is the clearest regulator of differentiation. The main role of the PI3K-PKB signaling module is in promoting expansion and survival of DC precursors rather than their differentiation, although its activity is also required to generate DCs with full functionality. Finally, activation of MEK/ERK and canonical NF-κB transcription factors is associated with differentiation and survival.

Close modal
Table 1

Interdependency of distinct signaling pathways

ProteinSubstrateAction
JAK2 STATs Signaling initiation 
 MAPK Signaling initiation 
 PI3K Signaling initiation 
 IκB Degradation (phosphorylation) 
PDK1 PKB Activation (phosphorylation) 
 IKKβ Activation (phosphorylation) 
RAS RAF Activation (phosphorylation) 
 PI3K Activation 
PP2A IKK Inhibition (negative feedback) 
 PKB Inhibition (negative feedback) 
SOCS1 STAT5 Inhibition (negative feedback) 
 NF-κB Inhibition (negative feedback) 
PIAS1 JAK2 Inhibition (negative feedback) 
 RelA Inhibition DNA binding 
PI3K PKB Activation (indirect) 
 NF-κB Activation (phosphorylation) 
PKB mTORC1 Activation (phosphorylation and indirect) 
 TSC2 Inhibition (phosphorylation) 
 RelA Enhanced activity (phosphorylation) 
 IKKβ Activation (indirect) 
p38 MAPK NF-κB Regulation DNA binding 
ERK TSC2 Inhibition (phosphorylation) 
NF-κB SOCS1 Increased production 
STAT5 NF-κB Activation (phosphorylation) 
IKKβ IκBα Degradation (phosphorylation) 
 TSC1 Inhibition (phosphorylation) 
ProteinSubstrateAction
JAK2 STATs Signaling initiation 
 MAPK Signaling initiation 
 PI3K Signaling initiation 
 IκB Degradation (phosphorylation) 
PDK1 PKB Activation (phosphorylation) 
 IKKβ Activation (phosphorylation) 
RAS RAF Activation (phosphorylation) 
 PI3K Activation 
PP2A IKK Inhibition (negative feedback) 
 PKB Inhibition (negative feedback) 
SOCS1 STAT5 Inhibition (negative feedback) 
 NF-κB Inhibition (negative feedback) 
PIAS1 JAK2 Inhibition (negative feedback) 
 RelA Inhibition DNA binding 
PI3K PKB Activation (indirect) 
 NF-κB Activation (phosphorylation) 
PKB mTORC1 Activation (phosphorylation and indirect) 
 TSC2 Inhibition (phosphorylation) 
 RelA Enhanced activity (phosphorylation) 
 IKKβ Activation (indirect) 
p38 MAPK NF-κB Regulation DNA binding 
ERK TSC2 Inhibition (phosphorylation) 
NF-κB SOCS1 Increased production 
STAT5 NF-κB Activation (phosphorylation) 
IKKβ IκBα Degradation (phosphorylation) 
 TSC1 Inhibition (phosphorylation) 

Although the various signaling cascades regulated by the GM-CSF receptor are often regarded as separate entities, they are actually interrelated, sharing proteins regulating their activation or inhibition, or even directly influencing each other. Proteins involved in more than 1 signaling cascade are listed, together with the proteins they regulate and the actions they express toward them. If 1 of the regulated proteins is regarded as a principle target, this protein is underlined.

Besides processes specifically regulated by distinct signaling cascades, other processes, such as DC survival, are regulated by several GM-CSF–activated signaling modules (Figure 4). This broad regulation is helpful in ensuring DC survival under diverse circumstances, for example, when GM-CSF–induced survival signaling through a specific pathway is hindered by additional environmental stimuli. Furthermore, in a recent model of GM-CSF–induced hematopoiesis, low GM-CSF concentrations were suggested to only activate PI3K, and concentrations of more than or equal to 10pM were required to activate MAPK and JAK/STAT signaling and to induce phosphorylation of PKB.24  Because survival could be mediated through PI3K activated by low GM-CSF concentrations, hematopoietic cell survival is ensured under all circumstances, whereas additional PKB activation and MAPK and JAK/STAT signal transduction seem required to induce proliferation.24  Although the relevance of this finding for GM-CSF–induced DC development requires further evaluation, it is interesting to note that the relative contribution of the various modules may not be equal under all circumstances. Thus, DC development in response to GM-CSF is regulated through an integrated network of signaling pathways that act separately but also influence each other's activity. The combination of overlapping and separate functions of the distinct signaling proteins enables an appropriate response to a wide range of situations.

Finally, although GM-CSF is often associated with DC development, GM-CSF–activated signaling events could support the differentiation of other hematopoietic lineages. The GM-CSFR is expressed at high levels by (developing) macrophages and granulocytes in addition to DCs21 ; and, for example, GM-CSF–induced PI3K/PKB-dependent survival or progenitor expansion could support the development of these lineages. Furthermore, transcription factors, such as STAT5, promote differentiation of other myeloid cells besides DCs, such as granulocytes and erythrocytes.68  However, the principle cytokines inducing STAT5 activation in these lineages include G-CSF, IL-5, and erythropoietin rather than GM-CSF, and to induce differentiation of these lineages additional actions of these cytokines are required. Together, the actions of the complete GM-CSF–induced signal transduction network support the generation of DC.

