Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2012 Jan 23;109(6):E343–E352. doi: 10.1073/pnas.1115635109

Transforming growth factor beta-activated kinase 1 (TAK1)-dependent checkpoint in the survival of dendritic cells promotes immune homeostasis and function

Yanyan Wang a,1, Gonghua Huang a,1, Peter Vogel b, Geoffrey Neale c, Boris Reizis d, Hongbo Chi a,2
PMCID: PMC3277515  PMID: 22308391

Abstract

Homeostatic control of dendritic cell (DC) survival is crucial for adaptive immunity, but the molecular mechanism is not well defined. Moreover, how DCs influence immune homeostasis under steady state remains unclear. Combining DC-specific and -inducible deletion systems, we report that transforming growth factor beta-activated kinase 1 (TAK1) is an essential regulator of DC survival and immune system homeostasis and function. Deficiency of TAK1 in CD11c+ cells induced markedly elevated apoptosis, leading to the depletion of DC populations, especially the CD8+ and CD103+ DC subsets in lymphoid and nonlymphoid tissues, respectively. TAK1 also contributed to DC development by promoting the generation of DC precursors. Prosurvival signals from Toll-like receptors, CD40 and receptor activator of nuclear factor-κB (RANK) are integrated by TAK1 in DCs, which in turn mediated activation of downstream NF-κB and AKT-Foxo pathways and established a gene-expression program. TAK1 deficiency in DCs caused a myeloid proliferative disorder characterized by expansion of neutrophils and inflammatory monocytes, disrupted T-cell homeostasis, and prevented effective T-cell priming and generation of regulatory T cells. Moreover, TAK1 signaling in DCs was required to prevent myeloid proliferation even in the absence of lymphocytes, indicating a previously unappreciated regulatory mechanism of DC-mediated control of myeloid cell-dependent inflammation. Therefore, TAK1 orchestrates a prosurvival checkpoint in DCs that affects the homeostasis and function of the immune system.

Keywords: innate immunity, immune tolerance


Dendritic cells (DCs) are a heterogeneous population of immune cells specialized to capture, process, and present antigens to T lymphocytes (1). DCs can be divided into three main populations: conventional DCs (cDCs), type I IFN-secreting plasmacytoid DCs (pDCs), and migratory DCs (24). cDCs can be further divided into CD8+ and CD8 DC subsets in lymphoid organs, whereas the CD103+ DC subset in nonlymphoid organs serves as a functional equivalent of CD8+ cDCs in lymphoid organs (24). Homeostasis of DCs is maintained through a dynamic balance of three main mechanisms. First, DCs are continuously replenished by blood-borne precursors that are further derived from macrophage and DC precursor (MDP) and common DC precursor (CDP) populations in the bone marrow (BM) (2, 5). Second, ∼5% of DCs are dividing at any given time. Although limited, the self-renewal ability in situ contributes to the maintenance of the DC population (68). Finally, apoptotic cell death of DCs is a central mechanism for down-modulating DC numbers to ensure immune homeostasis (9). Recent studies have provided important insight into the regulation of DC survival by Bcl-2 family members, including the antiapoptotic members Bcl-2 and Bcl-xL and the proapoptotic Bim (1012). Expression of these Bcl-2 family proteins is dynamically regulated by signals transduced from prosurvival cytokines, ligands for Toll-like receptors (TLRs), and other extracellular stimuli, thereby dictating the decision between survival and death (9). However, it is largely unexplored how the expression of Bcl-2–related proteins and apoptosis of DCs are regulated by the intracellular signaling network.

Despite a well-established role for DCs to induce antigen-specific immune activation and tolerance (13, 14), how DC survival affects immune homeostasis in the steady state, in the absence of encounter with foreign antigens, remains incompletely understood. Enhancing DC survival through transgenic expression of the baculoviral caspase inhibitor p35 or deletion of Fas leads to chronic lymphocyte activation and systemic autoimmunity (15, 16), whereas increasing DC numbers by FMS-like tyrosine kinase 3 ligand (FLT3L) administration protects mice from autoimmunity via inducing regulatory T-cell (Treg) accumulation (1719). Conversely, constitutive ablation of cDCs through the expression of diphtheria toxin in CD11c+ cells has been reported to cause fatal T-cell–mediated autoimmune disease (20) or a myeloid proliferative syndrome without obvious alterations of T-cell homeostasis (21). Mechanistically, peripheral DCs have been implicated in promoting homeostatic proliferation and survival of T cells (2224) as well as inducing a low-level tonic signaling in naive T cells to program their responsiveness to foreign antigens (25). Moreover, although a direct role for DCs to promote Treg expansion and accumulation has been established by recent elegant studies (17, 26), how this effect contributes to immune homeostasis is not fully understood (20, 21, 27, 28). Because many of the previous studies have used nonphysiological approaches to modulate DC survival, how endogenous regulators of DC survival influence immune homeostasis and function remains to be established.

Transforming growth factor beta-activated kinase 1 (TAK1) (encoded by Map3k7) is arguably the most widely used MAP kinase kinase kinase in the immune system (29). The essential role of TAK1 in host immune defense was first demonstrated in Drosophila (30, 31) and later confirmed in murine cells following stimulation through TLRs and proinflammatory cytokine receptors (32, 33). TAK1 also mediates the intracellular sensor pathway mediated by nucleotide-binding oligomerization domain 1 (NOD1) and NOD2 (34, 35), but TLR8-induced activation of NF-κB and JNK is independent of TAK1 (36). In lymphocytes, TAK1 is an essential component of antigen receptor signaling and promotes lymphocyte proliferation and survival and adaptive immune functions (33, 3740). Moreover, TAK1 is critical for the survival of hematopoietic stem cells and progenitors (41). These results indicate a cell context-dependent function for TAK1 in the immune and hematopoietic systems.

Whereas a role for TAK1 in the initiation of innate immune responses upon pathogen recognition is well established, its role in the homeostatic control of innate immune cells such as DCs has not been examined. To investigate the function of TAK1 in DCs, we generated DC-specific TAK1-deficient mice and found that TAK1 was essential for the homeostasis of DCs by promoting their survival. Using an inducible deletion system, we further identified a direct role of TAK1 to actively maintain mature DCs and BM precursors. Moreover, TAK1 deficiency in DCs caused a myeloid proliferative disorder, disrupted T-cell homeostasis under steady state, and prevented effective T-cell priming and Treg generation. Our studies demonstrate that a TAK1-mediated checkpoint in DC survival has a key role in the homeostasis and function of the innate and adaptive immune systems.

Results

Cell-Autonomous Role of TAK1 in Regulating DC Populations.

To investigate the function of TAK1 in DCs, we generated DC-specific TAK1-deficient mice by crossing mice bearing floxed and null alleles of the Map3k7 gene with transgenic mice expressing Cre under the control of the CD11c promoter to generate Map3k7flox/null CD11c-Cre mice (called “Map3k7DC mice” hereafter) (37, 42). TAK1 was efficiently deleted in splenic DCs but not T cells from these mice (SI Appendix, Fig. S1A). Flow cytometry analysis of splenic DC populations in Map3k7DC mice showed that the percentage and cell number of cDC (CD11c+MHC-II+) and pDC (CD11clomPDCA-1+) populations were markedly reduced compared with those of wild-type (WT) mice (Fig. 1A). Within cDCs, the CD8+ subset was more profoundly affected than the CD8 (CD11b+) subset (Fig. 1A). Similar defects were observed in other lymphoid organs, including mesenteric lymph nodes (MLNs) and thymus (Fig. 1B and SI Appendix, Fig. S1B). Moreover, nonlymphoid organs, such as the liver, had lower DC frequency with a preferential reduction of the CD103+ DC subset (Fig. 1C). This result is consistent with the recent findings that nonlymphoid CD103+ DCs and lymphoid organ-resident CD8+ cDCs are developmentally and functionally related (8, 4345). These results reveal a key role for TAK1 to regulate DC populations in both lymphoid and nonlymphoid organs.

Fig. 1.

Fig. 1.

