We describe here that lineage phenotype- negative (Lin)c-kit+ hematopoietic progenitor cells (HPCs) from day 13 postcoitus (dpc) murine fetal liver (FL) can generate dendritic cell (DC) precursors when cultured in vitro in the presence of PA6 stromal cells plus granulocyte/macrophage colony-stimulating factor (GM-CSF) + stem cell factor (SCF) + Flt3 ligand (Flt3L) for 12 to 14 days, and develop into mature DCs when stimulated with GM-CSF plus mouse tumor necrosis factor  (mTNF) for an additional 3 to 5 days. A transwell culture system showed that the generation of DC precursors depended on the support of PA6 cell-secreted soluble factor(s). The mature DCs derived from 13 dpc FL Linc-kit+ HPCs showed characteristic morphology and function of DCs and expressed high levels of Ia, CD86, and CD40 molecules, low levels of DEC205, E-cadherin, and F4/80 molecules, but barely detectable CD11c antigen. Once FL-derived HPCs were cultured without GM-CSF, NK1.1+ cells developed in the presence of PA6 cells + SCF + Flt3L. These NK1.1+ cells could develop into DC precursors at an earlier stage of differentiation by reculturing with PA6 cells + SCF + Flt3L + GM-CSF, but they would be irreversibly committed to NK cell precursors without GM-CSF after 3 days, suggesting that GM-CSF plays a critical role in controlling the transition of DC and NK cell precursors from 13 dpc FL-derived Linc-kit+ HPCs. This study represents the first success in generating mature DCs in vitro from murine FL HPCs. (Blood. 2000;95:138-146)

Dendritic cells (DCs) are professional antigen-presenting cells that are distributed throughout tissues and organs, where they show functional and phenotypic heterogeneity.1-4 DCs can uptake, process, and present antigens on major histocompatibility complex (MHC) class II to induce T cell immune responses, in particular to initiate primary antigen-specific immune reaction.5,6 They also participate in B cell-mediated immune responses,7-9 constituting an integral part of the immune system.10 Recent studies also suggest an important role for DCs in the induction of T cell tolerance. When antigen-bearing DCs are directly injected into the developing thymus or fetal thymic organ cultures, reactive T cells were selectively deleted.11 Moreover, if MHC class II molecules were only expressed by cortical epithelium, but not by DCs in thymic medulla, the propensity to autoimmunity increases.12 All these data suggest that DCs may be involved in the induction of tolerance by deleting autoreactive T cells and the tolerance of T cells to self-antigens during the fetal development.

During embryonic development of hematopoiesis, hematopoietic progenitor cells (HPCs) sequentially appear in yolk sac, paraaortic splanchnopleura, aorta-gonad-mesonephros region (AGM), fetal liver (FL), and finally in the bone marrow (BM).13-18 The FL is considered to be the principle hematopoietic organ during the murine fetal stage until the early neonatal stage, and serves as a reservoir of founder hematopoietic cells generated at early hematopoietic sites within the conceptus.13,17-19 It has been reported that the progenitor cells derived from FL can differentiate in vitro into T lymphocytes,20 natural killer (NK) cells, B lymphocytes, macrophages and mast cells in mouse,21 and T lymphocytes and thymic NK cells in human.22 However, it remains elusive whether murine FL-derived HPCs can differentiate into mature DCs.

It is well established now that DCs can be generated from blood monocytes,23 BM-derived HPCs24-26 and cord blood HPCs27 stimulated with granulocyte/macrophage colony-stimulating factor (GM-CSF), interleukin-4 (IL-4), and tumor necrosis factor α (TNFα). In addition, CD4lo thymic precursors obtained from adult thymus can develop into T cells and thymic DCs bearing CD8α molecule after intrathymic transplantation into irradiated congeneic mice,28,29 indicating the existence of lymphoid-committed DC progenitors. In the fetus, thymic DCs start to develop in the rat embryonic thymus as early as day 17.30 All these observations imply that the development of different DC subsets from embryonic HPCs may be regulated under distinct mechanisms from those of adult BM HPCs.

Because the thymus and spleen form relatively late in gestation and are involved in the production of highly differentiated more mature hematopoietic cells, HPCs presumably seed these organs from other embryonic hematopoietic sites, mostly FL.18,31 To explore the capacity of generating DC from embryonic HPCs, lineage phenotype-negative (Lin)c-kit+HPCs were isolated from day 13 postcoitus (dpc) FL, and cultured in the presence of growth factors and stromal cell line PA6. We found that these FL HPCs possess the capacity to generate DC precursors, which can finally differentiate into mature DCs, in the presence of GM-CSF + stem cell factor (SCF) + Flt3 ligand (Flt3L) and PA6 cells. We also describe that DC precursors in FL-derived HPCs may share progenitor cells in common with NK cells.

Cytokines, antibodies, and cell lines

Recombinant murine SCF and anti-c-kit antibody (ACK-2) were kindly provided by Dr T. Sudo, Basic Research Institute of Toray Co (Kanagawa, Japan), murine GM-CSF by Kirin Brewery Co (Tokyo, Japan), murine macrophage colony-stimulating factor (M-CSF) by Morinaga Co (Tokyo, Japan), and Flt3L by Immunex Co (Seattle, WA). Mouse TNFα (mTNFα) was produced in house.32 These cytokines were used at the optimal concentrations as previously described.26,33 DEC-205 (NLDC145), a rat monoclonal antibody (MoAb) to murine DCs, was a generous gift of Dr R. M. Steinman (Rockefeller University, New York, NY).34,35 The MoAb to mouse E-cadherin was purchased from Dainipon Pharmaceutical Co (Osaka, Japan). Other MoAbs and reagents used for immunostaining were obtained from PharMingen (San Diego, CA) unless otherwise indicated.

BM-derived stromal cell line, PA6 and M-CSF defective stromal cell line, OP9 were kindly provided by Dr S. Nishikawa (Kyoto University, Kyoto, Japan) and Dr T. Nakano (Osaka University, Osaka, Japan), respectively.

Mice

C57BL/6 and BALB/c mice were obtained from CLEA Animal Co (Tokyo, Japan) and maintained under specific pathogen-free conditions in the Animal Facility of the Department of Molecular Preventive Medicine, School of Medicine, The University of Tokyo, Tokyo, Japan. FL were obtained from 13 dpc fetuses of female BALB/c or C57BL/6 mice mated with male C57BL/6 mice. The presence of a postcoital plug was used to determine day 0 of the embryonic age. All animal experiments complied with the standards set out in the Guideline for Care and Use of Laboratory Animals of The University of Tokyo.

Suspension culture of Linc-kit+HPCs

BM Linc-kit+ HPCs were obtained and cultured to induce DCs as previously described.26,33 FL cells were obtained by aspirating FL from 13 dpc murine embryos. Linc-kit+ HPCs were isolated from FL mononuclear cells (MNCs) using a cell sorter (EPICS ELITE, Coulter Electronics, Hialeah, FL) as previously described.26,33 In brief, FL MNCs were subjected to indirect staining using a biotin-conjugated anti-c-kit MoAb and phycoerythrin (PE)-labeled streptavidin, followed by a set of fluorescein isothiocyanate (FITC)-labeled MoAbs to CD(145-2C11), CD4 (H129.19), CD(53-6.7), B220 (RA3-6B2), Gr-1 (Ly-6G), CD11a(2D7), and CD11b (M1/70). The degree of contamination by other types of cells in these preparations was consistently < 0.5% as revealed by an immunofluorescence analysis.

