Você está na página 1de 11

Role of chemokines in the biology of natural killer cells

Michael J. Robertson
Bone Marrow and Stem Cell Transplantation Program and Division of Hematology/Oncology, Indiana University
School of Medicine, Indianapolis

Abstract: Natural killer (NK) cells participate in virus-infected cells [1–3]. NK cells participate in innate im-
innate and adaptive immune responses to obligate mune responses that can inhibit intracellular pathogens while
intracellular pathogens and malignant tumors. Two antigen-specific T- and B-cell responses are generated [4 – 6].
major NK cell subsets have been identified in hu- Moreover, NK cells secrete cytokines, such as interferon-␥
mans: CD56dim CD16ⴙ and CD56bright CD16ⴚ. (IFN-␥), which promote the differentiation of activated CD4 T
Resting CD56dim CD16ⴙ NK cells express cells into Th1 helper effector cells [4, 5, 7]. NK cells can also
CXCR1, CXCR2, CXCR3, CXCR4, and CX3CR1 contribute to the elimination of infected cells during the effec-
but no detectable levels of CC chemokine recep- tor phase of adaptive immune responses [1–3]. NK cells can
tors on the cell surface. They migrate vigorously in recognize and destroy cancer cells that have evaded cytotoxic
response to CXCL12 and CXC3L1. In contrast, T lymphocytes (CTL) [8]. Thus, NK cells participate in innate
resting CD56bright CD16ⴚ NK cells express little and adaptive immune responses to intracellular pathogens and
CXCR1, CXCR2, and CXC3R1 but high levels of malignant tumors.
CCR5 and CCR7. Chemotaxis of CD56bright The cytolytic activity of NK cells is clearly distinguishable
CD16ⴚ NK cells is stimulated most potently by from that mediated by typical CTL: It occurs spontaneously
CCL19, CCL21, CXCL10, CXCL11, and CXCL12. (i.e., in the absence of deliberate, prior immunization) and does
Following activation, NK cells can migrate in re- not require expression of syngeneic major histocompatibility
sponse to additional CC and CXC chemokines. Cy- complex (MHC) antigens by target cells. Recently, the molec-
tolytic activity of NK cells is augmented by CCL2, ular basis for target-cell recognition by human and murine NK
CCL3, CCL4, CCL5, CCL10, and CXC3L1. More- cells has been elucidated [2, 8 –10]. Unlike CTL, NK cells do
over, proliferation of CD56dim CD16ⴙ NK cells is not appear to express one dominant receptor that dictates the
costimulated by CCL19 and CCL21. Activated NK specificity of cytolysis. Rather, triggering of NK cell cytotox-
cells produce XCL1, CCL1, CCL3, CCL4, CCL5, icity reflects a balance between activating and inhibitory sig-
CCL22, and CXCL8. Chemokines secreted by NK nals mediated by cell-surface receptors that belong to several
cells may recruit other effector cells during im- different gene families. Inhibitory MHC class I receptors have
mune responses. Furthermore, CCL3, CCL4, and a central role in current paradigms of target-cell recognition by
CCL5 produced by NK cells can inhibit in vitro NK cells [2, 8 –10]. Ligation of these inhibitory receptors by
replication of HIV. CCL3 and CXL10 expression specific MHC class I allotypes delivers a dominant-negative
appear to be required for protective NK cell re- signal to NK cells that prevents natural killing. Down-regula-
sponses in vivo to murine cytomegalovirus or tion of MHC class I molecules on the target-cell surface, which
Leishmania major, respectively. Moreover, NK commonly occurs during viral infection or neoplastic transfor-
cells participate in the in vivo rejection of trans- mation, releases the NK cell from inhibitory signals and allows
duced tumor cells that produce CCL19 or CCL21.
lysis of the aberrant target cell. Positive signals for cytolysis
Thus, chemokines appear to play an important role
are provided by ligation of several activating receptors (e.g.,
in afferent and efferent NK cell responses to in-
CD2, CD16, NKR-P1, 2B4, NKp30, NKp44, and NKp46)
fected and neoplastic cells. J. Leukoc. Biol. 71:
expressed by NK cells [2, 9, 10]. However, the contribution of
173–183; 2002.
these putative activating receptors to triggering NK cytolysis in
vivo has not been well-defined.
Key Words: chemotaxis 䡠 cytotoxicity 䡠 proliferation
Like T cells, NK cells are heterogeneous with respect to
functional activity and cell-surface antigen expression [11–14].
Two major NK cell subsets have been identified in humans.
INTRODUCTION CD56dim NK cells, comprising about 90% of peripheral blood
NK cells, express the CD16 antigen at high density but rela-
Natural killer (NK) cells, such as T and B cells, are one of the
major lymphocyte subsets that have been identified in all
vertebrate species examined [1–3]. Unlike T and B lympho-
cytes, however, NK cells do not productively rearrange T-cell Correspondence: Michael J. Robertson, M.D., Bone Marrow and Stem Cell
Transplantation Program, 1044 W. Walnut Street, Room R4-202, Indianapolis,
receptor or immunoglobulin genes and do not appear to express IN 46202. E-mail: mjrobert@iupui.edu
highly variable, antigen-specific receptors. Nevertheless, NK Received November 1, 2001; revised November 29, 2001; accepted Novem-
cells can discriminate between normal cells and neoplastic or ber 30, 2001.

Journal of Leukocyte Biology Volume 71, February 2002 173


tively low levels of CD56. Freshly isolated, unstimulated Chemokines play a crucial role in coordinating adaptive
CD56dim NK cells mediate natural killing and antibody-depen- immune responses. Chemokines regulate the migration of im-
dent cellular cytotoxicity (ADCC). CD56dim NK cells express ␤ mature lymphoid progenitor cells, the recirculation of mature
␥ common chains of the interleukin (IL)-2 and IL-15 receptors naive T and B lymphocytes, and the homing of antigen-specific
and demonstrate augmented cytotoxicity, proliferation, and cy- effector T cells. Chemokines also control the migration of
tokine secretion after stimulation with IL-2 or IL-15 [11–13, antigen-presenting cells, including dendritic cells and cells of
15]. However, CD56dim NK cells produce relatively little monocyte/macrophage lineage [22–27]. Other important phys-
IFN-␥ after stimulation with IL-2 or IL-15 in combination with iologic and pathophysiologic activities of chemokines include
IL-12. CD56bright NK cells express high-affinity IL-2 receptor inhibition or promotion of angiogenesis, regulation of tumor
␣ ␤ ␥ heterotrimers but little or no CD16. CD56bright NK cells metastasis, inhibition of hematopoietic progenitor cell prolif-
proliferate vigorously in response to IL-2 alone [11–13] and eration, and modulation of HIV infection. A detailed discus-
produce abundant amounts of IFN-␥ after stimulation with IL-2 sion of chemokine biology is beyond the scope of this review.
or IL-15, together with IL-12. In contrast, CD56bright NK cells Interested readers are referred to recent review articles devoted
exhibit weak cytolytic activity when freshly isolated. These to chemokines and their receptors [22–28].
observations support the hypothesis that CD56bright NK cells, Inconsistency and bewildering complexity have character-
like CD4 T cells, predominantly regulate other cells through ized the nomenclature of chemokines and their receptors in the
cytokine production [14, 16]. In contrast, CD56dim NK cells, past. It is not uncommon to encounter from three to more than
like CD8⫹ CTL, are terminally differentiated cytotoxic- five different synonyms being used in the literature to refer to
effector cells. the same chemokine. Recently, a uniform and widely accepted
The CD56bright and CD56dim NK cell subsets can be reliably nomenclature has been adapted [23, 27, 29]. Common syn-
distinguished only for resting human NK cells. The CD56 onyms for some chemokines that have been shown to affect NK
antigen is up-regulated on CD56dim NK cells following activa- cell function are summarized in Table 1.
tion in vitro or in vivo [17–21]. Thus, the density of CD56 on
the cell surface cannot be used to discriminate unstimulated
CD56bright NK cells from activated CD56dim NK cells. More- CHEMOKINE RECEPTORS EXPRESSED BY
over, subsets analogous to CD56bright and CD56dim NK cells in NK CELLS
humans have not been identified in mice or other species. It
should be noted that murine NK cells do not express CD56, so The expression of chemokine receptors by human NK cells is
this molecule cannot be required for NK cell function in vivo. a subject of controversy. The two most comprehensive studies
To mediate their cytolytic function effectively, NK cells that have been published to date provide contradictory results,
must be recruited to the site of infected or neoplastic cells. particularly with respect to the CXC chemokine receptors.
Moreover, to regulate the adaptive immune response, cytokine- Campbell et al. [30] found that most human NK cells express
secreting NK cells must be in intimate proximity to antigen- high levels of CXCR1, CXCR4, and CX3CR1; CXCR2 and
stimulated T and/or B cells. Nevertheless, until recently, very CXCR3 were expressed at lower levels. Several other groups
little was known about the regulation of NK cell migration and have also described expression of CXCR1 and CXCR2 [31, 32]
trafficking. Work done in several laboratories over the past few or CX3CR1 [33, 34] by resting human NK cells. In contrast,
years has implicated chemokines as key regulators of NK cell
migration and function.
TABLE 1. Synonyms for Selected Chemokines
with NK Cell Activity