Regulation of DC differentiation and subset distribution

Cell fate decisions in the hematopoietic system are initiated by cytokine-induced signal transduction and established by the resultant actions of transcription factors that regulate lineage commitment and differentiation of hematopoietic progenitors. DC differentiation is driven by a specific set of regulatory transcription factors, as reviewed elsewhere,2,69  but how the intracellular signaling pathways regulating DC development control this transcriptional program remains ill-defined. GM-CSF–activated canonical NF-κB and STAT5 transcription factors directly regulate genes involved in DC differentiation (Figure 5). For example, NF-κB p50 induces transcription of C/EBPα.70  Although this transcription factor may promote the development of interstitial DCs through potentiating the commitment of multipotent hematopoietic progenitors to the granulocyte/macrophage lineage, C/EBPα activity inhibits the development of Langerhans cells.71,72  The promoter of IRF4, a gene involved in subset-specific DC development, also contains several putative NF-κB binding sites.73  RelA, c-Rel, and p50 bind IRF4 promoter elements during human monocyte-derived DC differentiation.73  For murine DCs, IRF4 expression was shown to be required for GM-CSF–driven conventional DC differentiation in vitro, whereas spleens of IRF4−/− mice lack CD8α DCs, but CD8α+ DCs are unaffected and numbers of plasmacytoid DCs are only modestly reduced.74,75  Another subset-specific regulator is RelB. Activation of this noncanonical member of the NF-κB family is induced independently from the canonical NF-κB signaling pathway, but interaction between canonical and noncanonical NF-κB proteins has been described.76,78  Similar to IRF4, RelB is crucial in the differentiation of CD8α splenic DCs,79  and within the human immune system RelB has been shown to be required for interstitial DCs but not Langerhans cell differentiation.66  Besides affecting lineage decisions, canonical NF-κB–mediated regulation of RelB may contribute to the development of DCs with a more activated phenotype because this protein has also been associated with DC immunogenicity.80  Finally, NF-κB is known as a major inducer of inflammatory cytokine and costimulatory molecule expression in terminally differentiated DCs67  and its activation by GM-CSF may therefore affect the functions of GM-CSF–differentiated DCs.

Figure 5

GM-CSF–induced regulation of the DC transcriptional program mediated through STAT5 and canonical NF-κB. STAT5 and canonical NF-κB transcription factors can directly regulate the DC transcriptional program. Regulation of RelB, IRF4, and PU.1 expression not only promotes DC differentiation in general but may also influence subset distribution among the DCs generated. The role of C/EBPα is also subset-specific, promoting development of interstitial DCs through potentiating the commitment of multipotent hematopoietic progenitors to the granulocyte/macrophage lineage, but inhibiting Langerhans cell development. The inhibitory actions of STAT5 toward IRF8 and SpiB explain the negative impact GM-CSF has on the plasmacytoid DC lineage, whereas effects on genes involved in DC functionality may promote the immunogenicity of DC differentiated with GM-CSF.

Figure 5

GM-CSF–induced regulation of the DC transcriptional program mediated through STAT5 and canonical NF-κB. STAT5 and canonical NF-κB transcription factors can directly regulate the DC transcriptional program. Regulation of RelB, IRF4, and PU.1 expression not only promotes DC differentiation in general but may also influence subset distribution among the DCs generated. The role of C/EBPα is also subset-specific, promoting development of interstitial DCs through potentiating the commitment of multipotent hematopoietic progenitors to the granulocyte/macrophage lineage, but inhibiting Langerhans cell development. The inhibitory actions of STAT5 toward IRF8 and SpiB explain the negative impact GM-CSF has on the plasmacytoid DC lineage, whereas effects on genes involved in DC functionality may promote the immunogenicity of DC differentiated with GM-CSF.

Close modal

In addition to NF-κB, STAT5 has been suggested to influence subset-specific DC development through induction of IRF4 and RelB mRNA expression, although this may occur indirectly.29,36  More direct may be its negative effect on PU.1 and C/EBPα.32,81,82  For these proteins, expression levels have been suggested to affect hematopoietic lineage choices,71,83,84  and STAT5-mediated down-regulation of PU.1 and C/EBPα32,81,82  may be involved in maintaining the correct expression levels submissive for DC development. Furthermore, regulation of expression levels could affect DC subset decisions. In an in vitro system simultaneously generating human Langerhans cells and interstitial DC from CD34+ hematopoietic progenitors, Langerhans cell development is favored by inhibiting C/EBPα activity or enhancing PU.1 expression levels, at the cost of interstitial DC development.71,85  In addition, murine CD8α but not CD8α+ splenic DCs are dependent on PU.1 for their development.86  Other mechanisms by which STAT5 may regulate subset distribution include the inhibition of IRF8 and SpiB mRNA expression.29  These transcription factors are mainly associated with plasmayctoid DC development,2,69  and the inhibitory actions of STAT5 toward these genes could explain the negative impact of GM-CSF on the plasmacytoid DC lineage. Indeed, spleens of IRF8 null mice show specific reductions in plasmacytoid DCs and CD8α+ DCs.75  However, human subjects with IRF8 mutations resulting in strongly reduced IRF8 activity show general loss of DCs in peripheral blood,87  implicating a broader effect of IRF8 on human DC development. In addition to regulating subset distribution, STAT5 promotes expression of MHC class II transactivator protein (CIITA),35  which is essential for transcriptional activity of the MHC class II promoter. Because STAT5 also stimulates DNA binding and transcriptional activity of NF-κB,88  GM-CSF–induced STAT5 activation may increase the intrinsic immunogenicity of DCs generated in the presence of GM-CSF.

The PI3K/PKB and the MEK/ERK module have both been demonstrated to influence NF-κB and STAT5 transcriptional activity,89,90  but NF-κB/STAT5–independent effects on DC regulatory transcription factors also exist. PKB could affect DC differentiation through inhibition of GSK-3β, which directly inhibits C/EBPα activity.91  In addition, direct inhibitory effects of ERK on MHC class II transactivator protein expression have been proposed.92  In conclusion, GM-CSF–activated JAK2/STAT5 and canonical NF-κB are more directly involved in regulating the DC transcriptional program than PI3K/PKB and MEK/ERK (Figure 5). Through regulation of RelB, IRF4, PU.1, C/EBPα, IRF8, and SpiB, NF-κB and STAT5 not only promote DC differentiation in general but may also influence DC subset distribution. Moreover, by affecting genes involved in DC functionality, STAT5 and NF-κB may increase the immunogenicity of GM-CSF–differentiated DCs.

