The mononuclear phagocyte system (MPS), composed of monocytes/macrophages and dendritic cells (DCs), plays a critical role at the interface of the innate and adaptive immune systems. However, the simplicity of MPS has been challenged recently by discoveries of novel cellular components. In the current study, we identified the CD135+ subset of monocytes as a novel class of APCs in mice. CD135+ monocytes were readily found in the bone marrow, spleen, and peripheral blood at steady state, and they expressed markers specific to DCs, including MHC class II and CD209a, along with markers for monocytes/macrophages. In addition, this subset phagocytosed bacteria and activated naive T lymphocytes, fulfilling the criteria for APCs. CD135+ monocytes were derived directly from macrophage DC progenitors, not from common monocyte progenitors or other monocytes, suggesting that these are distinct from conventional monocytes. These findings facilitate our understanding of the MPS network that regulates immune responses for host defense.

Visual Abstract

Dendritic cells (DCs) play a central role in the immune system by connecting innate and adaptive immunity. Conventional DCs (cDCs) sense and ingest external Ags and present them on MHC class II, which activates naive T lymphocytes (14). In addition to cDCs, monocyte-derived DCs (moDCs) and plasmacytoid DCs (pDCs) are present in both mice and humans (57). All of these DC types are crucial for immune responses and maintenance of homeostasis. moDCs are induced in response to microbial infections and regulate the function of CD4+ or CD8+ T lymphocytes by presenting ingested Ags (79). pDCs, originating from both myeloid and lymphoid progenitors (10), secrete type I IFN upon their activation by signals mediated through TLR7 and TLR9 during viral infections (11, 12).

DCs and monocytes/macrophages compose the mononuclear phagocyte system (MPS), and they are generally considered to originate from common progenitor cells in the bone marrow (BM), although arguments still remain (1, 1315). Macrophage DC progenitors (MDPs) are considered to reside at the top of the hierarchy within the MPS, giving rise to both monocyte and DC lineages (16). Common DC progenitors (CDPs) (17) and common monocyte progenitors (cMoPs) (18) have been identified as exclusive progenitors for DCs and monocytes, respectively. MDPs and CDPs, both of which are progenitors of cDCs and pDCs, express Flt3 (also known as CD135) on their surface. Flt3 mRNA was detected in short-term hematopoietic stem cells, and the expression peaked in multipotent progenitors (19). Progenitor cells retaining surface CD135 expression have the potential to give rise to DCs, whereas progenitors, which no longer express CD135, lose their ability to differentiate into DCs. Also, the number of DCs is significantly reduced in the absence of Flt3 signaling, suggesting a critical role for the Flt3 ligand (Flt3L)–Flt3 interaction in the development of DC lineages (2, 20, 21).

moDCs are distinct from other DCs as they are generally considered to originate from monocytes rather than from undifferentiated progenitors (6, 22). Under inflammatory conditions, monocytes acquire CD11c and MHC class II expression, which indicates their differentiation into moDCs (2326). In addition, large numbers of moDCs can be obtained by in vitro culture of monocytes (7, 27, 28). Therefore, moDCs have been widely used for intensive characterization of DCs and have been used in immunotherapies (29, 30).

Accumulating evidence has shown that monocytes are a heterogeneous population in mice and humans (14, 31, 32). In mice, monocytes are classified based on the expression level of Ly6C. Ly6Chigh and Ly6Clow monocytes represent “classical” and “nonclassical” monocytes, respectively (33, 34). In addition, recent technical advances, including single-cell analysis, have further extended our understanding of MPS diversity (3537). Hence, elucidation of the cellular components within the MPS is necessary for the precise understanding of the immune system and for their clinical application. During our attempts to characterize monocyte subpopulations, we have identified a unique CD135+ population among cells expressing CD115 and CD11b, the markers that conventionally define monocytes. We propose that this population, based on its functions and differentiation pathway, represents a novel class of APCs.

C57BL/6 mice (CD45.2+, 7–12 wk old) were purchased from CLEA Japan (Tokyo, Japan) or Tokyo Laboratory Animals Science (Tokyo, Japan). BALB/c mice (7–12 wk old) were purchased from Japan SLC (Shizuoka, Japan). CD45.1+ in C57BL/6 background mice were a gift from Shigekazu Nagata (Osaka University, Osaka, Japan). All of the animal protocols were approved by the Committee on Animal Research of the Kyoto University Faculty of Medicine and by the Tokyo University of Pharmacy and Life Sciences Animal Use Committee.

Csf2−/− (38), Csf2rb−/− (39), Flt3l−/− (40), CD11c–diphtheria toxin receptor (DTR) (41), and CX3CR1-GFP mice (42) were maintained in the specific pathogen-free facility at Kanazawa Medical University, and all experiments with these animals were also approved by the Institutional Animal Care Committees of Kanazawa Medical University.

To obtain BM cells, tibia, femur, and humerus were flushed with PBS containing 2% FCS. Peripheral blood (PB) samples were collected from the orbital venous plexus under anesthesia. To lyse RBCs, BM, spleen, or PB cells were treated with Pharm Lyse reagent (BD Biosciences, San Jose, CA). For surface marker staining, cells were stained with the fluorescent marker-conjugated Abs listed in Supplemental Table I. Propidium iodide was used to exclude dead cells.

To analyze the effect of LPS, 5 μg of LPS (Escherichia coli O111:B4, Sigma-Aldrich, St. Louis, MO) was injected i.p. 12 h prior to analysis.

The CD11c-DTR mice were injected i.p. with 4 ng/g body weight DT (Sigma-Aldrich) in PBS. BM cells were analyzed by flow cytometry 1, 2, 3, 4, and 5 d after the DT injection.

BM cells were collected from wild-type, Csf2−/−, Csf2rb−/−, and Flt3l−/− mice and analyzed by flow cytometry.

A BD Cytofix/Cytoperm plus fixation/permeabilization kit with GolgiStop (BD Biosciences) was used for intracellular staining of cytokines. To investigate the effect of LPS on the production of inducible NO synthase (iNOS) or TNF-α, purified cells were incubated in cell culture medium in the presence of 1 μg/ml LPS for 16 h. Flow cytometric analyses or sorting were performed using FACSCanto II or FACSAria III (BD Biosciences). Data were analyzed using FlowJo software (BD Biosciences).

Total RNA was extracted using a RNeasy micro kit (Qiagen, Valencia, CA). Quantity and quality of RNA samples from each mouse were confirmed using an Agilent 2200 TapeStation (Agilent Technologies). The cDNA libraries for next-generation sequencing (NGS) were constructed using a Smart-Seq v4 ultra low input RNA kit for sequencing (Clontech), a Nextera XT DNA library preparation kit (Illumina), and a Nextera XT index kit (Illumina), according to the manufacturers’ instructions. The quality of NGS libraries was assessed using a fragment analyzer (Agilent) and sequenced on a NovaSeq 6000 System (Illumina) using a NovaSeq Xp 4-Lane Kit. Base calls were converted to the fastq file format by bcl2fastq2 conversion software v2.20 (Illumina). Library preparation and sequencing were performed at Takara Bio (Shiga, Japan), and the fastq files were provided to the NGS Core Facility of Kyoto Prefectural University of Medicine for further analyses.

RNA sequencing (RNA-seq) reads from the fastq files were aligned to Ensembl GRCm38/mm10 genome assembly by using Tophat 2.0.9 with Bowtie2 version 2.1.0 and samtools 0.1.19 after performing the quality control with FASTX-Toolkit 0.0.13, FastQC version 0.11.2, and PRINSEQ lite version 0.20.4. Gene expression analysis was performed on FPKM (fragments per kilobase of transcript per million mapped reads) data calculated from the RNA-seq data using Cufflinks 2.2.1. For the cell clustering, principal component and heatmap analyses were carried out using R version 3.6.0. Finally, 4891 genes were selected for analyses based on the results from the Cuffdiff option of Cufflinks under the following quality control conditions: 1) excluding genes where the “status” was not “OK,” 2) excluding genes that showed an error value for “log2(fold_change),” 3) including genes that showed “yes” for “significant,” 4) including genes that resulted in a |log2(fold change)| ≥ 2.0, and 5) excluding genes when more than six samples resulted in FPKM = 0. For the heatmap analysis of myeloid transcription factor genes, the heatmap images were drawn using the pheatmap package after normalizing the FPKM values derived from 27 selected genes by using the zFPKM R/Bioconductor package. The images of RNA-seq data were drawn using the Integrative Genomics Viewer tool (43).