CHEMOKINES AND THEIR RECEPTORS Chemokine Synonyms

XCL1 Lymphotactin ␣, SCM-1␣, ATAC


Chemokines are a group of at least 47 related proteins, making XCL2 Lymphotactin ␤, SCM-1␤, ATAC
them the largest family of cytokines known [22–27]. Depending CCL1 I-309, TCA3
on the number and spacing of conserved cysteine residues in CCL2 MCP-1, MCAF, JE
their amino acid sequence, chemokines have been classified CCL3 MIP-1␣
CCL4 MIP-1␤
into four major groups: the CXC (or ␣), CC (or ␤), CX3C, and
CCL5 RANTES
C subfamilies. The CXC and CC subfamilies have been sub- CCL7 MCP-3
divided further according to structural homologies and biolog- CCL8 MCP-2
ical activities. Chemokines exert their biologic effects by bind- CCL19 MIP-3␤, ELC, CK␤-11, Exodus-3
ing to specific cell-surface receptors. The chemokine receptors, CCL20 MIP-3␣, LARC, Exodus-1, ST38
CCL21 6Ckine, Exodus-2, SLC, CK␤-9, TCA4
which have also been classified into four subfamilies, are
CCL22 MDC, STCP-1, ABCD-1
G-protein-coupled seven-transmembrane-spanning molecules. CXCL8 IL-8
A single chemokine can bind to more than one receptor, and a CXCL9 Mig
given receptor can interact with multiple chemokines. Together CXCL10 IP-10, crg-2
with the large number of chemokines and receptors, this poses CXCL11 I-TAC, IP-9, H174, ␤-R1
CXCL12 SDF-1␣, SDF-1␤, PBSF
a formidable challenge for investigators seeking to elucidate
CX3CL1 Fractalkine, neurotactin
the physiologic role of chemokines in vivo.

174 Journal of Leukocyte Biology Volume 71, February 2002 http://www.jleukbio.org


Inngjerdingen et al. [35] found that purified, resting human NK not CD16⫹ resting human NK cells express CCR7 on the cell
cells expressed CXCR4 but not CXCR1, CXCR2, CXCR3, or surface and migrate vigorously in response to CCL19 and
CX3CR1. It is not clear why Inngjerdingen et al. [35] failed to CCL21 [30]. Results from the author’s laboratory (unpublished
detect CXCR1, CXCR2, and CX3CR1 on resting human NK results) are concordant with those of Campbell et al. [30] with
cells. It is possible that levels of these chemokine receptors respect to expression of CCR6 and CCR7 by resting CD56bright
were down-regulated during the isolation of purified NK cells and CD56dim NK cells.
by their methods; this phenomenon has been noted by another In contrast to results of others [30, 41] and our unpublished
group [36]. data, Inngjerdingen et al. [35] have demonstrated that the
The published data are more consistent with respect to NK majority of resting human NK cells expresses CCR7. However,
cell expression of the CC chemokine receptors. As assessed by Inngjerdingen et al. [35] used a polyclonal antibody recogniz-
flow cytometry using specific antibodies, CCR1, CCR2, CCR3, ing the carboxy terminus of CCR7 to stain permeabilized NK
CCR4, CCR5, CCR6, CCR7, CCR8, and CCR9 are not ex- cells. Thus, the CCR7 molecules detected by this method may
pressed at significant levels on the surface of most resting be located intracellularly rather than on the cell surface.
human NK cells [30, 35, 37]. CCR2, CCR4, CCR5, and CCR8 In addition to CCR7, other chemokine receptors are ex-
are expressed by human NK cells after in vitro activation with pressed differentially by the CD56bright CD16⫺ and CD56dim
IL-2 or IL-15 [35, 37, 38]. CD16⫹ subsets of human NK cells (Table 2). Unlike the great
Recent studies have clarified the expression of CCR7 and majority of resting NK cells, the rare CD56bright CD16⫺ subset
response to the CCR7 ligands, CCL19 and CCL21, by human does not express CXCR1, CXCR2, or CX3CR1 but does ex-
NK cells. Several groups have demonstrated that CCL19 and press CCR5 [30]. As expected, based on the known biology of
CCL21 do not stimulate significant chemotaxis of resting pe- chemokine receptors, chemotaxis of NK cells stimulated by
ripheral blood NK cells [39 – 41]. Moreover, message for chemokines is inhibitable by Bordetella pertussis toxin [42, 44,
CCR7, as assessed by Northern blot or reverse transcriptase- 45]. Chemokine signaling in NK cells involves several guanine
polymerase chain reaction (RT-PCR) analysis, was not de- nucleotide-binding proteins and leads to intracellular calcium
tected in resting NK cells [40, 41]. In contrast, Kim et al. [42] ion mobilization [45– 48].
found that CCL19 and CCL21 stimulated the migration of the
CD16⫺ subset of cord blood and adult peripheral blood NK
cells. CD56bright CD16⫺ NK cells normally comprise only REGULATION OF NK CELL MIGRATION BY
⬃10% of the total peripheral blood NK cell population, and CHEMOKINES
selective chemotaxis of this subset is obscured readily by the
relatively high, spontaneous migration of the much more nu- Numerous chemokines have been shown to stimulate the mi-
merous CD56dim CD16⫹ NK cells [39]. However, Kim et al. gration of NK cells as determined by in vitro chemotaxis assays
[42] isolated adult and cord blood NK cells by positive selec- (Tables 3 and 4). Results with the CXC chemokines agree
tion using CD56 beads, which greatly enriches the proportion well with the data on CXC chemokine receptor expression by
of CD56bright CD16⫺ NK cells among the total NK cell pop- NK cells. Thus, resting human NK cells have been found to
ulation [43]. Subsequent work has confirmed that CD16⫺ but migrate in response to known ligands for CXCR3 (CXCL9,

TABLE 2. Expression of Chemokine Receptors by Resting Human NK Cells

Chemokine Receptor CD56dimCD16⫹ NK Cells CD56brightCD16-NK Cells Unfractionated NK Cellsa References

XCR1 NR NR NR
CCR1 0b 0 0 [30, 35]
CCR2 0 0 0 [30, 35, 37]
CCR3 0 0 0 [30, 35]
CCR4 ⫹/⫺ 0 0 [30, 35]
CCR5 0 ⫹ 0 [30, 35, 37]
CCR6 0 0 0 [30, 35]
CCR7 0 ⫹ ⫹/⫺ [30, 35]
CCR8 NR NR 0 [35]
CCR9 0 0 NR [30]
CCR10 NR NR NR
CCR11 NR NR NR
CXCR1 ⫹ 0 ⫹ [30–32]
CXCR2 ⫹ 0 ⫹ [30–32]
CXCR3 ⫹ ⫹ ⫹/⫺ [30, 35, 104]
CXCR4 ⫹ ⫹ ⫹ [30, 35]
CXCR5 0 0 0 [30, 35]
CXCR6 0 0 NR [30]
CX3CR1 ⫹ ⫹/⫺ ⫹ [30, 33–35]
a
Unfractionated human peripheral blood NK cell preparations are expected to contain ⬃10% CD56bright NK cells and ⬃90% CD56dim NK cells.
b
⫹, positive results reported; 0, negative results reported; ⫹/⫺, equivocal or contradictory results reported; NR, results not reported.

Robertson Chemokine role in biology of NK cells 175


TABLE 3. Migration of Resting NK Cells in Response to Chemokines

Chemokine Total NK cells CD56dim/CD16⫹ NK cells CD56bright/CD16⫺ NK cells References

XCL1 ⫹a NR NR [50, 54]


CCL1 0 NR NR [35]
CCL2 ⫹/⫺ ⫹/⫺ ⫹/⫺ [30, 34, 36, 44, 50]
CCL3 ⫹ ⫹ ⫹ [36, 51]
CCL4 ⫹ 0 ⫹/⫺ [30, 36]
CCL5 ⫹ 0 ⫹ [30, 36]
CCL7 ⫹/⫺ ⫹/⫺ ⫹ [30, 36, 44]
CCL8 ⫹/⫺ NR NR [36, 44]
CCL17 0 0 NR [34]
CCL19 ⫹/⫺ 0 ⫹ [30, 35, 40–42]
CCL20 ⫹/⫺ ⫹/⫺ ⫹/⫺ [30, 35, 40]
CCL21 0 0 ⫹ [30, 39, 40, 42]
CCL22 ⫹/⫺ 0 0 [30, 35]
CCL25 NR 0 0 [30]
CXCL1 ⫹/⫺ NR NR [35, 36]
CXCL8 ⫹/⫺ ⫹ 0 [30, 35, 36]
CXCL9 ⫹ NR NR [49]
CXCL10 ⫹ ⫹/⫺ ⫹ [30, 35, 36]
CXCL11 NR ⫹/⫺ ⫹ [30]
CXCL12 ⫹ ⫹ ⫹ [30, 35, 39, 42]
CX3CL1 ⫹ ⫹ 0 [30, 34, 35]
a
See key to symbols in footnotes to Table 2.