Purified FL-derived Linc-kit+HPCs were incubated at a cell concentration of 3 × 104 cells/mL in Iscove's modified Dulbecco's medium (IMDM; GiBCO, Rockville, MD), supplemented with 20% fetal calf serum (FCS), penicillin G (100 U/mL), and streptomycin (100 μg/mL) in the presence of GM-CSF (4 ng/mL) + SCF (10 ng/mL) + Flt3L (10 ng/mL) + mTNFα (50 ng/mL). Optimal conditions were maintained by splitting these cultures at day 7 and every 2 to 3 days replacing 50% of the medium with a new medium containing fresh cytokines. At the indicated time intervals, morphologic observation with an inverted microscope and immunophenotypical analyses were performed on the cultured cells.

FL-derived Linc-kit+ HPCs were cocultured with PA6 or OP9 stromal cell line as shown in Figure1. In some experiments, FL-derived HPCs were cultured with or without stromal cells in the presence or absence of M-CSF (100 ng/mL) under the above described conditions. In other experiments, the NK 1.1+ cell subset was sorted at day 3, 5, and 7 from Linc-kit+ HPCs, which had been cocultured with PA6 cells in the upper compartments of transwell plates in the presence of SCF and Flt3L. The purity of the sorted cells was more than 99%. The sorted NK1.1+ cell subset was recultured for 10 to 12 days, supplemented with GM-CSF. These cells were collected for analyses after restimulation by GM-CSF + mTNFα for an additional 3 to 5 days. Meanwhile, the sorted NK1.1+ cell subset was also cultured continuously in the absence of GM-CSF for an additional 10 to 12 days. All the staining and sorting procedures were performed in the presence of 1 mM EDTA to avoid cell aggregation.

Fig. 1.

Schematic representation of culture conditions of purified murine FL-derived Linc-kit+ HPCs.

Fig. 1.

Schematic representation of culture conditions of purified murine FL-derived Linc-kit+ HPCs.

Close modal

Immunofluorescence analysis

Immunofluorescence analyses were performed as previously described.26,33 In 2-color analyses, 4 × 105 cells were sequentially incubated with optimal concentrations of biotinylated hamster anti-CD86 or anti-CD11c, and rat anti-DEC-205, anti-E-cadherin or anti-F4/80 MoAbs, followed by FITC-labeled streptavidin and by FITC-conjugated goat antirat IgG (Fab′)2 antibodies (Caltag, Camarillo, CA), respectively, or directly with FITC-labeled anti-CD8α or NK1.1 MoAbs. These cells were finally stained with PE-conjugated mouse antimouse Ia MoAb (AF6-120.1). In other experiments, the cells were first incubated with the appropriate concentration of biotinylated anti-Ly49c MoAb and subsequently labeled by PE-conjugated streptavidin. Then the cells were stained with FITC-conjugated anti-NK1.1 MoAb. In tri-color analysis, the cells were sequentially incubated with rabbit antimouse asialo-GM1 antibody (Wako Co, Osaka, Japan) and PE-conjugated goat antirabbit IgG (Fab′)2 antibody, followed by the incubation with FITC-labeled anti-CD3ε or CD4 and biotinylated anti-NK1.1 MoAbs. The cells were finally stained with Cy-chrome-conjugated streptavidin. The instrument compensation was set in each experiment using single-color or 2-color stained samples. In some experiments, the corresponding cell subpopulations were isolated using a cell sorter.

Endocytosis

The endocytosis experiment was performed as previously described.36,37 Briefly, in the endocytosis test the cells were incubated with 0.1 mg/mL FITC-Dextran (FITC-DX; 4,000 daltons; Sigma Chemicol Co, St Louis, MO) at 0°C or 37°C for 60 minutes. The reaction was stopped by adding ice-cold phosphate-buffered saline (PBS) containing 5% bovine serum albumin and 0.02% sodium azide, and the cells were washed 3 times with 2.5% FCS-0.02% sodium azide-PBS, respectively. Finally, the percentage and density of FITC-positive cells were examined with a cell sorter as previously described.33 

Mixed leukocyte reaction (MLR)

Splenic MNCs were prepared from allogeneic mice as previously described.33 The adherent cells were first removed by incubating them at 37°C for 60 minutes in IMDM medium containing 10% FCS. To obtain highly purified T cells, the nonadherent splenic MNCs were incubated with superparamagnetic MicroBeads conjugated with hamster anti-mouse CD4 MoAb (Miltenyi Biotec, Germany), thereby isolating the CD4+ T cells by magnetic cell sorting. After treatment with mitomycin C (MMC; 15 μg/mL),38 the indicated stimulator cells from FL Linc-kit+ cell culture, BM Linc-kit+ cell culture or peritoneal macrophages (from 100 to 3 × 104 cells) were added to the T cells (3 × 105) in wells of 96-well round-bottomed microtest tissue-culture plates (Nunc, Roskilde, Denmark). After incubating at 37°C for 4 to 5 days, cell proliferation was determined by using 3-(4,5-dimethylthiazolyl-2yl-2,5-diphenyltetrazolium bromide (MTT; Sigma Chemical Co). In brief, 15 μL of MTT (5 mg/mL in PBS) was added into each well and the plates were incubated at 37°C for an additional 4 hours. The resultant absorbance at 550 nm was read by a microplate immunoreader.

Nonspecific esterase (NSE) staining

Cells were cytocentrifuged for 5 minutes at 500 rpm on a microscope slide and used for NSE staining (a-naphthyl acetate esterase staining kit; Sigma Chemical Co), according to the instructions of the manufacturer.

Statistical analysis

Significant differences were evaluated with the Student's ttest. P < .05 were considered to be statistically significant.

Culture conditions for generating DCs from 13 dpc FL-derived Linc-kit+HPCs

We first determined whether culture conditions that lead to the differentiation of adult murine BM HPCs into DCs would also be applicable in FL-derived progenitor cells. The capacity to generate DCs from 13 dpc FL-derived Linc-kit+HPCs in the presence of GM-CSF + SCF + Flt3L and mTNFα was examined after culturing for 7, 14, and 21 days. As shown in Figure2A, there was no typical DC aggregate formation in the cultures. The immunofluorescence analysis showed that < 1.5% cells expressed Ia and CD86 antigens, characteristics of mature DCs (Figure 2B). These results indicated that the combination of GM-CSF + SCF + Flt3L + mTNFα could not induce the generation of mature DCs from 13 dpc FL-derived Linc-kit+ HPCs, whereas combination of these growth factors could induce mature DC differentiation from adult murine BM Linc-kit+ HPCs as reported previously by us.26 

Fig. 2.

The culture of murine 13 dpc FL-derived Linc-kit+ HPCs.

(A) Using a phase contrast microscope, morphologic analyses were performed on cultured murine 13 dpc FL-derived Linc-kit+ HPCs stimulated with GM-CSF + SCF + Flt3L + mTNFα at day 7, 14, and 21. Original magnifications: × 200. (B) 2-color immunofluorescence analysis was performed on the cultured cells at the indicated time points. The cells were sequentially stained with biotinylated CD86 MoAb and PE-labeled Ia MoAb. CD86 was revealed by FITC-streptavidin. The quads were set up on the isotype-matched control dot plot. These results are representative of 3 independent experiments.

Fig. 2.

The culture of murine 13 dpc FL-derived Linc-kit+ HPCs.