TAK1 has a key cell-autonomous role in regulating DC populations. (A) Flow cytometry of splenic cDC, pDC, CD8+ cDC, and CD11b+ cDC populations in WT and Map3k7DC mice. MHC-II, MHC class II. (Right) The proportion and cell numbers of the splenic DC populations in WT and Map3k7DC mice. (B and C) Flow cytometry of DC populations in the MLNs (B) and liver (C) of WT and Map3k7DC mice. (D) Flow cytometry of the contributions of spike BM-derived (CD45.1.2+) and WT or Map3k7DC donor BM-derived cells (CD45.2.2+) in the mixed chimeras after 2 mo of reconstitution. Data are representative of three to five independent experiments. Data represent the mean ± SEM.

To address whether the reduction of DCs in Map3k7DC mice was an intrinsic defect, we generated mixed BM chimeras. Specifically, we transferred WT or Map3k7DC BM cells (CD45.2.2+; donor) and WT BM cells (CD45.1.2+; spike) at a ratio of 1:1 into lethally irradiated WT (CD45.1.1+) recipients. After reconstitution, we analyzed splenic DC populations in the mixed chimeras. Compared with WT BM-derived donor cells, Map3k7DC donor cells in the chimeras contained a greatly reduced DC population (Fig. 1D). Therefore, TAK1 has a key cell-autonomous function in maintaining the DC pool.

TAK1 Promotes DC Survival and Development but Not Proliferation.

The overall size of DC populations is dependent upon the rates of apoptosis and proliferation as well as replenishment from DC precursors (2, 5, 9). We first measured caspase activity, a hallmark of apoptotic cell death (46). The percentage of caspase-positive DCs from Map3k7DC mice was significantly higher compared with that in WT cells (Fig. 2A). Further, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay also revealed elevated apoptosis of Map3k7DC DCs (Fig. 2B).

Fig. 2.

Fig. 2.

Deletion of TAK1 results in excessive apoptosis of DCs and loss of MDPs. (A) Caspase activity in freshly isolated WT and Map3k7DC splenic DCs, assessed by staining with FITC-conjugated VAD-FMK. (Right) The proportion of caspase+ cells. (B) Measurement of apoptosis of WT and Map3k7DC splenic DCs by TUNEL staining. (C) Flow cytometry of BrdU incorporation into splenic DCs of WT and Map3k7DC mice after an in vivo BrdU pulse for 24 h. (Right) The percentage of proliferating splenic DCs in WT and Map3k7DC mice. (D) Flow cytometry of MDPs (LinSca-1CSF1R+) in the BM Lin cells of WT and Map3k7CreER mice after 3 d of tamoxifen treatment in vivo. (Right) The proportion of MDPs among BM Lin cells. (E) BM cells from WT and Map3k7CreER mice were cultured with FLT3L in the presence of 0.5 μM 4-OHT. The expression of CSF1R and CD11c was analyzed by flow cytometry at day 4 (Left) and the percentage of CSF1R+ cells in CD11c cells was calculated (Right). Data are representative of three independent experiments. Data represent the mean ± SEM.

We next tested the possibility that the decreased DC population in Map3k7DC mice was partly ascribed to defective DC proliferation in situ (68). Notably, TAK1 is essential for mediating cell cycle progression in lymphocytes (33, 3740). We therefore measured DC proliferation in vivo using the Bromodeoxyuridine (BrdU) incorporation assay. DCs in WT and Map3k7DC mice incorporated BrdU to a comparable degree (Fig. 2C), indicating the lack of a role for TAK1 in DC proliferation.

Finally, we determined whether TAK1 regulated DC development by acting on DC precursors. One of the earliest precursor populations identified for DCs is MDPs (4749). Because CD11c-Cre mice do not allow deletion in MDPs, we crossed Map3k7flox/null mice with Rosa26-Cre-ERT2 mice (a Cre-ER fusion gene was recombined into the ubiquitously expressed Rosa26 locus) to generate Map3k7flox/null Rosa26-Cre-ERT2 mice (called “Map3k7CreER mice” hereafter). Three days after treatment of WT and Map3k7CreER mice with tamoxifen in vivo, the percentage of MDPs, defined by LinSca-1CSF1R(CD115)+ in Map3k7CreER mice was significantly decreased compared with that in WT mice (Fig. 2D). Next, we cultured WT and Map3k7CreER BM cells with FLT3L in the presence of 4-hydroxytamoxifen (4-OHT). After 4 d of culture, Map3k7CreER BM cells showed considerable defects to up-regulate CSF1R, a defining molecule for MDPs (49) (Fig. 2E). These data indicate an important role of TAK1 in the generation of MDPs, thereby contributing to DC development. Altogether, TAK1 is critical for the survival and development but not proliferation of DCs.

TAK1 Is Essential for Maintaining DC Survival by Integrating Prosurvival Receptor Signals.

To directly investigate the effects of TAK1 on the homeostasis of mature DCs, we analyzed DCs in WT and Map3k7CreER mice after short-term treatment with tamoxifen. Acute systemic deletion of TAK1, although lacking cell type-specific precision, allowed us to exclude the compensatory effects that might have developed due to the continuous loss of TAK1 in DCs or precursors. Following in vivo tamoxifen-mediated TAK1 deletion, splenic cDCs were markedly reduced, with a more prominent reduction in the CD8+ subset. The pDC population was also greatly reduced (Fig. 3A). Accordingly, caspase activation in DCs was elevated in these cells (Fig. 3B). The effect of TAK1 deletion on DC maintenance was further tested in vitro. BM cells from WT or Map3k7CreER mice (CD45.2+) were mixed with an equal number of WT (CD45.1+) spike cells, and after 7 d of culture with FLT3L to derive DCs, 4-OHT was added. After culturing for 6 additional days, CD11c+ cells from Map3k7CreER BM cells were selectively reduced (Fig. 3C), associated with enhanced apoptosis (Fig. 3D), compared with WT or spike-derived cells. These results demonstrate that one essential mechanism for the prosurvival function of TAK1 is through the maintenance of mature DC survival.

Fig. 3.

Fig. 3.

TAK1 is essential for the maintenance of mature DCs. (A) Flow cytometry of splenic cDC, pDC, CD8+ cDC, and CD11b+ cDC populations in WT and Map3k7CreER mice after 3 d of tamoxifen treatment. (Right) The proportion and cell numbers of the splenic DC populations. (B) Caspase activity in splenic DCs from tamoxifen-treated WT and Map3k7CreER mice, assessed with FITC-VAD-FMK. (Right) The proportion of apoptotic VAD-FMK+ splenic DCs. (C and D) BM cells from WT or Map3k7CreER mice (CD45.2+) were cultured with WT CD45.1+ BM cells at a 1:1 ratio in the presence of FLT3L, and 0.5 μM 4-OHT was added at day 7. The distribution (C) and apoptosis (D) of CD45.1 and CD45.1+ populations in the gated CD11c+ cells were analyzed by flow cytometry at the indicated time points. Data are representative of three independent experiments. Data represent the mean ± SEM.

The life span of DCs is determined by signals from pathogens and T cells that are transduced through TLRs, CD40, and receptor activator of nuclear factor-κB (RANK) (9, 10). To test the intrinsic requirement of TAK1 in mediating these prosurvival receptor signals, we generated FLT3L-derived DCs from WT and Map3k7DC BM cells and treated purified DCs with LPS, α-CD40, or RANKL for 2 d. In the absence of treatment, TAK1-deficient DCs had more spontaneous death compared with WT cells. LPS treatment resulted in enhanced survival of WT cells, but this effect was substantially reduced in Map3k7DC DCs. Moreover, TAK1-deficient DCs remained nearly completely unresponsive to the survival effects induced by α-CD40 or RANKL stimulation (SI Appendix, Fig. S2). These findings identify TAK1 as an important sensor that links signaling activities of extracellular receptors and survival of DCs.

TAK1-Dependent Pro- and Antiapoptotic Signaling Mechanisms in DCs.

