Hlx homeobox transcription factor negatively regulates interferon-production in monokine-activated natural killer cells

Page created by Theodore Parker
 
CONTINUE READING
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

IMMUNOBIOLOGY

Hlx homeobox transcription factor negatively regulates interferon-␥ production in
monokine-activated natural killer cells
Brian Becknell,1,2 Tiffany L. Hughes,2 Aharon G. Freud,1,2 Bradley W. Blaser,1,2 Jianhua Yu,3 Rossana Trotta,3
Hsiaoyin C. Mao,3 Marie L. Caligiuri de Jesús,4 Mohamad Alghothani,4 Don M. Benson Jr,4 Amy Lehman,5 David Jarjoura,5
Danilo Perrotti,6,8 Michael D. Bates,7 and Michael A. Caligiuri2-4,8
1Medical Scientist Program, 2Integrated Biomedical Science Graduate Program, 3Department of Molecular Virology, Immunology, and Medical Genetics,
4Divisionof Hematology/Oncology, 5Department of Epidemiology and Biometrics, 6Human Cancer Genetics Program, Department of Internal Medicine, College
of Medicine and Public Health, The Ohio State University, Columbus; 7Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital
Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, OH; 8Comprehensive Cancer Center, The Ohio State University,
Columbus

Natural killer (NK) cells contribute to host              activated NK cells, but with delayed kinet-      of the active Y693-phosphorylated form
immunity, including tumor surveillance,                   ics compared to IFN-␥. Ectopic Hlx expres-       of STAT4. Thus, Hlx expression in acti-
through the production of interferon                      sion decreases IFN-␥ synthesis in primary        vated NK cells temporally controls and
gamma (IFN-␥). Although there is some                     human NK cells and IFN-␥ promoter activ-         limits the monokine-induced produc-
knowledge about molecular mechanisms                      ity in an NK-like cell line. Hlx protein         tion of IFN-␥, in part through the tar-
that induce IFN-␥ in NK cells, consider-                  levels inversely correlate with those of         geted depletion of STAT4. (Blood. 2007;
ably less is known about the mechanisms                   STAT4, a requisite factor for optimal IFN-␥      109:2481-2487)
that reduce its expression. Here, we inves-               transcription. Mechanistically, we pro-
tigate the role of the Hlx transcription                  vide evidence indicating that Hlx overex-
factor in IFN-␥ production by NK cells.                   pression accelerates dephosphorylation
Hlx expression is induced in monokine-                    and proteasome-dependent degradation             © 2007 by The American Society of Hematology

Introduction
Innate immunity is characterized by the ability of immune cells to               numerous trans-acting factors have been implicated as positive
rapidly detect an invading pathogen and to restrict its dissemination            regulators of IFN-␥ gene expression8; however, the negative
while targeting compromised host cells for elimination. This                     regulation of this process is far less understood at a molecular level.
complex task is achieved in part through the production of soluble                   The H2.0-like homeobox 1 (HLX1, HB24, Hlx) gene encodes a
cytokines and chemokines by natural killer (NK) cells.1 NK cells                 highly conserved putative homeobox transcription factor9 that is
elaborate interferon gamma (IFN-␥) in response to stimulation with               preferentially expressed by T-helper 1 (TH1) polarized CD4⫹ T
monokines, particularly IL-12 in combination with IL-15, IL-18, or               cells.10,11 Ectopic expression of Hlx during TH2 differentiation
IL-1␤.2 IFN-␥ signals through a heterodimeric receptor and STAT1                 leads to increased IFN-␥ mRNA and protein expression.10-12 In this
to promote the maturation and activation of monocytes, leading to                study, we report that Hlx is a negative regulator of IFN-␥
improved antigen presentation and the establishment of macro-                    production by NK cells and that its inhibitory function is achieved
phage effector functions.3                                                       at least in part through the proteasomal degradation of STAT4, a
    The importance of IFN-␥ is illustrated by naturally occurring                key transcription factor for IFN-␥ mRNA synthesis.13,14
mutations in IFN-␥ receptor subunits and STAT1 in humans, who
are highly susceptible to systemic disease after mycobacterial
infection or vaccination with bacillus Calmette-Guerin.4 In addi-                Materials and methods
tion, mice lacking IFN-␥ responsiveness exhibit increased inci-
dence of tumors, consistent with a role for IFN-␥ in tumor                       Human NK cell isolation and cell lines
surveillance by immune cells.5 Although the beneficial roles of                  Human NK cells were isolated from peripheral blood (American Red
IFN-␥ are unquestionable, excessive IFN-␥ can be detrimental to                  Cross) in accordance with The Ohio State University Institutional
the host. In fact, high levels of IFN-␥ are implicated in the etiology           Review Board. NK-92, Phoenix-Ampho, and DERL-7 cells were
of inflammatory bowel disease.6 In addition, unabated IFN-␥                      cultured as described.15,16
production leads to enhanced apoptosis of hematopoietic stem cells
and impaired NK cell development.7 Therefore, it is not surprising               Murine NK cell culture
that IFN-␥ synthesis is subject to stringent control in vivo, with               Hlx⫹/⫺ mice were maintained on an FVB/N background. Dams from
multiple checkpoints including transcriptional control.8 Indeed,                 Hlx⫹/⫺ breedings were killed at E13.5, and fetal livers were isolated. DNA

Submitted October 10, 2006; accepted November 6, 2006. Prepublished online       The publication costs of this article were defrayed in part by page charge
as Blood First Edition Paper, November 16, 2006; DOI 10.1182/blood-2006-10-      payment. Therefore, and solely to indicate this fact, this article is hereby
050096.                                                                          marked ‘‘advertisement’’ in accordance with 18 USC section 1734.