Downstream effectors regulating proliferation and survival

The only GM-CSF–activated signaling pathway that has been explicitly connected to DC precursor expansion is the PI3K/PKB module (Figure 4).42  This module generally directs proliferation through transcriptional and translational control,93,95  but the specific mechanisms used during GM-CSF–induced DC development remain undefined. Although direct evidence is currently lacking, it also appears highly likely that MEK/ERK and JAK2/STAT5 are involved in DC precursor expansion, considering their important role in other hematopoietic lineages.59,68,90  Knowledge on the molecular regulation of DC survival is little more available. The IKK complex inhibits proapoptotic Forkhead box O (FOXO) transcription factors96  and activates canonical NF-κB transcription factors that induce the expression of Bfl-1/A1, Bcl2, Bcl-xL, and IAP, proteins involved in antiapoptosis.97,,100  Similarly, MEK/ERK signaling has been shown to regulate the expression of BAD, Bcl-2, and Bcl-XL22,59 ; and although no role for JAK2/STAT5 signaling in the regulation of DC survival has yet been described, considering its widespread control of apoptosis regulators this module likely contributes.68,90  Although Bcl-2, FOXO1, and particularly Bcl-XL have been associated with DC survival,101  direct evidence that GM-CSF–activated signaling modules regulate DC survival through modulation of these factors is currently unavailable. In contrast, GM-CSF–induced mTORC1-mediated survival of human monocyte-derived DCs has been shown to depend on regulation of antiapoptotic Mcl-1.48  In addition, GSK-3β activity, which is inhibited by phosphorylation by PKB, can induce apoptosis by Mcl-1 destabilization.102  Furthermore, PI3K, PKB, and mTORC1 can regulate FOXO1, BAD, and Bcl-2, which could contribute to the maintenance of DCs.89  The aforementioned factors involved in DC survival regulation were mainly identified for differentiated DCs. Although tempting, extrapolation of these findings to DC precursors may be inappropriate because of the differential expression of apoptosis regulators at distinct DC differentiation stages.42  In addition, apoptosis regulators as well as signal transduction pathways regulating survival have been reported to be subset-specific,42,103  stressing the need to separately evaluate each DC subset. Overall, the molecular control of DC viability and expansion in general, and proliferation and survival during GM-CSF–induced development in particular, is very poorly resolved. Interesting findings on subset- and differentiation stage–specific differences have been reported and will hopefully be defined further in the future.

Similar to GM-CSF, Flt3L has a key role in the regulation of DC development, but important differences exist between these 2 cytokines. Whereas deficiency of GM-CSF or the GM-CSFR had only minor consequences on the DCs in mouse lymphoid organs and significantly reduced DC numbers were only observed when analysis was extended to nonlymphoid tissue,10,,13  the importance of Flt3L in DC homeostasis was more obvious. Studies analyzing Flt3L−/− or Flt3−/− mice described an up to 10-fold reduction in spleen, lymph node, and thymic DCs of different subtypes as well as a reduction in specific early DC progenitors, such as the MDP or CDP, although the latter was not confirmed in all studies.11,104,105  In contrast, GM-CSF−/− mice showed a specific reduction in lymphoid tissue and migratory DCs, whereas plasmacytoid DC numbers were unaffected.10,,13  In addition, inflammation-driven monocyte conversion to murine splenic DCs3,17  and DC generation during acute inflammatory arthritis and antigen-induced peritonitis18  were reported to depend on GM-CSF, emphasizing the increasing importance of GM-CSF under inflammatory conditions. In accordance with the data on cytokine and/or receptor deficiency, administration of GM-CSF results in a specific expansion of CD11chiCD11bhi murine spleen DCs, whereas massive expansion of DCs of all categories has been reported in mice and humans injected with Flt3L.9,106,107  The specific characteristics of GM-CSF and Flt3L can be explained by their molecular actions. Similar to GM-CSF, Flt3L activates STAT3, PI3K/PKB, and MEK/ERK signaling, but variations in the magnitude of activation of these modules and/or in their relative activity compared with each other may account for the differential DC expansion induced by injection of these cytokines.2  Major differences include the direct activation of canonical NF-κB and STAT5 by GM-CSF, events that are absent in Flt3L-stimulated cells. Whereas Flt3L supports the development of all DC subsets through activation of STAT3,30,33  STAT5 and canonical NF-κB transcription factor activity induces a different DC subset-distribution in GM-CSF–dependent cultures (Figure 5). However, although the actions of GM-CSF inhibit plasmacytoid DC development in vitro,14  plasmacytoid DC numbers were unchanged in GM-CSF−/− mice,11  indicating that the in vivo effects of GM-CSF on plasmacytoid DC development may be limited under steady-state conditions.

Besides their differential effects on the composition of the DC pool, GM-CSF and Flt3L induce functional differences in the DCs they generate. DCs differentiated in vitro in Flt3L-dependent cultures resemble the relatively tolerogenic lymphoid-resident DCs found during the steady state, whereas GM-CSF–differentiated DCs have a more immunogenic phenotype and functionality.19  Similarly, GM-CSF–driven DC expansion in vivo results in the generation of DCs with a robust immune function prone to the induction of Th1 immune responses against for example pulmonary infections.108,109  In the treatment of cancer, administration of either Flt3L or GM-CSF resulted in the generation of DCs, but better antitumor responses were observed when GM-CSF was used.110  The tolerogenic function of Flt3L-DC is further supported by the delayed onset of diabetes in NOD mice and the protection from inflammatory bowel disease in mice in response to Flt3L injection, which was associated with increased DC and regulatory T-cell numbers.20  Again, a molecular explanation is available for the differences between GM-CSF and Flt3L. In the previous section, GM-CSF–induced activation of STAT5 and canonical NF-κB transcription factors increases the intrinsic immunogenicity of the DCs generated (Figures 5 and 6). In contrast, Flt3L-activated STAT3 inhibits canonical NF-κB activity as well as the expression of MHC class II proteins and costimulatory molecules111,,114  and promotes transcription of IDO.115  These actions account for the tolerogenic function of DCs generated in response to Flt3L20  (Figure 6). Thus, steady-state Flt3L-induced DC development induced through STAT3 yields relatively tolerogenic DCs of all subsets. Under inflammatory conditions, GM-CSF–induced STAT5 and NF-κB activity leads to the generation of specific DC subsets with enhanced intrinsic immunogenicity.

Figure 6

A molecular explanation for the reciprocal effects of GM-CSF and Flt3L on DC immunogenicity. The complementary effects GM-CSF and Flt3L have on the DCs they generate can be explained by their molecular actions. Through activation of STAT3, Flt3L promotes expression of IDO but inhibits canonical NF-κB and MHC class II expression, thereby inhibiting DC immunogenicity. In contrast, GM-CSF activates STAT5 and canonical NF-κB besides STAT3. By promoting NF-κB activity and MHC class II expression, STAT5 overrules the tolerogenic function of STAT3, resulting in the development of more immunogenic DCs.

