Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Transcriptional Regulation of IL-15

Expression during Hematopoiesis


Sara L. Colpitts, Spencer W. Stonier, Thomas A. Stoklasek,
Sierra H. Root, Hector Leonardo Aguila, Kimberly S.
This information is current as Schluns and Leo Lefrançois
of September 20, 2017.
J Immunol 2013; 191:3017-3024; Prepublished online 21
August 2013;
doi: 10.4049/jimmunol.1301389
http://www.jimmunol.org/content/191/6/3017

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


Supplementary http://www.jimmunol.org/content/suppl/2013/08/21/jimmunol.130138
Material 9.DC1
References This article cites 59 articles, 39 of which you can access for free at:
http://www.jimmunol.org/content/191/6/3017.full#ref-list-1
Subscription Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2013 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Transcriptional Regulation of IL-15 Expression during


Hematopoiesis

Sara L. Colpitts,* Spencer W. Stonier,† Thomas A. Stoklasek,* Sierra H. Root,*


Hector Leonardo Aguila,* Kimberly S. Schluns,† and Leo Lefrançois*,1
Dendritic cells (DCs) are the most commonly studied source of the cytokine IL-15. Using an IL-15 reporter transgenic mouse, we
have recently shown previously unappreciated differences in the levels of IL-15 expressed by subsets of conventional DCs (CD8+ and
CD82). In this study, we show that IL-15 promoter activity was differentially regulated in subsets of hematopoietically derived
cells with IL-15 expression largely limited to myeloid lineages. In contrast, mature cells of the lymphoid lineages expressed little to
no IL-15 activity. Surprisingly, we discovered that hematopoietic stem cells (lineage2Sca-1+c-Kit+) expressed high levels of IL-15,
suggesting that IL-15 expression was extinguished during lymphoid development. In the case of T cells, this downregulation was
Notch-dependent and occurred in a stepwise pattern coincident with increasing maturation and commitment to a T cell fate.

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


Finally, we further demonstrate that IL-15 expression was also controlled throughout DC development, with key regulatory
activity of IL-15 production occurring at the pre-DC branch point, leading to the generation of both IL-15+CD8+ and IL-152/low
CD82 DC subsets. Thus, IL-15 expression is coordinated with cellular fate in myeloid versus lymphoid immune cells. The
Journal of Immunology, 2013, 191: 3017–3024.

nterleukin-15 is a common g chain (CD132) cytokine that

I
primary mechanism of IL-15 delivery. Whether cis-presentation of
functions through a unique mechanism of delivery termed trans- IL-15 occurs in vivo is currently unclear but remains feasible (9).
presentation. IL-15 has a very high affinity (1.4 3 10211 M) First, CD8 T cells are known to upregulate IL-15Ra expression
for its private receptor, IL-15Ra (1). The two proteins bind within following activation (10), and second, impaired development and
the cell and are shuttled to the surface as a complex (2). IL-15 is activation of IL-15–dependent cells is observed in transgenic mice
then presented in trans to responding cells, such as NK cells, CD8 engineered to express a chimeric cytokine receptor composed of
memory T cells, and intraepithelial lymphocytes (IELs), that ex- the external domain of IL-15Ra fused to the intracellular portion
press the other two shared chains of the receptor, CD132 and IL-2/ of IL-2Ra, suggesting that cell-intrinsic signaling through IL-
15Rb (CD122) (3). The generation of both IL-15 and IL-15Ra 15Ra is required for the generation of optimal immune responses
knockout (KO) mice has greatly facilitated our ability to study the (11).
dramatic downstream effects that exist in the absence of IL-15 Many elegant studies have addressed the cell–cell interactions
signaling (4, 5). Importantly, transferring NK cells or CD8 that drive IL-15–dependent responses, including the cellular
T cells from IL-15Ra KO mice to wild-type (WT) recipients sources of IL-15 in vivo (reviewed in Ref. 12). Early studies used
demonstrated that IL-15–dependent cells need not express the IL- the generation of bone marrow (BM) chimeras to distinguish IL-
15Ra chain to mediate the downstream effects of IL-15 signaling 15 derived from hematopoietic (radio-sensitive) versus non-
(6–8). For this reason, trans-presentation is largely accepted as the hematopoietic (radio-resistant) sources (6, 8, 13). In total, these
studies showed that individual populations of IL-15–dependent
cells are varied in their requirements for IL-15 from distinct
*Department of Immunology, University of Connecticut Health Center, Farmington, sources. For example, peripheral NK cells and memory CD8
CT 06030; and †Department of Immunology, University of Texas MD Anderson T cells are partially restored when IL-15 production is limited to
Cancer Center, Houston, TX 77054
1
hematopoietically derived cells whereas IL-15 trans-presentation
Dr. Lefrançois passed away during preparation of this article for print.
by radio-resistant intestinal epithelial cells is necessary and suf-
Received for publication May 24, 2013. Accepted for publication July 4, 2013.
ficient for the generation and maintenance of CD8aa+ IELs (14).
S.L.C. is supported by a postdoctoral fellowship from the American Cancer Society Consistent with their association intracellularly, these studies have
(Grant PF-11-152-01-LIB), and S.W.S. was supported by National Institutes of
Health Predoctoral Training Grant in Cancer Immunology CA009598. This work also shown that the same cell must express both IL-15 and the IL-
was also supported by National Institutes of Health Grants 1RC1HL100569 (National 15Ra to mediate activity (15, 16). Recent approaches have used
Heart, Lung and Blood Institute) (to H.L.A.), AI 070910 (to K.S.S.), and R01
AI76457 and U01 AI 095544 (to L.L.).
transgenic expression of IL-15Ra limited to certain subsets of
cells and conditional deletion of IL-15Ra using Cre-lox technol-
Address correspondence and reprint requests to Department of Immunology, c/o
Dr. Lynn Puddington, University of Connecticut Health Center, 263 Farmington Avenue, ogy to more closely examine the significance of IL-15 from se-
Room L3092, Farmington, CT 06030. E-mail address: Leolefrancois2013@gmail.com lected in vivo sources (17–21). These studies have implicated
The online version of this article contains supplemental material. a role for dendritic cells (DCs) in trans-presenting IL-15 to many
Abbreviations used in this article: BM, bone marrow; BMDC, bone marrow–derived subsets of immune cells, yet they also suggest that other cellular
dendritic cell; cDC, conventional dendritic cell; DC, dendritic cell; DL1, Delta-like 1; sources of IL-15 likely work in conjunction with DCs to maintain
DN, double-negative; EmGFP, emerald GFP; ETP, early thymic progenitor; Flt3L,
Flt3 ligand; GITR, glucocorticoid-induced TNFR; HSC, hematopoietic stem cell; overall peripheral homeostasis.
IEL, intraepithelial lymphocyte; KO, knockout; Lin, lineage; LSK, lineage2Sca-1+ The overt lymphoproliferative disease observed when IL-15 is
c-Kit+; MFI, mean fluorescence intensity; pDC, plasmacytoid DC; Tg+ or Tg2 , produced in excess is evidence that IL-15 expression must be
transgene positive or negative; WT, wild-type.
tightly regulated (22). However, IL-15 itself has proven much
Copyright Ó 2013 by The American Association of Immunologists, Inc. 0022-1767/13/$16.00 more difficult to study. This is likely the combined result of overall

