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:

CD69 controls the uptake of L-tryptophan through LAT1-CD98 and AhR-dependent secretion of IL-22 in psoriasis

An Erratum to this article was published on 20 September 2016

This article has been updated

Abstract

The activation marker CD69 is expressed by skin γδ T cells. Here we found that CD69 controlled the aryl hydrocarbon receptor (AhR)-dependent secretion of interleukin 22 (IL-22) by γδ T cells, which contributed to the development of psoriasis induced by IL-23. CD69 associated with the aromatic-amino-acid-transporter complex LAT1-CD98 and regulated its surface expression and uptake of L-tryptophan (L-Trp) and the intracellular quantity of L-Trp-derived activators of AhR. In vivo administration of L-Trp, an inhibitor of AhR or IL-22 abrogated the differences between CD69-deficient mice and wild-type mice in skin inflammation. We also observed LAT1-mediated regulation of AhR activation and IL-22 secretion in circulating Vγ9+ γδ T cells of psoriatic patients. Thus, CD69 serves as a key mediator of the pathogenesis of psoriasis by controlling LAT1-CD98-mediated metabolic cues.

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: CD69-deficient mice develop a mild form of IL-23-induced psoriasis.
Figure 2: CD69 deficiency weakens IL-23-triggered IL-22 expression and STAT3 signaling in the epidermis.
Figure 3: CD69 regulates the expression of AhR and IL-22 in dermal resident γδ T cells.
Figure 4: IL-22 mediates skin inflammation downstream of CD69.
Figure 5: IL-23-induced skin inflammation is prevented by inhibition of AhR.
Figure 6: CD69 associates with LAT1-CD98 and controls the uptake of amino acids.
Figure 7: CD69 regulates AhR-induced secretion of IL-22 by dermal γδ T cells in vitro and in vivo through control of L-Trp uptake.
Figure 8: CD69 expression is detected in skin and circulating Vγ9+ T cells from psoriatic patients.

Similar content being viewed by others

Change history

  • 11 July 2016

    In the version of this article initially published online, the identification of dermal and epidermal γδ T cells in the legend for Figure 3f was reversed; a label was missing above the far left column of Figure 4c; and the red and blue lines were switched in the keys for the far right plots in Figure 6i. The legend for Figure 3f should read "...identified by high expression (top right; epidermal) or low expression (bottom right; dermal) of the γδ TCR." The far left column in Figure 4c should include the label "CD69-KO" above. The correct keys for Figure 6i are as follows: blue line, FIZC (37 °C), and red line, FICZ + BCH (37 °C); and blue line, CD69-KO (37 °C), and red line, WT (37°C). These errors have been corrected for the print, PDF and HTML versions of this article.

References

  1. Di Meglio, P., Villanova, F. & Nestle, F.O. Psoriasis. Cold Spring Harb. Perspect. Med. 4, a015354 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Lowes, M.A., Suárez-Fariñas, M. & Krueger, J.G. Immunology of psoriasis. Annu. Rev. Immunol. 32, 227–255 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Leonardi, C. et al. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N. Engl. J. Med. 366, 1190–1199 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Chan, J.R. et al. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J. Exp. Med. 203, 2577–2587 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zheng, Y. et al. Interleukin-22, a TH17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 445, 648–651 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Wolk, K. et al. IL-22 and IL-20 are key mediators of the epidermal alterations in psoriasis while IL-17 and IFN-γ are not. J. Mol. Med. 87, 523–536 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Van Belle, A.B. et al. IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice. J. Immunol. 188, 462–469 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Prans, E. et al. Copy number variations in IL22 gene are associated with psoriasis vulgaris. Hum. Immunol. 74, 792–795 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Shimauchi, T. et al. Serum interleukin-22 and vascular endothelial growth factor serve as sensitive biomarkers but not as predictors of therapeutic response to biologics in patients with psoriasis. J. Dermatol. 40, 805–812 (2013).

