You are on page 1of 11

From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

IMMUNOBIOLOGY

Regulation of human NK-cell cytokine and chemokine production by target cell


recognition
Cyril Fauriat,1 Eric O. Long,2 Hans-Gustaf Ljunggren,1 and Yenan T. Bryceson1
1Center
for Infectious Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden; and 2Laboratory of
Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD

Natural killer (NK)–cell recognition of in- of predominately proinflammatory cyto- CD56bright NK cells. The present data dem-
fected or neoplastic cells can induce cyto- kines and chemokines. Release of chemo- onstrate how specific target cell ligands
toxicity and cytokine secretion. So far, it kines MIP-1␣, MIP-1␤, and RANTES was dictate qualitative and temporal aspects
has been difficult to assess the relative induced within 1 hour of stimula- of NK-cell cytokine and chemokine re-
contribution of multiple NK-cell activa- tion, whereas release of TNF-␣ and IFN-␥ sponses. Conceptually, the results point
tion receptors to cytokine and chemokine occurred later. Engagement of CD16, 2B4, to CD56dim NK cells as an important
production upon target cell recognition. or NKG2D sufficed for chemokine re- source of cytokines and chemokines upon
Using Drosophila cells expressing li- lease, whereas induction of TNF-␣ and recognition of aberrant cells, producing
gands for the NK-cell receptors LFA-1, IFN-␥ required engagement of additional graded responses depending on the mul-
NKG2D, DNAM-1, 2B4, and CD16, we stud- receptors. Remarkably, our results re- tiplicity of activating receptors engaged.
ied the minimal requirements for secre- vealed that, upon target cell recognition, (Blood. 2010;115:2167-2176)
tion by freshly isolated, human NK cells. CD56dim NK cells were more prominent
Target cell stimulation induced secretion cytokine and chemokine producers than

Introduction
Natural killer (NK) cells respond directly to infected or neoplastic and express high levels of perforin. In contrast, CD56bright NK cells
cells through engagement of a multitude of germline-encoded express no or low levels of CD16, exclusively express the
receptors by ligands on target cells.1-3 Beside their ability to kill inhibitory receptor CD94/NKG2A, and have 10-fold lower per-
aberrant cells, NK cells are also critical components of the innate forin expression than CD56dim NK cells.25-27 Because of these and
immune response by virtue of their capacity to produce a variety of other findings, CD56dim and CD56bright NK-cell subsets are consid-
cytokines and chemokines.4-6 Murine models have demonstrated a ered to be developmentally distinct and to occupy different
dependence on NK cell–derived cytokines in early responses to functional niches.12,28-30
obligate intracellular parasites such as Listeria, Toxoplasma, and Human NK-cell responses to exogenous cytokines have been
Leishmania and in resistance to cytomegalovirus infection.7-10 In extensively studied.21 In contrast, relatively less is known with
many of these systems, NK cells respond to cues from sentinel respect to NK-cell cytokine and chemokine production upon target
immune cells, including dendritic cells, macrophages, and pathogen- cell recognition. For example, the full spectrum of cytokines
infected tissue cells.11-13 These cues are communicated by release released by freshly isolated, resting NK cells upon target cell
of cytokines, including interleukin-1 (IL-1), IL-10, IL-12, IL-15, recognition has not been fully characterized. Furthermore, the
and IL-18.14 Thus, secondary to triggering of innate immune cells minimal requirements for induction of cytokine secretion upon
by pattern recognition receptors, NK cells can relay and amplify engagement of specific ligands on target cells are not known. To
cytokine signals. Among the most prominent cytokines produced understand how NK cells may contribute to, and maybe even act as
by NK cells are tumor necrosis factor-␣ (TNF-␣) and interferon ␥ primary initiators of, immune responses upon target cell recogni-
(IFN-␥). Moreover, NK cells have been reported to secrete several tion, studies on how receptor-ligand interactions dictate qualitative
other factors, including immunoregulatory cytokines such as IL-5, and temporal aspects of cytokine and chemokine secretion are
IL-10, IL-13, the growth factor GM-CSF, and the chemokines important. Here, we have set out to study in detail cytokine and
MIP-1␣, MIP-1␤, IL-8, and RANTES.15-22 chemokine production by human peripheral blood NK cells upon
In humans, NK cells are usually defined as CD3–CD56⫹ cells,23 target cell recognition.
and can be further subdivided based on CD56 expression. Typi- To overcome the complexity in receptor-ligand interactions
cally, CD56dim NK cells constitute the majority (90%) of peripheral between NK cells and target cells, we have developed a reconstitu-
blood NK cells, whereas CD56bright NK cells are more abundant in tion system using Drosophila cells as targets.31 A notable advantage
secondary lymphoid tissues.14,24 CD56dim NK cells express high of such a system is that cytokine and chemokine secretion by
levels of the low-affinity Fc receptor CD16, display variegated primary, unmanipulated NK cells can be studied in the context of
expression of several types of inhibitory receptors for MHC class I, specific receptor-ligand interactions. This system has recently

Submitted August 13, 2009; accepted November 3, 2009. Prepublished online The online version of this article contains a data supplement.
as Blood First Edition paper, December 1, 2009; DOI 10.1182/blood-2009-08-
The publication costs of this article were defrayed in part by page charge
238469.
payment. Therefore, and solely to indicate this fact, this article is hereby
A Inside Blood analysis of this article appears at the front of this issue. marked ‘‘advertisement’’ in accordance with 18 USC section 1734.

BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11 2167


From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2168 FAURIAT et al BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11

revealed cooperation among NK-cell receptors for discrete events A NK cells


in cytotoxicity, including NK-cell cytolytic granule polarization 5000
NK cells + K562 cells
4000
and exocytosis.32,33

Secretion (pg/mL)
3000
Here, we addressed how specific engagement of the receptors 2000
1000
NKG2D (CD314), DNAM-1 (CD226), 2B4 (CD244), LFA-1 250
(CD11a/CD18), and CD16, or combinations thereof, regulate 200

cytokine and chemokine production by freshly isolated, resting 150


100
human NK cells. The data provide insight into the regulation of
50
cytokine and chemokine secretion by different NK-cell subsets 0
upon target cell recognition.

