Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes

Subjects

Abstract

Afferent lymph–borne dendritic cells essentially rely on the chemokine receptor CCR7 for their transition from the subcapsular lymph node sinus into the parenchyma, a migratory step driven by putative gradients of CCR7 ligands. We found that lymph node fringes indeed contained physiological gradients of the chemokine CCL21, which depended on the expression of CCRL1, the atypical receptor for the CCR7 ligands CCL19 and CCL21. Lymphatic endothelial cells lining the ceiling of the subcapsular sinus, but not those lining the floor, expressed CCRL1, which scavenged chemokines from the sinus lumen. This created chemokine gradients across the sinus floor and enabled the emigration of dendritic cells. In vitro live imaging revealed that spatially confined expression of CCRL1 was necessary and sufficient for the creation of functional chemokine gradients.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: CCRL1 is expressed in the SCS by cLECs but not fLECs.
Figure 2: CCRL1-dependent patterning of CCL21 gradients in the SCS.
Figure 3: CCRL1 shapes functional gradients of CCR7 ligands in vitro.
Figure 4: CCR7 mediates the directional migration of BMDCs along CCL19 gradients shaped by CCRL1.
Figure 5: CCRL1 is required for the CCR7-dependent migration of endogenous skin-derived DCs from the SCS into the LN parenchyma.
Figure 6: CCRL1 is required for the migration of BMDCs from the SCS into the LN parenchyma after intralymphatic injection.
Figure 7: CCL21 immunoreactivity and distribution of LDCs in LNs of plt and plt–CCRL1-deficient mice.

Similar content being viewed by others

References

  1. von Andrian, U.H. & Mempel, T.R. Homing and cellular traffic in lymph nodes. Nat. Rev. Immunol. 3, 867–878 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Shakhar, G. et al. Stable T cell–dendritic cell interactions precede the development of both tolerance and immunity in vivo. Nat. Immunol. 6, 707–714 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lindquist, R.L. et al. Visualizing dendritic cell networks in vivo. Nat. Immunol. 5, 1243–1250 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999).

    Article  PubMed  Google Scholar 

  5. Schneider, M.A., Meingassner, J.G., Lipp, M., Moore, H.D. & Rot, A. CCR7 is required for the in vivo function of CD4+CD25+ regulatory T cells. J. Exp. Med. 204, 735–745 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ohl, L. et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21, 279–288 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Vander Lugt, B. et al. CCR7 plays no appreciable role in trafficking of central memory CD4 T cells to lymph nodes. J. Immunol. 191, 3119–3127 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Förster, R., Davalos-Misslitz, A.C. & Rot, A. CCR7 and its ligands: balancing immunity and tolerance. Nat. Rev. Immunol. 8, 362–371 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Weber, M. et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science 339, 328–332 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Braun, A. et al. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat. Immunol. 12, 879–887 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Gosling, J. et al. Cutting edge: Identification of a novel chemokine receptor that binds dendritic cell- and T cell-active chemokines including ELC, SLC, and TECK. J. Immunol. 164, 2851–2856 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Townson, J.R. & Nibbs, R.J. Characterization of mouse CCX–CKR, a receptor for the lymphocyte-attracting chemokines TECK/mCCL25, SLC/mCCL21 and MIP-3β/mCCL19: comparison to human CCX–CKR. Eur. J. Immunol. 32, 1230–1241 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Bachelerie, F. et al. International Union of Pharmacology. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol. Rev. 66, 1–79 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bachelerie, F. et al. New nomenclature for atypical chemokine receptors. Nat. Immunol. 15, 207–208 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Heinzel, K., Benz, C. & Bleul, C.C. A silent chemokine receptor regulates steady-state leukocyte homing in vivo. Proc. Natl. Acad. Sci. USA 104, 8421–8426 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mäkinen, T. et al. PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes Dev. 19, 397–410 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Comerford, I., Milasta, S., Morrow, V., Milligan, G. & Nibbs, R. The chemokine receptor CCX-CKR mediates effective scavenging of CCL19 in vitro. Eur. J. Immunol. 36, 1904–1916 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Comerford, I. et al. The atypical chemokine receptor CCX-CKR scavenges homeostatic chemokines in circulation and tissues and suppresses Th17 responses. Blood 116, 4130–4140 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Wendland, M. et al. Lymph node T cell homeostasis relies on steady state homing of dendritic cells. Immunity 35, 945–957 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Moussion, C. & Girard, J.P. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 479, 542–546 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Stein, J.V. et al. The CC chemokine thymus-derived chemotactic agent 4 (TCA-4, secondary lymphoid tissue chemokine, 6Ckine, exodus-2) triggers lymphocyte function-associated antigen 1-mediated arrest of rolling T lymphocytes in peripheral lymph node high endothelial venules. J. Exp. Med. 191, 61–76 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gunn, M.D. et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J. Exp. Med. 189, 451–460 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Vassileva, G. et al. The reduced expression of 6Ckine in the plt mouse results from the deletion of one of two 6Ckine genes. J. Exp. Med. 190, 1183–1188 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rot, A. & von Andrian, U.H. Chemokines in innate and adaptive host defense: Basic chemokinese grammar for immune cells. Annu. Rev. Immunol. 22, 891–928 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Sánchez-Madrid, F. & del Pozo, M.A. Leukocyte polarization in cell migration and immune interactions. EMBO J. 18, 501–511 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Okada, T. et al. Antigen-engaged B cells undergo chemotaxis toward the T zone and form motile conjugates with helper T cells. PLoS Biol. 10.1371/journal.pbio.0030150 (3 May 2005).

