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:

Selective programming of CCR10+ innate lymphoid cells in skin-draining lymph nodes for cutaneous homeostatic regulation

Abstract

Innate lymphoid cells (ILCs) 'preferentially' localize into barrier tissues, where they function in tissue protection but can also contribute to inflammatory diseases. The mechanisms that regulate the establishment of ILCs in barrier tissues are poorly understood. Here we found that under steady-state conditions, ILCs in skin-draining lymph nodes (sLNs) were continuously activated to acquire regulatory properties and high expression of the chemokine receptor CCR10 for localization into the skin. CCR10+ ILCs promoted the homeostasis of skin-resident T cells and, reciprocally, their establishment in the skin required T cell–regulated homeostatic environments. CD207+ dendritic cells expressing the transcription factor Foxn1 were required for the proper generation of CCR10+ ILCs. These observations reveal mechanisms that underlie the specific programming and priming of skin-homing CCR10+ ILCs in the sLNs.

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: Selective programming of skin-specific CCR10+ ILCs in sLNs.
Figure 2: The homeostatic establishment of CCR10+ ILCs in the skin depends on the continuous input of CCR10+MHCII+ ILCs from sLNs.
Figure 3: CCR10 and CCR6 co-regulate the specific localization and maintenance of ILCs in the skin.
Figure 4: Immunological homeostatic conditions and T cells regulate the programming and maintenance of CCR10+ ILCs in sLNs and the skin.
Figure 5: CCR10+ ILCs regulate the homeostasis of helper T cells in the skin.
Figure 6: Foxn1 and CD207+ DCs are involved in the programming of CCR10+ ILCs in sLNs.

Similar content being viewed by others

References

  1. Spits, H. et al. Innate lymphoid cells–a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Walker, J.A., Barlow, J.L. & McKenzie, A.N. Innate lymphoid cells–how did we miss them? Nat. Rev. Immunol. 13, 75–87 (2013).

    CAS  PubMed  Google Scholar 

  3. Monticelli, L.A. et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 12, 1045–1054 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Roediger, B. et al. Cutaneous immunosurveillance and regulation of inflammation by group 2 innate lymphoid cells. Nat. Immunol. 14, 564–573 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hepworth, M.R. et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 498, 113–117 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Nussbaum, J.C. et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Mortha, A. et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 343, 1249288 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Powell, N. et al. The transcription factor T-bet regulates intestinal inflammation mediated by interleukin-7 receptor+ innate lymphoid cells. Immunity 37, 674–684 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Geremia, A. et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J. Exp. Med. 208, 1127–1133 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Coccia, M. et al. IL-1β mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4+ Th17 cells. J. Exp. Med. 209, 1595–1609 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wilhelm, C. et al. An IL-9 fate reporter demonstrates the induction of an innate IL-9 response in lung inflammation. Nat. Immunol. 12, 1071–1077 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Chang, Y.J. et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat. Immunol. 12, 631–638 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Kim, H.Y. et al. Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity. J. Allergy Clin. Immunol. 129, 216–227 (2012).

    CAS  PubMed  Google Scholar 

  14. Kim, H.Y. et al. Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat. Med. 20, 54–61 (2014).

    CAS  PubMed  Google Scholar 

  15. Kim, B.S. et al. TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation. Sci. Transl. Med. 5, 170ra116 (2013).

    Google Scholar 

  16. Salimi, M. et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 210, 2939–2950 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Imai, Y. et al. Skin-specific expression of IL-33 activates group 2 innate lymphoid cells and elicits atopic dermatitis-like inflammation in mice. Proc. Natl. Acad. Sci. USA 110, 13921–13926 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Pantelyushin, S. et al. Rorγt+ innate lymphocytes and γδ T cells initiate psoriasiform plaque formation in mice. J. Clin. Invest. 122, 2252–2256 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Mjösberg, J.M. et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat. Immunol. 12, 1055–1062 (2011).

    PubMed  Google Scholar 

  20. Constantinides, M.G., McDonald, B.D., Verhoef, P.A. & Bendelac, A. A committed precursor to innate lymphoid cells. Nature 508, 397–401 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Hoyler, T. et al. The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity 37, 634–648 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Homey, B. et al. Cutting edge: the orphan chemokine receptor G protein-coupled receptor-2 (GPR-2, CCR10) binds the skin-associated chemokine CCL27 (CTACK/ALP/ILC). J. Immunol. 164, 3465–3470 (2000).