Cytospin specimens were stained using a Diff-Quik stain kit (Sysmex, Kobe, Japan), a modified Wright–Giemsa staining system (44). To analyze the dendrite formation by CD135+ monocytes, cells were incubated for 16 h in RPMI 1640 cell culture medium supplemented with 10 ng/ml GM-CSF (FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan), 10% FCS, 50 μM 2-ME, and penicillin-streptomycin. Images were captured using an Olympus BX43 microscope (Olympus, Tokyo, Japan) connected to a DP80 CCD camera, and processed using cellSens standard 1.12 software (Olympus).

Sorted cells were cultured in RPMI 1640 cell culture medium supplemented with 10% FCS, 50 μM 2-ME, and penicillin-streptomycin with or without 5 ng/ml LPS for 2 h. Then, pHrodo Red E. coli BioParticles conjugate for phagocytosis (Invitrogen, Carlsbad, CA), whose fluorescence was activated only when ingested and acidified by phagocytes, was added to the culture and incubated in a 37°C water bath or on ice for 1 h. The cells were directly analyzed by flow cytometry.

CD4+ T cells were isolated from the spleen of BALB/c (H-2d) or C57BL/6 (H-2b) mice for allogeneic and autologous reactions, respectively, using an EasySep mouse T cell isolation kit (STEMCELL Technologies, Vancouver, BC, Canada) in combination with biotin-conjugated CD8 Abs to remove CD8+ T cells. Purified CD135 monocytes, CD135+ monocytes, cDCs, and pDCs obtained from the BM of C57BL/6 mice were used as test cells. To investigate the effect of LPS, these sorted cells were incubated in cell culture medium containing 5 ng/ml LPS for 2 h and washed twice and then subjected to MLR. Test cells (1 × 104) were incubated with 5 × 104 autologous or allogeneic CD4+ T cells, which had been stained using a CellTrace Violet cell proliferation kit (Invitrogen, Carlsbad, CA) in wells of a 96-well U-bottom culture plate. After 4 d of coculture, proliferation of the T cells was assessed by flow cytometry.

Sorted cells from CD45.1+ or CD45.2+ mice were resuspended in 30 μl of PBS and injected directly into the tibial BM cavity of CD45.2+ or CD45.1+ mice, respectively (45). After 48 (for monocytes), 60 (for progenitors), or 108 h (for CD135+ monocytes and progenitors), BM from the injected tibia and spleen cells of the recipients were collected and subjected to flow cytometric analysis.

Statistical analyses were performed using Microsoft Excel. Statistical differences were determined using a Student t test or the Tukey test. Values of p < 0.05 were considered statistically significant.

Sequence data were deposited to the DNA Data Bank of Japan. Accession numbers for the Genomic Expression Archive and the DNA Data Bank of Japan Sequence Read Archive are E-GEAD-338 (https://ddbj.nig.ac.jp/public/ddbj_database/gea/experiment/E-GEAD-000/E-GEAD-338/) and DRA009516 (https://ddbj.nig.ac.jp/resource/sra-submission/DRA009516), respectively.

Mouse monocytes are identified as CD11b+CD115+ cells within lineage marker (CD3, CD19, NK1.1, Ter119, and Ly6G)–negative (Lin-) BM or PB cells, and they are composed of at least two subpopulations, Ly6Chigh classical and Ly6Clow nonclassical or “patrolling” monocytes (34, 46). During our attempts to identify monocyte subpopulations, we found that a small proportion of monocytes expressed CD135 in the BM (right lower panel in (Fig. 1A). Whereas CD135 monocytes were clearly divided into Ly6Chigh and Ly6Clow subsets, the expression level of Ly6C on CD135+ monocytes was continuously distributed from intermediate to high (i.e., Ly6Cint to Ly6Chigh). These CD135+ monocytes were consistently found in spleen and PB as shown in (Fig. 1B. Giemsa staining revealed that Ly6ChighCD135 monocytes were mononuclear cells with bean-shaped nuclei and that Ly6ClowCD135 monocytes were homogeneously smaller cells with horseshoe-shaped or bilobular nuclei (Fig. 1C). Both Ly6Chigh and Ly6Cint CD135+ monocytes were morphologically similar to the Ly6ChighCD135 monocytes. In this study, we focused on the Ly6Chigh and Ly6Cint CD135+ monocyte subsets.

FIGURE 1.

Identification of CD135+ subsets of monocytes. (A) Flow cytometric analysis of mouse bone marrow (BM) cells. Lineage (Lin) markers and propidium iodide (PI) CD115+ cells (left upper panel) were divided into CD11b and CD11b+ subpopulations (right upper panel). CD11b and CD11b+ subpopulations were further subdivided based on c-Kit/CD135 or Ly6C/CD135 expression, respectively (lower panels). CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor. (B) Flow cytometric analysis of mouse LinCD11b+CD115+ spleen (SPL) and peripheral blood (PB) monocytes. CD135+ cells were detected in the LinCD11b+CD115+ monocyte fraction of BM, spleen, and PB cells (A and B). (C) Giemsa staining of subpopulations of LinCD11b+CD115+ monocytes. Scale bars, 10 μm.

FIGURE 1.

Identification of CD135+ subsets of monocytes. (A) Flow cytometric analysis of mouse bone marrow (BM) cells. Lineage (Lin) markers and propidium iodide (PI) CD115+ cells (left upper panel) were divided into CD11b and CD11b+ subpopulations (right upper panel). CD11b and CD11b+ subpopulations were further subdivided based on c-Kit/CD135 or Ly6C/CD135 expression, respectively (lower panels). CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor. (B) Flow cytometric analysis of mouse LinCD11b+CD115+ spleen (SPL) and peripheral blood (PB) monocytes. CD135+ cells were detected in the LinCD11b+CD115+ monocyte fraction of BM, spleen, and PB cells (A and B). (C) Giemsa staining of subpopulations of LinCD11b+CD115+ monocytes. Scale bars, 10 μm.

Close modal

CD135 is the receptor of Flt3L, an indispensable cytokine for differentiation and maintenance of DCs (2, 21). To assess the significance of CD135 on CD135+ monocytes, we analyzed Flt3l−/− mice (3840) and found that both Ly6Chigh and Ly6Cint CD135+ monocytes in the BM of these mice were significantly decreased when compared with those in wild-type mice (Fig. 2A). Flt3l deficiency also caused significant reduction of MDPs and CDPs, in addition to CD135+ monocytes (Fig. 2D, 2F), whereas the number of Ly6Chigh, Ly6Clow CD135 monocytes or cMoPs was not affected (Fig. 2B, 2C, and 2E, respectively). The numbers of CD135+ monocytes and those of MDPs and CDPs were unchanged in the BM of Csf2−/− mice and Csf2rb−/− mice (3840), suggesting the dependence of CD135+ monocyte differentiation on Flt3L but not on GM-CSF.

FIGURE 2.

Differentiation of Ly6Chigh and Ly6Cint CD135+ monocytes is dependent on Flt3. (AF) The total numbers of Ly6Chigh and Ly6Cint CD135+ monocytes (A), Ly6Chigh and Ly6Clow CD135 monocytes (B and C, respectively), macrophage dendritic cell progenitors (MDPs) (D), common monocyte progenitors (cMoP) (E), and common dendritic cell progenitors (CDPs) (F) in the bone marrow of wild-type (WT), Flt3l−/−, Csf2−/−, and Csf2rb−/− mice are shown. Data are means ± SD of three mice (*p < 0.05, determined by Student t test).

FIGURE 2.

Differentiation of Ly6Chigh and Ly6Cint CD135+ monocytes is dependent on Flt3. (AF) The total numbers of Ly6Chigh and Ly6Cint CD135+ monocytes (A), Ly6Chigh and Ly6Clow CD135 monocytes (B and C, respectively), macrophage dendritic cell progenitors (MDPs) (D), common monocyte progenitors (cMoP) (E), and common dendritic cell progenitors (CDPs) (F) in the bone marrow of wild-type (WT), Flt3l−/−, Csf2−/−, and Csf2rb−/− mice are shown. Data are means ± SD of three mice (*p < 0.05, determined by Student t test).

Close modal

Next, we analyzed the expression of DC markers on CD135+ monocytes (Fig. 3, Supplemental Fig. 1, under the PBS-treated condition). CD11c and MHC class II were highly expressed by cDCs (CD3, CD19, NK1.1, CD11chigh B220 cells in the spleen) and expressed at a low level by CD135 monocytes (Supplemental Fig. 1). CD135+ monocytes expressed CD11c and MHC class II at an intermediate level (Fig. 3A). Accordingly, both Ly6Chigh and Ly6Cint CD135+ monocytes in the BM of CD11c-DTR mice, which express DTR under the control of a CD11c promoter (41), were transiently and significantly reduced by DT administration (Fig. 3B, left panel), whereas CD135 monocytes were not affected (Fig. 3B, right panel).

FIGURE 3.

