Requirement of CCL17 for CCR7- and CXCR4-dependent migration of cutaneous dendritic cells
Edited by Richard A. Flavell, Yale University School of Medicine, Howard Hughes Medical Institute, New Haven, CT, and approved April 1, 2010 (received for review June 8, 2009)
Abstract
Chemokines are known to regulate the steady-state and inflammatory migration of cutaneous dendritic cells (DCs). The β-chemokine CCL17, a ligand of CCR4, is inducibly expressed in a subset of DCs and is strongly up-regulated in atopic diseases. Using an atopic dermatitis model, we show that CCL17-deficient mice develop acanthosis as WT mice, whereas dermal inflammation, T helper 2-type cytokine production, and the allergen-specific humoral immune response are significantly decreased. Notably, CCL17-deficient mice retained Langerhans cells (LCs) in the lesional skin after chronic allergen exposure, whereas most LCs emigrated from the epidermis of allergen-treated WT controls into draining lymph nodes (LNs). Moreover, CCL17-deficient LCs showed impaired emigration from the skin after exposure to a contact sensitizer. In contrast, the absence of CCR4 had no effect on cutaneous DC migration and development of atopic dermatitis symptoms. As an explanation for the major migratory defect of CCL17-deficient DCs in vivo, we demonstrate impaired mobility of CCL17-deficient DCs to CCL19/21 in 3D in vitro migration assays and a blockade of intracellular calcium release in response to CCR7 ligands. In addition, responsiveness of CCL17-deficient DCs to CXCL12 was impaired as well. We demonstrate that the inducible chemokine CCL17 sensitizes DCs for CCR7- and CXCR4-dependent migration to LN-associated homeostatic chemokines under inflammatory conditions and thus plays an important role in cutaneous DC migration.
Sign up for PNAS alerts.
Get alerts for new articles, or get an alert when an article is cited.
Chemokines represent a family of low molecular weight chemotactic cytokines and are classified as either constitutive or inducible (1). Constitutive chemokines are generally implicated in the homeostasis of the immune system, whereas inducible chemokines are expressed mainly during inflammatory processes. The migration of dendritic cells (DCs) into and out of the skin is coordinated by multiple chemokine–chemokine receptor pairs. Thus, immigration of immature DCs into the skin appears to be mediated through many different chemokine receptors, including CCR1, CCR2, CCR5, CCR6, CXCR1, CXCR2, and CXCR4 (2–4). In contrast, the homeostatic as well as inducible emigration of DCs from the skin to the skin-draining lymph nodes (LNs) has been shown to depend mainly on CCR7 and CXCR4 (4–6).
The inducible chemokine CCL17, previously termed thymus and activation-regulated chemokine (TARC) (7, 8), is strongly up-regulated through Toll-like receptor stimulation (9). CCL17 and the closely related chemokine CCL22 (MDC) (10, 11) bind to the receptor CCR4, which is known to be expressed on activated/memory T cells with a Th2 preference (12), regulatory T cells (13), Langerhans cells (LCs) (11), basophils, and macrophages (14). CCL17 and its receptor have been reported to play a role in endotoxic shock (14), systemic lupus erythematosus, Hodgkin's lymphoma, and pulmonary fibrosis. Using a reporter mouse model in which the ccl17 promoter drives expression of EGFP, we previously showed that CCL17 is specifically expressed by CD8α−CD11b+DEC205+ DCs. CCL17+ DCs are located in peripheral LNs, mucosa, and various nonlymphoid organs, but normally are not found in the spleen. LCs and dermal DCs (dDCs) in healthy normal skin are CCL17-negative, whereas CCL17 expression is induced in these cells after activation and migration to the skin-draining LNs (9). With the help of CCL17-deficient mice, we previously showed that contact hypersensitivity (CHS) responses are diminished in the absence of CCL17 (9).
Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by eczematous skin lesions, flexural lichenification, and intense pruritus (15). Both dysregulation of the immune system and disturbances of the epidermal barrier have been implicated in the etiology of AD (16). The prevalence of AD is particularly high in young children compared with adults (10–20% vs. 1–3%) (17). Recently reported data show enhanced expression of various proallergic and proinflammatory cytokines and chemokines in the lesional skin and serum of AD patients (18). This also applies to the chemokine CCL17, the serum levels of which directly correlate with the severity of the disease (19, 20). The strong correlation of CCL17 expression with disease severity in AD patients prompted us to directly investigate the role of CCL17 in cutaneous immune responses. Using a mouse model of AD, we demonstrate that both local skin inflammation and the systemic humoral immune response are strongly reduced in CCL17-deficient mice compared with WT and CCR4-deficient mice. We provide an unanticipated explanation for the immunostimulatory function of CCL17, demonstrating that this chemokine plays an essential role in the emigration of DCs from the skin to the skin-draining LNs.
Results
Presence of Acanthosis but Reduced Dermal Inflammation in CCL17-Deficient Mice.