    CAS  PubMed  Google Scholar 

  23. Xia, M. et al. CCR10 regulates balanced maintenance and function of resident regulatory and effector T cells to promote immune homeostasis in the skin. J. Allergy Clin. Immunol. 134, 634–644 e610 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Jin, Y., Xia, M., Sun, A., Saylor, C.M. & Xiong, N. CCR10 is important for the development of skin-specific γδT cells by regulating their migration and location. J. Immunol. 185, 5723–5731 (2010).

    CAS  PubMed  Google Scholar 

  25. Spencer, S.P. et al. Adaptation of innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science 343, 432–437 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cyster, J.G. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu. Rev. Immunol. 23, 127–159 (2005).

    CAS  PubMed  Google Scholar 

  27. Eberl, G. et al. An essential function for the nuclear receptor RORγ(t) in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 5, 64–73 (2004).

    CAS  PubMed  Google Scholar 

  28. Sigmundsdottir, H. et al. DCs metabolize sunlight-induced vitamin D3 to 'program' T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 8, 285–293 (2007).

    CAS  PubMed  Google Scholar 

  29. Kiss, E.A. et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334, 1561–1565 (2011).

    CAS  PubMed  Google Scholar 

  30. Rennert, P.D., Browning, J.L., Mebius, R., Mackay, F. & Hochman, P.S. Surface lymphotoxin α/β complex is required for the development of peripheral lymphoid organs. J. Exp. Med. 184, 1999–2006 (1996).

    CAS  PubMed  Google Scholar 

  31. Moro, K. et al. Innate production of TH2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells. Nature 463, 540–544 (2010).

    CAS  PubMed  Google Scholar 

  32. Neill, D.R. et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464, 1367–1370 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Nehls, M. et al. Two genetically separable steps in the differentiation of thymic epithelium. Science 272, 886–889 (1996).

    CAS  PubMed  Google Scholar 

  34. Flanagan, S.P. 'Nude', a new hairless gene with pleiotropic effects in the mouse. Genet. Res. 8, 295–309 (1966).

    CAS  PubMed  Google Scholar 

  35. Kissenpfennig, A. et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 22, 643–654 (2005).

    CAS  PubMed  Google Scholar 

  36. Bobr, A. et al. Acute ablation of Langerhans cells enhances skin immune responses. J. Immunol. 185, 4724–4728 (2010).

    CAS  PubMed  Google Scholar 

  37. McCully, M.L. et al. Epidermis instructs skin homing receptor expression in human T cells. Blood 120, 4591–4598 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Sigmundsdottir, H. & Butcher, E.C. Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat. Immunol. 9, 981–987 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Moed, H. et al. Increased CCL27–CCR10 expression in allergic contact dermatitis: implications for local skin memory. J. Pathol. 204, 39–46 (2004).

    CAS  PubMed  Google Scholar 

  40. Kakinuma, T. et al. Increased serum cutaneous T cell-attracting chemokine (CCL27) levels in patients with atopic dermatitis and psoriasis vulgaris. J. Allergy Clin. Immunol. 111, 592–597 (2003).

    CAS  PubMed  Google Scholar 

  41. Gudjonsson, J.E. et al. Assessment of the psoriatic transcriptome in a large sample: additional regulated genes and comparisons with in vitro models. J. Invest. Dermatol. 130, 1829–1840 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Riis, J.L. et al. Kinetics and differential expression of the skin-related chemokines CCL27 and CCL17 in psoriasis, atopic dermatitis and allergic contact dermatitis. Exp. Dermatol. 20, 789–794 (2011).

    CAS  PubMed  Google Scholar 

  43. Hu, S., Yang, K., Yang, J., Li, M. & Xiong, N. Critical roles of chemokine receptor CCR10 in regulating memory IgA responses in intestines. Proc. Natl. Acad. Sci. USA 108, E1035–E1044 (2011).