We dissected the biochemical and molecular mechanisms by which TAK1 regulates DC survival. We first measured the effects of TAK1 deficiency on the activity of NF-κB, which is important for DC survival (50). Constitutive NF-κB activity, assessed by IκBα phosphorylation, was considerably diminished in freshly isolated Map3k7DC splenic DCs (Fig. 4A), indicating a role for TAK1 to mediate NF-κB activation in DCs in vivo. Activity of AKT (also known as protein kinase B, PKB), another antiapoptotic kinase in DCs (51), was also diminished in TAK1-deficient DCs. Accordingly, phosphorylation of the AKT downstream target Foxo1 (Ser256), which exerts a potent inhibitory effect on Foxo1 activity, was decreased (Fig. 4A). The AKT-Foxo axis is important for the expression of the proapoptotic molecule Bim (52). Indeed, Bim expression was up-regulated in TAK1-deficient DCs, whereas the level of the antiapoptotic Bcl2 was not altered (Fig. 4A). In addition, production of reactive oxygen species (ROS), which is under the control of TAK1 signaling in other cell types (53), was undisturbed in TAK1-deficient DCs (SI Appendix, Fig. S3). Therefore, TAK1 regulates the activities of the NF-κB pathway and the AKT-Foxo-Bim signaling axis, and the interplay of these pathways likely contributes to the physiological function of TAK1 in mediating DC survival.

Fig. 4.

Fig. 4.

TAK1 regulates the activities of NF-κB and AKT-Foxo signaling and the expression of apoptotic factors and immune response genes. (A) Western blot analyses of p-IκB, p-AKT, p-Foxo1, Bim, and Bcl-2 expression in splenic DCs from WT and Map3k7DC mice. Numbers below lanes indicate band intensity relative to that of β-actin (loading control). (B) WT and Map3k7CreER mice were treated with tamoxifen for 3 d and RNA of splenic DCs was analyzed for the comparison of expression profiles. A subset of genes differentially regulated in WT and TAK1-deficient DCs (false discovery rate < 0.05) is shown. Normalized gene expression is displayed. (Scale bar shows SD from the mean.) (C) WT and Map3k7CreER mice were treated with tamoxifen for 3 d and RNA of splenic DCs was analyzed for the expression of the indicated genes. Data are representative of three independent experiments. Data represent the mean ± SEM.

To further understand the function of TAK1 in DCs, we used a functional genomics approach to analyze TAK1-dependent gene signature. To obviate compensatory effects due to sustained loss of TAK1, we purified splenic DCs from WT and Map3k7CreER mice after acute tamoxifen treatment and used microarrays to compare their gene expression profiles. A total of 550 probe sets showed equal or greater than twofold change (with false discovery rate <0.05) between WT and TAK1-deficient DCs. TAK1-deficient DCs showed up-regulated levels of several proapoptotic factors including Apaf-1, Bim (encoded by Bcl2l11), Caspase 6, and Gadd45a and reduced expression of transcription factors Nfkb2 and Relb. In addition, expression of several cytokines, chemokines, and their receptors was altered in TAK1-deficient DCs (Fig. 4B). Real-time PCR analysis confirmed the altered expression of selected genes (Fig. 4C). Interestingly, IL-10 and the IL-10 target gene SOCS3 were found to be significantly up-regulated in DCs from tamoxifen-treated Map3k7CreER mice (Fig. 4 B and C). DCs from Map3k7DC mice showed a similar increase in the expression of IL-10 and SOCS3, as well as proapoptotic Bim and Gadd45a (SI Appendix, Fig. S4). Furthermore, elevated STAT3 phosphorylation was observed in DCs from Map3k7DC and tamoxifen-treated Map3k7CreER mice (SI Appendix, Fig. S5), thus indicating an active autocrine IL-10 signaling in TAK1-deficient DCs. Therefore, TAK1 orchestrates a prosurvival program in DCs by regulating expression of apoptotic molecules and further links them with expression of immune response genes.

TAK1 Functions in DCs to Promote T-Cell Priming and Treg Generation.

DCs are the most potent antigen-presenting cells (APCs) to activate naive T cells. To investigate the role of TAK1 signaling in DCs to mediate T-cell–dependent immune responses, we transferred antigen-specific CD8+ T cells from OT-I TCR-transgenic mice (specific for the OVA257–264 peptide) into WT and Map3k7DC mice, followed by immunization with the cognate antigen. T cells isolated from Map3k7DC hosts contained fewer donor-derived antigen-specific T cells (Fig. 5A). We obtained similar results for antigen-specific CD4+ T-cell responses using OT-II TCR-transgenic T-cells (specific for OVA323–339) (Fig. 5B). Moreover, OT-II T cells activated with TAK1-deficient DCs in vitro were substantially impaired in their expansion (SI Appendix, Fig. S6A). These findings demonstrate that TAK1 signaling in DCs is required for the activation of antigen-specific naive T cells.

Fig. 5.

Fig. 5.

TAK1 signaling in DCs is required for T-cell priming and Treg generation. (A and B) Antigen-specific OT-I CD8+ (CD45.1+) (A) or OT-II CD4+ T cells (Thy1.1+) (B) were transferred into WT and Map3k7DC mice followed by antigen immunization in the presence of IFA. At day 4 after immunization, DLN cells were examined for the percentages of donor T cells. (C) Proportion of Foxp3+ nTreg cells among CD4+ T cells in the thymus, spleen, PLNs, and MLNs of WT and Map3k7DC mice. (Right) The proportion of Foxp3+ cells in CD4+ T cells of WT and Map3k7DC mice. (D) WT and Map3k7DC mice were transferred with naive OT-II CD4+ T cells (Thy1.1+) and immunized as described in B, and DLN cells were examined for the percentage of Foxp3+ cells among donor cells. (E) After adoptive transfer of naive OT-II CD4+ T cells, WT and Map3k7DC mice were fed with water supplemented with OVA protein. Five days later, the spleen and PLN cells were examined for the percentages of donor CD4+ T cells (Thy1.1+) and of Foxp3+ cells among donor cells. Data are representative of two to four independent experiments. Data represent the mean ± SEM.

In addition to T-cell priming, DCs have been recently shown to promote Treg cell generation (17, 5457). Treg cells either develop in the thymus, known as naturally occurring Treg (nTreg) cells, or are induced from naive T cells in the periphery under subimmunogenic antigen stimulation, known as induced Treg (iTreg) cells (58). Whereas nTreg cells were undisturbed in the thymus of Map3k7DC mice, they were considerably reduced in peripheral lymph organs (Fig. 5C). Given the critical importance of TAK1 signaling in Treg cell generation (37, 38), it remained possible that the reduction of nTreg cells could result from an off-target deletion of TAK1 in T cells in Map3k7DC mice. However, in mixed chimeras as described above (Fig. 1D), the nTreg population derived from Map3k7DC BM cells developed normally (SI Appendix, Fig. S7), indicating a non-cell–autonomous defect triggered by the abnormality of DCs.

We next tested whether TAK1 regulated induction of iTreg cells. We adoptively transferred naive OT-II CD4+ T cells into WT and Map3k7DC hosts, followed by s.c. antigen immunization or administration of the antigen in the drinking water. In both models, donor cells developed into a considerably smaller Foxp3+ population in Map3k7DC recipients relative to WT hosts (Fig. 5 D and E), indicating a role of TAK1 in DCs to mediate induction of antigen-specific iTreg cells. This result was further confirmed by the impaired ability of TAK1-deficient DCs to mediate iTreg induction in vitro (SI Appendix, Fig. S6B). We conclude that TAK1 function in DCs is critical for both naive T-cell priming and Treg generation.

TAK1 Deficiency in DCs Disrupts Homeostasis of T Cells and Myeloid Cells.

How DCs affect homeostasis of the immune system remains incompletely understood. We therefore tested whether defective DC survival in Map3k7DC mice influenced T-cell homeostasis. Map3k7DC mice had reduced proportions of T cells, especially CD8+ T cells, in the thymus, spleen, and peripheral lymph nodes (PLNs) (Fig. 6A). Moreover, splenic T cells from Map3k7DC mice were partially impaired to produce IFN-γ and IL-2 upon acute polyclonal stimulation (Fig. 6B), indicative of altered homeostasis of T cells in the steady state.

Fig. 6.

Fig. 6.

Map3k7DC mice show diminished conventional T cells but expanded myeloid cells. (A) Distribution of CD4+ and CD8+ T cells in the thymus, spleen, and PLNs of WT and Map3k7DC mice. (Right) The proportion of CD4+ and CD8+ T cells in WT and Map3k7DC mice. (B) Expression of IFN-γ and IL-2 in splenic CD4+ and CD8+ T cells was determined by intracellular cytokine staining after stimulation with PMA and ionomycin. (C) Spleen sections from WT and Map3k7DC mice were analyzed by H&E, Masson's trichrome, and reticulin staining for fibrosis and by anti-CD3 and anti-Mac2 immunohistochemistry. (D) Flow cytometry of myeloid cells (Gr1+CD11b+) in the spleen, PLNs, and MLNs of WT and Map3k7DC mice. Data are representative of at least three independent experiments. Data represent the mean ± SEM.

Despite diminished DC and T-cell populations, the spleen and PLN sizes of Map3k7DC mice were markedly increased (SI Appendix, Fig. S8A). Histological analysis showed that the structure of the secondary lymphoid organs was disrupted, associated with altered distribution of CD3+ T cells. In addition, the spleen of Map3k7DC mice showed severe fibrosis, detected by trichrome and reticulin staining (Fig. 6C). Moreover, increased numbers of Mac2-reactive myeloid cells were evident in the mutant spleen and PLNs (Fig. 6C and SI Appendix, Fig. S8B). Flow cytometry analysis of spleen, MLNs, and PLNs in Map3k7DC mice revealed a marked increase in Gr1+CD11b+ myeloid cells that likely corresponded to neutrophils and Gr1+ inflammatory monocytes (Fig. 6D) (59). Analysis of additional markers in Map3k7DC lymphoid organs confirmed the expansion of neutrophils (Ly6G+Ly6C+) and monocytes (CD115+CD11b+), with the latter population mainly consisting of the Ly6C+ inflammatory subset rather than the Ly6C subset (SI Appendix, Fig. S9) (59). Flow cytometry and blood counts identified a similar shift toward neutrophils and monocytes in peripheral blood of Map3k7DC mice (SI Appendix, Fig. S10). Further, myeloid cell infiltration was also observed in various nonlymphoid organs from Map3k7DC mice, including the liver, lung, kidney, and colon (SI Appendix, Fig. S11). In contrast, Map3k7DC BM contained largely normal populations of neutrophils and monocytes (SI Appendix, Fig. S12). These results collectively indicate the development of a myeloid proliferative syndrome in Map3k7DC mice.

Myeloid Proliferation Results from Loss of TAK1 in DCs Independently of Lymphocytes.

We dissected the cellular mechanisms that caused defective homeostasis of myeloid cells in Map3k7DC mice. Such a disorder could reflect a direct role of TAK1 in myeloid cells (for example, by nonspecific deletion) or could be due to dysregulated functions of T cells, especially Treg cells, or triggered by a mechanism that directly sensed the abnormality of DCs in vivo. To distinguish these possibilities, we analyzed the mixed BM chimeras composed of Map3k7DC donor and spike cells, as described above (Fig. 1D). No myeloid expansion was observed in the mixed chimeras that contained normal DC populations derived from the spike cells (Fig. 7A), suggesting that a normal DC population is important to prevent myeloid expansion.

Fig. 7.

Fig. 7.

Myeloid expansion in Map3k7DC mice results from altered DC–myeloid cell crosstalk independently of lymphocytes. (A) Flow cytometry of spike BM-derived and WT or Map3k7DC BM-derived myeloid cells in the mixed chimeras (Fig. 1D). (B) Flow cytometry of myeloid cells in the spleen, PLNs, MLNs, and thymus of Rag1−/− and Map3k7DC Rag1−/− mice. (C) Analysis of FLT3L levels in the serum from WT and Map3k7DC mice. Each symbol represents an individual mouse and short horizontal lines indicate the mean.

We next determined whether myeloid proliferation in Map3k7DC mice resulted from altered T-cell function or tolerance. In particular, depletion of DCs has been implicated in the development of autoimmune diseases that is dependent upon dysregulated autoreactive T cells and/or defective Treg-mediated immune suppression (17, 20). However, T cells in Map3k7DC mice did not appear to be activated, indicated by the largely normal expression of the homeostasis markers CD62L and CD44 and the absence of elevated serum autoantibodies (SI Appendix, Fig. S13). To further dissect the contribution of T cells, we crossed Map3k7DC mice onto the alymphoid Rag1−/− background. Notably, Rag1 deficiency elevated the population of myeloid cells compared with that in immunocompetent WT mice (Fig. 7B; compare with Fig. 6D), probably triggered by the lymphopenic environment. However, deletion of TAK1 in DCs on the same Rag1−/− background further expanded the myeloid populations including neutrophils and Ly6C+ inflammatory monocytes (Fig. 7B and SI Appendix, Fig. S14). Therefore, defective control of lymphoid and myeloid populations in Map3k7DC mice was uncoupled after depletion of lymphocytes, which revealed an important role for TAK1 signaling in DCs to maintain myeloid homeostasis in a manner independently of lymphocytes.

We further explored the mechanism by which lack of DCs triggered myeloid expansion. Depletion of DCs has recently been shown to increase the levels of FLT3L, a key growth factor for myeloid cell generation (21, 60). Indeed, the serum concentration of FLT3L was significantly elevated in Map3k7DC mice compared with the WT controls (Fig. 7C). Thus, FLT3L was up-regulated in the absence of a normal DC population that likely contributed to the abnormal myeloid expansion.

Discussion

Homeostasis of DCs is essential for the generation of adaptive immunity and the maintenance of immune tolerance. Here we have identified TAK1 as a critical regulator of DC survival and homeostasis. Using DC-specific and acute deletion systems, we have established that TAK1 is required to maintain DC populations by promoting the survival of DCs and generation of their precursors. TAK1 integrates prosurvival receptor signals and in turn activates NF-κB and AKT-Foxo pathways while controlling expression of Bim and other proapoptotic molecules. TAK1 signaling in DCs facilitates T-cell priming and generation of Treg cells, indicating a role of TAK1 to mediate both the immunogenic and tolerogenic activities of DCs. Moreover, loss of this active control mechanism in DCs has profound effects on the homeostasis of T cells and myeloid populations, and additional analysis reveals that TAK1 acts in DCs to control myeloid cell-mediated inflammation in a lymphocyte-independent manner. Therefore, a TAK1-dependent checkpoint in DC survival orchestrates immune function and homeostasis.

DC homeostasis is dependent upon the interplay of three main processes: development from precursors, division in situ, and apoptotic cell death. Combining tissue-specific and inducible deletion systems, we found that both acute and sustained loss of TAK1 resulted in an extensive apoptotic death of DCs. This result contrasted with the observations in neuronal cells in which short-term inhibition of TAK1 protects neurons from apoptosis, whereas prolonged inhibition is not protective (61). Therefore, our findings indicate a direct effect of TAK1 to actively maintain DC survival. Moreover, TAK1 contributed to DC development by maintaining the MDP population. However, TAK1 was dispensable for DC division, which differed from a critical requirement of TAK1 in clonal expansion of lymphocytes (33, 3740). Therefore, TAK1 has a unique role in DCs to selectively regulate their survival and development.