  27. Castellino, F. et al. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4 T cell-dendritic cell interaction. Nature 440, 890–895 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Middleton, J. et al. Transcytosis and surface presentation of IL-8 by venular endothelial cells. Cell 91, 385–395 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. Pruenster, M. et al. The Duffy antigen receptor for chemokines transports chemokines and supports their promigratory activity. Nat. Immunol. 10, 101–108 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Haessler, U., Pisano, M., Wu, M.M. & Swartz, M.A. Dendritic cell chemotaxis in 3D under defined chemokine gradients reveals differential response to ligands CCL21 and CCL19. Proc. Natl. Acad. Sci. USA 108, 5614–5619 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Schumann, K. et al. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity 32, 703–713 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Chai, Q. et al. Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity. Immunity 38, 1013–1024 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Crick, F. Diffusion in embryogenesis. Nature 225, 420–422 (1970).

    Article  CAS  PubMed  Google Scholar 

  34. Naumann, U. et al. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS ONE 5, e9175 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Boldajipour, B. et al. Control of chemokine-guided cell migration by ligand sequestration. Cell 132, 463–473 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Venkiteswaran, G. et al. Generation and dynamics of an endogenous, self-generated signaling gradient across a migrating tissue. Cell 155, 674–687 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Donà, E. et al. Directional tissue migration through a self-generated chemokine gradient. Nature 503, 285–289 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Ulvmar, M.H., Hub, E. & Rot, A. Atypical chemokine receptors. Exp. Cell Res. 317, 556–568 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nibbs, R.J.B. & Graham, G.J. Immune regulation by atypical chemokine receptors. Nat. Rev. Immunol. 13, 815–829 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Graham, G.J., Locati, M., Mantovani, A., Rot, A. & Thelen, M. The biochemistry and biology of the atypical chemokine receptors. Immunol. Lett. 145, 30–38 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Colditz, I.G., Schneider, M.A., Pruenster, M. & Rot, A. Chemokines at large: In-vivo mechanisms of their transport, presentation and clearance. Thromb. Haemost. 97, 688–693 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Lee, K.M. et al. D6 facilitates cellular migration and fluid flow to lymph nodes by suppressing lymphatic congestion. Blood 118, 6220–6229 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Fra, A.M. et al. Cutting edge: scavenging of inflammatory CC chemokines by the promiscuous putatively silent chemokine receptor D6. J. Immunol. 170, 2279–2282 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Nakano, H. et al. Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses. Nat. Immunol. 10, 394–402 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tanaka, Y., Mamalaki, C., Stockinger, B. & Kioussis, D. In-vitro negative selection of αβ-T-cell receptor transgenic thymocytes by conditionally immortalized thymic cortical epithelial-cell lines and dendritic cells. Eur. J. Immunol. 23, 2614–2621 (1993).