Expression of dendritic cell markers on CD135+ monocytes. (A) Flow cytometric analysis of CD135+ monocytes. Bone marrow cells were harvested from mice i.p. injected either with PBS or 5 μg of LPS 12 h prior to analysis. Black lines indicate isotype controls. (B) CD11c promoter–diphtheria toxin receptor (DTR) transgenic mice were treated either with vehicle or DT. The numbers of Ly6Chigh and Ly6Cint CD135+ monocytes (left panel) or Ly6Chigh and Ly6Clow CD135 monocytes (right panel) in the bone marrow (BM) are shown. Data are means ± SD of three mice (*p < 0.05, determined by Student t test). (C) Giemsa staining of purified Ly6Chigh and Ly6Cint CD135+ monocytes after a 16-h incubation with recombinant mouse GM-CSF. Scale bars, 10 μm. Data are representative of at least six independent experiments.

FIGURE 3.

Expression of dendritic cell markers on CD135+ monocytes. (A) Flow cytometric analysis of CD135+ monocytes. Bone marrow cells were harvested from mice i.p. injected either with PBS or 5 μg of LPS 12 h prior to analysis. Black lines indicate isotype controls. (B) CD11c promoter–diphtheria toxin receptor (DTR) transgenic mice were treated either with vehicle or DT. The numbers of Ly6Chigh and Ly6Cint CD135+ monocytes (left panel) or Ly6Chigh and Ly6Clow CD135 monocytes (right panel) in the bone marrow (BM) are shown. Data are means ± SD of three mice (*p < 0.05, determined by Student t test). (C) Giemsa staining of purified Ly6Chigh and Ly6Cint CD135+ monocytes after a 16-h incubation with recombinant mouse GM-CSF. Scale bars, 10 μm. Data are representative of at least six independent experiments.

Close modal

CD209a (DC-specific ICAM-3–grabbing nonintegrin [DC-SIGN]), a C-type lectin receptor known to be specific to DCs (35), was highly expressed on CD135+ monocytes but at a marginal level on CD135 monocytes, and the pDC marker Siglec-H (47) was expressed on Ly6Chigh and Ly6Cint CD135+ monocytes at a low level comparable to that on CD135 monocytes (Fig. 3A, Supplemental Fig. 1A).

As previous reports have shown that infection and inflammation promote differentiation and maturation of moDCs (48), we investigated the effects of LPS administration on DC marker expression on CD135+ monocytes. When wild-type mice were administered LPS i.p., the numbers of Lin CD115-expressing cells, including MDPs, cMoPs, CDPs, and all subsets of monocytes, were significantly decreased (data not shown). In response to LPS, expression of CD11c and MHC class II on CD135+ monocytes was markedly upregulated (Fig. 3A). The costimulation molecules CD40, CD80, and CD86 were expressed at low levels by CD135+ monocytes in the steady state, but highly upregulated in response to in vivo LPS stimulation (Fig. 3A). When Ly6Chigh and Ly6Cint CD135+ monocytes were cultured in the presence of GM-CSF for 16 h, dendrites were induced, indicating differentiation toward DC-like cells (Fig. 3C).

In addition, we analyzed the expression of other myeloid cells markers. The macrophage marker F4/80 was expressed on both Ly6Chigh and Ly6Cint CD135+ monocytes at a level comparable to that found on Ly6Chigh and Ly6Clow CD135 monocytes (Fig. 3A, Supplemental Fig. 1A). CD16/32, SIRPα, PD-L1, and PD-L2 were also expressed on CD135+ monocytes at levels comparable to those on CD135 monocytes (Supplemental Fig. 2A). CD64, FcεRI, CCR2, and CX3CR1 are expressed on monocytes and/or moDCs (7, 14). We observed that expression levels of these molecules on CD135+ monocytes were similar (CD64, FcεRI, and CCR2) or slightly higher (CX3CR1) than those on Ly6Chigh CD135 monocytes (Supplemental Fig. 2B, 2C). When compared with cDCs type 2 (cDC2s), lower FcεRI and higher CCR2 expression were distinct phenotypes of CD135+ monocytes. cDC2s expressed CX3CR1 in a biphasic manner, but shared expression of other myeloid markers with CD135+ monocytes (Supplemental Fig. 2A, 2B). Collectively, both Ly6Chigh and Ly6Cint CD135+ monocytes harbored characteristics of DCs in addition to monocytic features, and these DC-like phenotypes were enhanced in response to LPS or GM-CSF.

Since we observed that CD135+ monocytes, composing a small minority of LinCD11b+CD115+ monocytes, possessed not only the characteristics of monocytes but also of DCs, we hypothesized that CD135 monocytes are the conventional monocytes and CD135+ monocytes represent a novel distinct subset. Therefore, we analyzed the comprehensive gene expression profiles of these populations by RNA-seq to define their hierarchical and relative position in the MPS (Fig. 4). To this end, we purified Ly6Chigh and Ly6Cint subsets of CD135+ monocytes from BM and compared their transcriptional profiles with those of conventional Ly6Chigh and Ly6Clow monocytes, MDPs, cMoPs, and CDPs obtained from BM and cDC2s (CD11b+CD8 cDCs) obtained from the spleen. cDC2s were chosen because of their similarity to CD135+ monocytes in terms of CD11b expression (49). First, unsupervised clustering of the gene expression showed very similar gene expression profiles for Ly6Chigh and Ly6Cint CD135+ monocytes (Fig. 4A). The first branching in the hierarchical clustering dendrogram was a division into two groups, the progenitor cluster (MDPs, cMoPs, and CDPs) and the differentiated cell cluster (Ly6Chigh and Ly6Clow CD135 monocytes, Ly6Chigh and Ly6Cint CD135+ monocytes, and cDC2s). Within the differentiated cell cluster, CD135+ monocytes were closer to CD135 monocytes than to cDC2s (Fig. 4A). Accordingly, principal component analysis showed that gene expression profiles of these populations were divided into the progenitor group and the differentiated cell group (Fig. 4B). Profiles of the Ly6Chigh and Ly6Cint CD135+ monocytes were distinct from other differentiated cells and resided between Ly6Chigh and Ly6Clow CD135 monocytes and cDC2s, which indicates that CD135+ monocytes possess characteristics intermediate between CD135 monocytes and cDC2s. Among the myeloid transcription factors analyzed (Fig. 4C), Klf4 was expressed by both CD135 and CD135+ monocytes at relatively high levels, while higher expression of Batf3, Irf4, and SpiB were shared by Ly6Chigh and Ly6Cint CD135+ monocytes and cDC2s. Irf8, Tcf4, and Slfn5 were highly expressed by CD135+ monocytes at the level similar to progenitors (Irf8 and Tcf4) or to Ly6ChighCD135 monocytes (Slfn5) (Fig. 4C). SpiC and Hes1 were highly expressed by CD135 monocytes and cDC2s, whereas CD135+ monocytes and progenitors expressed these genes at a lower level (Fig. 4C). The pattern of genes expressed by CD135+ monocytes suggests that they are a distinct subpopulation of monocytes that harbors characteristics intermediate between conventional monocytes and cDCs.

FIGURE 4.

Transcriptional profiling of CD135+ monocytes reveals their distinct identity. (A and B) Hierarchical tree clustering (A) and principal component analysis (B) of the normalized gene expression profiles of the indicated cells. (C) Heatmap of the mRNA expression of myeloid transcription factors in the indicated cells. 6C, Ly6C; cDC2, conventional dendritic cell type 2; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor.

FIGURE 4.

Transcriptional profiling of CD135+ monocytes reveals their distinct identity. (A and B) Hierarchical tree clustering (A) and principal component analysis (B) of the normalized gene expression profiles of the indicated cells. (C) Heatmap of the mRNA expression of myeloid transcription factors in the indicated cells. 6C, Ly6C; cDC2, conventional dendritic cell type 2; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor.

Close modal

Having confirmed that CD135+ monocytes represent a distinct population, we looked for genes highly expressed by these cells (Fig. 5). First, we focused on classical and nonclassical MHC class II genes, which are required for Ag presentation to CD4+ T cells (Fig. 5A, 5B). Ly6CintCD135+ monocytes expressed classical MHC class II genes, including H2-Aa, H2-Ab1, and H2-Eb1, at levels comparable to those of cDC2s, whereas Ly6ChighCD135+ monocytes were similar to Ly6ClowCD135 monocytes. Among the cells examined, nonclassical MHC class II genes such as H2-Oa, H2-Ob, and H2-DMb2 were expressed at the highest levels by cDC2s. The expression level of these genes in Ly6CintCD135+ monocytes was lower than in cDC2s, but higher than in Ly6ChighCD135+ monocytes or conventional CD135 monocytes, which is consistent with the intermediate characteristics of CD135+ monocytes shown in (Fig. 4. Next, we focused on DC-SIGN genes (Fig. 5C). Among the cells examined, Cd209a, Cd209b, Cd209c, Cd209d, and Cd209e were expressed at the highest levels by Ly6Chigh and Ly6Cint CD135+ monocytes. Other genes encoding Fc receptors, complement receptors, TLRs, and integrins were expressed by CD135+ monocytes at levels similar to those by Ly6ChighCD135 monocytes (data not shown).

