You are on page 1of 16

IFN Regulatory Factor 8 Represses GM-CSF

Expression in T Cells To Affect Myeloid Cell


Lineage Differentiation
This information is current as Amy V. Paschall, Ruihua Zhang, Chen-Feng Qi, Kankana
Bardhan, Liang Peng, Geming Lu, Jianjun Yang, Miriam

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
of June 5, 2015.
Merad, Tracy McGaha, Gang Zhou, Andrew Mellor, Scott I.
Abrams, Herbert C. Morse III, Keiko Ozato, Huabao Xiong
and Kebin Liu
J Immunol 2015; 194:2369-2379; Prepublished online 2
February 2015;
doi: 10.4049/jimmunol.1402412
http://www.jimmunol.org/content/194/5/2369

Supplementary http://www.jimmunol.org/content/suppl/2015/01/31/jimmunol.140241
Material 2.DCSupplemental.html
References This article cites 73 articles, 38 of which you can access for free at:
http://www.jimmunol.org/content/194/5/2369.full#ref-list-1
Subscriptions Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscriptions
Permissions Submit copyright permission requests at:
http://www.aai.org/ji/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/cgi/alerts/etoc

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright 2015 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

IFN Regulatory Factor 8 Represses GM-CSF Expression in


T Cells To Affect Myeloid Cell Lineage Differentiation

Amy V. Paschall,*,,,1 Ruihua Zhang,x,1 Chen-Feng Qi,{ Kankana Bardhan,*


Liang Peng,x Geming Lu,x Jianjun Yang,x Miriam Merad,x Tracy McGaha,
Gang Zhou, Andrew Mellor, Scott I. Abrams, Herbert C. Morse, III,{ Keiko Ozato,#
Huabao Xiong,x and Kebin Liu*,,

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
During hematopoiesis, hematopoietic stem cells constantly differentiate into granulocytes and macrophages via a distinct differ-
entiation program that is tightly controlled by myeloid lineage-specific transcription factors. Mice with a null mutation of IFN
regulatory factor 8 (IRF8) accumulate CD11b+Gr1+ myeloid cells that phenotypically and functionally resemble tumor-
induced myeloid-derived suppressor cells (MDSCs), indicating an essential role of IRF8 in myeloid cell lineage differentiation.
However, IRF8 is expressed in various types of immune cells, and whether IRF8 functions intrinsically or extrinsically in
regulation of myeloid cell lineage differentiation is not fully understood. In this study, we report an intriguing finding that,
although IRF8-deficient mice exhibit deregulated myeloid cell differentiation and resultant accumulation of CD11b+Gr1+ MDSCs,
surprisingly, mice with IRF8 deficiency only in myeloid cells exhibit no abnormal myeloid cell lineage differentiation. Instead,
mice with IRF8 deficiency only in T cells exhibited deregulated myeloid cell differentiation and MDSC accumulation. We further
demonstrated that IRF8-deficient T cells exhibit elevated GM-CSF expression and secretion. Treatment of mice with GM-CSF
increased MDSC accumulation, and adoptive transfer of IRF8-deficient T cells, but not GM-CSFdeficient T cells, increased
MDSC accumulation in the recipient chimeric mice. Moreover, overexpression of IRF8 decreased GM-CSF expression in T cells.
Our data determine that, in addition to its intrinsic function as an apoptosis regulator in myeloid cells, IRF8 also acts extrinsically
to repress GM-CSF expression in T cells to control myeloid cell lineage differentiation, revealing a novel mechanism that the
adaptive immune component of the immune system regulates the innate immune cell myelopoiesis in vivo. The Journal of
Immunology, 2015, 194: 23692379.

M
yeloid cells are among the most abundant and het- local microenvironment to maintain homeostasis (14). All mye-
erogeneous hematopoietic cell types that are virtually loid cells originate from the pluripotent hematopoietic stem cells
present in all mammalian tissues, where they monitor that undergo progressive restriction in their lineage potential to
give rise to mature granulocytes and macrophages (58). Lineage
restriction and differentiation are regulated by timely activation of
*Department of Biochemistry and Molecular Biology, Medical College of Georgia, specific set of lineage-specific transcription factors in concert with
Georgia Regents University, Augusta, GA 30912; Cancer Immunology, Inflamma-
tion and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, downregulation of other set(s) of transcription factors that are
GA 30912; Charlie Norwood VA Medical Center, Augusta, GA 30904; xImmunol- important for alternative cell lineage potential (9, 10). Deregula-
ogy Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029;
{
Laboratory of Immunogenetics, National Institute of Allergy and Infectious Dis- tion of activities of these lineage-specific transcription factors
eases, National Institutes of Health, Bethesda, MD 20892; Department of Immu- often leads to hematopoietic disorders and leukemia (11, 12).
nology, Roswell Park Cancer Institute, Buffalo, NY 14263; and #Programs in
Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child
Therefore, lineage-specific transcription factors are essential for
Health and Human Development, National Institutes of Health, Bethesda, myeloid cell lineage differentiation and maturation (11, 1315).
MD 20892 Several lineage-specific transcription factors have been found to
1
A.V.P. and R.Z. equally contributed to this study. be essential for regulation of myeloid cell differentiation. The
Received for publication September 23, 2014. Accepted for publication December E-twenty-six (ETS) family transcription factor PU.1 is necessary for
30, 2014.
the earliest steps of myeloid lineage commitment, and its deficiency
This work was supported by National Institutes of Health Grants CA133085,
CA185909, and CA182518 (to K.L.); DK072201, AI091871, and AI104688 (to
results in loss of monocytes and granulocytes (1618). Kr uppel-
H.X.); and CA140622 (to S.I.A.); and in part by the Intramural Research Program like factor-4 (KLF4) is expressed in a monocyte-restricted and
of the National Institutes of Health, National Institute of Allergy and Infectious stage-specific pattern during myelopoiesis and functions to pro-
Diseases (to H.C.M.), the National Institute of Child Health and Human Develop-
ment (to K.O.), and VA Merit Review Award BX001962 (to K.L.). mote monocyte differentiation (19). KLF4 deficiency inhibited
Address correspondence and reprint requests to Dr. Huabao Xiong or Dr. Kebin Liu, monocyte, but increased granulocyte differentiation (20). Fur-
Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029 thermore, hematopoietic KLF42/2 chimeras generated by trans-
(H.X.) or Department of Biochemistry and Molecular Biology, Medical College of
Georgia, Georgia Regents University, Augusta, GA 30912 (K.L.). E-mail addresses:
plantation of KLF42/2 fetal liver cells completely lack circulating
huabao.xiong@mssm.edu (H.X.) or kliu@gru.edu (K.L.) inflammatory Ly6G+ monocytes (21). In contrast, the nuclear or-
The online version of this article contains supplemental material. phan receptor NR4A1 controls later stages of monocyte devel-
Abbreviations used in this article: BM, bone marrow; ETS, E-twenty-six; IRF, IFN opment, and is required for the generation of Ly6C2 monocytes,
regulatory factor; KLF4, Kr uppel-like factor-4; KO, knockout; MDSC, myeloid- but not Ly6C+ monocytes (22). The bZip transcription factor
derived suppressor cell; RV, retroviral vector; WT, wild-type.
MafB is expressed specifically in the myeloid lineage of the he-
Copyright 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00 matopoietic system and is upregulated successively during myeloid

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1402412
2370 IRF8 REPRESSES GM-CSF TO CONTROL MDSC ACCUMULATION

differentiation from multipotent progenitors to macrophages, and stages of myelopoiesis, namely the differentiation and lineage
thus plays an essential role in early myeloid and monocytic dif- commitment of monocytes/macrophages versus granulocytes under
ferentiation (23). c-Maf has been observed to induce monocytic physiological conditions (36, 37). However, the mechanism un-
differentiation in bipotent myeloid progenitors (24), whereas the derlying IRF8 function in myeloid cell differentiation and MDSC
zinc finger transcription factor Egr-1 is essential for lineage dif- accumulation is still elusive. Furthermore, IRF8 is expressed in both
ferentiation of macrophages, but not granulocytes (25). lymphoid and myeloid cells, but little is known about its relative
IFN regulatory factor (IRF)8 is a member of the IRF family that functions in lymphoid and myeloid cells in regulation of myeloid
was identified in the late 1980s in the context of research into the cell differentiation and MDSC accumulation.
type I IFN system. Subsequent studies over the past three decades We report in this work a surprising finding that IRF8 expressed in
have revealed that IRF8 is an essential transcription factor myeloid cells regulates myeloid cell spontaneous apoptosis,
for myeloid-lineage restriction and differentiation (68, 2631). whereas IRF8 expressed in T cells represses GM-CSF expression to
IRF8 functions as either a transcriptional activator or repressor, suppress MDSC differentiation from myeloid progenitor cells. We
depending on its binding partners in which it forms different proposed that IRF8 deficiency in T cells leads to increased GM-