Figure 6

A molecular explanation for the reciprocal effects of GM-CSF and Flt3L on DC immunogenicity. The complementary effects GM-CSF and Flt3L have on the DCs they generate can be explained by their molecular actions. Through activation of STAT3, Flt3L promotes expression of IDO but inhibits canonical NF-κB and MHC class II expression, thereby inhibiting DC immunogenicity. In contrast, GM-CSF activates STAT5 and canonical NF-κB besides STAT3. By promoting NF-κB activity and MHC class II expression, STAT5 overrules the tolerogenic function of STAT3, resulting in the development of more immunogenic DCs.

Close modal

Because of its key role in the generation and function of in particular immunogenic DCs, GM-CSF is currently used in several therapeutic strategies. For instance, inclusion of GM-CSF in mobilization regimens for HSC transplantation has been reported to induce 3 to 10 times higher DC yields during mobilization and to improve DC reconstitution after autologous HSC transplantation.116,117  Besides aiding immune reconstitution, GM-CSF seems to potentiate antitumor responses by promoting DC generation, migration, and activation when (artificially) produced at the tumor site,118,120  and promising therapeutic vaccination strategies based on the ex vivo generation and tumor antigen loading of DCs mostly use GM-CSF–based protocols to generate DCs.121  Finally, GM-CSF is used as potent adjuvant in vaccination strategies for the prevention of hepatitis B virus infection in patient groups with immunization difficulties and for the treatment of hepatitis C virus, human immunodeficiency virus, and fungal or mycobacterial infections.122 

By addition of GM-CSF, downstream signaling cascades are activated without the possibility to distinguish between those with desired and adverse biologic consequences. Other complicating factors are negative feedback mechanisms and/or the presence of anti-inflammatory signals, for example in the tumor microenvironment, which counteract the effects of GM-CSF. To circumvent these problems, manipulation of the signaling proteins responsible for GM-CSF–driven effects on DCs seems a useful strategy. A potential target is the PI3K-PKB-mTOR signaling module, which could be used to boost DC yields, improve DC survival, and promote DC immunogenicity. Indeed, mouse bone marrow-derived conventional DCs and human monocyte–derived DCs expressing constitutively active PKB or lacking PTEN activity because of siRNA-mediated deletion are highly activated DCs with improved longevity that are able to eradicate tumors more efficiently than control DCs.49,50  Based on its decisive function in DC lineage commitment,28,29,32  STAT5 appears a candidate protein for the manipulation of subset specification. Alternatively, the STAT5-regulated factors that are responsible for its effects on lineage decisions, such as PU.1 or IRF8, or the subset-specific regulator RelB66,79  could be targeted. In mice, silencing of RelB has also been applied to induce tolerance to transplanted hearts and treating autoimmune myasthenia gravis.123,124  Tolerance has also been promoted through pharmacologic inhibition of mTOR,52  whereas deletion of tolerogenic STAT3 contributed to the development of immunogenic antitumor immune responses.125,126  Finally, manipulation of STAT5 and canonical NF-κB transcription factors could be attempted to promote the development of immunogenic DCs.

Although promising, direct targeting of GM-CSF–regulated signaling proteins does not completely resolve the side effects of GM-CSF administration. Specific targeting of the cells of interest remains required, but because a definition of human DC-specific progenitors is lacking, in vivo targeting to early progenitors is currently not possible in humans. Furthermore, interfering environmental signals, negative feedback, and adverse effects caused by interrelation of the different pathways cannot be avoided completely. Despite these remaining challenges, our knowledge on the molecular basis of GM-CSF–induced DC development contributes to the understanding of the specific characteristics of GM-CSF–differentiated DCs, and will hopefully lead to novel strategies to manipulate DC biology for therapeutic purposes.

The 4 principle signaling modules activated by GM-CSF, JAK2/STAT5, PI3K/PKB, MEK/ERK, and canonical NF-κB have separate as well as overlapping functions in the regulation of GM-CSF–induced DC development. Although often forgotten, rather than acting as separate entities, these cascades form an integrated network with the activity of one pathway also affecting others. This causes flexibility and enables adequate responses to distinct conditions but complicates the interpretation of experiments in which the activity of a specific signaling protein is manipulated. This is a subject that should be given more attention in the future.

The molecular actions of GM-CSF explain its effects on subset distribution of developing DC and the relatively immunogenic phenotype and functionality of DCs generated by GM-CSF compared with DCs differentiated in response to Flt3L. Further identification of GM-CSF–induced downstream signaling proteins and transcription factors will be of use in the improvement and/or development of DC-based therapies. In addition, further understanding of the ontogenetic background of DCs and of their contribution to immune responses under different pathophysiologic conditions is required. Eventually, this knowledge will enable the use of DCs with tightly controlled functional abilities in novel therapeutic applications.

The online version of this article contains a data supplement.

Contribution: L.v.d.L., P.J.C., and A.M.W. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Andrea M. Woltman, Erasmus MC-University Medical Center, Department of Gastroenterology and Hepatology, L-244, ‘s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands; e-mail: a.woltman@erasmusmc.nl.

