Abstract
In vitro culture of bone marrow (BM) with Fms-like tyrosine kinase 3 ligand (Flt3L) is widely used to study development and function of type 1 conventional dendritic cells (cDC1). Hematopoietic stem cells (HSCs) and many progenitor populations that possess cDC1 potential in vivo do not express Flt3 and thus may not contribute to Flt3L-mediated cDC1 production in vitro. Here, we present a KitL/Flt3L protocol that recruits such HSCs and progenitors into the production of cDC1. Kit ligand (KitL) is used to expand HSCs and early progenitors lacking Flt3 expression into later stage where Flt3 is expressed. Following this initial KitL phase, a second Flt3L phase is used to support the final production of DCs. With this two-stage culture, we achieved approximately tenfold increased production of both cDC1 and cDC2 compared to Flt3L culture. cDC1 derived from this culture are similar to in vivo cDC1 in their dependence on IRF8, ability to produce IL-12, and induction of tumor regression in cDC1-deficient tumor-bearing mice. This KitL/Flt3L system for cDC1 production will be useful in further analysis of cDC1 that rely on in vitro generation from BM.
Keywords: Dendritic cell, Flt3L, KitL, HSC, Tumor immunology
Introduction
Dendritic cells (DCs) comprise several related hematopoietic lineages that function in innate and adaptive immune responses [1]. Conventional DCs (cDCs) comprise two major subsets that serve as antigen-presenting cells to prime naïve T cells and activate innate lymphoid cells [2–4]. cDC1 require the transcription factors NFIL3, ID2, BATF3, and IRF8 for development and specialize in cross-presentation to prime CD8 T cells against many intracellular pathogens and tumors [3]. cDC2 supports both Th17 [5, 6] and Th2 responses to various other pathogens [7–9]. A third DC subset, plasmacytoid DC (pDC), serves as sentinels of viral infection and augment T-cell responses by the production of cytokines [10].
DCs can be generated from bone marrow (BM) progenitors cultured with the activating ligand for Fms-like tyrosine kinase 3 (Flt3) [11]. Flt3 encodes a class III receptor tyrosine kinase closely related to Kit and macrophage colony-stimulating factor 1 receptor (M-CSFR or Fms) [12]. Identification of Flt3 ligand (Flt3L) [13] inspired attempts using it in expanding hematopoietic cells. Subcutaneous Flt3L administration in mice dramatically expanded CD11c+ MHCII+ DC populations [14]. Flt3L alone could generate CD11c+ DCs from densely plated murine BM in vitro [15] and generates cDC1, cDC2, and pDC [11, 16]. cDC1 produced by this method resemble in vivo cDC1 in several ways, including the developmental requirement for IRF8 [11] and BATF3 [17], and induction of IL-12 by Toxoplasma gondii soluble tachyzoite antigen (STAg) [18]. However, Flt3L cultures of BM produce relatively small numbers of cDC1 compared with the substantially greater numbers of monocyte-derived DCs (Mo-DCs) generated GM-CSF culture [19, 20]. As a result, investigating cDC1 on a large scale has been challenging.
Hematopoietic stem cells (HSCs) were originally described as Lineage− Sca1+ Kit+ (LSK) cells [21, 22] which now include self-renewing long-term HSC (LT-HSC), short-term HSC (ST-HSC), and nonself-renewing multipotent progenitors (MPPs) [23–26]. MPPs can be further divided into MPP2, MPP3, and MPP4 [25, 26]. Among these, LT-HSC, ST-HSC, MPP2, and MPP3 do not express Flt3 [26]. As these populations all have in vivo DC potentials [27] and all express Kit, we wondered whether Kit ligand (KitL or SCF) could enhance in vitro cDC1 production via the recruitment of these HSCs and MPPs into Flt3L culture.
Results
KitL treatment leads to enhanced in vitro production of all DC subsets
We used two conditions to test whether KitL can enhance in vitro cDC1 generation. In a sequential protocol, we cultured BM in KitL for 3 days, followed by treatment with Flt3L for another 8 days. In a combined protocol, we cultured BM with KitL plus Flt3L for 3 days, followed by treatment with Flt3L for another 8 days (Fig. 1A). Both KitL/Flt3L protocols generated significantly more live cells and cDC1 compared to our standard 8-day culture with Flt3L alone (Fig. 1B and Supporting Information Fig. 1A–E). We also tested the effect of the initial plating density of BM on the overall output of cDC1. Using high density BM plating standard for our Flt3L protocol (2.5 × 106 cells/mL), KitL showed little impact on enhancing cDC1 output (Supporting Information Fig. 1A and B). By contrast, using low density BM plating (6.25 × 105 cells/mL), KitL caused a substantial increase in cDC1 output for both sequential and combined KitL/Flt3L protocols (Supporting Information Fig. 1A and B). Time course analyses showed that both KitL/Flt3L conditions increased the production of cDC1, cDC2, and pDC at all time points compared with Flt3L cultures (Fig. 1C). Notably, the expansion of cDC1 using KitL was sustained over a prolonged period without any apparent plateau until day 11 (Fig. 1C). KitL appeared to enhance cDC1 and cDC2 production to a greater extent than pDC production. Both KitL/Flt3L protocols increased cDC1 production by approximately tenfold compared to Flt3L alone on days 10 and 11 of culture.