An Inside Blood analysis of this article appears at the front of this issue.     © 2007 by The American Society of Hematology

BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6                                                                                                             2481
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

2482    BECKNELL et al                                                                                 BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6

was extracted from embryonic tissue for genotyping (Extract-N-Amp               Cruz Biotechnology, Santa Cruz, CA), anti–␤-actin (1:1000; Santa Cruz
Tissue PCR Rapid Genotyping Kit; Sigma, St Louis, MO).17 Fetal liver            Biotechnology), anti-Ku70 (1:1000; Santa Cruz Biotechnology), and
cells were cultured in 96-well, round-bottom plates (104 cells/well) in         anti-Brg1 (1:1000).
DMEM with 10% FBS (Invitrogen, Carlsbad, CA), antibiotic/antimycotic,
glutamine, pegylated rat stem cell factor (100 ng/mL; Amgen, Thousand           Reporter assays
Oaks, CA), murine IL-7 (10 ng/mL; Peprotech, Rocky Hill, NJ), human
Flt-3 ligand (100 ng/mL; Amgen), and human IL-2 (1000 IU/mL; Roche,             Five million DERL-7 were transfected by nucleofection (Amaxa, Cologne,
Basel, Switzerland). Successful derivation of NK cells was confirmed            Germany) using Solution V and DNA as follows: 5 ␮g empty pGL3-Basic
cytometrically with NK1.1-APC and DX5-PE, CD19-biotin, CD14-biotin,             (Promega, Madison, WI) or the IFN-␥ firefly luciferase vector (a kind gift of
CD3-biotin, Ter119-biotin, and streptavidin-PerCP-Cy5.5 (BD PharMin-            Howard Young, National Cancer Institute22), 5 ␮g pcDNA3.1-Hlx or empty
gen, San Diego, CA).                                                            pcDNA3.1 vector (Invitrogen), 50 ng TK-renilla luciferase vector (pRL-
                                                                                TK; Promega), and program O-17. After electroporation, cells were
Retroviral infection of primary NK and NK-92 cells                              immediately placed in 10% RPMI containing IL-12/IL-18 and were
                                                                                incubated for 6 hours at 37°C. Cells were subsequently washed with
The entire Hlx open-reading frame, containing its native start and stop         ice-cold PBS and lysed in 140 ␮L 1 ⫻ passive lysis buffer (Promega), and
codons, was amplified from human leukocyte cDNA and introduced into             luminescence was analyzed with the dual-luciferase reporter system
pCR2.1-TOPO (Invitrogen). Forward and reverse primers for PCR were              (Promega). All assays were performed in triplicate, and all firefly luciferase
modified at their 5⬘ ends with BamHI and EcoRI sites, respectively. After       values were normalized for transfection efficiency by subtracting the renilla
bidirectional sequencing, the Hlx cDNA was mobilized to the PINCO               luciferase activity derived from the cotransfected pRL-TK plasmid. The
retroviral transfer vector as a BamHI-EcoRI fragment. Alternatively, for        activity of the pGL3 basic reporter vector alone was subtracted from that of
IFN-␥ reporter assays, Hlx was subcloned into pcDNA3.1 (Invitrogen).            the vector with the IFN-␥ promoter, and the mean ⫾ SD of the triplicate
Retrovirus was produced, and cells were infected and purified as described.16   values of the difference is shown.

Monokine stimulation of NK cells                                                Statistical analysis
Human NK cells were stimulated at 2 million/mL in RPMI medium                   Data were analyzed using a Student 2-tailed t test. P values below .05 were
containing 10% FBS and IL-12 (10 ng/mL; Genetics Institute, Cambridge,          considered statistically significant.
MA), IL-15 (100 ng/mL; Amgen), or IL-18 (50 ng/mL; R&D Systems,
Minneapolis, MN). Before stimulation at 1 million/mL, NK-92 cells were
washed extensively with PBS and rested for 24 hours in RPMI containing
10% FBS in the absence of IL-2. Murine NK cells were stimulated at 2            Results
million/mL in DMEM containing 10% FBS with the cytokines murine
IL-12 (10 ng/mL; Genetics Institute) and human IL-15 (3 ␮g/mL; Amgen).          Hlx expression is induced in monokine-activated NK cells
For proteasomal inhibition, MG-132 (Calbiochem), epoxomicin (Sigma),
and lactacystin were reconstituted according to the manufacturer’s instruc-     We first investigated the relationship between Hlx protein and
tions and incubated at 20 ␮M final concentration with rested PINCO-Hlx–         IFN-␥ mRNA levels in primary human NK cells over time after
infected NK-92 cells for 1 hour at 37°C before IL-12/IL-18 was added for        treatment with IL-12 and IL-18 (IL-12/IL-18). Hlx protein expres-
the indicated times. To inhibit protein translation, cycloheximide (Sigma)      sion was readily detectable by immunoblotting in resting NK cells,
was reconstituted in 100% ethanol and incubated at 20 ␮g/mL final               increased on IL-12/IL-18 treatment, peaked at 72 hours, and was
concentration with rested PINCO-Hlx–infected NK-92 cells for 30 minutes         maintained after 96 hours of stimulation (Figure 1A, upper panel).
at 37°C before IL-12/IL-18 was added for the indicated times.
                                                                                In contrast, IFN-␥ mRNA expression—measured by quantitative
                                                                                reverse transcription–polymerase chain reaction (QRT-PCR)—
Detection of IFN-␥ and Y693 pSTAT4
                                                                                peaked at 24 hours and subsequently declined (Figure 1A, lower
Intracellular staining for IFN-␥ was performed as described.18 Y693             panel). Maximal induction of Hlx expression required costimu-
pSTAT4 was detected with Y693 pSTAT4 or IgG2b isotype control mAb               lation with different cytokines (eg, IL-12/IL-18) (Figure 1B)
(BD PharMingen).19 Human IFN-␥ protein was detected by ELISA.18                 that, as reported,2 synergistically promoted high IFN-␥ produc-
                                                                                tion. Treatment with individual monokines led to modest or
QRT-PCR                                                                         undetectable changes in Hlx protein levels (Figure 1B), suggest-
Total RNA was extracted and reverse transcribed.18 Sequences of all             ing that increased Hlx expression may have a role in the
QRT-PCR primers and probes are available on request. QRT-PCR reactions          regulation of IFN-␥ production.
were performed and analyzed by the ⌬Ct method after normalizing to                  Because combination monokine treatment (eg, IL-12/IL-18 or
internal 18S control reactions, as described.20 Results—expressed as the        IL-12/IL-15) strongly stimulates IFN-␥ production by CD56bright
mean ⫾ SEM of triplicate wells—were the n-fold difference of transcript         but not CD56dim human NK cells,2,20 we investigated whether the
levels in an 18S-normalized sample compared with calibrator cDNA.               expression of Hlx was restricted to a specific CD56 NK subset.
Calibrator cDNA was unstimulated NK (Figure 1A), unstimulated CD56dim           Indeed, though Hlx mRNA and protein levels were comparable in
NK (Figure 1C), or unstimulated PINCO NK-92 (Figure 3C).
                                                                                resting CD56 NK subsets, induction of Hlx mRNA and protein
                                                                                expression occurred preferentially in IL-12/IL-18– and IL-12/IL-
Detection of Hlx protein and immunoblotting
                                                                                15–stimulated CD56bright NK cells (Figure 1C). Thus, Hlx expres-
Hlx protein was detected using affinity-purified rabbit antisera raised         sion is induced by monokine costimulation and enhanced within
against a GST-Hlx fusion protein (Abgent, San Diego, CA). To construct          the CD56bright NK subset; however, higher levels of Hlx correlated
the GST-Hlx fusion protein, a human Hlx cDNA fragment corresponding to          with decreased IFN-␥ production by NK cells.
amino acids 333 to 488 was PCR amplified and cloned into the BamHI and
EcoRI sites of pGEX-6P-1 (Amersham Biosciences, Freiburg, Germany).             Hlx negatively regulates IFN-␥ production in
Affinity-purified Hlx antiserum was used at 1:1000 dilution. Immuno-            monokine-activated NK cells
blotting was performed with whole cell lysates18 or nuclear and
cytosolic protein.21 Antibodies were anti–pSTAT4 Y693 (1:5000 mouse             Based on the kinetics of Hlx expression relative to IFN-␥ transcrip-
IgG2b monoclonal; BD PharMingen), anti–total STAT4 (1:1000; Santa               tion, it is conceivable that Hlx acts as a negative regulator of IFN-␥
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6                                                                                    Hlx INHIBITS IFN-␥ PRODUCTION BY NK CELLS     2483