1
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature
1998
392
6673
245
252
2
Merad
M
Manz
MG
Dendritic cell homeostasis.
Blood
2009
113
15
3418
3427
3
Shortman
K
Naik
SH
Steady-state and inflammatory dendritic-cell development.
Nat Rev Immunol
2007
7
1
19
30
4
Inaba
K
Steinman
RM
Pack
MW
et al
Identification of proliferating dendritic cell precursors in mouse blood.
J Exp Med
1992
175
5
1157
1167
5
Sallusto
F
Lanzavecchia
A
Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha.
J Exp Med
1994
179
4
1109
1118
6
Caux
C
Vanbervliet
B
Massacrier
C
et al
CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to GM-CSF+TNF alpha.
J Exp Med
1996
184
2
695
706
7
Inaba
K
Inaba
M
Romani
N
et al
Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J Exp Med
1992
176
6
1693
1702
8
Caux
C
Dezutter-Dambuyant
C
Schmitt
D
Banchereau
J
GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells.
Nature
1992
360
6401
258
261
9
Daro
E
Pulendran
B
Brasel
K
et al
Polyethylene glycol-modified GM-CSF expands CD11b(high)CD11c(high) but not CD11b(low)CD11c(high) murine dendritic cells in vivo: a comparative analysis with Flt3 ligand.
J Immunol
2000
165
1
49
58
10
Vremec
D
Lieschke
GJ
Dunn
AR
Robb
L
Metcalf
D
Shortman
K
The influence of granulocyte/macrophage colony-stimulating factor on dendritic cell levels in mouse lymphoid organs.
Eur J Immunol
1997
27
1
40
44
11
Kingston
D
Schmid
MA
Onai
N
Obata-Onai
A
Baumjohann
D
Manz
MG
The concerted action of GM-CSF and Flt3-ligand on in vivo dendritic cell homeostasis.
Blood
2009
114
4
835
843
12
Bogunovic
M
Ginhoux
F
Helft
J
et al
Origin of the lamina propria dendritic cell network.
Immunity
2009
31
3
513
525
13
Varol
C
Vallon-Eberhard
A
Elinav
E
et al
Intestinal lamina propria dendritic cell subsets have different origin and functions.
Immunity
2009
31
3
502
512
14
Gilliet
M
Boonstra
A
Paturel
C
et al
The development of murine plasmacytoid dendritic cell precursors is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor.
J Exp Med
2002
195
7
953
958
15
Ghirelli
C
Zollinger
R
Soumelis
V
Systematic cytokine receptor profiling reveals GM-CSF as a novel TLR-independent activator of human plasmacytoid predendritic cells.
Blood
2010
115
24
5037
5040
16
Hamilton
JA
Colony-stimulating factors in inflammation and autoimmunity.
Nat Rev Immunol
2008
8
7
533
544
17
Naik
SH
Metcalf
D
van Nieuwenhuijze
A
et al
Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes.
Nat Immunol
2006
7
6
663
671
18
Campbell
IK
van Nieuwenhuijze
A
Segura
E
et al
Differentiation of inflammatory dendritic cells is mediated by NF-kappaB1-dependent GM-CSF production in CD4 T cells.
J Immunol
2011
186
9
5468
5477
19
Xu
Y
Zhan
Y
Lew
AM
Naik
SH
Kershaw
MH
Differential development of murine dendritic cells by GM-CSF versus Flt3 ligand has implications for inflammation and trafficking.
J Immunol
2007
179
11
7577
7584
20
Liu
K
Nussenzweig
MC
Origin and development of dendritic cells.
Immunol Rev
2010
234
1
45
54
21
Barreda
DR
Hanington
PC
Belosevic
M
Regulation of myeloid development and function by colony stimulating factors.
Dev Comp Immunol
2004
28
5
509
554
22
Martinez-Moczygemba
M
Huston
DP
Biology of common beta receptor-signaling cytokines: IL-3, IL-5, and GM-CSF.
J Allergy Clin Immunol
2003
112
4
653
665
23
Geijsen
N
Koenderman
L
Coffer
PJ
Specificity in cytokine signal transduction: lessons learned from the IL-3/IL-5/GM-CSF receptor family.
Cytokine Growth Factor Rev
2001
12
1
19
25
24
Hercus
TR
Thomas
D
Guthridge
MA
et al
The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease.
Blood
2009
114
7
1289
1298
25
Meads
MB
Li
ZW
Dalton
WS
A novel TNF receptor-associated factor 6 binding domain mediates NF-kappa B signaling by the common cytokine receptor beta subunit.
J Immunol
2010
185
3
1606
1615
26
Ebner
K
Bandion
A
Binder
BR
de Martin
R
Schmid
JA
GM-CSF activates NF-kappaB via direct interaction of the GMCSF receptor with IkappaB kinase beta.
Blood
2003
102
1
192
199
27
Zhong
J
Yang
P
Muta
K
et al
Loss of Jak2 selectively suppresses DC-mediated innate immune response and protects mice from lethal dose of LPS-induced septic shock.
PLoS One
2010
5
3
e9593
28
Cohen
PA
Koski
GK
Czerniecki
BJ
et al
STAT3- and STAT5-dependent pathways competitively regulate the pan-differentiation of CD34pos cells into tumor-competent dendritic cells.
Blood
2008
112
5
1832
1843
29
Esashi
E
Wang
YH
Perng
O
Qin
XF
Liu
YJ
Watowich
SS
The signal transducer STAT5 inhibits plasmacytoid dendritic cell development by suppressing transcription factor IRF8.
Immunity
2008
28
4
509
520
30
Laouar
Y
Welte
T
Fu
XY
Flavell
RA
STAT3 is required for Flt3L-dependent dendritic cell differentiation.
Immunity
2003
19
6
903
912
31
Welte
T
Koch
F
Schuler
G
Lechner
J
Doppler
W
Heufler
C
Granulocyte-macrophage colony-stimulating factor induces a unique set of STAT factors in murine dendritic cells.
Eur J Immunol
1997
27
10
2737
2740
32
van de Laar
L
van den Bosch
A
Wierenga
AT
Janssen
HL
Coffer
PJ
Woltman
AM
Tight control of STAT5 activity determines human CD34-derived interstitial dendritic cell and Langerhans cell development.
J Immunol
2011
186
12
7016
7024
33
Onai
N
Obata-Onai
A
Tussiwand
R
Lanzavecchia
A
Manz
MG
Activation of the Flt3 signal transduction cascade rescues and enhances type I interferon-producing and dendritic cell development.