C CL /M P-1

S
C XC 8/ in
L1 9/M -8
G /IP G
-C 0
IL SF

-2
-4
-5
-6
IL IL- 7
2p 0
40
IL 3
IL 5
IF 7
C L2/ NF γ
C M -α

C L 5 / /M 1 α
6 N β

IL 2Rα
IL α


β
C T N-

M -1

-
-1 1

-1
-1
-1
L2 A 1

R
-1
C /Eo TE
C CL tax

0 I
XC L IL

IL
IL
IL
IL
IL

N
C R IP
C 4 IP

-1

IL
IF

C L3 C

-
X
C
B 10000
Methods
Cells
1000

Secretion (pg/mL)
Human NK cells were isolated from peripheral blood by negative selection
(NK-cell isolation kit; Miltenyi Biotec). Approval was obtained from the
100
Regional Ethics Review Board for the use of peripheral blood mononuclear
IFN-γ
cells from healthy donors. CD56dim and CD56bright NK-cell subsets were
TNF-α
sorted based on CD56 expression by flow cytometry (FACSAria; BD
10 MIP-1α
Biosciences). Freshly isolated NK cells were maintained in complete
MIP-1β
medium (RPMI 1640 supplemented with 10% fetal bovine serum and 2mM
RANTES
L-glutamine; all from Invitrogen). NK-cell populations contained at least 1
99% CD3⫺CD56⫹ cells and were used within 2 days of isolation. The cell 0 1 2 3 4 5 6 7 8 9 10 11 12
line K562 (ATCC) was maintained in complete medium. The transfection Time (hrs)
and maintenance of Drosophila S2 cells has been previously described and Figure 1. Profile and kinetics of NK-cell secretion upon interaction with K562
expression of the ligands for NK receptors was monitored prior to cells. (A) Resting NK cells were incubated alone or with K562 cells for 6 hours at
experiments.31,33 A rabbit serum raised against S2 cells and used to coat 37°C. Supernatants were harvested, and the concentrations of indicated cytokines
S2 cells with IgG has been described.32 and chemokines were determined by a multiplex immunoassay. Values represent
mean ⫾ SD of 8 different donors. (B) NK cells were mixed with K562 cells and
incubated at 37°C. Supernatants were harvested at different time points, as
Antibodies and fluorescent reagents indicated, and the concentrations of IFN-␥, TNF-␣, MIP-1␤, MIP-1␤, and RANTES
were determined by a multiplex immunoassay. Values represent mean ⫾ SD of
Fluorochrome-conjugated monoclonal antibodies (mAbs) used for flow 5 different donors.
cytometry were anti-CD3 (clone UCHT1; Dako), anti-CD56 (clone NCAM
16.2; BD Biosciences), anti-CD107a (clone H4A3; BD Biosciences),
anti–MIP-1␣ (clone 93342; R&D Systems), anti–MIP-1␤ (clone D21- paraformaldehyde (Sigma-Aldrich) in phosphate-buffered saline, permeabil-
1351; BD Biosciences), anti–TNF-␣ (clone MP6-XT22; eBioscience), and ized, and stained intracellularly with fluorochrome-conjugated mAbs to
anti–IFN-␥ (clone 25723.11; BD Biosciences). Streptavidin-conjugated cytokines and chemokines. Finally, cells were washed and analyzed on a
Qdot 605 was used to detect biotinylated anti-CD107a and a fixable cell CyAn ADP LX 9-color flow cytometer (Dako).34 Data were analyzed with
viability dye (LIVE/DEAD 405 nm) was used to exclude dead cells from FlowJo 8.6 software (TreeStar). Pie charts were generated with SPICE
the analysis (both from Invitrogen). software, Version 4.1 (M. Roederer, Vaccine Research Center, National
Institute of Allergy and Infectious Diseases, National Institutes of Health).
Cytokine secretion measurements
Statistical analysis
Resting NK cells (2 ⫻ 105) were washed twice in and mixed with 4 ⫻ 105
S2 cells or K562 cells in 200 ␮L of complete medium. In experiments with All analyses were performed with GraphPad software (GraphPad Soft-
sorted cells, 1 ⫻ 105 CD56dim or CD56bright NK cells were mixed with ware). Statistical analyses of minimal requirements and comparisons
1 ⫻ 105 S2 cells. Cells were incubated for the indicated time at 37°C in between CD56dim and CD56bright responsiveness were performed using a
5% CO2. Thereafter, supernatants were collected and stored at ⫺20°C 2-way analysis of variance (nonparametric) with Bonferroni posttest.
pending measurement. The concentrations of cytokines were quantified by
a multiplex immunoassay (Luminex 100 IS; Invitrogen) using a kit that
detected 25 different cytokines (Biosource; listed in Figure 1A). To address Results
the kinetics of secretion of select cytokines, 5 simplexes (for IFN-␥, TNF-␣,
RANTES, MIP-1␣, and MIP-1␤) were mixed according to the manufactur- Secretion profile of resting NK cells upon interaction with K562
er’s instructions (Biosource). cells

Intracellular staining of cytokines We first set out to determine the factors released by freshly isolated
peripheral blood NK cells upon short incubations with susceptible
Resting NK cells (2 ⫻ 105) were added to 4 ⫻ 105 S2 cells in 200 ␮L of K562 target cells. Resting NK cells were mixed with K562 cells for
complete medium. IL-12, IL-15, and IL-18 (all from Peprotech) were added 6 hours. Supernatants were harvested and the concentration of
in some experiments. Cells were incubated for 1 hour at 37°C in 5% CO2.
25 different soluble factors was determined by a multiplex
Thereafter, Brefeldin A (GolgiPlug; Becton Dickinson) was added to the
cultures, which were incubated for 5 more hours. After 6 hours of incuba-
immunoassay (Figure 1A). As anticipated, stimulation of NK cells
tion, the cells were spun down and resuspended in 50 ␮L of staining buffer by K562 cells induced IFN-␥ and TNF-␣ secretion. In addition,
(phosphate-buffered saline supplemented with 2% fetal bovine serum and stimulation induced ample secretion of several chemokines, includ-
2mM ethylenediaminetetraacetic acid) added fluorochrome-conjugated ing MCP-1 (CCL2), MIP-1␣ (CCL3), MIP-1␤ (CCL4), RANTES
mAbs for surface staining. Thereafter, cells were washed, fixed with 2% (CCL5), IL-8 (CXCL8), and IP-10 (CXCL10). Low levels of
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11 NK-CELL RESPONSES TO TARGET CELL RECOGNITION 2169

Figure 2. Minimal requirements for receptor-ligand 300


interactions for secretion of cytokines and chemo- IFN-γ
kines by NK cells. Resting NK cells were mixed with *
S2 cells expressing ligands for NK-cell receptors, as 200
indicated, and incubated for 6 hours at 37°C. For some
stimulations, S2 cells were preincubated with diluted
*
100 *
anti–S2 cell serum (⫹ IgG). Supernatants were har-
vested, and the concentrations of cytokines and chemo-
kines were determined by a multiplex immunoassay. 0
Values represent mean ⫾ SD of 5 different donors. Data 800
are representative of 3 independent experiments. For TNF-α
clarity, selected statistical analyses are indicated. 600 *
*P ⬍ .05, ***P ⬍ .001.
400 *
200

3500
Secretion (pg/mL)
3000 MIP-1α
2500
2000 ***
*
1500
1000
*
500
0
12500
MIP-1β
10000 ***
***
7500

5000
***
2500 ***
0
2500
RANTES
2000

1500 ***
1000
***

500

0 – S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2
ICAM1 CD48 ULBP1 CD155 ICAM1 ICAM1 ICAM1 CD48 ICAM1 ICAM1 CD48 ULBP1 CD155 ICAM1 ICAM1 ICAM1 CD48 ICAM1
CD48 ULBP1 CD155 ULBP1 CD48 CD48 ULBP1 CD155 ULBP1 CD48
ULBP1 ULBP1
+ + + + + + + + + +
IgG IgG IgG IgG IgG IgG IgG IgG IgG IgG

chemokines MIP-1␣, MIP-1␤, and RANTES were also detected in Thus, interactions with a susceptible target cell line can induce a
supernatants from NK cells incubated without target cells, suggest- profoundly proinflammatory profile of chemokine and cytokine
ing a degree of constitutive secretion. However, incubation with secretion. Moreover, kinetic studies revealed a rapid release of
K562 cells resulted in a 44-, 181-, and 6.5-fold increase in secretion chemokines, relative to cytokine secretion.
of MIP-1␣, MIP-1␤, and RANTES, respectively, compared with
supernatants from NK cells alone. Notably, stimulation of NK cells Minimal receptor engagement requirements for secretion of
by K562 cells also induced release of soluble IL-2R␣ (CD25). proinflammatory cytokines and chemokines by resting NK cells
GM-CSF, IL-5, and IL-13 were weakly or not secreted after
Having identified soluble factors that were secreted by resting NK
interaction of NK cells with K562 cells, contrasting previous
cells upon interaction with a susceptible target cell, the contribution
studies using cytokine-cultured NK cells.15,17 Low levels of IL-1␤,
of individual receptor-ligand interactions to cytokine and chemo-
IL-6, IL-7, IL-10, IL-12p40, IFN-␣, and MIG (CXCL9) secretion
kine secretion was assessed. Resting NK cells were mixed with
were measured after target cell stimulation. Secretion of IL-2, IL-4,
Drosophila Schneider 2 (S2) cells expressing ICAM-1 (CD54),
IL-5, IL-13, IL-15, IL-17, and eotaxin (CCL26) was not detected. CD48, ULBP1, and CD155 (which bind receptors LFA-1, 2B4,
Next, the temporal profile of cytokine and chemokine secretion NKG2D, and DNAM-1, respectively), or combinations thereof,
was analyzed (Figure 1B). Chemokine secretion could be detected and incubated for 6 hours. In these experiments, the concentrations
within 1 hour of mixing NK cells with K562 cells. In contrast, of IFN-␥, TNF-␣, MIP-1␣, MIP-1␤, and RANTES were deter-
TNF-␣ and, in particular, IFN-␥ secretion occurred later. After mined using a multiplex immunoassay (Figure 2). Secretion of
6 hours of incubation, the highest levels of secreted cytokines were TNF-␣ required coexpression of CD48 and ULBP1 on S2 cells,
measured. Thereafter the concentrations of chemokines remained whereas IFN-␥ required combined expression of ICAM-1, CD48,
stable throughout the time course, whereas IFN-␥ and TNF-␣ and ULBP1. In contrast, expression of CD48 was sufficient to
concentrations, if anything, decreased. induce secretion of chemokines MIP-1␣, MIP-1␤, and RANTES.
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2170 FAURIAT et al BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11