    Article  CAS  PubMed  Google Scholar 

  46. Friedl, P. & Brocker, E.B. Reconstructing leukocyte migration in 3D extracellular matrix by time-lapse videomicroscopy and computer-assisted tracking. Methods Mol. Biol. 239, 77–90 (2004).

    PubMed  Google Scholar 

  47. Overwijk, W.W. et al. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Caamano for reading the manuscript and for suggestions; C. Bleul (Novartis) for CCRL1-eGFP mice; M. Gunn (Duke University) for plt mice; and D. Kioussis (Medical Research Council, National Institute for Medical Research) for the TEP cell line. Supported by the Medical Research Council (G0802838 to A.R. and G9818340 to the Medical Research Council Centre), The European Union Marie Curie Actions (CRITICS to M.H.U. and A.R.), the Wellcome Trust (WT090962MA to I.N.-B. and A.R.), the European Research Council (322645 to R.F.), Deutsche Forschungsgemeinschaft (SFB738-B5 and EXC62, 'Rebirth', to R.F.) and Boehringer Ingelheim Fonds (A.B.).

Author information

Authors and Affiliations

Authors

Contributions

M.H.U., K.W., A.B., E.H., K.E., T.W., R.F. and A.R. designed the experiments and evaluated the data; M.H.U., K.W., A.B., P.K., E.H., L.C. and I.N.-B. did the experiments; T.W. wrote evaluation software; M.H.U., K.W., A.B., P.K., E.H., R.F. and A.R. prepared the figures; M.H.U., K.W., A.B. and E.H. contributed to the preparation of the manuscript; B.L. and K.N. produced cell lines; R.J.B.N. provided a mouse strain; T.R. rederived mouse strains; and R.F. and A.R. conceived of the project, directed the research and wrote the manuscript.

Corresponding authors

Correspondence to Reinhold Förster or Antal Rot.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Specificity controls for staining with antibody to CCRL1.

Acetone-fixed frozen sections of WT LN stained (a) with anti-CCRL1 antibody (red) and (b) with control isotype IgG. Acetone-fixed cryo sections of WT LN (c) and CCRL1 ko LN (d) stained with anti-CCRL1 (red), anti-LYVE-1 (blue) and anti-gp38 (green) antibodies. Scale bars 50 μm. Representative of more than 3 studies.

Supplementary Figure 2 CCRL1 expression in the LNs is confined to the gp38hiLyve-1lo LEC subset.

Flow cytometry analysis of LECs in lymph nodes from CCRL1-eGFP mice. (a-b) Stromal cells in the live cell gate (a), gated from the CD45neg, Ter119neg cells (b). (c) LECs are defined as gp38high CD31high cells, BECs as gp38low CD31high, FRC as gp38high CD31neg, and DN stromal cells are defined as gp38neg CD31neg. (d) gp38high CD31high LECs analysed for expression of LYVE-1 and eGFP. (e-f) Analysis of the eGFPneg and eGFPpos gated LEC subsets for expression of gp38 (e) and Lyve-1 (f). Geometric mean (MFI) and LYVE-1 positive cells in gate L1 are shown. eGFP positive cells in the gp38high CD31low, FRCs (g), the gp38low CD31high, BECs, (h) the gp38low CD31low DN stromal cells (i) and CD45pos gated cells (j). Representative graphs from inguinal LN from 3 CCRL1-eGFP mice.

Supplementary Figure 3 TEP-CCRL1 cells bind and scavenge CCR7 ligands.

(a) CCRL1 expressed in TEP cells scavenges CCL19. TEP-CCRL1 or TEP-mock cells were incubated at 37°C with 10nM CCL19-Alexa647 for the time indicated. Cells were washed with cold PBS 3% FBS and analyzed by flow cytometry revealing accumulation of CCL19 over time in TEP-CCRL1 cells but not TEP-mock cells (b,c). The expression of CCRL1 in monolayers inhibits CCL19- (b) and CCL21-induced (c) in vitro transmigration of BM-DCs. Monolayers of TEP-CCRL1 and empty vector-transfected control TEP-mock cells were grown to confluence on the lower side of Transwell insert filters with 5μm pores. BM-DCs were added to triplicate inserts and allowed to migrate in response to 0.8nM murine recombinant CCL19 or CCL21 or RPMI in the lower well. After incubation at 37o C for 3 h, BM-DCs were collected from the bottom well, stained by an anti-CD11c Ab and analyzed by FACS using counting beads. Data shown are mean and STDEV for triplicate wells from one representative experiment each from three and four performed using CCL19 and CCL21, respectively. Significance indicated: *: P < 0.05. In all experiments migration indices (ratios of BM-DCs migrated to chemokine and to RPMI) were 1.6±0.2 and 3.5±0.3 for CCL19 and 1.7±0.9 and 6.6±4.2 for CCL21, across TEP-CCRL1 and TEP-mock monolayers, respectively (median±STDEV; for both chemokines significant difference between TEP-CCRL1 and TEP-mock, p<0.05, Mann-Whitney, U-test).