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1301389
3018 DEVELOPMENTAL REGULATION OF IL-15

low levels of IL-15 in the steady-state and possibly poor detection Results
reagents, which in some cases may be further hindered when IL-15 IL-15 is expressed at the highest levels in myeloid lineages
is bound to IL-15Ra. To overcome these hurdles in IL-15 detection,
To facilitate the detection of IL-15 in vivo, our laboratory has
we generated a bacterial artificial chromosome transgenic mouse
recently generated a transgenic bacterial artificial chromosome
strain in which emerald GFP (EmGFP) is produced under control of
reporter mouse in which EmGFP is expressed under the control of
the IL-15 promoter and all upstream regulatory elements (EmGFP/
the promoter and upstream regulatory elements of the IL-15 gene
IL-15). We employed this tool to identify previously unappreciated
locus (23). Our analysis of these mice confirmed previously
differences in the respective levels of IL-15 produced by individual
published studies that show IL-15 production by DCs and mono-
subsets of conventional DCs (cDCs) during homeostasis and suc-
cytes (26, 27). However, the ability to use flow cytometry and
cessfully measure changes in IL-15 expression in DCs following
EmGFP expression as a surrogate for IL-15 promoter activity has
virus infection (23). In this study, we use EmGFP/IL-15 reporter
revealed previously unappreciated differences in the relative levels
mice to further identify novel subsets of cells capable of producing
of EmGFP/IL-15 expression in individual subsets of cells, par-
IL-15 and examine developmental pathways that regulate IL-15
ticularly CD8+ and CD82 DCs (Ref. 23 and Fig. 1A), and these
expression in immune cells, which largely result in limiting IL-
results were recently corroborated using a second transgenic IL-15
15 production to cells of the myeloid lineage.
reporter line (28). To further identify potentially novel cellular
sources of IL-15, we performed a large-scale analysis of both
Materials and Methods innate and adaptive immune cell subsets in transgenic mice (Tg+)
Mice and transgene-negative (Tg2) littermate controls using the mean

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


C57BL/6 mice were purchased from The Jackson Laboratory (Bar Harbor, fluorescence intensity (MFI) of EmGFP as an indicator of IL-15
ME) and the National Cancer Institute–Charles River (Fredericksburg, transcription (Fig. 1A). The phenotypic definitions used to identify
MD), and IL-152/2 mice were purchased from Taconic (Germantown, each population of immune cells by flow cytometry are shown in
NY). EmGFP/IL-15 reporter transgenics were bred and maintained at the
University of Connecticut Health Center. All mice were used between 6 Supplemental Table I. We found that EmGFP/IL-15 was highly
and 20 wk of age and housed according to the guidelines at the University expressed in splenic myeloid cells, including neutrophils, baso-
of Connecticut Health Center and MD Anderson Cancer Center. All phils, and eosinophils, and mast cells isolated from the peritoneal
experiments were performed with the approval of the respective Institu- cavity (Fig. 1A, 1B). Alternatively, we found a lack of EmGFP/IL-
tional Animal Care and Use Committees.
15 expression in lymphoid lineages, particularly CD4+ T cells and
Tissue isolation follicular B cells (Fig. 1C, 1D). Interestingly, NK and NKT cells
Spleen and thymus were harvested in complete media and crushed through along with marginal zone B cells, which are described as more
a 70-mm filter. BM was isolated by rapid centrifugation directly into “innate-like” subsets of the immune system, maintained low but
complete media. Peritoneal exudate cells were collected by injecting ∼10 detectable levels of EmGFP/IL-15 expression (Fig. 1D, 1E). How-
ml PBS into the peritoneal cavity and collecting the solution after 5 min. ever, unlike their cDC counterparts, plasmacytoid DCs (pDCs)
RBC lysis was performed by incubating single-cell suspensions in Tris
ammonium chloride for 5 min at 37˚C. expressed EmGFP/IL-15 at very low levels (Fig. 1F). Thus, we
observed an apparent bifurcation regarding IL-15 promoter ac-
Flow cytometry and cell sorting tivity that was largely associated with myeloid versus lymphoid
All Abs and Live/Dead viability dye were purchased from BD Biosciences, cell fate.
BioLegend, and eBioscience. Fc block (anti-CD16/32; clone 2.4G2) was Initial attempts to measure cell-surface IL-15 expression using
purchased from Bio X Cell (West Lebanon, NH). IL-15 was detected with a flow cytometry–based detection method have been largely un-
rabbit anti–IL-15 biotin (PeproTech, Rocky Hill, NJ) followed by
streptavidin-allophycocyanin (Jackson ImmunoResearch Labratories, West
successful. However, two reports have detected IL-15 protein in
Grove, PA). Background staining was determined by examining analogous BMDCs and Gr-1+ polymorphonuclear cells (29, 30), and using
populations from IL-152/2 mice. the same reagents we have also detected surface IL-15 on subsets
of DCs (17). Given the success of this particular Ab in detecting
In vitro culture
trans-presented IL-15, we sought to measure IL-15 protein di-
For IL-15 detection, 107 whole splenocytes or BM cells were cultured rectly ex vivo on the populations of granulocytes that exhibited
overnight with or without LPS (1 mg/ml; Sigma-Aldrich, St. Louis, MO) or
high levels of IL-15 transcription based on EmGFP expression.
IFN-a (300 U/ml; PBL Interferon Source and provided by Willem Over-
wijk [MD Anderson]). For in vitro culture with OP9 stromal cells, ex- Unfortunately, we were unable to detect IL-15 expression directly
periments were performed as previously described (24). Briefly, ∼4–5 3 ex vivo on any of the cell populations identified in Fig. 1 isolated
104 sorted lineage (Lin)2Sca-1+c-Kit+ (LSK) cells (CD32CD192CD11b2 from the spleen or BM (Supplemental Fig. 1A). However, we were
CD11c2Gr-12NK1.12Ter1192Sca-1+CD117+) were seeded on confluent also unable to detect IL-15 directly ex vivo on CD11c+ spleno-
OP9 or OP9-Delta-like 1 (DL1) stromal cells in OP9 media (MEMa, 20%
FBS, penicillin/streptomycin) supplemented with 5 ng/ml Flt3 ligand cytes (Supplemental Fig. 1B). In our previous studies, IL-15 was
(Flt3L) and 1 ng/ml IL-7 (PeproTech). Cells were replenished with new only detectable when the cells were subjected to an overnight
media and cytokines on day 5. To generate BM-derived DCs (BMDCs), in vitro culture or exposed to TLR ligands (17). Therefore, we
8 3 106 whole BM cells were plated at 2 3 106 cells/ml in six-well plates decided to take a similar approach to detect IL-15 protein on
with media (IMDM, 10% FBS, 2-ME, nonessential amino acids, sodium
pyruvate, penicillin/streptomycin) supplemented with 100 ng/ml Flt3L granulocytic cells. When we cultured splenocytes overnight in
(Celldex Therapeutics, Needham, MA) for 9 d. media alone, we detected low levels of IL-15 on neutrophils and
eosinophils, using CD11chigh DCs and IL-152/2 mice as a control
Adoptive transfer for IL-15 staining (Fig. 2A). Interestingly, when we performed
Approximately 1–5 3 104 purified pre-DCs (CD32CD192B2202Ter1192 similar overnight cultures with BM cells, we were also able to
NK1.12CD11c+MHC II2CD135+CD172a2) isolated from the BM were detect surface IL-15 on neutrophils, eosinophils, and macrophages
transferred to congenic recipients, and the spleens of recipient mice were
harvested after 6 d, as previously described (25).
(Fig. 2B). To further explore the potential of these novel sources
of IL-15 to produce protein, we stimulated cells from the spleen
Statistics and BM with LPS or IFN-a to determine whether these known
Statistical analysis was performed using Prism software (GraphPad Soft- inducers of IL-15 could augment IL-15 expression in the above
ware, San Diego, CA) using a Student t test or ANOVA where appropriate. populations. Surprisingly, all subsets were able to upregulate IL-
The Journal of Immunology 3019