                   A       Time (h)                   0    24    48         72          96
                                                                                                                 C                  CD56bright CD56dim
                           50 kDa

                                                                                                                                    12+18
                                                                                             HLX

                                                                                                                                    12+15
                                                                                                                                    12+18
                                                                                                                                    12+15
                                                                                             β-actin

                                                                                                                                    UN
                                                                                                                                    UN
                           42 kDa
                                     15000
                                                                                                                 50 kDa                                  Hlx
                                                                                                                                                         β−actin
                             IFN-γ mRNA
                                                                                                                 42 kDa
                                     10000

                                          5000                                                                                        P < .0001          P < .0002

                                                                                                                             1000

                                                                                                                  Hlx mRNA
                                             0
                                                 0         24        48          72          96                               100

                                                                Time (h)
                                                                                                                               10

                   B         UN                  12       15    18        12+15 12+18 15+18
                                                                                                                                1
                                                                                                                                      UN       12+15       UN        12+15

                  50 kDa                                                                                                               CD56bright            CD56dim
                                                                                                       Hlx
                  42 kDa                                                                               β−actin
Figure 1. Monokine-dependent Hlx expression by human                       CD56bright
                                                                          NK cells is delayed with respect to IFN-␥ transcription. (A) Delayed induction of Hlx protein with
respect to IFN-␥ production in primary NK cells. (upper) Total NK cells were stimulated with IL-12/IL-18 for indicated time points, and Hlx protein levels were analyzed by
immunoblotting. ␤-Actin levels were analyzed to ensure equal loading (n ⫽ 4 experiments). (lower) In parallel, IFN-␥ mRNA levels were analyzed by QRT-PCR. Mean ⫾ SEM
from 4 separate experiments is shown. (B) Maximal Hlx protein induction requires monokine costimulation. Total NK cells were stimulated for 72 hours, as indicated, and Hlx
and ␤-actin protein levels were analyzed by immunoblotting (n ⫽ 3 experiments). (C) Preferential induction of Hlx protein in the CD56bright NK subset. (upper) FACS-purified
unstimulated NK subsets were lysed directly (UN) or after stimulation with the indicated monokine combinations (72 hours). Lysates were analyzed for Hlx and ␤-actin protein
by immunoblotting (n ⫽ 3 experiments). (lower) FACS-purified NK subsets were lysed immediately for RNA (UN) or stimulated with IL-12/IL-15 for 12 hours before lysis. Hlx
mRNA levels were analyzed by QRT-PCR. Mean ⫾ SEM from 5 separate donors is shown.

production in CD56bright NK cells. Thus, IFN-␥ production was                                          Consistent with the data obtained with primary CD56bright NK cells,
assessed by intracellular staining in IL-12/IL-18–stimulated pri-                                      Hlx overexpression led to a marked decrease in IFN-␥ protein
mary human NK cells previously transduced with the EGFP                                                levels in IL-12/IL-18–stimulated NK-92 cells after 24 hours of
(enhanced green fluorescence protein)–expressing PINCO-Hlx or                                          stimulation (Figure 3B). Moreover, levels of IFN-␥ mRNA, albeit
empty PINCO retrovirus (Figure 2). Interestingly, a significant                                        similar in unstimulated vector- and Hlx-transduced NK-92 cells,
decrease in the proportion of IFN-␥⫹ cells was observed in the                                         dramatically increased over time after IL-12/IL-18 stimulation of
Hlx-expressing EGFP⫹CD56bright NK cells (CD56bright PINCO-Hlx                                          vector-transduced cells but not in NK-92 cells overexpressing Hlx
compared with CD56bright PINCO; P ⬍ .02; n ⫽ 3 experiments)                                            (Figure 3C). Thus, our data not only indicated that sustained Hlx
and in the CD56dim fraction (CD56dim PINCO-Hlx compared with                                           expression impairs the ability of NK cells to produce IFN-␥ in
CD56dim PINCO; P ⬍ .006; n ⫽ 3 experiments). In similar experi-                                        response to monokine costimulation, they also suggested that a
ments, human NK-92 cells, which as reported have a phenotype                                           monokine-induced increase of Hlx level is most likely required to
that resembles the CD56bright NK cell subset,23 were infected with                                     temporally control the expression and, therefore, the production of
the Hlx or the empty retrovirus and were EGFP-sorted. As                                               IFN-␥ by activated NK cells. Indeed, Hlx-deficient murine NK
expected, anti-Hlx Western blots performed on subcellular frac-                                        cells show enhanced IFN-␥ mRNA and protein production after in
tions from EGFP⫹ PINCO– and EGFP⫹ PINCO-Hlx–transduced                                                 vitro monokine costimulation with IL-12 and IL-15 compared with
NK-92 cell lysates showed that Hlx was indeed overexpressed and                                        wild-type and heterozygous control NK cells (Figure 3D and data
primarily localized in the nuclear compartment (Figure 3A). Note                                       not shown).
that Brg1 and Ku70 were detected as controls for purity of the
nuclear fraction and equal loading, respectively (Figure 3A).                                          Hlx represses IFNG promoter activity in NK cells