J Exp Med
2006
203
1
227
238
34
Saito
M
Nagasawa
M
Takada
H
et al
Defective IL-10 signaling in hyper-IgE syndrome results in impaired generation of tolerogenic dendritic cells and induced regulatory T cells.
J Exp Med
2011
208
2
235
249
35
Choi
YE
Yu
HN
Yoon
CH
Bae
YS
Tumor-mediated down-regulation of MHC class II in DC development is attributable to the epigenetic control of the CIITA type I promoter.
Eur J Immunol
2009
39
3
858
868
36
Sebastian
C
Serra
M
Yeramian
A
Serrat
N
Lloberas
J
Celada
A
Deacetylase activity is required for STAT5-dependent GM-CSF functional activity in macrophages and differentiation to dendritic cells.
J Immunol
2008
180
9
5898
5906
37
Fujita
J
Mizuki
M
Otsuka
M
et al
Myeloid neoplasm-related gene abnormalities differentially affect dendritic cell differentiation from murine hematopoietic stem/progenitor cells.
Immunol Lett
2011
136
1
61
73
38
Fukao
T
Tanabe
M
Terauchi
Y
et al
PI3K-mediated negative feedback regulation of IL-12 production in DCs.
Nat Immunol
2002
3
9
875
881
39
Sathaliyawala
T
O'Gorman
WE
Greter
M
et al
Mammalian target of rapamycin controls dendritic cell development downstream of Flt3 ligand signaling.
Immunity
2010
33
4
597
606
40
Antignano
F
Ibaraki
M
Kim
C
et al
SHIP is required for dendritic cell maturation.
J Immunol
2010
184
6
2805
2813
41
Zaru
R
Mollahan
P
Watts
C
3-Phosphoinositide-dependent kinase 1 deficiency perturbs Toll-like receptor signaling events and actin cytoskeleton dynamics in dendritic cells.
J Biol Chem
2008
283
2
929
939
42
van de Laar
L
Buitenhuis
M
Wensveen
FM
Janssen
HL
Coffer
PJ
Woltman
AM
Human CD34-derived myeloid dendritic cell development requires intact phosphatidylinositol 3-kinase-protein kinase B-mammalian target of rapamycin signaling.
J Immunol
2010
184
12
6600
6611
43
Hackstein
H
Taner
T
Zahorchak
AF
et al
Rapamycin inhibits IL-4-induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo.
Blood
2003
101
11
4457
4463
44
Haidinger
M
Poglitsch
M
Geyeregger
R
et al
A versatile role of mammalian target of rapamycin in human dendritic cell function and differentiation.
J Immunol
2010
185
7
3919
3931
45
Monti
P
Mercalli
A
Leone
BE
Valerio
DC
Allavena
P
Piemonti
L
Rapamycin impairs antigen uptake of human dendritic cells.
Transplantation
2003
75
1
137
145
46
Woltman
AM
de Fijter
JW
Kamerling
SW
et al
Rapamycin induces apoptosis in monocyte- and CD34-derived dendritic cells but not in monocytes and macrophages.
Blood
2001
98
1
174
180
47
Xie
J
Qian
J
Yang
J
Wang
S
Freeman
ME
III
Yi
Q
Critical roles of Raf/MEK/ERK and PI3K/AKT signaling and inactivation of p38 MAP kinase in the differentiation and survival of monocyte-derived immature dendritic cells.
Exp Hematol
2005
33
5
564
572
48
Woltman
AM
van der Kooij
SW
Coffer
PJ
Offringa
R
Daha
MR
van Kooten
C
Rapamycin specifically interferes with GM-CSF signaling in human dendritic cells, leading to apoptosis via increased p27KIP1 expression.
Blood
2003
101
4
1439
1445
49
Kim
JH
Kang
TH
Noh
KH
et al
Enhancement of DC vaccine potency by activating the PI3K/AKT pathway with a small interfering RNA targeting PTEN.
Immunol Lett
2010
134
1
47
54
50
Park
D
Lapteva
N
Seethammagari
M
Slawin
KM
Spencer
DM
An essential role for Akt1 in dendritic cell function and tumor immunotherapy.
Nat Biotechnol
2006
24
12
1581
1590
51
Hackstein
H
Taner
T
Logar
AJ
Thomson
AW
Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells.
Blood
2002
100
3
1084
1087
52
Turnquist
HR
Raimondi
G
Zahorchak
AF
Fischer
RT
Wang
Z
Thomson
AW
Rapamycin-conditioned dendritic cells are poor stimulators of allogeneic CD4+ T cells, but enrich for antigen-specific Foxp3+ T regulatory cells and promote organ transplant tolerance.
J Immunol
2007
178
11
7018
7031
53
Rodionova
E
Conzelmann
M
Maraskovsky
E
et al
GSK-3 mediates differentiation and activation of proinflammatory dendritic cells.
Blood
2007
109
4
1584
1592
54
Ono
T
Yanagawa
Y
Iwabuchi
K
Nonomura
K
Onoe
K
Glycogen synthase kinase 3 activity during development of bone marrow-derived dendritic cells (DCs) essential for the DC function to induce T helper 2 polarization.
Immunology
2007
122
2
189
198
55
Bendix
I
Pfueller
CF
Leuenberger
T
et al
MAPK3 deficiency drives autoimmunity via DC arming.
Eur J Immunol
2010
40
5
1486
1495
56
Kandilci
A
Grosveld
GC
SET-induced calcium signaling and MAPK/ERK pathway activation mediate dendritic cell-like differentiation of U937 cells.
Leukemia
2005
19
8
1439
1445
57
Lindner
I
Torruellas-Garcia
J
Kolonias
D
et al
Modulation of dendritic cell differentiation and function by YopJ of Yersinia pestis.
Eur J Immunol
2007
37
9
2450
2462
58
Hayashi
F
Yanagawa
Y
Onoe
K
Iwabuchi
K
Dendritic cell differentiation with prostaglandin E results in selective attenuation of the extracellular signal-related kinase pathway and decreased interleukin-23 production.
Immunology
2010
131
1
67
76
59
Geest
CR
Coffer
PJ
MAPK signaling pathways in the regulation of hematopoiesis.
J Leukoc Biol
2009
86
2
237
250
60
Wang
S
Yang
J
Qian
J
Wezeman
M
Kwak
LW
Yi
Q
Tumor evasion of the immune system: inhibiting p38 MAPK signaling restores the function of dendritic cells in multiple myeloma.
Blood
2006
107
6
2432
2439
61
Xie
J
Qian
J
Wang
S
Freeman
ME
3rd
Epstein
J
Yi
Q