To determine the influence of CCL17 on the development of AD, we treated heterozygous CCL17/EGFP (CCL17E/+) reporter mice and homozygous CCL17-deficient (CCL17E/E) mice with three cycles of tape-stripping and application of an ovalbumine (OVA)-containing patch on the skin of the back (21, 22). Mice treated with tape-stripping but receiving a patch soaked with NaCl alone were used as negative controls. In general, CCL17E/+ mice responded like normal BALB/c mice and were used as controls (Fig. S1). Mechanical stress induced by tape-stripping and application of a NaCl-soaked patch led to only minor thickening of the epidermis. When the model allergen OVA was applied to the patch on the mechanically stressed skin, acanthosis was induced in both CCL17E/+ and CCL17E/E mice to a similar extent (Fig. S2 A and B), indicating that CCL17 has no influence on keratinocyte proliferation. In contrast, thickening of the dermis as a measure of leukocytic inflammation was strikingly diminished in the absence of CCL17 (Fig. S2 A and C). Mast cell and eosinophil infiltration also were significantly lower in the lesional skin of CCL17E/E mice compared with OVA-treated CCL17E/+ controls (Figs. S2 F and G and S3A). To test whether an absence of CCR4 also leads to a similar amelioration of AD symptoms, CCR4-KO mice and C57BL/6 WT mice also were analyzed in the AD model. In contrast to CCL17E/E mice, CCR4KO mice developed allergic skin inflammation to the same extent as WT mice, with strong thickening of both the epidermis and the dermis (Fig. S2 D and E).
As reported previously, CCL17 is not expressed in normal healthy skin, whereas both LCs and dDCs up-regulate CCL17 after maturation and entry into the skin-draining LNs (9). Using the AD mouse model, we tested whether mechanical irritation and OVA treatment were sufficient to induce CCL17 expression within the affected skin. While CCL17/EGFP-expressing cells were present at low frequency in the skin of saline-treated mice, a much higher frequency of dispersed EGFP+ cells was detected in the dermis of the OVA-treated mice (Fig. S3B). Because the detection of EGFP+ LC in histological sections is inefficient for technical reasons, we also prepared epidermal sheets from dinitrofluorobenzene (DNFB)-sensitized ears to demonstrate that LCs also up-regulate CCL17/EGFP expression in the epidermis after sensitization (Fig. S3C).
Infiltrating CD4+ cells were significantly reduced, but not entirely absent, in OVA-treated CCL17E/E mice compared with OVA-treated CCL17E/+ mice (Fig. S4 A and B). In contrast, CCR4-deficient mice demonstrated dermal infiltration with CD4+ cells similar to that in WT mice after OVA treatment for AD (Fig. S4C). The number of FcεRI-expressing cells also was significantly reduced in OVA-treated CCL17E/E mice compared with OVA-treated CCL17E/+ mice (Fig. S4 D and E). Taken together, these findings indicate that the allergic reaction was strongly ameliorated in the absence of CCL17, whereas an absence of CCR4 had no significant impact on the development of allergic skin inflammation.
Enhancement of the Allergen-Specific Humoral Immune Response by CCL17.
In agreement with previous reports using the AD mouse model (22), we observed a 5-fold increase in total serum IgE level in OVA-treated CCL17E/+ mice compared with saline-treated CCL17E/+ mice (Fig. S5A). In contrast, the total serum IgE level was ∼50% lower in OVA-treated CCL17E/E mice compared with OVA-treated CCL17E/+ controls. More specifically, we also assessed OVA-specific IgE levels and observed a significant reduction in the sera of OVA-treated CCL17E/E mice compared with OVA-treated CCL17E/+ mice (Fig. S5A). The OVA-specific IgG1 and IgG2a titers also were significantly lower in OVA-treated CCL17E/E mice compared with OVA-treated CCL17E/+ mice (Fig. S5A). This indicates that CCL17 plays an essential role in the induction of allergen-specific antibody responses in the AD mouse model.
To gain insight into the cytokine response pattern of lesional skin, we analyzed cytokine expression by quantitative RT-PCR. In contrast to cytokine production in OVA-treated CCL17E/+ mice, levels of Th2-related cytokines (e.g., IL-4, IL-13, IL-10) were significantly reduced in OVA-treated CCL17-deficient mice and were barely distinguishable from those in saline-treated mice (Fig. S5C). After chronic OVA-treatment, the Th1-cytokine IFN-γ also was significantly decreased in CCL17E/E mice compared with CCL17E/+ mice. OVA-treated CCL17E/+ mice showed a strong increase in IL-1β expression in the skin compared with saline-treated CCL17E/+ mice, whereas IL-1β levels were not significantly different in OVA-treated and saline-treated CCL17E/E mice. In line with the severe thickening of the dermis in OVA-treated CCR4KO mice, these mice developed OVA-induced humoral immune responses (Fig. S5B) and local cytokine production (Fig. S5D) similar to what was seen in WT controls. Taken together, we can conclude that Th1- and Th2-cytokine expression is strongly diminished in the lesional skin of CCL17-deficient mice, but not CCR4KO mice, compared with WT mice.