    PubMed  PubMed Central  Google Scholar 

  44. Li, M. et al. Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proc. Natl. Acad. Sci. USA 103, 11736–11741 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Bradfield (University of Wisconsin) for Ahr−/− mice; and X. Zang (Albert Einstein College of Medicine) for antibodies to mouse PDL-2, mouse B7x, mouse B7h and mouse B7H3. Supported by the National Institute of Allergy and Infectious Diseases and the National Institute of Arthritis, Musculoskeletal and Skin Diseases and National Institute of Environmental Health Sciences of the US National Institutes of Health (R01AI071043 and R01AR064831 to N.X., and ES019964 to G.H.P.) and matching institutional funds from Pennsylvania State University for T32 AI074551 from the National Institutes of Health (for J.Y.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

J.Y., S.H. and L.Z. performed experiments; D.H.K. and G.H.P. provided materials; J.Y. and N.X. designed the study; J.Y., S.H., L.Z. and N.X. analyzed the data and wrote the manuscript; and all read and commented on the manuscript.

Corresponding author

Correspondence to Na Xiong.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of CCR10+ and CCR10– ILCs of the skin and sLNs.

(a) Flow cytometric analysis of Gata-3 and T-bet expression in EGFP(CCR10)+ and EGFP– CD45+CD3–Lin– ILCs of the skin and sLN of Ccr10+/EGFP mice. (b) Flow cytometric analysis of Rorγt expression in CD45+CD3–Lin– ILCs of the skin and sLN of mice carrying one Rorc-EGFP allele for reporting Rorγt expression. The gray histogram is of wild-type ILCs as a negative control (-CTRL) for Rorγt-EGFP. The Rorγt expression in γdT cells is included as a positive control. (c) Comparison of average median fluorescence intensity (MFI) of the MHCII staining of EGFP+MHCII+ vs. EGFP–MHCII+ CD45+CD3–Lin– skin ILCs of Ccr10+/EGFP mice. N=5. (d) Flow cytometric analysis of co-expression of MHCII and IL-17 or IL-5 in CCR10+ CD45+CD3–Lin– ILCs of the sLN and skin of Ccr10+/EGFP mice. (e and f) Flow cytometric analysis of expression of indicated co-inhibitory and co-stimulatory molecules on gated EGFP(CCR10)+ CD45+CD3–Lin– ILCs in the skin (e) and sLN (f) of Ccr10+/EGFP mice. Gray histograms are of the staining with isotype control antibodies. **P<0.01.

Supplementary Figure 2 Homeostatic establishment of CCR10+ ILCs in the skin depends on continuous input of CCR10+MHCII+ ILCs from sLNs.

(a) Comparison of average numbers of EGFP+ CD45+CD3–Lin– ILCs in the sLN and skin of FTY720 (FTY)-treated and untreated (CTRL) Ccr10+/EGFP mice. Analyzed 2 weeks after the treatment. N=3. (b) Analysis of T cells of sLN and the skin for their expression of CCR10 (EGFP) in FTY720 (FTY)-treated and untreated (CTRL) Ccr10+/EGFP mice. Analyzed 2 weeks after the treatment. Bar graphs show average percentages and numbers of EGFP+ sub-populations of total T cells in the sLN and skin. N=3. (c) Comparison of numbers of CCR10 (EGFP)+ CD45+CD3–Lin– ILCs in the skin of Ccr10+/EGFP (WT), Rorc–/–Ccr10+/EGFP (Rorc–/–) and Ahr–/–Ccr10+/EGFP (Ahr–/–) mice. N=3. (d) Flow cytometric analysis of expression of CCR10 (EGFP) on CD3int skin T cells of Ccr10+/EGFP (WT) and Rorc–/–Ccr10+/EGFP (Rorc–/–) mice. (e) Flow cytometric analysis of expression of IL-17 and IL-5 on skin CD45+CD3–Lin– ILCs of Ccr10+/EGFP (WT) and Rorc–/–Ccr10+/EGFP (Rorc–/–) mice. (f) Flow cytometric analysis of expression of CCR10 (EGFP) and other indicated molecules on CD45+CD3–Lin– ILCs of sLN and the skin of Ahr–/–Ccr10+/EGFP mice. (g-h) Flow cytometric analysis of expression of CCR10 (EGFP) and IL-17 and IL-5 on CD45+CD3–Lin– ILCs of sLN (g) and the skin (h) of Ahr–/–Ccr10+/EGFP (Ahr–/–) and Ccr10+/EGFP (WT) mice. (i) Flow cytometric analysis of expression of CCR10 (EGFP) on CD3int skin T cells of Ccr10+/EGFP (WT) and Ahr–/–Ccr10+/EGFP (Ahr–/–) mice. (j) Comparison of average numbers of EGFP(CCR10)+ and EGFP+MHCII+ CD45+CD3–Lin– ILCs in the skin of Ccr10+/EGFP mice treated with LTβR-Ig in utero and untreated Ccr10+/EGFP control (CTRL) mice. N=6 for control and 10 for LTβR-Ig-treated mice. *P<0.05, **P<0.01, ***P<0.001, NS: no significant difference.

