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:

Oleic acid availability impacts thymocyte preprogramming and subsequent peripheral Treg cell differentiation

Abstract

The nature of activation signals is essential in determining T cell subset differentiation; however, the features that determine T cell subset preference acquired during intrathymic development remain elusive. Here we show that naive CD4+ T cells generated in the mouse thymic microenvironment lacking Scd1, encoding the enzyme catalyzing oleic acid (OA) production, exhibit enhanced regulatory T (Treg) cell differentiation and attenuated development of experimental autoimmune encephalomyelitis. Scd1 deletion in K14+ thymic epithelia recapitulated the enhanced Treg cell differentiation phenotype of Scd1-deficient mice. The dearth of OA permitted DOT1L to increase H3K79me2 levels at the Atp2a2 locus of thymocytes at the DN2–DN3 transition stage. Such epigenetic modification persisted in naive CD4+ T cells and facilitated Atp2a2 expression. Upon T cell receptor activation, ATP2A2 enhanced the activity of the calcium–NFAT1–Foxp3 axis to promote naive CD4+ T cells to differentiate into Treg cells. Therefore, OA availability is critical for preprogramming thymocytes with Treg cell differentiation propensities in the periphery.

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

Fig. 1: naive CD4+ T cells of Scd1−/− mice, but not CD4creScd1loxP mice, have a cell-intrinsic preference for Treg cell differentiation.
Fig. 2: The Scd1-deficient thymic environment imprints DN3 thymocytes with subsequent Treg cell differentiation propensity.
Fig. 3: OA mediates the effects of SCD1 on preprogramming DN3 thymocytes for peripheral Treg cell generation.
Fig. 4: Cre-mediated Scd1 deletion in thymic K14+ cells preprograms DN3 thymocytes with Treg cell propensity.
Fig. 5: Enhanced Treg cell differentiation and EAE resistance in nude mice transplanted with thymi from K14creScd1loxP mice.
Fig. 6: Scd1 deficiency augments ATP2A2 expression to facilitate Treg cell differentiation.
Fig. 7: OA inadequacy allows DOT1L to promote H3K79me2-dependent ATP2A2 expression.

Similar content being viewed by others

Data availability

The sequencing data generated and analyzed in this publication have been deposited in NCBI’s Gene Expression Omnibus. The Mus musculus reference genome (GRCm38/mm10 assembly; https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) was used for ChIP–seq and RNA-sequencing alignment. The genome-wide maps of chromatin accessibility in DN3 thymocytes and splenic naive CD4+ T cells are deposited under the accession number GSE209563. The mRNA-sequencing data for cortical and medullary thymic epithelial cells are available under accession GSE237819, while the H3K79me2 ChIP–seq analysis data on in vitro-developed DN3 cells are available under accession GSE237573. Source data are provided with this paper.

References

  1. Cooper, M. D. & Alder, M. N. The evolution of adaptive immune systems. Cell 124, 815–822 (2006).

    CAS  PubMed  Google Scholar 

  2. Irla, M. Instructive cues of thymic T cell selection. Annu. Rev. Immunol. 40, 95–119 (2022).

    CAS  PubMed  Google Scholar 

  3. Kadouri, N., Nevo, S., Goldfarb, Y. & Abramson, J. Thymic epithelial cell heterogeneity: TEC by TEC. Nat. Rev. Immunol. 20, 239–253 (2020).

    CAS  PubMed  Google Scholar 

  4. Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. A. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Taniuchi, I. CD4 helper and CD8 cytotoxic T cell differentiation. Annu. Rev. Immunol. 36, 579–601 (2018).

    CAS  PubMed  Google Scholar 

  6. Davenport, M. P., Smith, N. L. & Rudd, B. D. Building a T cell compartment: how immune cell development shapes function. Nat. Rev. Immunol. 20, 499–506 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Chapman, N. M. & Chi, H. Metabolic adaptation of lymphocytes in immunity and disease. Immunity 55, 14–30 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Campbell, C. & Rudensky, A. Roles of regulatory T cells in tissue pathophysiology and metabolism. Cell Metab. 31, 18–25 (2020).

    CAS  PubMed  Google Scholar 

  9. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat. Genet. 27, 20–21 (2001).

    CAS  PubMed  Google Scholar 

  10. Pohar, J., Simon, Q. & Fillatreau, S. Antigen−specificity in the thymic development and peripheral activity of CD4+FOXP3+ T regulatory cells. Front Immunol. 9, 1701 (2018).

    PubMed  PubMed Central  Google Scholar 

  11. Savage, P. A., Klawon, D. E. J. & Miller, C. H. Regulatory T cell development. Annu. Rev. Immunol. 38, 421–453 (2020).

    CAS  PubMed  Google Scholar 

  12. van der Veeken, J. et al. Genetic tracing reveals transcription factor Foxp3-dependent and Foxp3-independent functionality of peripherally induced Treg cells. Immunity 55, 1173–1184 (2022).

    PubMed  PubMed Central  Google Scholar 

  13. Kitagawa, Y. et al. Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat. Immunol. 18, 173–183 (2017).

    CAS  PubMed  Google Scholar 

  14. Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gutcher, I. et al. Autocrine transforming growth factor-β1 promotes in vivo Th17 cell differentiation. Immunity 34, 396–408 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).

    CAS  PubMed  Google Scholar 

  17. Newton, R., Priyadharshini, B. & Turka, L. A. Immunometabolism of regulatory T cells. Nat. Immunol. 17, 618–625 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Berod, L. et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 20, 1327–1333 (2014).

    CAS  PubMed  Google Scholar 

  19. Field, C. S. et al. Mitochondrial integrity regulated by lipid metabolism is a cell-intrinsic checkpoint for Treg suppressive function. Cell Metab. 31, 422–437 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. AM, A. L., Syed, D. N. & Ntambi, J. M. Insights into stearoyl-CoA desaturase-1 regulation of systemic metabolism. Trends Endocrinol. Metab. 28, 831–842 (2017).

    Google Scholar 

  21. Liu, K. et al. Scd1 controls de novo beige fat biogenesis through succinate-dependent regulation of mitochondrial complex II. Proc. Natl Acad. Sci. USA 117, 2462–2472 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lochner, M., Berod, L. & Sparwasser, T. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 36, 81–91 (2015).

    CAS  PubMed  Google Scholar 

  23. Mottet, C., Uhlig, H. H. & Powrie, F. Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J. Immunol. 170, 3939–3943 (2003).

    CAS  PubMed  Google Scholar 

  24. Owen, D. L. et al. Thymic regulatory T cells arise via two distinct developmental programs. Nat. Immunol. 20, 195–205 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bettelli, E. et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238 (2006).

    CAS  PubMed  Google Scholar 

  26. Schmitt, T. M. & Zuniga-Pflucker, J. C. Induction of T cell development from hematopoietic progenitor cells by delta-like-1 in vitro. Immunity 17, 749–756 (2002).

    CAS  PubMed  Google Scholar 

  27. den Braber, I. et al. Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. Immunity 36, 288–297 (2012).

    Google Scholar 

  28. Sukseree, S. et al. Autophagy in the thymic epithelium is dispensable for the development of self-tolerance in a novel mouse model. PLoS ONE 7, e38933 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Sampath, H. et al. Skin-specific deletion of stearoyl-CoA desaturase-1 alters skin lipid composition and protects mice from high fat diet-induced obesity. J. Biol. Chem. 284, 19961–19973 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Xue, Y. et al. SCD1 sustains homeostasis of bulge niche via maintaining hemidesmosomes in basal keratinocytes. Adv. Sci. 10, e2201949 (2023).

    Google Scholar 

  31. Vassar, R., Rosenberg, M., Ross, S., Tyner, A. & Fuchs, E. Tissue-specific and differentiation-specific expression of a human K14 keratin gene in transgenic mice. Proc. Natl Acad. Sci. USA 86, 1563–1567 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Trebak, M. & Kinet, J. P. Calcium signalling in T cells. Nat. Rev. Immunol. 19, 154–169 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Dixon, R. E. et al. Graded Ca2+/calmodulin-dependent coupling of voltage-gated CaV1.2 channels. Elife 4, e05608 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Vaeth, M. et al. Dependence on nuclear factor of activated T-cells (NFAT) levels discriminates conventional T cells from Foxp3+ regulatory T cells. Proc. Natl Acad. Sci. USA 109, 16258–16263 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, M. O. & Rudensky, A. Y. T cell receptor signalling in the control of regulatory T cell differentiation and function. Nat. Rev. Immunol. 16, 220–233 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Garcia, B. A. et al. Chemical derivatization of histones for facilitated analysis by mass spectrometry. Nat. Protoc. 2, 933–938 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ferrari, F. et al. DOT1L-mediated murine neuronal differentiation associates with H3K79me2 accumulation and preserves SOX2-enhancer accessibility. Nat. Commun. 11, 5200 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Bian, Y. et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 585, 277–282 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Suzuki, K., Nagao, K., Tokunaga, J., Katayama, N. & Uyeda, M. Inhibition of DNA methyltransferase by microbial inhibitors and fatty acids. J. Enzym. Inhib. 10, 271–280 (1996).

    CAS  PubMed  Google Scholar 

  40. Hu, G. et al. Transformation of accessible chromatin and 3D nucleome underlies lineage commitment of early T cells. Immunity 48, 227–242 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Yoshida, H. et al. The cis-regulatory atlas of the mouse immune system. Cell 176, 897–912 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lai, B. et al. Principles of nucleosome organization revealed by single-cell micrococcal nuclease sequencing. Nature 562, 281–285 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Baxter, A. G. & Hodgkin, P. D. Activation rules: the two-signal theories of immune activation. Nat. Rev. Immunol. 2, 439–446 (2002).

    CAS  PubMed  Google Scholar 

  44. Curtsinger, J. M. & Mescher, M. F. Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol. 22, 333–340 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Dikiy, S. & Rudensky, A. Y. Principles of regulatory T cell function. Immunity 56, 240–255 (2023).

    CAS  PubMed  Google Scholar 

  46. Prentice, A. M. The thymus: a barometer of malnutrition. Br. J. Nutr. 81, 345–347 (1999).

    CAS  PubMed  Google Scholar 

  47. Oh-Hora, M. et al. Dual functions for the endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat. Immunol. 9, 432–443 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kim, K. D. et al. Calcium signaling via Orai1 is essential for induction of the nuclear orphan receptor pathway to drive Th17 differentiation. J. Immunol. 192, 110–122 (2014).

    CAS  PubMed  Google Scholar 

  49. Guichard, V. et al. Calcium-mediated shaping of naive CD4 T cell phenotype and function. Elife 6, e27215 (2017).

    PubMed  PubMed Central  Google Scholar 

  50. Velardi, E., Tsai, J. J. & van den Brink, M. R. M. T cell regeneration after immunological injury. Nat. Rev. Immunol. 21, 277–291 (2021).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Xinhua Liao for providing K14cre mice, L. Qiu, J. Wu, X. Miao, Q. Chu, Zi Li, Y. Ma, K. Wang, Z. Weng, Zhigang Li, Y. Zhai, Y. Bu, S. Yan and F. Zhao from Institutional Center for Shared Technologies and Facilities of SINH, CAS for technical assistance. Ying Wang discloses support for the research of this work from the National Key R&D Program of China (2022YFA0807300), National Natural Science Foundation of China (32070872, 31771641) and the March of Dimes Birth Defects Foundation of China. Y.S. discloses support for the research of this work from the National Key R&D Program of China (2018YFA0107500), National Natural Science Foundation of China (81930085, 81530043, 31961133024, 81571612) and Scientific Innovation Project of the Chinese Academy of Sciences (XDA16020403). Liangyu Lin discloses support for the research of this work from the National Natural Science Foundation of China (32000626) and a Fellowship of China Postdoctoral Science Foundation (2020M671261). Liangyu Lin acknowledges the support of SANOFI Scholarship Program. Q.L. discloses support for the research of this work from the National Natural Science Foundation of China (82301571).

Author information

Authors and Affiliations

Authors

Contributions

Liangyu Lin and H.M. conceived the project, designed and performed experiments, analyzed the data, interpreted results and prepared the manuscript. Q.L., L.D., Y.X., F.Z., F.W., K.L., J.Y., X.J., X.W., Jiaqi Wang, J.Z., B.L. and H.X. helped to conduct experiments. Li Lin conducted bioinformatic analysis. Q.L. and Yu Wang assisted in constructing the manuscript. D.R.G., X.K., Y.Y. and D.F. advised on the project. Jinyong Wang provided OP9-DL1 cells. Ying Wang and Y.S. led the project and wrote the manuscript with the co-authors.

Corresponding authors

Correspondence to Yufang Shi or Ying Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Properties of Treg cells of Scd1-/- mice.

(a) EAE was induced in Scd1-/- and Scd1+/- mice as described in Fig. 1a. The levels of Th17 and Treg cells in the central nervous system (CNS) were analyzed on day 17 post EAE induction (n = 7 mice for Scd1+/-+Iso, Scd1-/-+Iso, n = 5 mice for Scd1+/-+α-CD25, n = 6 for Scd1-/-+α-CD25). (b) The absolute number of CD4+Foxp3+ Treg cells and CD4+Foxp3 conventional T cells in various organs of Scd1-/- and Scd1+/- mice (n = 4 mice per group). (c) CD4+CD25+ T cells were isolated from the spleen of Scd1-/- mice and Scd1+/- littermate control mice, and co-cultured with activated CFSE-labeled CD4+ T cells (1 × 106) from wild type (WT) mice at indicated ratio. The proliferation of CD4+ T cells were determined by the dilution of CFSE. (d) The expression levels of CD25, CTLA-4, and GITR in CD4+Foxp3+ Treg cells were determined by flow cytometry. (e) Gating strategies (with the spleen as a representative example) for analyzing Helios expression in Treg cells. (f-g) The percentage and absolute number of Helios+ and Helios Treg cells in the various tissues of Scd1-/- mice and Scd1+/- mice under steady status (n = 4 mice per group). Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 2 Newly generated CD4+ T cells in Scd1-/- mice are prone to differentiate to Treg cells in WT recipient mice.

(a) Thymocytes were depleted of double positive and CD8 single positive cells by magnetic beads and then subjected to sort CD4+CD8CD73CD25 thymocytes by flow cytometry. The purity of sorted thymocytes was depicted. (b) The gating strategies (with the spleen as a representative example) for analyzing Treg cells in CD45.1+CD45.2+ WT mice receiving the co-transfer of CD4+CD8CD73CD25 thymocytes from CD45.1+WT and CD45.2+ Scd1-/- mice. (c) The percentage of Foxp3+ cells in CD4+ cells of CD45.1+WT and CD45.2+Scd1-/- origin were determined 6 weeks post thymocyte co-transfer (n = 6 mice per group). (d) The absolute number of WT CD45.1+CD4+Foxp3+ and Scd1-/- CD45.2+CD4+Foxp3+ cells in various organs of WT recipient mice were numerated (n = 6 mice per group). (e) The absolute number of CD4+ T cells derived from CD45.1+ and CD45.2+ donor cells were assessed (n = 6 mice per group). Data are presented as Mean ± SEM. NS, not significant; P values were determined by paired two-tailed Student’s t-test.

Source data

Extended Data Fig. 3 Functionality of CD4+ T Cells isolated from Scd1+/- and Scd1-/- mice.

(a) The percentage of CD44CD62L+, CD44+CD62L, and CD44+CD62L+ CD4+ T cells in the spleen of Scd1+/- (n = 7) and Scd1-/- mice (n = 5) at steady status. (b) CFSE-labeled splenic CD4+CD25 conventional T cells from Scd1+/- and Scd1-/- mice were subjected to anti-CD3/CD28 stimulation in vitro for 3 days and cell proliferation was detected by assessing CFSE dilution. (c-f) Splenic CD4+CD62L+T cells from Scd1-/- and Scd1+/- mice were cultured in Th1, Th2, or Th17 induction medium for 3 days (n = 3 for Th1 and Th17 differentiation, n = 4 for Th2 differentiation). The percentages of IFNγ+, IL-4+, or IL-17+ CD4+T cells were determined by flow cytometry. Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 4 Treg cells are comparable in CD4creScd1loxP and Scd1loxP mice.

(a) mRNA levels of Scd1 in CD4+ T cells and hepatocytes isolated from CD4creScd1loxP mice and Scd1loxP littermate control mice (n = 5 mice for each group). (b) The percentages of stearic acid (18:0) and oleic acid (18:1n9) among total fatty acids in in vitro expanded CD4+ T cells isolated from CD4creScd1loxP mice and Scd1loxP mice (n = 3 for each group). (c) The CD4+ T cell reconstitution efficiency in Rag2-/- mice receiving co-transfer of CD45.1+WT CD4+ T cells and CD45.2+CD4creScd1loxP CD4+ T cells (n = 4 mice per group). (d) The percentage of Treg cells among CD4+ T cells in the spleen, auxiliary lymph node and central nervous system of CD4creScd1loxP mice and Scd1loxP mice on day 0 (steady, n = 3 mice per group), day 18 (peak, n = 5 for Scd1loxP mice, n = 3 for CD4creScd1loxP mice), and day 24 (recovery, n = 5 for Scd1loxP mice, n = 3 for CD4creScd1loxP mice) post EAE induction. Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 5 The T cell reconstitution efficiency of WT and Scd1-/- bone marrow cells were comparable.

(a) Representative gating strategies in analyzing the thymocyte populations of the recipient mice as treated in Fig. 2a. The reconstitution efficiency of CD45.1+ and CD45.1+CD45.2+ cells within thymocytes at DN, DP, and SP stages were depicted. (b) The reconstitution efficiency of CD45.1+WT and CD45.1+CD45.2+Scd1-/- bone marrow cells transplantation (BMT) in different tissues of recipient mice as treated in Fig. 2a. (c) The percentages of donor CD4+ cells among CD45+ cells in the blood and different organs of recipient mice in Fig. 2a. (d) The thymic CD4CD8 double negative (DN) subpopulation levels in Scd1+/- mice and Scd1-/- mice (n = 5 mice for each group). (e) The thymic DN, CD8SP, CD4SP, and DP levels in Scd1+/- mice and Scd1-/- mice (n = 6 mice for each group). Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 6 DN3 and DN4 cells of Scd1 deficient origin generated CD4+ T cells with intrinsic Treg propensities upon injection into the thymus of WT mice.

(a) Gating strategies (with the spleen as a representative sample) used to analyze the Treg cell proportion among donor-derived CD4+ T cells. (b-c) Thymocyte population from CD45.2+Scd1-/- mice and CD45.1+WT mice at the DN1, DN2, DN3, or DN4 stage were mixed at a 1:1 ratio and co-transferred into irradiated (3 Gy) CD45.1+CD45.2+ WT recipient mice via intrathymic injection (n = 5 mice for DN1, DN2 transfer; n = 6 mice for DN3, DN4 transfer). Four weeks post transfer, the recipient mice received WT and Scd1-/- DN thymocytes were euthanized to numerate the absolute number of CD4+ T cells (b) and CD4+Foxp3+ Treg cells (c) in various organs. Data are presented as Mean ± SEM. NS, not significant; P values were determined by paired (b, c) two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 The influence of SCD1 inhibitor on in vitro T cell development.

(a) The progression of in vitro T cell development in the OP9-DL1 and HSC co-culture system. The effect of SCD1 inhibitor at a concentration of 40 nM was tested. The expression of CD44 and CD25 was used to monitor T cell developmental stages. (b) The proliferation of OP9-DL1 cells treated with SCD1 inhibitor at indicated doses was measured by CCK8 (n = 5 for each group). (c) Apoptosis in OP9-DL1 cells treated with SCD1 inhibitor at indicated doses was determined by Propidium Iodide/Annexin V staining (n = 4 for each group). (d) Schematic representation of different treatment protocols used in the in vitro T cell developmental system. HSCs with indicated background cultured on OP9-DL1 cells with different treatments were matured into DN3 stages and then co-transferred to Rag2-/- mice. (e) Representative gating strategies (with the spleen as a representative example) for analyzing T cells isolated from Rag2-/- mice receiving DN3 co-transferred as described in Extended Data Fig. 7d. (f) DN3 derived Treg cells in different tissues of Rag2-/- recipient mice were analyzed by flow cytometry (n = 7 mice). (g) The levels of CD4+Foxp3+ Treg cells in the blood, liver, and lungs of mice in Fig. 3e. (h-i) The percentage and absolute number of HeliosRORγt+ and Helios+ RORγt cells in the colon of mice with treatment as described in Fig. 3e (n = 5 mice for WT-ND, WT-OA; n = 4 mice for KO-DN, KO-OA). Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test (b, c, g, h, i) or paired two-tailed Student’s t-test (f).

Source data

Extended Data Fig. 8 The impact of Scd1 deficiency on the health status and thymic epithelial cells.

(a) Weight of body, spleen, and MLNs of K14creScd1loxP and Scd1loxP mice (n = 5 mice for each group). (b-c) The whole image and H&E staining of various organs of K14creScd1loxP and Scd1loxP mice. (d) Gating strategies used for identifying cortical thymic epithelial cells (cTEC) and medullar thymic epithelial cells (mTEC) of Scd1+/- and Scd1-/- mice, with subsequent calculation of their absolute numbers (n = 3 mice for each group). (e-f) cTEC and mTEC of Scd1+/- and Scd1-/- mice were isolated and subjected to mRNA sequencing analysis. The expression levels of critical regulators governing thymocyte migration, development, and selection in thymic epithelial cells were shown (e). The differentially expressed genes in cTEC or mTEC of Scd1+/- and Scd1-/- mice were shown in a volcano plot (f). (g) The percentage and absolute number of HeliosRORγt+Foxp3+ and Helios+ RORγtFoxp3+ cells in the colon of K14creScdloxP (n = 5) and Scd1loxP (n = 7) mice. (h-i) Average MALDI-TOF mass spectra of oleic acid in Fig. 4a. Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 Evaluation on OT-II T cells developed in K14creScd1loxP mice.

(a) Schematic representation of the experimental design to assess the differentiation potential of OT-II cells developed in K14creScd1loxP or Scd1loxP mice. (b) Gating strategies (with the spleen as a representative example) for identifying transferred OT-II T cells in the recipient mice (c) OT-II T cells developed in K14creScd1loxP or Scd1loxP mice were labeled with CFSE and co-cultured with splenocytes from CD45.1+WT mice in the presence of OVA323-339 peptide. The CFSE dilution in OT-II T cells was measured 96 h post-stimulation. (d-g) CD45.2+CD4+CD62L+TCRVβ5.1+ cells were isolated from the spleen of Scd1loxP or K14creScd1loxP mice and intravenously transferred to CD45.1+ WT recipient mice at a dose of 1 × 107 cells per mouse. To test Treg cell generation, recipient mice were supplied with 2 mg/mL OVA in drinking water for 7 consecutive days and examined for the percentage and number of Foxp3+ OT-II T cells in the spleen, MLNs and colon (n = 5 mice for each group) (d). To test Th1 and Th17 cell induction, recipient mice were s.c. injected with OVA emulsified in CFA and examined for the percentage and number of IL-17- and IFNγ-secreting OT-II T cells in the draining lymph nodes (DLNs) and lungs (n = 7 mice for each group) (e-f). To test Th2 cell induction, recipient mice were i.p. injected with OVA absorbed to alum adjuvant and examined for the percentage and number of IL-4-secreting OT-II T cells in the spleen and liver (n = 4 mice for each group) (g). The percentage and absolute number of distinct OT-II cell subsets were determined. (h-k) OVA323-339 peptide was supplied to the co-culture of CD45.2+CD4+CD62L+TCRVβ5.1+ cells and CD45.1+WT splenocytes to activate OT-II T cells in Th1 (n = 4 biologically independent samples), Th2 (n = 4 biologically independent samples), Th17 (n = 3 biologically independent samples), or Treg (n = 3 biologically independent samples) cell induction medium. Ninety-six hours later, the expression of IFNγ, IL-4, IL-17, or Foxp3 was determined by flow cytometry. Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test.

Source data

Extended Data Fig. 10 Scd1 deficiency promotes ATP2A2 expression through modulating DOT1L activities.

(a) Heat map display of ATAC-seq data from naïve CD4+ T and DN3 cells. (b) Venn diagram of overlapped differential ATAC peaks related genes. (c) Atp2a2 mRNA levels in naïve CD4+ T cells (n = 4 per group). (d) SCD1 inhibitor A939572 (SCD1i, 40 nM) was supplied to the in vitro T cell developmental system. DN3 thymocytes were harvested for ATP2A2 analysis (repeated twice). (e-f) WT naïve T cells were cultured in the Treg induction medium with or without SCD1i. The ATP2A2 protein (e) and the Treg (f) levels were determined (repeated twice). (g) The statistical analysis of western blot as described in Fig. 6c from 3 independent experiments. (h) Scramble siRNA (NC) or Atp2a2-targeting siRNA was transfected to CD4+CD62L+ T cells. The knockdown efficiency was determined 24 h after transfection (repeated twice). (i) SOCE in Scd1-/-CD4+CD62L+ T cells transfected with Atp2a2 -targeting siRNA or NC was detected. (j-k) WT CD4+CD62L+ T cells were transfected with adenovirus particles to overexpress ATP2A2 (n = 3 for each group) (j). T cells were subsequently cultured in the Treg induction medium for another 3 days to evaluate Treg levels (k). (l) The heat map showing the H3K79me2 levels at the genome of DN3 developed with or without SCD1i. (m) The H3K79me2 levels at the genetic locus of Atp2a2. (n-o) The statistical analysis of Fig. 7f (n) and 7h (o) obtained from 4 independent experiments. (p) SCD1i and DOT1L inhibitor EPZ005676 were added to the in vitro T cell development system. The resultant DN3 thymocytes were examined for ATP2A2 expression (repeated twice). (q) The levels of oleic acid and stearic acid in the nucleus of thymocytes from Scd1-/- or Scd1+/- mice (n = 6 for each group). (r) Palmitoleic acid (n = 4) or Palmitic acid (n = 3) was diluted to the indicated concentrations with 10% DMSO and supplied to an enzymatic reaction catalyzed by DOT1L. Data are presented as Mean ± SEM. NS, not significant; P values were determined by unpaired two-tailed Student’s t-test (c, f, k, q) or paired two-tailed Student’s t-test (n, o).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5, Table 1 and protocols.

Reporting Summary

Peer Review File

Supplementary Data 1

Statistical source data.

Supplementary Data 2

Statistical source data.

Supplementary Data 3

Statistical source data.

Supplementary Data 4

Statistical source data.

Supplementary Data 5

Statistical source data.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, L., Hu, M., Li, Q. et al. Oleic acid availability impacts thymocyte preprogramming and subsequent peripheral Treg cell differentiation. Nat Immunol 25, 54–65 (2024). https://doi.org/10.1038/s41590-023-01672-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01672-1

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