Among DC populations, the CD8+ and CD8 cDC subsets efficiently present antigens to CD8+ and CD4+ T cells, respectively (62). The loss of CD8+ cDCs was more profound relative to the CD8 subset in Map3k7DC mice. However, the remaining CD8 cDCs probably exhibited qualitative changes, reflected by their up-regulated CD11b levels on a per cell basis (Fig. 1A). Accordingly, both CD8+ and CD4+ T-cell responses were diminished in response to antigen challenge, in agreement with a requirement of TAK1 in the contact hypersensitivity response (63). Deletion of TAK1 was also incompatible with the survival of pDCs, and a more severe reduction of pDCs upon acute deletion of TAK1 than DC-specific TAK1 deficiency probably reflected the significant but incomplete deletion efficiency in pDCs in the CD11c-Cre line (42). Finally, the CD103+ DCs in the nonlymphoid organs were as sensitive to TAK1 deficiency as CD8+ cDCs in the lymphoid organs, which provides added genetic evidence supporting the notion that these two populations are developmentally and functionally linked (8, 4345). Therefore, TAK1 is critically required for the survival of all major DC subsets.

The molecular mechanisms in DC survival remain to be fully established. Among the best understood pathways for the survival of DCs are the Bcl-2 family members, as previous work has implicated a pivotal role for Bcl-2, Bcl-xL, and Bim in DC survival (9). How these factors are regulated by the intracellular signaling network is not well defined. Here we found an elevated Bim expression in TAK1-deficient DCs, which likely resulted from the altered AKT-Foxo signaling axis (52). In addition, TAK1 was required for NF-κB activity in DCs. Moreover, TAK1-deficient DCs showed an enhanced autocrine IL-10 signaling, which likely further contributed to the increased cell death, given the important role of IL-10 in promoting DC apoptosis (64). Taken together, TAK1 affects signal transduction and gene expression of both proapoptotic and antiapoptotic pathways and further links them with expression of immune response genes, thereby orchestrating a prosurvival program in DCs. Consistent with this notion, TAK1 integrates multiple prosurvival receptor signals in DCs.

As the most potent type of APCs, DCs are essential for the initiation and propagation of adaptive immune responses, but how DCs are involved in the maintenance of nTreg cell homeostasis remains under debate. Map3k7DC mice contained a greatly reduced number of nTreg cells in the periphery under steady state. These results differ from the earlier reports excluding a role for DCs to regulate nTreg cells (20, 21), but are consistent with more recent studies implicating a requirement for DCs to maintain the nTreg population (17, 27, 65). Interestingly, despite defective iTreg generation and nTreg homeostasis, T cells in Map3k7DC mice exhibited no signs of prominent activation. Instead, we found reduced populations of CD4+ and CD8+ T cells and diminished production of IFN-γ and IL-2 in T cells from Map3k7DC mice. Our results are in agreement with a recent DC ablation study in lupus-prone MRL/lpr mice that reveals an important role for DCs to maintain conventional T cells and their ability to produce IFN-γ (65). Thus, TAK1 acts in DCs to orchestrate T-cell priming and homeostasis but not self-tolerance.

Unexpectedly, deficiency of TAK1 in DCs results in a prominent myeloid proliferative disease characterized by expansion of neutrophils and Ly6C+ inflammatory monocytes, suggesting that DCs provide a negative signal to restrain myeloid expansion and inflammation under physiological conditions. The myeloid defects largely recapitulate the phenotypes of mice with constitutive loss of cDCs (20, 21), although the precise mechanism involved remains in debate. In particular, dysregulated autoreactive T cells and impaired Treg cells have been suggested as important mechanisms for excessive inflammation (17, 20). However, deletion of TAK1 in DCs disrupted myeloid homeostasis even on a Rag1−/− background. Although our results do not exclude the contribution of Treg or conventional T cells, they provide definitive evidence that DC signaling can directly impact myeloid cell homeostasis independently of lymphocytes, thus establishing a previously unappreciated mechanism of DC-mediated active control of myeloid expansion and inflammation. The crosstalk between DCs and myeloid cells is likely mediated by FLT3L, given the key role of FLT3L in myeloid cell expansion (21, 60), although this action does not appear to affect myeloid populations in the BM. Recently, expansion of myeloid populations and increases of serum FLT3L levels have been reported in mice with DC-selective deletion of A20, a negative regulator of NF-κB signaling (66, 67). However, unlike DCs lacking TAK1, A20-deficient DCs show enhanced responses to CD40 and RANK-mediated survival signals and trigger profound activation of T cells and B cells and development of autoimmunity (66, 67). Therefore, immune homeostasis under steady state is dependent upon molecular signals in DCs, especially a delicate threshold of NF-κB signaling.

The life span of DCs is important for the strength of adaptive immunity (10, 68, 69). Our results have identified a TAK1-dependent checkpoint in DC survival that influences the magnitude and quality of adaptive immune responses, as well as homeostasis of the immune system. In Drosophila TAK1 has a crucial function in mounting host defense reactions (30, 31). Not only does the mammalian immune system retain this function for innate defense responses (32, 33), but also DCs have evolved to acquire this evolutionarily conserved pathway to regulate their life span and further imprint innate and adaptive immunity. This molecular pathway in DCs may be explored for the development of DC-based therapeutic strategies.

Materials and Methods

Mice and BM Chimeras.

C57BL/6, CD45.1, Thy1.1, Rag1−/−, OT-I, and OT-II mice were purchased from the Jackson Laboratory. Foxp3gfp mice were kindly provided by A. Rudensky (Memorial Sloan-Kettering Cancer Center, New York, NY) (70). Floxed Map3k7 (Map3k7flox/null) mice were bred with CD11c-Cre or Rosa26-Cre-ERT2 mice (37, 42, 46) and have been backcrossed to the C57BL/6 background for at least eight generations. WT controls were in the same genetic background and included Cre+ mice to account for Cre effects. For in vivo tamoxifen treatment, WT and Map3k7CreER mice were injected i.p. with 1 mg per mouse of tamoxifen (Sigma) for 3 consecutive days before further analysis. For mixed BM experiments, BM cells from WT or Map3k7DC CD45.2.2+ mice were mixed with cells from CD45.1.2+ mice at a 1:1 ratio and transferred into lethally irradiated (11 Gy, split dose) CD45.1.1+ mice, as described previously (71). Animal protocols were approved by Institutional Animal Care and Use Committee of St. Jude Children's Research Hospital.

Cell Purification and Cultures.

Mouse spleens were digested with Collagenase D (Roche), and DCs (CD11c+TCRCD19DX5) were sorted on a Reflection cell sorter (i-Cyt). DCs from nonlymphoid organs were isolated as described in ref. 8. Lymphocytes were sorted for naive T cells (CD4+CD62LhiCD44loCD25). For DC culture, BM cells were cultured in medium containing 10% FBS and mouse FLT3L (200 ng/mL); CD11c+ cells were sorted at day 7, washed extensively, and treated with 10 μg/mL LPS, 10 μg/mL α-CD40, 1 μg/mL RANKL, or mock for 2 d for the analysis of cell survival. For the mixed culture, WT or Map3k7CreER BM cells were cultured with CD45.1+ BM cells at a 1:1 ratio in the presence of FLT3L. From day 7, 0.5 μM 4-OHT (Sigma-Aldrich) was added to each well to delete TAK1. For DC–T-cell coculture, splenic DCs and OT-II T cells (Foxp3GFP) were mixed at a 1:10 ratio in the presence of 1 μg/mL OVA323–339 peptide and cultured for 5 d, followed by flow cytometry analysis.

Antigen Challenge.

Antigen-specific T cells from OT-I (CD45.1+; 1 × 106) or OT-II TCR-transgenic mice (Thy1.1+; 2 × 106) were sorted and transferred into WT and Map3k7DC mice intravenously. Twenty-four hours later, the mice were injected s.c. with OVA257–264 or OVA323–339 (5 μg) in the presence of incomplete Freund's adjuvant (IFA; Difco). At day 4 after immunization, draining lymph node (DLN) cells were isolated for further analyses (71). In the model of oral antigen stimulation, after adoptive transfer of naive OT-II T cells, mice were fed with water supplemented with 20 mg/mL OVA protein (Grade VI; Sigma-Aldrich) for a total of 5 d (71).

Flow Cytometry.