Impaired Migration of DCs from Lesional Skin in CCL17-Deficient Mice.
The total cellularity of LNs draining the site of skin inflammation was enhanced by more than 3-fold in OVA-treated CCL17E/+ mice and about 2-fold in CCL17E/E mice compared with the respective NaCl-treated control mice (Fig. S6A). The absolute numbers of CD11c+ DCs in these LNs increased by 4- to 5-fold in CCL17E/+ mice, but by only 2-fold in CCL17-deficient mice compared with the respective saline-treated mice (Fig. S6C). We found no major changes in the frequency of CD11c+ DCs in the draining LNs (Fig. S6B). To directly quantify the number of skin-derived DCs, we also determined the absolute numbers of LC and dDC descendants in the LNs, based on high expression of MHC class II (MHCII) and differential expression levels of CD24a (23). Both dDCs and LCs (Fig. S6 D and E) were significantly reduced in the draining LNs of CCL17E/E mice compared with CCL17E/+ mice after OVA treatment. In terms of LN size and numbers of skin-derived DCs, again no difference was found between OVA-treated WT and CCR4KO mice (Fig. S6 F–J). Considering that the migration of LCs and dDCs from the inflamed skin to the draining LNs initiates further recruitment of lymphocytes and enlargement of the LNs, we asked whether migration of the DCs themselves was impaired in CCL17-deficient mice. Therefore, we stained sections of the affected skin from all four groups of mice for MHCII and CD11c. Application of a NaCl-soaked patch to the mechanically stressed skin did not induce emigration of LCs in either CCL17E/+ or CCL17E/E mice (Fig. 1 A and B; Fig. S7 A and B). In contrast, after treatment with the OVA-containing patch, the frequency of LCs was reduced by 50% in CCL17E/+ mice (Fig. 1 A and B and Fig. S7 A and B), whereas the number of CD11c+ cells in the dermis increased by more than 100% (Fig. S7C). Unexpectedly, the emigration of LCs did not occur in OVA-treated CCL17E/E mice (Fig. 1 A and B and Fig. S7 A and B). In contrast, there was no significant difference in the number of skin-resident dDCs in OVA-treated CCL17E/+ and CCL17E/E mice (Fig. S7C) or in the number of LCs in OVA-treated WT and CCR4KO mice (Fig. 1C).
Fig. 1.

Emigration of LCs After Contact Sensitization Requires CCL17.
The observation that CCL17 affects DC migration in the AD model prompted us to also examine emigration of skin-resident DCs in a model of acute contact sensitization. For this purpose, we applied the contact sensitizer DNFB on the ears for 4 h, and then cultured them for 48 h to permit emigration of LCs from the epidermis. Remarkably, LC emigration was significantly attenuated in CCL17E/E mice (Fig. 2A). Under steady-state conditions, ≈1,000 LCs/mm2 were found in the epidermis of CCL17E/+ and CCL17E/E mice, and treatment with solvent only did not significantly change this number (Fig. 2 A and B). In contrast, DNFB application induced the emigration of more than 50% of all LCs in CCL17E/+ mice (Fig. 2 A and B) and in WT and CCR4KO mice (Fig. 2C), but not in CCL17E/E mice. In the latter, the number of LCs was not significantly reduced compared with the solvent control and only moderately reduced compared with the steady state (Fig. 2 A and B). These findings are in line with the higher frequency of EGFP+ LC in epidermal sheets from DNFB-treated CCL17E/E mice compared with CCL17E/+ mice (Fig. S3C). To further analyze the migration kinetics of skin-derived DCs after allergen exposure, we treated mice with DNFB on the dorsal skin and analyzed the numbers of both LCs and dDCs in the draining LNs after 0, 2 and 4 days. In CCL17E/E mice, the number of LCs was significantly reduced by 4 days after treatment; for dDCs, a significant difference could already be detected on day 2 as well as in the steady state (Fig. 2 D and E). These results are in line with our previous finding of impaired CHS responses in CCL17E/E mice (9).
Fig. 2.

CCL17 Enhances Directional Migration to CCR7 and CXCR4 Ligands.
CCR7 and CXCR4 are known to be key regulators of DC migration from the skin to the skin-draining LNs (5, 6, 24–26). Because migration of mature DCs through peripheral tissues involves direct interactions with the extracellular matrix (3, 27), we first analyzed the migration of bone marrow (BM)-derived DCs in 3D collagen gels using live cell imaging microscopy. In this assay, BM-DCs from CCL17E/E mice showed less random motility and migratory speed (velocity) in the absence of a chemokine gradient compared with CCL17E/+ BM-DCs (Fig. 3 A and B and Movie S1 and Movie S2). When a CCL19 gradient was applied, BM-DCs of CCL17E/+ mice responded with vigorous migration and good directionality (0.52 ± 0.15) (Fig. 3A, Upper Right and Movie S3). In contrast, the directionality of BM-DCs from CCL17E/E mice toward the CCR7 ligand CCL19 was strongly reduced to 0.28 ± 0.18 (Fig. 3A, Lower Right and Movie S4). Likewise, the y-forward migration index, which indicates the directed migration toward the chemokine gradient along the y axis, was reduced by almost 50% (Fig. 3C). Furthermore, the average velocity of CCL17E/E cells was significantly lower than that of CCL17E/+ cells in the presence and absence of a CCL19 gradient (Fig. 3B).