Coexpression of CD48 and ULBP1 on S2 cells did not enhance chemokines upon such stimulations. For example, CD48 expres-
secretion of chemokines compared with S2 cells expressing CD48 sion on S2 cells induced MIP-1␣ production in 9% (⫾ 6%) of
alone. However, expression of ICAM-1, in addition to CD48 and CD56bright NK cells, as opposed to 43% (⫾ 7%) of CD56dim NK
ULBP1, on S2 cells could augment chemokine secretion, as cells. Thus, strikingly, upon target cell recognition CD56dim NK
observed for IFN-␥. To engage CD16 on NK cells, S2 cells were cells were more prominent chemokine and cytokine producers than
preincubated with an anti–S2 cell polyclonal serum.32 IgG coating CD56bright NK cells. Strengthening this notion, greater proportions
of S2 cells was sufficient to induce significant TNF-␣ secretion, but of CD56dim NK cells than CD56bright NK cells produced IFN-␥,
much less IFN-␥ secretion (Figure 2). In regard to IFN-␥ secretion, TNF-␣, MIP-1␣, and MIP-1␤ upon K562 cell stimulation (Figure
expression of ICAM-1 or CD48 on S2 cells augmented IgG- 3B). Moreover, experiments comparing cytokine and chemokine
mediated secretion. Chemokine secretion was readily induced by secretion from equal numbers of sorted CD56dim and CD56bright NK
IgG-coated S2 cells. Expression of ICAM-1 or CD48 on S2 cells cells separately incubated with K562 cells or various S2 cell
could augment IgG-induced chemokine secretion. transfectants also showed greater secretion by CD56dim NK cells
Thus, in the absence of IgG, engagement of 2B4 was sufficient (Figure 3C and data not shown).
to induce chemokine secretion, whereas stronger signals evoked by Ig-coated S2 cells were sufficient to induce TNF-␣ and IFN-␥
2B4 and NKG2D were required for secretion of TNF-␣ or IFN-␥. production by CD56dim, but not CD56bright, NK cells (supplemental
Coengagement of LFA-1 augmented cytokine and chemokine Figures 2-3). However, Ig-coated S2 cells induced 29% (⫾ 11%)
secretion induced by 2B4 and NKG2D. CD16 engagement could MIP-1b⫹ CD56bright NK cells, demonstrating that engagement of
induce secretion of chemokines and cytokines. CD16-mediated CD16 can elicit responses by a subset of CD56bright NK cells
secretion of IFN-␥ and chemokine secretion, but not TNF-␣, was (supplemental Figure 3).
augmented by LFA-1 coengagement. Reports defining CD56bright NK cells as the major source of NK
Longer time points were also assessed and secretion patterns cell–derived cytokines have relied on exogenous cytokines for
resembled that induced by K562 cell stimulation (supplemental stimulation.26 To ensure that the low cytokine response of CD56bright
Figure 1, available on the Blood website; see the Supplemental NK cells upon target cell recognition was not an artifact of our
Materials link at the top of the online article). Notable features were experimental procedure, we evaluated the response of freshly
a slight induction of chemokines by S2 cells expressing ULBP1, as isolated NK cells to 24 hours of stimulation with cytokines IL-12,
previously described with cytokine-cultured NK cells,35 and that IL-15, or IL-18, or combinations thereof (Figure 3D). These
costimulation could accelerate chemokine secretion. cytokines induce strong IFN-␥ secretion by CD56bright NK cells in
A range of S2 cell combinations engaging several activating 72-hour assays.26 In agreement with other reports, a greater
receptors was assessed for the 25 factors listed in Figure 1A. frequency of CD56bright NK cells, relative to CD56dim NK cells,
Corroborating findings with NK-cell stimulation by K562 cells, produced IFN-␥ and TNF-␣ in response to stimulation by exog-
profiles of secreted cytokines and chemokines were proinflamma- enous cytokines. In comparison, production of MIP-1␣ and MIP-1␤
tory (data not shown). Thus, NK-cell activation by CD16, 2B4, and was equal or greater in CD56dim NK cells. Next, we tested whether
NKG2D, in addition to costimulation by DNAM-1 and LFA-1, the greater frequency of cytokine-producing CD56bright NK cells
induced secretion of proinflammatory, but not immunoregulatory, after 24 hours of stimulation could reflect temporal differences in
cytokines. responsiveness between the 2 subsets. After 6 hours of stimulation,
somewhat higher frequencies of CD56bright NK cells, relative to
CD56dim NK cells are major producers of cytokines and CD56dim NK cells, produced IFN-␥ and TNF-␣ in response to
chemokines upon target cell recognition IL-12 plus IL-18 (Figure 4). Remarkably, as with 24 hours of
Whereas analysis of supernatants from cell-mixing experiments stimulation, production of MIP-1␣ and MIP-1␤ was, if anything,
identified the predominant chemokines and cytokines secreted by more pronounced in CD56dim NK cells relative to CD56bright NK
freshly isolated NK cells in response to target cell recognition, a cells. Moreover, after 24 hours of stimulation with K562 cells,
different approach was required to identify the phenotypic features the frequency of cytokine- and chemokine-producing CD56bright
of the cytokine- and chemokine-producing cells, to quantify the NK cells was still lower than that of CD56dim NK cells (data not
frequency of responding cells, and to examine to what extent shown). Together, these results suggest that CD56bright NK cells
individual cells could produce multiple soluble factors. To this end, are not inherently slower to respond to target cell recognition.
multicolor flow cytometry was performed. Production of IFN-␥, Rather, CD56dim NK cells excel in cytokine and chemokine
TNF-␣, MIP-1␣, and MIP-1␤ was compared in CD56dim and production upon recognition of target cells. Furthermore, a
CD56bright NK-cell subsets after 6 hours of incubation with differ- greater proportion of the CD56dim than the CD56bright subset of
ent S2 cells (Figure 3A). Coexpression of ICAM-1, CD48, and NK cells produced chemokines in response to exogenous
ULBP1 on S2 cells was required to induce IFN-␥ production by cytokine stimulation.
CD56dim NK cells. Expression of CD48 on S2 cells was sufficient
IL-12 plus IL-18 potentiate cytokine secretion upon target cell
to induce TNF-␣ production. Interestingly, despite LFA-1, 2B4,
recognition by CD56dim NK cells
and NKG2D being expressed at similar levels on different NK-cell
subsets (Bryceson et al36; data not shown), very few CD56bright NK Studies have shown that cytokines, such as IL-2, IL-12, IL-15, and
cells expressed IFN-␥ or TNF-␣ after incubation with target cells IL-21, can enhance IFN-␥ and chemokine secretion induced by
expressing ligands for these receptors. Expression of CD48 or target cell recognition.20,22,37-39 Therefore, we examined how exog-
ULBP1 on S2 cells was sufficient to induce MIP-1␣ and MIP-1␤ enous cytokine stimulation by IL-12 plus IL-18 influenced NK-cell
production by CD56dim NK cells (Figure 3A and supplemental production of IFN-␥, TNF-␣, MIP-1␣, and MIP-1␤ upon interac-
Figure 2), corroborating results quantifying chemokine secretion. tion with K562 cells (Figure 4). Addition of IL-12 and IL-18
CD56bright NK cells did produce some MIP-1␣ and MIP-1␤ upon augmented the frequency of IFN-␥–producing CD56dim NK cells
stimulation by S2 cells coexpressing CD48 and ULBP1. However, after 6 hours of incubation with K562 cells. Interestingly, IL-12
a consistently higher frequency of CD56dim NK cells produced and IL-18 did not significantly increase MIP-1␣ and MIP-1␤
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11 NK-CELL RESPONSES TO TARGET CELL RECOGNITION 2171

A
30
CD56dim NK cells
IFN-γ TNF-α ***
20 CD56bright NK cells
***
Positive cells (%) ***
10 ** *
0

100 MIP-1α MIP-1β


80 *** ***
60 *** ***
40 **
20 *
0 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2 S2
CD48 CD48 ICAM1 CD48 CD48 ICAM1 CD48 CD48 ICAM1 CD48 CD48 ICAM1
ULBP1 CD48 ULBP1 CD48 ULBP1 CD48 ULBP1 CD48
ULBP1 ULBP1 ULBP1 ULBP1

B 100
MIP-1β
Positive cells (%)

IFN-γ TNF-α MIP-1α ***


80 ***
***
60 ***
40 **
*
20

0
– K562 – K562 – K562 – K562 – K562 – K562 – K562 – K562
C
500 500 1500 5000
Secretion (pg/mL)

IFN-γ TNF-α MIP-1α MIP-1β


400 400 4000
1000
300 300 3000

200 200 2000


500
100 100 1000

0
– K562 – K562 – K562 – K562 – K562 – K562 – K562 – K562

D
100
IFN-γ TNF-α MIP-1α MIP-1β
Positive cells (%)

80

60

40

20

0 – – – – – – – –
IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

IL-12
IL-15
IL-18
IL-12+IL-15
IL-15+IL-18
IL-12+IL-18

Figure 3. CD56dim NK cells produce cytokines and chemokines upon target cell recognition. Resting NK cells were mixed with S2 cells expressing ligands for NK-cell
receptors (A) or K562 cells (B), as indicated, and incubated for 6 hours at 37°C. After stimulation, the cells were surface stained with fluorochrome-conjugated anti-CD56 mAb,
fixed, permeabilized, and stained intracellularly with fluorochrome-conjugated mAbs to cytokines and chemokines. The percentage of CD56dim or CD56bright NK cells producing
IFN-␥, TNF-␣, MIP-1␣, and MIP-1␤, as indicated, was determined by flow cytometry. Values represent mean ⫾ SD of at least 6 different donors. (C) Sorted CD56dim or
CD56bright NK cells were incubated alone or with K562 cells for 6 hours at 37°C. Supernatants were harvested, and the concentrations of cytokines and chemokines were
determined by a multiplex immunoassay. Values represent mean ⫾ SD of 5 different donors. (D) Resting NK cells were incubated alone or stimulated with 10 ng/mL IL-12,
100 ng/mL IL-15, or 100 ng/mL IL-18, or combinations thereof, for 24 hours at 37°C. After stimulation, the cells were surface stained with fluorochrome-conjugated anti-CD56
mAb, fixed, permeabilized, and stained intracellularly with fluorochrome-conjugated mAbs to cytokines and chemokines. The percentage of CD56dim or CD56bright NK cells
producing IFN-␥, TNF-␣, MIP-1␣, and MIP-1␤, as indicated, was determined by flow cytometry. Values represent mean ⫾ SD of 5 different donors. For clarity, selected
statistical analyses are indicated. *P ⬍ .05, **P ⬍ .01, ***P ⬍ .001.

production by CD56dim NK cells in response to K562 cells. K562 pathways for cytokine and chemokine production in CD56bright and
cells alone did not induce cytokine and chemokine production by CD56dim NK-cell subsets.
CD56bright NK cells, and addition of IL-12 and IL-18 did not
increase the frequencies of cytokine- and chemokine-producing Costimulation signals lower the threshold for CD16-induced
CD56bright NK cells relative to IL-12 plus IL-18 alone. In conclu- NK-cell cytokine and chemokine production
sion, exogenous cytokines potentiated cytokine production by
CD56dim NK cells in response to K562 cells. In such settings, Next, we analyzed how the strength of stimulation through
chemokine production by CD56dim NK cells could be ascribed increased CD16 engagement affected the frequency of cytokine-
mainly to target cell recognition, whereas cytokine and chemokine and chemokine-producing CD56dim NK cells (Figure 5). By
production by CD56bright NK cells could be ascribed to exogenous varying the concentration of anti-S2 serum with which S2 cells
cytokines. These data suggest different requirements and signaling were coated, the density of ligands for CD16 could be modulated
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2172 FAURIAT et al BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11

CD56dim NK cells Figure 4. Costimulation of NK cells by exogenous


CD56bright NK cells cytokines enhances cytokine production by CD56dim
100 NK cells upon interaction with K562 cells. Resting NK
IFN-γ TNF-α MIP-1α MIP-1β cells were incubated alone or stimulated with cytokines
***
Positive cells (%)

80 IL-12 and IL-18 (10 ng/mL and 100 ng/mL, respectively)


for 6 hours at 37°C with or without K562 cells. After stimula-
*** ***
60 tion, the cells were surface stained with fluorochrome-
* conjugated anti-CD56 mAb, fixed, permeabilized, and
***
40 stained intracellularly with fluorochrome-conjugated mAbs
*** * **
* * to cytokines and chemokines. The percentage of CD56dim
20 or CD56bright NK cells producing IFN-␥, TNF-␣, MIP-1␣,
and MIP-1␤, as indicated, was determined by flow cytom-
0 etry. Values represent mean ⫾ SD of 7 different donors.
IL-12+IL-18 – + – + – + – + – + – + – + – + – + – + – + – + – + – + – + – +
For clarity, selected statistical analyses are indicated.
K562 – – + + – – + + – – + + – – + + – – + + – – + + – – + + – – + +
*P ⬍ .05, **P ⬍ .01, ***P ⬍ .001.

on target cells.32 Experiments assessing degranulation of CD56dim Interrelationships between different NK-cell responses induced
NK cells upon modulation of IgG density on target cells produced by target cell recognition
similar results as have been reported previously (supplemental
Figure 4; Bryceson et al32). Increasing the concentration of IgG In addition to determining the frequency and phenotype of
augmented cytokine and chemokine responses in a nonlinear responding cells, multicolor flow cytometric analysis can also
fashion. Expression of CD48 or ICAM-1 on S2 cells lowered the provide information on how many different responses an individual
concentrations of IgG required to augment the frequency of cell can mediate. To gain insight into the relationships between
cytokine- and chemokine-producing CD56dim NK cells, with different NK-cell responses and to what extent individual cells can
coexpression of CD48 and ICAM-1 on S2 cells providing the perform multiple responses, intracellular expression of TNF-␣,
greatest response to low concentrations of IgG. NK-cell cytokine IFN-␥, MIP-1␣, and MIP-1␤ and surface expression of CD107a
and chemokine responses reached a plateau at the highest concen- were simultaneously assessed on CD56dim NK cells (Figure 6).
trations of IgG. At a 10⫺3 dilution of anti-S2 serum, coexpression Surface expression of CD107a (lysosome-associated membrane
of ICAM-1 and CD48 increased the frequency of IFN-␥– and protein-1) is a marker of lytic granule exocytosis.32 In line with
TNF-␣–producing CD56dim NK cells 2.8- and 2.3-fold, respec- previous findings, untransfected S2 cells did not induce cytokine or
tively. Nonlinear regression analysis revealed that the half- chemokine expression, and CD107a was not expressed on the cell
maximal percentage of IFN-␥–producing CD56dim NK cells in- surface (Figure 6A). Stimulation of resting NK cells by S2 cells
duced by CD16 engagement alone could be achieved with 10- and expressing ICAM-1, CD48, and ULBP1 induced intracellular
63-fold lower IgG concentration when S2 cells expressed CD48 or expression of TNF-␣, IFN-␥, MIP-1␣, and MIP-1␤ and surface
ICAM-1 and CD48, respectively (data not shown). S2 cells expression of CD107a on subsets of CD56dim NK cells (Figure 6A).
expressing ICAM-1 achieved the half-maximal level of response of
The greatest frequency of responding cells was consistently
untransfected S2 cells with a 3-fold lower concentration of IgG.
observed upon staining with MIP-1␤, closely followed by that of
Similar observations were made for TNF-␣ secretion. In addition,
MIP-1␣. Expression of MIP-1␤ correlated strongly with expres-
the half-maximal MIP-1␣ or MIP-1␤ responses could be reached
sion of MIP-1␣. Expression of TNF-␣, IFN-␥, and CD107a was
with 7- and 10-fold lower concentrations of IgG upon S2 cell
expression of ICAM-1 and CD48, respectively. Thus, coengage- confined to a MIP-1␤⫹ NK-cell subset. CD107a expression did not
ment of different activating receptors can lower the activation necessarily correlate with expression of TNF-␣ or IFN-␥. How-
threshold for CD56dim NK-cell cytokine and chemokine produc- ever, IFN-␥ expression was contained mostly within a TNF-␣⫹
tion, increasing the frequency of responding cells even in the NK-cell subset. Thus, the data demonstrate specific interrelation-
context of high ligand densities for powerful NK cell–activating ships and a considerable degree of cellular overlap among different
receptors such as CD16. responses enacted by CD56dim NK cells. For example, IFN-␥