(A) Using a phase contrast microscope, morphologic analyses were performed on cultured murine 13 dpc FL-derived Linc-kit+ HPCs stimulated with GM-CSF + SCF + Flt3L + mTNFα at day 7, 14, and 21. Original magnifications: × 200. (B) 2-color immunofluorescence analysis was performed on the cultured cells at the indicated time points. The cells were sequentially stained with biotinylated CD86 MoAb and PE-labeled Ia MoAb. CD86 was revealed by FITC-streptavidin. The quads were set up on the isotype-matched control dot plot. These results are representative of 3 independent experiments.

Close modal

To explore what conditions might be essential for inducing DCs from FL-derived Linc-kit+ HPCs, we first cocultured them with BM-derived stromal cell line PA6 or M-CSF defective stromal cell line OP9 in the presence of GM-CSF + SCF + Flt3L for 12 to 14 days. Because addition of mTNFα significantly inhibited the proliferation of FL Linc-kit+ HPCs (data not shown), this was excluded from the cultures. On the coculture with the PA6 or OP9 cells in the presence of GM-CSF + SCF + Flt3L, FL-derived HPCs did not develop DC aggregates (Figure 3A), and only a few cells were Ia+CD86+ cells (Figure3B). Because our previous findings showed that mTNFα plays a critical role in stimulating the differentiation of DCs from adult murine BM-derived Linc-kit+HPCs,26 the nonadherent cells from FL-derived HPCs cocultured with PA6 stromal cells in the presence of GM-CSF + SCF + Flt3L were stimulated with GM-CSF + mTNFα for an additional 3 to 5 days. As shown in Figure 3A, this resulted in the development of DC-like aggregates surrounded by cells with large sheet-like processes in the periphery. Phenotypically, such cells expressed high levels of Ia, CD86, and CD40, and low levels of F4/80, DEC205, and E-cadherin antigens. However, CD11c molecule was barely detected, and CD8α and NK1.1 antigens were negative (Figures 3B and4A). Giemsa-Wright staining showed that the aggregated cells were morphologically DCs with eccentric nuclei and polarized lamellipodia. They were negative for nonspecific esterase activity (Figure 4B).39 Functionally, these cells could enhance the proliferation of allogeneic T lymphocytes in an MLR assay at a comparable level with DCs generated from adult BM Linc-kit+ HPCs (Figure5). On the contrary, cells derived from OP9 cells + GM-CSF + SCF + Flt3L-induced FL-derived HPCs did not differentiate into mature DCs when restimulated with GM-CSF + mTNFα (Figure 3). These results suggested that DC precursors were generated from murine FL-derived Linc-kit+ HPCs cocultured with PA6 stromal cells in the presence of GM-CSF + SCF + Flt3L, whereas mTNFα and GM-CSF were essential for the final maturation of DCs from these precursors.

Fig. 3.

Generation of DCs from 13dpc FL-derived Linc-kit+ HPCs.

13 dpc FL-derived Linc-kit+ HPCs were cocultured with stromal cell lines PA6 or OP9 cells in the presence of GM-CSF + SCF + Flt3L for 12 to 14 days, and restimulated by GM-CSF + mTNFα for an additional 3 to 5 days. (A) A phase contrast microscopic observation was performed on cocultured FL-derived HPCs with PA6 or OP9 cells in the presence of GM-CSF + SCF + Flt3L for 12 to 14 days before and after replanted into new plates and restimulated by GM-CSF + mTNFα for an additional 3 to 5 days. Original magnification: × 200. (B) Immunophenotypic detection was performed on nonadherent cells from FL-derived HPC cultures at the indicated time points and culture conditions. These cells were sequentially stained with biotinylated CD86 MoAb and PE-labeled Ia MoAb, whereas CD86 was revealed by FITC-streptavidin. The quads were set up on the isotype-matched control dot plot. These results are representative of 3 independent experiments.

Fig. 3.

Generation of DCs from 13dpc FL-derived Linc-kit+ HPCs.

13 dpc FL-derived Linc-kit+ HPCs were cocultured with stromal cell lines PA6 or OP9 cells in the presence of GM-CSF + SCF + Flt3L for 12 to 14 days, and restimulated by GM-CSF + mTNFα for an additional 3 to 5 days. (A) A phase contrast microscopic observation was performed on cocultured FL-derived HPCs with PA6 or OP9 cells in the presence of GM-CSF + SCF + Flt3L for 12 to 14 days before and after replanted into new plates and restimulated by GM-CSF + mTNFα for an additional 3 to 5 days. Original magnification: × 200. (B) Immunophenotypic detection was performed on nonadherent cells from FL-derived HPC cultures at the indicated time points and culture conditions. These cells were sequentially stained with biotinylated CD86 MoAb and PE-labeled Ia MoAb, whereas CD86 was revealed by FITC-streptavidin. The quads were set up on the isotype-matched control dot plot. These results are representative of 3 independent experiments.

Close modal
Fig. 4.

Immunophenotypic and morphologic analyses of DC-like cells.

13dpc FL-derived Linc-kit+ HPCs were first supplemented with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days and then stimulated with GM-SCF + mTNFα for an additional 3 to 5 days. (A) The phenotype of the nonadherent cells was analyzed by 2-color immunofluorescence staining as described in the Materials and Methods. The indicated FITC-labeled MoAbs (CD8α, NK1.1, F4/80, CD40, CD11c, DEC205, and E-cadherin) were used to demonstrate the phenotypic characteristics of the generated DCs. The quads were set up on the isotype-matched control dot plot. (B) Giemsa staining and nonspecific esterase activity analyses were performed on GM-CSF + mTNFα-stimulated DC precursors at day 3 to day 5 that were derived from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days. Original magnification: × 400. These results are representative of 3 independent experiments.

Fig. 4.

Immunophenotypic and morphologic analyses of DC-like cells.

13dpc FL-derived Linc-kit+ HPCs were first supplemented with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days and then stimulated with GM-SCF + mTNFα for an additional 3 to 5 days. (A) The phenotype of the nonadherent cells was analyzed by 2-color immunofluorescence staining as described in the Materials and Methods. The indicated FITC-labeled MoAbs (CD8α, NK1.1, F4/80, CD40, CD11c, DEC205, and E-cadherin) were used to demonstrate the phenotypic characteristics of the generated DCs. The quads were set up on the isotype-matched control dot plot. (B) Giemsa staining and nonspecific esterase activity analyses were performed on GM-CSF + mTNFα-stimulated DC precursors at day 3 to day 5 that were derived from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days. Original magnification: × 400. These results are representative of 3 independent experiments.

Close modal
Fig. 5.

The capacity of the cultured cells to enhance allogeneic MLR.

Allogeneic MLR was performed using purified T cells (3 × 105 cells/well in 96-round-well plate) as responder cells. The unfractionated nonadherent cells, which were generated from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days before and after restimulation by GM-CSF + mTNFα for an additional 3 to 5 days, were treated by MMC and used as stimulator cells at the indicated cell numbers. DCs derived from BM-derived HPCs stimulated with SCF + Flt3L + GM-CSF + mTNFα, and peritoneal macrophages were used as controls. The proliferation of T cells was measured using MTT after 5 days of culture. Results are expressed as the mean ± 1 SD of triplicate cultures and are representative of 3 independent experiments. Black squares indicate macrophages; red diamonds, DCs from BM HPCs; green circles, DCs from FL HPCs; and purple triangles, FL-derived DC precursors.

Fig. 5.

The capacity of the cultured cells to enhance allogeneic MLR.