CXCL10, and CXCL11) and CXCR4 (CXCL12) [30, 35, 36, Resting human NK cells also migrate in response to several
39, 42, 49]. Furthermore, chemotaxis of resting NK cells is CC chemokines, including CCL2, CCL3, CCL4, CCL5, CCL7,
stimulated by XCL1 and CX3CL1 [30, 35, 50]. Although and CCL8 [35, 36, 44, 51], although known receptors for the
CXCR1 and CXCR2 appear to be expressed by most resting latter are generally undetectable on most resting NK cells [30,
human NK cells [30 –32], the responsiveness of the latter to 35, 37]. However, results with the CC chemokines are not
CXCL1 or CXCL8 is a subject of controversy. Some investiga- consistent in the published literature: Some investigators have
tors have shown that resting NK cells can migrate in response demonstrated positive findings using activated but not resting
to CXCL1 [35] or CXCL8 [30]; other investigators have found NK cells [44, 47]. It is possible that some of the known CC
these chemokines to have inconsistent, donor-dependent ef- chemokine receptors are actually expressed on the surface of
fects on resting NK cells [36]. Furthermore, CXCL8 may not resting NK cells but at levels below the limits of detection by
stimulate the chemotaxis of human NK cells that have been routine flow cytometry. Alternatively, resting NK cells may
activated in vitro [35, 47]. express as-yet uncharacterized novel receptors for some of the

TABLE 4. Chemokines Reported to Stimulate Migration of Activated NK Cells

Chemokine Cell preparation Stimulusa References

XCL1 Polyclonal human and murine NK cells; human NK clones IL-2; IL-2 ⫹ SC [45, 50, 54]
CCL1 Polyclonal human NK cells IL-2 [35, 52]
CCL2 Polyclonal human NK cells and NK clones IL-2; IL-2 ⫹ SC [35, 44, 47]
CCL3 Human NK clones IL-2 [47, 105]
CCL4 Human NK clones IL-2 [47]
CCL5 Human NK clones IL-2 [47, 105]
CCL7 Polyclonal human NK cells and NK clones IL-2; IL-2 ⫹ SC [44, 47]
CCL8 Polyclonal human NK cells and NK clones IL-2; IL-2 ⫹ SC [44, 47]
CCL17 Polyclonal human NK cells IL-2 [35, 52]
CCL19 Polyclonal human NK cells; NKL cellsb IL-2; LCM ⫹ lono [35, 40, 42]
CCL21 Polyclonal human NK cells; NKL cells IL-2; LCM ⫹ lono [40, 42]
CCL22 Polyclonal human NK cells IL-2; IL-2 ⫹ SC [35, 52, 53]
CXCL1 Polyclonal human NK cells IL-2 [35]
CXCL8 Polyclonal human NK cells IL-2 [46]
CXCL10 Polyclonal human NK cells IL-2 [35, 45]
CXCL12 Polyclonal human NK cells; NKL cells IL-2 [35, 40]
CX3CL1 Polyclonal human NK cells IL-2 [35]
a
SC, stimulator cells (e.g., irradiated B lymphoblastoid cell lines); LCM ⫹ lono, leukocyte conditioned medium plus ionomycin.
b
NKL is an IL-2-dependent neoplastic human NK cell line [106].

176 Journal of Leukocyte Biology Volume 71, February 2002 http://www.jleukbio.org


CC chemokines. After in vitro activation, human NK cells TABLE 5. Effect of Chemokines on NK Cell Cytotoxicity
up-regulate CCR2, CCR4, CCR7, and CCR8 [35, 37, 38, 40,
52] and demonstrate increased chemotactic responses to Resting NK Activated
Chemokine Cells NK Cells References
known ligands of these receptors, including CCL1, CCL2,
CCL3, CCL4, CCL5, CCL7, CCL8, CCL19, CCL21, and CCL2 ⫹a NR [36,57,58]
CCL22 [35, 40, 44, 47, 53]. CCL3 ⫹ NR [36,57,58,69]
Differences in the chemotactic responses of resting CCL4 ⫹ NR [36,57,58]
CD56bright and CD56dim NK cells correlate well with known CCL5 ⫹ NR [36,57,58]
CCL19 0 0 [40]
differences in their expression of chemokine receptors [30]. CCL20 0 0 [40]
Compared with CD56dim NK cells, CD56bright NK cells express CCL21 0 0 [40]
higher levels of CXCR3 and respond more vigorously to CXCL1 0 NR [36,57]
CXCL10 and CXCL11. Conversely, CD56bright NK cells ex- CXCL2 0 NR [36,57]
press little or no CXCR1, CXCR2, and CX3CR1 and do not CXCL4 0 NR [36,57]
CXCL8 0 NR [36,58]
migrate in response to CXCL8 or CX3CL1. As discussed CXCL10 ⫹ NR [36]
above, CD56bright NK cells but not CD56dim NK cells express CX3CL1 ⫹ NR [33]
CCR7 and respond to CCL19 and CCL21 [30, 42]. a
In contrast to the abundant in vitro studies, published data See key to symbols in footnotes to Table 2.
on in vivo migration of NK cells in response to chemokines are
sparse. Intraperitoneal (i.p.) injection of murine XCL1 induces
transient influx of lymphocytes into the peritoneal cavity [54]. found to augment ADCC or induced the lysis of NK-resistant
The majority of these lymphocytes are mature NK cells. How- targets by human NK cells. CX3CL1 has also been shown to
ever, freshly isolated murine NK cells did not migrate in vitro modestly increase NK cell cytotoxicity toward NK-sensitive
in response to XCL1 [54]. Therefore, additional factors, per- target cells [33].
haps induced by the local trauma of i.p. injection, may act in Maghazachi et al. [58] have shown that CCL2, CCL3, CCL4,
concert with XCL1 to stimulate chemotaxis of murine NK cells and CCL5 can augment cytotoxicity of enriched CD56⫹ hu-
in vivo. As discussed in detail below, CCL3 promotes in vivo man NK cells. In contrast to the results of Taub et al. [36],
migration of activated NK cells into the livers of mice infected these investigators found that the effects of CCL2, CCL3, and
with murine cytomegalovirus (MCMV) [6, 55]. CCL5 were comparable with those of optimal concentrations of
IL-2 and that chemokines could stimulate lysis of NK-sensitive
and NK-resistant target cells.
REGULATION OF NK CELL CYTOTOXICITY The mechanisms by which chemokines augment NK cell
BY CHEMOKINES cytolytic activity have not been fully elucidated. Lysis of target
cells by NK cells occurs in three phases [1, 2, 9, 10]. First, NK
NK cells mediate antibody-dependent and -independent lysis cells must bind to potential target cells via interactions be-
of target cells [1, 2, 9, 10]. Resting NK cells can lyse antibody- tween NK cell-surface adhesion molecules [e.g., lymphocyte
coated target cells by ADCC. The receptor on NK cells that function-associated antigen-1 (LFA-1) and CD2] and their
triggers ADCC is a multimolecular complex composed of cognate target-cell ligands [e.g., intercellular adhesion mole-
CD16, a low-affinity receptor for the Fc portion of immuno- cules (ICAM)-1, -2, and -3 and CD58]. Next, positive signals
globulin, in noncovalent association with homodimers or het- from ligation of activating receptors (e.g., CD16) must outweigh
erodimers of the ␨ family of signaling molecules. Resting NK negative signals from ligation of inhibitory receptors. Finally,
cells can also spontaneously lyse NK-sensitive target cells by NK cells must express apoptosis-inducing ligands (e.g., CD95
an antibody-independent process known as natural killing or ligand) and/or must discharge their cytotoxic granules against
NK activity [1, 2, 9, 10]. Sensitivity to natural killing is the the target-cell membrane [59]. NK cell cytotoxic granules
property of certain virus-infected and neoplastic-hematopoietic contain perforin and granzymes, which can induce apoptosis
cells; most normal cells and malignant epithelial cells are and necrosis of target cells.
NK-resistant. Nevertheless, after exposure to exogenous cyto- CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CXCL10, and
kines (such as IL-2, IL-12, or IL-15), NK cells can lyse solid CX3CL1 have been shown to promote cytotoxic granule release
tumor cells that are resistant to lysis by unstimulated NK cells by resting polyclonal human NK cells or NK cell clones [33,
[15, 18, 56]. This cytolytic activity has been called lympho- 36, 47]. Concentrations of chemokines that stimulate granule
kine-activated killer (LAK) activity. exocytosis were similar to those that enhance NK cell cytolytic
Relatively few chemokines have been shown to enhance the activity. Thus, chemokines may augment NK cell lysis of target
cytolytic activity of NK cells (Table 5). Taub et al. [36, 57] cells, in part, by facilitating the discharge of NK cell cytotoxic
found that CCL3 and CXCL10 consistently augmented the lysis granules. Augmentation of NK cell cytotoxicity by CX3CL1
of NK-sensitive K562 cells by purified, human NK cells. was not associated with up-regulation of LFA-1, CD2, or other
Enhancement of natural killing by these cytokines was inferior adhesion molecules on NK cells [33]. However, several CC
to that produced by optimal concentrations of IL-2. CCL2, chemokines known to augment NK cell cytotoxicity have been
CCL4, and CCL5 were also found to stimulate greater levels of found to induce redistribution of adhesion molecules on the NK
natural killing, albeit less consistently and in a donor-depen- cell surface [37]. Nevertheless, the effects of chemokines on
dent fashion. Unlike IL-2, none of these chemokines were NK cell adhesion-molecule expression and their conjugate

Robertson Chemokine role in biology of NK cells 177


formation with target cells have not been described in detail TABLE 6. Production of Chemokines by NK Cellsa
and merit further investigation. Moreover, it is currently not
known whether chemokines affect signals that are transduced Resting NK Activated
Chemokine Cells NK Cells References
after ligation of activating or inhibitory NK cell receptors.
XCL1 0b ⫹ [54]
CCL1 NR ⫹ [74]
REGULATION OF NK CELL PROLIFERATION CCL3 ⫹/⫺ ⫹ [37, 65, 66, 68, 69, 73]
BY CHEMOKINES CCL4 ⫹ ⫹ [37, 65, 66, 68, 72, 73]
CCL5 ⫹ ⫹ [37, 65–67]
CCL22 ⫹ ⫹ [67]
Human NK cells can be induced to proliferate in response to CXCL8 ⫹c ⫹ [71, 75, 76]
IL-2, IL-4, IL-7, IL-12, and IL-15 [15, 18, 60, 61]. However, a
NK cell proliferation to IL-2 and IL-15 is about tenfold greater Chemokines included in this table are those for which protein production
by NK cells has been reported.
than proliferation to other mitogenic cytokines. Thus, signals b
See key to symbols in footnotes to Table 2.
mediated through the common IL-2/IL-15 ␤ ␥ receptor appear c
Results pertain to freshly isolated uterine decidual CD56bright/CD16-NK
to provide the strongest proliferative stimulus for human NK cells.
cells. Like proliferation of T and B lymphocytes, the prolifer-
ation of NK cells appears to require costimulatory signals as
well as primary mitogenic signals. Costimulation of NK cell
CXCL8 protein [71]. It is currently not known whether periph-
proliferation can be provided by soluble cytokines, ligation of
defined cell-surface receptors, or contact with certain stimula- eral blood CD56bright CD16⫺ or CD56dim CD16⫹ NK cells
tor cells [60 – 64]. spontaneously produce CXCL8.
None of the chemokines that have been tested so far, in- After in vitro activation, NK cells produce greater amounts
cluding CCL2, CCL3, CCL4, CCL5, CCL19, CCL20, CCL21, of CCL4, CCL5, CCL22, and CXCL8 and also produce XCL1,
and CXCL8, have been found to stimulate the proliferation of CCL1, and CCL3 [37, 54, 65– 69, 72–76]. Based on these
purified, resting NK cells [40, 58]. CCL2, CCL3, CCL4, and results, it is expected that T cells, B cells, NK cells, neutro-
CCL5 can induce the proliferation of nylon-wool column-non- phils, and several other cell types could be attracted to the
adherent peripheral blood lymphocytes (PBL), which contain vicinity of activated NK cells. In fact, little has been published
mostly T and NK cells [58]. Purified CD4 and CD8 T cells did about the ability of activated NK cells to attract other cells in
not proliferate in response to CC chemokines. Furthermore, vitro or in vivo. Supernatants from activated human NK cells
CCL3- and CCL5-induced proliferation of PBL was abrogated stimulate the in vitro migration of CD4 T cells, CD8 T cells,
by the presence of neutralizing anti-IL-2 antibody. Thus, it has and neutrophils [76]. CXCL8 produced by activated NK cells
been speculated that CC chemokines can stimulate T-cell was found to be partially but not completely responsible for the
secretion of IL-2, which then indirectly stimulates the prolif- observed T-cell chemotaxis. Nieto et al. [37] have shown that
eration of NK cells [58]. IL-2-activated human NK cells can induce the chemotaxis of
Little has been published regarding the ability of chemo- other NK cells. Moreover, interaction between NK cells and
kines to costimulate NK cell proliferation in response to pri- NK-sensitive K562 target cells stimulated greater production
mary mitogens. The two known CCR7 ligands, CCL19 and of CCL3, CCL4, and CCL5 [37, 66] and a concomitant increase
CCL21, can costimulate IL-2-induced proliferation of CD56dim in the migration of bystander NK cells [37]. These data suggest
NK cells [40]. Such costimulation was dose-dependent and was that NK cell interaction with target cells can stimulate secre-
of moderate magnitude. No effect of CCL19 or CCL21 on the tion of chemokines that promote the recruitment of additional
more robust proliferation of CD56bright NK cells in response to NK cells to sites of infected or transformed cells.
IL-2 was detected [40]. Moreover, CCL20 in doses as high as CCR5 is a coreceptor for macrophage-tropic (M-tropic)
1000 ng/ml did not affect IL-2-induced proliferation of strains of HIV-1 [25]. Ligands for CCR5, including CCL3,
CD56dim or CD56bright NK cells. CCL4, and CCL5, have been shown to inhibit the infection of
human cells by M-tropic HIV-1 strains. Because activated
human NK cells can produce CCL3, CCL4, and CCL5, it is
CHEMOKINES PRODUCED BY NK CELLS possible that NK cells could be manipulated with therapeutic
intent in the treatment of HIV-1 infection. Indeed, activated
In addition to responding to numerous chemokines, NK cells NK cells from HIV-infected persons have been shown to sup-
have been found to produce several chemokines (Table 6). press HIV replication in autologous lymphocytes in vitro [66].
Human peripheral blood NK cells cultured in the absence of Furthermore, HIV-1 replication in normal PBL is inhibited
deliberate stimuli can spontaneously produce CCL4, CCL5, potently in vitro by supernatants of activated NK cells from
and CCL22 in vitro [37, 65– 68]. Unstimulated normal human HIV-infected or normal control donors [65]. The effects of
NK cells express mRNA for CCL3 [66], but appear to produce activated NK cells or their supernatants on HIV replication are
little if any CCL3 protein [65, 69]. Resting human NK cells partially but not completely reversed by the presence of neu-
also express mRNA for XCL1 [35, 70], but XCL1 protein was tralizing antibodies to CCL3, CCL4, and CCL5 [65, 66]. Thus,
not detected in the cytoplasm of unstimulated murine NK cells NK cells may inhibit HIV replication by secreting CCL3,
[54]. CD56bright CD16⫺ NK cells isolated from human uterine CCL4, and CCL5 as well as other chemokines or soluble-
decidua spontaneously express CXCL8 mRNA and secrete effector molecules.

178 Journal of Leukocyte Biology Volume 71, February 2002 http://www.jleukbio.org


PARTICIPATION OF CHEMOKINES IN NK Leishmania major is an obligate intracellular pathogen that
CELL RESPONSES TO INFECTION can cause cutaneous or disseminated disease. Resistant strains
AND CANCER of mice, such as C57BL/6, develop a Th1-dominated immune
response to this pathogen and recover from infection [4, 80,
Elegant work has demonstrated the crucial role of chemokines 81]. Conversely, susceptible strains, such as Balb/c, develop
in the adaptive immune response and the biology of T and B Th2 responses and succumb to disseminated disease. It has
lymphocytes [22–26]. For example, antigen-specific T- and been shown that IFN-␥ produced by activated NK cells in
B-cell responses are impaired severely in CCR7-deficient mice reactive lymph nodes promotes protective Th1 immune re-
[77]. This impairment does not appear to be caused by intrinsic sponses to L. major [4]. Moreover, parasite burden is correlated
defects in mature T or B lymphocytes, but rather the failure of inversely with the level of NK cell cytotoxicity present in
T cells, B cells, and dendritic cells to migrate appropriately to regional lymph nodes. Susceptibility of Balb/c mice appears to
secondary lymphoid tissues [77]. In contrast, the effects of be a result of, at least in part, defective NK cell activation at
deficiencies of specific chemokines or chemokine receptors on the site of infection as a consequence of inadequate, local
NK cell development and function have not been described in IL-12 production by macrophages [80]. However, failure to
detail. Therefore, the role of chemokines in NK cell biology in activate NK cells properly in draining lymph nodes could also
vivo must be inferred from indirect evidence. contribute to fatal leishmaniasis. Following infection with
An exception to this limitation is the clearly defined role of Leishmania, lower NK cell cytotoxicity is observed in the
CCL3 in NK cell-mediated protection from MCMV. NK cells draining lymph nodes of susceptible Balb/c mice as compared
become activated in vivo early in the course of MCMV infec- with resistant C57BL/6 mice [4, 82], although the number of
tion, and protective antiviral immune responses require IFN-␥ NK cells present does not differ between the two strains [82].
produced by NK cells [5]. Migration of NK cells into the liver Message for XCL1, CCL2, and CXCL10 is expressed in drain-
is necessary for successful clearance of MCMV infection [6, ing lymph nodes from resistant but not susceptible mice during
55]. Expression of CCL3 mRNA and protein is induced in the the early phases of Leishmania infection [82]. Furthermore,
murine liver following MCMV infection, and migration of adop- local injection of CXCL10 in vivo augmented NK cytotoxicity
tively transferred NK cells to the liver of MCMV-infected mice in the draining lymph nodes of Balb/c mice infected with
is abrogated by administration of neutralizing anti-CCL3 anti- Leishmania [82]. Thus, defective production CXCL10 and
body [55]. Moreover, migration of NK cells to the liver, local other chemokines in regional lymph nodes, with subsequent,
IFN-␥ production, and NK cell-dependent antiviral protection inadequate stimulation of NK cell cytotoxicity and/or cytokine
are markedly diminished in CCL3-deficient mice infected with production, may contribute to the susceptibility of Balb/c mice
MCMV [6, 55]. CCL3-deficient mice uniformly succumb to to Leishmania infection.
MCMV infection, whereas wild-type mice control the infection In addition to their crucial role in the response to some
and survive. Serum IFN-␥ levels are the same in CCL3- obligate intracellular pathogens, NK cells can contribute to the
deficient and wild-type mice, indicating that local IFN-␥ pro- rejection of malignant tumors. Preclinical animal models indi-
duction in the liver is critical for control of MCMV infection. cate that chemokines participate in the NK cell response to
Abundant production of CXCL9 occurred in the livers of cancer. Systemic administration of IL-12 can induce the com-
wild-type but not CCL3-deficient mice after MCMV infection plete regression of established tumors, inhibit the formation of
[6]. Depletion of NK cells strongly inhibited this CXCL9 distant metastases, and prolong the survival of tumor-bearing
production. Furthermore, CXCL9 production was not detected mice [83]. Depending on the experimental system used, CD4 T
in the livers of IFN-␥-deficient mice infected with MCMV. cells, CD8 T cells, and NK cells have been found to contribute
Therefore, it is likely that local secretion of IFN-␥ in the liver to the antitumor activity of IL-12. Furthermore, production of
by activated NK cells is responsible for CXCL9 production IFN-␥ in vivo is necessary but not sufficient for the antitumor
during MCMV infection. Furthermore, neutralization of CXCL9 effects of IL-12 [84, 85]. IFN-␥ secreted by IL-12-activated T
in vivo was associated with a marked increase in MCMV titers and NK cells can in turn stimulate the production of several
in the liver and a fatal outcome [6]. Thus, protective immune cytokines including CXCL9 and CXCL10. The latter are re-
responses to MCMV require expression of CCL3 in the liver to quired for tumor regression during IL-12 therapy [86]. The
attract activated NK cells, local IFN-␥ secretion by activated antitumor effects of CXCL9 and CXCL10 are due to recruit-
NK cells, and subsequent production of CXCL9 in the liver. ment of CXCR3-bearing antitumor-effector cells as well as the
Local production of CXCL9 and CXCL10 can also stimulate inhibition of tumor angiogenesis [86 – 89]. Thus, CXCL9,
NK cell-dependent protective responses to recombinant vac- CXCL10, and possibly other chemokines can participate in NK
cinia viruses in vivo [78]. cell-mediated antitumor effects during cytokine-based immu-
The cell types in the liver that produce CXCL9 during notherapy for cancer.
MCMV infection have not been identified clearly, but hepato- Systemic administration of immunostimulatory cytokines
cytes can produce this chemokine [79]. Moreover, a fourfold can be associated with substantial toxicity [90]. An alternative
increase in liver NK cell numbers was observed in MCMV- approach for cancer immunotherapy is vaccination with cyto-
infected as compared with uninfected CCL3-deficient mice [6]. kine gene-transduced tumor cells. Injection of tumor cells
Although this degree of NK cell infiltration is apparently not transduced with genes encoding one or more of several immu-
sufficient for ultimate control of MCMV, it suggests that factors nostimulatory cytokines can stimulate the rejection of estab-
other than CCL3 can stimulate NK cell migration into the liver lished, nontransduced tumors and promote durable, specific
in vivo. antitumor immunity [91, 92]. Antitumor effects have also been

Robertson Chemokine role in biology of NK cells 179


observed after vaccination with chemokine gene-transduced secondary lymphoid tissues [30, 41, 100 –103]. Because
tumor cells. Transduction of the C26 murine-colon adenocar- CD56bright NK cells produce large quantities of IFN-␥ and
cinoma cell line with a cDNA encoding CCL21 does not affect other cytokines after activation [11, 16, 62], it is also reason-
the in vitro growth of the malignant cells but does reduce their able to hypothesize that they can regulate adaptive T- and
tumorigenicity in vivo [93]. Depletion of CD8 T cells or NK B-cell responses in secondary lymphoid tissues. In contrast,
cells in vivo led to more rapid growth of tumors, indicating that resting CD56dim NK cells do not express CCR7 and express
both lymphocyte subsets participate in the response to CCL21- little or no L-selectin [11, 30, 100]; however, they express high
transduced C26 cells. CCR7 and CXCR3 are receptors for levels of LFA-1 and other adhesion molecules [17, 30, 40,
CCL21 in the mouse [39, 94]. Expression of CCR7 mRNA but 100]. Thus, CD56dim NK cells are expected to migrate to
not CXCR3 mRNA was increased dramatically in tumors peripheral nonlymphoid tissues rather than secondary lym-
formed by CCL21-transduced C26 cells as compared with phoid organs. However, CD56dim NK cells express CCR7 after
nontransduced cells [93]. These results suggest that paracrine activation [40], which could promote their migration to regional
secretion of CCL21 by transduced C26 cells recruits CCR7- lymph nodes after they have been stimulated at sites of inflam-
expressing effector cells, which then inhibit tumor cell growth. mation in peripheral tissues. Chemokines can also affect the
CCR7 is a receptor for CCL19 as well as CCL21. Transduc- cytolytic activity and proliferation of NK cells, potentially
tion of the C3L5 murine-breast adenocarcinoma cell line with indicating a major role for chemokines in the regulation of NK
a cDNA encoding CCL19 substantially reduces its tumorige- cell responses to tumors and infectious pathogens. Further
nicity in vivo without affecting its in vitro growth [95]. NK cells investigation is required to dissect the contribution of chemo-
and to a lesser degree CD4 T cells contribute to the rejection kines to the migration and effector function of NK cells in vivo.
of CCL19-transduced C3L5 cells; CD8 T cells do not appear to Such studies will further our understanding of basic NK cell
be involved. Vaccination with CCL19-transduced tumor cells biology and may facilitate the manipulation of NK cells in the
does not confer protection from subsequent rechallenge with treatment of human infectious and neoplastic diseases.
nontransduced C3L5 cells, although the latter grow more
slowly in vaccinated compared with nonvaccinated animals
[95]. In contrast to the initial rejection of CCL19-transduced ACKNOWLEDGMENTS
cells, partial immunity to rechallenge with nontransduced
C3L5 appears to be mediated entirely by CD4 T cells, without The author is supported in part by a National Institutes of
participation of NK cells or CD8 T cells. Health grant 3MO1 RR00750-27S3. I am grateful to Barrett
In preclinical models using the Meth A fibrosarcoma or Rollins, M.D., Ph.D., Hal Broxmeyer, Ph.D., and Robert Hro-
HM-1 ovarian carcinoma cell lines, injection with tumor cells mas, M.D., for providing helpful comments and suggestions.
transduced with cDNA encoding CCL19, CCL21, or CXCL12
appeared to augment antitumor immune responses evoked by
vaccination with tumor cells expressing IL-2 or granulocyte- REFERENCES
macrophage colony-stimulating factor (GM-CSF) [96]. In
agreement with the C26 and C3L5 models [93, 95], vaccination 1. Robertson, M. J., Ritz, J. (1990) Biology and clinical relevance of human
with chemokine-expressing tumor cells alone did not stimulate natural killer cells. Blood 76, 2421–2438.
2. Timonen, T. (1997) Natural killer cells: endothelial interactions, migra-
durable, protective antitumor immunity [96]. tion, and target cell recognition. J. Leukoc. Biol. 62, 693–701.
Transduction of the CCL2 gene has also been demonstrated 3. Trinchieri, G. (1989) Biology of natural killer cells. Adv. Immunol. 47,
to alter the tumorigenicity or immunogenicity of malignant 187–376.
4. Scharton, T. M., Scott, P. (1993) Natural killer cells are a source of
cells in several models [97–99]. Suppression of tumor forma- interferon ␥ that drives differentiation of CD4⫹ T cell subsets and
tion [98] and metastasis [99] by CCL2-transduced cells in induces early resistance to Leishmania major in mice. J. Exp. Med. 178,
T-cell-deficient nu/nu or SCID mice suggests a response by 567–577.
5. Orange, J. S., Wang, B., Terhorst, C., Biron, C. A. (1995) Requirement
innate immune effectors such as NK cells and monocytes. for natural killer cell-produced interferon ␥ in defense against murine
Indeed, NK cells have been shown to mediate the inhibition of cytomegalovirus infection and enhancement of this defense pathway by
metastasis by CCL2-transduced human lung adenocarcinoma interleukin 12 administration. J. Exp. Med. 182, 1045–1056.
6. Salazar-Mather, T. P., Hamilton, T. A., Biron, C. A. (2000) A chemokine-
cells in the SCID mouse model [99]. to-cytokine-to-chemokine cascade critical in antiviral defense. J. Clin.
Investig. 105, 985–993.
7. Abbas, A. K., Murphy, K. M., Sher, A. (1996) Functional diversity of
helper T lymphocytes. Nature 383, 787–793.
CONCLUDING REMARKS 8. Gumperz, J. E., Parham, P. (1995) The enigma of the natural killer cell.
Nature 378, 245–248.
Chemokines have been shown to play a central role in the 9. Lanier, L. L. (1997) Natural killer cells: from no receptors to too many.
Immunity 6, 371–378.
migration and trafficking of T and B lymphocytes in vivo 10. Moretta, A., Bottino, C., Vitale, M., Pende, D., Cantoni, C., Mingari,
[22–27]. It is likely that chemokines have a similar role in the M. C., Biassoni, R., Moretta, L. (2001) Activating receptors and core-
biology of NK cells. Differential expression of chemokine re- ceptors involved in human natural killler cell-mediated cytolysis. Annu.
Rev. Immunol. 19, 197–223.
ceptors [30] and adhesion molecules [30, 100, 101] by 11. Nagler, A., Lanier, L. L., Cwirla, S., Phillips, J. H. (1989) Comparative
CD56bright and CD56dim human NK cells suggests that these studies of human FcR III-positive and negative natural killer cells.
subsets may home to different microenvironments in vivo [14]. J. Immunol. 143, 3183–3191.
12. Caligiuri, M. A., Zmuidzinas, A., Manley, T. J., Levine, H., Smith, K. A.,
Indeed, expression of CCR7 and high levels of L-selectin by Ritz, J. (1990) Functional consequences of interleukin 2 receptor ex-
resting CD56bright NK cells [11, 30, 100] should direct them to pression on resting human lymphocytes: identification of a novel natural

180 Journal of Leukocyte Biology Volume 71, February 2002 http://www.jleukbio.org


killer cell subset with high affinity receptors. J. Exp. Med. 171, 1509 – 35. Inngjerdingen, M., Damaj, B., Maghazachi, A. A. (2001) Expression and
1526. regulation of chemokine receptors in human natural killer cells. Blood
13. Baume, D. M., Robertson, M. J., Levine, H., Manley, T. J., Schow, P. W., 97, 367–375.
Ritz, J. (1992) Differential responses to interleukin-2 define functionally 36. Taub, D. D., Sayers, T. J., Carter, C. R. D., Ortaldo, J. R. (1995) ␣ and
distinct subsets of human natural killer cells. Eur. J. Immunol. 22, 1– 6. ␤ Chemokines induce NK cell migration and enhance NK-mediated
14. Cooper, M. A., Fehniger, T. A., Caligiuri, M. A. (2001) The biology of cytolysis. J. Immunol. 155, 3877–3888.
human natural killer cell subsets. Trends Immunol. 22, 633– 640. 37. Nieto, M., Navarro, F., Perez-Villar, J. J., del Pozo, M. A., Gonzalez-
15. Carson, W. E., Giri, J. G., Lindemann, M. J., Linett, M. L., Ahdieh, M., Amaro, R., Mellado, M., Frade, J. M. R., Martinez-A, C., Lopez-Botet,
Paxton, R., Anderson, D., Eisenmann, J., Grabstein, K., Caligiuri, M. A. M., Sanchez-Madrid, F. (1998) Role of chemokines and receptor polar-
(1994) Interleukin (IL) 15 is a novel cytokine that activates human ization in NK-target cell interactions. J. Immunol. 161, 3330 –3339.
natural killer cells via components of the IL-2 receptor. J. Exp. Med. 180, 38. Polentarutti, N., Allavena, P., Bianchi, G., Giardina, G., Basile, A.,
1395–1403. Sozzani, S., Montovani, A., Introna, M. (1997) IL-2-regulated expression
16. Cooper, M. A., Fehniger, T. A., Turner, S. C., Chen, K. S., Ghaheri, B. A., of monocyte chemotactic protein-1 receptor (CCR2) in human NK cells:
Ghayur, T., Carson, W. E., Caligiuri, M. A. (2001) Human natural killer characterization of a predominant 3.4-kilobase transcript containing
cells: a unique immunoregulatory role for the CD56bright subset. Blood CCR2B and CCR2A sequences. J. Immunol. 158, 2689 –2694.
97, 3146 –3151. 39. Campbell, J. J., Bowman, E. P., Murphy, K., Youngman, K. R., Siani,
17. Robertson, M. J., Caligiuri, M. A., Manley, T. J., Levine, H., Ritz, J. M. A., Thompson, D. A., Wu, L., Zlotnick, A., Butcher, E. C. (1998)
6-C-kine (SLC), a lymphocyte adhesion-triggering chemokine expressed
(1990) Human natural killer cell adhesion molecules: differential ex-
by high endothelium, is an agonist for the MIP-3␤ receptor CCR7. J. Cell
pression after activation and participation in cytolysis. J. Immunol. 145,
Biol. 141, 1053–1059.
3194 –3201.
40. Robertson, M. J., Williams, B. T., Christopherson II, K., Brahmi, Z.,
18. Robertson, M. J., Soiffer, R. J., Wolf, S. F., Manley, T. J., Donahue, C.,
Hromas, R. (2000) Regulation of human natural killer cell migration and
Young, D., Herrmann, S. H., Ritz, J. (1992) Response of human natural proliferation by the exodus subfamily of CC chemokines. Cell. Immunol.
killer (NK) cells to NK cell stimulatory factor (NKSF): cytolytic activity 199, 8 –14.
and proliferation of NK cells is differentially regulated by NKSF. J. Exp. 41. Yoshida, R., Nagira, M., Imai, T., Baba, M., Tagaki, S., Tabira, Y., Akagi,
Med. 175, 779 –788. J., Nomiyama, H., Yoshie, O. (1998) EBI1-ligand chemokine (ELC)
19. Robertson, M. J., Cameron, C., Atkins, M. B., Gordon, M. S., Lotze, M. T., attracts a broad spectrum of lymphocytes: activated T cells strongly
Sherman, M. L., Ritz, J. (1999) Immunologic effects of interleukin 12 up-regulate CCR7 and efficiently migrate toward ELC. Int. Immunol. 10,
administered by bolus intravenous injection to patients with cancer. Clin. 901–910.
Cancer Res. 5, 9 –16. 42. Kim, C. H., Pelus, L. M., Appelbaum, E., Johanson, K., Anzai, N.,
20. Caligiuri, M. A., Murray, C., Robertson, M. J., Wang, E., Cochran, K., Broxmeyer, H. E. (1999) CCR7 ligands, SLC/6Ckine/Exodus2/TCA4 and
Cameron, C., Schow, P., Ross, M. E., Klumpp, T. R., Soiffer, R. J., Smith, CK␤-11/MIP-3␤/ELC, are chemoattractants for CD56(⫹)CD16(⫺) NK
K., Ritz, J. (1993) Selective modulation of human natural killer cells in cells and late stage lymphoid progenitors. Cell. Immunol. 193, 226 –235.
vivo after prolonged infusion of low dose recombinant interleukin 2. 43. Handgetinger, R., Welte, B., Dopfer, R., Niethammer, D. (1994) Rapid
J. Clin. Investig. 91, 123–132. method for purification of CD56⫹ natural killer cells with preferential
21. Ellis, T. M., Creekmore, S. P., McMannis, J. D., Braun, D. P., Harris, enrichment of the CD56bright⫹ subset. J. Clin. Lab. Anal. 8, 443– 446.
J. A., Fisher, R. I. (1988) Appearance and phenotypic characterization of 44. Allavena, P., Bianchi, G., Zhou, D., van Damme, J., Jilek, P., Sozzani, S.,
circulating Leu 19⫹ cells in cancer patients receiving recombinant Mantovani, A. (1994) Induction of natural killer cell migration by mono-
interleukin 2. Cancer Res. 48, 6597– 6602. cyte chemotactic protein-1, -2, and -3. Eur. J. Immunol. 24, 3233–3236.
22. Rollins, B. J. (1997) Chemokines. Blood 90, 909 –928. 45. Maghazachi, A. A., Skalhegg, B. S., Rolstad, B., Al-Aoukaty, A. (1997)
23. Rossi, D., Zlotnik, A. (2000) The biology of chemokines and their Interferon-inducible protein-10 and lymphotactin induce the chemotaxis
receptors. Annu. Rev. Immunol. 18, 217–242. and mobilization of intracellular calcium in natural killer cells through
24. Kim, C. H., Broxmeyer, H. E. (1999) Chemokines: signal lamps for pertussis toxin-sensitive and -insensitive heterotrimeric G-proteins.
trafficking of T and B cells for development and effector function. FASEB J. 11, 765–774.
J. Leukoc. Biol. 65, 6 –15. 46. Sebok, K., Woodside, D., Al-Aoukaty, A., Ho, A. D., Gluck, S., Maghaza-
25. Berger, E. A., Murphy, P. M., Farber, J. M. (1999) Chemokine receptors chi, A. A. (1993) IL-8 induces the locomotion of human IL-2-activated
as HIV-1 coreceptors: roles in viral entry, tropism, and disease. Annu. natural killer cells: involvement of a guanine binding (G0) protein. J.
Rev. Immunol. 17, 657–700. Immunol. 150, 1524 –1534.
26. Christopherson II, K., Hromas, R. (2001) Chemokine regulation of nor- 47. Loetscher, P., Seitz, M., Clark-Lewis, I., Bagglioni, M., Moser, B. (1996)
mal and pathologic immune responses. Stem Cells 19, 388 –396. Activation of NK cells by CC chemokines: chemotaxis, Ca2⫹ mobiliza-
27. Yoshie, O., Imai, T., Nomiyama, H. (2001) Chemokines in immunity. tion, and enzyme release. J. Immunol. 156, 322–327.
Adv. Immunol. 78, 57–110. 48. Al-Aoukaty, A., Schall, T. J., Maghazachi, A. (1996) Differential cou-
28. Rollins, B. J., ed. (1999) Chemokines and Cancer, Totowa, NJ, Humana. pling of CC chemokine receptors to multiple heterotrimeric G proteins in
29. Murphy, P. M., Baggiolini, M., Charo, I. F., Hebert, C. A., Horuk, R., human interleukin-2-activated natural killer cells. Blood 87, 4255–
Matsushima, K., Miller, L. H., Oppenheim, J. J., Power, C. A. (2000) 4260.
49. Romagnani, P., Annunziato, F., Lazzeri, E., Cosmi, L., Beltrame, C.,
Nomenclature for chemokine receptors. Pharmacol. Rev. 52, 145–176.
Lasagni, L., Galli, G., Francalanci, M., Manetti, R., Marra, F., Vanini, V.,
30. Campbell, J. J., Qin, S., Unutmaz, D., Soler, D., Murphy, K. E., Hodge,
Maggi, E., Romagnani, S. (2001) Interferon-inducible protein 10, mono-
M. R., Wu, L., Butcher, E. C. (2001) Unique subpopulations of CD56⫹
kine induced by interferon gamma, and interferon-inducible T-cell alpha
NK and NK-T peripheral blood lymphocytes identified by chemokine
chemoattractant are produced by thymic epithelial cells and attract
receptor expression. J. Immunol. 166, 6477– 6482.
T-cell receptor (TCR) ␣␤⫹ CD8⫹ single-positive T cells, TCR ␥␦⫹ T
31. Chuntharapai, A., Lee, J., Hebert, C. A., Kim, K. J. (1994) Monoclonal cells, and natural killer-type cells in human thymus. Blood 97, 601– 607.
antibodies detect different distribution patterns of IL-8 receptor A and 50. Bianchi, G., Sozzani, S., Zlotnick, A., Mantovani, A., Allavena, P. (1996)
IL-8 receptor B on human peripheral blood lymphocytes. J. Immunol. Migratory response of human natural killer cells to lymphotactin. Eur.
153, 5682–5688. J. Immunol. 26, 3238 –3241.
32. Morohashi, H., Miyawaki, T., Nomura, H., Kuno, K., Murakami, S., 51. Drake, P. M., Gunn, M. D., Charo, I. F., Tsou, C-L., Zhou, Y., Huang, L.,
Matsushima, K., Mukaida, N. (1995) Expression of both types of human Fisher, S. J. (2001) Human placental cytotrophoblasts attract monocytes
interleukin-8 receptors on mature neutrophils, monocytes, and natural and CD56bright natural killer cells via the actions of monocyte inflam-
killer cells. J. Leukoc. Biol. 57, 180 –187. matory protein 1␣. J. Exp. Med. 193, 1199 –1212.
33. Yoneda, O., Imai, T., Goda, S., Inoue, H., Yamauchi, A., Okazaki, T., 52. Inngjerdingen, M., Damaj, B., Maghazachi, A. A. (2000) Human NK cells
Imia, H., Yoshie, O., Bloom, E. T., Domae, N., Umehara, H. (2000) express CC chemokine receptors 4 and 8 and respond to thymus and
Fractalkine-mediated endothelial cell damage by NK cells. J. Immunol. activation-regulated chemokine, macrophage-derived chemokine, and
164, 4055– 4062. I-309. J. Immunol. 164, 4048 – 4054.
34. Imai, T., Hieshima, K., Haskell, C., Baba, M., Nagira, M., Nishimura, M., 53. Godiska, R., Chantry, D., Raport, C. J., Sozzani, S., Allavena, P., Leviten,
Kakizaki, M., Takagi, S., Nomiyama, H., Schall, T. J., Yoshie, O. (1997) D., Mantovani, A., Gray, P. W. (1997) Human macrophage-derived
Identification and molecular characterization of fractalkine receptor chemokine (MDC), a novel chemoattractant for monocytes, monocyte-
CX3CR1, which mediates both leukocyte migration and adhesion. Cell derived dendritic cells, and natural killer cells. J. Exp. Med. 185,
91, 521–530. 1595–1604.

Robertson Chemokine role in biology of NK cells 181


54. Hedrick, J. A., Saylor, V., Figueroa, D., Mizoue, L., Xu, Y., Menon, S., 74. Cosman, D., Mullberg, J., Sutherland, C. L., Chin, W., Armitage, R.,
Abrams, J., Handel, T., Zlotnick, A. (1997) Lymphotactin is produced by Fanslow, W., Kubin, M., Chalupny, N. J. (2001) ULBPs, novel MHC
NK cells and attracts both NK cells and T cells in vivo. J. Immunol. 158, class I-related molecules, bind to CMV glycoprotein UL16 and stimulate
1533–1540. NK cytotoxicity through NKG2D receptor. Immunity 14, 123–133.
55. Salazar-Mather, T. P., Orange, J. S., Biron, C. A. (1998) Early murine 75. Mainiero, F., Soriani, A., Stippoli, R., Jacobelli, J., Gismondi, A., Piccoli,
cytomegalovirus (MCMV) infection induces liver natural killer (NK) cell M., Frati, L., Santoni, A. (2000) RAC1/p38 MAPK signaling pathway
inflammation and protection through macrophage inflammatory protein controls ␤1 integrin-induced interleukin-8 production in human natural
1␣ (MIP-1␣)-dependent pathways. J. Exp. Med. 187, 1–14. killer cells. Immunity 12, 7–16.
56. Grimm, E. A., Mazumder, A., Zhang, H. Z., Rosenberg, S. A. (1982) 76. Somersalo, K., Carpen, O., Saksela, E. (1994) Stimulated natural killer
Lymphokine-activated killer cell phenomenon: lysis of natural killer- cells secrete factors with chemotactic activity, including NAP-1/IL-8,
resistant fresh solid tumor cells by interleukin 2-activated autologous which supports VLA-4- and VLA-5-mediated migration of T lympho-
human peripheral blood lymphocytes. J. Exp. Med. 155, 1823–1841. cytes. Eur. J. Immunol. 24, 2957–2965.
57. Taub, D. D., Ortaldo, J. R., Turcoviski-Corrales, S. M., Key, M. L., 77. Forster, R., Schubel, A., Breitfeld, D., Kremmer, E., Renner-Muller, I.,
Longo, D. L., Murphy, W. J. (1996) ␤ Chemokines costimulate lympho- Wolf, E., Lipp, M. (1999) CCR7 coordinates the primary immune re-
cyte cytolysis, proliferation, and cytokine production. J. Leukoc. Biol. 59, sponse by establishing functional microenvironments in secondary lym-
81– 89. phoid organs. Cell 99, 23–33.
58. Maghazachi, A., Al-Aoukaty, A., Schall, T. J. (1996) CC chemokines 78. Mahalingam, S., Farber, J. M., Karupiah, G. (1999) The interferon-
induce the generation of killer cells from CD56⫹ cells. Eur. J. Immunol. inducible chemokines MuMig and Crg-2 exhibit antiviral activity in vivo.
26, 315–319. J. Virol. 73, 1479 –1491.
59. Doherty, P. C. (1993) Cell-mediated cytotoxicity. Cell 75, 607– 612. 79. Park, J-W., Gruys, M. E., McCormick, K., Lee, J-K., Subleski, J.,
60. Robertson, M. J., Manley, T. J., Donahue, C., Levine, H., Ritz, J. (1993) Wigginton, J. M., Fenton, R. G., Wang, J-M., Wiltrout, R. H. (2001)
Costimulatory signals are required for optimal proliferation of human Primary hepatocytes from mice treated with IL-2/IL-12 produce T cell
natural killer cells. J. Immunol. 150, 1705–1714. chemoattractant activity that is dependent on monokine induced by
61. Robertson, M. J., Cameron, C., Lazo, S., Cochran, K. J., Voss, S. D., Ritz, IFN-␥ (Mig) and chemokine responsive to ␥-2 (Crg-2). J. Immunol. 166,
J. (1997) Costimulation of human natural killer cell proliferation: role of 3763–3770.
accessory cytokines and cell contact-dependent signals. Nat. Immunol. 80. Reiner, S. L., Zheng, S., Wang, Z-E., Stowring, L., Locksley, R. M.
15, 213–226. (1994) Leishmania promastigotes evade interleukin 12 (IL-12) induction
62. Voss, S. D., Daley, J., Ritz, J., Robertson, M. J. (1998) Participation of by macrophages and stimulate a broad range of cytokines from CD4⫹ T
the CD94 receptor complex in costimulation of human natural killer cells during initiation of infection. J. Exp. Med. 179, 447– 456.
cells. J. Immunol. 160, 1618 –1626. 81. Scharton-Kersten, T., Afonso, L. C. C., Wysocka, M., Trinchieri, G.,
63. Nandi, D., Gross, J. A., Allison, J. P. (1994) CD28-mediated costimula- Scott, P. (1995) IL-12 is required for natural killer cell activation and
tion is necessary for optimal proliferation of murine NK cells. J. Immu- subsequent T helper 1 development in experimental Leishmaniasis.
nol. 152, 3361–3369. J. Immunol. 154, 5320 –5330.
64. Rabinowich, H., Sedlmayr, P., Herberman, R. B., Whiteside, T. L. (1991) 82. Vester, B., Muller, K., Solbach, W., Laskay, T. (1999) Early gene
Increased proliferation, lytic activity, and purity of human natural killer expression of NK cell-activating chemokines in mice resistant to Leish-
cells cocultured with mitogen-activated feeder cells. Cell. Immunol. 135, mania major. Infect. Immun. 67, 3155–3159.
454 – 470. 83. Robertson, M. J., Ritz, J. (1996) Interleukin 12: basic biology and
65. Fehniger, T. A., Herbein, G., Yu, H., Para, M. I., Bernstein, Z. P., potential applications in cancer treatment. The Oncologist 1, 93–102.
O’Brien, W. A., Caligiuri, M. A. (1998) Natural killer cells from HIV-1⫹ 84. Nastala, C. L., Edington, H. D., McKinney, T. G., Tahara, H., Nalesnik,
patients produce C–C chemokines and inhibit HIV-1 infection. J. Im- M. A., Brunda, M. J., Gately, M. K., Wolf, S. F., Schreiber, R. D.,
munol. 161, 6433– 6438. Storkus, W. J., Lotze, M. T. (1994) Recombinant IL-12 administration
66. Oliva, A., Kinter, A. L., Vaccarezza, M., Rubbert, A., Catanzaro, A., induces tumor regression in association with IFN-␥ production. J. Im-
Moir, S., Monaco, J., Ehler, L., Mizell, S., Jackson, R., Li, Y., Romano, munol. 153, 1697–1706.
J. W., Fauci, A. S. (1998) Natural killer cells from human immunodefi- 85. Brunda, M. J., Luistro, L., Hendrzak, J. A., Fountoulakis, M., Garotta, G.,
ciency virus (HIV)-infected individuals are an important source of CC- Gately, M. K. (1995) Role of interferon-␥ in mediating the antitumor
chemokines and suppress HIV-1 entry and replication in vitro. J. Clin. efficacy of interleukin-12. J. Immunother. 17, 71–77.
Investig. 102, 223–231. 86. Tannenbaum, C. S., Tubbs, R., Armstrong, D., Finke, J., Bukowski,
67. Andrew, D. P., Chang, M. S., McNinch, J., Wathen, S. T., Rihanek, M., R. M., Hamilton, T. A. (1998) The CXC chemokines IP-10 and Mig are
Tseng, J., Spellberg, J. P., Elias III, C. G. (1998) STCP-1 (MDC) CC necessary for IL-12-mediated regression of the mouse RENCA tumor.
chemokine acts specifically on chronically activated Th2 lymphocytes J. Immunol. 161, 927–932.
and is produced by monocytes on stimulation with Th2 cytokines IL-4 87. Coughlin, C. M., Salhany, K. E., Gee, M. S., LaTemple, D. C., Kotenko,
and IL-13. J. Immunol. 161, 5027–5038. S., Ma, X., Gri, G., Wysocka, M., Kim, J. E., Liu, L., Laio, F., Farber,
68. Fehniger, T. A., Shah, M. H., Turner, M. J., VanDeusen, J. B., Whitman, J. M., Pestka, S., Trinchieri, G., Lee, W. M. F. (1998) Tumor cell
S. P., Cooper, M. A., Suzuki, K., Wechser, M., Goodsaid, F., Caligiuri, responses to IFN-␥ affect tumorigenicity and response to IL-12 therapy
M. A. (1999) Differential cytokine and chemokine gene expression by and antiangiogenesis. Immunity 9, 25–34.
human NK cells following activation with IL-18 or IL-15 in combination 88. Sgadari, C., Angiolillo, A. L., Tosato, G. (1996) Inhibition of angiogenesis
with IL-12: implications for the innate immune response. J. Immunol. by interleukin-12 is mediated by interferon-inducible protein 10. Blood
162, 4511– 4520. 87, 3877–3882.
69. Bluman, E. M., Bartynski, K. J., Avalos, B. R., Caligiuri, M. A. (1996) 89. Yao, L., Sgadari, C., Furuke, K., Bloom, E. T., Teruya-Feldstein, J.,
Human natural killer cells produce abundant macrophage inflammatory Tosato, G. (1999) Contribution of natural killer cells to inhibition of
protein-1␣ in response to monocyte-derived cytokines. J. Clin. Investig. angiogenesis by interleukin-12. Blood 93, 1612–1621.
97, 2722–2727. 90. Siegel, J. P., Puri, R. K. (1991) Interleukin-2 toxicity. J. Clin. Oncol. 9,
70. Henneman, B., Tam, Y. K., Tonn, T., Klingemann, H-G. (1999) Expres- 694 –704.
sion of SCM-1␣/lymphotactin and SCM-1␤ in natural killer cells is 91. Cornetta, K. G., Robertson, M. J. (2000) Basic principles of gene therapy:
upregulated by IL-2 and IL-12. DNA Cell Biol. 18, 565–571. basic principles and safety considerations. In Principles and Practice of
71. Saito, S., Kasahara, T., Sakakura, S., Enomoto, M., Umekage, H., the Biologic Therapy of Cancer (S. A. Rosenberg, ed.), Philadelphia,
Harada, N., Morii, T., Nishikawa, K., Narita, N., Ichijo, M. (1994) Lippincott, Williams and Wilkins, 733–747.
Interleukin-8 production by CD16⫺ CD56bright natural killer cells in the 92. Vieweg, J., Gilboa, E. (1995) Considerations for the use of cytokine-
human early pregnancy decidua. Biochem. Biophys. Res. Comm. 200, secreting tumor cell preparations for cancer treatment. Cancer Investig.
378 –383. 13, 193–201.
72. Kubin, M., Cassiano, L., Chalupny, J., Chin, W., Cosman, D., Fanslow, 93. Vicari, A. P., Ait-Yahia, S., Chemin, K., Mueller, A., Zlotnik, A., Caux,
W., Mullberg, J., Rousseau, A-M., Ulrich, D., Armitage, R. (2001) C. (2000) Antitumor effects of the mouse chemokine 6Ckine/SLC through
ULBP1, 2, 3: novel MHC class I-related molecules that bind to human angiostatic and immunological mechanisms. J. Immunol. 165, 1992–
cytomegalovirus glycoprotein UL16, activate NK cells. Eur. J. Immunol. 2000.
31, 1428 –1437. 94. Soto, H., Wang, W., Strieter, R. M., Copeland, N. G., Gilbert, D. J.,
73. Ortaldo, J. J., Bere, E. W., Hodge, D., Young, H. A. (2001) Activating Jenkins, N. A., Hedrick, J., Zlotnick, A. (1998) The CC chemokine
Ly-49 NK receptors: central role in cytokine and chemokine production. 6Ckine binds the CXC chemokine receptor CXCR3. Proc. Natl. Acad.
J. Immunol. 166, 4994 – 4999. Sci. USA 95, 8205– 8210.

182 Journal of Leukocyte Biology Volume 71, February 2002 http://www.jleukbio.org


95. Braun, S. E., Chen, K., Foster, R. G., Orchard, P. J., Kim, C. H., Hromas, R., 101. Andre, P., Spertini, O., Guia, S., Rihet, P., Dignat-George, F., Brailly, H.,
Kaplan, M. H., Broxmeyer, H. E., Cornetta, K. (2000) The CC chemokine Sampol, J., Anderson, P. J., Vivier, E. (2000) Modification of P-selectin
CK␤-11/MIP-3␤/ELC/Exodus 3 mediates tumor rejection of murine glycoprotein ligand-1 with a natural killer cell-restricted sulfated lac-
breast cancer cells through NK cells. J. Immunol. 164, 4025– 4031. tosamine creates an alternate ligand for L-selectin. Proc. Natl. Acad. Sci.
96. Nomura, T., Hasegawa, H., Kohno, M., Sasaki, M., Fujita, S. (2001) USA 97, 3400 –3405.
Enhancement of anti-tumor immunity by tumor cells transfected with the 102. Gunn, M. D., Tangemann, K., Tam, C., Cyster, J. G., Rosen, S. D.,
secondary lymphoid tissue chemokine, EBI-1-ligand chemokine and Williams, L. T. (1998) A chemokine expressed in lymphoid high endo-
stromal cell-derived factor-1␣ chemokine genes. Int. J. Cancer 91, thelial venules promotes the adhesion and chemotaxis of naive T lym-
597– 606. phocytes. Proc. Natl. Acad. Sci. USA 95, 258 –263.
97. Manome, Y., Wen, P. Y., Hershowitz, A., Tanaka, T., Rollins, B. J., Kufe, 103. Rossi, D. L., Vicari, A. P., Franz-Bacon, K., McClanahan, T. K., Zlot-
D. W., Fine, H. A. (1995) Monocyte chemoattractant protein-1 (MCP-1) nick, A. (1997) Identification through bioinformatics of two new macro-
gene transduction: an effective tumor vaccine strategy for non-intracra-
phage proinflammatory human chemokines: MIP-3␣ and MIP-3␤. J. Im-
nial tumors. Cancer Immunol. Immunother. 41, 227–235.
munol. 158, 1033–1036.
98. Rollins, B. J., Sunday, M. E. (1991) Suppression of tumor formation in
vivo by expression of the JE gene in malignant cells. Mol. Cell. Biol. 11, 104. Qin, S., Rottman, J. B., Myers, P., Kassam, N., Weinblatt, M., Loetscher,
3125–3131. M., Koch, A. E., Moser, B., Mackay, C. R. (1998) The chemokine
99. Nokihara, H., Yanagawa, H., Nishioka, Y., Yano, S., Mukaida, N., receptors CXCR3 and CCR5 mark subsets of T cells associated with
Matsushima, K., Sone, S. (2000) Natural killer cell-dependent suppres- certain inflammatory reactions. J. Clin. Investig. 101, 746 –753.
sion of systemic spread of human lung adenocarcinoma cells by mono- 105. Maghazachi, A. A., Al-Aoukaty, A., Schall, T. J. (1994) C–C chemokines
cyte chemoattractant protein-1 gene transfection in severe combined induce the chemotaxis of NK and IL-2-activated NK cells. J. Immunol.
immunodeficient mice. Cancer Res. 60, 7002–7007. 153, 4969 – 4977.
100. Frey, M., Packianathan, N. B., Fehniger, T. A., Ross, M. E., Wang, W-C., 106. Robertson, M. J., Cochran, K. J., Cameron, C., Le, J-M., Tantravahi, R.,
Stewart, C. C., Caligiuri, M. A., Evans, S. S. (1998) Differential expres- Ritz, J. (1996) Characterization of a cell line, NKL, derived from an
sion and function of L-selectin on CD56bright and CD56dim NK cell aggressive human natural killer cell leukemia. Exp. Hematol. 24, 406 –
subsets. J. Immunol. 161, 400 – 408. 415.

Robertson Chemokine role in biology of NK cells 183

Você também pode gostar