Supplementary Figure 3 Characterization of ILCs in Ccr6+/–Ccr10+/EGFP, Ccr6+/–Ccr10EGFP/EGFP, Ccr6–/–Ccr10+/EGFP and Ccr6–/–Ccr10EGFP/EGFP mice.

(a) Comparison of numbers of EGFP(CCR10)+ CD45+CD3–Lin– ILCs in sLN, skin and spleens of Ccr6+/–Ccr10+/EGFP, Ccr6+/–Ccr10EGFP/EGFP, Ccr6–/–Ccr10+/EGFP, and Ccr6–/–Ccr10EGFP/EGFP mice. N=5. (b) Flow cytometric identification of CD45+CD3–Lin– ILCs in the skin of Ccr6+/–Ccr10+/EGFP, Ccr6+/–Ccr10EGFP/EGFP, Ccr6–/–Ccr10+/EGFP, and Ccr6–/–Ccr10EGFP/EGFP mice. Gated on CD45+ lymphocytes. The bar graph on the right shows average percentages of ILCs in total CD45+ lymphocytes. N=4. (c) Flow cytometric analysis of expression of CCR10 (EGFP) and other indicated molecules on CD45+CD3–Lin– ILCs from sLN and skin of Ccr10EGFP/EGFP mice. Performed twice. (d) Representative flow cytometric analysis of donor Ccr10+/EGFP and Ccr10EGFP/EGFP EGFP+ CD45+CD3–Lin– sLN ILCs recovered in the skin of wild-type recipients 2 days after transfer of similar numbers of Ccr10+/EGFP (+/-) (CD45.1+CD45.2+) and Ccr10EGFP/EGFP (-/-) (CD45.1–CD45.2+) EGFP+ sLN ILCs (After). sLN ILCs were labeled with Violet dye before transfer for identification. Gated on Violet+45.2+CD3–Lin– EGFP+ cells. The mixture of donor sLN ILCs before transfer (Before) was shown as a control. *P<0.05, **P<0.01, NS: no significant difference.

Supplementary Figure 4 Different programming and maintenance of CCR10+ and CCR10– ILCs in sLNs and the skin in homeostatic and inflammatory conditions.

(a) Comparison of numbers of EGFP (CCR10)+ CD45+CD3–Lin– ILCs in the skin of Ccr10+/EGFP (WT) and Rag1–/–Ccr10+/EGFP (Rag1-/-) mice. N=5. (b) Flow cytometric analysis of expression of CCR10 (EGFP) and other indicated molecules on CD45+CD3–Lin– ILCs from sLN and skin of Rag1–/–Ccr10+/EGFP mice. N=3. (c) Flow cytometric analysis of expression of IL-17 and IL-5 on CD45+CD3–Lin– ILCs in the skin of Ccr10+/EGFP (WT) and Rag1–/–Ccr10+/EGFP (Rag1–/–) mice. (d) Flow cytometric analysis of donor CD45+CD3–Lin– ILCs and their expression of CCR10 and MHCII in the skin of Rag1–/– (CD45.1+CD45.2+) mice that received transfer of total sLN cells of Rag1–/–Ccr10+/EGFP (CD45.2+) mice or sorter-purified EGFP (CCR10)+ CD45+CD3–Lin– sLN ILCs of Ccr10+/EGFP (WT) or Rag1–/–Ccr10+/EGFP (Rag1–/–) (CD45.2+) mice. Analyzed two days or four weeks after transfer. Representative of two experiments. (e) Flow cytometric analysis of expression of CD44 and MHCII on EGFP+ vs. EGFP– CD45+CD3–Lin– host ILCs from sLN of Rag1–/–Ccr10+/EGFP mice transferred with Ccr10+/EGFP CD4+ splenic T cells. Analyzed 6 weeks after transfer. (f) Comparison of average numbers of EGFP(CCR10)+ CD45+CD3–Lin– ILCs in the sLN and skin of Ccr10+/EGFP (WT) and Foxp3–/–Ccr10+/EGFP (Foxp3–/–) mice. N=4. (g) Comparison of average numbers of EGFP(CCR10)+ CD45+CD3–Lin– ILCs in sLNs and the skin of calcipotriol-treated (Calcipotriol, N=7) versus untreated (CTRL, N=6) Ccr10+/EGFP mice. *P<0.05, **P<0.01.