Because T cells largely lacked EmGFP/IL-15 expression, and the


pathways that drive T cell commitment are well known, we ex-
amined the regulation of IL-15 during T cell development in the
thymus. We harvested thymocytes from Tg+ reporters and litter-
mate controls and determined the MFI of EmGFP/IL-15 expres-
sion in the CD32CD42CD82 double-negative (DN), CD32CD4+
CD8+ double-positive, and CD3+ single-positive CD4+ and CD8+
subsets. Similar to the LSK cells, early thymic progenitors (ETPs),
the c-Kit+ fraction of the CD44highCD252 DN1 subset (31),
expressed high levels of EmGFP/IL-15 (Fig. 3B, 3C). Such ele-
vated levels were maintained within the c-Kit+ fraction of CD25-
expressing DN2 cells as well. The downregulation of EmGFP/IL-
15 began as DN2 cells lost c-Kit expression (Fig. 3C), and
EmGFP/IL-15 expression was significantly reduced at the transi-
tion between DN2 and DN3 cells (Fig. 3B, 3C). By the DN4 stage,
EmGFP/IL-15 expression was completely lost in committed
T cells, and it remained absent from all downstream subsets in-
cluding double-positive and single-positive cells (Fig. 3B, 3C).
Thus, these data suggested that the signaling pathways controlling

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


the progression of DN2 to DN3 cells could be responsible for
initiating the downregulation of IL-15.
Notch signaling mediates IL-15 downregulation during
thymopoiesis
The role of Notch in driving T cell development has been well
studied (32), and it is thought that Notch signaling not only ini-
tiates the necessary program for T cell development but also turns
off any residual expression of myeloid-specific factors (33). To
test whether Notch signaling was sufficient to drive IL-15 down-
regulation, we cultured purified LSK cells in vitro with OP9
stromal cells expressing the Notch ligand DL1. We sorted
EmGFP+ LSK cells from Tg+ mice and EmGFP2 LSKs from Tg2
littermate controls and cultured them in the presence of Flt3L and
IL-7 for 9 d (24). At that time, a large population of CD25+ DN2
FIGURE 1. IL-15 is broadly expressed in myeloid cells. (A) Individual
and DN3 cells could easily be identified. Similar to our in vivo
subsets of immune cells were identified in the spleens of EmGFP reporter
analysis, we found that the DN2 to DN3 transition driven by
Tg+ and Tg2 littermate controls. The gating strategy for each subset is
described in Supplemental Table I. Data are presented graphically as the Notch signaling included a significant reduction in the MFI of
MFI of EmGFP/IL-15 in each subset of immune cells (mean 6 SEM). EmGFP/IL-15 (Fig. 4A). Interestingly, CD25+ DN2 cells grown
Representative histograms depicting relative levels of EmGFP/IL-15 ex- on OP9-DL1 cells had significantly reduced EmGFP/IL-15 ex-
pression in each population are shown in (B)–(F). Data are representative pression compared with the small number of CD25+ DN2 cells
of at least three experiments. Gray histograms show Tg2, black line that emerge when LSKs are cultured on control OP9 stromal cells.
indicates Tg+. Thus, in OP9-DL1 cultures, the abundance of Notch ligands may
have initiated some degree of EmGFP/IL-15 downregulation in
15 expression to some degree compared with both unstimulated DN2 cells that preceded the complete silencing of IL-15 expression
controls and stimulated IL-15 KO cells (Fig. 2). Thus, in vitro and additional phenotypic changes associated with the DN3 stage.
detection of IL-15 in neutrophils and eosinophils correlated with However, the presence of Notch ligands did not universally down-
the findings from our reporter mice indicating that these granu- regulate EmGFP/IL-15 expression because the small population of
locytic populations have the capacity to produce IL-15 protein. Gr-1+ cells that escaped Notch-driven differentiation signals and
developed on the OP9-DL1 stromal cells expressed identical levels
Hematopoietic progenitors express high levels of IL-15 of EmGFP/IL-15 as Gr-1+ cells that emerged after coculture with
All immune cells are derived from a small population of hemato- control OP9 cells lacking DL1 expression (Fig. 4B). This finding
poietic stem cells (HSCs) maintained in the adult BM. Because we suggested that even in the presence of Notch signals, acquisition of
observed such dramatic variation in the levels of EmGFP/IL-15 a myeloid fate includes the maintained expression of IL-15.
expressed by mature cells of the hematopoietic system, we asked
to what degree EmGFP/IL-15 was expressed by hematopoietic pro- Coordinated regulation of IL-15 throughout DC development
genitors in the BM of transgenic reporter mice. We hypothesized that Thus far, our results provided new evidence that IL-15 expression
HSCs would lack IL-15 expression and that IL-15 would be up- was controlled as immune cells progress toward a mature lymphoid
regulated in myeloid cells, thus rendering them capable of trans- fate. Considering the unique differences in EmGFP/IL-15 pro-
presenting IL-15 and delivering a survival signal to responding NK duction between CD8+ and CD82 DCs, we hypothesized that IL-15
and T cells. Surprisingly, LSK cells, which constitute a heteroge- would also be regulated throughout DC development, rather than
neous population of both long-term and short-term HSCs, expressed being controlled exclusively by extrinsic inflammatory mediators.
uniformly high levels of EmGFP/IL-15 (Fig. 3A). Thus, contrary to Therefore, we tracked EmGFP/IL-15 expression in BM precur-
our hypothesis, this result suggested that alternative mechanisms sor cells downstream of LSKs but upstream of DC development.
selectively limited IL-15 expression in lymphoid lineages. Both macrophage/DC progenitors and common DC progenitors
3020 DEVELOPMENTAL REGULATION OF IL-15