                                                                                                       Because Hlx overexpression in NK-92 cells resulted in decreased
                                                                                                       IFN-␥ mRNA levels and because Hlx is a homeobox transcription
                                                                                                       factor, we reasoned that Hlx may regulate the transcriptional
                                                                                                       activity of the IFNG promoter in NK cells. To begin to test this
                                                                                                       hypothesis, we sought to determine whether Hlx could inhibit
                                                                                                       IFNG promoter activity in vivo. We made use of luciferase reporter
                                                                                                       assays in the DERL-7 NK-like cell line,24 which is easily trans-
                                                                                                       duced by electroporation.15,25 DERL-7 was electroporated with a
                                                                                                       previously characterized full-length human IFNG promoter-firefly
                                                                                                       luciferase reporter construct containing an approximately 3.6-kb
                                                                                                       IFNG 5⬘ flanking sequence and an 0.8-kB enhancer element from
                                                                                                       the first intron of IFNG following the luciferase open-reading
                                                                                                       frame (Figure 4A).22,26 After electroporation, cells were treated
Figure 2. Hlx inhibits IFN-␥ production by primary CD56bright NK. PINCO- or                            with IL-12/IL-18 for 6 hours. After normalizing for transfection
PINCO-Hlx–infected primary human NK cells were stimulated for 24 hours with                            efficiency, we determined that Hlx overexpression repressed IFNG
IL-12/IL-18 and were subjected to staining for CD56 and IFN-␥. (left) Infected cells                   promoter activity (Figure 4B). However, on subsequent deletion
were gated on CD56brightEGFP⫹ or CD56dimEGFP⫹. (right) IFN-␥ staining in each
population compared with isotype controls. Results shown are representative of 3                       analysis of the 3.6-kb IFNG promoter and the 0.8-kb intron 1
experiments (CD56bright PINCO-Hlx compared with CD56bright PINCO; P ⬍ .02).                            enhancer element within the construct, we were unable to map Hlx
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

2484      BECKNELL et al                                                                                                            BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6

                                                                                             negatively regulate IFN-␥ gene transcription by suppressing STAT4
                                                                                             expression. Thus, levels of total STAT4 protein were also evaluated
                                                                                             in NK-92 cells transduced with Hlx or empty retrovirus, EGFP-
                                                                                             sorted and stimulated with IL-12/IL-18. Before stimulation, Hlx-
                                                                                             and vector-transduced NK-92 cells expressed similar levels of
                                                                                             unphosphorylated STAT4 (lower, faster-migrating band at 0 hour;
                                                                                             Figure 5B). Similarly, both populations demonstrated comparable
                                                                                             induction of Y693 pSTAT4 levels after IL-12/IL-18 stimulation
                                                                                             (upper, slower-migrating band at 0.5 hours; Figure 5B). However,
                                                                                             Hlx overexpression inhibited long-term monokine-induced STAT4
                                                                                             activation as levels of Y693 pSTAT4 progressively decreased over
                                                                                             time in Hlx-expressing NK-92 cells but not in vector-transduced
                                                                                             NK-92 cells (Figure 5B). In parallel experiments, we did not
                                                                                             observe an effect of Hlx on total and tyrosine-phosphorylated
                                                                                             STAT1 and STAT5, indicating that Hlx exerts its effect specifically
                                                                                             on STAT4. We also found that retroviral expression of Hlx in
                                                                                             primary NK reduced the proportion of Y693 pSTAT4⫹ cells after
                                                                                             IL-12/IL-18 treatment (from 78.0% ⫾ 5.4% in vector-transduced
                                                                                             cells to 67.1% ⫾ 5.4% in Hlx-transduced cells; n ⫽ 5 independent
                                                                                             experiments; P ⬍ .005).
                                                                                                 Interestingly, at later time points, we observed that ectopic Hlx
                                                                                             expression not only interfered with STAT4 activation, it also
                                                                                             significantly reduced the level of unphosphorylated STAT4 protein
                                                                                             (Figure 5B). This reduction in total STAT4 protein levels led us to
                                                                                             test whether Hlx overexpression is associated with a decrease in
                                                                                             STAT4 mRNA; however, no difference in STAT4 transcript levels
                                                                                             was detectable by QRT-PCR in Hlx compared with vector-infected
                                                                                             NK-92 cells during time-course experiments in IL-12/IL-18 (n ⫽ 4
                                                                                             experiments; data not shown). Alternatively, it has been reported
                                                                                             that Y693 pSTAT4 undergoes proteasome-dependent degradation
                                                                                             in T lymphocytes after IL-12 treatment.29,30 We therefore attempted
Figure 3. Hlx inhibits IFN-␥ mRNA and protein expression in NK-92 cells. (A)
Overexpression of Hlx protein in NK-92 cells. FACS-purified NK-92 cells transduced           to reverse the loss of Y693 pSTAT4 protein through the application
with PINCO or PINCO-Hlx were subjected to nucleocytoplasmic fractionation and                of MG-132, an inhibitor of the 26S proteasome subunit. Indeed,
immunoblotting for Hlx. Enrichment of nuclear protein was confirmed by Brg1
                                                                                             MG-132 rescued the loss of Y693 pSTAT4 after short-term (3 or 6
staining. Equal loading was confirmed by Ku70 staining. (B) Inhibition of IFN-␥
production by Hlx. PINCO- or PINCO-Hlx–infected NK-92 cells were stimulated with             hours) treatment of Hlx-expressing NK-92 cells with IL-12/IL-18
IL-12/IL-18, and supernatants were harvested after 24 hours for IFN-␥ ELISA. Shown           (Figure 5C). Similar results were obtained with the additional
are the mean ⫾ SEM from 5 separate experiments. (C) Time course of IFN-␥ mRNA                proteasome inhibitors epoxomicin and lactacystin (data not shown).
in PINCO- compared with PINCO-Hlx–transduced NK-92 in response to IL-12/IL-18.
Shown are results of 1 of 4 representative experiments; error bars represent SD of           Interestingly, proteasome inhibition also rescued expression of the
triplicate PCR reactions. (D) Increased IFN-␥ production by Hlx⫺/⫺ NK in response to         unphosphorylated STAT4 protein, particularly after 6-hour stimula-
monokine costimulation. Fetal liver–derived NK of indicated genotypes were sub-              tion with IL-12/IL-18 (Figure 5C). The E3 ubiquitin ligase SLIM
jected to 24-hour stimulation with IL-12 and IL-15, followed by intracellular staining for
IFN-␥. Mean ⫾ SEM percentages of IFN-␥⫹ NK1.1⫹ cells from 9 experiments with
                                                                                             has been shown to catalyze polyubiquitylation of Y693 pSTAT4 in
NK cells derived from 9 or more livers of each genotype are shown. *P ⬍ .001 Hlx⫹/⫹          vitro and to attenuate STAT4 protein expression in vivo.30 There-
compared with Hlx⫺/⫺. **P ⬍ .005 Hlx⫹/⫺ compared with Hlx⫺/⫺).                               fore, we sought to determine whether SLIM protein levels were

repressive activity to any particular region (data not shown). Thus,
the mechanism of IFNG promoter regulation by Hlx is likely                                             A                            3.6 kB                       0.8 kB
complex and dependent on multiple cis-regulatory elements.
                                                                                                                  IFNG promoter                                IFNG intron 1
                                                                                                                                                       luc
Hlx promotes proteasome-dependent STAT4 degradation
after monokine stimulation
                                                                                                      B
                                                                                                               promoter activity