30 40
IFN-γ TNF-α
30
20
20
10
Positive cells (%)

10

Figure 5. Increasing activating ligand density on 0 0


target cells augments the frequency of cytokine- and 10–9 10–8 10–7 10–6 10–5 10–4 10–3 10–2 10–9 10–8 10–7 10–6 10–5 10–4 10–3 10–2
chemokine-producing CD56dim NK cells. Before mix- 100 100
ing with NK cells, S2 cells expressing ligands for NK-cell MIP-1α MIP-1β
receptors, as indicated, were preincubated with serial 80 80
dilutions of anti–S2 cell serum. Resting NK cells were
incubated with S2 cells for 6 hours at 37°C. After stimula- 60 60 S2
tion, the cells were surface stained with fluorochrome- S2–ICAM1
conjugated anti-CD56 mAb, fixed, permeabilized, and 40 40
stained intracellularly with fluorochrome-conjugated mAbs S2–CD48
20 20
to cytokines and chemokines. The percentage of CD56dim S2–ICAM1–CD48
NK cells producing IFN-␥, TNF-␣, MIP-1␣, and MIP-1␤,
0 0
as indicated, was determined by flow cytometry. Values
10–9 10–8 10–7 10–6 10–5 10–4 10–3 10–2 10–9 10–8 10–7 10–6 10–5 10–4 10–3 10–2
represent mean ⫾ SD of 3 different donors.
[IgG]
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11 NK-CELL RESPONSES TO TARGET CELL RECOGNITION 2173

A
30K 30K
3
10

20K 20K
SSC-H

FSC-A
2
10

CD56
1
10K 10K 10

0
Lymphocytes Singlets CD56dim NK cells
0 0
1 2 3
0 10K 20K 30K 0 10K 20K 30K 0 10 10 10
FSC-H FSC-H DCM Stimulation

1.7 4.7 5.8 0.7 6.3 0.2 5.79 0.7


3 3 3 3
10 10 10 10
MIP-1β

2 2 2 2
10 10 10 10
S2
1 1 1 1
10 10 10 10

0 2.2 0 0.3 0 0.2 0 0.3

5.9 49.9 41 14.8 47.9 7.9 43 12.8


103 103 103 103
S2
ICAM1
MIP-1β

102 102 102 102


CD48
101 101 101 101 ULBP1

0 3.9 0 0.6 0 0.2 0 1.3


1 2 3 1 2 3 1 2 3 1 2 3
0 10 10 10 0 10 10 10 0 10 10 10 0 10 10 10
MIP-1α TNF-α IFN-γ CD107a
1.3 12.8 2 14 7.3 6.8 9.5 3.8 8.9 6.2
10
3
10
3
10
3
10
3
103 S2
ICAM1
CD107a

TNF-α
10
2
10
2
10
2
10
2
102
CD48
1 1 1 1 10
1 ULBP1
10 10 10 10

0 0 0 0
43 39.8 9 4.9 0 1.9

1 2 3 1 2 3 1 2 3 1 2 3 1 2 3
0 10 10 10 0 10 10 10 0 10 10 10 0 10 10 10 0 10 10 10

MIP-1β MIP-1α TNF-α IFN-γ IFN-γ


B
MIP-1β TNF-α IFN-γ CD107a
4 responses

3 responses

2 responses

1 response

No response
S2 S2 S2 S2 S2 S2 S2
ICAM-1 CD48 ULBP1 CD48 ICAM1 CD48
ULBP1 CD48 ULBP1
ULBP1 +
IgG
Figure 6. Interrelationships between different NK-cell responses induced by target cell recognition. Resting NK cells were mixed with S2 cells expressing ligands for
NK-cell receptors, as indicated, and incubated for 6 hours at 37°C. For some stimulations, S2 cells were preincubated with diluted anti–S2 cell serum (⫹ IgG). After stimulation,
the cells were surface stained with fluorochrome-conjugated anti-CD56 and anti-CD107a mAbs, fixed, permeabilized, and stained intracellularly with fluorochrome-conjugated
mAbs to cytokines and chemokines. (A) Lymphocytes were gated on forward scatter height (FSC-H) versus side scatter height plots (SSC-H). Single-cell events were gated on
forward scatter height (FSC-H) versus forward scatter area plots (FSC-A). CD56dim NK cells were gated on CD56 versus dead cell marker (DCM) plots. The second and third
rows show MIP-1␣, TNF-␣, IFN-␥, and CD107a staining in relation to MIP-1␤ staining after stimulation with S2 cells, as indicated. The bottom row shows MIP-1␤, MIP-1␣,
TNF-␣, and IFN-␥ staining in relation to CD107a staining, and IFN-␥ staining in relation to TNF-␣ staining (right panel). Gates were set using fluorochrome-conjugated isotype
control mAbs. The plots are derived from one representative donor. (B) CD56dim NK cells were gated as described in panel A, and a Boolean gating strategy was used for
analysis. Pie charts represent the frequency of cells positive for the given number of measured responses (MIP-1␤, TNF-␣, IFN-␥, and CD107a). Thus, cells can be categorized
into the number of responses they display. Arcs depict the relative frequency of cells specifically positive for MIP-1␤, TNF-␣, IFN-␥, and/or CD107a staining, as indicated.
Values represent the mean of 6 different donors.

production by individual CD56dim NK cells is likely to be nor TNF-␣ was increased relative to S2 cells only expressing
accompanied by MIP-1␤ and TNF-␣ production by the same cell. CD48. A small fraction of cells coexpressed MIP-1␤, TNF-␣, and
Further analysis was performed to enumerate responses by surface CD107a. Coexpression of ICAM-1, CD48, and ULBP1
individual CD56dim NK cells upon recognition of target cells further increased the frequency of cells producing MIP-1␤ and
expressing increasing numbers of ligands for activating receptors TNF-␣. Moreover, IFN-␥ was produced by a sizable fraction of
(Figure 6B). Given the high correlation between expression of CD56dim NK cells. If anything, surface expression of CD107a
MIP-1␣ and MIP-1␤, MIP-1␣ was omitted from this analysis. decreased, compared with stimulation with S2 cells coexpressing
Incubation of resting NK cells alone, with untransfected S2 cells, or CD48 and ULBP1. Upon stimulation with S2 cells coexpressing
S2 cells expressing ICAM-1 did not induce responses by CD56dim ICAM-1, CD48, and ULBP1, a small fraction of cells responded
NK cells. Expression of CD48 or ULBP1 on S2 cells induced with all 4 responses examined, whereas a greater fraction of cells
MIP-1␤ and weak degranulation by CD56dim NK cells. Coexpres- responded with 3 responses. Last, when NK cells were stimulated
sion of CD48 and ULBP1 on S2 cells specifically synergized for with IgG-coated S2 cells expressing CD48 and ULBP1, more than
degranulation, as neither the frequency of cells producing MIP-1␤ 90% of the CD56dim NK cells responded in terms of at least one
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2174 FAURIAT et al BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11

A A current view of NK-cell biology regards CD56bright and

M 7a
IP α
β
CD56dim NK cells as developmentally distinct subsets occupying

α
M P-1
-1
10


F-

N
different functional niches.41,42 In general, CD56bright NK cells are
CD

TN
I

IF
considered to be the major source of cytokines, whereas CD56dim
0 1 2 3 4 5 6 NK cells are regarded as specialized for cytotoxic function.12,30,43
Stimulation time (hours) The anatomic distribution of the 2 subsets underlies this perceived
dichotomy. Enriched in secondary lymphoid tissues, CD56bright NK
B IFN-γ cells can intimately interact with other immune cells, influencing
TNF-α ongoing adaptive immune responses through secretion of soluble
CD107a factors. Conversely, in the periphery, CD56dim NK-cell cytotoxic
MIP-1α activity can provide immunosurveillance of infected or neoplastic
MIP-1β
cells. Notably, CD56bright NK cells produce more cytokines in
response to phorbol myristate acetate and ionomycin stimulation
Strength of stimulation
than CD56dim NK cells.26,27 This fact has been ascribed to the
Figure 7. Schematic representation of activation thresholds and kinetics of
resting CD56dim NK-cell responses. (A) Approximate times required for induction of
relatively low expression of the phosphatase SHIP-1 and the high
different NK-cell responses such as degranulation (surface expression of CD107a), expression of the phosphatase inhibitor SET in CD56bright NK cells,
chemokine secretion (MIP-1␣ and MIP-1␤), and cytokine secretion (IFN-␥ and facilitating a lower activation threshold for cytokine secretion.44,45
TNF-␣) are indicated on the time scale. (B) The figure depicts the relative signal
strength required for induction of different NK-cell responses such as degranulation
Although we also found that exogenous cytokines induced a
(surface expression of CD107a), chemokine secretion (MIP-1␣ and MIP-1␤), and greater frequency of IFN-␥– and TNF-␣–producing CD56bright NK
cytokine secretion (IFN-␥ and TNF-␣). cells in 6- and 24-hour assays, our data highlight a prominent role
for CD56dim NK cells in producing cytokines and chemokines upon
parameter and 16% of the cells responded in terms of all target cell recognition. In fact, CD56dim NK cells responded more
parameters examined. Analogously, increasing densities of IgG on vigorously in terms of cytokine and chemokine production relative
target cells augmented the frequencies of CD56dim NK cells to CD56bright NK cells. These data substantiate and extend previous
displaying multiple different responses, and costimulation reduced findings demonstrating that CD56dim NK cells can produce cyto-
the density of IgG required for NK cells to display multiple kines in response to a tumor cell line.46 Here, cytokines and
responses (supplemental Figure 5). chemokines were induced by 2B4 and NKG2D synergy or K562
Together, these data provide insights into the heterogeneous cells that express ligands for the natural cytotoxicity receptor
response of NK cells to target cell recognition, revealing specific NKp30, NKG2D, and DNAM-1.47,48 Comparatively, CD56bright and
interrelationships and a hierarchy among the NK-cell responses for CD56dim NK-cell subsets express similar levels of activating
degranulation of lytic granules and production of chemokines and receptors.36 Therefore, differential expression of molecules convey-
cytokines. Moreover, the data illustrate how increasing signal ing signals from receptors such as 2B4, NKG2D, and NKp30 might
strength can induce multiple different effector responses from underlie the relatively more potent response of CD56dim NK cells to
individual NK cells. target cell recognition. Division of NK-cell subsets on the basis of
variegated inhibitory receptor expression or maturation markers
such as CD27 may further distinguish CD56dim NK-cell subsets in
terms of cytokine production and will be addressed in further
Discussion experiments.46,49,50
NK cells have previously been reported to secrete a plethora of
With a multiplicity of receptors evolved to sense cellular homeosta-
proinflammatory and immunoregulatory cytokines. In our experi-
sis and distress, NK cells are well equipped to act as primary
mental setting, using resting human NK cells freshly isolated from
initiators of immune responses upon recognition of infected or
peripheral blood, K562 target cell recognition consistently induced
neoplastic cells.40 Such responses are not confined to cytotoxic
effector mechanisms, but also involve the secretion of cytokines secretion of a proinflammatory cytokine profile characterized by
and chemokines. Here, we define the cytokines and chemokines MIP-1␣, MIP-1␤, RANTES, TNF-␣, and IFN-␥. This profile was
that are secreted by normal, freshly isolated human peripheral observed also with S2 cells expressing ligands for NKG2D and
blood NK cells upon interaction with target cells expressing 2B4, IgG-coated S2 cells triggering CD16, and K562 cells
complex or defined sets of ligands for specific activating receptors. triggering NKp30. NKG2D and 2B4 synergy is immunoreceptor
We demonstrate that engagement of the activating receptors CD16, tyrosine-based activation motif independent, whereas CD16 and
2B4, or NKG2D sufficed for rapid secretion of chemokines, and NKp30 are associated with immunoreceptor tyrosine-based activa-
that coengagement of additional activating receptors could acceler- tion motif–containing adaptor proteins.51-53 Thus, diverse signals
ate and increase chemokine secretion. In contrast, secretion of induce proinflammatory cytokine and chemokine secretion by NK
IFN-␥ required engagement of multiple different receptors and cells. Strikingly, upon NK-cell recognition of target cells, we did
occurred later, revealing a tighter control and a higher activation not detect secretion of immunoregulatory cytokines, such as IL-5,
threshold for induction of cytokine secretion (Figure 7). Unexpect- IL-10, IL-13, or GM-CSF. Such immunoregulatory cytokines are
edly, the results revealed CD56dim, rather than CD56bright, NK cells more often reported to be secreted by NK cells upon stimulation
to be more prominent producers of cytokines upon target cell with exogenous cytokines. For example, IL-10 is secreted predomi-
recognition. Furthermore, upon both target cell recognition and nately by CD56bright NK cells upon stimulation by IL-12 with IL-2
exogenous cytokine stimulation, CD56dim NK cells excelled in or IL-15 (Fehniger et al21; Wolk et al54; C.F. and Y.T.B. unpublished
production of the chemokine MIP-1␤. These experiments highlight observations, March 2009). In mice, such NK-cell secretion of
CD56dim NK cells as an important proinflammatory cytokine IL-10 requires STAT4,55 which signals downstream of the IL-12
source during early immune responses to aberrant cells. and IL-23 receptors.56 Our data suggest that NK cells are initially
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11 NK-CELL RESPONSES TO TARGET CELL RECOGNITION 2175