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
heterodimeric protein complexes and on its target DNA elements CSF expression to induce MDSC differentiation, whereas IRF8
(10, 19, 32). For example, IRF8 forms a protein complex with deficiency in myeloid cells leads to decreased myeloid cell
IRF1 or IRF2 and binds to the IFN-stimulated response element spontaneous apoptosis. Thus, cooperative IRF8 functions in T cells
(A/GNGAAANNGAAACT) to repress gene transcription. Con- and myeloid cells mediate myeloid cell differentiation and ho-
versely, the IRF8-PU.1 heterodimer complex specifically binds to meostasis in vivo.
the ETS-IRF composite element (GGAANNGAAA) or the IRF-
ETS composite sequence (GAAANN[N]GGAA) to activate gene Materials and Methods
transcription (19, 27, 33, 34). In humans, a loss-of-function mu- Mice
tation of IRF8 leads to monocytic and dendritic cell immunode-
IRF8 knockout (KO) mice were generated as previously described (36). The
ficiency (35). A hallmark of IRF8 null mice is deregulated IRF8 T KO mice were generated as previously described (39). The loxp-
myeloid cell lineage differentiation, as characterized by lack of flanked Irf8 gene [B6(Cg)-Irf8tm1.1Hm/J] were generated as previously de-
myeloid cell maturation, including lack of bone marrow (BM) scribed (40). The Lyz2-cre mice [B6.129P2-Lyz2tm1(cre)Ifo/J] were obtained
resident macrophages, CD8a+ dendritic cells, and plasmacytoid from The Jackson Laboratory. Use of mice was performed according to
approved protocols by institutional animal use and care committee in Icahn
dendritic cells in lymphoid organs (36, 37). This deregulated School of Medicine at Mount Sinai and Georgia Regents University.
myeloid cell differentiation causes massive accumulation of het-
erogeneous CD11b+Gr1+ myeloid cells that phenotypically and Antibodies
functionally resemble myeloid-derived suppressor cells (MDSCs) The following Abs were purchased from BD Biosciences, BioLegend
(38). Therefore, IRF8 is essential for the regulation of diverse (San Diego, CA), or eBiosciences, as conjugated to FITC, PE, PE-Cy5,

FIGURE 1. Creation of mice with IRF8 deficiency


only in myeloid cells. (A) Diagram of creation and
analysis of mice with IRF8 deficiency only in the my-
eloid cells (IRF8 MKO mice). Mice with loxp-flanked Irf8
gene [B6(Cg)-Irf8tm1.1Hm/J] (irf8loxp+/+, top left) were
crossed to the Lyz2-cre mice [B6.129P2-Lyz2tm1(cre)Ifo/J]
(Lyz2cre+/+, bottom left). irf8loxp+/+ mice: exon 2 (E2)
was flanked with loxp. E1 and E3 indicate exons 1 and
3 of the irf8 gene. Lyz2cre+/+ mice: cre coding sequence
was inserted in Lyz2 gene exon 1. E2: Lyz2 exon 2. Lyz2
P: Lyz2 promoter. (B) Myeloid cells of IRF8 MKO
mice express mutant IRF8 mRNA. Gr1+ (lane 1), CD11b+
(lane 2), and CD11b+Gr1+ (lane 3) cells were sorted from
WT and IRF8 MKO mice; stimulated with IFN-g
(100 U/ml) and LPS (1 mg/ml) overnight; and ana-
lyzed by RT-PCR for IRF8 mRNA levels. The WT
IRF8 (IRF8WT) and truncated mutant IRF8 mRNA
(IRF8mmt) are indicated at the right. (C) IRF8 is
functionally deficient in myeloid cells of IRF8 MKO
mice. Cells as shown in (B) were analyzed for Il12b
(IL12p40), Nos2 (inducible NO synthase), Cxcr3 (IFN-g
inducible protein 10), and Ccl3 (MIP1a) transcripts by
real-time RT-PCR using b-actin as internal controls.
The Journal of Immunology 2371

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
FIGURE 2. Mice with IRF8 deficiency only in myeloid cells exhibit normal myelopoiesis. (A) Spleen morphology of 3-mo-old WT, IRF8 MKO,
and IRF8 KO mice. (B) Percentages of CD11b+Gr1+ MDSCs in thymus (Thy), spleen (SP), lymph node (LN), and BM of WT and IRF8 MKO mice.
Shown are representative results from one mouse of three mice. (C) Quantitation of CD11b+Gr1+ in SP and BM of WT (n = 3) and IRF8 MKO (n = 3)
mice, as shown in (B). (D) Subsets of Gr1+ (Ly6C+ and Ly6G+) myeloid cells SP and BM. Shown are representative results from one mouse of three
mice. Column, mean; bar, SD. (E) Quantification of subsets of Gr1+ myeloid cells in SP and BM of WT (n = 3) and IRF8MKO (n = 3) mice. (F) The
indicated tissues were collected from WT and IRF8 MKO mice, stained with the CD4- and CD8-specific mAbs. Shown are representative images
CD4+ and CD8+ T cell profiles. (G) Quantification of CD4+ and CD8+ T cells in the indicated tissues from WT (n = 3) and IRF8 MKO (n = 3) mice.
Column, mean; bar, SD.

PerCP-Cy5.5, or allophycocyanin: CD4 (L3T4), CD8 (53-6.7), CD3e (145- T cell transfer models
2C11), Gr-1(RB6-8C5), CD11b(M1/70), H-2kb (AF6-88.5.5.3), GM-CSF
(MP1-22E9), and the respective IgG isotype controls. T cell transfer was performed as previously described (Powrie et al. 1993). In
brief, purified CD4+ T cells from wild-type (WT) and IRF8 KO mice were
Intracellular staining and flow cytometry injected i.p. into Rag1 KO recipients (5 3 105 cells per mouse in 200 ml
sterile PBS per injection). Mice were weighed every week throughout the
Cells were stimulated with anti-CD3 and anti-CD28 mAbs for 3 d and course of experiments. After 4 wk, mice were sacrificed; spleen, mesenteric
restimulated with PMA and ionomycin for 5 h in the presence of brefeldin A lymph node, and BM were picked up for flow cytometer analysis.
before intracellular staining. Cells were fixed with IC Fixation Buffer (BD
Biosciences), incubated with permeabilization buffer, and stained with PE IRF8 overexpression and GM-CSF expression in T cells
anti-mouse, allophycocyanin anti-mouse, and FITC anti-mouse Abs. Flow
cytometry was performed on a FACSCalibur (BD Biosciences) and LSR The retroviral vector (RV) used in this study was the MIG retroviral vector
Fortessa (BD Biosciences). that consisted of a MSCV-IRES-GFP vector. Mouse naive CD4+ T cells
2372 IRF8 REPRESSES GM-CSF TO CONTROL MDSC ACCUMULATION

from C57BL/6 WT and IRF8 KO mouse spleens were transduced with Chromatin immunoprecipitation
empty RV control (RV-MIG) and the RV containing the IRF8 coding
sequence (RV-MIG-IRF8), respectively. The cells were activated under CD4+ T cells were isolated from spleens of WT mice using negative selection
anti-CD3 and anti-CD28 Abs for 3 d; then restimulated with PMA/ method (BioLegend) and stimulated with anti-CD3 and anti-CD8 mAbs for
ionomycin for 5 h; and stained for surface CD4 and intracellular GM- 3 d. Chromatin immunoprecipitation was performed using anti-IRF8 Ab
CSF. Total RNA was also isolated and analyzed for GM-CSF expression (C19; Santa Cruz, Dallas, TX) and the protein A agarose, according to the
by real-time RT-PCR. manufacturers instructions (Millipore, Temecula, CA). Goat IgG was used
as negative control for the Ab. The immunoprecipitated genomic DNA was
Acute graft-versus-host disease model amplified by real-time PCR using five pairs of PCR primers (Supplemental
Table I) covering the region approximately from 24000 to +1000 relative to
All donor BM cells were isolated from C57BL/6 (H-2b) mice. T cell de- the csf2 transcription initiation site in the csf2 promoter region.
pletion of BM was performed with auto-MACS by using anti-CD90.2
microbeads (Miltenyi Biotec). Donor CD4+ T cells (purity 95%) were
purified from the spleens of C57BL/6 or GM-CSF2/2 mice (H-2b) by Results
using the CD4 beads (Miltenyi Biotec). The BALB/c (H-2d) hosts were A key phenotype of IRF8 null mice is deregulation of myeloid
irradiated with 9 Gy from 137Cs source. One day later, the hosts were cell lineage differentiation
injected i.v. with 2 3 106 BM cells only or combined with 0.3 3 106 CD4+

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
T cells isolated from C57BL/6 or GM-CSF KO mice. The hosts were IRF8 is a transcription factor of the IRF family. Mice with a null
weighed every 2 d and monitored for survival. The H-2Kb+ myeloid cells mutation of IRF8 exhibit two prominent phenotypes (36). The first is
were gated and analyzed for CD11b+Gr1+ cells by FACS in spleen and BM enhanced susceptibility to virus infections associated with impaired
in host at 11 and 14 d after donor BM cell transfer.
IFN-g production. The second is deregulated myeloid cell lineage
Cytokine ELISA differentiation, characterized by splenomegaly (Supplemental
Fig. 1A) and massive accumulation of CD11b+Gr1+ MDSCs in
Supernatants from cell cultures were collected after activation under various
conditions, and secreted cytokines in the supernatants were measured by BM and spleen (Supplemental Fig. 1B). Therefore, IRF8 is a key
ELISA kits with purified coating and biotinylated detection Ab antiGM- transcription factor for myeloid cell lineage differentiation and is
CSF (eBioscience). essential for the proliferation and differentiation of hematopoietic
RT-PCR analysis progenitor cells into mature myeloid cells (36, 37).