We further hypothesized that Hlx might negatively regulate IFN-␥                                                                   40
                                                                                                                Relative IFNG

mRNA levels by inhibiting the function or activity of a positive                                                                   30
regulator of IFN-␥ production. STAT4 is a known trans-activator                                                                    20
                                                                                                                                   10
of the IFN-␥ promoter in NK cells.13,14 STAT4 is activated by                                                                       0
tyrosine phosphorylation at tyrosine residue 693 (Y693) by Jak2/                                                                             Vector      Hlx
Tyk2 kinases within seconds of IL-12 stimulation, resulting in its                           Figure 4. Hlx inhibits IFNG promoter activity in DERL-7 cells. (A) The human
rapid nuclear translocation and direct association with multiple                             IFNG luciferase construct used in this study consisted of 3.6 kB of a 5⬘ flanking
cis-regulatory elements within the IFN-␥ promoter.26-28 In IL-12/IL-                         sequence upstream of the transcriptional start site, the firefly luciferase open-reading
                                                                                             frame, and 0.8 kB of the intron 1 enhancer element. (B) DERL-7 cells with Hlx or
18–stimulated primary human NK or NK-92 cells, we observed
                                                                                             empty vector were transfected with the IFNG luciferase construct, and luciferase
that the increase of Hlx expression was accompanied by a decrease                            activity was measured after 6-hour stimulation with IL-12/IL-18. Mean ⫾SD of 1 of 3
in total STAT4 protein levels (Figure 5A), suggesting that Hlx may                           representative experiments is shown.
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6                                                                            Hlx INHIBITS IFN-␥ PRODUCTION BY NK CELLS                      2485

   A                                                     B                                                                         D
                     Primary Human NK                                      PINCO                       Hlx                                      PINCO            Hlx
  Time (h)   0         24     48    72   96                       0   0.5 1   3   6 12 18 24 0 0.5 1   3    6 12 18 24 Time (h)
                                                                                                                                           0 1 3 6 9 12 24 0 1 3 6 9 12 24
   50 kDa                                     Hlx                                                                                 80 kDa
                                                         80 kDa                                                         STAT4                                                    STAT4
   80 kDa                                     STAT4      42 kDa                                                        β−actin                                                   β-Actin
                                                                                                                                  42 kDa
   42 kDa                                     β-actin

                                                         C                                                                         E       _   _   +    +   _    _     +   +       CHX
                            NK-92
                                                             IL-12 + IL-18:         3h                     6h                              +   +   _    _   +    +     _   _       EtOH
  Time (h)       0       24        48    72
                                                                              -      V    MG      -         V    MG                        P   Hlx P    Hlx P   Hlx P      Hlx
   50 kDa                                      Hlx
                                                                  80 kDa                                               STAT4
   80 kDa                                      STAT4                                                                                                                             STAT4
   42 kDa                                      β-actin            42 kDa                                               Actin                                                     β-Actin

                                                                                                                                                12 h            24 h
Figure 5. Hlx overexpression leads to proteasome-dependent degradation of STAT4. (A) Endogenous Hlx levels increased as total STAT4 levels declined. Primary
human NK or NK-92 cells were harvested at the indicated time points after IL-12/IL-18 stimulation, and Hlx, STAT4, and ␤-actin protein levels were determined by
immunoblotting. (n ⫽ 4 experiments in primary NK, n ⫽ 2 experiments in NK-92). (B) Hlx overexpression decreased total and Y693 pSTAT4 levels in NK-92 after IL-12/IL-18
treatment. FACS-purified PINCO- or Hlx-expressing NK-92 cells were harvested at indicated time points after IL-12/IL-18 stimulation. Total STAT4 and ␤-actin levels were
assessed by immunoblotting (n ⫽ 4 experiments). (C) Proteasome inhibition rescued loss of STAT4 in NK-92 cells overexpressing Hlx. Cells were preincubated for 1 hour with
20 mM MG-132 (MG), an equal amount of DMSO vehicle, or medium alone (—), followed by IL-12/IL-18 treatment for the indicated times. Total protein was harvested, and total
STAT4 and ␤-actin levels were assessed by immunoblotting (n ⫽ 3 experiments). (D) Diminished Y693 pSTAT4 levels in Hlx-overexpressing NK-92 cells despite inhibition of
new protein synthesis. PINCO- or Hlx-expressing NK-92 cells were preincubated for 30 minutes with 10 ␮M CHX, followed by IL-12/IL-18 treatment for the indicated times.
Total cellular protein was harvested, and total STAT4 and ␤-actin levels were assessed by immunoblotting (the arrowhead indicates the position of the 80 kDa marker). (E)
Comparison of CHX compared with ethanol (EtOH) carrier effects on STAT4 expression in PINCO- (P) and Hlx-expressing NK-92 after IL-12/IL-18 stimulation for the indicated
times (n ⫽ 3 experiments).