Figure 1.
KitL/Flt3L cultures generate large numbers of type 1 conventional DC (cDC1), type 2 conventional DC (cDC2), and plasmacytoid DC (pDC). (A) Scheme of different BM culture methods. (B) Representative flow cytometry plots of 2.5 × 105 BM cells plated at 6.25 × 105 cells/mL and cultured with indicated methods as in (A). Data shown are one of four similar experiments. (C) Numbers of cDC1, cDC2, and pDC derived from per 105 BM with indicated methods at indicated time points.Data shown are one of four similar experiments with four biologically independent samples.Data are mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (one-way ANOVA).
Thrombopoietin (TPO) has been used to expand human CD34+ HSCs and progenitors [28, 29], and an Flt3L + TPO protocol has been shown to derive all three DC subsets from human CD34+ cells [30]. Therefore, we decided to test if TPO could enhance cDC1 generation from mouse BM. Compared to Flt3L alone, Flt3L + TPO culture enhanced cDC1 and pDC production by twofold and fourfold, respectively (Supporting InformationFig. 1D and F). We then attempted multiple combinations to incorporate the Flt3L + TPO protocol to our KitL/Flt3L protocol. In all combinations tested, the incorporation of TPO did not further enhance or only slightly further enhanced cDC1 or pDC production from KitL/Flt3L culture (Supporting Information Fig. 1E and G). As the major enhancement of cDC1 production came from KitL, we decided to focus on the KitL/Flt3L protocols.
KitL/Flt3L-derived cDC1 are developmentally and functionally related to in vivo cDC1
cDC1 development requires Irf8 autoactivation mediated by the Irf8 +32 kb enhancer, as Irf8 +32−/− mice lack cDC1 in vivo and cannot generate cDC1 in Flt3L culture [31]. We cultured BM from WT and Irf8 +32−/− mice using both the sequential and combined KitL/Flt3L protocols. WT but not Irf8 +32−/− BM generated cDC1 (Fig. 2A–C). Conversely, retroviral expression of IRF8 strongly increased cDC1/cDC2 ratios in WT BM KitL/Flt3L culture (Supporting Information Fig. 1H), consistent with previous results from Flt3L culture [18]. Therefore, cDC1 derived from KitL/Flt3L culture are developmentally similar to in vivo cDC1 in terms of IRF8 dependence.
Figure 2.
KitL/Flt3L type 1 conventional DC (cDC1) require the Irf8 +32 kb enhancer. (A) Representative flow cytometry plots of sequential KitL/Flt3L cultures from WT and Irf8 +32−/− BM (pregate: B220− cells). (B) % cDC1 of live singlets from WT and Irf8 +32−/− BM from indicated cultures. Data are pooled from two-independent experiments. Center values indicate mean. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (unpaired, two-tailed Student’s t-test).
Next, we wondered if cDC1 derived from KitL/Flt3L culture are functionally similar to in vivo cDC1 with regard to cytokine production and antigen cross-presentation. Consistent with DC subsets from spleens and Flt3L cultures, cDC1 but not cDC2 derived from KitL/Flt3L protocols could produce IL-12 in response to STAg (Fig. 3A and B, Supporting Information Fig. 2B and C). In the same experiments, we also examined DCs derived from iCD103 culture, a culture system using Flt3L plus GM-CSF that generates CD103+ cDC1 [32] (Supporting Information Fig. 2A). In agreement with spleens, Flt3L alone, and KitL/Flt3L cultures, iCD103-derived cDC1 but not cDC2 produced IL-12 upon STAg stimulation (Fig. 3A and B, Supporting Information Fig. 2B and C). In vitro cross-presentation assays demonstrated that only cDC1, not cDC2, derived from KitL/Flt3L cultures could induce OT-1 T-cell division upon incubation with Abelson-OVA, a form of dead cell-associated antigen (Fig. 3C–E). In summary, cDC1 generated from KitL/Flt3L cultures show functional similarities to bona fide in vivo cDC1.
Figure 3.