FIGURE 5.

CD135+ monocytes highly express MHC class II genes and Cd209 genes. (AC) RNA expression of genes in the indicated cells are shown. n = 3 for each cell type. 6C, Ly6C; cDC2, conventional dendritic cell type 2; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor. *p < 0.05 (comparison with Ly6ChighCD135+ monocytes determined by the Tukey test); #p < 0.05 (comparison with Ly6CintCD135+ monocytes determined by the Tukey test).

FIGURE 5.

CD135+ monocytes highly express MHC class II genes and Cd209 genes. (AC) RNA expression of genes in the indicated cells are shown. n = 3 for each cell type. 6C, Ly6C; cDC2, conventional dendritic cell type 2; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor. *p < 0.05 (comparison with Ly6ChighCD135+ monocytes determined by the Tukey test); #p < 0.05 (comparison with Ly6CintCD135+ monocytes determined by the Tukey test).

Close modal

Then, we sought for CD135+ monocytes specific markers and found that Cd301a (Clec-10a) and Cd206 (Mrc1) genes were highly expressed by Ly6Chigh and Ly6Cint CD135+ monocytes (Supplemental Fig. 3). CD301a is a C-type lectin, and CD206, also a C-type lectin, is a mannose receptor. We evaluated the expression of these molecules at the protein level by flow cytometry and confirmed their high expression in Ly6Chigh and Ly6Cint CD135+ monocytes (Supplemental Fig. 3). However, CD301a was also expressed by Ly6ChighCD135 monocytes and the expression of CD206 on CD135+ monocytes was not high enough, suggesting the need for more ideal specific markers. Taken together, the unique expression of MHC class II genes, DC-SIGNs, and other molecules related to recognition of microorganisms, phagocytosis, and Ag presentation by the CD135+ monocyte subpopulation suggests its functional properties within the immune system.

We next analyzed the phagocytosis and Ag presentation abilities of these cells (Fig. 6). Since Ly6Chigh and Ly6Cint CD135+ monocytes share almost identical gene expression profiles, we considered them together, using CD135+ monocytes for further functional analysis.

FIGURE 6.

CD135+ monocytes harbor the potential for phagocytosis and Ag presentation. (A and B) Phagocytic activity of the indicated cells was assessed using fluorescence-labeled bacteria (pHrodo). Purified cells were incubated for 2 h with or without LPS and then subjected to the phagocytosis assay. (C and D) Indicated cells were subjected to an autologous (Auto) or allogeneic (Allo) MLR assay. Flow cytometric analysis of allogeneic T cells cocultured with the indicated cells is shown in (C), and the frequencies of proliferating autologous or those of allogeneic T cells (cells with lower fluorescence than the control cells) are summarized in (D). Dotted lines in (C) indicate the peak position of each cell division. (EH) IFN-γ production by the indicated cells (G and H) during the MLR using allogeneic T cells with or without prior LPS treatment (E and F). cDC, conventional dendritic cell; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor; pDC, plasmacytoid dendritic cell. Dot plots and histograms are representatives of four to nine independent experiments. Dotted lines in (E) indicate the peak position of each cell division. Data are means ± SD of four to nine independent experiments (#p < 0.05 comparison with between LPS(−) and LPS(+); *p < 0.05, comparison between autologous and allogeneic reactions determined by Student t test).

FIGURE 6.

CD135+ monocytes harbor the potential for phagocytosis and Ag presentation. (A and B) Phagocytic activity of the indicated cells was assessed using fluorescence-labeled bacteria (pHrodo). Purified cells were incubated for 2 h with or without LPS and then subjected to the phagocytosis assay. (C and D) Indicated cells were subjected to an autologous (Auto) or allogeneic (Allo) MLR assay. Flow cytometric analysis of allogeneic T cells cocultured with the indicated cells is shown in (C), and the frequencies of proliferating autologous or those of allogeneic T cells (cells with lower fluorescence than the control cells) are summarized in (D). Dotted lines in (C) indicate the peak position of each cell division. (EH) IFN-γ production by the indicated cells (G and H) during the MLR using allogeneic T cells with or without prior LPS treatment (E and F). cDC, conventional dendritic cell; CDP, common dendritic cell progenitor; cMoP, common monocyte progenitor; MDP, macrophage dendritic cell progenitor; pDC, plasmacytoid dendritic cell. Dot plots and histograms are representatives of four to nine independent experiments. Dotted lines in (E) indicate the peak position of each cell division. Data are means ± SD of four to nine independent experiments (#p < 0.05 comparison with between LPS(−) and LPS(+); *p < 0.05, comparison between autologous and allogeneic reactions determined by Student t test).

Close modal

To evaluate phagocytotic activity, CD135+ monocytes (BM), Ly6Chigh and Ly6Clow CD135 monocytes (BM), cDCs (spleen), and pDCs (CD3CD19NK1.1CD11cintB220+ cells in spleen) were purified and incubated for an hour with E. coli labeled with pH-sensitive fluorescence (Fig. 6A, 6B). Ly6Chigh and Ly6Clow CD135 monocytes became fluorescence positive after the incubation, suggesting active phagocytosis and acidification of the phagosome. In contrast, cDCs and pDCs did not show any signs of phagocytosis or phagosome acidification under the same conditions. A marginal but statistically significant increase in the number of fluorescence-positive CD135+ monocytes were detected, although fewer than of Ly6ChighCD135 monocytes. When sorted cells were subjected to longer preincubation on tissue culture plates and then to longer incubation with the labeled bacteria (2 h), cDCs revealed phagocytic activities and the phagocytic activity of CD135 monocytes was enhanced (Supplemental Fig. 4A). Even under this condition, CD135+ monocytes showed phagocytic activities at an intermediate level between CD135 monocytes and cDCs. LPS did not enhance such phagocytic activity of Ly6Chigh and Ly6Clow CD135 or CD135+ monocytes (Fig. 6A, 6B).

For evaluation of Ag-presenting ability, we used an allogeneic MLR assay (50). Purified cells were cocultured with allogeneic CD4+ T cells (Fig. 6C, 6D) or autologous (Fig. 6D) CD4+ T cells that had been labeled with a fluorescent dye in advance. When allogeneic CD4+ T cells were cocultured with cDCs for 4 d, a significant number of T cells with diluted fluorescence emerged, which indicates T cell proliferation in response to Ag presentation by cDCs (Fig. 6C, 6D). As expected, Ly6Chigh and Ly6Clow CD135 monocytes failed to induce any T cell proliferation. In contrast, coculture with allogeneic CD135+ monocytes resulted in proliferation of T cells, demonstrating their Ag-presenting ability (Fig. 6C, 6D). Proliferation index (the total number of divisions divided by the number of cells that went into division) for CD135+ monocytes and cDCs was 1.96 ± 0.28 and 2.24 ± 0.21, respectively (mean ± SD, p = 0.08, n = 6 for CD135+ monocytes and n = 7 for cDCs). Next, we evaluated the changes in the ability of Ag presentation and IFN-γ production in response to LPS (Fig. 6E–H). Allogeneic T cells stimulated by CD135+ monocytes or cDCs proliferated (Fig. 6E, 6F) and produced IFN-γ (Fig. 6G, 6H). The Ag-presenting functions and IFN-γ production of cDCs and CD135+ monocytes were not further enhanced by prestimulating them with LPS, suggesting that Ag presentation is a de novo function of these cells (Fig. 6E–H). These results clearly indicate that CD135+ monocytes possess the ability to present Ags and stimulate T cell proliferation and induce IFN-γ production by the T cells.

Production of iNOS and TNF-α are the specific features common to Ly6Chigh inflammatory monocytes and moDCs (49, 51). Indeed, we found that exposure of Ly6ChighCD135 monocytes to LPS induced iNOS and TNF-α production (Supplemental Fig. 4B–E). CD135+ monocytes also produced iNOS and TNF-α in response to LPS, although at a lower level. Only a few cells of the Ly6ClowCD135 monocyte population expressed TNF-α. Neither cDCs nor pDCs produced iNOS or TNF-α regardless of the presence of LPS (Supplemental Fig. 4B–E).