Novel and detrimental effects of lipopolysaccharide on in vitro generation of immature dendritic cells: involvement of mitogen-activated protein kinase p38.
J Immunol
2003
171
9
4792
4800
62
Ouaaz
F
Arron
J
Zheng
Y
Choi
Y
Beg
AA
Dendritic cell development and survival require distinct NF-kappaB subunits.
Immunity
2002
16
2
257
270
63
Gu
XY
Zhou
LF
Zhang
MS
et al
Targeted NF-kappaB inhibition of asthmatic serum-mediated human monocyte-derived dendritic cell differentiation in a transendothelial trafficking model.
Cell Immunol
2009
260
1
14
20
64
van de Laar
L
van den Bosch
A
van der Kooij
SW
et al
A nonredundant role for canonical NF-kappaB in human myeloid dendritic cell development and function.
J Immunol
2010
185
12
7252
7261
65
Zhou
LF
Zhang
MS
Yin
KS
et al
Effects of adenoviral gene transfer of mutated IkappaBalpha, a novel inhibitor of NF-kappaB, on human monocyte-derived dendritic cells.
Acta Pharmacol Sin
2006
27
5
609
616
66
Platzer
B
Jorgl
A
Taschner
S
Hocher
B
Strobl
H
RelB regulates human dendritic cell subset development by promoting monocyte intermediates.
Blood
2004
104
12
3655
3663
67
Hayden
MS
West
AP
Ghosh
S
NF-kappaB and the immune response.
Oncogene
2006
25
51
6758
6780
68
Coffer
PJ
Koenderman
L
de Groot
RP
The role of STATs in myeloid differentiation and leukemia.
Oncogene
2000
19
21
2511
2522
69
Wu
L
Liu
YJ
Development of dendritic-cell lineages.
Immunity
2007
26
6
741
750
70
Wang
D
Paz-Priel
I
Friedman
AD
NF-kappa B p50 regulates C/EBP alpha expression and inflammatory cytokine-induced neutrophil production.
J Immunol
2009
182
9
5757
5762
71
Iwama
A
Osawa
M
Hirasawa
R
et al
Reciprocal roles for CCAAT/enhancer binding protein (C/EBP) and PU.1 transcription factors in Langerhans cell commitment.
J Exp Med
2002
195
5
547
558
72
Zhang
P
Iwasaki-Arai
J
Iwasaki
H
et al
Enhancement of hematopoietic stem cell repopulating capacity and self-renewal in the absence of the transcription factor C/EBP alpha.
Immunity
2004
21
6
853
863
73
Lehtonen
A
Veckman
V
Nikula
T
et al
Differential expression of IFN regulatory factor 4 gene in human monocyte-derived dendritic cells and macrophages.
J Immunol
2005
175
10
6570
6579
74
Suzuki
S
Honma
K
Matsuyama
T
et al
Critical roles of interferon regulatory factor 4 in CD11bhighCD8alpha− dendritic cell development.
Proc Natl Acad Sci U S A
2004
101
24
8981
8986
75
Tamura
T
Tailor
P
Yamaoka
K
et al
IFN regulatory factor-4 and -8 govern dendritic cell subset development and their functional diversity.
J Immunol
2005
174
5
2573
2581
76
Dejardin
E
Droin
NM
Delhase
M
et al
The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways.
Immunity
2002
17
4
525
535
77
Jacque
E
Tchenio
T
Piton
G
Romeo
PH
Baud
V
RelA repression of RelB activity induces selective gene activation downstream of TNF receptors.
Proc Natl Acad Sci U S A
2005
102
41
14635
14640
78
Saccani
S
Pantano
S
Natoli
G
Modulation of NF-kappaB activity by exchange of dimers.
Mol Cell
2003
11
6
1563
1574
79
Wu
L
D'Amico
A
Winkel
KD
Suter
M
Lo
D
Shortman
K
RelB is essential for the development of myeloid-related CD8alpha− dendritic cells but not of lymphoid-related CD8alpha+ dendritic cells.
Immunity
1998
9
6
839
847
80
Neumann
M
Fries
H
Scheicher
C
et al
Differential expression of Rel/NF-kappaB and octamer factors is a hallmark of the generation and maturation of dendritic cells.
Blood
2000
95
1
277
285
81
Schuringa
JJ
Chung
KY
Morrone
G
Moore
MA
Constitutive activation of STAT5A promotes human hematopoietic stem cell self-renewal and erythroid differentiation.
J Exp Med
2004
200
5
623
635
82
Wierenga
AT
Schepers
H
Moore
MA
Vellenga
E
Schuringa
JJ
STAT5-induced self-renewal and impaired myelopoiesis of human hematopoietic stem/progenitor cells involves down-modulation of C/EBPalpha.
Blood
2006
107
11
4326
4333
83
Carotta
S
Wu
L
Nutt
SL
Surprising new roles for PU.1 in the adaptive immune response.
Immunol Rev
2010
238
1
63
75
84
Gangenahalli
GU
Gupta
P
Saluja
D
et al
Stem cell fate specification: role of master regulatory switch transcription factor PU.1 in differential hematopoiesis.
Stem Cells Dev
2005
14
2
140
152
85
Heinz
LX
Platzer
B
Reisner
PM
et al
Differential involvement of PU.1 and Id2 downstream of TGF-beta1 during Langerhans-cell commitment.
Blood
2006
107
4
1445
1453
86
Guerriero
A
Langmuir
PB
Spain
LM
Scott
EW
PU.1 is required for myeloid-derived but not lymphoid-derived dendritic cells.
Blood
2000
95
3
879
885
87
Hambleton
S
Salem
S
Bustamante
J
et al
IRF8 mutations and human dendritic-cell immunodeficiency.
N Engl J Med
2011
365
2
127
138
88
Nakamura
T
Ouchida
R
Kodama
T
et al
Cytokine receptor common beta subunit-mediated STAT5 activation confers NF-kappa B activation in murine proB cell line Ba/F3 cells.
J Biol Chem
2002
277
8
6254
6265
89
Datta
SR
Brunet
A
Greenberg
ME
Cellular survival: a play in three Akts.
Genes Dev
1999
13
22
2905
2927
90
Rane
SG
Reddy
EP
JAKs, STATs and Src kinases in hematopoiesis.
Oncogene
2002
21
21
3334
3358
91
Ross
SE
Erickson
RL
Hemati
N
MacDougald
OA
Glycogen synthase kinase 3 is an insulin-regulated C/EBPalpha kinase.
Mol Cell Biol
1999
19
12
8433
8441
92
Yao
Y
Xu
Q
Kwon
MJ
et al
ERK and p38 MAPK signaling pathways negatively regulate CIITA gene expression in dendritic cells and macrophages.
J Immunol
2006
177
1
70
76
93
Bhaskar
PT
Hay
N
The two TORCs and Akt.
Dev Cell
2007
12
4
487
502
94
Deane
JA