Supplementary Figure 4 Lower proportions of CCR7+CD86+ DCs in skin-draining LNs of CCRL1-deficient mice.

(a) Gating strategy to identify “migratory” CD86pos CCR7pos DC in inguinal LNs of wild type (WT)and CCRL1 deficient (KO) mice. After single cell gate and exclusion of dead cells, total CD45+ cells were analysed for CD11c and CD11b expression as displayed for representative wild type and CCRL1 deficient LNs. Populations of CD11bhigh (blue gate) and CD11blow (green gate) DCs were further analysed for CCR7 and CD86 expression to identify CCR7+CD86+ “migratory” DCs. To identify LDCs total DCs (red gate) were analysed for langerin against CCR7. This gate encompasses both epidermal-derived Langerhans cells and langerinpos dermal DCs. (b) Proportions of both CD11bhighCD11c+CD86highCCR7+ and CD11blowCD11c+CD86highCCR7+ DCs are significantly lower in CCRL1 deficient mice (KO, white box) compared to wild-type (WT grey box), Significance indicated: *, P < 0.00. Data are from four litters of 8 week old F2 female mice. n=9 per group. (c) Representative histogram of CCR7 expression in WT (blue) versus CCRL1 ko (red) gated on all DCs, representative of 3 independent experiments. (e) Geometric mean fluorescent intensity of CCR7 in gated CD86high cells from WT (grey box) and KO (white box). F2 littermates, n=4 and 5 for CCRL1 ko and WT mice, respectively. Box-plot graphs show median values with quartiles and minimum/maximum.

Supplementary Figure 5 Cell populations in LNs of wild-type and CCRL1-deficient mice.

(a) LN cellularity in WT (grey box) and CCRL1 ko mice (white box) determined by FACS using counting beads after LN digestion. (b-e) Absolute cell numbers (left) and relative proportions (right) of main immune cell populations. (b) T cells (CD3+), (c) B cells (B220+CD19+) (d) Myeloid cells (CD11c-CD11b+) and (e) DC (CD11c+). Data are from F2 generation 8 week old females; n=6-7 in a-c and n=5 in d-e across three and two litters, respectively. Significance indicated: *, P < 0.05, **, P < 0.01. Box-plot graphs show median values with quartiles and minimum/maximum.

Supplementary Figure 6 DC-borne antigen–induced T cell proliferation in wild-type and CCRL1-deficient LNs

(a-d) Representative proliferation curves of CellTrace violet labelled pmel (Thy1.1+ Vbeta13 TCR+) T-cells after transfer of 5x104 cognate antigen loaded, LPS matured WT BM-DCs into the footpad of WT or CCRL1 deficient mice. Proliferating T cells in draining LNs of WT (a) and CCRL1 deficient (b), and non-draining LNs of WT (c) and CCRL1 deficient (d) mice. Data expressed as % of pmel cells showing dilution of the CellTrace dye. (e-f) Cumulative data from draining (e) and non-draining (f) LNs of WT and CCRL1 deficient mice; n=18 and 19 (e) and 9 and 8 (f), respectively. (g) Homing of pmel T-cells into resting WT and CCRL1 ko LNs after i.v. transfer. Cumulative data from 10 WT and 8 CCRL1 deficient mice. Significance indicated,*, P < 0.001. Box-plot graphs show median values with quartiles and minimum/maximum.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ulvmar, M., Werth, K., Braun, A. et al. The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes. Nat Immunol 15, 623–630 (2014). https://doi.org/10.1038/ni.2889

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2889

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing