BCR-ABL tyrosine kinase inhibitors (TKI) have dramatically improved therapy for chronic myelogenous leukemia (CML). However, several problems leading to TKI resistance still impede a complete cure of this disease. IFN regulatory factor-8 (IRF8) is a transcription factor essential for the development and functions of immune cells, including dendritic cells. Irf8−/− mice develop a CML-like disease and IRF8 expression is downregulated in patients with CML, suggesting that IRF8 is involved in the pathogenesis of CML. In this study, by using a murine CML model, we show that BCR-ABL strongly inhibits a generation of dendritic cells from an early stage of their differentiation in vivo, concomitant with suppression of Irf8 expression. Forced expression of IRF8 overrode BCR-ABL (both wild-type and T315I-mutated) to rescue dendritic cell development in vitro, indicating that the suppression of Irf8 causes dendritic cell deficiency. Gene expression profiling revealed that IRF8 restored the expression of a significant portion of BCR-ABL–dysregulated genes and predicted that BCR-ABL has immune-stimulatory potential. Indeed, IRF8-rescued BCR-ABL–expressing dendritic cells were capable of inducing CTLs more efficiently than control dendritic cells. Altogether, our findings suggest that IRF8 is an attractive target in next-generation therapies for CML. Cancer Res; 73(22); 6642–53. ©2013 AACR.

Chronic myelogenous leukemia (CML) is a myeloproliferative disorder that accounts for approximately 15% of newly diagnosed cases of adult leukemia (1, 2). The causal molecule of this disease is BCR-ABL, a 210 kDa oncoprotein encoded by the BCR-ABL fusion gene, which is generated by a t(9;22) chromosomal translocation (3). BCR-ABL possesses deregulated ABL tyrosine kinase activity that stimulates many downstream signals including the MYC, NF-κB, RAS, mitogen-activated protein kinase, phosphoinositide 3-kinase (PI3K), and STAT pathways. These signals lead to stimulation of cell proliferation, inhibition of apoptosis, and alteration of adhesion to stroma cells and extracellular matrix. CML begins with a chronic phase in which clonal BCR-ABL+ hematopoietic stem cells give rise to increased numbers of their progenies, particularly myeloid precursors and mature cells (mainly neutrophils) in the bone marrow, blood, and extramedullary tissues. After 3 to 5 years, if not treated, the disease progresses into a fatal blast crisis, characterized by accumulation of immature precursor cells with differentiation arrest.

During the last decade, therapy with BCR-ABL–selective tyrosine kinase inhibitors (TKI), such as imatinib, nilotinib, and dasatinib, has greatly improved the prognosis of patients with CML (4). However, several problems still impede a complete cure of this disease. First, a minor but significant subset of patients fails to respond to or becomes resistant to TKIs mainly due to acquisition of point mutations in the ABL gene, such as T315I. Second, because leukemic stem cells are not efficiently eliminated by TKIs, discontinuing TKI therapy often results in CML recurrence (4). Third, while CML is highly sensitive to tumor immunity, TKIs may suppress functions of T cells and dendritic cells (5–7). Therefore, next-generation therapies for CML are required.

Dendritic cells are the most potent professional antigen-presenting cells and are essential for the initiation and regulation of innate and adaptive immunity against pathogens and tumors (8). Dendritic cells are a heterogeneous population that can be divided into classical dendritic cells (cDC; further divided into CD8α+ dendritic cells, CD4+ dendritic cells, and double-negative dendritic cells in mice) and plasmacytoid dendritic cells (pDC) both in humans and mice (9). CD8α+ dendritic cells (BDCA3+ in humans) are a unique subset capable of priming CTLs by cross-presentation of dead cell materials and are thus critical for tumor immunity. pDCs produce a large amount of type I IFN, that is, IFN-α/β, upon Toll-like receptor (TLR)-7/8/9 signaling. Type I IFN elicits antiviral and antitumor responses. Of note, IFN-α was once used as a major therapeutic agent for CML (10, 11).

CML has long been suggested to be highly sensitive to T cell–mediated tumor immunity (12). CML responds to immune-mediated therapies, such as IFN-α, allogeneic stem cell transplantation (alloSCT), and donor lymphocyte infusion (13). IFN-α induces a specific T-cell response in CML (10, 11). In alloSCT, T-cell depletion from donor bone marrow cells results in a significant increase in relapse, especially in patients with CML. Yet, the fact that the disease develops in patients with CML suggests that tumor immunity is insufficient in patients with CML. Because T cells per se in patients with CML rarely express BCR-ABL, it is reasonable to suspect dendritic cells as a culprit. Indeed, several studies have shown that patients with CML have reduced numbers of cDCs and pDCs in the chronic phase, as compared with healthy individuals (14, 15). Other studies have reported that dendritic cells generated in vitro from monocytes or CD34+ cells of patients with CML are functionally defective in multiple aspects, such as actin organization, antigen processing, migration, maturation, and cytokine production (16, 17). In a murine CML model, defective homing and impaired induction of CTLs by BCR-ABL–expressing dendritic cells have been reported (18). However, the mechanism of impaired dendritic cell development in CML remains unknown.

IFN regulatory factor-8 (IRF8) is a hematopoietic transcription factor that regulates the development of multiple immune cell types (19). IRF8 is required for differentiation of mouse cDCs (particularly CD8α+ dendritic cells; refs. 20, 21), pDCs (22), and monocytes (particularly Ly6C+ monocytes; ref. 23), while inhibiting myeloid cell proliferation and neutrophil differentiation (24). IRF8 is also indispensable for the differentiation of CTLs (25). Thus, Irf8−/− mice develop immunodeficiency and a CML-like syndrome (26). Importantly, mutations in the human IRF8 gene are associated with dendritic cell immunodeficiency (27). Furthermore, IRF8 expression is dramatically decreased in patients with CML (28, 29). IRF8 also overcomes the mitogenic activity of BCR-ABL in differentiating myeloid progenitors in vitro (30). Coexpression of IRF8 in bone marrow progenitors can ameliorate BCR-ABL–induced myeloproliferative disorder in mice (31). Coexpression of IRF8 in a BCR-ABL–transformed mouse pro-B cell line induces a chemokine-dependent antileukemia immunity in mice (31–33). These findings imply that there is an antagonistic relationship between IRF8 and CML pathogenesis. However, this idea has not been tested in terms of dendritic cell biology yet. In this study, we have investigated how BCR-ABL and IRF8 are involved in the development and function of dendritic cells in CML using a murine model.

Mice

C57BL/6-Ly5.1 or -Ly5.2 congenic mice and OT-I or OT-II T cell receptor transgenic mice (The Jackson Laboratory) were used at 8 to 12 weeks of age. All animal experiments were carried out in accordance with the Guidelines for Proper Conduct of Animal Experiments (Science Council of Japan), and all protocols were approved by the Institutional Review Boards of Yokohama City University (Yokohama, Japan; Protocol #F11-85).

Flow cytometry

Flow cytometry was performed using FACSCanto II (BD Biosciences), and data were analyzed using the FlowJo software (TreeStar). For antibodies and their clone names, see Supplementary Materials and Methods.

Separation of hematopoietic progenitors

Murine lineage marker–negative (Lin) cells were purified from bone marrow cells by the magnetic-activated cell sorting (MACS) system using the Lineage Cell Depletion Kit (Miltenyi Biotec) and anti-interleukin (IL)-7 receptor (IL-7R/CD127) antibody. For isolation of LinSca-1+c-Kit+ (LSK) cells, MACS-purified Lin cells were stained with antibodies against Sca-1 and c-Kit, and were then purified by the fluorescence-activated cell sorting (FACS) system using a FACSAria II (BD Biosciences).

Retroviral transduction and CML model mice

The following murine stem cell virus (MSCV) retroviral vectors were used: MIG [MSCV-internal ribosome entry site (IRES)- GFP], MIG-p210BCR-ABL (MSCV-p210BCR-ABL-IRES-GFP), MICD8 [MSCV-IRES-human truncated CD8 (hCD8t)], and MICD8-IRF8 (MSCV-IRF8-IRES-hCD8t). Lin cells were precultured for 24 hours with stem cell factor (SCF) and thrombopoietin (TPO) for transplantation, or SCF, IL-6, and IL-3 for in vitro experiments, and were then transduced with MSCVs by spinoculation for 2 consecutive days. All cytokines were purchased from PeproTech. For generating CML model mice, C57BL/6-Ly5.1 LSK cells transduced with MIG or MIG-p210BCR-ABL were injected intravenously into lethally irradiated syngenic C57BL/6-Ly5.2 recipients.

qRT-PCR, ELISA, and immunoblot analysis

Quantitative PCR (qPCR) with quantitative reverse transcription PCR (qRT-PCR) was performed using RNAiso Plus (Takara Bio), DNase I (Invitrogen), PrimeScript (Takara Bio), Thunderbird SYBR qPCR Mix (Toyobo), and an ABI PRISM 7900 sequence detection system (Applied Biosystems) according to the manufacturers' protocols. Primer sequences are described in Supplementary Materials and Methods. Data were analyzed using the ΔΔCT method and normalized against Gapdh levels. ELISA for IFN-α and IL-12p40 was performed using commercially available kits (PBL and BioLegend, respectively). For immunoblot analysis, anti-IRF8 (C-19; Santa Cruz Biotechnology), c-ABL (Cell Signaling Technology), phosphotyrosine (4G10; Millipore), and β-actin (AC-74; Sigma-Aldrich) antibodies were used.

Dendritic cell culture

Bone marrow Lin cells transduced with MSCVs were washed 24 hours after the last spinoculation and cultured with human fms-like tyrosine kinase-3 (Flt3)-ligand (Flt3L) for 7 days to induce dendritic cell differentiation. TKIs and a STAT5 inhibitor [N′-((4-oxo-4H-chromen-3-yl)methylene)nicotinohydrazide] were purchased from Santa Cruz Biotechnology. Trichostatin A (TSA) and 5-azacytidine (5-Aza) were purchased from Focus Biomolecules and Sigma-Aldrich, respectively.

Microarray

RNAs from two independent experiments were analyzed using a Whole Mouse Genome 8 × 60 K Microarray (Agilent) according to the manufacturer's protocol. Microarray data are available at the GEO/NCBI database (GSE44920). For details, see Supplementary Materials and Methods.

T-cell response assays

Antigen presentation capability of dendritic cells via MHC class II (MHC-II) was evaluated using OT-II T cells essentially as previously described (21). In vitro CTL assays were performed using OT-I T cells essentially by the method previously established (34).

BCR-ABL inhibits dendritic cell development and Irf8 expression

To investigate how dendritic cell development is affected in CML, we first used a murine CML model in which hematopoietic progenitors (LSK cells) were transduced with bicistronic MIG-p210BCR-ABL retrovirus (carrying cDNAs encoding BCR-ABL and GFP) in the presence of SCF and TPO, and then transplanted these cells into lethally irradiated mice. These mice (BCR-ABL mice), but not mice transplanted with empty MIG-transduced cells (MIG mice), exhibited splenomegaly and died by 4 to 8 weeks after the transplantation (Fig. 1A and Supplementary Fig. S1A). Wright–Giemsa staining and flow-cytometric analyses of splenic cells revealed that BCR-ABL mice had significant increases in the percentage and number of GFP+ (therefore BCR-ABL+) neutrophils, a hallmark of CML, as compared with MIG mice (Fig. 1B and C).

Figure 1.

Inhibition of dendritic cell development and Irf8 expression by BCR-ABL in vivo. LSK cells transduced with empty MIG or MIG-p210BCR-ABL were transplanted into lethally irradiated mice. Spleens and bone marrow cells in recipient mice were analyzed 4 to 5 weeks after transplantation (n = 5 each except for Fig. 1G, in which n = 3 each). A, spleens (left) and splenic cell numbers (right). B, Wright–Giemsa stains of FACS-sorted GFP+ spleen cells. C–E, flow-cytometric analyses of splenic neutrophils (C), splenic dendritic cells (D), and bone marrow MDPs and CDPs (E). F, Irf8 mRNA expression levels in FACS-sorted GFP+ bone marrow (BM) Lin cells and splenic cells determined by qRT-PCR in triplicate. G, IRF8 protein expression levels in splenic GFP+CD11chigh cDCs and GFP+PDCA-1+CD11clow pDCs determined by flow cytometry. The mean fluorescent intensity (MFI) was calculated by subtracting the autofluorescence intensity. All values in bar graphs are the mean ± SD. *, P < 0.05 and **, P < 0.01 (Student t test).

Figure 1.

Inhibition of dendritic cell development and Irf8 expression by BCR-ABL in vivo. LSK cells transduced with empty MIG or MIG-p210BCR-ABL were transplanted into lethally irradiated mice. Spleens and bone marrow cells in recipient mice were analyzed 4 to 5 weeks after transplantation (n = 5 each except for Fig. 1G, in which n = 3 each). A, spleens (left) and splenic cell numbers (right). B, Wright–Giemsa stains of FACS-sorted GFP+ spleen cells. C–E, flow-cytometric analyses of splenic neutrophils (C), splenic dendritic cells (D), and bone marrow MDPs and CDPs (E). F, Irf8 mRNA expression levels in FACS-sorted GFP+ bone marrow (BM) Lin cells and splenic cells determined by qRT-PCR in triplicate. G, IRF8 protein expression levels in splenic GFP+CD11chigh cDCs and GFP+PDCA-1+CD11clow pDCs determined by flow cytometry. The mean fluorescent intensity (MFI) was calculated by subtracting the autofluorescence intensity. All values in bar graphs are the mean ± SD. *, P < 0.05 and **, P < 0.01 (Student t test).

Close modal

We found that both cDCs (CD11chigh cells) and pDCs (CD11clowPDCA-1+ cells) were dramatically diminished in BCR-ABL mice as compared with MIG mice; the percentage and number of GFP+ cDCs showed 16.7- and 6.5-fold reductions, respectively, and those of pDCs showed 18.1- and 8.1-fold reductions, respectively (Fig. 1D). More detailed analysis demonstrated that all cDC subsets, that is, CD8α+ dendritic cells, CD4+ dendritic cells, and CD8αCD4 (double-negative) dendritic cells, were significantly diminished (Supplementary Fig. S1B). In addition, we noted that the remaining GFP+ cDCs in BCR-ABL mice expressed lower levels of MHC-II compared with those in MIG mice (Supplementary Fig. S1C). In fact, these spared dendritic cells exhibited a reduced capability of presenting ovalbumin (OVA) antigen, as demonstrated by the proliferation of and IFN-γ production by OT-II T cells (Supplementary Fig. S1D). These results indicate that BCR-ABL strongly and globally inhibits the generation of dendritic cells in vivo.

BCR-ABL inhibits an early stage of dendritic cell differentiation

We next analyzed progenitor populations in the bone marrow. Dendritic cells originate from bone marrow hematopoietic stem cells through monocyte-dendritic cell progenitors (MDP; LinSca-1CD127c-Kitint/+CD115+) and then common dendritic cell progenitors (CDP; LinSca-1CD127Flt3+c-KitintCD115+; refs. 9, 35). MDPs are a CD115+ population in myeloid progenitors (LinCD127Sca-1c-Kit+), and overlap with granulocyte-monocyte progenitors (GMP; LinCD127Sca-1c-Kit+CD16/32+CD34+) and to a lesser extent common myeloid progenitors (CMP; LinCD127Sca-1c-Kit+CD16/32lowCD34+). Within myeloid progenitors, CMPs progress toward either GMPs or megakaryocyte-erythroid progenitors (MEP; LinCD127Sca-1c-Kit+CD16/32CD34).

Interestingly, the bone marrow GFP+ population in BCR-ABL mice lacked MDPs and CDPs (Fig. 1E). The percentages of total GFP+ myeloid progenitors were comparable between BCR-ABL mice and MIG mice (Supplementary Fig. S1E). GFP+ myeloid progenitors in BCR-ABL mice had relatively low percentages of CMPs and GMPs, which could be partially due to the lack of MDPs, while they contained a modestly increased percentage of MEPs and harbored an aberrant CD34 CD16/32+ population. Despite the sharp decrease in MDP counts, we noted that monocyte counts were not significantly affected in BCR-ABL mice (data not shown), consistent with the fact that monocyte counts are not reduced in patients with CML. The developmental route of these BCR-ABL+ monocytes is unknown, but one possibility is that they are derived from upstream myeloid progenitors without transiting MDPs. Overall, these data suggest that BCR-ABL affects the early stage of dendritic cell differentiation, particularly the generation of MDPs.

We analyzed IRF8 expression in BCR-ABL and MIG mice. qRT-PCR analysis revealed that BCR-ABL suppressed Irf8 mRNA expression both in Lin bone marrow progenitors and splenic cells (Fig. 1F). Immunostaining for IRF8 in splenic dendritic cells demonstrated that the remaining few BCR-ABL+ dendritic cells expressed lower levels of IRF8 than control dendritic cells (Fig. 1G and Supplementary Fig. S1F).

IRF8 overrides BCR-ABL to rescue dendritic cell differentiation

To further investigate the mechanisms involved in this process, we examined whether BCR-ABL inhibits dendritic cell differentiation in vitro. We transduced MIG-p210BCR-ABL or empty MIG retrovirus into bone marrow Lin progenitors in the presence of SCF, IL-6, and IL-3, and the transduced cells were then cultured with Flt3L for 7 days to induce differentiation toward cDCs (CD11chighMHC-II+) and pDCs (CD11c+B220+CD11blow; ref. 21). In MIG-transduced control cultures, cDCs and pDCs were efficiently generated (Fig. 2A). In BCR-ABL–transduced cultures, however, the development of both cDCs and pDCs was significantly inhibited in terms of percentages and absolute cell counts, even though BCR-ABL–transduced cells showed a greater increase in cell number during the Flt3L culture than MIG-transduced cells. Wright–Giemsa staining revealed that while many of the MIG-transduced cells showed typical dendritic cell morphology, BCR-ABL–transduced cells exhibited neutrophil-like, macrophage-like, or immature morphologies (Fig. 2B). Furthermore, the induction of Irf8 mRNA expression during the Flt3L culture was almost completely abrogated by BCR-ABL (Fig. 2C). When a kinase-dead K1172R mutant of BCR-ABL was transduced into cells, neither dendritic cell development nor IRF8 expression was affected, suggesting that the kinase activity of BCR-ABL is required for the observed inhibitory effects (Fig. 2D). The expression levels of wild-type (WT) and K1172R BCR-ABL were comparable, and K1172R lacked autophosphorylation at tyrosine residues.

Figure 2.

Inhibition of dendritic cell development and Irf8 expression by BCR-ABL in vitro. Bone marrow Lin cells were transduced with MIG or MIG-p210BCR-ABL and then cultured for 7 days in the presence of Flt3L to induce dendritic cell differentiation. A, flow-cytometric analysis of dendritic cells. Numbers in middle left indicate percentages of pDCs relative to GFP+CD11c+ cells (without parentheses) and those relative to total GFP+ cells (in parentheses). For evaluating cell numbers, FACS-purified GFP+–transduced cells were seeded at 2 × 105 cells/mL at the beginning of Flt3L culture. B, Wright–Giemsa stains of FACS-sorted transduced GFP+ cells. C, Irf8 mRNA expression levels in FACS-sorted GFP+ cells determined by qRT-PCR. D, flow cytometry, qRT-PCR, and immunoblot analysis in cells transduced with K1172R BCR-ABL. Tyr-P BCR-ABL indicates tyrosine-phosphorylated BCR-ABL. β-Actin expression is shown as a loading control. All values in bar graphs are the mean ± SD from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Figure 2.

Inhibition of dendritic cell development and Irf8 expression by BCR-ABL in vitro. Bone marrow Lin cells were transduced with MIG or MIG-p210BCR-ABL and then cultured for 7 days in the presence of Flt3L to induce dendritic cell differentiation. A, flow-cytometric analysis of dendritic cells. Numbers in middle left indicate percentages of pDCs relative to GFP+CD11c+ cells (without parentheses) and those relative to total GFP+ cells (in parentheses). For evaluating cell numbers, FACS-purified GFP+–transduced cells were seeded at 2 × 105 cells/mL at the beginning of Flt3L culture. B, Wright–Giemsa stains of FACS-sorted transduced GFP+ cells. C, Irf8 mRNA expression levels in FACS-sorted GFP+ cells determined by qRT-PCR. D, flow cytometry, qRT-PCR, and immunoblot analysis in cells transduced with K1172R BCR-ABL. Tyr-P BCR-ABL indicates tyrosine-phosphorylated BCR-ABL. β-Actin expression is shown as a loading control. All values in bar graphs are the mean ± SD from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Close modal

We next asked whether restoration of IRF8 expression could improve dendritic cell differentiation impaired by BCR-ABL. To this end, we cotransduced Lin cells with MICD8-IRF8 (that expresses IRF8 and hCD8t) together with MIG-p210BCR-ABL, and treated them with Flt3L. hCD8t, used as a transduction marker, does not transmit any signals because it contains no cytoplasmic domain. Flow-cytometric analysis of doubly transduced (i.e., GFP+hCD8t+) cells revealed that the forced expression of IRF8 efficiently rescued differentiation of both cDCs and pDCs (Fig. 3A). The expression levels of Irf8 mRNA and IRF8 protein in IRF8-transduced BCR-ABL+ cells were comparable with those of endogenous IRF8 in control cells (Fig. 3B and C, compare the first and fourth lanes). Importantly, IRF8 also efficiently overrode the TKI-resistant T315I-mutant BCR-ABL to rescue dendritic cell differentiation (Fig. 3D). WT and T315I BCR-ABL were expressed at comparable levels (Fig. 3B and C). These results indicate that the suppression of Irf8 is a cause, rather than a result, of dendritic cell deficiency by BCR-ABL in CML.

Figure 3.

Dendritic cell differentiation impaired by BCR-ABL can be restored by forced expression of IRF8 in vitro. Bone marrow Lin cells were cotransduced with empty MIG, MIG-p210BCR-ABL or MIG-T315IBCR-ABL, and empty MICD8 or MICD8-IRF8 and then cultured for 7 days in the presence of Flt3L. A, cotransduction with IRF8 and BCR-ABL. Numbers in bottom left indicate percentages of pDCs relative to GFP+hCD8+CD11c+ cells (without parentheses) and those relative to total GFP+hCD8+ cells (in parentheses). B, qRT-PCR for Irf8 mRNA expression levels in FACS-sorted GFP+hCD8t+ cells. C, immunoblot analysis of IRF8 and BCR-ABL in GFP+hCD8t+ cells. D, cotransduction with IRF8 and T315I BCR-ABL. All values in bar graphs are from three independent experiments.

Figure 3.

Dendritic cell differentiation impaired by BCR-ABL can be restored by forced expression of IRF8 in vitro. Bone marrow Lin cells were cotransduced with empty MIG, MIG-p210BCR-ABL or MIG-T315IBCR-ABL, and empty MICD8 or MICD8-IRF8 and then cultured for 7 days in the presence of Flt3L. A, cotransduction with IRF8 and BCR-ABL. Numbers in bottom left indicate percentages of pDCs relative to GFP+hCD8+CD11c+ cells (without parentheses) and those relative to total GFP+hCD8+ cells (in parentheses). B, qRT-PCR for Irf8 mRNA expression levels in FACS-sorted GFP+hCD8t+ cells. C, immunoblot analysis of IRF8 and BCR-ABL in GFP+hCD8t+ cells. D, cotransduction with IRF8 and T315I BCR-ABL. All values in bar graphs are from three independent experiments.

Close modal

IRF8 normalizes the expression of a significant portion of BCR-ABL–dysregulated genes

We next performed transcriptome analysis by microarray in bone marrow Lin cells transduced with empty vectors, MIG-p210BCR-ABL, or MIG-p210BCR-ABL + MICD8-IRF8 followed by 5-day Flt3L culture (see Supplementary Fig. S2A and S2B for quality control data). We first calculated the number of genes whose expression was significantly affected by BCR-ABL [fold change (FC) > 3; false discovery rate (FDR) < 0.05] and those normalized by IRF8 (Fig. 4A and Supplementary Tables S1 and S2). The criteria of normalization were as follows: IRF8 significantly reverses the expression in BCR-ABL+ cells (FC > 2; FDR < 0.05), and expression of the gene becomes comparable between control dendritic cells and IRF8-rescued BCR-ABL+ dendritic cells (i.e., FC is no longer > 3 and FDR < 0.05). BCR-ABL decreased the expression of 1,433 genes and increased the expression of 1,915 genes (FC > 3). Among these genes, despite the stringent criteria, IRF8 normalized the expression of 688 genes (48%) and 564 genes (29%), respectively. These results indicate that BCR-ABL impairs the expression of many genes via suppressing IRF8 expression.

Figure 4.

Gene expression profiling of cells transduced with BCR-ABL and/or IRF8. Bone marrow Lin cells were cotransduced with empty MIG or MIG-p210BCR-ABL and empty MICD8 or MICD8-IRF8, and FACS-sorted GFP+hCD8t+ cells were then cultured for 5 (for microarray) or 7 days (for qRT-PCR) in the presence of Flt3L. Microarray and qRT-PCR were performed in biologic duplicates. A, Venn diagrams for the numbers of genes that displayed more than a 3-fold change in expression by BCR-ABL and those normalized by IRF8. B, GSEA for dendritic cell (DC) signature genes. NES, normalized enrichment score. C, qRT-PCR. Values are from two independent experiments, in which each PCR was performed in triplicate. D, GSEA for dendritic cell maturation-related signature genes.

Figure 4.

Gene expression profiling of cells transduced with BCR-ABL and/or IRF8. Bone marrow Lin cells were cotransduced with empty MIG or MIG-p210BCR-ABL and empty MICD8 or MICD8-IRF8, and FACS-sorted GFP+hCD8t+ cells were then cultured for 5 (for microarray) or 7 days (for qRT-PCR) in the presence of Flt3L. Microarray and qRT-PCR were performed in biologic duplicates. A, Venn diagrams for the numbers of genes that displayed more than a 3-fold change in expression by BCR-ABL and those normalized by IRF8. B, GSEA for dendritic cell (DC) signature genes. NES, normalized enrichment score. C, qRT-PCR. Values are from two independent experiments, in which each PCR was performed in triplicate. D, GSEA for dendritic cell maturation-related signature genes.

Close modal

Gene set enrichment analysis (GSEA; ref. 36) showed that expression of dendritic cell signature genes (37) was indeed significantly suppressed by BCR-ABL, and was efficiently reinduced by coexpression of IRF8 (Fig. 4B). qRT-PCR analysis of several of these genes (Ciita, H2-Eb2, and H2-Aa) in the same set of transduced cells as well as cells transduced with IRF8 alone on day 7 and in freshly isolated Lin cells confirmed the findings (Fig. 4C, top). Expression of other genes critical for the function of CD8α+ dendritic cells (e.g., cross-presentation), such as Clec9a, Xcr1, and Tlr3, was also suppressed by BCR-ABL and restored by IRF8 (Fig. 4C, bottom). Conversely, GSEA showed that expression of granulocyte signature genes (see Supplementary Materials and Methods) was significantly induced by BCR-ABL, but suppressed by IRF8 (Supplementary Fig. S2C).

We also paid attention to the genes dysregulated by BCR-ABL but not normalized by IRF8. We used Ingenuity Pathway Analysis (IPA) software to predict transcription factors that may act upstream of these genes. The results showed that the genes induced by BCR-ABL, irrespective of IRF8 supplementation, were significantly enriched with those inducible by NF-κB, a transcription factor activated by BCR-ABL and involved in many activation signals in dendritic cells (Supplementary Fig. S2D). Furthermore, GSEA showed that IRF8-rescued BCR-ABL+ dendritic cells expressed higher levels of dendritic cell maturation-related genes (38) than control dendritic cells (Fig. 4D, right). This was even more apparent in IRF8-untransduced BCR-ABL+ cells compared with control dendritic cells and IRF8-rescued BCR-ABL+ dendritic cells (Fig. 4D, left and middle), suggesting that dendritic cell maturation-related genes were upregulated by BCR-ABL rather than exogenous IRF8. These results lead us to speculate that IRF8-rescued, BCR-ABL–expressing dendritic cells may be in a “preactivated” state. Other predicted transcription factors included GATA1 and STAT5 (see Discussion). Indeed, the expression of Gata1 itself was 15-fold higher in BCR-ABL–transduced cells than in control dendritic cells, regardless of IRF8 supplementation.

IRF8-rescued BCR-ABL+ dendritic cells have higher cytokine production and CTL induction capacities

We next evaluated the functionality of the rescued dendritic cells. To examine their ability to produce IFN-α and IL-12p40, transduced Lin cells followed by 7-day Flt3L culture were stimulated with TLR9 ligands CpG-A (for pDCs) or CpG-B (for cDCs), or a TLR4 ligand lipopolysaccharide (LPS; Fig. 5A). TLRs recognize not only the pathogen-associated molecular patterns (PAMP) but also damage-associated molecular patterns (DAMP) that may be involved in antitumor immunity (39). BCR-ABL almost completely abolished production of these cytokines, but their expression was restored by coexpression of IRF8. Interestingly, IRF8-rescued BCR-ABL+ dendritic cells produced even higher levels of these cytokines than control dendritic cells, especially for IL-12p40, whose expression is dependent on NF-κB and IRF8 (19, 40).

Figure 5.

Functionality of IRF8-rescued BCR-ABL+ dendritic cells. Double transduction of Lin cells followed by 7-day Flt3L culture was performed as in Fig. 3. A, cytokine production. FACS-sorted GFP+hCD8t+ cells were stimulated with the indicated TLR ligands for 24 hours and their supernatants were analyzed by ELISA. B, expression levels of antigen presentation–related molecules determined by flow cytometry. C, induction of CTLs. Antigen-specific cytotoxicity induced by FACS-sorted GFP+hCD8t+ cells was analyzed. All values are from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Figure 5.

Functionality of IRF8-rescued BCR-ABL+ dendritic cells. Double transduction of Lin cells followed by 7-day Flt3L culture was performed as in Fig. 3. A, cytokine production. FACS-sorted GFP+hCD8t+ cells were stimulated with the indicated TLR ligands for 24 hours and their supernatants were analyzed by ELISA. B, expression levels of antigen presentation–related molecules determined by flow cytometry. C, induction of CTLs. Antigen-specific cytotoxicity induced by FACS-sorted GFP+hCD8t+ cells was analyzed. All values are from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Close modal

We also examined the expression of MHC-related molecules by flow cytometry. BCR-ABL strongly inhibited MHC-II expression, but increased MHC-I expression 4.1-fold (Fig. 5B and Supplementary Fig. S3). When IRF8 expression was supplemented, MHC-II expression was recovered, and MHC-I expression remained high. We also found that expression of the costimulatory molecules CD80 and CD86 was significantly induced by BCR-ABL and remained high in IRF8-cotransduced cells. We noted that the mRNA levels of MHC-I, CD80, and CD86 did not correlate with protein expression levels on the cell surface, suggesting the occurrence of posttranscriptional control. The expression level of another costimulatory molecule CD40 was low and not affected by BCR-ABL. Thus, IRF8-rescued BCR-ABL+ dendritic cells expressed higher levels of MHC-I and some costimulatory molecules than regular dendritic cells.

CD8α+ dendritic cells have been shown to be critical for eliminating tumor cells by priming CTLs via cross-presentation in mice (41). In addition, CD8α+ dendritic cells require type I IFN to induce CTLs that reject tumors (42, 43). These previous findings, together with the above data on cytokine production and MHC-related molecules, prompted us to examine the ability to cross-present an antigen (OVA) to induce a CTL response (Fig. 5C). Transduced Lin cells followed by 7-day Flt3L culture were pulsed with OVA protein, and then mixed with T cells from OT-I mice. The primed T cells were then mixed with target splenocytes loaded with OVA peptide, and OVA-specific CTL activity was calculated. Although control dendritic cells efficiently induced CTLs, BCR-ABL–transduced cells failed to do so. Strikingly, IRF8-rescued BCR-ABL+ dendritic cells regained the ability to induce CTLs, and the observed response was even stronger than that of control dendritic cells.

Transduction of IRF8 without BCR-ABL did not augment the above functional parameters. Thus, these results suggest that BCR-ABL harbors an immune-stimulatory potential that is manifested when dendritic cell differentiation is restored by supplementation with IRF8.

Imatinib partially restores Irf8 expression, but cancels the enhancement of functionality in IRF8-rescued BCR-ABL+ dendritic cells

To investigate whether clinically available TKIs can restore Irf8 expression as well as dendritic cell differentiation and function, imatinib, nilotinib, and dasatinib were added at concentrations two times higher than those required for 90% inhibition (IC90) of BCR-ABL kinase activity during the Flt3L treatment of transduced Lin cells. These TKIs partially restored the expression of Irf8 expression in BCR-ABL–transduced cells (Fig. 6A, top). Efficient inhibition of BCR-ABL kinase activity by imatinib was confirmed by immunoblot analysis for autophosphorylation of BCR-ABL (Fig. 6A, bottom). Presumably, due to the modest but significant increase in Irf8 expression, imatinib restored cDC differentiation (Fig. 6B). However, the recovery of pDC differentiation and MHC-II expression was not complete (Fig. 6B and C). In addition, the recovery of cytokine production and CTL induction was very modest (Fig. 6D). Interestingly, imatinib canceled the enhancement of dendritic cell functionality by BCR-ABL in IRF8-rescued BCR-ABL+ dendritic cells; MHC-I, CD80, and CD86 expression, cytokine production, and CTL induction all returned to unenhanced levels. Thus, BCR-ABL augments dendritic cell functionality via its tyrosine kinase activity. We also noted that in control dendritic cells, TKIs modestly inhibited Irf8 expression, and that imatinib slightly suppressed the ability of control dendritic cells to of induce CTLs (Fig. 6E). Overall, these data suggest that dendritic cells rescued by IRF8 are more functional than those rescued by TKIs, at least under the condition used in this study.

Figure 6.

The effects of TKIs on dendritic cell differentiation, function, and Irf8 expression. Lin cells were singly transduced as in Fig. 2 or doubly transduced as in Fig. 3 and cultured in the presence of Flt3L for 7 days. TKIs (imatinib at 2 μmol/L, nilotinib at 50 nmol/L, or dasatinib at 5 nmol/L) were added during the Flt3L culture. Their differentiation status and function were analyzed as in Figs. 2 and 5. In A, D, and E, transduced cells were FACS-sorted before the Flt3L culture. A, qRT-PCR (top) and immunoblot analysis (bottom). B, representative data of flow-cytometric analysis. C, expression levels of antigen presentation–related molecules. D, cytokine production. E, induction of CTLs. Imatinib was added to the Flt3L culture of dendritic cells but not to the coculture with T cells. All values in bar graphs are from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Figure 6.

The effects of TKIs on dendritic cell differentiation, function, and Irf8 expression. Lin cells were singly transduced as in Fig. 2 or doubly transduced as in Fig. 3 and cultured in the presence of Flt3L for 7 days. TKIs (imatinib at 2 μmol/L, nilotinib at 50 nmol/L, or dasatinib at 5 nmol/L) were added during the Flt3L culture. Their differentiation status and function were analyzed as in Figs. 2 and 5. In A, D, and E, transduced cells were FACS-sorted before the Flt3L culture. A, qRT-PCR (top) and immunoblot analysis (bottom). B, representative data of flow-cytometric analysis. C, expression levels of antigen presentation–related molecules. D, cytokine production. E, induction of CTLs. Imatinib was added to the Flt3L culture of dendritic cells but not to the coculture with T cells. All values in bar graphs are from three independent experiments. *, P < 0.05 and **, P < 0.01 (Student t test).

Close modal

To elucidate the pathway through which BCR-ABL inhibits Irf8 expression, we tested whether a STAT5 inhibitor, a histone deacetylase inhibitor (TSA), or a DNA methytransferase inhibitor (5-Aza) improved Irf8 expression and dendritic cell differentiation. TSA and 5-Aza were tested because we speculated that epigenetic mechanisms may be involved, given the discrepancy between the effects of the kinase-dead BCR-ABL–mutant and of imatinib treatment after BCR-ABL expression on Irf8 expression. However, none of these agents restored Irf8 expression or dendritic cell differentiation in BCR-ABL–transduced cells, and instead tended to suppress these events in control cells (Supplementary Fig. S4). Combinations of TSA or 5-Aza with imatinib also failed to rescue Irf8 expression and dendritic cell differentiation.

In this study, the antagonistic relationship between BCR-ABL and IRF8 was investigated in the context of dendritic cell development in CML. Our results revealed that BCR-ABL abolishes dendritic cell development at the level of generating MDPs. This causes severe reductions in dendritic cell counts for all subsets in CML mice, consistent with previous findings in patients with CML (14, 15). BCR-ABL inhibits Irf8 expression, and supplementation with IRF8 restores the expression of a significant portion of BCR-ABL–dysregulated genes, leading to efficient recovery of dendritic cell differentiation. Thus, the suppression of Irf8 expression is the cause of impaired dendritic cell development by BCR-ABL, which we infer accounts, at least in part, for the inability of patients with CML to establish antileukemic immunity.

Gene expression profiling indicated that the expression of a significant numbers of NF-κB target genes was increased by BCR-ABL, and predicted that IRF8-rescued BCR-ABL+ dendritic cells may be in a preactivated state. In fact, we found that the rescued dendritic cells had higher cytokine production and CTL induction capacity than normal dendritic cells, and such enhancement was completely canceled by imatinib. These results indicate that BCR-ABL harbors an immune-stimulatory potential, which would not be elicited in the natural course of CML or during the therapy with TKIs. Thus, IRF8 seems to “convert” BCR-ABL from a dendritic cell suppressor to a dendritic cell activator.

We have previously shown that IRF8 overrides the mitogenic activity of BCR-ABL in differentiating myeloid progenitor cells (30). It has also been shown that IRF8 antagonizes the antiapoptotic effect of BCR-ABL (44). Moreover, coexpression of IRF8 inhibits BCR-ABL–induced myeloid hyperplasia in vivo (31). Neutrophil differentiation is promoted by BCR-ABL (45), but potently inhibited by IRF8 (24). Therefore, we expect the restoration of IRF8 to have two distinct effects. One is to directly reduce tumor burden by inhibiting abnormally enhanced growth and survival of myeloid cells as well as disproportionate neutrophil differentiation. The other is to induce effective anti-CML immunity by restoring dendritic cell development and by cooperating with BCR-ABL to strengthen dendritic cell functionality. In this regard, IRF8 may be the key to establishing multidisciplinary therapy for CML.

We propose that the selective restoration of IRF8 will serve as a hallmark for new CML therapies that may overcome the problems with current TKI therapy. First, the effects of IRF8 supplementation are not affected by TKI-resistant mutations in BCR-ABL. Second, unlike TKIs, IRF8 therapy would generate BCR-ABL+ dendritic cells with enhanced functionality to induce anti-CML CTL responses. Finally, once anti-CML CTLs are induced, it would be possible to discontinue the IRF8 therapy. It should be noted, however, that the possibility of autoimmune reactions by these hyperfunctional dendritic cells will need to be evaluated.

To develop a method for restoration of IRF8 expression, it will be essential to understand the mechanism through which BCR-ABL inhibits IRF8 expression and dendritic cell development. Our data seem to provide some insights into these mechanisms. Unlike Irf8−/− mice, which show severe reduction in CDP counts but not in MDP counts (23, 46), CML model mice lack both MDPs and CDPs. Thus, the dendritic cell differentiation stage affected by BCR-ABL is upstream of that affected by the loss of IRF8. This suggests that BCR-ABL acts on machinery that is required for the expression of Irf8 (the mechanism of which has not been clarified yet) as well as the generation of MDPs.

One possibility raised from the above context is inhibition of the transcription factor PU.1 by GATA1. PU.1 is expressed in similar sets of hematopoietic cell lineages as IRF8 and is required for the generation of MDPs (35). PU.1 activity is inhibited by GATA1, which is essential for the generation of MEPs (47). Our data showed that BCR-ABL strongly induced Gata1 expression. Moreover, GATA1 targets were indeed significantly enriched in genes induced by BCR-ABL and that the percentage of MEPs was increased in BCR-ABL mice. Interestingly, GATA1 has been shown to inhibit Irf8 expression, possibly by preventing PU.1 recruitment to the Irf8 promoter in dendritic cells (48). Moreover, GATA1 activity can be augmented via PI3K/AKT, a signaling pathway stimulated by BCR-ABL (49). Therefore, inhibition of PU.1 through induction and activation of GATA1 by BCR-ABL is an attractive possibility. Other possibilities included the involvement of STAT5. It has been reported that GM-CSF inhibits Irf8 expression via STAT5 (50). Thus, it was tempting to speculate that BCR-ABL–activated STAT5 suppresses IRF8 expression. However, our experiments using the STAT5 inhibitor did not support this. Finally, even though we observed that TSA or 5-Aza, as single agents or in combination with imatinib, failed to rescue Irf8 expression, it is still important to consider the possibility that other histone modifications, such as methylation, may be involved.

In conclusion, our present study provides new insights into CML pathogenesis and the basis of new therapy for CML in terms of dendritic cell biology. Further investigation of the mechanisms through which BCR-ABL suppresses IRF8 expression will be required to develop a method to selectively restore IRF8 expression.

No potential conflicts of interest were disclosed.

Conception and design: T. Tamura

Development of methodology: T. Watanabe, C. Hotta, T. Tamura

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Watanabe, C. Hotta, S.-I. Koizumi, M. Nakazawa, H. Fujita, R. Sakai, S. Fujisawa, Z. Ikezawa, M. Aihara, Y. Ishigatsubo

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.-I. Koizumi, J. Nakabayashi, A. Nishiyama, M. Aihara, T. Tamura

Writing, review, and/or revision of the manuscript: T. Watanabe, S.-I. Koizumi, T. Tamura

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C. Hotta, K. Miyashita, D. Kurotaki, G.R. Sato, M. Yamamoto, A. Nishiyama

Study supervision: T. Tamura

The authors thank Drs. Warren Pear, Ricardo A. Feldman, and Huabao Xiong for providing retroviral vectors.

This work was supported by KAKENHI Grants-in-Aid 24390246 and 24118002 from the Japan Society for the Promotion of Science, a Special Coordination Fund for Promoting Science and Technology from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan and grants from the Yokohama Foundation for Advancement of Medical Science and the Kato Memorial Bioscience Foundation (T. Tamura).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Sawyers
CL
. 
Chronic myeloid leukemia
.
N Engl J Med
1999
;
340
:
1330
40
.
2.
Jabbour
E
,
Kantarjian
H
. 
Chronic myeloid leukemia: 2012 Update on diagnosis, monitoring, and management
.
Am J Hematol
2012
;
87
:
1037
45
.
3.
Wong
S
,
Witte
ON
. 
The BCR-ABL story: bench to bedside and back
.
Annu Rev Immunol
2004
;
22
:
247
306
.
4.
O'Hare
T
,
Zabriskie
MS
,
Eiring
AM
,
Deininger
MW
. 
Pushing the limits of targeted therapy in chronic myeloid leukaemia
.
Nat Rev Cancer
2012
;
12
:
513
26
.
5.
Appel
S
,
Balabanov
S
,
Brummendorf
TH
,
Brossart
P
. 
Effects of imatinib on normal hematopoiesis and immune activation
.
Stem Cells
2005
;
23
:
1082
8
.
6.
Krusch
M
,
Salih
HR
. 
Effects of BCR-ABL inhibitors on anti-tumor immunity
.
Curr Med Chem
2011
;
18
:
5174
84
.
7.
Fujita
H
,
Kitawaki
T
,
Sato
T
,
Maeda
T
,
Kamihira
S
,
Takaori-Kondo
A
, et al
The tyrosine kinase inhibitor dasatinib suppresses cytokine production by plasmacytoid dendritic cells by targeting endosomal transport of CpG DNA
.
Eur J Immunol
2013
;
43
:
93
103
.
8.
Steinman
RM
,
Banchereau
J
. 
Taking dendritic cells into medicine
.
Nature
2007
;
449
:
419
26
.
9.
Liu
K
,
Nussenzweig
MC
. 
Origin and development of dendritic cells
.
Immunol Rev
2010
;
234
:
45
54
.
10.
Molldrem
JJ
,
Lee
PP
,
Wang
C
,
Felio
K
,
Kantarjian
HM
,
Champlin
RE
, et al
Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia
.
Nat Med
2000
;
6
:
1018
23
.
11.
Burchert
A
,
Wölfl
S
,
Schmidt
M
,
Brendel
C
,
Denecke
B
,
Cai
D
, et al
Interferon-α, but not the ABL-kinase inhibitor imatinib (STI571), induces expression of myeloblastin and a specific T-cell response in chronic myeloid leukemia
.
Blood
2003
;
101
:
259
64
.
12.
Lim
SH
,
Coleman
S
. 
Chronic myeloid leukemia as an immunological target
.
Am J Hematol
1997
;
54
:
61
7
.
13.
Kolb
HJ
,
Mittermuller
J
,
Clemm
C
,
Holler
E
,
Ledderose
G
,
Brehm
G
, et al
Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients
.
Blood
1990
;
76
:
2462
5
.
14.
Mohty
M
,
Isnardon
D
,
Vey
N
,
Brière
F
,
Blaise
D
,
Olive
D
, et al
Low blood dendritic cells in chronic myeloid leukaemia patients correlates with loss of CD34+/CD38 primitive haematopoietic progenitors
.
Br J Haematol
2002
;
119
:
115
8
.
15.
Boissel
N
,
Rousselot
P
,
Raffoux
E
,
Cayuela
JM
,
Maarek
O
,
Charron
D
, et al
Defective blood dendritic cells in chronic myeloid leukemia correlate with high plasmatic VEGF and are not normalized by imatinib mesylate
.
Leukemia
2004
;
18
:
1656
61
.
16.
Eisendle
K
,
Lang
A
,
Eibl
B
,
Nachbaur
D
,
Glassl
H
,
Fiegl
M
, et al
Phenotypic and functional deficiencies of leukaemic dendritic cells from patients with chronic myeloid leukaemia
.
Br J Haematol
2003
;
120
:
63
73
.
17.
Dong
R
,
Cwynarski
K
,
Entwistle
A
,
Marelli-Berg
F
,
Dazzi
F
,
Simpson
E
, et al
Dendritic cells from CML patients have altered actin organization, reduced antigen processing, and impaired migration
.
Blood
2003
;
101
:
3560
7
.
18.
Mumprecht
S
,
Claus
C
,
Schürch
C
,
Pavelic
V
,
Matter
MS
,
Ochsenbein
AF
. 
Defective homing and impaired induction of cytotoxic T cells by BCR/ABL-expressing dendritic cells
.
Blood
2009
;
113
:
4681
9
.
19.
Tamura
T
,
Yanai
H
,
Savitsky
D
,
Taniguchi
T
. 
The IRF family transcription factors in immunity and oncogenesis
.
Annu Rev Immunol
2008
;
26
:
535
84
.
20.
Aliberti
J
,
Schulz
O
,
Pennington
DJ
,
Tsujimura
H
,
Reis e Sousa
C
,
Ozato
K
, et al
Essential role for ICSBP in the in vivo development of murine CD8α+ dendritic cells
.
Blood
2003
;
101
:
305
10
.
21.
Tamura
T
,
Tailor
P
,
Yamaoka
K
,
Kong
HJ
,
Tsujimura
H
,
O'Shea
JJ
, et al
IFN regulatory factor-4 and -8 govern dendritic cell subset development and their functional diversity
.
J Immunol
2005
;
174
:
2573
81
.
22.
Schiavoni
G
,
Mattei
F
,
Sestili
P
,
Borghi
P
,
Venditti
M
,
Morse
HC
 III
, et al
ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8α(+) dendritic cells
.
J Exp Med
2002
;
196
:
1415
25
.
23.
Kurotaki
D
,
Osato
N
,
Nishiyama
A
,
Yamamoto
M
,
Ban
T
,
Sato
H
, et al
Essential role of the IRF8-KLF4 transcription factor cascade in murine monocyte differentiation
.
Blood
2013
;
121
:
1839
49
.
24.
Tamura
T
,
Nagamura-Inoue
T
,
Shmeltzer
Z
,
Kuwata
T
,
Ozato
K
. 
ICSBP directs bipotential myeloid progenitor cells to differentiate into mature macrophages
.
Immunity
2000
;
13
:
155
65
.
25.
Miyagawa
F
,
Zhang
H
,
Terunuma
A
,
Ozato
K
,
Tagaya
Y
,
Katz
SI
. 
Interferon regulatory factor 8 integrates T-cell receptor and cytokine-signaling pathways and drives effector differentiation of CD8 T cells
.
Proc Natl Acad Sci U S A
2012
;
109
:
12123