Allogeneic MLR was performed using purified T cells (3 × 105 cells/well in 96-round-well plate) as responder cells. The unfractionated nonadherent cells, which were generated from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days before and after restimulation by GM-CSF + mTNFα for an additional 3 to 5 days, were treated by MMC and used as stimulator cells at the indicated cell numbers. DCs derived from BM-derived HPCs stimulated with SCF + Flt3L + GM-CSF + mTNFα, and peritoneal macrophages were used as controls. The proliferation of T cells was measured using MTT after 5 days of culture. Results are expressed as the mean ± 1 SD of triplicate cultures and are representative of 3 independent experiments. Black squares indicate macrophages; red diamonds, DCs from BM HPCs; green circles, DCs from FL HPCs; and purple triangles, FL-derived DC precursors.

Close modal

Mediators required for generating DC precursors from FL-derived Linc-kit+HPCs

To elucidate whether the cell-cell contact is required for the generation of DC precursors, transwell plates with 0.4-μm pore size were used in the coculture system. The direct interaction of HPCs and stromal cells could be blocked in the transwell system, while soluble factors could diffuse into the cell cultures. After coculturing with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days, the nonadherent cells in the upper compartments of transwell plates from FL-derived HPCs were transferred into a new 6-well plate and restimulated with GM-CSF + mTNFα. As shown in Figure 6, DC precursors could be generated in the transwell culture system, and these cells could subsequently differentiate into mature DCs by restimulation with GM-CSF + mTNFα.

Fig. 6.

The role of stromal cells in the induction of DCs from FL-derived Linc-kit+ HPCs.

13 dpc FL-derived HPCs were cultured in the presence of cytokines, GM-CSF + SCF + Flt3L, with or without stromal cell line OP9 or PA6 cells in a transwell culture system. M-CSF was added into these cultures as indicated. After 12 to 14 days of culture, the nonadherent cells were replanted in a new 6-well plate, restimulated with GM-CSF + mTNFα for an additional 3 to 5 days and then collected for immunofluorescence analysis. Ia+CD86+ cells were examined by staining with PE-labeled Ia MoAb and biotinylated CD86 MoAb, and revealed by FITC-streptavidin. Results are expressed as the mean ± SD of 3 independent experiments. *P < .05 significance compared with cultures lacking stromal cells or M-CSF addition, or between different stromal cells. Gray bars represent cultures without M-CSF; colored bars, with M-CSF.

Fig. 6.

The role of stromal cells in the induction of DCs from FL-derived Linc-kit+ HPCs.

13 dpc FL-derived HPCs were cultured in the presence of cytokines, GM-CSF + SCF + Flt3L, with or without stromal cell line OP9 or PA6 cells in a transwell culture system. M-CSF was added into these cultures as indicated. After 12 to 14 days of culture, the nonadherent cells were replanted in a new 6-well plate, restimulated with GM-CSF + mTNFα for an additional 3 to 5 days and then collected for immunofluorescence analysis. Ia+CD86+ cells were examined by staining with PE-labeled Ia MoAb and biotinylated CD86 MoAb, and revealed by FITC-streptavidin. Results are expressed as the mean ± SD of 3 independent experiments. *P < .05 significance compared with cultures lacking stromal cells or M-CSF addition, or between different stromal cells. Gray bars represent cultures without M-CSF; colored bars, with M-CSF.

Close modal

In contrast, coculture with OP9 cells in the same condition could not generate DC precursors from FL-derived Linc-kit+ HPCs. Because OP9 cells are deficient in the production of functional M-CSF, we asked whether M-CSF might account for the generation of DC precursors in PA6-stimulated culture. Addition of M-CSF to OP9 cells + GM-CSF + SCF + Flt3L resulted in significant induction of FL HPCs-derived DC precursors that subsequently differentiated into mature DCs by restimulation with GM-CSF + mTNFα. However, the effect of M-CSF on the generation of DC precursors was significantly less than that of PA6 cells + GM-CSF + SCF + Flt3L-stimulated culture. Moreover, addition of M-CSF alone to the combination of cytokines, GM-CSF + SCF + Flt3L, could not induce the generation of DC precursors from FL-derived HPCs. Furthermore, addition of M-CSF to PA6 cells + GM-CSF + SCF + Flt3L-induced culture did not enhance the generation of DC precursors (Figure 6).

Further characterization of DC precursors derived from 13 dpc FL Linc-kit+HPCs

To better understand the cellular basis for mature DC development from 13 dpc FL-derived Linc-kit+HPCs, we analyzed the features of the cells generated at day 12 to day14 in culture with PA6 cells + SCF + Flt3L + GM-CSF. Morphologically, these cells showed monocyte-like cells. Phenotypic analysis demonstrated that these cells expressed high level of CD11b (data not shown). When these nonadherent cells were further examined with a panel of MoAbs, the analysis revealed that these cells expressed high levels of CD40 and E-cadherin and low levels of NK1.1 and Gr-1, but lacked certain DC-associated markers Ia, CD86, and DEC205, whereas CD11c and F4/80 molecules were barely detected. However, CD8α antigen was negative (Figure7). The above evidence indicates that culture with PA6 cells + GM-CSF + SCF + Flt3L is not enough to support the generation of mature DCs from 13 dpc FL-derived Linc-kit+ HPCs.

Fig. 7.

Immunofluorescence analysis on DC precursors generated from murine FL-derived Linc-kit+HPCs.

Immunofluorescence analysis was performed on the nonadherent cells from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days. The indicated markers of FITC-labeled MoAbs were used to demonstrate the phenotypic characteristics of the DC precursors. Solid and dotted lines indicated the immunofluoresence intensity of cells as a control and the test of MoAbs, respectively. Representative results from 3 independent experiments are shown.

Fig. 7.

Immunofluorescence analysis on DC precursors generated from murine FL-derived Linc-kit+HPCs.

Immunofluorescence analysis was performed on the nonadherent cells from FL-derived HPCs cocultured with PA6 cells + GM-CSF + SCF + Flt3L for 12 to 14 days. The indicated markers of FITC-labeled MoAbs were used to demonstrate the phenotypic characteristics of the DC precursors. Solid and dotted lines indicated the immunofluoresence intensity of cells as a control and the test of MoAbs, respectively. Representative results from 3 independent experiments are shown.

Close modal

Functionally, these DC precursors were active in endocytic uptake of FITC-DX at 37°C, but not at 0°C (Figure8). These cells were not effectively able to stimulate allogeneic T-cell proliferation in MLR (Figure 5). When these DC precursors were further stimulated by GM-CSF and mTNFα to differentiate into mature DCs, such endocytic capacity was significantly reduced (Figure 8), and the capacity to stimulate T cell proliferation in allogeneic MLR was considerably enlarged (Figure 5).

Fig. 8.

Endocytic activity of FL HPC-derived DC precursors and mature DCs.

The capacity of FITC-DX uptake was analyzed by a cell sorter as described in “Materials and Methods.” The FITC intensity of cells as a control or the test of FITC-DX uptake was indicated. The results are representative of 3 independent experiments.

Fig. 8.

Endocytic activity of FL HPC-derived DC precursors and mature DCs.

The capacity of FITC-DX uptake was analyzed by a cell sorter as described in “Materials and Methods.” The FITC intensity of cells as a control or the test of FITC-DX uptake was indicated. The results are representative of 3 independent experiments.

Close modal

DCs derived from FL Linc-kit+HPCs share progenitor cells in common with NK cells

Previous reports demonstrated that Sca-1+c-kit+ FL cells could differentiate into lymphocytes.20,21 In our studies, culture of FL-derived Linc-kit+HPCs with PA6 alone for 7 days led to the generation of NK1.1+ cells accounted for 75% of the suspended cells. Addition of SCF + Flt3L further increased the proliferation of NK1.1+ cells. However, GM-CSF significantly inhibited the generation of NK1.1+ cells in PA6 cells + SCF + Flt3L cocultures (Figure 9). The cultured cells derived from 13 dpc FL Linc-kit+HPCs in the presence of PA6 cells + SCF + Flt3L at day 14 were immunophenotypically analyzed. These NK1.1+ cells were CD3ε and CD4 negative, and expressed similar level of asialo-GM1 antigen (Figure 10A) to that of adult murine spleen NK cells (data not shown). They also partly expressed a mature NK cell marker, Ly49c antigen (Figure 10B). The results suggest that these cultured cells possess characteristics of NK cells, but not of NK T cells.

Fig. 9.

Generation of NK1.1+ cells from FL-derived Linc-kit+ HPCs.

13 dpc FL-derived Linc-kit+ HPCs were cocultured with PA6 cells in the presence or absence of SCF + Flt3L with or without GM-CSF in a transwell culture system. The nonadherent cells were collected at the indicated time points and stained with FITC-conjugated anti-NK1.1 MoAb. The data represent mean value ± SD of NK1.1+ cell percentage in the cultures from 5 independent experiments. Black squares represent PA6 cells; red diamonds, PA6 cells with SCF and Flt3L; and green circles, PA6 cells with GM-CSF, SCF, and Flt3L.

Fig. 9.

Generation of NK1.1+ cells from FL-derived Linc-kit+ HPCs.

13 dpc FL-derived Linc-kit+ HPCs were cocultured with PA6 cells in the presence or absence of SCF + Flt3L with or without GM-CSF in a transwell culture system. The nonadherent cells were collected at the indicated time points and stained with FITC-conjugated anti-NK1.1 MoAb. The data represent mean value ± SD of NK1.1+ cell percentage in the cultures from 5 independent experiments. Black squares represent PA6 cells; red diamonds, PA6 cells with SCF and Flt3L; and green circles, PA6 cells with GM-CSF, SCF, and Flt3L.

Close modal
Fig. 10.

Immunophenotypic analysis on NK1.1+ cells generated from FL-derived Linc-kit+ HPCs.

Linc-kit+ HPCs from 13 dpc FL were cultured in the presence of PA6 cells + SCF + Flt3L for 14 days. The phenotypes of the nonadherent cells were analyzed by 2- and tri-color immunofluorescence staining as described in “Materials and Methods.” (A) In the tri-color analyses, expression of asialo-GM1 and CD3ε or CD4 antigens on NK1.1+ cells was analyzed. (B) The 2-color analysis was performed on expression of NK1.1 and Ly49c antigens from the cultured cells. The quads were set up on the isotype-matched control dot plot and the results are representative of 3 independent experiments.

Fig. 10.

Immunophenotypic analysis on NK1.1+ cells generated from FL-derived Linc-kit+ HPCs.

Linc-kit+ HPCs from 13 dpc FL were cultured in the presence of PA6 cells + SCF + Flt3L for 14 days. The phenotypes of the nonadherent cells were analyzed by 2- and tri-color immunofluorescence staining as described in “Materials and Methods.” (A) In the tri-color analyses, expression of asialo-GM1 and CD3ε or CD4 antigens on NK1.1+ cells was analyzed. (B) The 2-color analysis was performed on expression of NK1.1 and Ly49c antigens from the cultured cells. The quads were set up on the isotype-matched control dot plot and the results are representative of 3 independent experiments.

Close modal

To clarify whether DCs share a common progenitor with NK cells, we sorted NK1.1+ cells from PA6 cells + SCF + Flt3L-stimulated FL Linc-kit+ HPCs cultures at the indicated time points, cultured them again in the presence of the PA6 cells + SCF + Flt3L + GM-CSF for an additional 10 to 12 days, and restimulated them with GM-CSF + mTNFα. Mature DCs (> 26%) could be generated from the NK1.1+ cells sorted at day 3 of original culture, but not from a prolonged identical culture of more than 5 days duration (Figure 11). In contrast, flow cytometry analysis revealed that more than 92% of the cells expressed NK1.1 antigen from the continuous culture of the sorted NK1.1+ cells in the absence of GM-CSF for an additional 10 to 12 days. These results suggest that FL Linc-kit+ HPC-derived DCs may share a common progenitor with NK cells.

Fig. 11.

Capacity to generate DC precursors from NK1.1+ cells.

13 dpc FL-derived HPCs were co-cultured with PA6 cells + SCF + Flt3L in transwell plates. At the indicated time points, the nonadherent cells were collected and NK1.1+ cells were sorted and recultured with PA6 cells + GM-CSF + SCF + Flt3L. After being recultured for 10 to 12 days, the nonadherent cells were restimulated with GM-CSF + mTNFα for an additional 3 to 5 days. The expression of Ia and CD86 antigens on these cells was examined as described in “Materials and Methods.” Solid and dotted lines indicated the immunofluoresence intensity of cells stained with a control and the test MoAbs, respectively. The quads were set up on the isotype-matched control dot plot. Representative results from 3 independent experiments are shown.

Fig. 11.

Capacity to generate DC precursors from NK1.1+ cells.

13 dpc FL-derived HPCs were co-cultured with PA6 cells + SCF + Flt3L in transwell plates. At the indicated time points, the nonadherent cells were collected and NK1.1+ cells were sorted and recultured with PA6 cells + GM-CSF + SCF + Flt3L. After being recultured for 10 to 12 days, the nonadherent cells were restimulated with GM-CSF + mTNFα for an additional 3 to 5 days. The expression of Ia and CD86 antigens on these cells was examined as described in “Materials and Methods.” Solid and dotted lines indicated the immunofluoresence intensity of cells stained with a control and the test MoAbs, respectively. The quads were set up on the isotype-matched control dot plot. Representative results from 3 independent experiments are shown.

Close modal

Identification of the progenitors of DCs in the early stages of fetal hematopoiesis is of particular interest in understanding the origin and development of DCs in the establishment of the immune system.1,10-12,40,41 We have described here that purified murine 13 dpc FL Linc-kit+ HPCs can differentiate into DC precursors in vitro after coculture with PA6 stromal cells for 12 to 14 days in the presence of GM-CSF + SCF + Flt3L. These DC precursors can subsequently be induced to develop into mature DCs by further stimulation with GM-CSF + mTNFα for an additional 3 to 5 days. The resultant DCs not only show typical morphologic characteristics and immunophenotype of DCs, but also markedly stimulate T cell proliferation in allogeneic MLR as effectively as adult murine BM Linc-kit+ HPC-derived DCs do,26 suggesting that these cells are mature DCs with antigen-presenting function.

Generation of DC precursors from 13 dpc murine FL Linc-kit+ HPCs in vitro depends on the support of PA6 stromal cell line even in the presence of a cytokine mixture of GM-CSF + SCF + Flt3L, which suffices to induce adult BM Linc-kit+ HPCs to develop into DC precursors.26 In transwell culture, we showed that some soluble factors secreted by PA6 stromal cells together with the cytokine cocktail could induce FL-derived HPCs to develop into DC precursors. Previous studies have shown that the number of Langerhans cells (LCs) is reduced in the skin of osteopetrotic (op/op) mice which is defective in the production of functional M-CSF protein.42 This implied that OP9 stromal cells might induce 13 dpc FL-derived Linc-kit+ HPCs to develop into DCs by the addition of M-CSF. Addition of M-CSF to OP9 cells + GM-CSF + SCF + Flt3L generated DC precursors from FL-derived HPCs to a much less extent than that of PA6 cells and the cytokines-stimulated culture. This result indicates that M-CSF partially compensates for the incapability of OP9 cells to induce the differentiation of DC precursors from FL-derived HPCs. However, the addition of M-CSF to the culture of PA6 cells + GM-CSF + SCF + Flt3L did not enhance the yield of DCs generated from FL-derived Linc-kit+ HPCs. Moreover, M-CSF plus the cytokines GM-CSF + SCF + Flt3L without stromal cells could not induce FL-derived HPCs into DC precursors, indicating that M-CSF cannot substitute for the role of PA6-derived soluble factor(s) which play an essential role in stimulating FL Linc-kit+ HPCs to differentiate into DC precursors.

Several other cytokines have been considered to be the candidates accounting for the generation of DC precursors from FL-derived HPCs. For example, IL-4 and TGF-β have been shown to be essential factors for DC differentiation.43,44 However, addition of IL-4 to the combination of GM-CSF + SCF + Flt3L failed to generate DC precursors from FL-derived HPCs, or anti–IL-4 MoAb did not block the generation of FL-derived DC precursors in the PA6 cells and cytokine cocktail culture system (data not shown). We have recently observed that TGF-β polarizes adult murine BM-derived HPCs to differentiate into monocytes/macrophages that can eventually differentiate into LC-like DCs expressing high level of E-cadherin.45Interestingly, DC precursors generated in our cultures expressed E-cadherin, which is believed to be a specific marker expressed on LCs and to be tightly regulated by TGF-β.46 However, without PA6 cells, addition of TGF-β to the combination of GM-CSF + SCF + Flt3L did not stimulate the generation of DC precursors from FL-derived HPCs. Moreover, both OP9 and PA6 cells expressed IL-4 mRNA, whereas TGF-β mRNA was undetectable in either of them (data not shown). Taken together, these observations indicate that neither IL-4 nor TGF-β substitutes for PA6 cells to induce the generation of DCs from FL-derived HPCs. Thereafter, the collaborative role of IL-4 or serum-derived TGF-β with some PA6 cell-secreted factors may not be ruled out.

Most recently, it has been demonstrated that generation of T cells, NK cells, and DCs in vitro from human FL-derived CD34+D38 cells required the fetal thymic organ cultures,47 in- consistent with our findings that the generation of DC precursors from murine FL HPCs needs the support of PA6 cells. Obviously, the culture conditions reported here are artificial and might not exactly reflect the physiological situation in vivo. PA6 stromal cells were derived from BM.48 It has been shown that PA6 cells can promote the proliferation of BM hematopoietic cells through a short range cell-to-cell interaction by providing an in vitro microenvironment similar to that for in vivo hematopoiesis.49 It is anticipated that the unidentified factor produced by PA6 cells may be available in bone marrow or thymus environments that may skew development of HPCs toward the DC lineage. Biochemical purification of the factor(s) that are responsive for the generationof DC precursors from FL HPCs is in progress, but so far unsuccessful.

FL HPC-derived DCs in this study displayed low levels of DEC205 and E-cadherin and barely detectable CD11c antigen. Previous studies demonstrated that murine splenic DCs express high level of CD11c and can be classified into at least 2 subpopulations CD8α+DEC205+ and CD8αDEC205 cells. On culture in vitro, these mature splenic DCs express high level of DEC205.2 This phenotype of splenic DCs significantly differs from that of mature DCs or their precursors generated from FL-derived Linc-kit+ HPCs in our culture system. We have recently reported that the development of BM Linc-kit+ HPC-derived DCs could be mediated through CD11b−/dullCD11c+ and CD11b+/hi CD11c+ precursors.33 When we evaluated FL Linc-kit+ HPCs, it was found that the cytokerastic kinetics differ between BM- and FL-derived Linc-kit+ HPCs. FL-derived DCs were generated from the precursors on which CD11c antigen was barely expressed. Except for Gr-1 marker, these cells lacked the phenotypic characteristics of other hematopoietic cell lineages, whereas BM Linc-kit+HPC-derived ones expressed high levels of DEC205 and CD11c molecules.26,33 These findings would be reminiscent of the generation of CD11c DC from FL-derived Linc-kit+ HPCs. In human blood, CD11c+ and CD11c DC subsets have been identified.50A recent study demonstrated that CD11c+ and CD11c DC subsets possibly localize in the B-cell and T-cell areas, respectively, and may exert distinct roles in initiating cellular and humoral immune responses.50 Further characterization of the phenotype of DC precursors in vivo in murine fetus will be helpful for understanding DC development and its function in educating the immune system during fetal stage.

Evidence has accumulated indicating that CD40 molecule, a member of TNF receptor family, could be expressed on tonsil DCs,51 blood DCs,52 and LCs53 in human and some murine DCs.33 Moreover, triggering of CD40 ligand and CD40 could stimulate human CD34+ HPCs to differentiate into mature DCs and activate the function of mature DCs.54 Interestingly, our results show that CD40 molecule was highly expressed on DC precursors generated from FL Linc-kit+ HPCs in the presence of PA6 cells + GM-CSF + SCF + Flt3L. Because CD40 and other members of TNF receptor family can bind to their ligands expressed on activated and memory T cells, it is presumed that the stimulatory effects of DCs and T cells are mutual because DCs induce T cells into immune or tolerance state by presenting antigens,1,11,12,54and T cells in turn promote the development of precursors into functionally mature DCs.10,54-56 

It has been reported that NK cells are developmentally close to lymphoid cells in mouse57-59 and in human.60Murine FL-derived Sca-1+c-kit+ cells generated in vitro mixed NK colonies with B220+cells.21 A T cell precursor subset in murine thymus has been shown to be tripotential with full potential to generate DCs and also NK cells although the ability to develop B cells has been lost.59,61,62 Further study has demonstrated that NK cells and DCs may branch off the T cell lineage from a common intermediate bipotential progenitor in human postnatal thymus.63However, it remains to be elucidated which factors may determine the branch-off of DCs from NK and T cells. A remarkable feature from our findings is that murine FL-derived HPCs are able to differentiate into either DC or NK cell precursors, depending on the presence of GM-CSF. When cocultured with PA6 cells, murine FL-derived HPCs could generate DC precursors in the presence of GM-CSF, whereas the development of NK1.1+ cells was drastically suppressed. In the absence of GM-CSF, a large number of NK1.1+ cells proliferated in PA6 cells + SCF + Flt3L-stimulated cultures. Interestingly, the generated NK1.1+ cells could revert to DC precursors at an early stage of differentiation by reculturing with GM-CSF + PA6 cells + SCF + Flt3L. These observations reveal that unlike IL-2, which can simultaneously induce bipotential differentiation of human thymic progenitors into DCs and NK cells in similar proportion in vitro,63 GM-CSF plays a critical role not only in promoting the viability, proliferation, survival, function, and mobilization of DC precursors and DCs, but also in the commitment into DC precursors at the expense of NK cell precursors from FL-derived HPCs. The transition between DC and NK cell precursors suggests that DCs and NK cells may share a common progenitor of fetal hematopoietic origin. Thus, owing to the relationship of NK cells and DCs as well as their dependence on stromal cells in differentiation, we speculate that 13 dpc murine FL Linc-kit+ HPCs may be able to generate thymic DCs under appropriate culture condition. It is believed that CD8α+ DCs represent murine lymphoid DCs that may derive from lymphoid progenitor cells.64 However, it remains to establish a culture system for generating CD8α+ lymphoid DCs in vitro.65 We therefore could not directly compare the NK1.1+ precursor-derived DCs with CD8α+ lymphoid DCs. Further experiments will be performed in vivo to address the relationship of NK1.1+precursor-derived DCs with CD8α+ lymphoid DCs; these experiments are in progress in our laboratory.

It has been demonstrated that the primary stromal cultures from 16 dpc rat embryonic thymus can differentiate into morphologically and phenotypically mature DCs after several days of culture.30In agreement with these results, our findings suggest that FL HPCs gain the capacity to generate DCs as early as 13 dpc. Probably, the embryonic DC precursors have developed in the early embryonic hematopoietic organs such as fetal liver and thymus possibly with the phenotype different from that of the adult counterparts; for example, embryonic DC precursors barely display CD11c molecule. However, the differentiation of these DC precursors may require other factors during the embryonic development. FL Linc-kit+ HPCs may migrate into various tissues where they produce essential factors required for DC development and function in the fetal stage, to generate diverse DC precursors. This study would provide a novel insight into the mechanism of DC ontogeny and their role in the establishment of the immune system.

We express our gratitude to Dr R. M. Steinman (Rockefeller University, New York, NY) for his kind gift of MoAb to DEC-205 (NLDC 145); and Dr Tohru Nakano (Osaka University, Osaka, Japan) and Dr Shin-ichi Nishikawa (Kyoto University, Kyoto, Japan) for their generous gifts of PA6 and OP9 stromal cell lines. We highly appreciate Dr J. J. Oppenheim (NCI-FCRDC, Frederick, MD) for his critical review of the manuscript. We also thank Drs V. Christian, H. Iizasa, and Hongyan Dong for their kind assistance.

Reprints:Kouji Matsushima, Department of Molecular Preventive Medicine, School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Steinman
RM
The dendritic cell system and its role in immunogenicity.
Annu Rev Immunol.
9
1991
271
296
2
Vremec
D
Shortman
K
Dendritic cell subtypes in mouse lymphoid organ: cross-correlation of surface markers, changes with incubation, and difference among thymus, spleen, and lymph nodes.
J Immunol.
159
1997
565
573
3
Vremec
D
Zorbas
M
Scollay
R
et al
The surface phenotype of dendritic cells purified from mouse thymus and spleen: investigation of the CD8 expression by a subpopulation of dendritic cells.
J Exp Med.
176
1992
47
58
4
Steinman
RM
Lustig
DS
Cohn
ZA
Identification of a novel cell type in peripheral lymphoid organs of mice. III. Functional properties in vivo.
J Exp Med.
139
1974
1431
1445
5
Pierre
P
Turley
SJ
Gatti
E
et al
Development regulation of MHC class II transport in mouse dendritic cells.
Nature.
388
1997
787
792
6
Hart
DNJ
Dendritic cells: unique leukocyte populations which control the primary immune response.
Blood.
90
1997
3245
3287
7
Dubois
B
Vanbervliet
B
Fayette
J
et al
Dendritic cells enhance growth and differentiation of CD40-activated B lymphocytes.
J Exp Med.
185
1997
941
951
8
Wykes
M
Pombo
A
Jenkins
C
MacPherson
GG
Dendritic cells interact directly with naive B lymphocytes to transfer antigen and initiate class switching in a primary T-dependent response.
J Immunol.
161
1998
1313
1319
9
Clark
EA
Regulation of B lymphocytes by dendritic cells.
J Exp Med.
185
1997
801
803
10
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature.
392
1998
245
252
11
Brocker
T
Riedinger
M
Karjalainen
K
Targeted expression of major histocompatibility complex (MHC) class II molecules demonstrates that dendritic cells can induce negative but not positive selection of thymocytes in vivo.
J Exp Med.
185
1997
541
550
12
Laufer
TM
Dekoning
J
Markowits
JS
Lo
D
Glimcher
LH
Unopposed positive selection and autoreactivity in mice expressing class II MHC only on thymic cortex.
Nature.
383
1996
81
85
13
Moore
MAS
Metcalf
D
Ontogeny of the haemopoietic system: yolk sac origin of in vivo and in vitro colony forming cells on the developing.
Br J Haematol.
18
1970
279
296
14
Godin
I
Dieterlen-Lievre
F
Cumano
A
Emergence of multipotent hemopoietic cells in the yolk sac and paraaortic splanchnopleura in mouse embryos, beginning at 8.5 days postcoitus.
Proc Natl Acad Sci U S A.
92
1995
773
777
15
Medvinsky
A
Dzierzak
E
Definitive hematopoiesis is autonomously initiated by the AGM region.
Cell.
86
1996
897
906
16
Müller
AM
Medvinsky
A
Strouboulis
J
Grosveld
F
Development of hematopoietic stem cell activity in the mouse embryo.
Immunity.
1
1994
291
301
17
Civin
CI
Gore
SD
Antigenic analysis of hematopoiesis.
J Hematother.
2
1993
137
144
18
Bonifer
C
Faust
N
Geiger
H
Müller
AM
Developmental changes in the differentiation capacity of hematopietic stem cells.
Immunol Today.
19
1998
236
241
19
Johnson
GR
Moore
MAS
Role of stem cell migration in initiation of mouse foetal liver haemopoiesis.
Nature.
258
1975
726
728
20
Kawamoto
H
Ohmura
K
Katsura
Y
Direct evidence for the commitment of hematopoietic stem cells to T, B, and myeloid lineages in murine fetal liver.
Int Immunol.
9
1997
1011
1019
21
Aiba
Y
Ogawa
M
Development of natural killer cells, B lymphocytes, macrophages, and mast cells from single hematopoietic progenitors in culture of murine fetal liver cells.
Blood.
90
1997
3923
3930
22
Jaleco
AC
Blom
B
Res
P
et al
Fetal liver contains committed NK progenitors, but is not a site for development of CD34+ cells into T cells.
J Immunol.
159
1997
694
702
23
Zhou
L-J
Tedder
TF
CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells.
Proc Natl Acad Sci U S A.
93
1996
2588
2592
24
Inaba
K
Inaba
M
Deguchi
M
et al
Granulocytes, macrophages, and dendritic cells arise from a common major histocompatibility complex class II-negative progenitor in mouse bone marrow.
Proc Natl Acad Sci U S A.
90
1993
3038
3042
25
Inaba
K
Inaba
M
Romani
N
et al
Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J Exp Med.
176
1992
1693
1702
26
Zhang
Y
Mukaida
N
Wang
J-B
Harada
A
Akiyama
M
Matsushima
K
Induction of dendritic cell differentiation by granulocyte-macrophage colony-stimulating factor, stem cell factor, and tumor necrosis factor α in vitro from lineage phenotypes-negative c-kit+ murine hematopoietic progenitor cells.
Blood.
90
1997
4842
4853
27
Caux
C
Vanbervliet
B
Massacrier
C
et al
CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to GM-CSF + TNFα.
J Exp Med.
184
1996
695
706
28
Ardavin
C
Wu
L
Li
C-L
Shortman
K
Thymic dendritic cells and T cells develop simultaneously in the thymus from a common precursor population.
Nature.
362
1993
761
763
29
Wu
L
Vremec
D
Ardavin
C
et al
Mouse thymus dendritic cells: kinetics of development and changes in surface markers during maturation.
Eur J Immunol.
25
1995
418
425
30
Vicente
A
Varas
A
Alonso
L
de Moral
MG
Zapata
AG
Ontogeny of rat thymic dendritic cells.
Immunology.
82
1994
75
81
31
Dzierzak
E
Medvinsky
A
de Buijn
M
Qualitative and quantitative aspects of hematopoietic cell development in the mammalian embryo.
Immunol Today.
19
1998
228
236
32
Goeddel
DV
Aggaewal
BB
Gray
PW
et al
Tumor necrosis factors: gene structure and biological activities.
Cold Spring Harb Symp Quant Biol.
1
1986
597
609
33
Zhang
Y
Harada
A
Wang
J-B
et al
Bifurcated dendritic cell differentiation in vitro from murine lineage phenotype-negative c-kit+ bone marrow hematopoietic progenitor cells.
Blood.
92
1998
118
128
34
Kraal
G
Breel
M
Jaanse
M
Bruin
G
Langerhans cells, veiled cells, and interdigitating cells in the mouse recognized by a monoclonal antibody.
J Exp Med.
163
1986
981
997
35
Swiggard
WJ
Mirza
A
Nussenzweig
MC
Steinman
RM
DEC-205, a 250 KD protein abundant on mouse dendritic cells and thymic epithelium that is detected by the monoclonal antibody NLDC-145: purification, characterization and N-terminal amino acid sequence.
Cell Immunol.
165
1995
302
311
36
Winzler
C
Rovere
P
Rescigno
M
et al
Maturation stage of mouse dendritic cells in growth factor-dependent long-term culture.
J Exp Med.
185
1997
317
328
37
Lutz
MB
Rovere
P
Kleijmeer
MJ
et al
Intracellular routes and selective retention of antigens in mildly acidic cathepsin D/lysosome-associated membrane protein-1/MHC class II-positive vesicles in immature dendritic cells.
J Immunol.
159
1997
3707
3716
38
Cohen
PJ
Cohen
PA
Rosenberg
SA
Katz
SI
Mule
JJ
Murine epidermal Langerhans cells and splenic dendritic cells present tumor associated antigens to primed T cells.
Eur J Immunol.
24
1994
315
319
39
MacPherson
G
Properties of lymph-borne (veiled) cells in culture. I. Modulation of phenotype, survival and function: partial dependence on GM-CSF.
Immunology.
68
1989
102
107
40
Swat
W
Ignatowicz
L
von Boehmer
H
Kisielow
P
Colonal deletion of immature CD4+8+ thymocytes in suspension culture by extrathymic antigen-presenting cells.
Nature.
35
1991
150
153
41
Ardavin
C
Thymic dendritic cells.
Immunol Today.
18
1997
350
361
42
Witmer-Pack
MD
Hughes
DA
Schuler
G
et al
Identification of macrophages and dendritic cells in the osteopetrotic (op/op) mouse.
J Cell Sci.
104
1993
1021
1029
43
Palucka
KA
Taquet
N
Sanchez-Chapuis
F
Gluckman
JC
Dendritic cells as the terminal stage of monocyte differentiation.
J Immunol.
160
1998
4587
4595
44
Strobl
H
Riedl
E
Scheinecker
C
et al
TGF-β 1 promotes in vitro development of dendritic cells from CD34+ hematopoietic progenitors.
J Immunol.
157
1996
1499
1507
45
Zhang
Y
Zhang
Y-Y
Ogata
M
et al
Transforming growth factor-β 1 polarizes murine hematopoietic progenitor cells to generate Langerhans cell-like dendritic cells through a monocyte/macrophage differentiation pathway.
Blood.
93
1999
1208
1220
46
Borkowski
TA
Letterio
JJ
Farr
AG
Udey
MC
A role for endogeneous transforming growth factor β 1 in Langerhans cell biology: the skin of transforming growth factor β 1 null mice is devoid of epidermal Langerhans cells.
J Exp Med.
184
1996
2417
2422
47
Plum
J
De Smedt
M
Verhasselt
B
et al
In vitro intrathymic differentiation kinetics of human fetal liver CD34+CD38− progenitors reveals a phenotypically defined dendritic/T-NK precursor split.
J Immunol.
162
1999
60
68
48
Kodama
H
Amagai
Y
Koyama
H
Kasai
S
Hormonal responsiveness of a preadipose cell line derived from newborn mouse calvaria.
J Cell Physiol.
112
1982
83
88
49
Kodama
H
Amagai
Y
Koyama
H
Kasai
S
A new preadipose cell line derived from newborn mouse calvaria can promote the proliferation of pluripotent hemopoietic stem cells in vitro.
J Cell Physiol.
112
1982
89
95
50
Brown
KA
Bedford
P
Macey
M
et al
Human blood dendritic cells: binding to vascular endothelium and expression of adhesion molecules.
Clin Exp Immunol.
107
1997
601
607
51
Hart
DNJ
McKenzie
JL
Isolation and characterization of human tonsil dendritic cells.
J Exp Med.
168
1988
157
170
52
Freudenthal
PS
Steiman
RM
The distinct surface of human blood dendritic cells, as observed after an improved isolation method.
Proc Natl Acad Sci U S A.
87
1990
7698
7702
53
Romani
N
Lenz
A
Glassel
H
et al
Cultured human Langerhans cells resemble lymphoid dendritic cells in phenotype and function.
J Invest Dermatol.
93
1989
600
609
54
Caux
C
Massacrier
C
Vanbervliet
B
et al
Activation of human dendritic cells through CD40 cross-linking.
J Exp Med.
180
1994
1263
1272
55
Wong
BR
Josien
R
Lee
SY
Sauter
B
Li
H-L
Steinman
RM
Choi
Y
TRANCE, a new TNF family member predominantly expressed in T cells is a dendritic cell specific survival factor.
J Exp Med.
186
1997
2075
80
56
Josien
R
Wong
BR
Li
H-L
Steinman
RM
Choi
Y
TRANCE, a TNF family member, is differentially expressed on T cell subsets and induces cytokine production in dendritic cells.
J Immunol.
162
1999
2562
2568
57
Matsuzaki
Y
Gyotoku
J
Ogawa
M
et al
Characterization of c-kit positive intrathymic stem cells that are restricted to lymphoid differentiation.
J Exp Med.
178
1993
1283
1292
58
Rodewald
H-R
Moingeon
P
Lucich
JL
Dosiou
C
Lopez
P
Reinherz
EL
A population of early fetal thymocytes expressing Fcγ RII/III contains precursors of T lymphocytes and natural killer cells.
Cell.
69
1992
139
150
59
Moore
TA
Zlotnik
A
T-cell lineage commitment and cytokine responses of thymic progenitors.
Blood.
86
1995
1850
1860
60
Sanchez
MJ
Muench
MO
Roncarolo
MG
Lanier
LL
Phillips
JH
Identification of a common T/natural killer cell progenitor in human fetal thymus.
J Exp Med.
180
1994
569
576
61
Wu
L
Li
C-L
Shortman
K
Thymic dendritic cell precursors: relationship to the T lymphocyte lineage and phenotype of the dendritic cell progeny.
J Exp Med.
184
1996
903
911
62
Fehling
HJ
von Boehmer
H
Early α β T cell development in the thymus of normal and genetically altered mice.
Curr Opin Immunol.
9
1997
263
275
63
Marquez
C
Trigueros
C
Fraco
JM
et al
Identification of a common developmental pathway for thymic natural killer cells and dendritic cells.
Blood.
91
1998
2760
2771
64
Winkel
K
Sotzik
F
Vremec
D
Cameron
PU
Shortman
K
CD4 and CD8 expression by human and mouse thymic dendritic cells.
Immunol Lett.
40
1994
93
99
65
Varas
A
Vicente
A
Sacedon
R
Zapata
AG
Interleukin-7 influences the development of thymic dendritic cells.
Blood.
92
1998
93
100
Sign in via your Institution