wired to promote immune responses by secreting proinflammatory responses. Chemokine production is an early feature of NK-cell
cytokines upon target cell recognition. responses and is triggered with relatively weak activating signals,
With Drosophila S2 cells expressing ligands for NK-cell requiring less stimulation than that necessary for degranulation or
receptors, the minimal requirements for cytokine and chemokine cytokine production. Induction of TNF-␣ and IFN-␥ is more
production were dissected. Interestingly, IFN-␥ secretion required stringently controlled, with a greater requirement for receptor
the coengagement by LFA-1, 2B4, and NKG2D, reflecting a cooperation and release at later time points after stimulation.
minimal requirement previously identified for natural cytotoxic- Thus, the data reveal a hierarchy among factors released upon
ity.33 Likewise TNF-␣ secretion also required receptor coengage- NK-cell interactions with target cells, with graded responses
ment, although here 2B4 and NKG2D synergy sufficed. Data according to the degree of ligand expression for activating
corroborate an earlier study using antibody-coated beads, where receptors (Figure 7B).
mAbs for different NK cell activating receptors could not induce Altogether, this study provides detailed insight into regulation
secretion of TNF-␣ and IFN-␥ on their own by freshly isolated NK of NK-cell cytokine secretion upon target cell recognition. Engage-
cells.36 However, S2 cells revealed that secretion of IFN-␥ is ment of individual activating receptors on resting NK cells suffices
particularly dependent on LFA-1 engagement, as opposed to for chemokine secretion, to alert and recruit other immune cells.
secretion of TNF-␣. This might reflect not only LFA-1–mediated More complex interactions, upon which multiple activating NK-
adhesion, but also signaling.57 Remarkably, chemokines such as cell receptors are engaged, can induce production of TNF-␣ and
MIP-1␣, MIP-1␤, and RANTES could be induced by engagement IFN-␥, to promote cellular resistance to infections and shape
of 2B4 or NKG2D. In contrast to other activating receptors, adaptive immune responses. These chemokines and cytokines are
engagement of CD16 was sufficient to induce some IFN-␥ and readily produced by the CD56dim NK-cell subset. Chemokine and
TNF-␣ secretion, in addition to chemokine secretion. The strong cytokine production by CD56dim NK cells may thus be an important
propensity of CD16 to induce NK-cell activation reflects the role of component of immunosurveillance.
antibody-dependent cell-mediated cytotoxicity as an effector arm
of the adaptive immune system. Thus, in vivo, selection of B cells
can safeguard robust activation mediated by CD16 engagement. Acknowledgments
Notably, upon stimulation of receptors such as 2B4 and NKG2D,
intracellular cytokine and chemokine expression was apparent, We thank S. Wood for critical reading of the paper and
sometimes without much secretion detected. The divergence in J. Michaëlsson for helpful discussions.
intracellular expression versus secretion after stimulation might This work was supported by grants from the Swedish Founda-
reflect regulation of cytokine and chemokine exocytosis. This facet tion for Strategic Research, Research Council, and Cancer Society
of regulation will be interesting to explore in more detail. (C.F., H.-G.L., and Y.T.B.), by the Intramural Research Program at
Temporally, the secretion of chemokines always preceded that National Institutes of Health, National Institute of Allergy and
of TNF-␣ and IFN-␥. Comparing the frequency of cytokine- and Infectious Diseases (E.O.L.), and Mary Beve’s Foundation, David
chemokine-producing cells revealed that a higher proportion of NK & Astrid Hagelen’s Foundation, the Karolinska Institute Research
cells produced chemokines relative to cytokines after 6 hours of Foundation, and Jonas Söderquist’s Stipend (Y.T.B.).
stimulation. Costimulation of receptors by engagement of LFA-1 or
other coactivation receptors could, in many instances, accelerate
and increase cytokine and chemokine secretion, and increase the Authorship
frequency of cells producing these factors. As we could simulta-
neously monitor production of MIP-1␣, MIP-1␤, TNF-␣, and Contribution: C.F. designed research, performed experimental
IFN-␥, as well as CD107a, in individual cells, the relationships work, analyzed and interpreted data, and drafted the paper; E.O.L
between production of specific chemokines, cytokines, and degranu- and H.-G.L. designed research and contributed to drafting the
lation could be assessed. These experiments revealed that produc- paper; and Y.T.B. designed research, analyzed and interpreted data,
tion of TNF-␣ and IFN-␥ and degranulation were contained within and drafted the paper.
the chemokine-producing NK-cell subset. Moreover, most IFN-␥– Conflict-of-interest disclosure: The authors declare no compet-
producing cells also expressed TNF-␣, but the inverse relationship ing financial interests.
did not hold. Production of TNF-␣ and IFN-␥ did not necessarily Correspondence: Cyril Fauriat or Yenan T. Bryceson, Center for
correlate with degranulation, but this could in part reflect differ- Infectious Medicine, Department of Medicine, Karolinska Institute,
ences in kinetics of these responses (Figure 7A). Taken together, Karolinska University Hospital Huddinge, S-14186 Stockholm, Swe-
these results reveal different activation thresholds for distinct den; e-mail: cfauriat@wanadoo.fr or yenan.bryceson@ki.se.

References
1. Lanier LL. NK cell recognition. Annu Rev Immu- highly regulated antiviral warfare. Trends Immu- T-cell-deficient hosts. Proc Natl Acad Sci U S A.
nol. 2005;23:225-274. nol. 2007;28(6):252-259. 1993;90(13):6115-6119.
2. Bottino C, Castriconi R, Moretta L, Moretta A. 6. Vivier E, Tomasello E, Baratin M, Walzer T, 9. Scharton TM, Scott P. Natural killer cells are a
Cellular ligands of activating NK receptors. Ugolini S. Functions of natural killer cells. Nat Im- source of interferon gamma that drives differen-
Trends Immunol. 2005;26(4):221-226. munol. 2008;9(5):503-510. tiation of CD4⫹ T cell subsets and induces early
7. Wherry JC, Schreiber RD, Unanue ER. Regula- resistance to Leishmania major in mice. J Exp
3. Bryceson YT, March ME, Ljunggren HG, Long tion of gamma interferon production by natural Med. 1993;178(2):567-577.
EO. Activation, coactivation, and costimulation of killer cells in scid mice: roles of tumor necrosis 10. Orange JS, Wang B, Terhorst C, Biron CA. Re-
resting human natural killer cells. Immunol Rev. factor and bacterial stimuli. Infect Immun. 1991; quirement for natural killer cell-produced inter-
2006;214(1):73-91. 59(5):1709-1715. feron gamma in defense against murine cytomeg-
4. Lodoen MB, Lanier LL. Natural killer cells as an 8. Gazzinelli RT, Hieny S, Wynn TA, Wolf S, Sher A. alovirus infection and enhancement of this
initial defense against pathogens. Curr Opin Im- Interleukin 12 is required for the T-lymphocyte- defense pathway by interleukin 12 administration.
munol. 2006;18(4):391-398. independent induction of interferon gamma by an J Exp Med. 1995;182(4):1045-1056.
5. Lee SH, Miyagi T, Biron CA. Keeping NK cells in intracellular parasite and induces resistance in 11. Biron CA, Nguyen KB, Pien GC, Cousens LP,
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2176 FAURIAT et al BLOOD, 18 MARCH 2010 䡠 VOLUME 115, NUMBER 11