KitL/Flt3L type 1 conventional DC (cDC1) can produce IL-12 and can cross-present cell-associated antigen. (A) Representative flow cytometry plots of splenocytes or sequential KitL/Flt3L cultures stimulated with Toxoplasma gondii soluble tachyzoite antigen (STAg). Numbers represent % IL-12 p40+ cells within gated cDC1 and type 2 conventional DC (cDC2) populations, respectively. STAg-stimulated cells are depicted in orange, unstimulated cells are depicted in blue, and unstimulated cells stained with isotype control antibodies are depicted in gray. (B) % IL-12 p40+ cells within gated cDC1 and cDC2 populations from indicated cultures treated with STAg. Data are pooled from two independent experiments. (C) Cell-Trace Violet dilution of OT-1 T cells after in vitro exposure to indicated cDC1 or cDC2 with polyI:C alone (no antigen) or polyI:C plus Abelson-OVA (Abelson-OVA). Numbers represent % divided OT1 T cells (pregate: CD45.2− CD45.1+ CD8α+ Vα2+ cells). (D and E) % divided OT-1 T-cell from indicated conditions. Data are pooled from three independent experiments. Data are mean ± SD.*p < 0.05,**p < 0.01,***p < 0.001,****p < 0.0001 (unpaired, two-tailed Student’s t-test).
Not all in vitro generated DCs that can cross-present in vitro are able to induce tumor regression in vivo. For example, Mo-DCs can cross-present in vitro but cannot induce tumor regression when used as an in situ vaccine in cDC1-deficient Irf8 +32−/− mice due to their inability to migrate to tdLNs [33]. In addition, although iCD103 DCs promote antitumor responses when coadministered with Flt3L, poly I:C, and irradiation [34], it was unknown whether they could induce tumor regression upon in situ vaccination. Therefore, we tested whether KitL/Flt3L and iCD103 cDC1 could induce tumor regression in Irf8 +32−/− mice. As previously described [33], Irf8 +32−/− mice were implanted with an immunogenic fibrosarcoma, 1956-mOVA, and vaccinated intratumorally with three doses of cDC1 or phosphate buffered saline (PBS) (Fig. 4A, Supporting Information Fig. 3A). As expected, PBS-treated Irf8 +32−/− mice failed to control tumor growth and failed to prime antigen-specific CD8 T cells (Fig. 4B–D). In contrast, antigen-specific CD8 T cells were generated in mice treated with cDC1 derived from Flt3L, KitL/Flt3L, or iCD103 cultures, and tumors regressed (Fig. 4B–D). These results suggest that, like Flt3L cDC1, KitL/Flt3L and iCD103 cDC1 can capture tumor antigens and migrate to tumor-draining lymph nodes (tdLNs) to prime CD8 T cells, resulting in tumor regression.
Figure 4.
Both KitL/Flt3L type 1 conventional DC (cDC1) and iCD103 cDC1 can mediate tumor regression in vivo. (A) Scheme of WT BM cultures for cDC1 injections into Irf8 +32−/− mice on day 1, 4, and 8 after tumor inoculation. I.T., intratumoral. (B) Irf8 +32−/− mice were injected with 106 1956-mOVA fibrosarcoma and treated by three doses of 106 cDC1 generated from indicated cultures or PBS. Shown are tumor growth curves for individual mice. Data are pooled from four independent experiments. (C and D) Spleens from tumor-beading mice as in (B) were harvested on day 15 and analyzed for SIINFEKL-Kb tetramer+ CD8 T cells. (C) Representative flow cytometry plots from KitL/Flt3L cDC1- and PBS-treated mice, respectively (pregate: B220− cells). (D) % SIINFEKL-Kb tetramer+ of CD8 T cells from indicated conditions. CD8 T cells are gated as B220− TCRβ+ CD8α+. Data are pooled from four-independent experiments. Center values indicate mean. *p < 0.05 (one-way ANOVA).
KitL treatment recruits Flt3− HSCs and MPPs into Flt3L-mediated in vitro cDC1 generation
We hypothesized that KitL treatment allowed Flt3− HSCs and progenitors to develop in vitro and acquire Flt3 expression, thus contributing to cDC1 generation. To test this, we used cell sorting to purify Flt3− LT-HSC, ST-HSC, MPP2, MPP3, as well as Flt3+ MPP4, MDP, and CDP. We then cultured these populations with Flt3L, KitL/Flt3L, or iCD103 method (Fig. 5A–C). In Flt3L-only cultures, most Flt3− cells failed to survive, and cDC1 arose only from Flt3+ progenitors such as MPP4, MDP, and CDP (Fig. 5E, Supporting Information Fig. 3B and C). In contrast, KitL/Flt3L culture generated cDC1 from all Flt3− populations as well as Flt3+ MPP4 and MDP (Fig. 5D and E, Supporting InformationFig. 3B and C). cDC1 generation from Flt3+ MPP4 was also significantly enhanced by KitL/Flt3L culture (Fig. 5E). iCD103 cultures showed trends toward enhanced cDC1 generation from ST-HSC and MPP4, which did not reach statistical significance. To investigate if Flt3− HSCs and progenitors developed into Flt3+ progenitors in vitro, we performed shorter versions of Flt3L and KitL/Flt3L cultures. Consistent with observations in standard Flt3L and KitL/Flt3L cultures, short-term KitL/Flt3L treatment supported survival of Flt3− HSCs and MPPs, whereas Flt3-only treatment did not (Fig. 5F). Due to exposure to high amounts of Flt3L, no surface Flt3 expression could be detected in either Flt3L or KitL/Flt3L condition (Fig. 5F and G). Upon fixing and permeabilization, Lin− progenitors expressing cytoplasmic Flt3 were identified. Notably, some originally Flt3− HSCs and MPPs started to express intracellular Flt3 in KitL/Flt3L cultures (Fig. 5F and H). KitL/Flt3L culture also significantly expanded MPP4 to generate more Flt3+ progenitors (Fig. 5G and H). These in vitro Flt3+ progenitors also expressed M-CSFR, resembling in vivo MDP and CDP. In summary, these observations demonstrate that KitL can recruit Flt3− HSCs and MPPs, as well as expand MPP4, thereby enhancing cDC1 generation.