8
.
26.
Holtschke
T
,
Lohler
J
,
Kanno
Y
,
Fehr
T
,
Giese
N
,
Rosenbauer
F
, et al
Immunodeficiency and chronic myelogenous leukemia-like syndrome in mice with a targeted mutation of the ICSBP gene
.
Cell
1996
;
87
:
307
17
.
27.
Hambleton
S
,
Salem
S
,
Bustamante
J
,
Bigley
V
,
Boisson-Dupuis
S
,
Azevedo
J
, et al
IRF8 mutations and human dendritic-cell immunodeficiency
.
N Engl J Med
2011
;
365
:
127
38
.
28.
Schmidt
M
,
Nagel
S
,
Proba
J
,
Thiede
C
,
Ritter
M
,
Waring
JF
, et al
Lack of interferon consensus sequence binding protein (ICSBP) transcripts in human myeloid leukemias
.
Blood
1998
;
91
:
22
9
.
29.
Schmidt
M
,
Hochhaus
A
,
Nitsche
A
,
Hehlmann
R
,
Neubauer
A
. 
Expression of nuclear transcription factor interferon consensus sequence binding protein in chronic myeloid leukemia correlates with pretreatment risk features and cytogenetic response to interferon-alpha
.
Blood
2001
;
97
:
3648
50
.
30.
Tamura
T
,
Kong
HJ
,
Tunyaplin
C
,
Tsujimura
H
,
Calame
K
,
Ozato
K
. 
ICSBP/IRF-8 inhibits mitogenic activity of p210 Bcr/Abl in differentiating myeloid progenitor cells
.
Blood
2003
;
102
:
4547
54
.
31.
Hao
SX
,
Ren
R
. 
Expression of interferon consensus sequence binding protein (ICSBP) Is downregulated in Bcr-Abl-induced murine chronic myelogenous leukemia-like disease, and forced coexpression of ICSBP inhibits Bcr-Abl-induced myeloproliferative disorder
.
Mol Cell Biol
2000
;
20
:
1149
61
.
32.
Deng
M
,
Daley
GQ
. 
Expression of interferon consensus sequence binding protein induces potent immunity against BCR/ABL-induced leukemia
.
Blood
2001
;
97
:
3491
7
.
33.
Nardi
V
,
Naveiras
O
,
Azam
M
,
Daley
GQ
. 
ICSBP-mediated immune protection against BCR-ABL-induced leukemia requires the CCL6 and CCL9 chemokines
.
Blood
2009
;
113
:
3813
20
.
34.
Nakagawa
Y
,
Watari
E
,
Shimizu
M
,
Takahashi
H
. 
One-step simple assay to determine antigen-specific cytotoxic activities by single-color flow cytometry
.
Biomed Res
2011
;
32
:
159
66
.
35.
Geissmann
F
,
Manz
MG
,
Jung
S
,
Sieweke
MH
,
Merad
M
,
Ley
K
. 
Development of monocytes, macrophages, and dendritic cells
.
Science
2010
;
327
:
656
61
.
36.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
37.
Miller
JC
,
Brown
BD
,
Shay
T
,
Gautier
EL
,
Jojic
V
,
Cohain
A
, et al
Deciphering the transcriptional network of the dendritic cell lineage
.
Nat Immunol
2012
;
13
:
888
99
.
38.
Lindstedt
M
,
Johansson-Lindbom
B
,
Borrebaeck
CA
. 
Global reprogramming of dendritic cells in response to a concerted action of inflammatory mediators
.
Int Immunol
2002
;
14
:
1203
13
.
39.
Green
DR
,
Ferguson
T
,
Zitvogel
L
,
Kroemer
G
. 
Immunogenic and tolerogenic cell death
.
Nat Rev Immunol
2009
;
9
:
353
63
.
40.
Murphy
TL
,
Cleveland
MG
,
Kulesza
P
,
Magram
J
,
Murphy
KM
. 
Regulation of interleukin 12 p40 expression through an NF-κB half-site
.
Mol Cell Biol
1995
;
15
:
5258
67
.
41.
Hildner
K
,
Edelson
BT
,
Purtha
WE
,
Diamond
M
,
Matsushita
H
,
Kohyama
M
, et al
Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity
.
Science
2008
;
322
:
1097
100
.
42.
Fuertes
MB
,
Kacha
AK
,
Kline
J
,
Woo
SR
,
Kranz
DM
,
Murphy
KM
, et al
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells
.
J Exp Med
2011
;
208
:
2005
16
.
43.
Diamond
MS
,
Kinder
M
,
Matsushita
H
,
Mashayekhi
M
,
Dunn
GP
,
Archambault
JM
, et al
Type I interferon is selectively required by dendritic cells for immune rejection of tumors
.
J Exp Med
2011
;
208
:
1989
2003
.
44.
Burchert
A
,
Cai
D
,
Hofbauer
LC
,
Samuelsson
MK
,
Slater
EP
,
Duyster
J
, et al
Interferon consensus sequence binding protein (ICSBP; IRF-8) antagonizes BCR/ABL and down-regulates bcl-2
.
Blood
2004
;
103
:
3480
9
.
45.
Kobayashi
S
,
Kimura
F
,
Ikeda
T
,
Osawa
Y
,
Torikai
H
,
Kobayashi
A
, et al
BCR-ABL promotes neutrophil differentiation in the chronic phase of chronic myeloid leukemia by downregulating c-Jun expression
.
Leukemia
2009
;
23
:
1622
7
.
46.
Becker
AM
,
Michael
DG
,
Satpathy
AT
,
Sciammas
R
,
Singh
H
,
Bhattacharya
D
. 
IRF-8 extinguishes neutrophil production and promotes dendritic cell lineage commitment in both myeloid and lymphoid mouse progenitors
.
Blood
2012
;
119
:
2003
12
.
47.
Zhang
P
,
Behre
G
,
Pan
J
,
Iwama
A
,
Wara-Aswapati
N
,
Radomska
HS
, et al
Negative cross-talk between hematopoietic regulators: GATA proteins repress PU.1
.
Proc Natl Acad Sci U S A
1999
;
96
:
8705
10
.
48.
Shimokawa
N
,
Nishiyama
C
,
Nakano
N
,
Maeda
K
,
Suzuki
R
,
Hara
M
, et al
Suppressive effects of transcription factor GATA-1 on cell type-specific gene expression in dendritic cells
.
Immunogenetics
2010
;
62
:
421
9
.
49.
Zhao
W
,
Kitidis
C
,
Fleming
MD
,
Lodish
HF
,
Ghaffari
S
. 
Erythropoietin stimulates phosphorylation and activation of GATA-1 via the PI3-kinase/AKT signaling pathway
.
Blood
2006
;
107
:
907
15
.
50.
Esashi
E
,
Wang
YH
,
Perng
O
,
Qin
XF
,
Liu
YJ
,
Watowich
SS
. 
The signal transducer STAT5 inhibits plasmacytoid dendritic cell development by suppressing transcription factor IRF8
.
Immunity
2008
;
28
:
509
20
.