Total RNA was isolated from cells using TRIzol (Invitrogen, San Diego, Myeloid cellspecific IRF8 deficiency does not ablate myeloid
CA), according to the manufacturers instructions, and used for the first- cell lineage differentiation
strand cDNA synthesis using the Moloney murine leukemia virus reverse
transcriptase (Promega). The cDNA was then used as template for semi- As mentioned above, IRF8-deficient mice exhibit deregulated
quantitative and real-time PCR analysis. The sequences of primers are myeloid cell lineage differentiation, resulting in accumulation of
listed in Supplemental Table I. MDSCs (Supplemental Fig. 1). In keeping with earlier studies (13,

FIGURE 3. MDSCs from IRF8 MKO mice exhibit


decrease spontaneous apoptosis. (A) MDSCs from IRF8
KO mice exhibit decreased spontaneous apoptosis.
Spleen and BM were collected from 3-mo-old WT (n =
3) and IRF8 KO (n = 3) mice. Cells were stained im-
mediately under cold conditions with CD11b- and Gr1-
specific mAbs plus annexin V and DAPI. CD11b+Gr1+
cells were gated for annexin V+DAPI+ cells. Shown are
representative results of one of three pairs of mice. Ap-
optosis was quantified as percentage of CD11b+Gr1+
cells that are also annexin V+DAPI+ cells and presented
at the right. Column, mean; bar, SD. (B) MDSCs from
IRF8 MKO mice also exhibit decreased spontaneous
apoptosis. Spleen and BM cells were collected from WT
(n = 5) and IRF8 MKO (n = 5) mice. Spontaneous apo-
ptosis was determined as in (A). *p , 0.05, **p , 0.01.
The Journal of Immunology 2373

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
FIGURE 4. IRF8 deficiency in T cells results in accumulation of MDSCs. (A) Spleen morphology of 3-mo-old WT and IRF8 TKO mice. (B)
Percentages of CD11b+Gr1+ MDSCs in thymus (Thy), spleen (SP), lymph node (LN), BM, and blood of WT and IRF8 TKO mice. Shown are rep-
resentative results from one mouse of three mice. (C) Quantitation of CD11b+Gr1+ MDSCs in the indicated tissues of WT (n = 3) and IRF8 TKO (n = 3)
mice. Column, mean; bar, SD. **p , 0.01. (D) Subsets of Gr1+ (Ly6C+ and Ly6G+) myeloid cells in SP and BM. Shown are representative results from
one mouse of three mice. (E) Quantification of subsets of Gr1+ myeloid cells in SP and BM of WT (n = 3) and IRF8 TKO (n = 3) mice, as shown in (D).
Column, mean; bar, SD. **p , 0.01.

19, 41, 42), this indicates that IRF8 functions in myeloid cells to IP1a expression levels are higher in Gr1+ cells from IRF8 MKO
regulate myeloid cell lineage differentiation. However, whether mice as compared with those from WT mice (Fig. 1C). IL12p40
IRF8 expressed in myeloid cells regulates myeloid cell lineage levels were also lower in CD11b+ and CD11b+Gr1+ cells in IRF8
differentiation is still a hypothesis to be tested. Therefore, we MKO mice as compared with WT mice (Fig. 1C). Our data thus
created mice with IRF8 deficiency only in myeloid cells by indicate that IRF8 is functionally deficient in these myeloid cells.
crossing mice with a loxp-flanked Irf8 gene [B6(Cg)-Irf8tm1.1Hm/J] Therefore, we have created mice with irf8 mutation and IRF8
(9, 40) with the Lyz2-cre mice [B6.129P2-Lyz2tm1(cre)Ifo/J] functional deficiency only in myeloid cells.
(Fig. 1A). Exon 2 of Irf8 in the B6(Cg)-Irf8tm1.1Hm/J mouse is flanked Surprisingly, analyses of IRF8 MKO mice revealed that they do
by loxp sites, and it has been shown that deletion of exon 2 leads not develop the splenomegaly characteristic of IRF8 KO mice
to depletion of IRF8 protein in cre-expressing tissues (40). We (Fig. 2A). No significant differences were observed in the per-
selected Irf8loxp+/+ and Lyz2cre+/+ mice based on genotypes centages of CD11b+, Gr1+, and CD11b+Gr1+ cells in Thy, spleen,
(termed IRF8 MKO) (Supplemental Fig. 2). To confirm that exon lymph nodes, and BM of WT and IRF8 MKO mice (Fig. 2B, 2C).
2 is indeed deleted from these mice, we designed PCR primers In addition, the subsets of monocytic and granulocytic MDSCs
that flank exon 2 of Irf8 mRNA. CD11b+, Gr1+, and CD11b+Gr1+ (Ly6G+ and Ly6C+) also did not differ significantly between WT
cells were sorted from WT and IRF8 MKO mice and treated with and IRF8 MKO cells (Fig. 2D, 2E). There were no significant
IFN-g and LPS for 24 h. RT-PCR analysis of IRF8 mRNA indi- differences between these mice in the percentages of CD4+ and
cated that exon 2 was indeed deleted from Irf8 mRNA in IRF8 CD8+ T cells (Fig. 2F, 2G). These data thus indicate that IRF8
MKO mice (Fig. 1B). To determine whether the myeloid cells in expressed in myeloid cells is not sufficient enough to mediate
IRF8 MKO mice are functionally deficient, the expression levels CD11b+Gr1+ MDSC differentiation.
of IRF8 target genes in these cells were analyzed. IRF8 is a tran-
scription activator of IL12p40 and inducible NO synthase and is IRF8 deficiency results in decreased spontaneous apoptosis
a transcriptional repressor of IFN-ginducible protein 10 and IP1a in vivo
(43, 44). CD11b+, Gr1+, and CD11b+Gr1+ cells were sorted from The above observation is a surprising one because IRF8-deficient
WT and IRF8 MKO mice. The cells were treated with IFN-g and myeloid cells from IRF8 MKO mice are functionally deficient
LPS overnight and then analyzed for the expression levels of these (Fig. 1C) and IRF8 conventional KO mice exhibit deregulated
four IRF8 target genes. IL12p40 and inducible NO synthase ex- myeloid cell lineage differentiation and resultant MDSC accu-
pression levels are lower, whereas IFN-ginducible protein 10 and mulation (36) (Supplemental Fig. 1). Therefore, we further func-
2374 IRF8 REPRESSES GM-CSF TO CONTROL MDSC ACCUMULATION

tionally examined these IRF8 MKO mice. Like T cells, myeloid


cell turnover is mediated by the FasFas ligand apoptosis pathway
(45). It has been shown that IRF8 regulates the expression of
multiple apoptosis-regulatory genes, including Bcl-xL, Bax, FLIP,
and Fas, in myeloid cells (4649), to regulate apoptosis (50, 51).
Based on these observations, we then hypothesized that IRF8
deficiency at least should lead to decreased spontaneous apoptosis
of MDSCs in IRF8 MKO mice. To test this hypothesis, we ex-
amined spontaneous apoptosis of MDSCs in WT and IRF8 MKO
mice with IRF8 KO mice as positive controls. As expected,
MDSCs from spleens and BM of IRF8 KO mice exhibited sig-
nificantly reduced spontaneous apoptosis as compared with those
from WT mice (Fig. 3A). Similar to what was observed in IRF8