Supplementary Figure 5 CCR10+ ILCs regulate the homeostasis of resident helper T cells in the skin.

(a) Flow cytometric analysis of donor CD4+ T cells and their expression of IL-17, IFN-γ and IL-5 in the skin of Rag1–/– and Rag1–/–Ccr10EGFP/EGFP recipient mice that were transferred with total CD4+ splenic T cells from Ccr10+/EGFP mice. Bar graphs show average percentages of IL-17, IFN-g and IL-5-producing CD4+ T cells of the donor T cells in the skin of Rag1–/– and Rag1–/–Ccr10EGFP/EGFP recipients. N=3. NS: no significant difference.

Supplementary Figure 6 Transcription factor Foxn1 and CD207+ dendritic cells are involved in the programming of CCR10+ ILCs.

(a) Comparison of average numbers of EGFP(CCR10)+ CD45+CD3–Lin– ILCs in the sLN and skin of Ccr10+/EGFP (WT) and Foxn1–/–Ccr10+/EGFP (Foxn1–/–) mice. N=4. (b) Flow cytometric analysis of EGFP+ and EGFP– CD45+CD3–Lin– ILCs of Ccr10+/EGFP donor BM cells in the skin of Foxn1–/– vs. wild-type recipient mice (CD45.2+) that were irradiated and transferred with similar numbers of total BM cells from Ccr10+/EGFP (CD45.1+CD45.2+) mice. Analyzed eight weeks after transfer. Bar graphs show average percentages of EGFP+ and EGFP+MHCII+ subpopulations of total donor cell-derived CD45+CD3–Lin– ILCs in the skin of recipients. N=5 for wild-type and 4 for Foxn1–/– mice. (c) Flow cytometric analysis of Foxn1 (RFP) expression in CD103+ and CD103– CD207+CD45+CD3–CD11c+ DCs of the sLN and skin of Foxn1+/CreGt(ROSA)26Sor+/RFP mice. Cells of Foxn1+/+Gt(ROSA)26SorRFP/RFP mice were included as negative control for RFP signals. (d) Flow cytometric analysis of CD45+CD3–Lin– ILCs and their EGFP and MHCII expression in sLNs and the skin of diphtheria toxin (DT)-treated and untreated (CTRL) MuLanDTRCcr10+/EGFP mice. Analyzed 8 days after DT treatment. (e) Comparison of average percentages of EGFP+ T cells in sLNs and the skin of MuLanDTRCcr10+/EGFP mice treated with DT for indicated periods of time. CTRL: untreated controls. N=5 for CTRL and 7 for DT for the 8-day sLN samples, 3 for CTRL and 6 for DT for the 17-21-day sLN and skin samples. (f) Comparison of percentages of EGFP+ T cells in sLNs and the skin of DT-treated HuLanDTRCcr10+/EGFP mice, analyzed similarly as in the panel (e). N=7 for CTRL and 6 for DT for the 8-day sLN samples, 4 each for the 17-21-day sLN and skin samples. *P<0.05, **P<0.01, ***P<0.001, NS: no significant difference.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 1455 kb)

Supplementary Table 1

Excel table of antibodies (XLS 47 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, J., Hu, S., Zhao, L. et al. Selective programming of CCR10+ innate lymphoid cells in skin-draining lymph nodes for cutaneous homeostatic regulation. Nat Immunol 17, 48–56 (2016). https://doi.org/10.1038/ni.3312

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3312

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