altered by Hlx overexpression by immunoblotting, but no change in                              between the onset of detectable Hlx mRNA and appreciable Hlx
SLIM expression was observed (data not shown).                                                 protein expression (Figure 1A). In future studies, it will be
    To confirm that the loss of STAT4 protein was caused by                                    insightful to analyze the HLX promoter itself and to determine the
decreased protein stability, new protein synthesis was inhibited                               direct influence of monokines on its activity and the potential
using cycloheximide (CHX), and the kinetics of STAT4 degrada-                                  effects of NK-derived cytokines that have been shown to inhibit
tion were monitored in NK-92 expressing Hlx compared with                                      IFN-␥ production, such as transforming growth factor-␤ and
empty vector. As expected, in the presence of CHX, Hlx overexpres-                             TWEAK,15,31 on Hlx mRNA and protein expression.
sion resulted in decreased Y693 pSTAT4 levels beginning after 6                                    In addition, we identified the STAT4 transcription factor as a
hours of IL-12/IL-18 stimulation compared with empty vector                                    key molecular target of Hlx action in NK cells. Multiple lines of
(Figure 5D). Surprisingly, however, CHX treatment resulted in an                               evidence have established the role of STAT4 as a requisite, direct
accumulation of unphosphorylated STAT4 in cells expressing Hlx                                 transactivator of the IFNG promoter in NK cells after monokine
and empty vector but with more rapid kinetics with Hlx overexpres-                             stimulation.13,14,27 Constitutive STAT4 activation is strongly impli-
sion (Figure 5D). Thus, it appears that Hlx overexpression acceler-                            cated in autoimmune disease, including rheumatoid arthritis and
ates the dephosphorylation of Y693 STAT4 and that its subsequent                               inflammatory bowel disease.6,32 The inhibitory effect of Hlx on
degradation is reliant on new protein synthesis. To confirm these                              STAT4 levels identifies Hlx as a potentially relevant molecular
findings were specific to CHX treatment, we treated Hlx and vector                             target to exploit in autoimmunity. Ultimately, the physiological and
expressing NK-92 with CHX or ethanol carrier for 12 hours or 24                                pathophysiological roles of Hlx in the attenuation of STAT4
hours and repeated the STAT4 Western blot (Figure 5E). Again, we                               transcriptional activity and IFN-␥ production must be determined
found that the inhibitory effect of Hlx overexpression on Y693                                 in vivo.
pSTAT4 levels was insensitive to CHX treatment. Furthermore, we                                    Mechanistically, we found that Hlx promotes the degradation of
confirmed the stabilizing influence of CHX treatment on the levels                             Y693 pSTAT4. Previous work has identified SLIM as an E3
of unphosphorylated STAT4, regardless of Hlx or vector expression.                             ubiquitin ligase with substrate specificity toward Y693 pSTAT4.30
                                                                                               However, this study did not explore the possibility that Y693
                                                                                               pSTAT4 undergoes dephosphorylation before its degradation by
Discussion                                                                                     the proteasome. In our study, through the use of CHX, we
                                                                                               implicated Hlx specifically in the dephosphorylation of pSTAT4
In this study, we found that Hlx is a novel negative regulator of                              Y693 (Figure 4E), and we demonstrated that subsequent degrada-
IFN-␥ production in NK cells after monokine costimulation. The                                 tion of unphosphorylated STAT4 requires new protein synthesis.
delayed kinetics of Hlx protein induction with respect to IFN-␥                                These findings suggest that the process of Y693 pSTAT4 degrada-
itself are consistent with a model in which Hlx serves as a feedback                           tion is complex, consisting of a CHX-insensitive dephosphoryla-
inhibitor of IFN-␥ synthesis in activated CD56bright NK. This                                  tion step and a CHX-sensitive degradation step. The specific role of
Hlx-mediated negative feedback pathway is likely important in                                  Hlx in Y693 pSTAT4 dephosphorylation remains unclear. Although
vivo, given the potentially deleterious effects of unchecked IFN-␥                             the Hlx protein structure lacks sequence homology to known
production on the host.6,7 The process of Hlx protein induction after                          tyrosine phosphatase domains, it is conceivable that Hlx may
monokine costimulation is likely complex. Indeed, Hlx regulation                               recruit a protein tyrosine phosphatase to DNA-bound Y693 pSTAT4
must occur at transcriptional and posttranscriptional levels because                           dimers or tetramers. In addition, it is possible that Hlx may
parallel mRNA and protein expression studies have established                                  associate directly with DNA through its homeodomain and may
long lags between monokine costimulation and the onset of Hlx                                  displace DNA-bound Y693 pSTAT4 multimers, rendering them
mRNA expression (approximately 12 hours; data not shown) and                                   more susceptible to dephosphorylation and degradation. Moreover,
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

2486     BECKNELL et al                                                                                        BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6