Fig. 3.

We next analyzed the directional migratory capacity of BM-DCs from either CCL17E/+ or CCL17E/E mice to the CCR7 ligands CCL19 and CCL21 and to the CXCR4 ligand CXCL12 in transwell migration assays. Again, we found that BM-DCs generated from CCL17E/E mice had a strongly reduced ability to transmigrate toward all three chemokines compared with DCs from CCL17E/+ mice (Fig. 3D). Nevertheless, the migratory ability of CCL17E/E DCs was not entirely abrogated. Interestingly, CCL17E/E DCs also demonstrated reduced random migration when incubated without any chemokine and moderately reduced chemokinesis (Fig. S8 A and B). We then asked whether migration of CCL17E/E BM-DCs to CCL19 and CXCL12 could be restored by preincubation with recombinant murine (rec) CCL17. As shown in Fig. 3 E–G, recCCL17 had to be present for more than 2 h and at concentrations of >100 ng/mL to achieve full migratory capacity. In addition, emigration of LCs from the epidermis also could be induced in vivo in CCL17E/E mice by injection of recCCL17 into the ear pinna before challenge with DNFB (Fig. S8 E and F). In contrast, treatment with anti-CCL17 antibodies strongly inhibited CCL17E/+ DC migration (Fig. S8D). Thus, we can conclude that CCL17 is essential for the ability of DCs to respond to CCR7 or CXCR4 ligands with efficient directed chemotaxis in an inflammatory micro-milieu. In contrast, the absence of CCR4 had no influence on the migration of BM-DCs toward CCL19 (Fig. S8G). Although CCL22, which is thought to have similar biological activity as CCL17, is produced at normal amounts in CCL17E/E mice (Fig. S8H), it apparently does not compensate for CCL17-deficiency. But the addition of recCCL22 to CCL17-deficient DCs also restores migration to CCL19 (Fig. S8I), and antibody-mediated blockade of CCL22 slightly inhibits DC migration (Fig. S8J). Furthermore, knockdown of CCL22 by siRNA leads to reduced migration of CCL17-expressing, but not CCL17-deficient, BM-DCs to CCL19 and CXCL12 (Fig. S8 K and L). These findings indicate that CCL22 might have a similar function as CCL17, but nonetheless does not compensate for CCL17 in terms of cutaneous DC migration, perhaps because of local cell type–specific differences in production of CCL17 versus CCL22.
CCL17 Regulates CCR7 and CXCR4 Function Without Altering Their Expression Levels.
To test whether the impaired responsiveness of CCL17E/E DCs to CCR7 and CXCR4 ligands was caused by reduced expression of CCR7 or CXCR4, we stained BM-DCs for these receptors. Under normal conditions, CCR7 and CXCR4 are expressed by stimulated BM-DCs, and despite the impaired responsiveness of CCL17E/E DC to CCL19, CCL21, and CXCL12, we found no reduction in CCR7 or CXCR4 expression (Fig. 4A). We also stained for CCR4 and found that equivalent amounts of CCR4 were expressed on unstimulated DCs of CCL17E/E and CCL17E/+ mice (Fig. 4B, Left). After LPS stimulation, the expression levels of CCR4 on DCs from CCL17E/E mice were slightly higher than those on DCs from CCL17E/+ mice (Fig. 4B, Right). We also analyzed CCR7 expression on ex vivo isolated LCs of WT, CCL17E/+, CCL17E/E, and CCR4KO mice. Whereas CCR7 expression was not detectable directly after isolation of the cells (Fig. 4 C and D, Left), it was strongly up-regulated after LPS stimulation, with no difference in LCs from WT, CCL17E/+, CCL17E/E, and CCR4KO mice (Fig. 4 C and D, Right). This indicates that CCL17 deficiency does not affect surface expression of CCR7 and CXCR4, but rather alters their functionality. As a consequence of signal transduction through chemokine receptors, cells normally react with the release of intracellular calcium stores. Thus, we compared the ability of CCR7 to induce calcium flux in CCL17E/+ versus CCL17E/E DCs after stimulation with CCL19 or CXCL12 (Fig. 4 E–N). Remarkably, both CCL19- and CXCL12-induced Ca2+ flux was strongly reduced in CCL17E/E DCs, whereas Ca2+ flux could be induced in all genotypes by treatment with ionomycin (Fig. 4 E–N).
Fig. 4.