FIGURE 2. Detection of IL-15 pro-


tein from granulocytic myeloid cells.
(A and B) Cells (107) from the spleen
(A) or BM (B) of WT (black lines) and
IL-152/2 mice (gray filled histograms)
were incubated overnight in the ab-
sence or presence of 1 mg/ml LPS or
300 U/ml IFN-a. Numbers indicate the
MFI of IL-15 staining for WT (top)
and IL-152/2 samples (bottom). Plots
are representative of two to three ex-
periments.

expressed high levels of EmGFP/IL-15, albeit at significantly re- to determine whether EmGFP/IL-15+ pre-DCs were progenitors of
duced levels compared with the LSK cells (Fig. 5A, 5B). Thus, both EmGFP/IL-15+ CD8+ DCs and EmGFP/IL-152/low CD82
there was a progressive downregulation of EmGFP/IL-15 through DCs, sorted pre-DCs from the BM of Tg+ mice and Tg2 controls
each stage of DC development suggesting continuous regulatory were transferred to naive congenic recipients. After 6 d, EmGFP/
activity at the IL-15 locus. Pre-DCs, the final precursor population IL-15 expression by donor-derived DCs was measured. As pre-
prior to commitment to either the CD8+ or CD82 DC lineage (25), viously shown (25), pre-DCs gave rise to both DC subsets with

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


expressed low but uniform levels of EmGFP/IL-15 (Fig. 5C, 5D). preferential generation of CD82 DCs. Nevertheless, we detected
Because we observed homogeneous expression of IL-15 in the two populations of DCs (CD8+ and CD82) expressing signifi-
pre-DC precursor population, our results suggested that the overall cantly different levels of EmGFP/IL-15 (Fig. 5E, 5F) derived from
level of IL-15 expression by a given DC subset was linked to the a population of pre-DCs expressing uniform levels of EmGFP/IL-
final developmental program for that particular subset. Therefore, 15 (Fig. 5C). Thus, although little is known with regard to the
precise transcriptional elements that regulate the development of
CD8+ versus CD82 DCs, these results indicated that regulation at
the IL-15 locus occurred during this cell fate decision.
Just as the OP9-DL1 system can be used to mimic T cell de-
velopment in vitro, culturing BM cells in vitro with Flt3L can be
used to generate a diverse mixture of cDCs and pDCs. Importantly,
unlike BMDCs generated by culture with GM-CSF, the CD103+
and SIRPa+ (CD172a+) subsets of cDCs produced in Flt3L-
containing cultures more accurately reflect the CD8+ and CD82
subsets observed in vivo (34). We hypothesized that if the level of
IL-15 expressed by individual subsets of DCs was linked to their
cellular fate, the Flt3L-derived BMDCs should exhibit a similar
pattern of EmGFP/IL-15 expression between CD103+ and SIRPa+
DCs as did CD8+ and CD82 DCs, respectively. Indeed, the in vitro
generation of BMDCs with Flt3L exactly mimicked the pattern of
IL-15 expression by DC subsets in vivo (Fig. 5G). pDCs lacked
expression of EmGFP/IL-15, and CD103+ DCs expressed signif-
icantly higher levels of EmGFP/IL-15 than SIRPa+ DCs when
compared with the average MFI of EmGFP/IL-15 in BMDCs
grown from Tg2 controls (Fig. 5G). Thus, our findings suggested
that Flt3 signaling initiates a developmental program that includes
the regulation of IL-15 expression in developing DCs.

Discussion
IL-15 is an essential cytokine required for the development and
maintenance of several populations of immune cells. However, the
unfortunate combination of low expression levels and, perhaps, poor
detection reagents has made studying certain elements of IL-15
FIGURE 3. Murine HSCs express high levels of EmGFP/IL-15. (A) HSCs biology extremely difficult. By using a transgenic IL-15 reporter
were identified in the BM of reporter mice and littermate controls as LSK. mouse, we showed that IL-15 expression can be detected in numerous
Data are representative of at least two independent experiments. (Lin2 = subsets of hematopoietically derived immune cells. Our findings in-
CD32CD192CD11b2CD11c2Gr-12NK1.12Ter1192.) (B and C) Thymo- dicated that granulocytic cells such as neutrophils, eosinophils, and
cytes from reporter mice and littermate controls were examined for ex-
basophils have the potential to make IL-15 in vivo. However, it is
pression of EmGFP/IL-15. DN thymocyte populations were defined as
important to note that proven reagents for detecting IL-15 protein
follows: CD32CD42CD82; ETP, c-Kit+CD252; DN2, CD44highCD25+,
both c-Kit+ and c-Kit2; DN3, CD442CD25+; DN4, CD442CD252. Double- following in vitro culture failed to detect any measurable levels of
positive (DP) cells were CD32CD4+CD8+, and single positive (SP) cells IL-15 on any myeloid cells, including DCs, directly ex vivo. Thus,
were either CD3+CD42CD8+ or CD3+CD4+CD82. The MFI of EmGFP/IL- whereas the IL-15 reporter communicates transcriptional rather than
15 is shown graphically for each population in (B). Gray histograms show translational activity, it nevertheless remains an excellent tool for
Tg2, black line indicates Tg+. *p , 0.05. n.s., Not significant. dissecting the regulation of IL-15 expression in vivo.
The Journal of Immunology 3021

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


FIGURE 4. Notch-driven regulation of IL-15 gene expression during thymopoiesis. (A and B) LSK cells were sorted from Tg+ and Tg2 mice and
cultured with OP9 or OP9-DL1 stromal cells in the presence of Flt3L and IL-7 for 9 d, as previously described (24). In (A), contour plots (left) indicate the
level of CD44 and CD25 expression on the population of Gr-12 cells. The relative level of EmGFP/IL-15 in CD25+ DN2 (CD44high) or DN3 (CD44low)
cells grown on either OP9 or OP9-DL1 cells is shown in the histogram (center) and depicted graphically (right). *p , 0.05 using ANOVA followed by
a Tukey post test. In (B), cells were harvested and analyzed for expression of CD45 and Gr-1 to identify hematopoietically derived granulocytes (density
plots, left). The relative level of EmGFP/IL-15 expression in each population is shown in the histogram (center) and graphically (right). Results are
representative of two individual experiments. n.s., Not significant using Student t test.

