Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Batf3-Dependent CD11blow/− Peripheral Dendritic Cells Are GM-CSF-Independent and Are Not Required for Th Cell Priming after Subcutaneous Immunization

  • Brian T. Edelson,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Tara R. Bradstreet,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Wumesh KC,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Kai Hildner,

    Current address: Department of Internal Medicine, Medical Clinic 1, University of Erlangen-Nürnberg, Erlangen, Germany

    Affiliations Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Jeremy W. Herzog,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Julia Sim,

    Affiliation Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • John H. Russell,

    Affiliation Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Theresa L. Murphy,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Emil R. Unanue,

    Affiliation Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Kenneth M. Murphy

    kmurphy@wustl.edu

    Affiliations Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America

Abstract

Dendritic cells (DCs) subsets differ in precursor cell of origin, functional properties, requirements for growth factors, and dependence on transcription factors. Lymphoid-tissue resident CD8α+ conventional DCs (cDCs) and CD11blow/−CD103+ non-lymphoid DCs are developmentally related, each being dependent on FMS-like tyrosine kinase 3 ligand (Flt3L), and requiring the transcription factors Batf3, Irf8, and Id2 for development. It was recently suggested that granulocyte/macrophage colony stimulating factor (GM-CSF) was required for the development of dermal CD11blow/−Langerin+CD103+ DCs, and that this dermal DC subset was required for priming autoreactive T cells in experimental autoimmune encephalitis (EAE). Here, we compared development of peripheral tissue DCs and susceptibility to EAE in GM-CSF receptor deficient (Csf2rb−/−) and Batf3−/− mice. We find that Batf3-dependent dermal CD11blow/−Langerin+ DCs do develop in Csf2rb−/− mice, but that they express reduced, but not absent, levels of CD103. Further, Batf3−/− mice lacking all peripheral CD11blow/− DCs show robust Th cell priming after subcutaneous immunization and are susceptible to EAE. Our results suggest that defective T effector priming and resistance to EAE exhibited by Csf2rb−/− mice does not result from the absence of dermal CD11blow/−Langerin+CD103+ DCs.

Introduction

GM-CSF regulates the development and activity of several myeloid cell types and influences both the initiation and maintenance of adaptive immune responses [1]. Mice lacking GM-CSF or its receptor show decreased antigen specific T cell priming against the encephalitogenic peptide of myelin oligodendrocyte glycoprotein (MOG35–55) and are resistant to EAE [2]. Beyond this role in priming adaptive immune responses, GM-CSF acts to sustain ongoing effector responses, both in EAE initiated by MOG35–55 peptide [2] and in collagen-induced arthritis [3]. Similarly, in a murine model of autoimmune myocarditis, GM-CSF acts during priming of Th17 responses by promoting IL-6 and IL-23 production from DCs and during ongoing autoimmune responses by promoting survival of autoreactive CD4+ T cells [4].

Several recent studies indicate that it is GM-CSF rather than IL-17 that is the primary Th17-derived cytokine responsible for the development of EAE [5][7]. Initially, it was thought that IL-17 production would explain why Th17 cells and RORγt were required for EAE. However, in vivo neutralization of IL-17 only reduced EAE severity but did not eliminate disease, and non-IL-17 and non-IFN-γ pathways have been implicated [8][10]. IL-23, but not IL-6 or TGF-β, was found to be critical for regulating the pathogenicitiy of Th17 cells [11], [12]. GM-CSF derived from T cells, but not other CNS-resident or peripheral immune cells, was required for development of EAE [5], and acted during the effector phase to augment pathogenicity of Th17 cells [6], [7]. IL-23 induced Th17 cells to increase production of GM-CSF, which further enhanced IL-23 production by antigen-presenting cells [6]. RORγt was shown to induce GM-CSF, which acted on infiltrating myeloid cells entering the CNS, rather than resident microglia, during the development of EAE [7]. These studies suggest a model of EAE in which pathogenic T cells secrete GM-CSF that activates myeloid cells infiltrating the CNS to produce pathogenic lesions and further amplifies IL-23 production by DCs [13].

A recent report has suggested that the role for GM-CSF in EAE was based on its requirement for the development of dermal CD11blow/−Langerin+CD103+ DCs [14]. Dermal CD11blow/−Langerin+CD103+ DCs represent one anatomic subtype of peripheral tissue CD11blow/−CD103+ DCs, all of which share with lymphoid tissue CD8α+ cDCs developmental dependence on Batf3, Irf8, and Id2 [15], [16]. CD8α+ cDCs are cross-presenting DCs, characterized by consistent expression of DEC205, and variable expression of CD103 which can be regulated by GM-CSF, IL-3, and TGF-β1 [17][19]. Mice lacking GM-CSF or the GM-CSF receptor were reported to lack dermal CD11blow/−Langerin+CD103+ DCs in the skin and peripheral lymph nodes under steady state and inflammatory conditions, and were resistant to EAE [14]. In addition, radiation chimeras expressing the human diphtheria toxin receptor (DTR) controlled by the Langerin promoter [20] and depleted of peripheral CD11blow/−Langerin+CD103+ DCs were resistant to EAE and had reduced priming of MOG35–55 -specific T cells [14]. This study concluded that GM-CSF was required for the initiation of EAE because it was required for the development of dermal CD11blow/−Langerin+CD103+ DCs, which were required to prime pathogenic T cells [14].

To test whether loss of GM-CSF receptor protects from EAE by eliminating dermal CD11blow/−Langerin+CD103+ DCs, we asked if Batf3−/− mice, which also lack this DC subset, are also protected from EAE. We find that Batf3−/− mice develop EAE after immunization and have increased numbers of MOG35–55 peptide-specific T cells, excluding a requirement for either peripheral CD11blow/−CD103+ DCs or lymphoid tissue-resident CD8α+ DCs in EAE development. Direct comparison of Batf3−/− and GM-CSF receptor deficient mice revealed that GM-CSF signaling was not required for the development of dermal CD11blow/−Langerin+CD103+ DCs, but rather regulated the expression level of CD103 on all peripheral DCs.

Results

Batf3−/− mice develop EAE following MOG immunization

To compare Batf3−/− mice with mice lacking the GM-CSF receptor (Csf2rb−/−) on the same genetic background, we backcrossed Batf3−/− mice onto the C57BL/6 background for 10 generations. Csf2rb−/− mice lack the common β-chain for signaling through receptors for GM-CSF, IL-5, and IL-3. However, mice have a second β-chain, Csf2rb2, which selectively pairs with the IL-3 receptor α-chain. Thus, Csf2rb−/− mice lose responsiveness to GM-CSF and IL-5, but not to IL-3 [21], [22]. Wild type (C57BL/6), Batf3−/−, and Csf2rb−/− mice were subcutaneously immunized with MOG35–55 peptide as previously described and followed for clinical disease [14]. Csf2rb−/− mice remained disease free as reported [14], but Batf3−/− mice developed EAE with the same severity and kinetics as wild type C57BL/6 mice (Fig. 1A).

thumbnail
Figure 1. Batf3−/− mice are susceptible to EAE.

(A) Clinical course of EAE in C57BL/6 mice, Batf3−/− mice (C57BL/6 background), and Csf2rb−/− mice. Data are combined from two experiments (n = 10 mice per group). Points represent the mean clinical score. For clarity, error bars are not displayed. (B) ELISPOT assays on popliteal lymph node cells at day 7 following footpad immunization (MOG35–55 peptide in CFA) and restimulation with MOG35–55 peptide. Data are from one of two similar experiments (n = 3 per group). Mean ± SEM, p values by unpaired student's t test.

https://doi.org/10.1371/journal.pone.0025660.g001

In draining lymph nodes, examination by ELISPOT assay showed that immunized Batf3−/− mice had an increased frequency of MOG35–55-specific T cells producing IL-17 and IFN-γ (Fig. 1B), while Csf2rb−/− mice had a significantly reduced frequency of such cells, as previously reported [14]. Thus, Batf3−/− mice, which lack all peripheral CD11blow/−CD103+ DCs [16], are able to prime pathogenic CD4+ T cells, and remain susceptible to EAE. These findings would disagree with the interpretation that resistance to EAE in Csf2rb−/− mice results from the absence of dermal CD11blow/−Langerin+CD103+ DCs as was recently suggested [14].

Csf2rb−/− mice retain development of Batf3-dependent dermal CD11blow/−Langerin+ DCs

GM-CSF was recently reported to directly regulate CD103 expression on splenic CD8α+ DCs [17][19], so conceivably Csf2rb−/− mice might retain dermal CD11blow/−Langerin+ DCs that simply lack CD103. To test this, we examined DCs from C57BL/6, Batf3−/−, and Csf2rb−/− mice for expression of markers that distinguish between Batf3-dependent and Batf3-independent DC subsets (Fig. 2A).

thumbnail
Figure 2. Batf3-dependent dermal CD11blow/−Langerin+ DCs develop in Csf2rb−/− mice, but lack expression of CD103.

(A) FACS analysis of SDLN (inguinal) DCs from C57BL/6, Batf3−/− (C57BL/6 background), and Csf2rb−/− mice. Left plots are gated on CD11chigh cells. The second column is gated on migratory (DEC205+CD8α) DCs. The third, fourth, and fifth columns are gated on Langerin+ migratory DCs. Numbers represent the percentage of cells within the indicated gates. MFI are shown for the indicated gates. Data are representative of eight to ten individual inguinal lymph nodes from four to five mice per genotype over two experiments. (B) FACS analysis of SDLN (inguinal) DCs from C57BL/6, Batf3−/−, and Csf2rb−/− mice. Left plots are gated on CD11chigh cells. Middle plots are gated on migratory (DEC205+CD8α) DCs. Right plots are gated on all Langerin+ migratory DCs, independent of expression of CD11b. Numbers represent the percentage of cells within the indicated gates. Data are representative of inguinal lymph nodes from at least five mice per genotype for all stains except CD24, which has been performed on one mouse per genotype. (C) Absolute cell numbers per individual inguinal lymph nodes calculated from the FACS analysis in (A). Langerhans cells were gated as Langerin+CD11b+EpCAMhigh DCs, and dermal Langerin+ DCs were gated as Langerin+CD11blow/−EpCAMmid DCs. Horizontal bars represent the geometric mean; p values by unpaired student's t test. (D) Clinical course of EAE in 129S6/SvEv mice and Batf3−/− mice (129S6/SvEv background). Data are from one of three similar experiments (n = 5 mice per group). Points represent the mean clinical score. For clarity, error bars are not displayed.

https://doi.org/10.1371/journal.pone.0025660.g002

In skin draining lymph nodes (SDLNs), migratory DCs are evident as DEC205+CD8α cells, which can be divided into at least three subsets based on CD11b, Langerin, and CD103 expression [23][25]. C57BL/6, Batf3−/−, and Csf2rb−/− mice each contained normal populations of dermal Langerin DCs and Langerhans cells of the epidermis (CD11b+Langerin+CD103EpCAMhigh) (Fig. 2A). However, differences were evident in the dermal CD11blow/−Langerin+CD103+ DCs between these strains. C57BL/6 mice contained the normal population of dermal CD11blow/−Langerin+CD103+EpCAMmid DCs, which were missing in Batf3−/− mice [16]. Notably, Csf2rb−/− mice retained this population of dermal DCs, but in this strain of mice, these DCs expressed slightly lower levels of CD103 relative to C57BL/6 mice. CD24 can also been used to distinguish between populations of dermal DCs [26], and similarly C57BL/6 mice contained a population of dermal Langerin+CD24+EpCAMmid DCs that were completely missing in Batf3−/− mice, but which were present in Csf2rb−/− mice (Fig. 2B). Csf2rb−/− mice showed reduced total lymph node cellularity and a reduced number of overall lymph node DCs, but normal numbers of Langerhans cells and dermal CD11blow/−Langerin+ DCs (Fig. 2C). Batf3−/− mice showed a selective ∼100-fold reduction in the number of dermal CD11blow/−Langerin+ DCs. These results indicate that Csf2rb−/− mice do contain dermal CD11blow/−Langerin+ DCs, but that in the absence of GM-CSF signaling, CD103 expression is reduced.

C57BL/6 Batf3−/− mice contain CD8α+ cDCs in SDLNs but not spleen

In analyzing SDLNs of C57BL/6 Batf3−/− mice, we noticed an unexpected population of DEC205+CD8α+ cDCs that were eliminated in the spleen and SDLNs of 129S6/SvEv Batf3−/− mice [16], [27] (Fig. 2A, 2C). Therefore, we directly compared DCs from SDLNs of Batf3−/− mice on both genetic backgrounds (Fig. S1A). C57BL/6 Batf3−/− mice lacked dermal CD11blow/−CD103+ DCs (Fig. S1A) but had a persistence of DEC205+CD8α+ cDCs in peripheral lymph nodes (Fig. S1A, S1B). In spleens, Batf3−/− mice showed a significant decrease in DEC205+CD8α+ cDCs on both C57BL/6 and 129S6/SvEv backgrounds compared to controls (Fig. S1C, S1D). On the 129S6/SvEv background, DEC205+CD8α+ cDCs were reduced from 4.8% of splenic cDCs in control mice to only 0.11% of splenic cDCs in Batf3−/− mice. On the C57BL/6 background, DEC205+CD8α+ cDCs were reduced from 14% of splenic cDCs in control mice to only 2.5% of splenic cDCs in Batf3−/− mice (Fig. S1C, S1D). Thus, for splenic cDCs, Batf3 is non-redundant in the development of DEC205+CD8α+ cDCs.

While loss of Batf3 does not eliminate the population of DEC205+CD8α+ cDCs in SDLNs, these cells did not express CD103 normally (Fig. S1E). 9% of the DEC205+CD8α+ cDCs in SDLNs of control mice expressed CD103, while only 1.8% expressed CD103 from Batf3−/− mice. Notably, CD103 expres sion was also reduced on this population from SDLNs of Csf2rb−/−mice. Conceivably, the persistence of DEC205+CD8α+ cDCs in SDLNs on the C57BL/6 background could result from genetic polymorphisms that impact the redundancy of Batf3 in the development of these cells. Alternately, variable inflammation between strains could be involved, since infection with Toxoplasma gondii can regulate the size of splenic CD11blow/−CD103+ DC populations [28].

129S6/SvEv Batf3−/− are susceptible to EAE

Conceivably, persistence of DEC205+CD8α+ cDCs in SDLNs of C57BL/6 Batf3−/− mice was the basis for their susceptibility to EAE, based on potentially overlapping functions for DEC205+CD8α+ cDCs and dermal CD11blow/−CD103+ DCs [29]. To test this, we examined Batf3−/− mice on the 129S6/SvEv background, which lack both DEC205+CD8α+ and dermal CD11blow/−CD103+ DCs in both spleen and SDLNs (Fig. 2D). As with C57BL/6 Batf3−/− mice, Batf3−/− mice on the129S6/SvEv background were equally as susceptible to EAE as wild type controls. Thus, loss of dermal CD11blow/−CD103+ DCs could not have explained the resistance to EAE that was observed in Csf2rb−/− mice.

Csf2rb−/− mice retain Batf3-dependent CD11blow/− peripheral tissue DCs with reduced CD103 expression

We next compared C57BL/6, Batf3−/−, and Csf2rb−/− mice for the presence of DC subsets in peripheral tissues, specifically the lung and kidney. In the lung, DCs exist as two populations, characterized as Batf3-dependent CD11blow/−LangerinmidCD103+ DCs and Batf3-independent CD11b+LangerinCD103 DCs. Lungs from Batf3−/− mice completely lacked CD11blow/−LangerinmidCD103+ DCs (Figure 3A) as expected [16]. Importantly, lungs from Csf2rb−/− mice retained a normal population of pulmonary CD11blow/− DCs, which expressed reduced levels of CD103 and undetectable levels of Langerin (Fig. 3A). Csf2rb−/− mice develop alveolar proteinosis due to defective surfactant handling by lung macrophages in the absence of GM-CSF signaling [21]. Conceivably, this inflammatory process could have influenced CD103 expression by CD11blow/− lung DCs. To test this, we examined CD103 expression by DCs of the kidney, since this organ shows no pathology in Csf2rb−/− mice. Again, while Batf3−/− mice completely lacked CD11blow/−CD103+ kidney DCs, Csf2rb−/− mice had normal numbers of CD11blow/− kidney DCs, but which expressed moderately reduced levels of CD103 (Fig. 3B). In summary, it appears that Csf2rb−/− mice retain normal development of peripheral tissue CD11blow/− which express reduced levels of CD103.

thumbnail
Figure 3. GM-CSF controls the level of CD103 expression on Batf3-dependent CD11blow/− peripheral DCs.

(A) FACS analysis of lung DCs from C57BL/6, Batf3−/− (C57BL/6 background), and Csf2rb−/− mice. Left plots are gated on live, single cells. The second column of plots is gated on CD11c+CD45.2+ cells, consisting of a mixture of lung macrophages and DCs. Note that in Csf2rb−/− mice, lung macrophages lack CD11c expression [38], and therefore this gate includes only DCs. The third and fourth columns of plots are gated on I-Abhigh autofluorescencelow DCs. Numbers represent the percentage of cells within the indicated gates. Data are representative of six individual lungs from three mice per genotype over two experiments. (B) FACS analysis of kidney DCs from C57BL/6, Batf3−/− (C57BL/6 background), and Csf2rb−/− mice. Left plots are gated on live, single cells. The second column of plots is gated on CD11chighCD45.2+ cells, consisting of DCs. The third column of plots is gated on CD11chighI-Ab+ DCs. Numbers represent the percentage of cells within the indicated gates. Data are representative of six individual kidneys from three mice per genotype over two experiments.

https://doi.org/10.1371/journal.pone.0025660.g003

Basal CD103 expression by CD8α+-equivalent cDCs requires Batf3 but not GM-CSF signaling

While CD103 has been used as a marker for the dermal CD11blow/−Langerin+ DC lineage [14], recent work shows that CD103 expression can be dynamically regulated by cytokines [17], [18]. Therefore, we wished to re-examine the relationships between CD103 expression, Batf3, and GM-CSF signaling. We selected the Flt3L-induced DC differentiation from bone marrow cultures for this purpose since it has allowed the analysis of CD103 expression as dependent on both Batf3 and GM-CSF [17], [18] (Fig. 4).

thumbnail
Figure 4. GM-CSF induces CD103 expression on Flt3L-derived CD8α-equivalent DCs.

FACS analysis of developing DCs in bone marrow cells from C57BL/6, Batf3−/− (C57BL/6 background), and Csf2rb−/− mice cultured with Flt3L for 9 days, with or without the addition of GM-CSF to the culture on day 7. The left column is gated on live single cells, the second on CD11c+CD45RA cDCs, the third on I-Ab+ cDCs, and the fourth on CD24highSirpαlow cells. Numbers represent the percentage of cells within the indicated gates. Data are representative of bone marrow cells from two mice per genotype over two experiments.

https://doi.org/10.1371/journal.pone.0025660.g004

Previously, CD24high SIRPαlow DCs were considered an “equivalent” of CD8α+ cDCs in Flt3L-treated bone marrow cultures [30], despite the fact that only a subpopulation of these cells express CD103 (Fig. 4). The CD103+ fraction of CD24high SIRPαlow DCs was more potent than the CD103 fraction for cross-presentation [17], suggesting a correlation between CD103 expression and CD8α+ cDC function. We find that addition of GM-CSF during day 7 through 9 of Flt3L culture induces CD103 expression on all CD24high SIRPαlow DCs (Fig. 4), in agreement with recent reports which showed that such GM-CSF-induced cells exhibit robust cross-presentation [17], [18]. However, while addition of IL-3 and TGF-β1 to Flt3L cultures can both induce expression of CD103, only IL-3 induced the capacity for cross-presentation [17], dissociating CD103 expression from cross-presentation. Likewise, we found that GM-CSF treatment induced CD103 expression on all CD24highSIRPαlow DCs, even in bone marrow cultures from Batf3−/− mice (Fig. 4), which completely lack CD103+ DCs in vivo [16], further dissociating CD103 expression from the properties of true CD8α+ cDC equivalents. In agreement, a recent report found that GM-CSF-induced CD103+CD24highSIRPαlow Flt3L-derived DCs from Batf3−/− mice lacked the capacity for cross-presentation [18]. Finally, Csf2rb−/− bone marrow generated a subpopulation of CD103+CD24highSIRPαlow DCs, indicating development that is independent of GM-CSF signaling, although whether these arise from the actions of cytokines such as IL-3, or arise independently of soluble factors present in the culture remains unknown. In summary, these results indicate that CD103 expression can be regulated by GM-CSF, independently of Batf3, and may not represent a static marker of Batf3-dependent DC lineages.

Discussion

Our study makes two new observations. First, we show that CD11blow/− non-lymphoid DCs develop in the absence of GM-CSF signaling. Second, we show that Batf3-dependent CD11blow/− non-lymphoid DCs are not required for T effector cell priming or clinical EAE after subcutaneous immunization. King et al. recently concluded that mice deficient for GM-CSF or GM-CSF receptor were resistant to EAE because they lacked dermal CD11blow/−Langerin+CD103+ DCs, which they proposed were required for priming effector CD4 T cells with MOG35–55 [14]. Dermal CD11blow/−Langerin+CD103+ DCs are a conventional DC subset that requires the transcription factor Batf3 for their development [16]. Here, we directly compared Csf2rb−/− with Batf3−/− mice for their susceptibility to EAE and for their development of various DC subsets using a broad panel of DC markers, including Langerin, CD11b, EpCAM, CD24, and CD103. We confirm that the dermal CD11blow/−Langerin+CD103+ DC subset is absent in Batf3−/− mice, but surprisingly find that Csf2rb−/− mice harbor normal numbers of this DC subset in SDLNs as a CD11blow/− Langerin+EpCAMmidCD24high cell that expresses reduced, but not absent, levels of CD103. Moreover, in other peripheral tissues, Csf2rb−/− mice have similar CD11blow/−CD103+ DCs that express reduced, but not absent, levels of CD103.

Our data agree with recent reports that GM-CSF directly regulates CD103 expression on Flt3L-derived CD8α+ equivalent cDCs and splenic DEC205+CD8α+ cDCs [17][19]. Beyond this, we show that GM-CSF also regulates levels of CD103 expression on peripheral tissue-resident CD11blow/− DCs, which were not examined in the previous studies. Notably, a separate report has suggested that GM-CSF signaling is required for the development of small intestinal lamina propria CD11b+CD103+ DCs [31], although their data could be reinterpreted as simply representing reduced CD103 expression by these DCs. In that report, Csf2rb−/−mice had half as many lamina propria CD11b+CD103+ DCs as wild type mice, but 1.7-fold more CD11b+CD103 DCs than wild type mice, consistent with a reduction of CD103 expression as an explanation for the “missing” subset.

King et al. also found decreased T cell priming to subcutaneous immunization with MOG35–55 peptide and partial EAE resistance after in vivo depletion of peripheral Langerin+CD103+ DCs using radiation chimeras reconstituted with bone marrow from transgenic mice expressing DTR under the control of the Langerin promoter [20]. The use of radiation chimeras allowed the selective depletion of dermal CD11blow/−Langerin+ DCs, avoiding depletion of epidermal CD11b+Langerin+ Langerhans cells, because the latter are radiation-resistant, and were therefore of host origin and did not express the DTR. Our results indicate that Csf2rb−/− mice are not deficient in dermal CD11blow/−Langerin+ DCs, but rather that these cells have reduced CD103 expression. However, Batf3−/− mice lack this dermal DC subset, and yet are susceptible to EAE induction and display increased, not decreased, T cell priming to MOG35–55 peptide immunization. It is unclear why DTR-mediated depletion of donor-derived, radiation-sensitive Langerin+ DCs reduces priming and delays EAE, when genetic ablation of these cells in Batf3−/− mice leaves priming and EAE intact. Their use of DTR-mediated depletion involved irradiation of mice prior to immunization, which may have had unintended consequences. Furthermore, a study by Henri et al. has identified additional dermal DC subsets [26], including one that is Langerin+ but CD103, although it's Batf3-dependence was not characterized. Conceivably, the depletion of this subset by diphtheria toxin treatment could contribute to differences between our results and those of King et al. Notably, DTR-mediated depletion reduced the severity of EAE, but with a profound decrease only in MOG35–55-specific IFN-γ-secreting T cells, with minimal change in the number of IL-17-secreting T cells. In contrast, MOG35–55-immunized Csf2rb−/− mice had profound reductions in antigen-specific IFN-γ-secreting and in IL-17-secreting T cells, suggesting that Csf2rb−/− mice differ from mice in which Langerin+ DCs are depleted using DTR.

Our results suggest that the absence of dermal CD11blow/−Langerin+CD103+ DCs leads to augmented T cell priming of both Th17 and Th1 responses. Igyártó et al. have recently shown that skin infection of Batf3−/− mice with recombinant Candida albicans led to reduced Th1 but increased Th17 responses compared to control mice [32]. These authors demonstrated that dermal Langerin+CD103+ DCs expressed IL-12, explaining their ability to promote Th1 cell responses, and IL-27, a cytokine known to inhibit Th17 cell responses [33], [34]. While we found increased Th17 responses in Batf3−/− mice immunized with MOG35–55 and CFA, we also observed increased Th1 responses compared to control mice. Conceivably, the different Th1 responses between our study and Igyártó et al. could arise from differences in the form of immunization, relying on CFA versus fungal infection. Notably, blood-derived inflammatory monocytes have been reported to serve as a critical source of IL-12 after CFA-based subcutaneous immunization [35]. While we observed between a 2 to 3-fold higher frequency of IL-17-producing T cells in MOG35–55-immunized Batf3−/− mice relative to controls, the clinical EAE scores were similar between these groups. Conceivably, the frequency of IL-17-producing T cells is unrelated to the severity of EAE when Th17 cells are above some threshold frequency. Alternatively, IL-17 itself may not be the limiting factor in regulating the intensity of EAE, since GM-CSF also recently was implicated as the major Th17-derived cytokine responsible for driving pathogenic lesions in this model [5][7]. In fact, however, we do observe a slightly more rapid onset of EAE in Batf3−/− mice, which was more pronounced in the 129S6/SvEv genetic background (Fig. 2D), which could relate to increased Th17 development.

If a lack of peripheral CD11blow/− DCs does not abrogate priming of T effector cells after subcutaneous immunization, why then does this priming defect occur in Csf2rb−/− mice? One possibility is that other DC subsets besides dermal CD11blow/− DCs can prime CD4 T cells [35], but that the ability of these other DCs to prime requires that they receive a GM-CSF signal. Indeed, GM-CSF is essential for IL-6 and IL-23 production by DCs during the priming phase of autoimmune myocarditis [4] and is required for inflammatory DC development from monocytes in a model of repeated immunization with methylated BSA and CFA [36]. T cells do not express GM-CSF receptor, so an intrinsic T cell defect seems unlikely. While the source of GM-CSF after subcutaneaous immunization is still unclear, antigen-specific T cells can produce GM-CSF [5][7] and may augment responses by DCs during initial priming of T effector cells.

Materials and Methods

Ethics statement

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Animal Studies Committee of Washington University (#20090320).

Mice

Wild-type 129S6/SvEv and C57BL/6 mice were purchased from Taconic. Batf3−/− mice on a 129S6/SvEv background were previously generated in our laboratory [27], and were backcrossed for 10 generations to the C57BL/6 background. Csf2rb−/− mice on the C57BL/6 background were purchased from Jackson Laboratory. Experiments were performed with sex-matched mice at 8–20 weeks of age. Mice were bred and maintained in our specific pathogen-free animal facility according to institutional guidelines.

Induction of EAE

EAE was induced as previously described [14]. Briefly, mice were immunized subcutaneously with 100 micrograms MOG35–55 peptide (Sigma Genosys) emulsified in complete Freund's adjuvant (CFA) (made with 5 mg/ml heat-killed Mycobacterium tuberculosis H37Ra (BD Difco) in incomplete Freund's adjuvant (BD Difco)). Pertussis toxin (List Biological Laboratories) was injected intraperitoneally (300 ng) on days zero and two. Mice were observed for signs of EAE and graded on a standard 0–5 scale as described [37].

T cell priming and ELISPOT assays

Mice were immunized subcutaneously in the hind footpads with 10 nanomoles MOG35–55 peptide (Sigma Genosys) emulsified in complete Freund's adjuvant (CFA) (BD Difco). Popliteal lymph nodes were collected at day 7, and single cell suspensions were used in ELISPOT assays with the IFNγ ELISPOT antibody pair from BD Biosciences on Multiscreen Filter Plates from Millipore. An IL-17 ELISPOT was designed using anti-IL17A capture (clone TC11-18H10) and detection (clone TC11-8H4.1) antibodies (BD Biosciences). Cells were plated at 1×106 cells/well, in duplicate, and stimulated with either no antigen, or 10 micromolar MOG35–55 peptide. Plates were developed after 16 hours of stimulation at 37°C, and spots were counted on an Immunospot counter (Cellular Technology Ltd.).

Antibodies

The following antibodies were purchased from BD Biosciences: PE-Cy7 anti-CD11b (M1/70), PerCP-Cy5.5 anti-CD8α (53-6.7), V500 anti-CD8α (53-6.7), PE anti-CD45RA (14.8), biotin anti-CD24 (30F1), FITC CD24 (M1/69), APC anti-CD172a/SIRPα (P84). FITC anti-CD11b (M1/70), APC eFluor 780-anti-CD11c (N418), PE anti-CD103 (2E7), biotin anti-CD103 (2E7), eFluor450 anti-CD317/BST2 (eBio927), eFluor450 anti-MHCII (I-A/I-E) (M5/114.15.2), APC anti-CD45.2 (104), and streptavidin-eFluor450 were purchased from eBioscience. PE anti-CD205/DEC205 (NLDC-145) and APC anti-CD205/DEC205 (NLDC-145) were purchased from Miltenyi. Alexa Fluor 488 anti-CD207/Langerin (929F3.01) was purchased from Imgenex. PE anti-CD326/EpCAM (G8.8) was purchased from Biolegend.

Flow cytometry

Inguinal lymph nodes and spleens were minced and digested in 5 ml Iscove's modified Dulbecco's media +10% FCS (cIMDM) with 250 µg/ml collagenase B (Roche) and 30 U/ml DNase I (Sigma-Aldrich) for 1 h at 37°C with stirring. Lungs and kidneys were perfused with 10 ml DPBS via injection into the right ventricle after transection of the lower aorta. Dissected lungs and kidneys were minced and digested in 5 ml of cIMDM with 4 mg/ml collagenase D (Roche) for 1 h at 37°C with stirring. EDTA (5 mM final concentration) was added to cell suspensions, and cells were incubated on ice for 5 min. Cells were passed through an 80 µm strainer before red blood cells lysis with ACK lysis buffer. Cells were counted on a Vi-CELL analyzer, and 2–5×106 cells were used per antibody staining reaction.

Staining was performed at 4°C in the presence of Fc Block (clone 2.4G2, BD Biosciences or BioXcell) in FACS buffer (DPBS+0.5% BSA+2 mm EDTA+0.02% sodium azide). For experiments involving intracellular anti-Langerin staining, cells were surface antibody stained prior to fixation in 4% formaldehyde (Thermo Scientific) for 15 minutes at room temperature. Cells were subsequently permeabilized (DPBS+0.1% BSA+0.5% saponin) for 5 min at 4°C and stained in the same permeabilization buffer with anti-Langerin antibody. Cells were washed twice in permeabilization buffer before returning them to FACS buffer. Flow cytometry was performed on a FACSCantoII (BD Biosciences), and data was analyzed with FlowJo software (Tree Star, Inc.).

Bone marrow cultures

Bone marrow cells from femurs and tibias were collected and red blood cells were lysed in ACK lysis buffer. Cells were cultured in four ml cIMDM in six-well plates at 2×106 cells/ml containing 160 ng/ml murine Flt3L (Peprotech). At day seven of culture, murine GM-CSF (Peprotech) was added to some wells at 20 ng/ml. Non-adherent cells were collected at day nine for flow cytometry.

Supporting Information

Figure S1.

Flow cytometric analyses of DCs in Batf3−/− mice. (A) FACS analysis of SDLN (inguinal) DCs from Batf3+/+ and Batf3−/− mice on both the C57BL/6 and 129S6/SvEv backgrounds. Left plots from mice of each background are gated on CD11c+BST2 cDCs. Right plots from mice of each background are gated on migratory (DEC205+CD8α) DCs. Numbers represent the percentage of cells within the indicated gates. Data are representative of at least six mice per genotype and background obtained in several independent experiments. (B) Absolute cell numbers of DEC205+CD8α+ DCs per individual inguinal lymph nodes from C57BL/6 and Batf3−/− mice (C57BL/6 background) calculated from the FACS analysis performed in Figure 2A. Horizontal bars represent the geometric mean, p value by unpaired student's t test. (C) and (D) FACS analysis of splenic DCs from Batf3+/+ and Batf3−/− mice on both the (C) C57BL/6 and (D) 129S6/SvEv backgrounds. Plots are gated on CD11c+I-Ab+ cDCs. Numbers represent the percentage of cells within the indicated gates. Data are representative of at least six mice per genotype and background obtained in several experiments. (E) FACS analysis of SDLN (inguinal) DCs from C57BL/6, Batf3−/− (C57BL/6 background), and Csf2rb−/− mice. Left plots are gated on CD11chigh cells. Right plots are gated on DEC205+CD8α+ cDCs. Numbers represent the percentage of cells within the indicated gates. Data are representative of eight to ten individual inguinal lymph nodes from four to five mice per genotype obtained in two experiments. Data is depicted in these plots are derived from analysis of the same mice used in Figure 2A, but gated to analyze CD103 expression on DEC205+CD8α+ cDCs.

https://doi.org/10.1371/journal.pone.0025660.s001

(TIF)

Author Contributions

Conceived and designed the experiments: KMM BTE ERU TLM JHR. Performed the experiments: BTE TRB WKC KH JWH JS. Analyzed the data: BTE KMM TLM ERU JHR. Contributed reagents/materials/analysis tools: BTE KMM. Wrote the paper: BTE KMM.