Discussion
Chemokines are known to exacerbate inflammatory reactions in allergic skin diseases. In AD, serum levels of several chemokines were found to correlate with disease activity (18). Here we studied the pathogenic role of CCL17 in a murine AD model using CCL17-deficient mice. Although CCL17 and CCL22 are structurally homologous, and both bind to CCR4, the absence of CCL17 alone, and not the absence of CCR4, leads to major amelioration of disease symptoms in the AD model. Our results demonstrate an association of CCL17 with cutaneous DC migration. We show that emigration of DCs, in particular LCs, from the skin to skin-draining LNs is inhibited in models of AD and CHS in CCL17-deficient mice. This finding of a major migratory defect of CCL17-deficient DCs in vivo is further substantiated by direct live cell imaging in vitro, demonstrating that CCL17 is required to sensitize activated BM-DCs for CCR7- and CXCR4-dependent migration and allow Ca2+ flux. Thus, chemokine-dependent emigration of DCs from the skin under inflammatory conditions is strongly enhanced by the inducible chemokine CCL17 through sensitization of at least two different receptors for homeostatic chemokines.
The molecular mechanisms that regulate DC migration from the skin to the draining LNs are only partially understood. Triggering of CCR7 was shown to be required for DC migration under both steady-state and inflammatory conditions (5). In addition, signaling via CXCR4 was found to contribute to the migration of cutaneous DCs after contact sensitization (6, 26). In steady state, the number of LCs within the epidermis is similar in CCL17E/E and CCL17E/+ mice (Fig. 2B). In contrast to CCR7KO mice, which lack skin-derived DCs in peripheral LNs (5), peripheral LNs of CCL17E/E mice have only slightly reduced numbers of skin-derived DCs (Fig. S6 L and M). This slightly reduced skin-derived DC migration might be the result of CCL17 production induced by unintentional environmental stimuli under steady-state conditions. On the other hand, there is an obvious deficiency in the mobilization of LCs from the epidermis after allergic sensitization in CCL17-deficient mice, but not in CCR7KO mice (5). This difference is likely explained by the fact that CXCR4-dependent DC migration is impaired in CCL17E/E mice as well.
In vivo, the CCL17-dependent migratory defect was observed primarily in LCs. We cannot exclude the possibility that migration of dDCs is affected as well, because the number of CD11c+ DCs in the dermis might be kept high in the inflamed skin through a continuous influx of DCs from the circulation. As for the absolute numbers of LC and dDC descendants in the draining LNs, both subsets were similarly reduced in OVA-treated CCL17E/E mice compared with CCL17E/+ controls. The concept that CCR7 is not constitutively able to signal cell migration in the presence of its ligands CCL19/CCL21 has been reported for CD47-deficient DCs (28) and WT DCs that have been differentiated in vitro in the absence of prostaglandin E2 (PGE2) (29, 30). Like CCL17-deficient DCs, these cells express normal levels of CCR7, but fail to undergo efficient directional migration to CCR7 ligands. Thus, the expression of CCR7 on the cell surface alone is not sufficient to guarantee its full functionality (31). The molecular mechanisms by which PGE2 or CCL17 act on CCR7 and potentially other chemokine receptors remain enigmatic, however. In line with previous findings regarding the influence of PGE2 on the responsiveness of human DCs to CCR7 ligands (29), we have shown that the release of intracellular Ca2+ stores in response to CCL19 and CXCL12 is markedly impaired in the absence of CCL17. Therefore, CCL17 may be required to engage important signal transduction components downstream of CCR7 and CXCR4. At present, we also cannot exclude the possibility that CCL17 deficiency leads to a general impairment of DC mobility, because chemokinesis was reduced in CCL17E/E DCs as well (Fig. S8 A and B). A similar phenomenon of sensitization of chemokine receptor function has been described in vitro for CXCR4 and CXCR3, which act synergistically to induce migration of plasmacytoid DCs (pDCs) (32, 33). In the present study, we now provide an in vivo proof of the relevance of cooperation of inducible and homeostatic chemokines during allergic inflammation.
CCL17 has long been implicated in the preferential attraction of Th2 cells (8, 12), which upon recognition of allergens, release IL-4, IL-5, IL-10, and IL-13. We have demonstrated that cytokine expression in the skin is strongly attenuated in the absence of CCL17. In addition, the numbers of mast cells and eosinophils are reduced in OVA-treated CCL17E/E mice, correlating with decreased production of allergen-specific IgE and IgG antibodies. We anticipate that the amelioration of CD4+ T cell infiltration in the skin of CCL17E/E mice is mainly a consequence of reduced T-cell priming in the skin-draining LNs due to deficient DC migration. Although the possibility of a direct skin-homing deficiency of activated T cells in CCL17E/E mice cannot be excluded, this is quite unlikely, because CCR4 and CCR10 are known to play redundant roles in the attraction of memory T cells to the skin (34), in line with the present observation that CCR4KO mice develop allergic skin inflammation similar to WT mice.
Our finding that deficiency of CCL17 alone leads to a blockade of cutaneous DC migration also sheds some light on the relationship and possible redundancy of CCL17 and CCL22, which both bind and activate the receptor CCR4. As shown in this study, recCCL22 can be used like recCCL17 to reconstitute DC migration in vitro, and inhibition of CCL22 leads to reduced DC mobility. CCL22 protein is normally expressed in our CCL17E/E knock-in mice (Fig. S8H), excluding the possibility that the genetic manipulation of ccl17 in these mice also affected the closely linked ccl22 gene. However, under physiological conditions, CCL17 and CCL22 appear to be locally produced by different cell types and in different amounts (35, 36). Furthermore, CCL22 has a greater affinity to the D6 decoy receptor compared with CCL17, and it is susceptible to cleavage by the dipeptidyl-peptidase IV (CD26) (37). These differences may explain why CCL22 does not fully compensate for CCL17 deficiency in vivo. The finding that CCR4KO mice show no defect in cutaneous DC migration and no amelioration of AD pathology suggests that CCL17 and CCL22 must bind to an additional receptor besides CCR4. The only other chemokine receptor that has been postulated to bind CCL17 is CCR8 (38), although this was not seen by another group (39). Therefore, it will be important to identify the as-yet unknown additional interaction partners of CCL17 and CCL22 in DCs.
Consistent with our findings in the mouse model, the number of epidermal LCs is reduced in lesional skin of AD patients compared with normal skin, and LC migration from the epidermis to the dermis correlates with enhanced expression of thymic stromal lymphopoietin (TSLP) in the epidermis (40, 41). In this context, it is interesting to note that TSLP induces maturation and CCL17 expression in DCs. In addition, transgenic expression of CCL17 in keratinocytes was shown to enhance Th2-dependent CHS and AD-like symptoms (42).
In conclusion, we have demonstrated that the chemokine CCL17 plays a unique and essential role in cutaneous DC migration and the pathogenesis of allergic skin inflammation. Regarding the design of novel therapeutic drugs for treatment of AD, it has been suggested that blockade of CCR4 represents a promising target (43). Based on our present findings that CCL17-deficient mice, but not CCR4KO mice, display a defect in cutaneous DC migration and are protected from AD, we propose that local inhibition of CCL17 might be more suitable for prevention of allergic skin inflammation than CCR4 blockade.
Materials and Methods
AD Mouse Model.
Atopic skin inflammation was induced as described previously (21, 22). In brief, mice were tape-stripped four times using Tegaderm 1624W (3M Medica). First, 100 μg of OVA (grade V; Sigma-Aldrich) in NaCl or NaCl alone was placed on a patch of sterile gauze and attached to the skin. The patches were applied on days 1 and 4 and maintained for a total of 1 week. After 2 weeks of rest, mice were tape-stripped again, and an identical patch was reapplied to the same site. Mice received a total of three 1-week patch exposures.
Contact Sensitization.
Mice were treated on both sides of the ear with 10 μL of 0.5% DNFB in acetone/olive oil (5:1). After 4 h, the ears were removed and cultured for 2 days at 37 °C. Epidermal sheets were stained with mouse anti-Langerin (clone 929F3.01; Acris Antibodies). To investigate the in vivo migration of DCs, the dorsal skin was shaved, and 80 μL of 0.5% DNFB was applied. After 2 and 4 days, skin-draining LNs were analyzed by flow cytometry.
In Vitro Migration Assay.
BM-DCs were generated as described previously (9). On day 6, nonadherent cells were harvested and activated with LPS overnight. DC migration was measured in 24-well Costar transwell chambers (Corning). After a 3-h incubation at 37 °C, the migrated cells were counted and stained for FACS analysis.
Three-Dimensional Collagen Gel Chemotaxis Assay.
The 3D chemotaxis assays were performed as described previously (27) with some modifications. Collagen-BM-DC mixtures were placed in custom-made chemotaxis chambers and incubated at 37 °C in 5% CO2 for 90 min. After collagen polymerization, 600 ng/mL of CCL19 (R&D Systems) was added on top of the gel. Time-lapse series were recorded using a fully automated inverted TE Eclipse microscope (Nikon). BM-DC were tracked over 3 h (5 min/frame) using the ImageJ manual tracking plug-in.
Other Methods.
Detailed descriptions of all methods used are provided in SI Materials and Methods.
Acknowledgments
We thank Ingo Uthe and Markus Korkowski for technical help; Cora Schild for technical advice; Heike Weighardt, Charlotte Esser, Jean Krutmann, and Natalija Novak for discussion and comments on the manuscript; and Klaus Pfeffer for generous support. This work was supported by the Deutsche Forschungsgemeinschaft (Grant SFB 704 to I.F. and W.K. and Grant FOR729 to I.F. and B.H.).