Studies have shown that certain subsets of IL-15–dependent cells T cell behavior in the BM can also be influenced by CXCL12 (36).
require IL-15 to be trans-presented by a specific subset of cells. Furthermore, IL-15 signaling can induce CXCR4 expression on
For example, limiting IL-15 trans-presentation to CD11c+ cells in vitro–generated CD4 memory T cells, which perhaps results
was largely sufficient to restore memory CD8 T cells in the pe- in the juxtaposition of memory T cells with IL-15–producing
riphery but failed to reconstitute the IEL compartment of the in- hematopoietic progentiors (43). Regardless, evidence clearly sug-
testine (17). Alternatively, IELs require IL-15 to be delivered by gests that some IL-15 is derived from hematopoietic sources in
intestinal epithelial cells (18, 21). Because many of the progenitor the BM, enough to significantly increase the presence of NK
and granulocytic populations that we have shown in this study cells in the BM of IL-15 KO mice reconstituted with WT BM
to express IL-15 are present at extremely low frequencies, it will (14). Interestingly, however, deletion of IL-15Ra from DCs had
likely be difficult to examine their individual contributions in no effect on the presence of memory CD8 T cells specifically
maintaining IL-15–dependent homeostasis in vivo. Additionally, it within the BM (21). This may not be surprising considering the
is highly likely that some degree of compensatory mechanisms relatively low numbers of DCs present in this location and the
exists when manipulation of IL-15/IL-15Ra expression occurs. expression of IL-15 by a number of other cell types that likely act
For example, when IL-15Ra is conditionally deleted from both in conjunction with stromal cells to drive IL-15–dependent re-
CD11c+ and LysM+ cells, the defect in NK cell homeostasis is not sponses in the BM.
exacerbated above deletion in either subset alone, suggesting that Our studies show that steady-state IL-15 expression is linked to
CD11c+ cells and LysM+ cells provide similar functions in the a myeloid cell fate. There are numerous transcriptional networks at
absence of the other (21). play in the myeloid versus lymphoid cell fate decision, and it is
The BM has been suggested as a site of long-term memory CD8 highly unlikely that a single transcription factor will be identified
and CD4 T cell maintenance (35–38), and this is perhaps the result that regulates all instances of IL-15 expression in hematopoietic
of IL-15 reservoirs present in that site. Snell et al. (39) identified cells. However, one potential candidate is the ets family member
a subset of memory CD8 T cells in the BM that expressed high PU.1 (encoded by Sfpi1), which is essential for myeloid cell de-
levels of glucocorticoid-induced TNFR (GITR) but were absent velopment, including macrophages and DCs (44–48). Also im-
from secondary lymphoid tissues in the periphery and also from portant for B cell development, gradations of PU.1 expression
the BM of IL-15–deficient mice. They further showed that in progenitor cells drive the generation of B lymphocytes versus
memory CD8 T cells upregulated GITR in response to IL-15 macrophages (49). Alternatively, concurrent with commitment to
signaling in vitro and upregulated GITR upon migration into the the T cell lineage is a loss of expression of myeloid-specific
BM, suggesting that IL-15 signals were delivered specifically transcription factors (33, 50). In fact, constitutive expression of
within the BM. Similarly, studies in humans have demonstrated PU.1 is sufficient to block T cell development at the DN to double-
the close proximity of CD8 and CD4 memory T cells to IL-15– positive transition (51). However, recent studies have shown that
producing cells in the BM (40). Whereas others have shown that ETPs maintain a significant level of PU.1 expression and retain
IL-15 can be expressed by BM stromal cells (26, 41), to our myeloid potential, whereas DN3 cells, which are committed to
knowledge ours is the first study describing IL-15 expression by T cell development, lack PU.1 expression (33). We observed
hematopoietic progenitor cells residing in the BM. Interestingly, a similar pattern of IL-15 expression in developing T cells where
HSCs have been shown to reside in a specific BM niche using IL-15 was expressed at high levels by ETPs, but Notch signaling
a CXCL12/CXCR4-dependent mechanism (42), and memory led to the downregulation of IL-15 in DN3 and DN4 cells, perhaps
3022 DEVELOPMENTAL REGULATION OF IL-15

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


FIGURE 5. IL-15 expression is tightly regulated throughout DC development. BM cells were harvested from Tg+ mice (black line) and Tg2 littermate
controls (gray histograms) to determine EmGFP/IL-15 expression by (A, B) Lin2Sca-12CD135+ macrophage-DC progenitors (MDP), common DC
progenitors (CDP), and (C, D) Lin22CD11c+MHC class II2CD135+CD172a2 pre-DC cells. Gating and histograms of EmGFP/IL-15 expression are shown
in (A) and (C) and depicted graphically in (B) and (D). The level of EmGFP/IL-15 expressed by LSK cells (see Fig. 3) is shown for statistical comparison in
(B). Lin22 = CD32CD192B2202Ter1192NK1.12. (E and F) Pre-DCs were purified by FACS from Tg+ and Tg2 controls and transferred to naive CD45.1
recipients between 3 and 4 wk of age. After 6 d, the spleens of recipient mice were harvested to determine the phenotype of donor-derived cells. Spleens
were first stained with anti–CD45.2-allophycocyanin followed by incubation with anti-allophycocyanin microbeads to enrich for the donor-derived pop-
ulation by MACS prior to flow cytometric analysis. One mouse that received no donor cells is shown as a control. Data in (E) are concatenated from one of
two independent experiments with three to four recipient mice per group per experiment. The data in (F) are pooled from the two independent experiments.
(G) BM cells from Tg+ and Tg2 mice were cultured in vitro with Flt3L, and after 9 d, cells were harvested and analyzed by flow cytometry. pDCs were
gated as B220+CD11cint, and cDC populations (either CD103+ or SIRPa+) were first gated as B2202CD11c+. The data are presented as the fold increase in
MFI of EmGFP over the average MFI of EmGFP from Tg2 controls for each population (F, G). *p , 0.05.