Figure 5.
KitL recruit Flt3− HSCs and progenitors and expand MPP4 to enhance in vitro type 1 conventional DC (cDC1) generation by Flt3L. (A) Scheme of HSC/progenitor sorting and culture. (B) Gating strategies for LT-HSC, ST-HSC, MPP2, MPP3, and MPP4. (C) Gating strategies for MDP and CDP. (D) Representative flow cytometry plots of sequential KitL/Flt3L cultures from ST-HSC. (E) Numbers of cDC1 generated from per 103 indicated HSCs or progenitors under culture conditions depicted in (A).Data are pooled from four independent experiments.(F-H) Flt3− HSCs and multipotent progenitors (MPPs) (indicated by the red gate in (B)) or MPP4 were sorted and cultured in Flt3L for 2 days (Flt3L) or in KitL for 3 days and then in Flt3L for 2 days (KitL/Flt3L) and analyzed for surface or intracellular Flt3 expression. (F and G) Representative flow cytometry plots showing surface (top) and intracellular (bottom) Flt3 expression (pregate: Lin− cells).(H) Numbers of intracellular-Flt3+ cells generated from per 103 indicated populations under indicated conditions. Data are pooled from three independent experiments. Data are mean ± SD.*p < 0.05,**p < 0.01,***p < 0.001,****p < 0.0001 (one-way ANOVA in (E), unpaired, two-tailed Student’s t-test in (H)).
Discussion
In summary, we present two KitL/Flt3L protocols that increase in vitro production of cDC1 and cDC2 by tenfold compared to the established Flt3L culture. We also showed that cDC1 generated from either KitL/Flt3L cultures or a previously published iCD103 method are sufficient for tumor regression in cDC1-deficient hosts. KitL/Flt3L protocols have the following technical distinctions from iCD103 protocols. First, KitL/Flt3L cultures generate all three DC subsets in large numbers compared to predominantly cDC1 generated by iCD103 culture [32]. Hence, the KitL/Flt3L system provides a useful tool for studying not only cDC1 but also cDC2 and pDC. Second, KitL/Flt3L protocols are somewhat shorter than iCD103 protocols, generating large numbers of cDC1 in 11 days as compared to 16 days. Moreover, our study demonstrates a clear mechanism for the enhanced cDC1 production with KitL/Flt3L protocols, where KitL supports in vitro development of Flt3− HSCs and MPPs into Flt3+ progenitors to participate in Flt3L-mediated cDC1 generation.
Several protocols have been reported to generate human cDC1 [35–39]. These use combinations of Flt3L, KitL, GM-CSF, IL-4 (FSG4) [35, 36, 38] or combinations of cytokines and stromal cells, such as MS5 cells with Flt3L, KitL, GM-CSF (MS5 + FSG) [37, 38], and OP9-DL1 cells together with FSG [39]. Although KitL is a common component in these protocols, no mechanistic basis has been reported for its inclusion. Our data with mouse BM suggest that KitL may act to recruit and expand human HSCs and other Flt3− progenitors into cDC1 generation. However, further studies are necessary to validate this hypothesis.
Materials and methods
Mice
WT C57BL6/J mice were obtained from the Jackson laboratory. Irf8 +32−/− mice have been described [31]. Both sexes were used between 6 and 12 weeks of age for all experiments. All in vivo experiments were performed in our specific-pathogen free facility. All experiments were performed in accordance with procedures approved by the AAALAC-accredited Animal Studies Committee of Washington University in St Louis and were in compliance with all relevant ethical regulations.
Isolation and culture of bulk BM
BM were harvested from femurs, tibias, and pelvises. Each bone was cut open on both ends and placed in a 0.5 mL microcentrifuge tube with a hole at the bottom, which is placed in a 1.5 mL microcentrifuge tube. The inner 0.5 mL tube carrying the bones is filled with 120 μL MACS buffer (5% fetal calf serum (FCS), 2 mM EDTA in PBS), and BM were flushed into the outer 1.5 mL tube by centrifuging at 5000g for 2 min. Red blood cells were lysed using ammonium chloride-potassium bicarbonate (ACK) buffer. After RBC lysis, cells were washed with MACS buffer and then passed through a 70 μm strainer.
For Flt3L cultures, BM were plated at 2.5 × 106 cells/mL in I10F (Iscove’s Modified Dulbecco’s Medium supplemented with 10% heat-inactivated FCS, L-glutamine, sodium pyruvate, MEM nonessential amino acid, penicillin/streptomycin, and 55 μM β-mercaptoethanol) supplemented with 5% Flt3L-Fc conditioned media and cultured for 8 days unless otherwise specified.
For sequential KitL/Flt3L cultures, BM were plated at 6.25 × 105 cells/mL in I10F supplemented with 5% KitL conditioned media and cultured for 3 days. The medium is then replaced with the same volume of I10F supplemented with 5% Flt3L-Fc conditioned media, and the cells were cultured for another 7–8 days unless otherwise specified. Combined KitL/Flt3L cultures were performed similarly as sequential cultures except that I10F supplemented with 5% KitL conditioned media plus 5% Flt3L-Fc conditioned media were used for the first 3 days. For experiments with recombinant KitL (rKitL/Flt3L cultures), BM were plated in I10F supplemented with 200 ng/mL recombinant KitL (PeproTech) and cultured for 3 days. The medium is then replaced with the same volume of I10F supplemented with 5% Flt3L-Fc conditioned media, and the cells were cultured for another 8 days.
iCD103 cultures were performed as described previously [32] but with a few modifications. Briefly, BM were plated at 7.5 × 105 cells/mL in I10F supplemented with 5% Flt3L-Fc conditioned media and 2.5 ng/mL GM-CSF (PeproTech) and cultured for 9 days. Nonadherent cells were then replated at 1.5 × 105 cells/mL in I10F supplemented with 5% Flt3L-Fc conditioned media plus 2.5 ng/mL GM-CSF for another 7 days unless otherwise specified.
For conditions with TPO, 50 ng/mL recombinant murine TPOs (PeproTech) were used.
Flow cytometry and antibodies
Flow cytometry was performed using a FACSAria Fusion (BD Biosciences). Data were analyzed using FlowJo software. Surface staining was performed at 4°C in the presence of Fc block (2.4G2, BD Biosciences) in MACS buffer.
For the analysis of IL-12 p35 and p40, splenocytes or cultured cells were plated at 5 × 105 cells/mL and stimulated with 1 μg/mL STAg for 6 h in the presence of 250 ng/mL Brefeldin A. The cells were stained for surface markers, fixed with 2% paraformaldehyde in PBS, washed with PBS, and then permeabilized with 0.5% saponin while staining for IL-12 p35 and p40 at 4°C overnight. The procedure for Flt3 intracellular staining was similar to IL-12 p35, p40 intracellular staining except that in the last step, cells were permeabilized with 0.5% saponin while staining for Flt3 at room temperature for 1 h.
The following antibodies/stains were used from BD Biosciences: 7AAD, Kit (2B8), Flt3 (A2F10.1), Clec9A (10B4), streptavidin-FITC; Biolegend: CD3ε (145–2C11), CD11b (M1/70),CD11c (N418), CD19 (6D5), B220 (RA3–6B2), IL-7Rα (A7R34), Ly6C (HK1.4), Ly6G (1A8), Ter119 (Ter-119), MHCII (M5/114.15.2), CD8α (53–6.7), CD24 (M1/69), CD48 (HM48–1), M-CSFR (AFS98), CD150 (TC15–12F12.2), Sirpα (P84), XCR1 (ZET), Sca-1 (E13–161.7), SiglecH (551), TCRβ (H57–597), CD103 (2E7), streptavidin-PE, CD45.1 (A20), CD45.2 (104), Vα2 (B20.1); eBioscience: IL-12/IL-23 p40 (C17.8), rat IgG2a isotype control (sBR2a), streptavidin-APC; Invitrogen: CD11c (N418) CD105 (MJ7/18); Tonbo Biosciences: MHCII (M5/114.15.2); R & D systems: IL-12/IL-35 p35 (27537), mouse IgG1 isotype control (11711).
Isolation and culture of BM HSCs and progenitors
BM were isolated as described above and depleted Lin+ cells by staining with corresponding biotinylated antibodies followed by depletion with MojoSort Streptavidin Nanobeads (Biolegend). The remaining cells were stained with fluorescent antibodies/stains before sorting. Lin markers for ST-HSC, LT-HSC, MPP2, and MPP3 sorting include CD3ε, CD11b, CD11c, CD19, B220, IL-7Rα, Ly6C, Ly6G, Ter119, and MHCII. Lin markers for MPP4, MDP, and CDP sorting include CD3ε, CD11c, CD19, B220, CD105, IL-7Rα, Ly6G, Ter119, and MHCII. A FACSAria Fusion was used for sorting, and cells were sorted into I10F and kept at 4°C until plating. A number of 1.5–2.5 × 103 cells were plated in 200 μL in U-bottom 96-well plates and cultured as described in the isolation and culture of bulk BM section. For experiments that investigate Flt3+ progenitor generation, shorter versions of the culture protocols were used. In those experiments, Flt3L cultures were done for 2 days, and KitL/Flt3L cultures involved 3 days of KitL treatment followed by 2 days of Flt3L treatment.
Tumor lines and growth experiments
Tumor lines were used, and growth experiments were performed as described [33]. Briefly, 1956-mOVA fibrosarcomas derived from frozen stocks were propagated for 2 days in RPMI media supplemented with 10% FCS, washed three times with PBS, and resuspended at 1 × 107 cells/mL in PBS. Irf8 +32−/− mice were subcutaneously injected into the flanks with 106 tumor cells. Tumor growth was measured with a caliper, and tumor area was calculated by the multiplication of two perpendicular diameters. In accordance with our IACUC-approved protocol, maximal tumor diameter was 20 mm in one direction and in no experiments was this limit exceeded. For intratumoral DC injections, 1 × 106 sorted cDC1 were injected intratumorally on days 1, 4, and 8 after tumor implantation, and tumor growth was measured as described above. For analysis of OVA-specific CD8 T cells, spleens were harvested 15 days after tumor transplantation, and SIINFEKL-H2-Kb staining was performed as described [33].
Retroviral transduction
Retroviral constructs expressing Thy1.1 and IRF8-T2A-Thy1.1 have been described [18]. For retroviral packaging, Plat-E cells were plated in 6-well plates at 0.4 × 106 cells/mL and incubated overnight. Retroviral plasmids mixed with TransIT-LT1 (Mirus Bio) in Opti-MEM reduced serum medium were transfected into the Plat-E cells. The culture media were changed the next day, and the supernatant containing retroviruses were collected 48 h posttransfection. For retroviral transduction, ACK-lysed BM were cultured in I10F supplemented with 5% KitL conditioned media or 5% KitL conditioned media together with 5% Flt3L-Fc conditioned media overnight. After removing the culture media, the cells were transduced with the supernatant containing retroviruses in the presence of 2 mg/mL polybrene by centrifugation at 729 × g for 1 h at room temperature. The culture media were changed 24 h later, and the cells were further cultured following the KitL/Flt3L protocols as described in the isolation and culture of bulk BM section.
In vitro cross-presentation assays
For the preparation of Abelson-OVA, MHCI TKO BM transformed into Abelson TKO cell line with v-abl supernatant. Abelson TKO cell line was transduced with MSCV-mOVA-IRES-Thy1.1 retrovirus vector and sorted for Thy1.1+ cells. The cells were cultured and then killed by 3 cycles of freezing in liquid nitrogen and thawing at 37°C.
For in vitro cross-presentation assays, sorted OT-1 T cells were labeled with 1 μM CellTrace Violet (Invitrogen). A number of 12,500 labeled OT1 T cells were then cocultured with 12,500 sorted cDC1 or cDC2 at the presence of 125 ng/mL polyI:C (Sigma-Aldrich) with or without 25,000 Abelson-OVA in U-bottom 96-well plates. Cultures were analyzed on day 3 for CellTrace Violet dilution.
Statistics
Statistical analysis was performed using GraphPad Prism software version 8. When two groups were compared, unpaired, two-tailed Student’s t-test was used to determine significant differences. When multiple groups were compared, one-way ANOVA was used to determine significant differences followed by post hoc comparisons. Means ± SD are displayed in scatter plots with bars, and means are displayed in scatter plots without bars.
Supplementary Material
Acknowledgments:
This work was supported by grants from the NIH (R01AI150297, R01CA248919, and R21AI164142, R01AI162643, and R21AI163421 to K.M.M., and T32CA009621 to M.Y.C.), and a gift from the 1440 Foundation to W.E.G. and K.M.M. S.T.F. is a Cancer Research Institute Irvington Fellows supported by the Cancer Research Institute. We thank Dr. K. Choi for providing CHO cells expressing KitL, Dr. J. Kipnis for providing some of the Irf8 +32−/− mice. We thank R.A. Ohara for advice on in vitro cross-presentation assays.
Abbreviations:
- cDC1
type 1 conventional DC
- cDC2
type 2 conventional DC
- Flt3
Fms-like tyrosine kinase 3
- Flt3L
Flt3 ligand
- KitL
Kit ligand
- LT-HSC
long-term HSC
- MPP
multipotent progenitor
- pDC
plasmacytoid DC
- STAg
Toxoplasma gondii soluble tachyzoite antigen
- ST-HSC
short-term HSC
- tdLN
tumordraining lymph node
- TPO
thrombopoietin
Footnotes
Additional supporting information may be found online in the Supporting Information section at the end of the article.
Conflict of interest statement: The authors declare no commercial or financial conflict of interest.
Data availability statement:
The data that support the findings of this study are available in the supplementary material of this article. Further inquiries are available from the corresponding author upon reasonable request.
References
- 1.Liu K. and Nussenzweig MC, Origin and development of dendritic cells. Immunol. Rev 2010. 234: 45–54. [DOI] [PubMed] [Google Scholar]
- 2.Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, Goldszmid RS, Sher A. et al. , CD8a+ dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 2011. 35: 249–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Murphy TL, Grajales-Reyes GE, Wu X, Tussiwand R, Briseno CG, Iwata A, Kretzer NM et al. , Transcriptional control of dendritic cell development. Annu. Rev. Immunol 2016. 34: 93–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Anderson DA III, Dutertre CA, Ginhoux F. and Murphy KM, Genetic models of human and mouse dendritic cell development and function. Nat. Rev. Immunol 2021. 21: 101–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lewis KL, Caton ML, Bogunovic M, Greter M, Grajkowska LT, Ng D, Klinakis A. et al. , Notch2 receptor signaling controls functional differentiation of dendritic cells in the spleen and intestine. Immunity 2011. 35: 780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Satpathy AT, Briseno CG, Lee JS, Ng D, Manieri NA, KC W, Wu X. et al. , Notch2-dependent classical dendritic cells orchestrate intestinal immunity to attaching-and-effacing bacterial pathogens. Nat. Immunol. 2013. 14: 937–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kumamoto Y, Linehan M, Weinstein JS, Laidlaw BJ, Craft JE and Iwasaki A, CD301b(+) dermal dendritic cells drive T helper 2 cell-mediated immunity. Immunity 2013. 39: 733–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tussiwand R, Everts B, Grajales-Reyes GE, Kretzer NM, Iwata A, Bagaitkar J, Wu X. et al. , Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 2015. 42: 916–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu TT, Kim S, Desai P, Kim DH, Huang X, Ferris ST, Wu R. et al. , Ablation of cDC2 development by triple mutations within the Zeb2 enhancer. Nature 2022. 607: 142–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Swiecki M. and Colonna M, Unraveling the functions of plasmacy-toid dendritic cells during viral infections, autoimmunity and tolerance. Immunol. Rev 2010. 234: 142–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Naik SH, Proietto AI, Wilson NS, Dakic A, Schnorrer P, Fuchsberger M, Lahoud MH et al. , Cutting edge: generation of splenic CD8+ and CD8− dendritic cell equivalents in Fms-like tyrosine kinase 3 ligand bone marrow cultures. J. Immunol 2005. 174: 6592–6597. [DOI] [PubMed] [Google Scholar]
- 12.Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland NG and Lemischka IR, A receptor tyrosine kinase cDNA isolated from a population of enriched primitive hematopoietic cells and exhibiting close genetic linkage to c-kit. Proc. Natl. Acad. Sci. USA 1991. 88: 9026–9030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lyman SD, James L, Zappone J, Sleath PR, Beckmann MP and Bird T, Characterization of the protein encoded by the flt3 (flk2) receptor-like tyrosine kinase gene. Oncogene 1993. 8: 815–822. [PubMed] [Google Scholar]
- 14.Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K. and McKenna HJ, Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J. Exp. Med 1996. 184: 1953–1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Brasel K, De Smedt T, Smith JL and Maliszewski CR, Generation of murine dendritic cells from flt3-ligand-supplemented bone marrow cultures. Blood 2000. 96: 3029–3039. [PubMed] [Google Scholar]
- 16.Kreisel FH, Blasius A, Kreisel D, Colonna M. and Cella M, Interferon-producing cells develop from murine CD31(high)/Ly6C(-) marrow progenitors. Cell. Immunol 2006. 242: 91–98. [DOI] [PubMed] [Google Scholar]
- 17.Grajales-Reyes GE, Iwata A, Albring J, Wu X, Tussiwand R, KC W, Kretzer NM et al. , Batf3 maintains autoactivation of Irf8 for commitment of a CD8alpha(+) conventional DC clonogenic progenitor. Nat. Immunol 2015. 16: 708–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kim S, Bagadia P, Anderson DA III, Liu TT, Huang X, Theisen DJ, O’Connor KW et al. , High amount of transcription factor IRF8 engages AP1-IRF composite elements in enhancers to direct type 1 conventional dendritic cell identity. Immunity 2020. 53: 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara S, Muramatsu S. et al. , Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J. Exp. Med. 1992. 176: 1693–1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sallusto F. and Lanzavecchia A, Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J. Exp. Med 1994. 179: 1109–1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Spangrude GJ, Heimfeld S. and Weissman IL, Purification and characterization of mouse hematopoietic stem cells. Science 1988. 241: 58–62. [DOI] [PubMed] [Google Scholar]
- 22.Ikuta K. and Weissman IL, Evidence that hematopoietic stem cells express mouse c-kit but do not depend on steel factor for their generation. Proc. Natl. Acad. Sci. USA 1992. 89: 1502–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Adolfsson J, Mansson R, Buza-Vidas N, Hultquist A, Liuba K, Jensen CT, Bryder D. et al. , Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 2005. 121: 295–306. [DOI] [PubMed] [Google Scholar]
- 24.Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C. and Morrison SJ, SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005. 121: 1109–1121. [DOI] [PubMed] [Google Scholar]
- 25.Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W, Jaworski M, Offner S. et al. , Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 2008. 135: 1118–1129. [DOI] [PubMed] [Google Scholar]
- 26.Pietras EM, Reynaud D, Kang YA, Carlin D, Calero-Nieto FJ, Leavitt AD, Stuart JM et al. , Functionally distinct subsets of lineage-biased multipotent progenitors control blood production in normal and regenerative conditions. Cell Stem Cell 2015. 17: 35–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Klein F, Roux J, Cvijetic G, Rodrigues PF, von Muenchow L, Lubin R, Pelczar P. et al. , Dntt expression reveals developmental hierarchy and lineage specification of hematopoietic progenitors. Nat. Immunol 2022. 23: 505–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chen W, Antonenko S, Sederstrom JM, Liang X, Chan AS, Kanzler H, Blom B. et al. , Thrombopoietin cooperates with FLT3-ligand in the generation of plasmacytoid dendritic cell precursors from human hematopoietic progenitors. Blood 2004. 103: 2547–2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Balan S, Arnold-Schrauf C, Abbas A, Couespel N, Savoret J, Imperatore F, Villani AC et al. , Large-scale human dendritic cell differentiation revealing notch-dependent lineage bifurcation and heterogeneity. Cell Rep. 2018. 24: 1902–1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Proietto AI, Mittag D, Roberts AW, Sprigg N. and Wu L, The equivalents of human blood and spleen dendritic cell subtypes can be generated in vitro from human CD34(+) stem cells in the presence of Fms-like tyrosine kinase 3 ligand and thrombopoietin. Cell. Mol. Immunol 2012. 9: 446–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Durai V, Bagadia P, Granja JM, Satpathy AT, Kulkarni DH, Davidson JT, Wu R. et al. , Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat. Immunol 2019. 20: 1161–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mayer CT, Ghorbani P, Nandan A, Dudek M, Arnold-Schrauf C, Hesse C, Berod L. et al. , Selective and efficient generation of functional Batf3-dependent CD103+ dendritic cells from mouse bone marrow. Blood 2014. 124: 3081–3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ferris ST, Ohara RA, Ou F, Wu R, Huang X, Kim S, Chen J. et al. , cDC1 vaccines drive tumor rejection by direct presentation independently of host cDC1. Cancer Immunol. Res. 2022. 10: 920–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hammerich L, Marron TU, Upadhyay R, Svensson-Arvelund J, Dhainaut M, Hussein S, Zhan Y. et al. , Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med 2019. 25: 814–824. [DOI] [PubMed] [Google Scholar]
- 35.Poulin LF, Salio M, Griessinger E, Anjos-Afonso F, Craciun L, Chen JL, Keller AM et al. , Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J. Exp. Med 2010. 207: 1261–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Poulin LF, Reyal Y, Uronen-Hansson H, Schraml B, Sancho D, Murphy KM, Hakansson UK et al. , DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and non-lymphoid tissues. Blood 2012. 119: 6052–6062. [DOI] [PubMed] [Google Scholar]
- 37.Lee J, Breton G, Aljoufi A, Zhou YJ, Puhr S, Nussenzweig MC and Liu K, Clonal analysis of human dendritic cell progenitor using a stromal cell culture. J. Immunol. Methods 2015. 425: 21–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Helft J, Bottcher J, Chakravarty P, Zelenay S, Huotari J, Schraml BU, Goubau D. et al. , GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c(+)MHCII(+) macrophages and dendritic cells. Immunity 2015. 42: 1197–1211. [DOI] [PubMed] [Google Scholar]
- 39.Kirkling ME, Cytlak U, Lau CM, Lewis KL, Resteu A, Khodadadi-Jamayran A, Siebel CW et al. , Notch signaling facilitates in vitro generation of cross-presenting classical dendritic cells. Cell Rep.. 2018. 23: 3658–3672. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support the findings of this study are available in the supplementary material of this article. Further inquiries are available from the corresponding author upon reasonable request.