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
KO mice, MDSCs from both spleens and BM of IRF8 MKO mice
also exhibit significantly decreased spontaneous apoptosis (Fig.
3B). We should point out in this work that MDSCs from IRF8 KO
and IRF8 MKO mice are all sensitive to apoptosis; only the level
of spontaneous apoptosis in MDSCs of IRF8-deficient mice de-
creased as compared with the respective control WT mice.
Therefore, we conclude that IRF8 deficiency leads to decreased
MDSC spontaneous apoptosis. However, this deficiency alone
apparently is not sufficient enough to cause MDSC accumulation.
IRF8 deficiency in T cells leads to increased MDSCs
Based on the above observations, we hypothesized that IRF8 FIGURE 5. IRF8-deficient T cells induce MDSC accumulation. (A)
expressed in other type(s) of cells plays a role in myeloid cell CD4+ T cells were purified from WT and IRF8 TKO mice and adoptively
differentiation. To test this hypothesis, we analyzed mice with IRF8 transferred to Rag1 KO mice. Mice were analyzed 14 d after cell transfer.
deficiency only in T cells (termed IRF8 TKO) (39). In IRF8 TKO Spleen morphology (left panel) and total spleen cells (right panel) are
mice, cre expression is controlled by lck promoter that drives shown. (B) MDSC profiles in the Rag1 KO mice, Rag1 KO mice that have
received WT T cells, and Rag1 KO mice that have received IRF8
T cellspecific cre expression. IRF8 TKO mice have enlarged
KO T cells, as shown in (A). Spleen cells were stained with H2Kb-,
spleens as compared with WT mice (Fig. 4A). The percentages of CD11b-, and Gr1-specific mAbs. H-2Kb+ cells were gated and analyzed for
CD11b+Gr1+ cells (Fig. 4B, 4C) in spleen, BM, and blood, and CD11b+Gr1+ cells. Shown are representative results of one of three mice
Ly6C+Ly6G+ (Fig. 4D, 4E) cells in spleen and BM were signifi- (top panel). MDSCs in each type of mice were quantified and presented at
cantly higher in IRF8 TKO mice as compared with WT mice. the bottom panel. Column, mean; bar, SD. **p , 0.01.
Next, we took a complementary approach to determine the role
of IRF8 expressed in T cells in regulation of MDSC differentiation. cells ex vivo (Fig. 6A). Next, purified CD4+ T cells from WT and
We purified CD4+ T cells from WT and IRF8 TKO mice and IRF8 KO mice were cultured in the presence of anti-CD3 and anti-
adoptively transferred these T cells to Rag1 KO mice. The ratio- CD28 mAbs. Analysis of intracellular GM-CSF protein levels
nale is that if IRF8 deficiency in T cells regulates myeloid cell using flow cytometry revealed that GM-CSF is higher in T cells
lineage differentiation, then T cells from IRF8 KO mice should from IRF8 KO mice as compared with T cells from WT mice
increase MDSC accumulation in the Rag1 KO recipients. Indeed, (Fig. 6B). Real-time RT-PCR analysis determined that GM-CSF
adoptive transfer of IRF8-deficient T cells to Rag1 KO mice transcript levels were also significantly higher in T cells from
resulted in increased spleen size and spleen cell number (Fig. 5A). IRF8 KO mice than WT mice (Fig. 6C). Analysis of GM-CSF
Analysis of CD11b+Gr1+ cells indicated that adoptive transfer of protein level in cell culture medium indicated that IRF8-deficient
IRF8-deficient T cells significantly increased CD11b+Gr1+ MDSC T cells secrete significantly more GM-CSF than WT T cells
accumulation in the Rag1 KO mouse spleens (Fig. 5B). Taken (Fig. 6C). Taken together, these observations indicate that IRF8
together, our data indicate that IRF8 expressed in T cells mediates functions as a repressor of GM-CSF in T cells.
CD11b+Gr1+ MDSC lineage differentiation in vivo.
T cellproduced GM-CSF promotes MDSC differentiation
IRF8 represses GM-CSF expression in T cells to regulate To functionally determine the role of GM-CSF in myeloid cell
myeloid cell differentiation differentiation in vivo, rGM-CSF protein was injected into WT
It is well documented that proinflammatory cytokines mediate mice. Analysis of GM-CSFtreated mice revealed that CD11b+
CD11b+Gr1+ immature myeloid cell differentiation under patho- Gr1+ MDSC increased significantly in blood, spleen, and BM
logical conditions (5254). In particular, GM-CSF has been shown (Fig. 6D, 6E). To further determine that activated T cells can
to induce CD11b+Gr1+ MDSC accumulation in tumor-bearing enhance myeloid cell development in vivo, we then made use of
mice (53, 5557). Recent studies have shown that tumor- an acute graft-versus-host disease model to determine the function
secreted GM-CSF induces differentiation of MDSCs that pheno- of T cellproduced GM-CSF in myeloid cell differentiation.
typically and functionally resemble tumor-induced MDSCs BALB/c (H-2d) recipient mice were irradiated. T celldepleted
(5557). Based on these observations, we hypothesized that IRF8 BM cells were prepared from C57BL/6 mice (H-2Kb). CD4+
represses GM-CSF expression and IRF8 deficiency in T cells T cells were purified from WT, IRF8 KO, and GM-CSF KO mice
results in increased GM-CSF expression to induce deregulated (C57BL/6, H-2Kb). BM alone or BM in combination with the
myeloid cell differentiation. To test this hypothesis, we first cul- purified T cells was then adoptively transferred to the recipient
tured BM cells from WT mice in the presence of GM-CSF. In- mice. In this graft-versus-host disease model, transferred T cells
deed, GM-CSF induced CD11b+Gr1+ cell differentiation from BM could be activated by three ways, as follows: 1) direct Ag pre-
The Journal of Immunology 2375

FIGURE 6. IRF8 represses GM-CSF ex-


pression in T cells to suppress MDSC ac-
cumulation. (A) GM-CSF induces BM cell
differentiation to CD11b+Gr1+ MDSCs. BM
cells were prepared (left panel) and cultured
in vitro in the presence of GM-CSF for 4 d
(right panel) and analyzed for CD11b+ and
Gr1+ cells. (B) Purified CD4+ T cells from
WT and IRF8 KO mice were stimulated
with anti-CD3 and anti-CD28 mAbs for 3 d.
Cells were stained intracellularly with GM-
CSFspecific mAb and analyzed by flow
cytometry. (C) Left panel, CD4+ T cells
were purified from spleens of WT and IRF8

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
KO mice and stimulated with anti-CD3 and
anti-CD28 mAbs for 3 d. RNAs were pre-
pared and analyzed by real-time RT-PCR
for GM-CSF mRNA level using b-actin as
internal control. Right panels, CD4+ T cells
were purified from spleens of WT, IRF8
KO, and IRF8 TKO mice and stimulated
with anti-CD3 and anti-CD28 mAbs for 3 d.
The culture supernatants were analyzed by
ELISA for GM-CSF protein level. Column,
mean; bar, SD. **p , 0.01. (D) WT mice
were injected i.p. with rGM-CSF protein
(1 mg in 100 ml PBS per mouse) every 2 d
three times. Myeloid cells in the indicated
tissues were analyzed by flow cytometry.
Shown are representative results from one
mouse of three mice. (E) Quantification of
CD11b+Gr1+ cells, as shown in (D). Col-
umn, mean; bar, SD. **p , 0.01.

sentation by donor APCs; 2) indirect Ag presentation by recipient To determine whether IRF8 also regulates GM-CSF expression
APCs (cross presenting); and 3) MLR. The rationale is that if in MDSCs, MDSCs were purified from spleen cells of WT and
IRF8 is a repressor of GM-CSF and activated T cellproduced IRF8 KO mice. Total RNA was isolated from the purified MDSCs
GM-CSF promotes CD11b+Gr1+ cell differentiation, then more and analyzed for GM-CSF mRNA expression by real-time RT-
H-2Kb+ CD11b+Gr1+ cells should accumulate in the recipient H-2d PCR. It is clear that, unlike in T cells, IRF8 deficiency in
mice. Analysis of the chimeric mice indicated that adoptive transfer MDSCs clearly does not increase GM-CSF expression in MDSCs
of T cells increased spleen size and spleen cell number as compared (Fig. 9). Taken together, our data demonstrate that IRF8 expressed
with BM-only transfer. However, both spleen sizes and number of in T cells functions as a GM-CSF repressor to control myeloid cell
spleen cells were greater in the recipient H-2d mice that received lineage differentiation, and IRF8 deficiency in T cells leads to
IRF8 KO T cells and BM as compared with recipients of WT increased GM-CSF expression to deregulate myeloid cell lineage
T cells and BM (Fig. 7A). Furthermore, T cells from GM-CSF KO differentiation and resultant MDSC accumulation.
mice did not induce increased spleen sizes and spleen cell numbers In summary, we determined that, under physiological conditions,
(Fig. 7A). IRF8 expressed in T cells represses GM-CSF expression to control
Consistent with the increased spleen sizes and spleen cell myeloid cell lineage differentiation, whereas IRF8 expressed in
numbers, the percentages of donor H-2Kb+ CD11b+Gr1+ cells were myeloid cells mediates myeloid cell spontaneous apoptosis to
higher in the recipient H-2d mice receiving IRF8 KO T cells than regulate myeloid cell turnover. Loss of IRF8 function in T cells
those receiving WT T cells. In contrast, mice receiving GM-CSF leads to increased GM-CSF expression to induce deregulated
KO T cells exhibited no differences in H-2Kb+ CD11b+Gr1+ cells myeloid cell differentiation, which acts in concert with loss of IRF8
as compared with control mice (Fig. 7B, 7C). function in myeloid cell and resultant decreased spontaneous ap-
A complementary approach was then used to further determine optosis to result in MDSC accumulation (Fig. 10).
the function of IRF8 in repression of GM-CSF expression. Naive
CD4+ T cells from C57BL/6 WT and IRF8 KO mice were Discussion
transduced with retroviral vector encoding IRF8 or empty vector, The mammalian immune system consists of an innate and an
and were activated with anti-CD3 and anti-CD28 Abs. The cells adaptive component. The innate immune system that comprises
were restimulated with PMA/ionomycin for 5 h and stained for primarily myeloid cells uses a limited number of germline-encoded
surface CD4 and intracellular GM-CSF. It is apparent that over- patternrecognition receptors to detect the relatively invariant
expression of IRF8 significantly decreases the percentage of GM- pathogen-associated molecular patterns (58). In contrast, the
CSFexpressing cells in both WT and IRF8 KO CD4+ T cells adaptive immune system, which is composed of T and B cells,
(Fig. 8A, 8B). Analysis of GM-CSF mRNA level indicates that employs Ag receptors that are not encoded in the germline but
overexpression of IRF8 significantly decreased GM-CSF mRNA are generated de novo. Although the adaptive immune system
level (Fig. 8C). depends on the generation of almost unlimited repertoire of Ag
2376 IRF8 REPRESSES GM-CSF TO CONTROL MDSC ACCUMULATION