Supporting Information
Supporting Information (PDF)
Supporting Information
- Download
- 2.11 MB
sm01.mov
- Download
- 4.16 MB
sm02.mov
- Download
- 4.88 MB
sm03.mov
- Download
- 4.67 MB
sm04.mov
- Download
- 4.15 MB
References
1
SJ Allen, SE Crown, TM Handel, Chemokine: Receptor structure, interactions, and antagonism. Annu Rev Immunol 25, 787–820 (2007).
2
JG Cyster, Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol 23, 127–159 (2005).
3
S Koch, K Kohl, E Klein, D von Bubnoff, T Bieber, Skin homing of Langerhans cell precursors: Adhesion, chemotaxis, and migration. J Allergy Clin Immunol 117, 163–168 (2006).
4
GJ Randolph, J Ochando, S Partida-Sánchez, Migration of dendritic cell subsets and their precursors. Annu Rev Immunol 26, 293–316 (2008).
5
L Ohl, et al., CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21, 279–288 (2004).
6
K Kabashima, et al., CXCL12-CXCR4 engagement is required for migration of cutaneous dendritic cells. Am J Pathol 171, 1249–1257 (2007).
7
T Imai, et al., Molecular cloning of a novel T cell–directed CC chemokine expressed in thymus by signal sequence trap using Epstein-Barr virus vector. J Biol Chem 271, 21514–21521 (1996).
8
I Lieberam, I Förster, The murine beta-chemokine TARC is expressed by subsets of dendritic cells and attracts primed CD4+ T cells. Eur J Immunol 29, 2684–2694 (1999).
9
J Alferink, et al., Compartmentalized production of CCL17 in vivo: Strong inducibility in peripheral dendritic cells contrasts selective absence from the spleen. J Exp Med 197, 585–599 (2003).
10
A Mantovani, PA Gray, J Van Damme, S Sozzani, Macrophage-derived chemokine (MDC). J Leukoc Biol 68, 400–404 (2000).
11
F Katou, et al., Macrophage-derived chemokine (MDC/CCL22) and CCR4 are involved in the formation of T lymphocyte–dendritic cell clusters in human inflamed skin and secondary lymphoid tissue. Am J Pathol 158, 1263–1270 (2001).
12
T Imai, et al., Selective recruitment of CCR4-bearing Th2 cells toward antigen-presenting cells by the CC chemokines thymus and activation-regulated chemokine and macrophage-derived chemokine. Int Immunol 11, 81–88 (1999).
13
A Iellem, et al., Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4(+)CD25(+) regulatory T cells. J Exp Med 194, 847–853 (2001).
14
Y Chvatchko, et al., A key role for CC chemokine receptor 4 in lipopolysaccharide-induced endotoxic shock. J Exp Med 191, 1755–1764 (2000).
15
N Novak, T Bieber, DY Leung, Immune mechanisms leading to atopic dermatitis. J Allergy Clin Immunol 112, S128–S139 (2003).
16
PM Elias, Y Hatano, ML Williams, Basis for the barrier abnormality in atopic dermatitis: Outside-inside-outside pathogenic mechanisms. J Allergy Clin Immunol 121, 1337–1343 (2008).
17
DY Leung, M Boguniewicz, MD Howell, I Nomura, QA Hamid, New insights into atopic dermatitis. J Clin Invest 113, 651–657 (2004).
18
B Homey, M Steinhoff, T Ruzicka, DY Leung, Cytokines and chemokines orchestrate atopic skin inflammation. J Allergy Clin Immunol 118, 178–189 (2006).
19
Y Shimada, K Takehara, S Sato, Both Th2 and Th1 chemokines (TARC/CCL17, MDC/CCL22, and Mig/CXCL9) are elevated in sera from patients with atopic dermatitis. J Dermatol Sci 34, 201–208 (2004).
20
H Saeki, K Tamaki, Thymus and activation regulated chemokine (TARC)/CCL17 and skin diseases. J Dermatol Sci 43, 75–84 (2006).
21
JM Spergel, et al., Epicutaneous sensitization with protein antigen induces localized allergic dermatitis and hyperresponsiveness to methacholine after single exposure to aerosolized antigen in mice. J Clin Invest 101, 1614–1622 (1998).
22
G Wang, et al., Repeated epicutaneous exposures to ovalbumin progressively induce atopic dermatitis–like skin lesions in mice. Clin Exp Allergy 37, 151–161 (2007).
23
S Stutte, B Jux, C Esser, I Förster, CD24a expression levels discriminate Langerhans cells from dermal dendritic cells in murine skin and lymph nodes. J Invest Dermatol 128, 1470–1475 (2008).
24
L Riol-Blanco, et al., The chemokine receptor CCR7 activates in dendritic cells two signaling modules that independently regulate chemotaxis and migratory speed. J Immunol 174, 4070–4080 (2005).
25
SA Kellermann, S Hudak, ER Oldham, YJ Liu, LM McEvoy, The CC chemokine receptor-7 ligands 6Ckine and macrophage inflammatory protein-3 beta are potent chemoattractants for in vitro– and in vivo–derived dendritic cells. J Immunol 162, 3859–3864 (1999).