by overriding PU.1-initiated lineage commitment (52–54). Thus, subsets. The Flt3/Flt3L signaling axis plays a dominant role in
silencing of IL-15 in the lymphoid lineages occurred as progeni- controlling DC development (55). This conclusion has been
tors were directed toward a lymphoid fate. Because preliminary established in vivo (56, 57) but is also apparent using Flt3L (as
studies have suggested that PU.1 can bind to a site in the first opposed to GM-CSF) to generate DCs from BM progenitors
intron of the IL-15 gene locus (E.V. Rothenberg, personal com- in vitro (34). Considering these two commonly used approaches
munication), the possibility exists that PU.1 can modulate IL-15 for generating BMDCs, it is interesting that previous work shows
expression in vivo. Interestingly, retroviral transduction of DN3 or that CD8+ memory T cells are better maintained by Flt3L-derived
DN4 cells with PU.1 enforces a myeloid cell fate when cultured DCs than those grown in GM-CSF (58), which could suggest
on OP9 stromal cells (50). Whether or not this transcriptional re- a preferential role for Flt3 signaling in driving optimal IL-15
programming would include the upregulation of IL-15 could re- expression by terminally differentiated DCs. Flt3/Flt3L is also
veal a direct role for PU.1 in driving IL-15 transcription. critical for the generation of pDCs, which we have shown lack IL-
We further found that IL-15 expression was continuously modu- 15 expression. pDCs are unique from their conventional coun-
lated during myelopoiesis, particularly in the development of DC terparts because they share similarities with developing lympho-
The Journal of Immunology 3023

cytes, including the expression of RAG genes (59). Because we 8. Burkett, P. R., R. Koka, M. Chien, S. Chai, F. Chan, A. Ma, and D. L. Boone.
2003. IL-15Ra expression on CD8+ T cells is dispensable for T cell memory.
have shown that IL-15 expression is turned off in lymphoid lin- Proc. Natl. Acad. Sci. USA 100: 4724–4729.
eages during their development, perhaps the transcriptional net- 9. Olsen, S. K., N. Ota, S. Kishishita, M. Kukimoto-Niino, K. Murayama,
work that is responsible for communicating lymphoidesque H. Uchiyama, M. Toyama, T. Terada, M. Shirouzu, O. Kanagawa, and
S. Yokoyama. 2007. Crystal structure of the interleukin-15·interleukin-15 re-
characteristics on to this population of innate cells could also be ceptor a complex: insights into trans and cis presentation. J. Biol. Chem. 282:
responsible for IL-15 downregulation during their development. 37191–37204.
Finally, which signals and transcription factors collaborate to 10. Schluns, K. S., K. Williams, A. Ma, X. X. Zheng, and L. Lefrançois. 2002.
Cutting edge: requirement for IL-15 in the generation of primary and memory
control the differentiation of pre-DCs into CD8+ versus CD82 antigen-specific CD8 T cells. J. Immunol. 168: 4827–4831.
cDCs is currently unknown. However, our data, both in vivo and 11. Wu, Z., H. H. Xue, J. Bernard, R. Zeng, D. Issakov, J. Bollenbacher-Reilley,
I. M. Belyakov, S. Oh, J. A. Berzofsky, and W. J. Leonard. 2008. The IL-15
in vitro, indicated that CD8+ DCs express higher levels of IL-15. receptor a chain cytoplasmic domain is critical for normal IL-15Ralpha function
Id2, IRF8, and Batf3 have all been shown to be required for CD8+ but is not required for trans-presentation. Blood 112: 4411–4419.
DC development, but whether these specific transcription factors 12. Castillo, E. F., and K. S. Schluns. 2012. Regulating the immune system via IL-15
transpresentation. Cytokine 59: 479–490.
act at the IL-15 locus remains to be examined. The differential 13. Schluns, K. S., K. D. Klonowski, and L. Lefrançois. 2004. Transregulation of
expression of IL-15 by cDC subsets has consequences in vivo, as memory CD8 T-cell proliferation by IL-15Ra+ bone marrow-derived cells.
both CD8+ DCs and IL-15Ra were required to maintain a unique Blood 103: 988–994.
14. Schluns, K. S., E. C. Nowak, A. Cabrera-Hernandez, L. Puddington,
population of CD122+ Ag–inexperienced CD8 T cells that exhibit L. Lefrançois, and H. L. Aguila. 2004. Distinct cell types control lymphoid
a memory phenotype (28). Importantly, our previous studies have subset development by means of IL-15 and IL-15 receptor a expression. Proc.
Natl. Acad. Sci. USA 101: 5616–5621.
demonstrated that both subsets of cDCs have the capacity to up- 15. Sandau, M. M., K. S. Schluns, L. Lefrancois, and S. C. Jameson. 2004. Cutting
regulate IL-15 expression following virus-induced inflammation edge: transpresentation of IL-15 by bone marrow-derived cells necessitates ex-

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


in an IFN-a receptor–dependent fashion, suggesting that the pression of IL-15 and IL-15R alpha by the same cells. J. Immunol. 173: 6537–
6541.
downregulation of IL-15 by immature CD82 DCs does not in- 16. Burkett, P. R., R. Koka, M. Chien, S. Chai, D. L. Boone, and A. Ma. 2004.
volve permanent modifications to the gene locus (23). Coordinate expression and trans presentation of interleukin (IL)-15Ra and IL-15
In conclusion, the use of an IL-15 reporter transgenic system has supports natural killer cell and memory CD8+ T cell homeostasis. J. Exp. Med.
200: 825–834.
facilitated the identification of immune cells with the potential to 17. Stonier, S. W., L. J. Ma, E. F. Castillo, and K. S. Schluns. 2008. Dendritic cells
produce IL-15 in vivo. We have shown a large-scale bifurcation in drive memory CD8 T-cell homeostasis via IL-15 transpresentation. Blood 112:
4546–4554.
the two lineages of hematopoietically derived cells whereby IL-15 18. Ma, L. J., L. F. Acero, T. Zal, and K. S. Schluns. 2009. Trans-presentation of IL-
expression is primarily limited to myeloid cells and down- 15 by intestinal epithelial cells drives development of CD8aa IELs. J. Immunol.
modulated during lymphoid cell development. Surprisingly, IL-15 183: 1044–1054.
19. Castillo, E. F., S. W. Stonier, L. Frasca, and K. S. Schluns. 2009. Dendritic cells
was expressed at high levels by HSCs in the BM, and IL-15 support the in vivo development and maintenance of NK cells via IL-15 trans-
regulation occurred during both thymopoiesis and DC develop- presentation. J. Immunol. 183: 4948–4956.
ment. In total, our findings suggested that molecular mechanisms 20. Castillo, E. F., L. F. Acero, S. W. Stonier, D. Zhou, and K. S. Schluns. 2010.
Thymic and peripheral microenvironments differentially mediate develop-
responsible for the development of cells in both myeloid and ment and maturation of iNKT cells by IL-15 transpresentation. Blood 116:
lymphoid lineages were also able to exert regulatory control at the 2494–2503.
21. Mortier, E., R. Advincula, L. Kim, S. Chmura, J. Barrera, B. Reizis, B. A. Malynn,
IL-15 gene locus. and A. Ma. 2009. Macrophage- and dendritic-cell-derived interleukin-15 recep-
tor alpha supports homeostasis of distinct CD8+ T cell subsets. Immunity 31:
811–822.
Acknowledgments 22. Fehniger, T. A., K. Suzuki, A. Ponnappan, J. B. VanDeusen, M. A. Cooper,
S. M. Florea, A. G. Freud, M. L. Robinson, J. Durbin, and M. A. Caligiuri. 2001.
We thank Quynh-Mai Pham and the University of Connecticut Health Cen-
Fatal leukemia in interleukin 15 transgenic mice follows early expansions in
ter Flow Cytometry Core for technical assistance and Juan Carlos Zuniga- natural killer and memory phenotype CD8+ T cells. J. Exp. Med. 193: 219–231.
Pflücker for OP9 and OP9-DL1 stromal cells. 23. Colpitts, S. L., T. A. Stoklasek, C. R. Plumlee, J. J. Obar, C. Guo, and
L. Lefrançois. 2012. Cutting edge: the role of IFN-a receptor and MyD88 sig-
naling in induction of IL-15 expression in vivo. J. Immunol. 188: 2483–2487.
Disclosures 24. Holmes, R., and J. C. Zuniga-Pflucker. 2009. The OP9-DL1 system: generation
T.A.S. and L.L. have a patent related to the IL-15 complex and have re- of T-lymphocytes from embryonic or hematopoietic stem cells in vitro. Cold
ceived royalties from Marine Polymers Technology, Inc. The other authors Spring Harb. Protoc. 2009: pdb.prot5156.
25. Liu, K., G. D. Victora, T. A. Schwickert, P. Guermonprez, M. M. Meredith, K. Yao,
have no financial conflicts of interest. F. F. Chu, G. J. Randolph, A. Y. Rudensky, and M. Nussenzweig. 2009. In vivo
analysis of dendritic cell development and homeostasis. Science 324: 392–397.
26. Grabstein, K. H., J. Eisenman, K. Shanebeck, C. Rauch, S. Srinivasan, V. Fung,
References C. Beers, J. Richardson, M. A. Schoenborn, M. Ahdieh, et al. 1994. Cloning of
1. Giri, J. G., S. Kumaki, M. Ahdieh, D. J. Friend, A. Loomis, K. Shanebeck, a T cell growth factor that interacts with the beta chain of the interleukin-2
R. DuBose, D. Cosman, L. S. Park, and D. M. Anderson. 1995. Identification and receptor. Science 264: 965–968.
cloning of a novel IL-15 binding protein that is structurally related to the a chain 27. Mattei, F., G. Schiavoni, F. Belardelli, and D. F. Tough. 2001. IL-15 is expressed
of the IL-2 receptor. EMBO J. 14: 3654–3663. by dendritic cells in response to type I IFN, double-stranded RNA, or lipo-
2. Duitman, E. H., Z. Orinska, E. Bulanova, R. Paus, and S. Bulfone-Paus. 2008. polysaccharide and promotes dendritic cell activation. J. Immunol. 167: 1179–
How a cytokine is chaperoned through the secretory pathway by complexing 1187.
with its own receptor: lessons from interleukin-15 (IL-15)/IL-15 receptor a. Mol. 28. Sosinowski, T., J. T. White, E. W. Cross, C. Haluszczak, P. Marrack, L. Gapin,
Cell. Biol. 28: 4851–4861. and R. M. Kedl. 2013. CD8a+ dendritic cell trans presentation of IL-15 to naive
3. Dubois, S., J. Mariner, T. A. Waldmann, and Y. Tagaya. 2002. IL-15Ra recycles CD8+ T cells produces antigen-inexperienced T cells in the periphery with
and presents IL-15 in trans to neighboring cells. Immunity 17: 537–547. memory phenotype and function. J. Immunol. 190: 1936–1947.
4. Kennedy, M. K., M. Glaccum, S. N. Brown, E. A. Butz, J. L. Viney, M. Embers, 29. Miyazaki, S., F. Ishikawa, K. Shimizu, T. Ubagai, P. H. Edelstein, and
N. Matsuki, K. Charrier, L. Sedger, C. R. Willis, et al. 2000. Reversible defects K. Yamaguchi. 2007. Gr-1high polymorphonuclear leukocytes and NK cells act
in natural killer and memory CD8 T cell lineages in interleukin 15-deficient via IL-15 to clear intracellular Haemophilus influenzae in experimental murine
mice. J. Exp. Med. 191: 771–780. peritonitis and pneumonia. J. Immunol. 179: 5407–5414.
5. Lodolce, J. P., D. L. Boone, S. Chai, R. E. Swain, T. Dassopoulos, S. Trettin, and 30. Mortier, E., T. Woo, R. Advincula, S. Gozalo, and A. Ma. 2008. IL-15Ra
A. Ma. 1998. IL-15 receptor maintains lymphoid homeostasis by supporting chaperones IL-15 to stable dendritic cell membrane complexes that activate NK
lymphocyte homing and proliferation. Immunity 9: 669–676. cells via trans presentation. J. Exp. Med. 205: 1213–1225.
6. Koka, R., P. R. Burkett, M. Chien, S. Chai, F. Chan, J. P. Lodolce, D. L. Boone, 31. Allman, D., A. Sambandam, S. Kim, J. P. Miller, A. Pagan, D. Well, A. Meraz,
and A. Ma. 2003. Interleukin (IL)-15Ra-deficient natural killer cells survive in and A. Bhandoola. 2003. Thymopoiesis independent of common lymphoid
normal but not IL-15Ra-deficient mice. J. Exp. Med. 197: 977–984. progenitors. Nat. Immunol. 4: 168–174.
7. Lodolce, J. P., P. R. Burkett, D. L. Boone, M. Chien, and A. Ma. 2001. T cell- 32. Maillard, I., T. Fang, and W. S. Pear. 2005. Regulation of lymphoid development,
independent interleukin 15Ra signals are required for bystander proliferation. J. differentiation, and function by the Notch pathway. Annu. Rev. Immunol. 23:
Exp. Med. 194: 1187–1194. 945–974.
3024 DEVELOPMENTAL REGULATION OF IL-15