FIGURE 7. T cellproduced GM-CSF


induces MDSC differentiation. (A) BALB/c
mice (H-2d) were irradiated with 9 Gy from
a 137Cs source and were injected i.v. with
2 3 106 T celldepleted BM cells alone, or
combined with 0.3 3 106 CD4+ T cells
isolated from C57BL/6 (H-2kb), C57BL/6-

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
IRF8 KO, C57BL/6-GM-CSF KO mice.
Mice were analyzed 14 d after cell transfer.
Spleens of the indicated chimera mice are
shown. Spleen cell numbers are presented in
the right panel. (B) The H-2kb+CD11b+ and
H-2kb+Gr1+ myeloid cells in the indicated
chimera mice were analyzed by flow cytometry
in spleens 11 d after donor cell transfer. Shown
are representative results from one mouse of
three mice. (C) Quantification of CD11b+ and
Gr1+ cells in the indicated mice as shown in
(B). Column, mean; bar, SD. **p , 0.01.

receptors on T and B cells and subsequent activation and clonal the pathogen-associated molecular patterns to deliver essential
expansion of T and B cells carrying the Ag-specific receptors, the danger signals to activate the adaptive immune system (29). Thus,
innate immune system is the first line of defense that recognizes the innate immune cells, primarily the myeloid cells, regulate the

FIGURE 8. Restoration of IRF8 expres-


sion decreases GM-CSF+ T cells. Naive
CD4+ T cells from C57BL/6 WT and
IRF82/2 mice were transduced with retro-
virus encoding IRF8 or empty vector, and
the cells were activated with anti-CD3 and
anti-CD28 Abs for 3 d. The cells were
restimulated with PMA/ionomycin for 5 h,
stained for intracellular GM-CSF, and ana-
lyzed by flow cytometry. Representative
FACS dot plots gated on CD4+ T cells
and the percentage of GM-CSFproducing
CD4+ T cells are shown. (B) Quantification
of CD4+GM-CSF+ cells in three indepen-
dent experiments, as shown in (A). Column,
mean; bar, SD. Overexpression of IRF8
significantly decreased percentage of GM-
CSF+ cells in both WT and IRF8 KO CD4+
T cells (**p , 0.01). (C) Total RNA was
prepared from cells, as shown in (A), and
analyzed for GM-CSF mRNA levels by re-
al-time RT-PCR using b-actin as internal
control. Overexpression of IRF8 signifi-
cantly decreased GM-CSF mRNA levels in
both WT and IRF8 KO CD4+ T cells
(**p , 0.01).
The Journal of Immunology 2377

represses GM-CSF expression in T cells. Analysis of the csf2


promoter region revealed two putative IFN-stimulated response
element sites (Supplemental Fig. 3A); however, our initial attempts
have failed to detect direct IRF8 binding to the csf2 gene promoter
(Supplemental Fig. 3B), suggesting that IRF8 may regulate GM-
CSF expression through an indirect mechanism. Further studies are
needed to determine the mechanisms underlying csf2 transcription
regulation by IRF8.
The lyz2 promoter used in generating the IRF8 MKO mice is
the most commonly used one for myeloid cellspecific gene ex-
pression in the literature (6670). The current expressed sequence
FIGURE 9. GM-CSF is not downregulated in MDSCs from IRF8 KO
mice. CD11b+Gr1+ MDSCs were purified from spleens of WT (n = 4) and
tag data indicate that the expression level of lysozyme 2 is highest
IRF8 KO (n = 3) mice. Total RNA was isolated from these purified MDSCs in BM as compared with other tissues in mice (http://www.ncbi.

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
and analyzed for IRF8 expression levels by real-time RT-PCR using nlm.nih.gov/UniGene/ESTProfileViewer.cgi?uglist=Mm.45436).
b-actin as the internal control. The IRF8 expression level in MDSCs from Therefore, it is unlikely that lyz2 promoter is not activated in
the first WT mice was arbitrarily set at 1.0. The expression levels of IRF8 BM. In the literature, it is well demonstrated that lyz2-driven Cre
in MDSCs of the rest samples were the levels relative to the IRF8 ex- expression induces specific and highly efficient deletion of loxp-
pression level in MDSCs of first WT mice. flanked gene segments in mature myeloid cells (6669). How-
ever, it was also observed that lyz2-driven Cre-mediated deletion
adaptive immune response under pathophysiological conditions of loxp-flanked gene segments is less efficient in immature
(4, 29, 5961). In this study, we show that T cells regulate myeloid myeloid cells as compared with that in mature amyloid cells
cell differentiation through an IRF8-mediated and GM-CSFde- (6668). Therefore, it is possible that IRF8 is only partially
pendent extrinsic mechanism in vivo. Our findings therefore deleted in myeloid progenitor cells. In contrast, IRF8 expression
revealed that the adaptive immune component of the immune
system (i.e., T cells) regulates the innate myeloid cell lineage
differentiation.
Decades ago, a seminal study with Irf82/2 mice revealed IRF8 as
an essential transcription factor in regulating myeloid cell lineage
differentiation (36). One of the prominent phenotypes of IRF8 null
mice is the massive accumulation of CD11b+Gr1+ cells in BM,
spleen, blood, and peripheral lymphoid organs as a consequence of
altered myelopoiesis (36). Interestingly, pathological conditions,
that is, cancer, also induce massive accumulation of CD11b+Gr1+
cells termed MDSCs (CD11b+Gr1+ in mice and CD11b+CD33+ in
humans) (6264). Furthermore, glioblastoma multiforme and pan-
creatic cancer cells have been shown to secrete GM-CSF to induce
tumor-associated macrophage and MDSC accumulation in the
tumor-bearing host (55, 57, 65). Moreover, IRF8 expression is often
silenced in CD11b+Gr1+ MDSCs in the tumor-bearing hosts (38,
49). These observations clearly demonstrate an inverse correlation
between IRF8 expression/function and CD11b+Gr1+ MDSC accu-
mulation under both physiological and pathological conditions.
These observations thus suggest that IRF8 functions in myeloid cells
mediate myeloid cell differentiation and loss of IRF8 expression or
function causes deregulation of myelopoiesis and resultant MDSC
accumulation. However, our present data strongly support a model
showing that myeloid IRF8 levels can also be controlled extrinsi-
cally to regulate myeloid cell differentiation and MDSC accumu-
lation in vivo. In this study, we showed that IRF8 expression in T
cells is critical for the production of GM-CSF which, in turn,
impacts IRF8 expression levels in the receptive myeloid populations
and the resultant myeloid phenotype. The relationship between
IRF8 expression and GM-CSF production in tumor cells warrants
further study, as well as detailing IRF8 expression levels in the
different myeloid progenitor populations of the IRF8 MKO model. FIGURE 10. Model of IRF8 deficiency and MDSC accumulation. Under
A previous study has convincingly demonstrated that mixed BM physiological conditions, IRF8 represses GM-CSF expression in T cells.
chimeras generated from a mixture of WT and IRF8 KO BM Loss of IRF8 expression and function leads to elevated GM-CSF expres-
display defects only in myeloid cells differentiated from the IRF8 sion and secretion. T cellproduced GM-CSF binds to its receptor on
myeloid progenitor cells to induce MDSC differentiation. In contrast, loss
KO BM, indicating a myeloid cellintrinsic abnormality in IRF8-
of IRF8 expression or function in myeloid cells in IRF8 null mice leads to
deficient myeloid progenitors (31). In this study, we observed decreased spontaneous apoptosis of myeloid cells to reduce MDSC turn-
IRF8 intrinsic function in myeloid cells in apoptosis regulation. over. Thus, loss of IRF8 expression and function in T cells results in in-
However, it seems that the extrinsic function of IRF8 as a regu- creased GM-CSF expression and resultant increased MDSC differentiation,
lator of GM-CSF expression plays a more dominant role in MDSC which acts in concert with decreased myeloid cell spontaneous apoptosis and
accumulation under our conditions. It is clear that IRF8 clearly turnover to lead to MDSC accumulation.
2378 IRF8 REPRESSES GM-CSF TO CONTROL MDSC ACCUMULATION