Flow cytometry was performed as described previously (46, 71, 72). For intracellular cytokine detection, the cells were stimulated for 5 h with phorbol 12-myristate 13-acetate (PMA) and ionomycin in the presence of monensin before staining according to the manufacturer's instructions (BD Biosciences). For caspase activity detection, cells were stained with CaspACE FITC-VAD-FMK in situ marker according to the manufacturer's instructions (Promega). For TUNEL staining, freshly isolated splenocytes were used according to the protocol supplied (BD Biosciences). ROS were measured by incubation for 30 min at 37 °C with 10 μM CM-H2DCFDA (Invitrogen). For BrdU labeling, mice were injected i.p. with 1 mg BrdU (5-bromo-2-deoxyuridine); 24 h later, mice were killed and BrdU incorporation was analyzed according to the manufacturer's instructions (BD Biosciences) (46).

RNA and Protein Analyses.

Real-time PCR analysis was done with primers and probe sets from Applied Biosystems as described in refs. 46 and 71. Microarray analysis was performed as described previously (46, 72), and the results have been deposited in the GEO database (GSE34417). The heat maps were generated to show relative expression of genes after z-transformation, which subtracts the mean of each from each individual value for that gene and then divides by the SD. This method sets all of the genes into the same scale with a mean of 0 and SD of 1. The scale bar shows the distance from the mean expression as SD units. Immunoblot was performed as described in refs. 46 and 71, using the following antibodies: p-STAT3, p-AKT (Ser473), p-Foxo1 (Ser256), and p-IκBα (all from Cell Signaling Technology); Bcl-2 (Santa Cruz); Bim (Abcam); and β-actin (Sigma). The serum levels of FLT3L and autoantibodies were measured by ELISA as described in ref. 73.

Histopathology and Immunohistochemistry.

The spleen and PLNs were processed as described previously (72) and stained with hematoxylin and eosin (H&E), Masson's trichrome, and reticulin stains for the visualization of collagen deposition. Immunohistochemistry was performed as described previously (72). Peripheral blood samples were collected for blood counts.

Statistical Analysis.

P values were calculated using Student's t test. P values <0.05 were considered significant.

Supplementary Material

Supporting Information

Acknowledgments

This work is supported by the National Institutes of Health (R01 NS064599 and K01 AR053573), the Cancer Research Institute, the National Multiple Sclerosis Society (RG4180-A-1), and the Hartwell Foundation.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission. K.M.M. is a guest editor invited by the Editorial Board.

Data deposition: The microarray results reported in this paper have been deposited in the Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE34417).

See Author Summary on page 1834.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1115635109/-/DCSupplemental.