    CAS  PubMed  Google Scholar 

  10. Wolk, K. et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur. J. Immunol. 36, 1309–1323 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Martin, B., Hirota, K., Cua, D.J., Stockinger, B. & Veldhoen, M. Interleukin-17-producing γδ T cells selectively expand in response to pathogen products and environmental signals. Immunity 31, 321–330 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Qiu, J. et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Veldhoen, M., Hirota, K., Christensen, J., O'Garra, A. & Stockinger, B. Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells. J. Exp. Med. 206, 43–49 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Smirnova, A. et al. Evidence for new light-independent pathways for generation of the endogenous aryl hydrocarbon receptor agonist FICZ. Chem. Res. Toxicol. 29, 75–86 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Sinclair, L.V. et al. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cai, Y. et al. Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation. Immunity 35, 596–610 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Laggner, U. et al. Identification of a novel proinflammatory human skin-homing Vγ9Vδ2 T cell subset with a potential role in psoriasis. J. Immunol. 187, 2783–2793 (2011).

    Article  CAS  PubMed  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ahlfors, H. et al. IL-22 fate reporter reveals origin and control of IL-22 production in homeostasis and infection. J. Immunol. 193, 4602–4613 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Sumaria, N. et al. Cutaneous immunosurveillance by self-renewing dermal γδ T cells. J. Exp. Med. 208, 505–518 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. González-Amaro, R., Cortés, J.R., Sánchez-Madrid, F. & Martín, P. Is CD69 an effective brake to control inflammatory diseases? Trends Mol. Med. 19, 625–632 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Sancho, D. et al. CD69 downregulates autoimmune reactivity through active transforming growth factor-β production in collagen-induced arthritis. J. Clin. Invest. 112, 872–882 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Martín, P. et al. The leukocyte activation antigen CD69 limits allergic asthma and skin contact hypersensitivity. J. Allergy Clin. Immunol. 126, 355–365, 365.e1–365.e3 (2010).

    Article  PubMed  CAS  Google Scholar 

  24. Cruz-Adalia, A. et al. CD69 limits the severity of cardiomyopathy after autoimmune myocarditis. Circulation 122, 1396–1404 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Radulovic, K. et al. CD69 regulates type I IFN-induced tolerogenic signals to mucosal CD4 T cells that attenuate their colitogenic potential. J. Immunol. 188, 2001–2013 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Rutz, S., Eidenschenk, C. & Ouyang, W. IL-22, not simply a Th17 cytokine. Immunol. Rev. 252, 116–132 (2013).

    Article  PubMed  CAS  Google Scholar 

  27. Martín, P. et al. CD69 association with Jak3/Stat5 proteins regulates Th17 cell differentiation. Mol. Cell. Biol. 30, 4877–4889 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Mackay, L.K. et al. The developmental pathway for CD103+CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14, 1294–1301 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Awasthi, A. et al. Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells. J. Immunol. 182, 5904–5908 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Shiow, L.R. et al. CD69 acts downstream of interferon-α/β to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440, 540–544 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Ribot, J.C. et al. CD27 is a thymic determinant of the balance between interferon-γ- and interleukin 17-producing gammadelta T cell subsets. Nat. Immunol. 10, 427–436 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kashiwagi, H., Yamazaki, K., Takekuma, Y., Ganapathy, V. & Sugawara, M. Regulatory mechanisms of SNAT2, an amino acid transporter, in L6 rat skeletal muscle cells by insulin, osmotic shock and amino acid deprivation. Amino Acids 36, 219–230 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Taylor, P.M. Role of amino acid transporters in amino acid sensing. Am. J. Clin. Nutr. 99, S223–S230 (2014).

    Article  CAS  Google Scholar 

  34. Harden, J.L. et al. The tryptophan metabolism enzyme L-kynureninase is a novel inflammatory factor in psoriasis and other inflammatory diseases. J. Allergy Clin. Immunol. 137, 1830–1840 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Rizzo, H.L. et al. IL-23-mediated psoriasis-like epidermal hyperplasia is dependent on IL-17A. J. Immunol. 186, 1495–1502 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Boniface, K. et al. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. J. Immunol. 174, 3695–3702 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Mitra, A., Raychaudhuri, S.K. & Raychaudhuri, S.P. IL-22 induced cell proliferation is regulated by PI3K/Akt/mTOR signaling cascade. Cytokine 60, 38–42 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Finlay, D.K. et al. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 209, 2441–2453 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mascanfroni, I.D. et al. Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α. Nat. Med. 21, 638–646 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fenczik, C.A. et al. Distinct domains of CD98hc regulate integrins and amino acid transport. J. Biol. Chem. 276, 8746–8752 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Lauzurica, P. et al. Phenotypic and functional characteristics of hematopoietic cell lineages in CD69-deficient mice. Blood 95, 2312–2320 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Di Meglio, P. et al. Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity 40, 989–1001 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Walter, A. et al. Aldara activates TLR7-independent immune defence. Nat. Commun. 4, 1560 (2013).

    Article  PubMed  CAS  Google Scholar 

  44. van den Bogaard, E.H. et al. Genetic and pharmacological analysis identifies a physiological role for the AHR in epidermal differentiation. J. Invest. Dermatol. 135, 1320–1328 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Furue, M. et al. Gene regulation of filaggrin and other skin barrier proteins via aryl hydrocarbon receptor. J. Dermatol. Sci. 80, 83–88 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Serre, K. & Silva-Santos, B. Molecular mechanisms of differentiation of murine pro-inflammatory γδ T cell subsets. Front. Immunol. 4, 431 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Shibata, K. et al. Notch-Hes1 pathway is required for the development of IL-17-producing γδ T cells. Blood 118, 586–593 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Mukai, M. & Tischkau, S.A. Effects of tryptophan photoproducts in the circadian timing system: searching for a physiological role for aryl hydrocarbon receptor. Toxicol. Sci. 95, 172–181 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Kondo, M. et al. Expansion of human peripheral blood gammadelta T cells using zoledronate. JoVE 55, 3152 (2011).

    Google Scholar 

Download references

Acknowledgements

We thank D. Rotin (University of Toronto) for the plasmid for the expression of LAT1-mCherry; P. Taylor (University of Dundee) for antiserum to human LAT1; M. Navarro (Universidad Autónoma de Madrid, Spain) for IL-23R-GFP reporter mice; S. Bartlett for English editing; and T. Hernandez and R. Brid Doohan for technical assistance with immunohistochemistry. Supported by the Spanish Ministry of Economy and Competitiveness (SAF2011-25834 and SAF2014-55579-R to F.S.-M.; SAF2011-27330 to P.M.; and SAF2013-42850 to M.F.), Comunidad de Madrid (INDISNET-S2011/BMD-2332 to F.S.-M.; and 2010/BMD-2332 from M.F. and F.S.-M.), Instituto Salud Carlos III (Red Cardiovascular RD 12-0042-0056 to F.S.-M.; BIOIMID to M.F. and F.S.-M.), the European Research Council (ERC-2011-AdG 294340-GENTRIS to F.S.-M.) and the Ramón Areces foundation (M.F. and F.S.-M.).

Author information

Authors and Affiliations

Authors

Contributions

D.C. performed mice experimentation, analyzed and interpreted data and wrote the manuscript; M.L.S. collaborated on mice experimentation, data interpretation and writing of the manuscript; H.d.l.F. performed analysis of psoriatic patients; R.S.-D. and O.M.-G. performed quantitative PCR; I.J., A.F. and J.V. performed proteomic and metabolic-mass spectrometry analyses; C.P. contributed expertise in radioactive assays; M.F., M.V.-M. and P.M.F.-S. provided reagents and helped with the revision of the manuscript; E.D. provided biopsies from psoriatic patients and their clinical diagnostics; P.M. helped to design research, provided reagents, collaborated in data interpretation and manuscript writing; and F.S.-M. planned research, discussed results and collaborated in writing the manuscript.

Corresponding author

Correspondence to Francisco Sánchez-Madrid.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Resident skin γδ T cell populations are not affected by CD69 expression.

(a) Flow cytometry (left) analyzing the expression of IL-22 and IL-17 by CD69-deficient and wild-type TH17 cells obtained in vitro. Numbers in quadrants indicates percent cells in each (through). Middle, frequency of IL-22+ and IL-17+ cells gated as above. Right, ELISA quantification of IL-22 and IL-17 levels in the supernatants of cells described above. (b) Expression of AhR in TH17 cells as in a. (c) Quantitative PCR analysis of Il22, AhR, Ahrr, Cyp1a1 and Cyp1b1 mRNA in TH17 cells from CD69-deficient and wild-type mice (Key) cultured by 48h as in a; results calculated as in Fig. 2a,b are present relative to those of CD69-deficient cells. (d) Flow cytometry of total or epidermal or dermal skin cells suspensions from ears of wild-type mice (e) Expression of CD69 in epidermal and dermal γδ T cells sorted from the skin of mice. (f) Frequency of CD3+ populations of cells detected in the skin of CD69-deficient and wild-type mice, in steady state. NS, * P < 0.05, ** P < 0.01, *** P < 0.001; data was analyzed with unpaired t-test for a-c; and two way ANOVA with bonferroni multiple comparison test for f. Three independent experiments were performed with similar results for a, b (n=4 mice per group) and for f (n=5 mice per group). Error bars represent the mean + SEM for a-c and f.

Supplementary Figure 2 Dermal γδ T cells are the main target of IL-23-induced psoriasis.

(a) Flow cytometry (left) analyzing IL-22 and IL-17 secretion by wild-type dermal γδ T cells five hours after a single intradermal IL-23 (500 ng) or PBS injection, and with an intraperitoneal injection of Brefeldin A (250 µg/mice). Right, frequency of IL-22+ and IL-17+ cells gated from dermal γδ T cells as above, and total number of cells detected per ear after IL-23 and PBS treatment. (b) Flow cytometry (left) analyzing IL-22 and IL-17 secretion by sorted wild-type dermal γδ T cells stimulated in vitro with IL-23 plus IL-1β from 24h and 48h. Middle, frequency of IL-22+ and IL-17+ cells gated as above. Right, expression of AhR in wild-type dermal γδ T cells in vitro stimulated as above. (c) Expression of RORγt, pSTAT3 and pSTAT5 detected by flow cytometry in sorted dermal γδ T cells from CD69-deficient and wild-type mice after 24h of stimulation with IL-23+IL-1β. (d) Flow cytometry (left) of skin suspensions from wild-type and IL23R-GFP+ reporter mice showing the expression of GFP+ restricted to dermal γδ T cells in homeostatic conditions. Right, expression of IL-23R (GFP) in epidermal and dermal γδ T cells sorted from the skin of wild-type and IL-23R-GFP reporter mice. (e) Flow cytometry analyzing expression of IL-23R in dermal γδ T cells gated on live cells from CD69-deficient and wild-type mice skin suspensions. Right, expression of IL-23R in sorted dermal γδ T cells from CD69-deficient and wild-type mice. NS, * P < 0.05, ** P < 0.01, *** P < 0.001, unpaired t-test was used for panel a, c and e. Two independent experiments were performed with similar results for panels a-e. Data is showed as mean+s.e.m.; n=5 mice per group for panels a, c, e; n=4 mice per group for panel b.

Supplementary Figure 3 AhR regulates skin inflammation induced by IL-23.

(a) Analysis of ear thickness of CD69-deficient and wild-type mice during the administration of eight doses of IL-22 (500ng/dose) (horizontal axis) (left). Quantitative PCR analysis of Il1b, mS100a8 and mS100a9 genes detected after completed study indicated above (right) (presented as in Fig. 2a,b). (b) Flow cytometry (left) of live-gated dermal γδ T cells from AhR-deficient (AhR-KO) and wild-type littermates mice after three doses of IL-23 (500ng/dose) or PBS and sacrificed upon systemic administration of Brefeldin A. Right, percent and total number per ear of IL-22+ and IL-17+ cells gated as above. (c) Flow cytometry (left) of sorted dermal γδ T cells from CD69-deficient and wild-type mice stimulated in vitro with IL-23 plus IL-1β in the presence of CH-223191(3µM) or its vehicle (DMSO) by 24 hours. Right, frequency of IL-22+ and IL-17+ cells gated as above (d) Expression of CD69 in wild-type CD27- γδ T cells sorted from spleen and lymph-nodes suspensions and stimulated in vitro by 24h with IL-23, IL-1β or combination of both. (e) CD69-deficient and wild-type spleen CD27-γδ T cells were stimulated in vitro with IL-23 plus CH-223191 or DMSO. Flow cytometry (left) and frequency (right) of IL-22+ and IL-17+ γδ T cells gated from live cells. US, unstimulated. AU, arbitrary units, NS, not significant; *** P < 0.001. Two way ANOVA with bonferroni multiple comparison test was used for panels a, c, e; and one way ANOVA with newman-keuls multiple comparison test was used for panels b, d. Two independent experiment were performed for each panels; n=6 mice per group for panel a (mean ± s.d), n=5 mice per group for panel b (each symbol represent an individual mice and small horizontal lines indicate the mean ± s.d.) and n=4 mice per group for panels c, d, e (mean+s.e.m.).

Supplementary Figure 4 CD69 binds specifically to LAT1 and CD98.

(a) Extracted ion chromatogram traces of the monoisotopic peaks from the doubly-charged ions corresponding to four selected peptides from LAT1 and CD98 or from two non-specifically binding proteins (DNPEP and KV2A4). (b-c) Quantitative PCR of nutrient transporters were assessed in CD69-deficient and wild-type CD4+ T cells activated by 24 hours with anti-CD3 and anti-CD28 antibodies. NS, * P < 0.05, data was analyzed with two way ANOVA with bonferroni multiple comparison test for panel b, and unpaired t-test for panel c. Three independent proteomic assays were performed and two experiments for panels b, c, (n=5 mice per group, mean + s.e.m.) with similar results.

Supplementary Figure 5 Internalization of CD69 reduces the uptake of amino acids through LAT1.

(a) Uptake of 3H-labeled amino acids (AAs) by CD4+ T cells from CD69-deficient and wild-type mice activated by 24h with anti-CD3 and anti-CD28, at various times (horizontal axes, top row). Uptake of each labeled amino acid after 15 min, in the presence or absence of BCH (horizontal axes, bottom row). (b) Flow cytometry analyzing membrane and total expression of LAT1 and CD98 in CD69-deficient and wild-type CD4+ T cells activated as above and treated with anti-CD69 blocking Ab (2.2.) or its control isotype Ab (2.8). (c) Confocal image of a HEK cell co-transfected with CD69-GFP and LAT1-Cherry expressing plasmids that was incubated with Alexa-647 Zenon labeled anti-CD69 mAb. (d) Uptake of 3H-labeled L-Trp and L-Phe by wild-type and CD69-deficient activated CD4+ T cells treated as in b at various time (horizontal axes, left) and uptake of those amino acids after 15 minutes in the presence or absence of BCH (horizontal axes, right). NS ** P < 0.01; *** P < 0.001. Data were analyzed with two way ANOVA and Bonferroni multiple comparison test. For amino acid uptake curves a linear regression analysis with slopes comparison were conducted. At least three independent experiments were conducted for each panel, with similar results (mean + s.e.m, n = 5 replicates from each group).

Supplementary Figure 6 CD69 controls mTORC and the secretion of IL-22 by TH17 cells.

Immunoblot analysis of mTORC signaling in CD69-deficient and wild-type TH17 cells obtained after 24 hours (a) and 96 hours (b) cultured in RPMI medium. (c) Flow cytometry (above plots) analyzing the expression of IL-22 and IL-17 by CD69-deficient and wild-type TH17 cells polarized in vitro in RPMI medium alone or supplemented with BCH or FICZ, or cultured in IMDM medium. Frequency (bottom row) of IL-22+ and IL-17+ CD4+ T cells gated as above. (d) Quantitative PCR of L-Trp degrading enzymes mRNA expression were tested in the skin of CD69-deficient and wild-type mice that received IL-23 or IL23 plus IL-22, at the end of the studies (presented as in Fig. 2 a.b). NS, * P < 0.05, results were compared using two way ANOVA followed by bonferroni multiple comparison test. Two independent experiments were conducted for each panel, n=3 or 4 mice per group (panels a, b,c), n=5 mice per group (panel d). Error bar mean + s.e.m. for c,d.

Supplementary Figure 7 CD69 controls the uptake of L-Trp, activation of AhR and IL-22.

CD69 expression is required for the optimal expression of LAT1 in the membrane and uptake of aromatic amino acid by activated lymphocytes. L-Trp is photoconverted or catabolized in the cytosol to several AhR ligands, including FICZ, that regulates AhR-mediated signaling and IL-22 secretion.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–3 (PDF 2977 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cibrian, D., Saiz, M., de la Fuente, H. et al. CD69 controls the uptake of L-tryptophan through LAT1-CD98 and AhR-dependent secretion of IL-22 in psoriasis. Nat Immunol 17, 985–996 (2016). https://doi.org/10.1038/ni.3504

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3504

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