level in hematopoietic stem cells is negligible, but increases with 4. Gabrilovich, D. I., S. Ostrand-Rosenberg, and V. Bronte. 2012. Coordinated
regulation of myeloid cells by tumours. Nat. Rev. Immunol. 12: 253268.
differentiation of hematopoietic stem cells into immature myeloid 5. Metcalf, D. 1989. The molecular control of cell division, differentiation com-
cells. Differentiation of hematopoietic stem cells into the multi- mitment and maturation in haemopoietic cells. Nature 339: 2730.
potent progenitorcommon lymphoid progenitorgranulocyte- 6. Milanovic, M., G. Terszowski, D. Struck, O. Liesenfeld, and D. Carstanjen.
2008. IFN consensus sequence binding protein (Icsbp) is critical for eosinophil
myeloid progenitor pathway coincided with sequential increases development. J. Immunol. 181: 50455053.
in IRF8 expression (9). This IRF8 expression pattern correlates 7. Tsujimura, H., T. Tamura, and K. Ozato. 2003. Cutting edge: IFN consensus
at least partially with lyz2 promoter activity and lysozyme 2 ex- sequence binding protein/IFN regulatory factor 8 drives the development of type
I IFN-producing plasmacytoid dendritic cells. J. Immunol. 170: 11311135.
pression patterns during myeloid cell lineage differentiation. It is 8. Jaiswal, H., M. Kaushik, R. Sougrat, M. Gupta, A. Dey, R. Verma, K. Ozato, and
apparent that further studies are warranted to determine the lyz2 P. Tailor. 2013. Batf3 and Id2 have a synergistic effect on Irf8-directed classical
CD8a+ dendritic cell development. J. Immunol. 191: 59936001.
promoter activity and irf8 deletion during myeloid cell lineage 9. Wang, H., M. Yan, J. Sun, S. Jain, R. Yoshimi, S. M. Abolfath, K. Ozato,
differentiation in the IRF8 MKO mice. W. G. Coleman, Jr., A. P. Ng, D. Metcalf, et al. 2014. A reporter mouse reveals
Although IRF8 expressed in myeloid cells is not sufficient lineage-specific and heterogeneous expression of IRF8 during lymphoid and
myeloid cell differentiation. J. Immunol. 193: 17661777.
enough to intrinsically regulate myeloid cell differentiation, the 10. Kanno, Y., B. Z. Levi, T. Tamura, and K. Ozato. 2005. Immune cell-specific

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
fact that IRF8 is often silenced in MDSCs in tumor-bearing mice amplification of interferon signaling by the IRF-4/8-PU.1 complex. J. Interferon
and human cancer patients suggests that IRF8 expressed in myeloid Cytokine Res. 25: 770779.
11. Tenen, D. G., R. Hromas, J. D. Licht, and D. E. Zhang. 1997. Transcription
cells should play a role in MDSC accumulation (38, 49, 71). IRF8 factors, normal myeloid development, and leukemia. Blood 90: 489519.
is a transcription factor previously shown to regulate the expres- 12. Satpathy, A. T., C. G. Briseno, X. Cai, D. G. Michael, C. Chou, S. Hsiung,
sion of multiple apoptosis-regulatory genes, including Bcl-xL, D. Bhattacharya, N. A. Speck, and T. Egawa. 2014. Runx1 and Cbfb regulate the
development of Flt3+ dendritic cell progenitors and restrict myeloproliferative
Bax, FLIP, and Fas, in myeloid cells (4649). Furthermore, as in disorder. Blood 123: 29682977.
T cells, it has been shown that MDSC turnover is mediated by the 13. Tamura, T., T. Nagamura-Inoue, Z. Shmeltzer, T. Kuwata, and K. Ozato. 2000.
FasFas ligand apoptosis pathway of the hosts immune cells (45). ICSBP directs bipotential myeloid progenitor cells to differentiate into mature
macrophages. Immunity 13: 155165.
It is well known that deregulation of Fas-mediated apoptosis in 14. Novershtern, N., A. Subramanian, L. N. Lawton, R. H. Mak, W. N. Haining,
T cells leads to autoimmune lymphoproliferative syndrome with M. E. McConkey, N. Habib, N. Yosef, C. Y. Chang, T. Shay, et al. 2011. Densely
interconnected transcriptional circuits control cell states in human hematopoie-
massive T cell accumulation (72), which resembles massive sis. Cell 144: 296309.
MDSC accumulation in IRF8 null mice. Therefore, by parallel, it 15. Shivdasani, R. A., and S. H. Orkin. 1996. The transcriptional control of hema-
is reasonable to assume that IRF8 deficiency-induced deregulation topoiesis. Blood 87: 40254039.
16. Singh, H., R. P. DeKoter, and J. C. Walsh. 1999. PU.1, a shared transcriptional
of the Fas-mediated apoptosis pathway in MDSCs might decrease regulator of lymphoid and myeloid cell fates. Cold Spring Harb. Symp. Quant.
MDSC sensitivity to host immune cell-mediated apoptosis. In- Biol. 64: 1320.
deed, MDSCs from IRF8 KO mice exhibit decreased spontaneous 17. DeKoter, R. P., J. C. Walsh, and H. Singh. 1998. PU.1 regulates both cytokine-
dependent proliferation and differentiation of granulocyte/macrophage progen-
apoptosis. MDSCs from IRF8 MKO mice also exhibit decreased itors. EMBO J. 17: 44564468.
spontaneous apoptosis. Therefore, in addition to regulating MDSC 18. Iwasaki, H., and K. Akashi. 2007. Myeloid lineage commitment from the he-
differentiation in an extrinsic manner, IRF8 also mediates MDSC matopoietic stem cell. Immunity 26: 726740.
19. Kurotaki, D., N. Osato, A. Nishiyama, M. Yamamoto, T. Ban, H. Sato,
spontaneous apoptosis to control their turnover. Thus, increased J. Nakabayashi, M. Umehara, N. Miyake, N. Matsumoto, et al. 2013. Essential
differentiation induced by T cellproduced GM-CSF, acting in role of the IRF8-KLF4 transcription factor cascade in murine monocyte differ-
entiation. Blood 121: 18391849.
concert with decreased MDSC spontaneous apoptosis/turnover, 20. Feinberg, M. W., A. K. Wara, Z. Cao, M. A. Lebedeva, F. Rosenbauer,
leads to MDSC accumulation in IRF8-deficient host. H. Iwasaki, H. Hirai, J. P. Katz, R. L. Haspel, S. Gray, et al. 2007. The Kruppel-
The molecular mechanism underlying MDSC accumulation has like factor KLF4 is a critical regulator of monocyte differentiation. EMBO J. 26:
41384148.
been subject of extensive studies (38, 46, 73). In this study, we 21. Alder, J. K., R. W. Georgantas, III, R. L. Hildreth, I. M. Kaplan, S. Morisot,
observed that IRF8 functions extrinsically as a GM-CSF repressor X. Yu, M. McDevitt, and C. I. Civin. 2008. Kruppel-like factor 4 is essential for
in T cells. We show that IRF8-deficient T cells express elevated inflammatory monocyte differentiation in vivo. J. Immunol. 180: 56455652.
22. Hanna, R. N., L. M. Carlin, H. G. Hubbeling, D. Nackiewicz, A. M. Green,
level of GM-CSF. Based on our observations, we propose that J. A. Punt, F. Geissmann, and C. C. Hedrick. 2011. The transcription factor
IRF8 is a GM-CSF repressor. Loss of IRF8 function in T cells NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C-
results in increased GM-CSF expression and secretion. GM-CSF monocytes. Nat. Immunol. 12: 778785.
23. Kelly, L. M., U. Englmeier, I. Lafon, M. H. Sieweke, and T. Graf. 2000. MafB is
binds to its receptor on myeloid progenitor cells to increase an inducer of monocytic differentiation. EMBO J. 19: 19871997.
MDSC differentiation. At the same time, loss of IRF8 function in 24. Hegde, S. P., J. Zhao, R. A. Ashmun, and L. H. Shapiro. 1999. c-Maf induces
monocytic differentiation and apoptosis in bipotent myeloid progenitors. Blood
the differentiating myeloid cells decreases their spontaneous ap- 94: 15781589.
optosis and turnover. GM-CSFinduced increase in MDSC dif- 25. Nguyen, H. Q., B. Hoffman-Liebermann, and D. A. Liebermann. 1993. The zinc
ferentiation, in concert with IRF8 deficiencymediated decrease in finger transcription factor Egr-1 is essential for and restricts differentiation along
the macrophage lineage. Cell 72: 197209.
MDSC turnover, causes massive MDSC accumulation (Fig. 10). 26. Savitsky, D., T. Tamura, H. Yanai, and T. Taniguchi. 2010. Regulation of im-
munity and oncogenesis by the IRF transcription factor family. Cancer Immunol.
Immunother. 59: 489510.
Acknowledgments 27. Tamura, T., H. Yanai, D. Savitsky, and T. Taniguchi. 2008. The IRF family
transcription factors in immunity and oncogenesis. Annu. Rev. Immunol. 26:
We thank Dr. Jeanene Pihkala in the Medical College of Georgia Flow 535584.
Cytometry Core Facility for the excellent technical assistance in cell sorting. 28. Taniguchi, T., K. Ogasawara, A. Takaoka, and N. Tanaka. 2001. IRF family of
transcription factors as regulators of host defense. Annu. Rev. Immunol. 19: 623655.
29. Ikushima, H., H. Negishi, and T. Taniguchi. 2013. The IRF family transcription
Disclosures factors at the interface of innate and adaptive immune responses. Cold Spring
The authors have no financial conflicts of interest. Harb. Symp. Quant. Biol. 78: 105116.
30. Salem, S., D. Langlais, F. Lefebvre, G. Bourque, V. Bigley, M. Haniffa,
J.-L. Casanova, D. Burk, A. Berghuis, K. M. Butler, et al. 2014. Functional
characterization of the human dendritic cell immunodeficiency associated with the
References IRF8K108E mutation. Blood 124: 18941904.
1. Okabe, Y., and R. Medzhitov. 2014. Tissue-specific signals control reversible pro- 31. Becker, A. M., D. G. Michael, A. T. Satpathy, R. Sciammas, H. Singh, and
gram of localization and functional polarization of macrophages. Cell 157: 832844. D. Bhattacharya. 2012. IRF-8 extinguishes neutrophil production and promotes
2. Wynn, T. A., A. Chawla, and J. W. Pollard. 2013. Macrophage biology in de- dendritic cell lineage commitment in both myeloid and lymphoid mouse pro-
velopment, homeostasis and disease. Nature 496: 445455. genitors. Blood 119: 20032012.
3. Gabrilovich, D. I., and S. Nagaraj. 2009. Myeloid-derived suppressor cells as 32. Tamura, T., and K. Ozato. 2002. ICSBP/IRF-8: its regulatory roles in the de-
regulators of the immune system. Nat. Rev. Immunol. 9: 162174. velopment of myeloid cells. J. Interferon Cytokine Res. 22: 145152.
The Journal of Immunology 2379