References

  • 1.Steinman RM. Lasker Basic Medical Research Award. Dendritic cells: Versatile controllers of the immune system. Nat Med. 2007;13:1155–1159. doi: 10.1038/nm1643. [DOI] [PubMed] [Google Scholar]
  • 2.Liu K, Nussenzweig MC. Origin and development of dendritic cells. Immunol Rev. 2010;234:45–54. doi: 10.1111/j.0105-2896.2009.00879.x. [DOI] [PubMed] [Google Scholar]
  • 3.Helft J, Ginhoux F, Bogunovic M, Merad M. Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice. Immunol Rev. 2010;234:55–75. doi: 10.1111/j.0105-2896.2009.00885.x. [DOI] [PubMed] [Google Scholar]
  • 4.Watowich SS, Liu YJ. Mechanisms regulating dendritic cell specification and development. Immunol Rev. 2010;238:76–92. doi: 10.1111/j.1600-065X.2010.00949.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Liu K, et al. In vivo analysis of dendritic cell development and homeostasis. Science. 2009;324:392–397. doi: 10.1126/science.1170540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kabashima K, et al. Intrinsic lymphotoxin-beta receptor requirement for homeostasis of lymphoid tissue dendritic cells. Immunity. 2005;22:439–450. doi: 10.1016/j.immuni.2005.02.007. [DOI] [PubMed] [Google Scholar]
  • 7.Liu K, et al. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat Immunol. 2007;8:578–583. doi: 10.1038/ni1462. [DOI] [PubMed] [Google Scholar]
  • 8.Ginhoux F, et al. The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med. 2009;206:3115–3130. doi: 10.1084/jem.20091756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen M, Wang J. Programmed cell death of dendritic cells in immune regulation. Immunol Rev. 2010;236:11–27. doi: 10.1111/j.1600-065X.2010.00916.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hou WS, Van Parijs L. A Bcl-2-dependent molecular timer regulates the lifespan and immunogenicity of dendritic cells. Nat Immunol. 2004;5:583–589. doi: 10.1038/ni1071. [DOI] [PubMed] [Google Scholar]
  • 11.Chen M, Huang L, Wang J. Deficiency of Bim in dendritic cells contributes to overactivation of lymphocytes and autoimmunity. Blood. 2007;109:4360–4367. doi: 10.1182/blood-2006-11-056424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen M, Huang L, Shabier Z, Wang J. Regulation of the lifespan in dendritic cell subsets. Mol Immunol. 2007;44:2558–2565. doi: 10.1016/j.molimm.2006.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jung S, et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity. 2002;17:211–220. doi: 10.1016/s1074-7613(02)00365-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040. [DOI] [PubMed] [Google Scholar]
  • 15.Chen M, et al. Dendritic cell apoptosis in the maintenance of immune tolerance. Science. 2006;311:1160–1164. doi: 10.1126/science.1122545. [DOI] [PubMed] [Google Scholar]
  • 16.Stranges PB, et al. Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity. 2007;26:629–641. doi: 10.1016/j.immuni.2007.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Darrasse-Jèze G, et al. Feedback control of regulatory T cell homeostasis by dendritic cells in vivo. J Exp Med. 2009;206:1853–1862. doi: 10.1084/jem.20090746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Swee LK, Bosco N, Malissen B, Ceredig R, Rolink A. Expansion of peripheral naturally occurring T regulatory cells by Fms-like tyrosine kinase 3 ligand treatment. Blood. 2009;113:6277–6287. doi: 10.1182/blood-2008-06-161026. [DOI] [PubMed] [Google Scholar]
  • 19.O'Keeffe M, et al. Fms-like tyrosine kinase 3 ligand administration overcomes a genetically determined dendritic cell deficiency in NOD mice and protects against diabetes development. Int Immunol. 2005;17:307–314. doi: 10.1093/intimm/dxh210. [DOI] [PubMed] [Google Scholar]
  • 20.Ohnmacht C, et al. Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity. J Exp Med. 2009;206:549–559. doi: 10.1084/jem.20082394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Birnberg T, et al. Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome. Immunity. 2008;29:986–997. doi: 10.1016/j.immuni.2008.10.012. [DOI] [PubMed] [Google Scholar]
  • 22.Brocker T. Survival of mature CD4 T lymphocytes is dependent on major histocompatibility complex class II-expressing dendritic cells. J Exp Med. 1997;186:1223–1232. doi: 10.1084/jem.186.8.1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gruber A, Brocker T. MHC class I-positive dendritic cells (DC) control CD8 T cell homeostasis in vivo: T cell lymphopenia as a prerequisite for DC-mediated homeostatic proliferation of naive CD8 T cells. J Immunol. 2005;175:201–206. doi: 10.4049/jimmunol.175.1.201. [DOI] [PubMed] [Google Scholar]
  • 24.Zaft T, Sapoznikov A, Krauthgamer R, Littman DR, Jung S. CD11chigh dendritic cell ablation impairs lymphopenia-driven proliferation of naive and memory CD8+ T cells. J Immunol. 2005;175:6428–6435. doi: 10.4049/jimmunol.175.10.6428. [DOI] [PubMed] [Google Scholar]
  • 25.Hochweller K, et al. Dendritic cells control T cell tonic signaling required for responsiveness to foreign antigen. Proc Natl Acad Sci USA. 2010;107:5931–5936. doi: 10.1073/pnas.0911877107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Suffner J, et al. Dendritic cells support homeostatic expansion of Foxp3+ regulatory T cells in Foxp3.LuciDTR mice. J Immunol. 2010;184:1810–1820. doi: 10.4049/jimmunol.0902420. [DOI] [PubMed] [Google Scholar]
  • 27.Bar-On L, Birnberg T, Kim KW, Jung S. Dendritic cell-restricted CD80/86 deficiency results in peripheral regulatory T-cell reduction but is not associated with lymphocyte hyperactivation. Eur J Immunol. 2011;41:291–298. doi: 10.1002/eji.201041169. [DOI] [PubMed] [Google Scholar]
  • 28.Bar-On L, Jung S. Defining dendritic cells by conditional and constitutive cell ablation. Immunol Rev. 2010;234:76–89. doi: 10.1111/j.0105-2896.2009.00875.x. [DOI] [PubMed] [Google Scholar]
  • 29.Huang G, Shi LZ, Chi H. Regulation of JNK and p38 MAPK in the immune system: Signal integration, propagation and termination. Cytokine. 2009;48:161–169. doi: 10.1016/j.cyto.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Vidal S, et al. Mutations in the Drosophila dTAK1 gene reveal a conserved function for MAPKKKs in the control of rel/NF-kappaB-dependent innate immune responses. Genes Dev. 2001;15:1900–1912. doi: 10.1101/gad.203301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Silverman N, et al. Immune activation of NF-kappaB and JNK requires Drosophila TAK1. J Biol Chem. 2003;278:48928–48934. doi: 10.1074/jbc.M304802200. [DOI] [PubMed] [Google Scholar]
  • 32.Shim JH, et al. TAK1, but not TAB1 or TAB2, plays an essential role in multiple signaling pathways in vivo. Genes Dev. 2005;19:2668–2681. doi: 10.1101/gad.1360605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sato S, et al. Essential function for the kinase TAK1 in innate and adaptive immune responses. Nat Immunol. 2005;6:1087–1095. doi: 10.1038/ni1255. [DOI] [PubMed] [Google Scholar]
  • 34.Hasegawa M, et al. A critical role of RICK/RIP2 polyubiquitination in Nod-induced NF-kappaB activation. EMBO J. 2008;27:373–383. doi: 10.1038/sj.emboj.7601962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kim JY, Omori E, Matsumoto K, Núñez G, Ninomiya-Tsuji J. TAK1 is a central mediator of NOD2 signaling in epidermal cells. J Biol Chem. 2008;283:137–144. doi: 10.1074/jbc.M704746200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Qin J, et al. TLR8-mediated NF-kappaB and JNK activation are TAK1-independent and MEKK3-dependent. J Biol Chem. 2006;281:21013–21021. doi: 10.1074/jbc.M512908200. [DOI] [PubMed] [Google Scholar]
  • 37.Wan YY, Chi H, Xie M, Schneider MD, Flavell RA. The kinase TAK1 integrates antigen and cytokine receptor signaling for T cell development, survival and function. Nat Immunol. 2006;7:851–858. doi: 10.1038/ni1355. [DOI] [PubMed] [Google Scholar]
  • 38.Sato S, et al. TAK1 is indispensable for development of T cells and prevention of colitis by the generation of regulatory T cells. Int Immunol. 2006;18:1405–1411. doi: 10.1093/intimm/dxl082. [DOI] [PubMed] [Google Scholar]
  • 39.Liu HH, Xie M, Schneider MD, Chen ZJ. Essential role of TAK1 in thymocyte development and activation. Proc Natl Acad Sci USA. 2006;103:11677–11682. doi: 10.1073/pnas.0603089103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schuman J, et al. A critical role of TAK1 in B-cell receptor-mediated nuclear factor kappaB activation. Blood. 2009;113:4566–4574. doi: 10.1182/blood-2008-08-176057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tang M, et al. TAK1 is required for the survival of hematopoietic cells and hepatocytes in mice. J Exp Med. 2008;205:1611–1619. doi: 10.1084/jem.20080297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Caton ML, Smith-Raska MR, Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J Exp Med. 2007;204:1653–1664. doi: 10.1084/jem.20062648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Varol C, et al. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009;31:502–512. doi: 10.1016/j.immuni.2009.06.025. [DOI] [PubMed] [Google Scholar]
  • 44.Bedoui S, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009;10:488–495. doi: 10.1038/ni.1724. [DOI] [PubMed] [Google Scholar]
  • 45.Bogunovic M, et al. Origin of the lamina propria dendritic cell network. Immunity. 2009;31:513–525. doi: 10.1016/j.immuni.2009.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yang K, Neale G, Green DR, He W, Chi H. The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function. Nat Immunol. 2011;12:888–897. doi: 10.1038/ni.2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fogg DK, et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science. 2006;311:83–87. doi: 10.1126/science.1117729. [DOI] [PubMed] [Google Scholar]
  • 48.Varol C, et al. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J Exp Med. 2007;204:171–180. doi: 10.1084/jem.20061011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Waskow C, et al. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat Immunol. 2008;9:676–683. doi: 10.1038/ni.1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ouaaz F, Arron J, Zheng Y, Choi Y, Beg AA. Dendritic cell development and survival require distinct NF-kappaB subunits. Immunity. 2002;16:257–270. doi: 10.1016/s1074-7613(02)00272-8. [DOI] [PubMed] [Google Scholar]
  • 51.Del Prete A, et al. Defective dendritic cell migration and activation of adaptive immunity in PI3Kgamma-deficient mice. EMBO J. 2004;23:3505–3515. doi: 10.1038/sj.emboj.7600361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fu Z, Tindall DJ. FOXOs, cancer and regulation of apoptosis. Oncogene. 2008;27:2312–2319. doi: 10.1038/onc.2008.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lam CR, et al. TAK1 regulates SCF expression to modulate PKBα activity that protects keratinocytes from ROS-induced apoptosis. Cell Death Differ. 2011;18:1120–1129. doi: 10.1038/cdd.2010.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yamazaki S, et al. Dendritic cells are specialized accessory cells along with TGF- for the differentiation of Foxp3+ CD4+ regulatory T cells from peripheral Foxp3 precursors. Blood. 2007;110:4293–4302. doi: 10.1182/blood-2007-05-088831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yamazaki S, et al. CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells. J Immunol. 2008;181:6923–6933. doi: 10.4049/jimmunol.181.10.6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Coombes JL, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–1764. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sun CM, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–1785. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: More of the same or a division of labor? Immunity. 2009;30:626–635. doi: 10.1016/j.immuni.2009.05.002. [DOI] [PubMed] [Google Scholar]
  • 59.Auffray C, Sieweke MH, Geissmann F. Blood monocytes: Development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009;27:669–692. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
  • 60.Hochweller K, et al. Homeostasis of dendritic cells in lymphoid organs is controlled by regulation of their precursors via a feedback loop. Blood. 2009;114:4411–4421. doi: 10.1182/blood-2008-11-188045. [DOI] [PubMed] [Google Scholar]
  • 61.Neubert M, Ridder DA, Bargiotas P, Akira S, Schwaninger M. Acute inhibition of TAK1 protects against neuronal death in cerebral ischemia. Cell Death Differ. 2011;18:1521–1530. doi: 10.1038/cdd.2011.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.den Haan JM, Lehar SM, Bevan MJ. CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo. J Exp Med. 2000;192:1685–1696. doi: 10.1084/jem.192.12.1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhao YG, Wang Y, Hao W, Wan YY. An essential role for TAK1 in the contact hypersensitivity response. Cell Mol Immunol. 2011;8:315–324. doi: 10.1038/cmi.2011.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chang WL, et al. Exposure of myeloid dendritic cells to exogenous or endogenous IL-10 during maturation determines their longevity. J Immunol. 2007;178:7794–7804. doi: 10.4049/jimmunol.178.12.7794. [DOI] [PubMed] [Google Scholar]
  • 65.Teichmann LL, et al. Dendritic cells in lupus are not required for activation of T and B cells but promote their expansion, resulting in tissue damage. Immunity. 2010;33:967–978. doi: 10.1016/j.immuni.2010.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kool M, et al. The ubiquitin-editing protein A20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity. Immunity. 2011;35:82–96. doi: 10.1016/j.immuni.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 67.Hammer GE, et al. Expression of A20 by dendritic cells preserves immune homeostasis and prevents colitis and spondyloarthritis. Nat Immunol. 2011;12:1184–1193. doi: 10.1038/ni.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nopora A, Brocker T. Bcl-2 controls dendritic cell longevity in vivo. J Immunol. 2002;169:3006–3014. doi: 10.4049/jimmunol.169.6.3006. [DOI] [PubMed] [Google Scholar]
  • 69.Josien R, et al. TRANCE, a tumor necrosis factor family member, enhances the longevity and adjuvant properties of dendritic cells in vivo. J Exp Med. 2000;191:495–502. doi: 10.1084/jem.191.3.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Fontenot JD, et al. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 2005;22:329–341. doi: 10.1016/j.immuni.2005.01.016. [DOI] [PubMed] [Google Scholar]
  • 71.Huang G, Wang Y, Shi LZ, Kanneganti TD, Chi H. Signaling by the phosphatase MKP-1 in dendritic cells imprints distinct effector and regulatory T cell fates. Immunity. 2011;35:45–58. doi: 10.1016/j.immuni.2011.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Shi LZ, et al. HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011;208:1367–1376. doi: 10.1084/jem.20110278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chi H, et al. Dynamic regulation of pro- and anti-inflammatory cytokines by MAPK phosphatase 1 (MKP-1) in innate immune responses. Proc Natl Acad Sci USA. 2006;103:2274–2279. doi: 10.1073/pnas.0510965103. [DOI] [PMC free article] [PubMed] [Google Scholar]
Proc Natl Acad Sci U S A. 2012 Feb 7;109(6):1834–1835.