we have not fully rejected the hypothesis that Hlx functions as a                  IL-4 is a potent inhibitor of IFN-␥ production by T cells.35
transcription factor and that it is in fact the transcriptional target(s)          Therefore, by decreasing the responsiveness of a CD4⫹ T cell to
of Hlx, translated before CHX and monokine treatment, that trigger                 IL-4 during early TH2 polarization, Hlx may indirectly promote the
Y693 pSTAT4 dephosphorylation after IL-12/IL-18 stimulation.                       production of IFN-␥ by this lymphocyte population. If the effect of
Indeed, it is conceivable that SLIM is one such target of Hlx.                     Hlx on IFN-␥ is attributed entirely to IL-4R␣ regulation, it will be
    Besides identifying a role for STAT4 in Hlx-dependent regula-                  important in the future to determine whether the loss of IL-4R␣
tion of IFN-␥, our work identifies the IFNG promoter as a potential                expression eliminates the increase in IFN-␥ seen on ectopic Hlx
direct target of Hlx transcriptional repression. However, in prelimi-              expression in TH2 cells. In addition, it will be interesting to
nary experiments, we were unable to identify the specific region                   determine whether Hlx influences IL-4R␣ expression by NK cells
required for Hlx repression (data not shown). This difficulty may be               given that IL-4 plays a very different role in NK cells than in T
attributable to a complex requirement for multiple cis-regulatory                  cells, namely to costimulate IFN-␥ production.36,37
motifs throughout the IFNG promoter for Hlx repression of IFN-␥                        Thus, it is likely that Hlx serves different roles in the T and NK
to occur. Therefore, in subsequent work, it will be essential to                   lineages, and the extent of its function may be limited to discrete
distinguish the effects of Hlx on STAT4 from its direct effect, if                 developmental windows. It is not without precedent that transcrip-
any, at the IFNG promoter. Stimulation of Hlx overexpressing                       tion factors perform divergent functions in multiple lineages.
NK-92 cells with IL-2 or IL-15, alone or in combination with                       GATA-3 is one transcriptional regulator shown to have apparently
IL-18, did not influence IFN-␥ mRNA or protein levels compared                     opposing functions in IFN-␥ regulation by NK and CD4⫹ T cells.38
with cells infected with empty retroviral vector (data not shown).                 Alternatively, one can envision multiple scenarios that could
Because STAT4 is activated minimally under these conditions (data                  account for opposing Hlx activities in CD4⫹ T and NK cells, such
not shown), this finding is consistent with a model in which the                   as posttranslational modification of Hlx or unique expression of
effects of Hlx are mediated primarily by STAT4. STAT4 is required                  Hlx interacting proteins in lineage-specific fashion. It is anticipated
for IFN-␥ production in response to IL-12,13,14 making it prohibi-                 that more detailed studies of the Hlx protein and its interactions
tive to explore the consequence of Hlx overexpression in STAT4-                    will elucidate its mechanism of action in T- and NK-cell lineages in
deficient NK cells on IFN-␥ production after IL-12/IL-18 treatment                 the future.
or to determine the effect of Hlx on the activity of IFNG promoter
constructs lacking STAT4 enhancer elements. Given these chal-
lenges, it may be insightful instead to perform a detailed structure–              Acknowledgments
function analysis of Hlx itself so as to identify Hlx mutants that
                                                                                   This work was supported by National Cancer Institute grants P30
maintain IFN-␥ repression without affecting STAT4 levels and
                                                                                   CA16059, CA68458, and CA95426 (M.A.C.) and by National
vice versa.
                                                                                   Institutes of Health grants DK02791 and DK61219 (M.D.B.).
    Our interest in Hlx was prompted by published accounts of its
                                                                                      We thank Dr Howard Young (National Cancer Institute, Freder-
role as a positive regulator of IFN-␥ production in CD4⫹ T
                                                                                   ick, MD) for providing the IFNG promoter luciferase construct; Dr
lymphocytes.10-12 Although the data presented here are not consis-
                                                                                   Michael Grusby (Harvard University) for the generous donation of
tent with this role in NK cells, considerable caution should be
                                                                                   SLIM antisera, and Dr Denis Guttridge (The Ohio State University)
exercised in comparing this work with the T-cell studies given the
                                                                                   and Dr Wei-ping Zheng (University of Rochester) for helpful advice.
distinctly different mechanisms of IFN-␥ production in NK and
CD4⫹ T-cell populations. Although NK cells produce IFN-␥ within
minutes of monokine stimulation,20 CD4⫹ T cells must undergo a                     Authorship
process of polarization, entailing cell division and active chromatin
remodeling at the IFNG locus, before IFN-␥ transcription can                       Contribution: B.B. designed and performed the research and wrote
occur.33 Ectopic expression of Hlx during TH2 polarization of                      the manuscript. T.L.H. performed the research. A.G.F. designed the
CD4⫹ T cells has been shown to promote IFN-␥ production, but                       research. B.W.B. performed the research. J.Y designed the re-
Hlx expression during TH1 polarization and in fully TH1- and                       search. R.T. designed the research. H.C.M. collected the data.
TH2-polarized CD4⫹ T cells reportedly does not influence IFN-␥                     M.L.C. performed the research. M.A. performed the research.
synthesis.10-12 These findings have led to the hypothesis that Hlx                 D.M.B. performed the research. A.L. analyzed the data. D.J.
functions in a discrete window during early TH-cell differentiation                analyzed the data. D.P. designed the research. M.D.B. designed the
to promote IFN-␥ production.10-12                                                  research. M.A.C. designed the research and edited the original and
    Interestingly, recent work from the Zheng34 laboratory clearly                 revised versions of the manuscript.
implicates Hlx as a negative regulator of the IL-4 receptor alpha                     Conflict-of-interest disclosure: The authors declare no compet-
chain (IL-4R␣). Because IL-4 is required for normal TH2 differen-                  ing financial interests.
tiation, Hlx deficiency results in impaired production of TH2                         Correspondence: Michael A. Caligiuri, A458 Starling-
cytokines, and this phenotype can be reversed by ectopic retroviral                Loving Hall, 320 W 10th Ave, Columbus, OH 43210; e-mail:
expression of IL-4R␣.34 Apart from promoting TH2 differentiation,                  michael.caligiuri@osumc.edu.

References
 1. Dorner BG, Smith HR, French AR, et al. Coordi-      3. Schroder K, Hertzog PJ, Ravasi T, Hume DA. In-          5. Dunn GP, Old LJ, Schreiber RD. The immunobiol-
    nate expression of cytokines and chemokines by         terferon-gamma: an overview of signals, mecha-             ogy of cancer immunosurveillance and immu-
    NK cells during murine cytomegalovirus infection.      nisms and functions. J Leukoc Biol. 2004;75:163-           noediting. Immunity. 2004;21:137-148.
    J Immunol. 2004;172:3119-3131.                         189.
                                                                                                                   6. Bouma G, Strober W. The immunological and ge-
 2. Cooper MA, Fehniger TA, Turner SC, et al. Hu-       4. Doffinger R, Dupuis S, Picard C, et al. Inherited          netic basis of inflammatory bowel disease. Nat
    man natural killer cells: a unique innate immuno-      disorders of IL-12- and IFN␥-mediated immunity:
                                                                                                                      Rev Immunol. 2003;3:521-533.
    regulatory role for the CD56bright subset. Blood.      a molecular genetics update. Mol Immunol. 2002;
    2001;7:3146-3151.                                      38:903-909.                                             7. Shimozato O, Ortaldo JR, Komschlies KL, Young
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

BLOOD, 15 MARCH 2007 䡠 VOLUME 109, NUMBER 6                                                                       Hlx INHIBITS IFN-␥ PRODUCTION BY NK CELLS                 2487