33. Bell, J. J., and A. Bhandoola. 2008. The earliest thymic progenitors for T cells 47. Guerriero, A., P. B. Langmuir, L. M. Spain, and E. W. Scott. 2000. PU.1 is
possess myeloid lineage potential. Nature 452: 764–767. required for myeloid-derived but not lymphoid-derived dendritic cells. Blood 95:
34. Brasel, K., T. De Smedt, J. L. Smith, and C. R. Maliszewski. 2000. Generation of 879–885.
murine dendritic cells from flt3-ligand-supplemented bone marrow cultures. 48. Carotta, S., A. Dakic, A. D’Amico, S. H. Pang, K. T. Greig, S. L. Nutt, and
Blood 96: 3029–3039. L. Wu. 2010. The transcription factor PU.1 controls dendritic cell development
35. Becker, T. C., S. M. Coley, E. J. Wherry, and R. Ahmed. 2005. Bone marrow is and Flt3 cytokine receptor expression in a dose-dependent manner. Immunity 32:
a preferred site for homeostatic proliferation of memory CD8 T cells. J. 628–641.
Immunol. 174: 1269–1273. 49. DeKoter, R. P., and H. Singh. 2000. Regulation of B lymphocyte and macro-
36. Mazo, I. B., M. Honczarenko, H. Leung, L. L. Cavanagh, R. Bonasio, phage development by graded expression of PU.1. Science 288: 1439–1441.
W. Weninger, K. Engelke, L. Xia, R. P. McEver, P. A. Koni, et al. 2005. Bone 50. Laiosa, C. V., M. Stadtfeld, H. Xie, L. de Andres-Aguayo, and T. Graf. 2006.
marrow is a major reservoir and site of recruitment for central memory CD8+ Reprogramming of committed T cell progenitors to macrophages and den-
T cells. Immunity 22: 259–270. dritic cells by C/EBPa and PU.1 transcription factors. Immunity 25: 731–
37. Parretta, E., G. Cassese, P. Barba, A. Santoni, J. Guardiola, and F. Di Rosa. 2005. 744.
CD8 cell division maintaining cytotoxic memory occurs predominantly in the 51. Anderson, M. K., A. H. Weiss, G. Hernandez-Hoyos, C. J. Dionne, and
bone marrow. J. Immunol. 174: 7654–7664. E. V. Rothenberg. 2002. Constitutive expression of PU.1 in fetal hematopoietic
38. Tokoyoda, K., S. Zehentmeier, A. N. Hegazy, I. Albrecht, J. R. Grün, progenitors blocks T cell development at the pro-T cell stage. Immunity 16: 285–
M. Löhning, and A. Radbruch. 2009. Professional memory CD4+ T lymphocytes 296.
preferentially reside and rest in the bone marrow. Immunity 30: 721–730. 52. Franco, C. B., D. D. Scripture-Adams, I. Proekt, T. Taghon, A. H. Weiss,
39. Snell, L. M., G. H. Lin, and T. H. Watts. 2012. IL-15-dependent upregulation of M. A. Yui, S. L. Adams, R. A. Diamond, and E. V. Rothenberg. 2006. Notch/
GITR on CD8 memory phenotype T cells in the bone marrow relative to spleen Delta signaling constrains reengineering of pro-T cells by PU.1. Proc. Natl.
and lymph node suggests the bone marrow as a site of superior bioavailability of Acad. Sci. USA 103: 11993–11998.
IL-15. J. Immunol. 188: 5915–5923. 53. Rothenberg, E. V., and D. D. Scripture-Adams. 2008. Competition and collab-
40. Herndler-Brandstetter, D., K. Landgraf, B. Jenewein, A. Tzankov, R. Brunauer, oration: GATA-3, PU.1, and Notch signaling in early T-cell fate determination.
S. Brunner, W. Parson, F. Kloss, R. Gassner, G. Lepperdinger, and B. Grubeck- Semin. Immunol. 20: 236–246.
Loebenstein. 2011. Human bone marrow hosts polyfunctional memory CD4+ 54. Del Real, M. M., and E. V. Rothenberg. 2013. Architecture of a lymphomyeloid
and CD8+ T cells with close contact to IL-15-producing cells. J. Immunol. 186: developmental switch controlled by PU.1, Notch and Gata3. Development 140:

Downloaded from http://www.jimmunol.org/ by guest on September 20, 2017


6965–6971. 1207–1219.
41. Mrózek, E., P. Anderson, and M. A. Caligiuri. 1996. Role of interleukin-15 in the 55. Schmid, M. A., D. Kingston, S. Boddupalli, and M. G. Manz. 2010. Instructive
development of human CD56+ natural killer cells from CD34+ hematopoietic cytokine signals in dendritic cell lineage commitment. Immunol. Rev. 234: 32–
progenitor cells. Blood 87: 2632–2640. 44.
42. Sugiyama, T., H. Kohara, M. Noda, and T. Nagasawa. 2006. Maintenance of the 56. McKenna, H. J., K. L. Stocking, R. E. Miller, K. Brasel, T. De Smedt,
hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone E. Maraskovsky, C. R. Maliszewski, D. H. Lynch, J. Smith, B. Pulendran, et al.
marrow stromal cell niches. Immunity 25: 977–988. 2000. Mice lacking flt3 ligand have deficient hematopoiesis affecting hemato-
43. Jourdan, P., J. P. Vendrell, M. F. Huguet, M. Segondy, J. Bousquet, J. Pène, and poietic progenitor cells, dendritic cells, and natural killer cells. Blood 95: 3489–
H. Yssel. 2000. Cytokines and cell surface molecules independently induce CXCR4 3497.
expression on CD4+ CCR7+ human memory T cells. J. Immunol. 165: 716–724. 57. Waskow, C., K. Liu, G. Darrasse-Jèze, P. Guermonprez, F. Ginhoux, M. Merad,
44. Scott, E. W., M. C. Simon, J. Anastasi, and H. Singh. 1994. Requirement of T. Shengelia, K. Yao, and M. Nussenzweig. 2008. The receptor tyrosine kinase
transcription factor PU.1 in the development of multiple hematopoietic lineages. Flt3 is required for dendritic cell development in peripheral lymphoid tissues.
Science 265: 1573–1577. Nat. Immunol. 9: 676–683.
45. McKercher, S. R., B. E. Torbett, K. L. Anderson, G. W. Henkel, D. J. Vestal, 58. Frasca, L., S. W. Stonier, W. W. Overwijk, and K. S. Schluns. 2010. Differential
H. Baribault, M. Klemsz, A. J. Feeney, G. E. Wu, C. J. Paige, and R. A. Maki. mechanisms of memory CD8 T cell maintenance by individual myeloid cell
1996. Targeted disruption of the PU.1 gene results in multiple hematopoietic types. J. Leukoc. Biol. 88: 69–78.
abnormalities. EMBO J. 15: 5647–5658. 59. Shigematsu, H., B. Reizis, H. Iwasaki, S. Mizuno, D. Hu, D. Traver, P. Leder,
46. Scott, E. W., R. C. Fisher, M. C. Olson, E. W. Kehrli, M. C. Simon, and H. Singh. N. Sakaguchi, and K. Akashi. 2004. Plasmacytoid dendritic cells activate
1997. PU.1 functions in a cell-autonomous manner to control the differentiation lymphoid-specific genetic programs irrespective of their cellular origin. Immu-
of multipotential lymphoid-myeloid progenitors. Immunity 6: 437–447. nity 21: 43–53.

You might also like