References

  1. 1. Hamilton JA (2008) Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 8: 533–544.
  2. 2. McQualter JL, Darwiche R, Ewing C, Onuki M, Kay TW, et al. (2001) Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis. J Exp Med 194: 873–882.
  3. 3. Cook AD, Braine EL, Campbell IK, Rich MJ, Hamilton JA (2001) Blockade of collagen-induced arthritis post-onset by antibody to granulocyte-macrophage colony-stimulating factor (GM-CSF): requirement for GM-CSF in the effector phase of disease. Arthritis Res 3: 293–298.
  4. 4. Sonderegger I, Iezzi G, Maier R, Schmitz N, Kurrer M, et al. (2008) GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J Exp Med 205: 2281–2294.
  5. 5. Ponomarev ED, Shriver LP, Maresz K, Pedras-Vasconcelos J, Verthelyi D, et al. (2007) GM-CSF production by autoreactive T cells is required for the activation of microglial cells and the onset of experimental autoimmune encephalomyelitis. J Immunol 178: 39–48.
  6. 6. El Behi M, Ciric B, Dai H, Yan Y, Cullimore M, et al. (2011) The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 12: 568–575.
  7. 7. Codarri L, Gyulveszi G, Tosevski V, Hesske L, Fontana A, et al. (2011) RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol 12: 560–567.
  8. 8. Hofstetter HH, Ibrahim SM, Koczan D, Kruse N, Weishaupt A, et al. (2005) Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol 237: 123–130.
  9. 9. Lovett-Racke AE, Yang Y, Racke MK (2011) Th1 versus Th17: are T cell cytokines relevant in multiple sclerosis? Biochim Biophys Acta 1812: 246–251.
  10. 10. Kroenke MA, Chensue SW, Segal BM (2010) EAE mediated by a non-IFN-gamma/non-IL-17 pathway. Eur J Immunol 40: 2340–2348.
  11. 11. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, et al. (2007) TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol 8: 1390–1397.
  12. 12. Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, et al. (2010) Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature 467: 967–971.
  13. 13. McGeachy MJ (2011) GM-CSF: the secret weapon in the T(H)17 arsenal. Nat Immunol 12: 521–522.
  14. 14. King IL, Kroenke MA, Segal BM (2010) GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization. J Exp Med 207: 953–961.
  15. 15. Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, et al. (2009) The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med 206: 3115–3130.
  16. 16. Edelson BT, KC W, Juang R, Kohyama M, Benoit LA, et al. (2010) Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells. J Exp Med 207: 823–836.
  17. 17. Sathe P, Pooley J, Vremec D, Mintern J, Jin JO, et al. (2011) The acquisition of antigen cross-presentation function by newly formed dendritic cells. J Immunol 186: 5184–5192.
  18. 18. Jackson JT, Hu Y, Liu R, Masson F, D'Amico A, et al. (2011) Id2 expression delineates differential checkpoints in the genetic program of CD8alpha(+) and CD103(+) dendritic cell lineages. EMBO J 30: 2690–2704.
  19. 19. Zhan Y, Carrington EM, van Nieuwenhuijze A, Bedoui S, Seah S, et al. (2011) GM-CSF increases cross presentation and CD103 expression by mouse CD8(+) spleen dendritic cells. Eur J Immunol. In press.
  20. 20. Kissenpfennig A, Henri S, Dubois B, Laplace-Builhe C, Perrin P, et al. (2005) Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 22: 643–654.
  21. 21. Robb L, Drinkwater CC, Metcalf D, Li RL, Kontgen F, et al. (1995) Hematopoietic and Lung Abnormalities in Mice with A Null Mutation of the Common Beta-Subunit of the Receptors for Granulocyte-Macrophage Colony-Stimulating Factor and Interleukin-3 and Interleukin-5. Proc Natl Acad Sci U S A 92: 9565–9569.
  22. 22. Nicola NA, Robb L, Metcalf D, Cary D, Drinkwater CC, et al. (1996) Functional inactivation in mice of the gene for the interleukin-3 (IL-3)-specific receptor beta-chain: implications for IL-3 function and the mechanism of receptor transmodulation in hematopoietic cells. Blood 87: 2665–2674.
  23. 23. Bursch LS, Wang L, Igyarto B, Kissenpfennig A, Malissen B, et al. (2007) Identification of a novel population of Langerin+ dendritic cells. J Exp Med 204: 3147–3156.
  24. 24. Ginhoux F, Collin MP, Bogunovic M, Abel M, Leboeuf M, et al. (2007) Blood-derived dermal langerin+ dendritic cells survey the skin in the steady state. J Exp Med 204: 3133–3146.
  25. 25. Poulin LF, Henri S, de Bovis B, Devilard E, Kissenpfennig A, et al. (2007) The dermis contains langerin+ dendritic cells that develop and function independently of epidermal Langerhans cells. J Exp Med 204: 3119–3131.
  26. 26. Henri S, Poulin LF, Tamoutounour S, Ardouin L, Guilliams M, et al. (2010) CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J Exp Med 207: 189–206.
  27. 27. Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, et al. (2008) Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322: 1097–1100.
  28. 28. Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, et al. (2011) CD8a+ Dendritic Cells Are the Critical Source of Interleukin-12 that Controls Acute Infection by Toxoplasma gondii Tachyzoites. Immunity 35: 249–259.
  29. 29. Bedoui S, Whitney PG, Waithman J, Eidsmo L, Wakim L, et al. (2009) Cross-presentation of viral and self antigens by skin-derived CD103(+) dendritic cells. Nat Immunol 10: 488–495.
  30. 30. Naik SH, O'Keeffe M, Proietto A, Hochrein H, Shortman K, et al. (2010) CD8+, CD8−, and plasmacytoid dendritic cell generation in vitro using flt3 ligand. Methods Mol Biol 595: 167–176.
  31. 31. Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, et al. (2009) Origin of the lamina propria dendritic cell network. Immunity 31: 513–525.
  32. 32. Igyártó BZ, Haley K, Ortner D, Bobr A, Gerami-Nejad M, et al. (2011) Skin-Resident Murine Dendritic Cell Subsets Promote Distinct and Opposing Antigen-Specific T Helper Cell Responses. Immunity 35: 260–272.
  33. 33. Stumhofer JS, Laurence A, Wilson EH, Huang E, Tato CM, et al. (2006) Interleukin 27 negatively regulates the development of interleukin 17-producing T helper cells during chronic inflammation of the central nervous system. Nat Immunol 7: 937–945.
  34. 34. Diveu C, McGeachy MJ, Boniface K, Stumhofer JS, Sathe M, et al. (2009) IL-27 blocks RORc expression to inhibit lineage commitment of Th17 cells. J Immunol 182: 5748–5756.
  35. 35. Nakano H, Lin KL, Yanagita M, Charbonneau C, Cook DN, et al. (2009) Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses. Nat Immunol 10: 394–402.
  36. 36. Kamphorst AO, Guermonprez P, Dudziak D, Nussenzweig MC (2010) Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J Immunol 185: 3426–3435.
  37. 37. Lees JR, Sim J, Russell JH (2010) Encephalitogenic T-cells increase numbers of CNS T-cells regardless of antigen specificity by both increasing T-cell entry and preventing egress. J Neuroimmunol 220: 10–16.
  38. 38. Guth AM, Janssen WJ, Bosio CM, Crouch EC, Henson PM, et al. (2009) Lung environment determines unique phenotype of alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 296: L936–L946.