Salazar-Mather TP. Natural killer cells in antiviral regulatory role for the CD56(bright) subset. 42. Romagnani C, Juelke K, Falco M, et al.
defense: function and regulation by innate cyto- Blood. 2001;97(10):3146-3151. CD56brightCD16- killer Ig-like receptor- NK cells
kines. Annu Rev Immunol. 1999;17:189-220. 27. Jacobs R, Hintzen G, Kemper A, et al. display longer telomeres and acquire features of
12. Caligiuri MA. Human natural killer cells. Blood. CD56bright cells differ in their KIR repertoire and CD56dim NK cells upon activation. J Immunol.
2008;112(3):461-469. cytotoxic features from CD56dim NK cells. Eur 2007;178(8):4947-4955.
13. Zucchini N, Crozat K, Baranek T, Robbins SH, J Immunol. 2001;31(10):3121-3127. 43. Di Santo JP. Functionally distinct NK-cell subsets:
Altfeld M, Dalod M. Natural killer cells in immuno- 28. Münz C. Non-cytotoxic protection by human NK developmental origins and biological implications.
defense against infective agents. Expert Rev Anti cells in mucosal secondary lymphoid tissues. Eur Eur J Immunol. 2008;38(11):2948-2951.
Infect Ther. 2008;6(6):867-885. J Immunol. 2008;38(11):2946-2948. 44. Trotta R, Parihar R, Yu J, et al. Differential ex-
14. Cooper MA, Fehniger TA, Caligiuri MA. The biol- 29. Di Santo JP. Natural killer cells: diversity in pression of SHIP1 in CD56bright and CD56dim
ogy of human natural killer-cell subsets. Trends search of a niche. Nat Immunol. 2008;9(5):473- NK cells provides a molecular basis for distinct
Immunol. 2001;22(11):633-640. 475. functional responses to monokine costimulation.
Blood. 2005;105(8):3011-3018.
15. Cuturi MC, Anegon I, Sherman F, et al. Produc- 30. Colonna M. Interleukin-22-producing natural killer
45. Trotta R, Ciarlariello D, Dal Col J, et al. The PP2A
tion of hematopoietic colony-stimulating factors cells and lymphoid tissue inducer-like cells in mu-
inhibitor SET regulates natural killer cell IFN-
by human natural killer cells. J Exp Med. 1989; cosal immunity. Immunity. 2009;31(1):15-23.
gamma production. J Exp Med. 2007;204(10):
169(2):569-583. 31. Barber DF, Long EO. Coexpression of CD58 or 2397-2405.
16. Smyth MJ, Zachariae CO, Norihisa Y, Ortaldo JR, CD48 with intercellular adhesion molecule 1 on
46. Anfossi N, Andre P, Guia S, et al. Human NK cell
Hishinuma A, Matsushima K. IL-8 gene expres- target cells enhances adhesion of resting NK
education by inhibitory receptors for MHC class I.
sion and production in human peripheral blood cells. J Immunol. 2003;170(1):294-299.
Immunity. 2006;25(2):331-342.
lymphocyte subsets. J Immunol. 1991;146(11): 32. Bryceson YT, March ME, Barber DF, Ljunggren
3815-3823. 47. Brandt CS, Baratin M, Yi EC, et al. The B7 family
HG, Long EO. Cytolytic granule polarization and member B7-H6 is a tumor cell ligand for the acti-
17. Warren HS, Kinnear BF, Phillips JH, Lanier LL. degranulation controlled by different receptors in vating natural killer cell receptor NKp30 in hu-
Production of IL-5 by human NK cells and regula- resting NK cells. J Exp Med. 2005;202(7):1001- mans. J Exp Med. 2009;206(7):1495-1503.
tion of IL-5 secretion by IL-4, IL-10, and IL-12. 1012.
J Immunol. 1995;154(10):5144-5152. 48. Carlsten M, Bjorkstrom NK, Norell H, et al. DNAX
33. Bryceson YT, Ljunggren HG, Long EO. Minimal accessory molecule-1 mediated recognition of
18. Bluman EM, Bartynski KJ, Avalos BR, Caligiuri requirement for induction of natural cytotoxicity freshly isolated ovarian carcinoma by resting
MA. Human natural killer cells produce abundant and intersection of activation signals by inhibitory natural killer cells. Cancer Res. 2007;67(3):1317-
macrophage inflammatory protein-1 alpha in re- receptors. Blood. 2009;114(13):2657-2666. 1325.
sponse to monocyte-derived cytokines. J Clin In- 34. Gonzalez VD, Bjorkstrom NK, Malmberg KJ, et 49. Kim S, Sunwoo JB, Yang L, et al. HLA alleles de-
vest. 1996;97(12):2722-2727. al. Application of nine-color flow cytometry for de- termine differences in human natural killer cell
19. Peritt D, Robertson S, Gri G, Showe L, tailed studies of the phenotypic complexity and responsiveness and potency. Proc Natl Acad Sci
Aste-Amezaga M, Trinchieri G. Differentiation of functional heterogeneity of human lymphocyte U S A. 2008;105(8):3053-3058.
human NK cells into NK1 and NK2 subsets. J Im- subsets. J Immunol Methods. 2008;330(1-2):64-
50. Silva A, Andrews DM, Brooks AG, Smyth MJ,
munol. 1998;161(11):5821-5824. 74.
Hayakawa Y. Application of CD27 as a marker for
20. Oliva A, Kinter AL, Vaccarezza M, et al. Natural 35. Sutherland CL, Chalupny NJ, Schooley K, distinguishing human NK cell subsets. Int Immu-
killer cells from human immunodeficiency virus VandenBos T, Kubin M, Cosman D. UL16-binding nol. 2008;20(4):625-630.
(HIV)-infected individuals are an important source proteins, novel MHC class I-related proteins, bind
51. Lanier LL, Yu G, Phillips JH. Co-association of
of CC-chemokines and suppress HIV-1 entry and to NKG2D and activate multiple signaling path-
CD3 zeta with a receptor (CD16) for IgG Fc on
replication in vitro. J Clin Invest. 1998;102(1):223- ways in primary NK cells. J Immunol.
human natural killer cells. Nature.
231. 2002;168(2):671-679.
1989;342(6251):803-805.
21. Fehniger TA, Shah MH, Turner MJ, et al. Differen- 36. Bryceson YT, March ME, Ljunggren HG, Long 52. Kurosaki T, Ravetch JV. A single amino acid in the
tial cytokine and chemokine gene expression by EO. Synergy among receptors on resting NK glycosyl phosphatidylinositol attachment domain
human NK cells following activation with IL-18 or cells for the activation of natural cytotoxicity and determines the membrane topology of Fc gamma
IL-15 in combination with IL-12: implications for cytokine secretion. Blood. 2006;107(1):159-166. RIII. Nature. 1989;342(6251):805-807.
the innate immune response. J Immunol. 1999; 37. Anegón I, Cuturi MC, Trinchieri G, Perussia B. 53. Pende D, Parolini S, Pessino A, et al. Identifica-
162(8):4511-4520. Interaction of Fc receptor (CD16) ligands induces tion and molecular characterization of NKp30, a
22. Roda JM, Parihar R, Magro C, Nuovo GJ, transcription of interleukin 2 receptor (CD25) and novel triggering receptor involved in natural cyto-
Tridandapani S, Carson WE III. Natural killer cells lymphokine genes and expression of their prod- toxicity mediated by human natural killer cells. J
produce T cell-recruiting chemokines in response ucts in human natural killer cells. J Exp Med. Exp Med. 1999;190(10):1505-1516.
to antibody-coated tumor cells. Cancer Res. 1988;167(2):452-472.
54. Wolk K, Kunz S, Asadullah K, Sabat R. Cutting
2006;66(1):517-526. 38. Carson WE, Giri JG, Lindemann MJ, et al. Inter- edge: immune cells as sources and targets of the
23. Lanier LL, Phillips JH, Hackett J Jr, Tutt M, Kumar leukin (IL) 15 is a novel cytokine that activates IL-10 family members? J Immunol. 2002;168(11):
V. Natural killer cells: definition of a cell type human natural killer cells via components of the 5397-5402.
rather than a function. J Immunol. 1986;137(9): IL-2 receptor. J Exp Med. 1994;180(4):1395-
55. Grant LR, Yao ZJ, Hedrich CM, et al. Stat4-
2735-2739. 1403.
dependent, T-bet-independent regulation of IL-10
24. Ferlazzo G, Munz C. NK cell compartments and 39. Roda JM, Parihar R, Lehman A, Mani A, in NK cells. Genes Immun. 2008;9(4):316-327.
their activation by dendritic cells. J Immunol. Tridandapani S, Carson WE III. Interleukin-21 56. Watford WT, Hissong BD, Bream JH, Kanno Y,
2004;172(3):1333-1339. enhances NK cell activation in response to anti- Muul L, O’Shea JJ. Signaling by IL-12 and IL-23
25. Lanier LL, Le AM, Civin CI, Loken MR, Phillips body-coated targets. J Immunol. 2006;177(1): and the immunoregulatory roles of STAT4. Immu-
JH. The relationship of CD16 (Leu-11) and 120-129. nol Rev. 2004;202:139-156.
Leu-19 (NKH-1) antigen expression on human 40. Bryceson YT, Long EO. Line of attack: NK cell 57. Perez OD, Mitchell D, Jager GC, et al. Leukocyte
peripheral blood NK cells and cytotoxic T lympho- specificity and integration of signals. Curr Opin functional antigen 1 lowers T cell activation
cytes. J Immunol. 1986;136(12):4480-4486. Immunol. 2008;20(3):344-352. thresholds and signaling through cytohesin-1 and
26. Cooper MA, Fehniger TA, Turner SC, et al. Hu- 41. Freud AG, Caligiuri MA. Human natural killer cell Jun-activating binding protein 1. Nat Immunol.
man natural killer cells: a unique innate immuno- development. Immunol Rev. 2006;214:56-72. 2003;4(11):1083-1092.
From www.bloodjournal.org by guest on September 11, 2016. For personal use only.

2010 115: 2167-2176


doi:10.1182/blood-2009-08-238469 originally published
online December 1, 2009

Regulation of human NK-cell cytokine and chemokine production by


target cell recognition
Cyril Fauriat, Eric O. Long, Hans-Gustaf Ljunggren and Yenan T. Bryceson

Updated information and services can be found at:


http://www.bloodjournal.org/content/115/11/2167.full.html
Articles on similar topics can be found in the following Blood collections
Immunobiology (5415 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 might also like