Fruman
DA
Phosphoinositide 3-kinase: diverse roles in immune cell activation.
Annu Rev Immunol
2004
22
563
598
95
Franke
TF
PI3K/Akt: getting it right matters.
Oncogene
2008
27
50
6473
6488
96
Chapuis
N
Park
S
Leotoing
L
et al
IkappaB kinase overcomes PI3K/Akt and ERK/MAPK to control FOXO3a activity in acute myeloid leukemia.
Blood
2010
116
20
4240
4250
97
Catz
SD
Johnson
JL
Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer.
Oncogene
2001
20
50
7342
7351
98
Chen
C
Edelstein
LC
Gelinas
C
The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L).
Mol Cell Biol
2000
20
8
2687
2695
99
Wang
CY
Mayo
MW
Korneluk
RG
Goeddel
DV
Baldwin
AS
Jr
NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
Science
1998
281
5383
1680
1683
100
Zong
WX
Edelstein
LC
Chen
C
Bash
J
Gelinas
C
The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis.
Genes Dev
1999
13
4
382
387
101
Chen
M
Wang
J
Programmed cell death of dendritic cells in immune regulation.
Immunol Rev
2010
236
11
27
102
Maurer
U
Charvet
C
Wagman
AS
Dejardin
E
Green
DR
Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1.
Mol Cell
2006
21
6
749
760
103
Chen
M
Huang
L
Shabier
Z
Wang
J
Regulation of the lifespan in dendritic cell subsets.
Mol Immunol
2007
44
10
2558
2565
104
McKenna
HJ
Stocking
KL
Miller
RE
et al
Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells.
Blood
2000
95
11
3489
3497
105
Waskow
C
Liu
K
Darrasse-Jeze
G
et al
The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues.
Nat Immunol
2008
9
6
676
683
106
Maraskovsky
E
Daro
E
Roux
E
et al
In vivo generation of human dendritic cell subsets by Flt3 ligand.
Blood
2000
96
3
878
884
107
Maraskovsky
E
Brasel
K
Teepe
M
et al
Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified.
J Exp Med
1996
184
5
1953
1962
108
Miller
G
Pillarisetty
VG
Shah
AB
Lahrs
S
Xing
Z
DeMatteo
RP
Endogenous granulocyte-macrophage colony-stimulating factor overexpression in vivo results in the long-term recruitment of a distinct dendritic cell population with enhanced immunostimulatory function.
J Immunol
2002
169
6
2875
2885
109
Wang
J
Snider
DP
Hewlett
BR
et al
Transgenic expression of granulocyte-macrophage colony-stimulating factor induces the differentiation and activation of a novel dendritic cell population in the lung.
Blood
2000
95
7
2337
2345
110
Mach
N
Gillessen
S
Wilson
SB
Sheehan
C
Mihm
M
Dranoff
G
Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or Flt3-ligand.
Cancer Res
2000
60
12
3239
3246
111
Hoentjen
F
Sartor
RB
Ozaki
M
Jobin
C
STAT3 regulates NF-kappaB recruitment to the IL-12p40 promoter in dendritic cells.
Blood
2005
105
2
689
696
112
Kitamura
H
Kamon
H
Sawa
S
et al
IL-6-STAT3 controls intracellular MHC class II alphabeta dimer level through cathepsin S activity in dendritic cells.
Immunity
2005
23
5
491
502
113
Nefedova
Y
Cheng
P
Gilkes
D
et al
Activation of dendritic cells via inhibition of Jak2/STAT3 signaling.
J Immunol
2005
175
7
4338
4346
114
Yu
Z
Zhang
W
Kone
BC
Signal transducers and activators of transcription 3 (STAT3) inhibits transcription of the inducible nitric oxide synthase gene by interacting with nuclear factor kappaB.
Biochem J
2002
367
1
97
105
115
Sun
Y
Chin
YE
Weisiger
E
et al
Cutting edge: negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3.
J Immunol
2009
182
10
5899
5903
116
Gazitt
Y
Comparison between granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in the mobilization of peripheral blood stem cells.
Curr Opin Hematol
2002
9
3
190
198
117
Eksioglu
EA
Kielbasa
J
Eisen
S
Reddy
V
Granulocyte-macrophage colony-stimulating factor increases the proportion of circulating dendritic cells after autologous but not after allogeneic hematopoietic stem cell transplantation.
Cytotherapy
2011
13
7
888
896
118
Burke
JM
GM-CSF-armed, replication-competent viruses for cancer.
Cytokine Growth Factor Rev
2010
21
2
149
151
119
Dranoff
G
GM-CSF-based cancer vaccines.
Immunol Rev
2002
188
147
154
120
Waller
EK
The role of sargramostim (rhGM-CSF) as immunotherapy.
Oncologist
2007
12
suppl 2
22
26
121
Ueno
H
Schmitt
N
Klechevsky
E
et al
Harnessing human dendritic cell subsets for medicine.
Immunol Rev
2010
234
1
199
212
122
Fabrizi
F
Ganeshan
SV
Dixit
V
Martin
P
Meta-analysis: the adjuvant role of granulocyte macrophage-colony stimulating factor on immunological response to hepatitis B virus vaccine in end-stage renal disease.
Aliment Pharmacol Ther
2006
24
5
789
796
123
Li
M
Zhang
X
Zheng
X
et al
Immune modulation and tolerance induction by RelB-silenced dendritic cells through RNA interference.
J Immunol
2007
178
9
5480
5487
124
Zhang
Y
Yang
H
Xiao
B
et al
Dendritic cells transduced with lentiviral-mediated RelB-specific ShRNAs inhibit the development of experimental autoimmune myasthenia gravis.
Mol Immunol
2009
46
4
657
667
125
Herrmann
A
Kortylewski
M
Kujawski
M
et al
Targeting Stat3 in the myeloid compartment drastically improves the in vivo antitumor functions of adoptively transferred T cells.
Cancer Res
2010
70
19
7455
7464
126
Melillo
JA
Song
L
Bhagat
G
et al
Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function.
J Immunol
2010
184
5
2638
2645
127
Vallabhapurapu
S
Karin
M
Regulation and function of NF-kappaB transcription factors in the immune system.
Annu Rev Immunol
2009
27
693
733

Supplemental data

Sign in via your Institution