      HA. Impaired NK cell development in an IFN-              18. Trotta R, Parihar R, Yu J, et al. Differential ex-         regulation of Stat4 activation and IFN-gamma
      gamma transgenic mouse: aberrantly expressed                 pression of SHIP1 in CD56bright and CD56dim NK             gene expression by IFN-alpha and IL-12. Eur
      IFN-gamma enhances hematopoietic stem cell                   cells provides a molecular basis for distinct func-        J Immunol. 2001;31:2236-2245.
      apoptosis and affects NK cell differentiation. J Im-         tional responses to monokine costimulation.           28. Barbulescu K, Becker C, Schlaak JF, et al. IL-12
      munol. 2002;168:1746-1752.                                   Blood. 2005;105:3011-3018.                                and IL-18 differentially regulate the transcriptional
 8.   Glimcher LH, Townsend MJ, Sullivan BM, Lord              19. Uzel G, Frucht DM, Fleisher TA, Holland SM. De-           activity of the human IFN-gamma promoter in pri-
      GM. Recent developments in the transcriptional               tection of intracellular phosphorylated STAT-4 by         mary CD4⫹ T lymphocytes. J Immunol. 1998;
      regulation of cytolytic effector cells. Nat Rev Im-          flow cytometry. Clin Immunol. 2001;100:270-276.           160:3642-3647.
      munol. 2004;4:900-911.                                                                                             29. Wang KS, Zorn E, Ritz J. Specific down-regula-
                                                               20. Fehniger TA, Shah MH, Turner MJ, et al. Differen-
 9.   Bates MD, Wells JM, Venkatesh B. Comparative                 tial cytokine and chemokine gene expression by            tion of interleukin-12 signaling through induction
      genomics of the Hlx homeobox gene and protein:               human NK cells following activation with IL-18 or         of phospho-STAT4 protein degradation. Blood.
      conservation of structure and expression from                IL-15 in combination with IL-12: implications for         2001;97:3860-3866.
      fish to mammals. Gene. 2005;352:45-56.                       the innate immune response. J Immunol. 1999;          30. Tanaka T, Soriano MA, Grusby MJ. SLIM is a
10.   Mullen AC, Hutchins AS, High FA, et al. Hlx is in-           162:4511-4520.                                            nuclear ubiquitin E3 ligase that negatively regu-
      duced by and genetically interacts with T-bet to         21. Dignam JD, Martin PL, Shastry BS, Roeder RG.              lates STAT signaling. Immunity. 2005;22:729-736.
      promote heritable T(H)1 gene induction. Nat Im-              Eukaryotic gene transcription with purified com-      31. Maecker H, Varfolomeev E, Kischkel F, et al.
      munol. 2002;3:652-658.                                       ponents. Methods Enzymol. 1983;101:582-598.               TWEAK attenuates the transition from innate to
11.   Zheng WP, Zhao Q, Zhao X, et al. Up-regulation                                                                         adaptive immunity. Cell. 2005;123:931-944.
                                                               22. Bream JH, Hodge DL, Gonsky R, et al. A distal
      of Hlx in immature Th cells induces IFN-gamma
                                                                   region in the interferon-gamma gene is a site of      32. Kaplan MH. STAT4: a critical regulator of inflam-
      expression. J Immunol. 2004;172:114-122.
                                                                   epigenetic remodeling and transcriptional regula-         mation in vivo. Immunol Res. 2005;31:231-242.
12.   Martins GA, Hutchins AS, Reiner SL. Transcrip-               tion by interleukin-2. J Biol Chem. 2004;279:
      tional activators of helper T cell fate are required                                                               33. Bird JJ, Brown DR, Mullen AC, et al. Helper T cell
                                                                   41249-41257.                                              differentiation is controlled by the cell cycle. Im-
      for establishment but not maintenance of signa-
      ture cytokine expression. J Immunol. 2005;175:           23. Gong JH, Maki G, Klingemann HG. Characteriza-             munity. 1998;9:229-237.
      5981-5985.                                                   tion of a human cell line (NK-92) with phenotypi-     34. Mikhalkevich N, Becknell B, Caligiuri MA, Bates
                                                                   cal and functional characteristics of activated           MD, Harvey R, Zheng WP. Responsiveness of
13.   Kaplan MH, Sun YL, Hoey T, Grusby MJ. Im-
                                                                   natural killer cells. Leukemia. 1994;8:652-658.           naive CD4 T cells to polarizing cytokine deter-
      paired IL-12 responses and enhanced develop-
      ment of Th2 cells in Stat4-deficient mice. Nature.       24. Di Noto R, Pane F, Camera A, et al. Characteriza-         mines the ratio of Th1 and Th2 cell differentiation.
      1996;382:174-177.                                            tion of two novel cell lines, DERL-2 (CD56⫹/              J Immunol. 2006;176:1553-1560.
                                                                   CD3⫹/Tcry5⫹) and DERL-7 (CD56⫹/CD3⫺/                  35. Jacobson NG, Szabo SJ, Weber-Nordt RM, et al.
14.   Thierfelder WE, van Deursen JM, Yamamoto K,
                                                                   TCR␥␦⫺), derived from a single patient with               Interleukin 12 signaling in T helper type 1 (Th1)
      et al. Requirement for Stat4 in interleukin-12-
                                                                   CD56⫹ non-Hodgkin’s lymphoma. Leukemia.                   cells involves tyrosine phosphorylation of signal
      mediated responses of natural killer and T cells.
                                                                   2001;15:1641-1649.                                        transducer and activator of transcription (Stat)3
      Nature. 1996;382:171-174.
                                                               25. Fedele M, Pentimalli F, Baldassarre G, et al.             and Stat4. J Exp Med. 1995;181:1755-1762.
15.   Yu J, Wei M, Becknell B, et al. Pro- and antiin-
      flammatory cytokine signaling: reciprocal antago-            Transgenic mice overexpressing the wild-type          36. Morris SC, Orekhova T, Meadows MJ, Heidorn
      nism regulates interferon-gamma production by                form of the HMGA1 gene develop mixed growth               SM, Yang J, Finkelman FD. IL-4 induces in vivo
      human natural killer cells. Immunity. 2006;24:575-           hormone/prolactin cell pituitary adenomas and             production of IFN-gamma by NK and NKT cells.
      590.                                                         natural killer cell lymphomas. Oncogene. 2005;            J Immunol. 2006;176:5299-5305.
                                                                   24:3427-3435.                                         37. Bream JH, Curiel RE, Yu CR, et al. IL-4 synergis-
16.   Becknell B, Trotta R, Yu J, et al. Efficient infection
      of human natural killer cells with an EBV/retroviral     26. Gonsky R, Deem RL, Young HA, Targan SR. CD2               tically enhances both IL-2- and IL-12-induced
      hybrid vector. J Immunol Methods. 2005;296:115-              mediates activation of the IFN-gamma intronic             IFN-gamma expression in murine NK cells.
      123.                                                         STAT binding region in mucosal T cells. Eur J Im-         Blood. 2003;102:207-214.
17.   Hentsch B, Lyons I, Li R, et al. Hlx homeo box               munol. 2003;33:1152-1162.                             38. Samson SI, Richard O, Tavian M, et al. GATA-3
      gene is essential for an inductive tissue interac-       27. Matikainen S, Paananen A, Miettinen M, et al.             promotes maturation, IFN-gamma production,
      tion that drives expansion of embryonic liver and            IFN-alpha and IL-18 synergistically enhance IFN-          and liver-specific homing of NK cells. Immunity.
      gut. Genes Dev. 1996;10:70-79.                               gamma production in human NK cells: differential          2003;19:701-711.
From www.bloodjournal.org by guest on May 20, 2015. For personal use only.

                                             2007 109: 2481-2487
                                             doi:10.1182/blood-2006-10-050096 originally published
                                             online November 16, 2006

Hlx homeobox transcription factor negatively regulates interferon-γ
production in monokine-activated natural killer cells
Brian Becknell, Tiffany L. Hughes, Aharon G. Freud, Bradley W. Blaser, Jianhua Yu, Rossana Trotta,
Hsiaoyin C. Mao, Marie L. Caligiuri de Jesús, Mohamad Alghothani, Don M. Benson, Jr, Amy
Lehman, David Jarjoura, Danilo Perrotti, Michael D. Bates and Michael A. Caligiuri

Updated information and services can be found at:
http://www.bloodjournal.org/content/109/6/2481.full.html
Articles on similar topics can be found in the following Blood collections
  Gene Expression (1086 articles)
  Immunobiology (5303 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.
You can also read