26
K Ouwehand, et al., CXCL12 is essential for migration of activated Langerhans cells from epidermis to dermis. Eur J Immunol 38, 3050–3059 (2008).
27
P Friedl, EB Bröcker, Reconstructing leukocyte migration in 3D extracellular matrix by time-lapse videomicroscopy and computer-assisted tracking. Methods Mol Biol 239, 77–90 (2004).
28
S Hagnerud, et al., Deficit of CD47 results in a defect of marginal zone dendritic cells, blunted immune response to particulate antigen and impairment of skin dendritic cell migration. J Immunol 176, 5772–5778 (2006).
29
E Scandella, et al., CCL19/CCL21-triggered signal transduction and migration of dendritic cells requires prostaglandin E2. Blood 103, 1595–1601 (2004).
30
JH Yen, T Khayrullina, D Ganea, PGE2-induced metalloproteinase-9 is essential for dendritic cell migration. Blood 111, 260–270 (2008).
31
N Sánchez-Sánchez, L Riol-Blanco, JL Rodríguez-Fernández, The multiple personalities of the chemokine receptor CCR7 in dendritic cells. J Immunol 176, 5153–5159 (2006).
32
A Krug, et al., IFN-producing cells respond to CXCR3 ligands in the presence of CXCL12 and secrete inflammatory chemokines upon activation. J Immunol 169, 6079–6083 (2002).
33
B Vanbervliet, et al., The inducible CXCR3 ligands control plasmacytoid dendritic cell responsiveness to the constitutive chemokine stromal cell–derived factor 1 (SDF-1)/CXCL12. J Exp Med 198, 823–830 (2003).
34
Y Reiss, AE Proudfoot, CA Power, JJ Campbell, EC Butcher, CC chemokine receptor (CCR)4 and the CCR10 ligand cutaneous T cell–attracting chemokine (CTACK) in lymphocyte trafficking to inflamed skin. J Exp Med 194, 1541–1547 (2001).
35
C Vestergaard, et al., Overproduction of Th2-specific chemokines in NC/Nga mice exhibiting atopic dermatitis–like lesions. J Clin Invest 104, 1097–1105 (1999).
36
TJ Curiel, et al., Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10, 942–949 (2004).
37
R Bonecchi, et al., Differential recognition and scavenging of native and truncated macrophage-derived chemokine (macrophage-derived chemokine/CC chemokine ligand 22) by the D6 decoy receptor. J Immunol 172, 4972–4976 (2004).
38
G Bernardini, et al., Identification of the CC chemokines TARC and macrophage inflammatory protein-1 beta as novel functional ligands for the CCR8 receptor. Eur J Immunol 28, 582–588 (1998).
39
JM Fox, et al., Structure/function relationships of CCR8 agonists and antagonists: Amino-terminal extension of CCL1 by a single amino acid generates a partial agonist. J Biol Chem 281, 36652–36661 (2006).
40
E Guttman-Yassky, et al., Major differences in inflammatory dendritic cells and their products distinguish atopic dermatitis from psoriasis. J Allergy Clin Immunol 119, 1210–1217 (2007).
41
YJ Liu, et al., TSLP: An epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol 25, 193–219 (2007).
42
Y Tsunemi, et al., CCL17 transgenic mice show an enhanced Th2-type response to both allergic and non-allergic stimuli. Eur J Immunol 36, 2116–2127 (2006).
43
AD Luster, Antichemokine immunotherapy for allergic diseases. Curr Opin Allergy Clin Immunol 1, 561–567 (2001).
Information & Authors
Information
Published in
Classifications
Submission history
Published online: April 26, 2010
Published in issue: May 11, 2010
Keywords
Acknowledgments
We thank Ingo Uthe and Markus Korkowski for technical help; Cora Schild for technical advice; Heike Weighardt, Charlotte Esser, Jean Krutmann, and Natalija Novak for discussion and comments on the manuscript; and Klaus Pfeffer for generous support. This work was supported by the Deutsche Forschungsgemeinschaft (Grant SFB 704 to I.F. and W.K. and Grant FOR729 to I.F. and B.H.).
Notes
This article is a PNAS Direct Submission.
Authors
Competing Interests
The authors declare no conflict of interest.
Metrics & Citations
Metrics
Altmetrics
Citations
Cite this article
Requirement of CCL17 for CCR7- and CXCR4-dependent migration of cutaneous dendritic cells, Proc. Natl. Acad. Sci. U.S.A.
107 (19) 8736-8741,
https://doi.org/10.1073/pnas.0906126107
(2010).
Copied!
Copying failed.
Export the article citation data by selecting a format from the list below and clicking Export.
Cited by
Loading...
View Options
View options
PDF format
Download this article as a PDF file
DOWNLOAD PDFLogin options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginRecommend to a librarian
Recommend PNAS to a LibrarianPurchase options
Purchase this article to access the full text.