Author Summary

Author Summary

Professional antigen-presenting cells display portions of foreign invaders, or antigens, thus playing an important role in initiating immune responses to infections. Dendritic cells are the most potent of those cells and are specialized to capture, process, and present antigens to T cells, white blood cells critical for the immune response. In contrast, the involvement of dendritic cells in maintaining the homeostasis of the immune system in the absence of infection remains poorly understood. Here, we show that transforming growth factor beta-activated kinase 1 (TAK1), an enzyme with important immune functions, regulates the survival of dendritic cells and plays a key role in the homeostasis and function of the immune system.

TAK1 is one of the immune system's most widely used kinases, enzymes that modify proteins by adding phosphate molecules (1). The essential role of TAK1 in immune defense was first demonstrated in the fruit fly and later confirmed in the mouse. TAK1 is an essential component of the signaling that takes place when T cells are presented with antigens and plays a role in orchestrating immune functions (2). The involvement of TAK1 in dendritic cell homeostasis and function under physiological conditions remains undefined.

Dendritic cells are continuously replenished by precursors present in the bone marrow and are further sustained by mechanisms that promote their survival and proliferation (3). To explore the roles of TAK1 in dendritic cells, we used a “loss-of-function” genetic approach. That is, we deleted the TAK1 gene specifically in dendritic cells. Loss of TAK1 depleted the majority of dendritic cells in all tissues examined, which was mainly ascribed to markedly elevated apoptosis (i.e., programmed cell death). In addition, TAK1 contributed to dendritic cell development by promoting the generation and survival of dendritic cell precursors in the bone marrow. By contrast, deletion of TAK1 did not affect the proliferation of dendritic cells. Using an inducible system to acutely delete TAK1 in mature dendritic cells, we further showed that maintenance of dendritic cells in vivo depended on TAK1. These results establish a crucial anti-apoptotic function for TAK1 in dendritic cells.

We next determined the molecular and signaling mechanisms by which TAK1 regulates dendritic cell survival. We found that TAK1 was required to convey prosurvival signals from receptors; in general, receptors are located on the surface of cells and bind to specific molecules, thereafter conveying information into the cell. Specifically, TAK1 had a role in integrating signals from certain receptors, namely Toll-like receptors, CD40 and RANK, to promote dendritic cell survival. This function depended on the ability of TAK1 to mediate the activation of specific downstream pathways including NF-κB and AKT-Foxo signaling. Moreover, TAK1 established a gene-expression signature by controlling the expression of apoptotic regulators and immune-response genes. Therefore, TAK1 bridges prosurvival receptors and downstream signaling pathways and gene expression, thereby orchestrating a prosurvival program in dendritic cells.

To investigate the role of TAK1 signaling in dendritic cells to mediate T-cell–dependent immune responses, we used both in vitro and in vivo systems. Compared with TAK1-sufficient dendritic cells, TAK1-deficient dendritic cells were impaired in driving T-cell expansion in vitro and in vivo. Moreover, TAK1 signaling in dendritic cells was required for the generation of regulatory T cells, a specialized T-cell subset critical for immune suppression (i.e., down-regulating immune responses when they are not necessary). Finally, in the absence of foreign invaders, loss of TAK1 in dendritic cells diminished the population of conventional T cells and their ability to produce immune signaling molecules known as cytokines. Altogether, TAK1 acts in dendritic cells to promote the homeostasis and function of T cells.

Surprisingly, despite the reduction of dendritic cells and T cells, the loss of TAK1 in dendritic cells resulted in a syndrome characterized by greatly expanded populations of myeloid cells, which are immune cells other than lymphocytes (i.e., white blood cells like T and B cells). This expansion, triggered directly by the lack of TAK1 in dendritic cells, remained prominent in a lymphocyte-deficient background, suggesting that myeloid-dependent inflammation can develop in the absence of T cells. Our results therefore demonstrate that dendritic cell signaling can influence myeloid homeostasis independently of lymphocytes, thereby establishing a previously unappreciated mechanism of dendritic cell-mediated active control of myeloid expansion and inflammation under physiological conditions. The crosstalk between dendritic cells and myeloid cells is likely mediated by an important cytokine known as FLT3L (4), and future work is required to identify the precise mechanism by which this occurs.

The life span of dendritic cells is important for the strength of T-cell responses to infections and other insults. We identify TAK1 as an important checkpoint in dendritic cell survival, thereby influencing both T-cell–mediated immune responses and the homeostasis of the immune system (Fig. P1). In the fruit fly, TAK1 has a crucial function in immune defense reactions. Our findings show that dendritic cells in mammals have evolved to adopt this pathway to regulate both their life span and the function and homeostasis of the immune system. This molecular pathway may be explored for the development of dendritic cell-based therapies against infections and tumors.

Fig. P1.

Fig. P1.

TAK1-mediated regulation of dendritic cell survival and immune system functions. Dendritic cells (DCs) are derived from macrophage and dendritic cell precursors (MDPs), whose maintenance requires the TAK1 enzyme. Moreover, TAK1 integrates prosurvival signals from certain receptors on the cell surface, namely Toll-like receptors (TLRs), CD40, and RANK, and activates downstream signaling pathways mediated by NF-κB and AKT-Foxo. TAK1 signaling in dendritic cells is required to promote T-cell homeostasis and activation, but it negatively controls the expansion of myeloid cells (immune cells other than T and B cells) and inflammation.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Data deposition: The microarray results reported in this paper have been deposited in the Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE34417).

See full research article on page E343 of www.pnas.org.

Cite this Author Summary as: PNAS 10.1073/pnas.1115635109.

References

  • 1.Huang G, Shi LZ, Chi H. Regulation of JNK and p38 MAPK in the immune system: Signal integration, propagation and termination. Cytokine. 2009;48:161–169. doi: 10.1016/j.cyto.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wan YY, Chi H, Xie M, Schneider MD, Flavell RA. The kinase TAK1 integrates antigen and cytokine receptor signaling for T cell development, survival and function. Nat Immunol. 2006;7:851–858. doi: 10.1038/ni1355. [DOI] [PubMed] [Google Scholar]
  • 3.Chen M, Wang J. Programmed cell death of dendritic cells in immune regulation. Immunol Rev. 2010;236:11–27. doi: 10.1111/j.1600-065X.2010.00916.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Birnberg T, et al. Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome. Immunity. 2008;29:986–997. doi: 10.1016/j.immuni.2008.10.012. [DOI] [PubMed] [Google Scholar]

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    Supporting Information
    1115635109_sapp.pdf (1.3MB, pdf)

    Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

    RESOURCES