33. Tamura, T., P. Thotakura, T. S. Tanaka, M. S. Ko, and K. Ozato. 2005. Identi- 53. Serafini, P., R. Carbley, K. A. Noonan, G. Tan, V. Bronte, and I. Borrello. 2004.
fication of target genes and a unique cis element regulated by IRF-8 in devel- High-dose granulocyte-macrophage colony-stimulating factor-producing vac-
oping macrophages. Blood 106: 19381947. cines impair the immune response through the recruitment of myeloid suppressor
34. Dror, N., M. Alter-Koltunoff, A. Azriel, N. Amariglio, J. Jacob-Hirsch, cells. Cancer Res. 64: 63376343.
S. Zeligson, A. Morgenstern, T. Tamura, H. Hauser, G. Rechavi, et al. 2007. 54. Waight, J. D., Q. Hu, A. Miller, S. Liu, and S. I. Abrams. 2011. Tumor-derived
Identification of IRF-8 and IRF-1 target genes in activated macrophages. Mol. G-CSF facilitates neoplastic growth through a granulocytic myeloid-derived
Immunol. 44: 338346. suppressor cell-dependent mechanism. PLoS One 6: e27690.
35. Hambleton, S., S. Salem, J. Bustamante, V. Bigley, S. Boisson-Dupuis, 55. Bayne, L. J., G. L. Beatty, N. Jhala, C. E. Clark, A. D. Rhim, B. Z. Stanger, and
J. Azevedo, A. Fortin, M. Haniffa, L. Ceron-Gutierrez, C. M. Bacon, et al. 2011. R. H. Vonderheide. 2012. Tumor-derived granulocyte-macrophage colony-
IRF8 mutations and human dendritic-cell immunodeficiency. N. Engl. J. Med. stimulating factor regulates myeloid inflammation and T cell immunity in pan-
365: 127138. creatic cancer. Cancer Cell 21: 822835.
36. Holtschke, T., J. Lohler, Y. Kanno, T. Fehr, N. Giese, F. Rosenbauer, J. Lou, 56. Marigo, I., E. Bosio, S. Solito, C. Mesa, A. Fernandez, L. Dolcetti, S. Ugel,
K. P. Knobeloch, L. Gabriele, J. F. Waring, et al. 1996. Immunodeficiency and N. Sonda, S. Bicciato, E. Falisi, et al. 2010. Tumor-induced tolerance and im-
chronic myelogenous leukemia-like syndrome in mice with a targeted mutation mune suppression depend on the C/EBPbeta transcription factor. Immunity 32:
of the ICSBP gene. Cell 87: 307317. 790802.
37. Tailor, P., T. Tamura, H. C. Morse, III, and K. Ozato. 2008. The BXH2 mutation 57. Pylayeva-Gupta, Y., K. E. Lee, C. H. Hajdu, G. Miller, and D. Bar-Sagi. 2012.
in IRF8 differentially impairs dendritic cell subset development in the mouse. Oncogenic Kras-induced GM-CSF production promotes the development of
Blood 111: 19421945. pancreatic neoplasia. Cancer Cell 21: 836847.

Downloaded from http://www.jimmunol.org/ at Georgia Regents University Greenblatt Library on June 5, 2015
38. Waight, J. D., C. Netherby, M. L. Hensen, A. Miller, Q. Hu, S. Liu, P. N. Bogner, 58. Iwasaki, A., and R. Medzhitov. 2010. Regulation of adaptive immunity by the
M. R. Farren, K. P. Lee, K. Liu, and S. I. Abrams. 2013. Myeloid-derived innate immune system. Science 327: 291295.
suppressor cell development is regulated by a STAT/IRF-8 axis. J. Clin. In- 59. Rutkowski, M. R., T. L. Stephen, and J. R. Conejo-Garcia. 2012. Anti-tumor
vest. 123: 44644478. immunity: myeloid leukocytes control the immune landscape. Cell. Immunol.
39. Ouyang, X., R. Zhang, J. Yang, Q. Li, L. Qin, C. Zhu, J. Liu, H. Ning, 278: 2126.
M. S. Shin, M. Gupta, et al. 2011. Transcription factor IRF8 directs a silencing 60. Schenten, D., and R. Medzhitov. 2011. The control of adaptive immune
programme for TH17 cell differentiation. Nat. Commun. 2: 314. responses by the innate immune system. Adv. Immunol. 109: 87124.
40. Feng, J., H. Wang, D. M. Shin, M. Masiuk, C. F. Qi, and H. C. Morse, III. 2011. 61. Wermeling, F., R. M. Anthony, F. Brombacher, and J. V. Ravetch. 2013. Acute
IFN regulatory factor 8 restricts the size of the marginal zone and follicular inflammation primes myeloid effector cells for anti-inflammatory STAT6 sig-
B cell pools. J. Immunol. 186: 14581466. naling. Proc. Natl. Acad. Sci. USA 110: 1348713491.
41. Yamamoto, M., T. Kato, C. Hotta, A. Nishiyama, D. Kurotaki, M. Yoshinari, 62. Sonda, N., F. Simonato, E. Peranzoni, B. Cal`, S. Bortoluzzi, A. Bisognin,
M. Takami, M. Ichino, M. Nakazawa, T. Matsuyama, et al. 2011. Shared and E. Wang, F. M. Marincola, L. Naldini, B. Gentner, et al. 2013. miR-142-3p
distinct functions of the transcription factors IRF4 and IRF8 in myeloid cell prevents macrophage differentiation during cancer-induced myelopoiesis. Im-
development. PLoS One 6: e25812. munity 38: 12361249.
42. Rosenbauer, F., and D. G. Tenen. 2007. Transcription factors in myeloid develop- 63. Ostrand-Rosenberg, S. 2013. Looking to the future of cancer immunotherapy:
ment: balancing differentiation with transformation. Nat. Rev. Immunol. 7: 105117. many questions to answer and many therapeutic opportunities. Cancer Immunol.
43. Zhao, J., H. J. Kong, H. Li, B. Huang, M. Yang, C. Zhu, M. Bogunovic, Immunother. 62: 12.
F. Zheng, L. Mayer, K. Ozato, et al. 2006. IRF-8/interferon (IFN) consensus 64. Youn, J. I., V. Kumar, M. Collazo, Y. Nefedova, T. Condamine, P. Cheng,
sequence-binding protein is involved in Toll-like receptor (TLR) signaling and A. Villagra, S. Antonia, J. C. McCaffrey, M. Fishman, et al. 2013. Epigenetic
contributes to the cross-talk between TLR and IFN-gamma signaling pathways. silencing of retinoblastoma gene regulates pathologic differentiation of myeloid
J. Biol. Chem. 281: 1007310080. cells in cancer. Nat. Immunol. 14: 211220.
44. Zhu, C., K. Rao, H. Xiong, K. Gagnidze, F. Li, C. Horvath, and S. Plevy. 2003. 65. Pyonteck, S. M., L. Akkari, A. J. Schuhmacher, R. L. Bowman, L. Sevenich,
Activation of the murine interleukin-12 p40 promoter by functional interactions D. F. Quail, O. C. Olson, M. L. Quick, J. T. Huse, V. Teijeiro, et al. 2013. CSF-
between NFAT and ICSBP. J. Biol. Chem. 278: 3937239382. 1R inhibition alters macrophage polarization and blocks glioma progression.
45. Sinha, P., O. Chornoguz, V. K. Clements, K. A. Artemenko, R. A. Zubarev, and Nat. Med. 19: 12641272.
S. Ostrand-Rosenberg. 2011. Myeloid-derived suppressor cells express the death 66. Clemens, R. A., L. E. Lenox, T. Kambayashi, N. Bezman, J. S. Maltzman,
receptor Fas and apoptose in response to T cell-expressed FasL. Blood 117: K. E. Nichols, and G. A. Koretzky. 2007. Loss of SLP-76 expression within
53815390. myeloid cells confers resistance to neutrophil-mediated tissue damage while
46. Gabriele, L., J. Phung, J. Fukumoto, D. Segal, I. M. Wang, P. Giannakakou, maintaining effective bacterial killing. J. Immunol. 178: 46064614.
N. A. Giese, K. Ozato, and H. C. Morse, III. 1999. Regulation of apoptosis in 67. Clausen, B. E., C. Burkhardt, W. Reith, R. Renkawitz, and I. Forster. 1999.
myeloid cells by interferon consensus sequence-binding protein. J. Exp. Med. Conditional gene targeting in macrophages and granulocytes using LysMcre
190: 411421. mice. Transgenic Res. 8: 265277.
47. Yang, J., X. Hu, M. Zimmerman, C. M. Torres, D. Yang, S. B. Smith, and K. Liu. 68. Sissons, J. R., J. J. Peschon, F. Schmitz, R. Suen, M. Gilchrist, and A. Aderem.
2011. Cutting edge: IRF8 regulates Bax transcription in vivo in primary myeloid 2012. Cutting edge: microRNA regulation of macrophage fusion into multinu-
cells. J. Immunol. 187: 44264430. cleated giant cells. J. Immunol. 189: 2327.
48. Yang, D., S. Wang, C. Brooks, Z. Dong, P. V. Schoenlein, V. Kumar, X. Ouyang, 69. Lalani, A. I., C. R. Moore, C. Luo, B. Z. Kreider, Y. Liu, H. C. Morse, III, and
H. Xiong, G. Lahat, A. Hayes-Jordan, et al. 2009. IFN regulatory factor 8 P. Xie. 2015. Myeloid cell TRAF3 regulates immune responses and inhibits
sensitizes soft tissue sarcoma cells to death receptor-initiated apoptosis via re- inflammation and tumor development in mice. J. Immunol. 194: 334348.
pression of FLICE-like protein expression. Cancer Res. 69: 10801088. 70. Gautier, E. L., A. Chow, R. Spanbroek, G. Marcelin, M. Greter, C. Jakubzick,
49. Hu, X., K. Bardhan, A. V. Paschall, D. Yang, J. L. Waller, M. A. Park, A. Nayak- M. Bogunovic, M. Leboeuf, N. van Rooijen, A. J. Habenicht, et al. 2012. Sys-
Kapoor, T. A. Samuel, S. I. Abrams, and K. Liu. 2013. Deregulation of apoptotic temic analysis of PPARg in mouse macrophage populations reveals marked
factors Bcl-xL and Bax confers apoptotic resistance to myeloid-derived sup- diversity in expression with critical roles in resolution of inflammation and
pressor cells and contributes to their persistence in cancer. J. Biol. Chem. 288: airway immunity. J. Immunol. 189: 26142624.
1910319115. 71. Stewart, T. J., K. M. Greeneltch, J. E. Reid, D. J. Liewehr, S. M. Steinberg,
50. Jiao, J., A. C. Dragomir, P. Kocabayoglu, A. H. Rahman, A. Chow, K. Liu, and S. I. Abrams. 2009. Interferon regulatory factor-8 modulates the
D. Hashimoto, M. Leboeuf, T. Kraus, T. Moran, G. Carrasco-Avino, et al. 2014. development of tumour-induced CD11b+Gr-1+ myeloid cells. J. Cell. Mol. Med.
Central role of conventional dendritic cells in regulation of bone marrow release 13: 39393950.
and survival of neutrophils. J. Immunol. 192: 33743382. 72. Siegel, R. M., F. K. Chan, H. J. Chun, and M. J. Lenardo. 2000. The multifaceted
51. Pathak, S., S. Ma, V. Shukla, and R. Lu. 2013. A role for IRF8 in B cell anergy. role of Fas signaling in immune cell homeostasis and autoimmunity. Nat.
J. Immunol. 191: 62226230. Immunol. 1: 469474.
52. Bunt, S. K., L. Yang, P. Sinha, V. K. Clements, J. Leips, and S. Ostrand- 73. Hu, X., D. Yang, M. Zimmerman, F. Liu, J. Yang, S. Kannan, A. Burchert,
Rosenberg. 2007. Reduced inflammation in the tumor microenvironment delays Z. Szulc, A. Bielawska, K. Ozato, et al. 2011. IRF8 regulates acid ceramidase
the accumulation of myeloid-derived suppressor cells and limits tumor pro- expression to mediate apoptosis and suppresses myelogeneous leukemia. Cancer
gression. Cancer Res. 67: 1001910026. Res. 71: 28822891.
Supplemental Figure 1. Mice with a null mutation of irf8 exhibit accumulation of
CD11b+Gr1+ MDSCs. A. Spleen morphology of wt and IRF8 KO mice (3 months of age).
Shown are representative images from one pair of wt and IRF8 KO mice of three pairs of mice.
B. CD11b+Gr1+ MDSC profiles in thymus, spleen, lymph nodes and bone marrow. Cells were
collected from the indicated tissues, and stained with CD11b- and Gr1-specific mAbs. Isotype
control mAbs were used as negative controls. Shown are representative results from one pair of
mice of three pairs of wt and IRF8 KO mice.
Supplemental Figure 2. Genotypes of wt and IRF8 MKO mice. PCR banding patterns of mice
with the indicated genotypes. PCR primer sequences and genotyping protocols are according to
the Jackson Laboratory.
Supplemental Figure 3. A. The mouse GM-CSF gene (csf2) promoter structure showing the
region of -4000 to +1000 relative to the transcription initiation site (black bar). The putative
ISRE consensus sequences and locations are indicated at the top of the bar. The PCR-amplified
genomic DNA regions for each of the five ChIP PCR primer pairs are indicated (short blue bars).
B. CD4+ T cells were isolated by negative selection. The cells were stimulated with anti-CD3
and anti-CD28 mAbs for 3 days and analyzed by chromatin immunoprecipitation (ChIP) using
goat IgG or anti-IRF8 antibody (C-19, Santa Cruz). The immunoprecipitated genomic DNA was
analyzed by real-time PCR using the mouse genomic DNA (gDNA, 2ng) as a positive control.
Shown are representative results from one of four independent experiments.
Supplemental Table 1: PCR Primer Sequences
Gene Use Sequence
IP-1a RT-PCR Forward 5'-GCTGTTTGCTGCCAAGTAGCC-3'
Reverse 5'-TGACCAACTGGGAGGGAGATGG-3'
IP10 RT-PCR Forward 5'-TCTCTCCATCACTCCCCTTTACC-3'
Reverse 5'-CTTGCTTCGGCAGTTACTTTTGTC-3'
iNOS RT-PCR Forward 5'-TTGGGGAGACAGCGAAATGC-3'
Reverse 5'-GGAGTGGAGAAGAAGGGAGGAAAG-3'
IL12p40 RT-PCR Forward 5'-GAGACCCTGCCCATTGAACTG-3'
Reverse 5'-GGAACGCACCTTTCTGGTTACAC-3'
IRF8 RT-PCR Forward 5'-CGTGGAAGACGAGGTTACGCTG-3'
Reverse 5'-GCTGAATGGTGTGTGTCATAGGC-3'
IRF8mt RT-PCR Forward 5'-GCGCGGGCAGCGTGGGAACCGGCG-3'
Reverse 5'-GTCACTTCTTCAAAATCTGGGCTC-3'
GM-CSF ChIP1 PCR Forward 5'-CTTCTCCCCTCATTTCTCCTTTG-3'
Reverse 5'-TGCCCCAAGCCTTATCTCCAAC-3'
GM-CSF ChIP2 PCR Forward 5'-GCTCTTCTGCCAGGTTAGGACTTC-3
Reverse 5'-GATGGATGCTGTATCTGTTGTTGG-3
GM-CSF ChIP3 PCR Forward 5'-CCCTCACTTCTGTCTGGTTTCATC-3'
Reverse 5'-CACTTGTTTTGCCTGCTTTGTG-3'
GM-CSF ChIP4 PCR Forward 5'-TGTGTGCGTGCCTGGTTATTG-3'
Reverse 5'-TTGACTGCTGATTCTGCCCTCC-3
GM-CSF ChIP5 PCR Forward 5'-GCTTTTGAAATAGTGCTTCCCCAC-3
Reverse 5'-TTCCCAGTTCCAAGTGCTGTCC-3
GM-CSF RT-PCR Forward 5'- AAACACAAGTTACCACCTATGCGG -3
Reverse 5'- TCCAAGTTCCTGGCTCATTACG -3
b-Actin RT-PCR Forward 5'-CTGGCACCACACCTTCTACAATG-3'
Reverse 5'-GGGTCATCTTTTCACGGTTGG-3'

You might also like