We next sought to identify the differentiation pathway of CD135+ monocytes. For this purpose, we used a CD45.1/CD45.2 congenic mouse system. In brief, MDPs, cMoPs, or CDPs (1618) obtained from CD45.1 mice were injected directly into the tibial BM cavity of nonirradiated CD45.2 mice (Fig. 7). Flow cytometric analysis of BM and spleen cells of the recipient mice 60 and 108 h after the injection revealed that the donor-derived MDPs gave rise to CD135+ monocytes in addition to CD115CD135+ DCs and CD135- conventional monocytes (Fig. 7A, 7D, 7E). Although CDPs emerged in the BM and the spleen after MDP transfer, MDP-derived cMoPs were observed only in the BM, suggesting that BM is the optimal environment for cMoP development from MDPs (Fig. 7A, 7D). Then, we tested the differentiation potential of cMoPs and CDPs, which are direct progenitors of monocytes and DCs, respectively. In the recipients of cMoPs, the donor-derived cells were all CD135 monocytes, and no donor-derived CD135+ monocytes were detected at both 60 and 108 h after the transfer (Fig. 7B, 7D, 7E). Consistent with a previous report (52), CDPs mostly differentiated into CD115CD135+ DCs in the recipients (Fig. 7C, 7E). CDPs gave rise to cDCs in the spleen of the recipient less efficiently than MDPs (Fig. 7E). Although donor-derived CD135+ monocytes were detectable in the recipients of CDPs, the contribution of CDPs as progenitors of CD135+ monocytes was almost negligible because the recovery of CDP-derived cells in the recipients was very low (Fig. 7C, 7D, 7E). These results indicate that MDPs are the main and direct progenitors of CD135+ monocytes and that CDPs poorly proliferate after transfer and the efficiency of differentiation from CDPs toward CD135+ monocytes is much less than from MDPs.

FIGURE 7.

CD135+ monocytes are mostly derived from MDPs. (AC) Representative flow cytometric analysis of bone marrow (BM) cells of CD45.2 recipient mice that had been adoptively transferred with (A) macrophage dendritic cell progenitors (MDPs), (B) common monocyte progenitors (cMoPs), or (C) common dendritic cell progenitors (CDPs) obtained from the BM of CD45.1 mice. (D) Flow cytometric analysis summary of BM cells or spleen (SPL) of CD45.2 recipient mice 60 h after respective transfers. Mo, monocytes. (E) Summary of flow cytometric analysis of BM cells or SPL cells of CD45.2 recipient mice 108 h after respective transfers of MDP, cMoP, CDP, or CD135+ Mo. Differentiation index was defined as (% in donor-derived indicated cells in tibial BM cells/the number of infused cells) × 108, which is supposed to reflect the differentiation efficiency of the infused cells. Data are means ± SD of six independent experiments (for D) or four independent experiments (for E). (*p < 0.05, **p < 0.01, determined by Student t test).

FIGURE 7.

CD135+ monocytes are mostly derived from MDPs. (AC) Representative flow cytometric analysis of bone marrow (BM) cells of CD45.2 recipient mice that had been adoptively transferred with (A) macrophage dendritic cell progenitors (MDPs), (B) common monocyte progenitors (cMoPs), or (C) common dendritic cell progenitors (CDPs) obtained from the BM of CD45.1 mice. (D) Flow cytometric analysis summary of BM cells or spleen (SPL) of CD45.2 recipient mice 60 h after respective transfers. Mo, monocytes. (E) Summary of flow cytometric analysis of BM cells or SPL cells of CD45.2 recipient mice 108 h after respective transfers of MDP, cMoP, CDP, or CD135+ Mo. Differentiation index was defined as (% in donor-derived indicated cells in tibial BM cells/the number of infused cells) × 108, which is supposed to reflect the differentiation efficiency of the infused cells. Data are means ± SD of six independent experiments (for D) or four independent experiments (for E). (*p < 0.05, **p < 0.01, determined by Student t test).

Close modal

Accumulating evidence shows that moDCs are induced from monocytes under stress conditions or in vitro. In contrast, we have shown that CD135+ monocytes, functioning as APCs such as DCs, readily exist at steady state. Then, we tested the upstream and downstream relationship between CD135 and CD135+ monocytes using adoptive transfer experiments with the CD45.1/CD45.2 congenic mouse system. Ly6ChighCD135 monocytes, Ly6ClowCD135 monocytes, or CD135+ monocytes were purified from BM cells and then adoptively transferred into the tibial BM cavity of nonirradiated mice as mentioned above (Fig. 8). In the recipients of Ly6ChighCD135 monocytes, donor-derived cells were detected in both Ly6Chigh and Ly6Clow CD135 monocytes as previously reported (53, 54), but not in CD135+ monocytes. Ly6ClowCD135 monocytes gave rise only to Ly6ClowCD135 monocytes. CD135+ monocytes did not differentiate into either Ly6Chigh or Ly6Clow CD135 monocytes (Fig. 8) or cDCs (Fig. 7E) in the recipients. These results were consistent with our finding that cMoPs did not give rise to CD135+ monocytes, and they clearly demonstrate that the differentiation pathway of CD135+ monocytes is distinct from that of conventional monocytes.

FIGURE 8.

CD135+ monocytes are neither progenitors nor progeny of CD135 monocytes. Flow cytometric analysis of bone marrow (BM) or spleen (SPL) of CD45.1 recipient mice that had been adoptively transferred with Ly6Chigh or Ly6Clow CD135 monocytes (Mo) or CD135+ Mo obtained from the BM of CD45.2 donor mice. The numbers are frequencies of donor-derived cells among the indicated cells in the recipients. Bar graphs indicate the frequencies among donor-derived CD11b+CD115+ monocytes. Dot plots are representatives of four to eight independent experiments. Data are means ± SD of four to eight independent experiments. (*p < 0.05, determined by Student t test).

FIGURE 8.

CD135+ monocytes are neither progenitors nor progeny of CD135 monocytes. Flow cytometric analysis of bone marrow (BM) or spleen (SPL) of CD45.1 recipient mice that had been adoptively transferred with Ly6Chigh or Ly6Clow CD135 monocytes (Mo) or CD135+ Mo obtained from the BM of CD45.2 donor mice. The numbers are frequencies of donor-derived cells among the indicated cells in the recipients. Bar graphs indicate the frequencies among donor-derived CD11b+CD115+ monocytes. Dot plots are representatives of four to eight independent experiments. Data are means ± SD of four to eight independent experiments. (*p < 0.05, determined by Student t test).

Close modal

In this study, we have identified CD135+ monocytes as novel APCs in the mouse. They are morphologically similar to classical monocytes, found in BM, spleen, and PB at steady state, and express surface markers specific to DCs, in addition to monocytic markers including CD115 and CD11b, which are the hallmarks of mouse monocytes (35, 53). In addition to the phenotypical resemblance to DCs, these CD135+ monocytes are equipped with the ability to phagocytose external Ags and to activate naive T lymphocytes.

In this study, we identified and characterized a CD135+ subpopulation within mouse CD115+CD11b+ monocytes. Moreover, we redefined Ly6Chigh and Ly6Clow conventional monocytes (33, 55) within the CD135 monocytes. The gene expression profile findings support CD135+ monocytes having a distinct identity among known cells within the MPS. We found that Ly6C expression on CD135+ monocytes ranged from intermediate to high levels in a continuous manner and, although Ly6Cint cells expressed higher levels of DC signature molecules, such as MHC class II and CD11c, there were no other significant differences between the gene expression profiles of Ly6Chigh and Ly6Cint cells. Thus, we consider that they are almost identical. In CD135+ monocytes, MHC class II genes were highly expressed at a level almost equivalent to that in cDC2s. To our surprise, CD209 genes known as DC-SIGNs were expressed at the highest levels in all the analyzed cells. These features of CD135+ monocytes reflected their function as APCs demonstrated by our analysis.

CD135+ monocytes exhibit properties of both monocytes and DCs. These features remind us of those of moDCs (1, 7, 9, 13, 14, 49). Typically, moDCs do not exist at steady state but are induced from monocytes under inflammatory conditions, including various infections or GM-CSF stimulation in vitro (7, 48, 56). During the induction of moDCs, monocytes acquire expression of general DC markers including CD11c and MHC class II. In contrast, the CD135+ monocytes in our study existed readily at steady state and expressed DC markers, suggesting that they are different from moDCs. As revealed by others, conventional monocytes are heterogeneous in both mice and humans, and mouse monocytes are classified according to their Ly6C expression level (14, 31, 55). Interestingly, candidates for moDCs precursors have been identified within a Ly6Cint subset (35). In another report, surface MHC class II (intracellular MHC class II+) CD11cCD209a+Ly6Chigh monocytes are shown as precursors of moDCs, giving rise to moDCs in response to GM-CSF and being independent of Flt3L (57). CD135+ monocytes in our study similarly expressed CD209a at a high level, but they also expressed CD11c and MHC class II on their surface. Furthermore, our results demonstrated that differentiation of CD135+ monocytes was clearly dependent on Flt3L. These results suggest that CD135+ monocytes are not identical to known precursors of moDCs. In addition, analysis in the steady state showed that intra-BM transfer of Ly6Chigh CD135 monocytes or CD135+ monocytes did not result in the emergence of the other population, suggesting that these cells are mutually distinct. These results strongly suggest that CD135+ monocytes are novel de novo APCs that exist at steady state. Further studies are needed to investigate the possibility that CD135+ monocytes are some forms of moDCs, and/or that moDCs are independently derived from CD135 monocytes under inflammatory conditions.

CD135+ monocytes have similarities with other cells within MPS. Menezes et al. (57) identified R2 and R3 cells within a CD135+CD115+ cellular fraction. CD135+ monocytes are also similar to pre-cDC2s as found by Schlitzer et al. (58). Pre-cDC2s expressed CD135 and Ly6C. However, R2, R3 cells, and pre-cDC2s were defined within the MHC class II population, whereas most CD135+ monocytes expressed MHC class II, as shown in (Fig. 3A. In addition, R2 cells were negative for CD11C, which is expressed by CD135+ monocytes. Functionally, transfer of CD135+ monocytes did not give rise to cDC2s in the spleen of the recipients (Fig. 7E). Yáñez et al. also identified CD135-expressing cells within CD11b+CD115+ cells (15). However, they were negative for Ly6C and F4/80, both of which were expressed by CD135+ monocytes (Figs. 1A and 3A, respectively). Although we cannot completely rule out the possibility of some overlaps, we have currently concluded that CD135+ monocytes are different from those cells reported by others. Monocytes, macrophages, and DCs, all members of the MPS, share many properties, gene expression profiles, and differentiation pathways (13, 49). Given that CD135+ monocytes are classified as monocytes, we expected that they would be descendants of cMoPs, as cMoPs are considered to be progenitors with limited potential to differentiate into all kinds of monocytes (18). Indeed, we verified that Ly6Chigh and Ly6Clow CD135+ conventional monocytes were derived from cMoPs. However, the vast majority of CD135+ monocytes were differentiated directly from MDPs but never from cMoPs, to our surprise. Considering that CD135+ monocytes have the ability to present Ags even at steady state and have a unique differentiation pathway, it might be better to avoid referring to them as monocytes and instead name them “monocytoid DCs.”

Development of cellular components of the MPS is governed by complex networks of transcriptional regulation, and cell type–specific requirements for transcription factors have been identified such as Klf4 and Irf8 for Ly6Chigh monocytes, and Nr4a1 and Cebpb for Ly6Clow monocytes (35, 5962). We have not identified any specific transcription factors required for the development of CD135+ monocytes so far. However, RNA-seq analysis revealed that CD135+ monocytes expressed Irf8 at a higher level than CD135 conventional monocytes or cDC2s. Because Ly6Chigh monocytes (including both CD135 and CD135+) are markedly reduced in Irf8-deficient mice (62), we hypothesize that Irf8 plays important roles in specification, differentiation, or maintenance of CD135+ monocytes in addition to its known roles in other monocytes and DCs (62, 63). We are currently investigating the molecular mechanisms that regulate the supply of CD135+ monocytes.

In summary, our results clearly showed the existence of a novel APC population with a distinct differentiation pathway among monocytes. CD135+ monocytes are circulating through lymphoid organs and PB in the steady state with the abilities to activate naive T cells by presenting Ags, indicating that they play an important role in protecting homeostasis from external Ags through their immune functions. Recently, novel DCs/DC-like cells have been identified in human PB as well (6466). Redefinition of conventional monocytes as a CD135 subset should be also of great impact on our understanding of MPS. We are currently working to further elucidate the differentiation and functions of CD135+ monocytes under healthy and diseased condition with the aim of better understanding the MPS complex and the immune responses.

We thank Dr. Shigekazu Nagata (Osaka University) for providing CD45.1 mice. We thank Drs. Hiroshi Kawamoto, Junji Uehori and Yosuke Nagahata (Kyoto University) for technical advice. We are also grateful to Yoko Nakagawa for excellent technical assistance.

This work was partly supported by KAKENHI Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science and University Grants 18K08354, 21K19386 and 21H02956 (to H.H.) and 21K08379 (to A.Y.).

N.K.: methodology, validation, formal analysis, investigation, writing – original draft and visualization; C.O.: investigation; Y.T., M. Nakano, and K.T.: validation, formal analysis, and writing – review and editing; A.Y.: validation and formal analysis, investigation, and writing – review and editing; M. Nagao and T.M.: supervision; N.O.: methodology, validation, formal analysis, and writing – review and editing; H.H.: conceptualization, methodology, validation, formal analysis, writing – review and editing, project administration, and funding acquisition.

The sequences presented in this article have been submitted to the DNA Data Bank of Japan under accession numbers E-GEAD-338 and DRA009516.

The online version of this article contains supplemental material.

Abbreviations used in this article:

BM

bone marrow

cDC

conventional DC

cDC2

cDC type 2

CDP

common DC progenitor

cMoP

common monocyte progenitor

DC

dendritic cell

DC-SIGN

DC-specific ICAM-3–grabbing nonintegrin

DTR

diphtheria toxin receptor

Flt3L

Flt3 ligand

FPKM

fragments per kilobase of transcript per million mapped reads

iNOS

inducible NO synthase

Lin

lineage

MDP

macrophage DC progenitor

moDC

monocyte-derived DC

MPS

mononuclear phagocyte system

NGS

next-generation sequencing

PB

peripheral blood

pDC

plasmacytoid DC

RNA-seq

RNA sequencing

1.
Mildner
A.
,
S.
Jung
.
2014
.
Development and function of dendritic cell subsets.
Immunity
40
:
642
656
.
2.
Merad
M.
,
P.
Sathe
,
J.
Helft
,
J.
Miller
,
A.
Mortha
.
2013
.
The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting.
Annu. Rev. Immunol.
31
:
563
604
.
3.
Steinman
R. M.
,
J.
Idoyaga
.
2010
.
Features of the dendritic cell lineage.
Immunol. Rev.
234
:
5
17
.
4.
Murphy
T. L.
,
G. E.
Grajales-Reyes
,
X.
Wu
,
R.
Tussiwand
,
C. G.
Briseño
,
A.
Iwata
,
N. M.
Kretzer
,
V.
Durai
,
K. M.
Murphy
.
2016
.
Transcriptional control of dendritic cell development.
Annu. Rev. Immunol.
34
:
93
119
.
5.
Nakano
H.
,
M.
Yanagita
,
M. D.
Gunn
.
2001
.
CD11c+B220+Gr-1+ cells in mouse lymph nodes and spleen display characteristics of plasmacytoid dendritic cells.
J. Exp. Med.
194
:
1171
1178
.
6.
Chapuis
F.
,
M.
Rosenzwajg
,
M.
Yagello
,
M.
Ekman
,
P.
Biberfeld
,
J. C.
Gluckman
.
1997
.
Differentiation of human dendritic cells from monocytes in vitro.
Eur. J. Immunol.
27
:
431
441
.
7.
Segura
E.
,
S.
Amigorena
.
2013
.
Inflammatory dendritic cells in mice and humans.
Trends Immunol.
34
:
440
445
.
8.
Mildner
A.
,
S.
Yona
,
S.
Jung
.
2013
.
A close encounter of the third kind: monocyte-derived cells.
Adv. Immunol.
120
:
69
103
.
9.
León
B.
,
M.
López-Bravo
,
C.
Ardavín
.
2007
.
Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania.
Immunity
26
:
519
531
.
10.
Rodrigues
P. F.
,
L.
Alberti-Servera
,
A.
Eremin
,
G. E.
Grajales-Reyes
,
R.
Ivanek
,
R.
Tussiwand
.
2018
.
Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells.
Nat. Immunol.
19
:
711
722
.
11.
Guiducci
C.
,
R. L.
Coffman
,
F. J.
Barrat
.
2009
.
Signalling pathways leading to IFN-α production in human plasmacytoid dendritic cell and the possible use of agonists or antagonists of TLR7 and TLR9 in clinical indications.
J. Intern. Med.
265
:
43
57
.
12.
Siegal
F. P.
,
N.
Kadowaki
,
M.
Shodell
,
P. A.
Fitzgerald-Bocarsly
,
K.
Shah
,
S.
Ho
,
S.
Antonenko
,
Y. J.
Liu
.
1999
.
The nature of the principal type 1 interferon-producing cells in human blood.
Science
284
:
1835
1837
.
13.
Guilliams
M.
,
F.
Ginhoux
,
C.
Jakubzick
,
S. H.
Naik
,
N.
Onai
,
B. U.
Schraml
,
E.
Segura
,
R.
Tussiwand
,
S.
Yona
.
2014
.
Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny.
Nat. Rev. Immunol.
14
:
571
578
.
14.
Guilliams
M.
,
A.
Mildner
,
S.
Yona
.
2018
.
Developmental and functional heterogeneity of monocytes.
Immunity
49
:
595
613
.
15.
Yáñez
A.
,
S. G.
Coetzee
,
A.
Olsson
,
D. E.
Muench
,
B. P.
Berman
,
D. J.
Hazelett
,
N.
Salomonis
,
H. L.
Grimes
,
H. S.
Goodridge
.
2017
.
Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes.
Immunity
47
:
890
902.e4
.
16.
Fogg
D. K.
,
C.
Sibon
,
C.
Miled
,
S.
Jung
,
P.
Aucouturier
,
D. R.
Littman
,
A.
Cumano
,
F.
Geissmann
.
2006
.
A clonogenic bone marrow progenitor specific for macrophages and dendritic cells.
Science
311
:
83
87
.
17.
Onai
N.
,
A.
Obata-Onai
,
M. A.
Schmid
,
T.
Ohteki
,
D.
Jarrossay
,
M. G.
Manz
.
2007
.
Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow.
Nat. Immunol.
8
:
1207
1216
.
18.
Hettinger
J.
,
D. M.
Richards
,
J.
Hansson
,
M. M.
Barra
,
A. C.
Joschko
,
J.
Krijgsveld
,
M.
Feuerer
.
2013
.
Origin of monocytes and macrophages in a committed progenitor.
Nat. Immunol.
14
:
821
830
.
19.
Karsunky
H.
,
M.
Merad
,
A.
Cozzio
,
I. L.
Weissman
,
M. G.
Manz
.
2003
.
Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo.
J. Exp. Med.
198
:
305
313
.
20.
Miller
J. C.
,
B. D.
Brown
,
T.
Shay
,
E. L.
Gautier
,
V.
Jojic
,
A.
Cohain
,
G.
Pandey
,
M.
Leboeuf
,
K. G.
Elpek
,
J.
Helft
, et al
Immunological Genome Consortium
.
2012
.
Deciphering the transcriptional network of the dendritic cell lineage.
Nat. Immunol.
13
:
888
899
.
21.
Waskow
C.
,
K.
Liu
,
G.
Darrasse-Jèze
,
P.
Guermonprez
,
F.
Ginhoux
,
M.
Merad
,
T.
Shengelia
,
K.
Yao
,
M.
Nussenzweig
.
2008
.
The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues.
Nat. Immunol.
9
:
676
683
.
22.
Romani
N.
,
S.
Gruner
,
D.
Brang
,
E.
Kämpgen
,
A.
Lenz
,
B.
Trockenbacher
,
G.
Konwalinka
,
P. O.
Fritsch
,
R. M.
Steinman
,
G.
Schuler
.
1994
.
Proliferating dendritic cell progenitors in human blood.
J. Exp. Med.
180
:
83
93
.
23.
Shortman
K.
,
S. H.
Naik
.
2007
.
Steady-state and inflammatory dendritic-cell development.
Nat. Rev. Immunol.
7
:
19
30
.
24.
Rivollier
A.
,
J.
He
,
A.
Kole
,
V.
Valatas
,
B. L.
Kelsall
.
2012
.
Inflammation switches the differentiation program of Ly6Chi monocytes from antiinflammatory macrophages to inflammatory dendritic cells in the colon.
J. Exp. Med.
209
:
139
155
.
25.
Zigmond
E.
,
C.
Varol
,
J.
Farache
,
E.
Elmaliah
,
A. T.
Satpathy
,
G.
Friedlander
,
M.
Mack
,
N.
Shpigel
,
I. G.
Boneca
,
K. M.
Murphy
, et al
2012
.
Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells.
Immunity
37
:
1076
1090
.
26.
Greter
M.
,
J.
Helft
,
A.
Chow
,
D.
Hashimoto
,
A.
Mortha
,
J.
Agudo-Cantero
,
M.
Bogunovic
,
E. L.
Gautier
,
J.
Miller
,
M.
Leboeuf
, et al
2012
.
GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells.
Immunity
36
:
1031
1046
.
27.
Posch
W.
,
C.
Lass-Flörl
,
D.
Wilflingseder
.
2016
.
Generation of human monocyte-derived dendritic cells from whole blood.
J. Vis. Exp.
118
:
54968
.
28.
Zarif
J. C.
,
J. R.
Hernandez
,
J. E.
Verdone
,
S. P.
Campbell
,
C. G.
Drake
,
K. J.
Pienta
.
2016
.
A phased strategy to differentiate human CD14+monocytes into classically and alternatively activated macrophages and dendritic cells.
Biotechniques
61
:
33
41
.
29.
Anguille
S.
,
E. L.
Smits
,
E.
Lion
,
V. F.
van Tendeloo
,
Z. N.
Berneman
.
2014
.
Clinical use of dendritic cells for cancer therapy.
Lancet Oncol.
15
:
e257
e267
.
30.
Palucka
K.
,
J.
Banchereau
.
2013
.
Dendritic-cell-based therapeutic cancer vaccines.
Immunity
39
:
38
48
.
31.
Wong
K. L.
,
J. J.
Tai
,
W. C.
Wong
,
H.
Han
,
X.
Sem
,
W. H.
Yeap
,
P.
Kourilsky
,
S. C.
Wong
.
2011
.
Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets.
Blood
118
:
e16
e31
.
32.
Ziegler-Heitbrock
L.
,
P.
Ancuta
,
S.
Crowe
,
M.
Dalod
,
V.
Grau
,
D. N.
Hart
,
P. J.
Leenen
,
Y. J.
Liu
,
G.
MacPherson
,
G. J.
Randolph
, et al
2010
.
Nomenclature of monocytes and dendritic cells in blood.
Blood
116
:
e74
e80
.
33.
Geissmann
F.
,
S.
Jung
,
D. R.
Littman
.
2003
.
Blood monocytes consist of two principal subsets with distinct migratory properties.
Immunity
19
:
71
82
.
34.
Gordon
S.
,
P. R.
Taylor
.
2005
.
Monocyte and macrophage heterogeneity.
Nat. Rev. Immunol.
5
:
953
964
.
35.
Mildner
A.
,
J.
Schönheit
,
A.
Giladi
,
E.
David
,
D.
Lara-Astiaso
,
E.
Lorenzo-Vivas
,
F.
Paul
,
L.
Chappell-Maor
,
J.
Priller
,
A.
Leutz
, et al
2017
.
Genomic characterization of murine monocytes reveals C/EBPβ transcription factor dependence of Ly6C cells.
Immunity
46
:
849
862.e7
.
36.
Villani
A. C.
,
R.
Satija
,
G.
Reynolds
,
S.
Sarkizova
,
K.
Shekhar
,
J.
Fletcher
,
M.
Griesbeck
,
A.
Butler
,
S.
Zheng
,
S.
Lazo
, et al
2017
.
Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors.
Science
356
:
eaah4573
.
37.
Hamers
A. A. J.
,
H. Q.
Dinh
,
G. D.
Thomas
,
P.
Marcovecchio
,
A.
Blatchley
,
C. S.
Nakao
,
C.
Kim
,
C.
McSkimming
,
A. M.
Taylor
,
A. T.
Nguyen
, et al
2019
.
Human monocyte heterogeneity as revealed by high-dimensional mass cytometry.
Arterioscler. Thromb. Vasc. Biol.
39
:
25
36
.
38.
Dranoff
G.
,
A. D.
Crawford
,
M.
Sadelain
,
B.
Ream
,
A.
Rashid
,
R. T.
Bronson
,
G. R.
Dickersin
,
C. J.
Bachurski
,
E. L.
Mark
,
J. A.
Whitsett
, et al
1994
.
Involvement of granulocyte-macrophage colony-stimulating factor in pulmonary homeostasis.
Science
264
:
713
716
.
39.
Nishinakamura
R.
,
N.
Nakayama
,
Y.
Hirabayashi
,
T.
Inoue
,
D.
Aud
,
T.
McNeil
,
S.
Azuma
,
S.
Yoshida
,
Y.
Toyoda
,
K.
Arai
, et al
1995
.
Mice deficient for the IL-3/GM-CSF/IL-5 β c receptor exhibit lung pathology and impaired immune response, while β IL3 receptor-deficient mice are normal.
Immunity
2
:
211
222
.
40.
Kingston
D.
,
M. A.
Schmid
,
N.
Onai
,
A.
Obata-Onai
,
D.
Baumjohann
,
M. G.
Manz
.
2009
.
The concerted action of GM-CSF and Flt3-ligand on in vivo dendritic cell homeostasis.
Blood
114
:
835
843
.
41.
Jung
S.
,
D.
Unutmaz
,
P.
Wong
,
G.
Sano
,
K.
De los Santos
,
T.
Sparwasser
,
S.
Wu
,
S.
Vuthoori
,
K.
Ko
,
F.
Zavala
, et al
2002
.
In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens.
Immunity
17
:
211
220
.
42.
Jung
S.
,
J.
Aliberti
,
P.
Graemmel
,
M. J.
Sunshine
,
G. W.
Kreutzberg
,
A.
Sher
,
D. R.
Littman
.
2000
.
Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion.
Mol. Cell. Biol.
20
:
4106
4114
.
43.
Robinson
J. T.
,
H.
Thorvaldsdóttir
,
W.
Winckler
,
M.
Guttman
,
E. S.
Lander
,
G.
Getz
,
J. P.
Mesirov
.
2011
.
Integrative genomics viewer.
Nat. Biotechnol.
29
:
24
26
.
44.
Satake
S.
,
H.
Hirai
,
Y.
Hayashi
,
N.
Shime
,
A.
Tamura
,
H.
Yao
,
S.
Yoshioka
,
Y.
Miura
,
T.
Inaba
,
N.
Fujita
, et al
2012
.
C/EBPβ is involved in the amplification of early granulocyte precursors during candidemia-induced “emergency” granulopoiesis.
J. Immunol.
189
:
4546
4555
.
45.
Baba
T.
,
K.
Naka
,
S.
Morishita
,
N.
Komatsu
,
A.
Hirao
,
N.
Mukaida
.
2013
.
MIP-1α/CCL3-mediated maintenance of leukemia-initiating cells in the initiation process of chronic myeloid leukemia.
J. Exp. Med.
210
:
2661
2673
.
46.
Yang
J.
,
L.
Zhang
,
C.
Yu
,
X. F.
Yang
,
H.
Wang
.
2014
.
Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases.
Biomark. Res.
2
:
1
.
47.
Blasius
A. L.
,
M.
Colonna
.
2006
.
Sampling and signaling in plasmacytoid dendritic cells: the potential roles of Siglec-H.
Trends Immunol.
27
:
255
260
.
48.
Cheong
C.
,
I.
Matos
,
J. H.
Choi
,
D. B.
Dandamudi
,
E.
Shrestha
,
M. P.
Longhi
,
K. L.
Jeffrey
,
R. M.
Anthony
,
C.
Kluger
,
G.
Nchinda
, et al
2010
.
Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209+ dendritic cells for immune T cell areas.
Cell
143
:
416
429
.
49.
Schlitzer
A.
,
N.
McGovern
,
F.
Ginhoux
.
2015
.
Dendritic cells and monocyte-derived cells: two complementary and integrated functional systems.
Semin. Cell Dev. Biol.
41
:
9
22
.
50.
Chen
J. C.
,
M. L.
Chang
,
M. O.
Muench
.
2003
.
A kinetic study of the murine mixed lymphocyte reaction by 5,6-carboxyfluorescein diacetate succinimidyl ester labeling.
J. Immunol. Methods
279
:
123
133
.
51.
Serbina
N. V.
,
T. P.
Salazar-Mather
,
C. A.
Biron
,
W. A.
Kuziel
,
E. G.
Pamer
.
2003
.
TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection.
Immunity
19
:
59
70
.
52.
Helft
J.
,
J.
Böttcher
,
P.
Chakravarty
,
S.
Zelenay
,
J.
Huotari
,
B. U.
Schraml
,
D.
Goubau
,
C.
Reis e Sousa
.
2015
.
GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells.
Immunity
42
:
1197
1211
.
53.
Yona
S.
,
K. W.
Kim
,
Y.
Wolf
,
A.
Mildner
,
D.
Varol
,
M.
Breker
,
D.
Strauss-Ayali
,
S.
Viukov
,
M.
Guilliams
,
A.
Misharin
, et al
2013
.
Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis.
Immunity
38
:
79
91
.
54.
Varol
C.
,
L.
Landsman
,
D. K.
Fogg
,
L.
Greenshtein
,
B.
Gildor
,
R.
Margalit
,
V.
Kalchenko
,
F.
Geissmann
,
S.
Jung
.
2007
.
Monocytes give rise to mucosal, but not splenic, conventional dendritic cells.
J. Exp. Med.
204
:
171
180
.
55.
Ginhoux
F.
,
S.
Jung
.
2014
.
Monocytes and macrophages: developmental pathways and tissue homeostasis.
Nat. Rev. Immunol.
14
:
392
404
.
56.
Briseño
C. G.
,
M.
Haldar
,
N. M.
Kretzer
,
X.
Wu
,
D. J.
Theisen
,
W.
Kc
,
V.
Durai
,
G. E.
Grajales-Reyes
,
A.
Iwata
,
P.
Bagadia
, et al
2016
.
Distinct transcriptional programs control cross-priming in classical and monocyte-derived dendritic cells.
Cell Rep.
15
:
2462
2474
.
57.
Menezes
S.
,
D.
Melandri
,
G.
Anselmi
,
T.
Perchet
,
J.
Loschko
,
J.
Dubrot
,
R.
Patel
,
E. L.
Gautier
,
S.
Hugues
,
M. P.
Longhi
, et al
2016
.
The heterogeneity of Ly6Chi monocytes controls their differentiation into iNOS+ macrophages or monocyte-derived dendritic cells.
Immunity
45
:
1205
1218
.
58.
Schlitzer
A.
,
V.
Sivakamasundari
,
J.
Chen
,
H. R.
Sumatoh
,
J.
Schreuder
,
J.
Lum
,
B.
Malleret
,
S.
Zhang
,
A.
Larbi
,
F.
Zolezzi
, et al
2015
.
Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow.
Nat. Immunol.
16
:
718
728
.
59.
Carlin
L. M.
,
E. G.
Stamatiades
,
C.
Auffray
,
R. N.
Hanna
,
L.
Glover
,
G.
Vizcay-Barrena
,
C. C.
Hedrick
,
H. T.
Cook
,
S.
Diebold
,
F.
Geissmann
.
2013
.
Nr4a1-dependent Ly6Clow monocytes monitor endothelial cells and orchestrate their disposal.
Cell
153
:
362
375
.
60.
Tamura
A.
,
H.
Hirai
,
A.
Yokota
,
A.
Sato
,
T.
Shoji
,
T.
Kashiwagi
,
M.
Iwasa
,
A.
Fujishiro
,
Y.
Miura
,
T.
Maekawa
.
2015
.
Accelerated apoptosis of peripheral blood monocytes in Cebpb-deficient mice.
Biochem. Biophys. Res. Commun.
464
:
654
658
.
61.
Tamura
A.
,
H.
Hirai
,
A.
Yokota
,
N.
Kamio
,
A.
Sato
,
T.
Shoji
,
T.
Kashiwagi
,
Y.
Torikoshi
,
Y.
Miura
,
D. G.
Tenen
,
T.
Maekawa
.
2017
.
C/EBPβ is required for survival of Ly6C monocytes.
Blood
130
:
1809
1818
.
62.
Kurotaki
D.
,
N.
Osato
,
A.
Nishiyama
,
M.
Yamamoto
,
T.
Ban
,
H.
Sato
,
J.
Nakabayashi
,
M.
Umehara
,
N.
Miyake
,
N.
Matsumoto
, et al
2013
.
Essential role of the IRF8-KLF4 transcription factor cascade in murine monocyte differentiation.
Blood
121
:
1839
1849
.
63.
Sichien
D.
,
C. L.
Scott
,
L.
Martens
,
M.
Vanderkerken
,
S.
Van Gassen
,
M.
Plantinga
,
T.
Joeris
,
S.
De Prijck
,
L.
Vanhoutte
,
M.
Vanheerswynghels
, et al
2016
.
IRF8 transcription factor controls survival and function of terminally differentiated conventional and plasmacytoid dendritic cells, respectively.
Immunity
45
:
626
640
.
64.
Dutertre
C. A.
,
E.
Becht
,
S. E.
Irac
,
A.
Khalilnezhad
,
V.
Narang
,
S.
Khalilnezhad
,
P. Y.
Ng
,
L. L.
van den Hoogen
,
J. Y.
Leong
,
B.
Lee
, et al
2019
.
Single-cell analysis of human mononuclear phagocytes reveals subset-defining markers and identifies circulating inflammatory dendritic cells.
Immunity
51
:
573
589.e8
.
65.
Tang-Huau
T. L.
,
E.
Segura
.
2019
.
Human in vivo-differentiated monocyte-derived dendritic cells.
Semin. Cell Dev. Biol.
86
:
44
49
.
66.
Rhodes
J. W.
,
O.
Tong
,
A. N.
Harman
,
S. G.
Turville
.
2019
.
Human dendritic cell subsets, ontogeny, and impact on HIV infection.
Front. Immunol.
10
:
1088
.

The authors have no financial conflicts of interest.

Supplementary data