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:

PD-1 signaling facilitates activation of lymphoid tissue inducer cells by restraining fatty acid oxidation

Abstract

Anti-programmed death-1 (PD-1) immunotherapy that aims to restore T cell activity in cancer patients frequently leads to immune-related adverse events such as colitis. However, the underlying mechanism is still elusive. Here, we find that Pdcd1-deficient mice exhibit disrupted gut microbiota and aggravated dextran sulfate sodium (DSS)-induced colitis. In addition to T cells, PD-1 is also substantially expressed in colonic lymphoid tissue inducer (LTi) cells. During DSS-induced colitis, LTi cell activation is accompanied by increased PD-1 expression, whereas PD-1 deficiency results in reduced interleukin-22 (IL-22) production by LTi cells and exacerbated inflammation. Mechanistically, activated LTi cells reprogram their metabolism toward carbohydrate metabolism and fatty acid synthesis, while fatty acid oxidation (FAO) is unchanged. However, PD-1 deficiency leads to significantly elevated FAO in LTi cells, which in turn attenuates their activation and IL-22 production. Consistently, FAO suppression efficiently restores IL-22 production in Pdcd1−/− LTi cells. Thus, our study provides unforeseen mechanistic insight into colitis occurrence during anti-PD-1 immunotherapy through LTi cell metabolic reconfiguration.

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: PD-1 deficiency results in gut dysbiosis and exacerbated colitis.
Fig. 2: LTi cell activation induces PD-1 upregulation.
Fig. 3: PD-1 deficiency reduces IL-22 production in LTi cells.
Fig. 4: PD-1 signaling facilitates LTi cell activation and metabolic reprogramming.
Fig. 5: Single-cell analysis unravels metabolic reprogramming during ILC3 activation.
Fig. 6: Elevated FAO suppresses IL-22 expression in LTi cells.
Fig. 7: FAO inhibition recovers IL-22 defect in Pdcd1−/− LTi cells.

Similar content being viewed by others

Data availability

RNA-seq and scRNA-seq data generated in this study are publicly available in the NCBI Gene Expression Omnibus repository under the accession number GSE144687. This study did not generate any unique codes. All other data can be made available from the authors on reasonable request. Source data are provided with this paper.

References

  1. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    Article  PubMed  Google Scholar 

  2. Keir, M. E., Butte, M. J., Freeman, G. J. & Sharpe, A. H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Haanen, J. et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 28, iv119–iv142 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Postow, M. A., Sidlow, R. & Hellmann, M. D. Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378, 158–168 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Macdonald, T. T. & Monteleone, G. Immunity, inflammation, and allergy in the gut. Science 307, 1920–1925 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. Fuchs, A. et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity 38, 769–781 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Klose, C. S. N. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Barlow, J. L. et al. Innate IL-13-producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity. J. Allergy Clin. Immunol. 129, 191-8.e1-4 (2012).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tang, Q. et al. Development of IL-22-producing NK lineage cells from umbilical cord blood hematopoietic stem cells in the absence of secondary lymphoid tissue. Blood 117, 4052–4055 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. 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).

    Article  CAS  PubMed  Google Scholar 

  16. Klose, C. S. N. et al. A T-bet gradient controls the fate and function of CCR6RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Zhong, C., Zheng, M. & Zhu, J. Lymphoid tissue inducer—a divergent member of the ILC family. Cytokine Growth Factor Rev. 42, 5–12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhong, C. et al. Differential expression of the transcription factor GATA3 specifies lineage and functions of innate lymphoid cells. Immunity 52, 83–95.e4 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Zhong, C. & Zhu, J. Transcriptional regulators dictate innate lymphoid cell fates. Protein Cell 8, 242–254 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Rankin, L. C. et al. Complementarity and redundancy of IL-22-producing innate lymphoid cells. Nat. Immunol. 17, 179–186 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Zheng, Y. et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 14, 282–289 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Goto, Y. et al. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science 345, 1254009 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Yu, Y. et al. Single-cell RNA-seq identifies a PD-1hi ILC progenitor and defines its development pathway. Nature 539, 102–106 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Taylor, S. et al. PD-1 regulates KLRG1+ group 2 innate lymphoid cells. J. Exp. Med. 214, 1663–1678 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pentcheva-Hoang, T., Chen, L., Pardoll, D. M. & Allison, J. P. Programmed death-1 concentration at the immunological synapse is determined by ligand affinity and availability. Proc. Natl Acad. Sci. USA 104, 17765–17770 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Michalek, R. D. et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J. Immunol. 186, 3299–3303 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Patel, C. H., Leone, R. D., Horton, M. R. & Powell, J. D. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat. Rev. Drug Discov. 18, 669–688 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Wilhelm, C. et al. Critical role of fatty acid metabolism in ILC2-mediated barrier protection during malnutrition and helminth infection. J. Exp. Med. 213, 1409–1418 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Karagiannis, F. et al. Lipid-droplet formation drives pathogenic group 2 innate lymphoid cells in airway inflammation. Immunity 52, 620–634.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Helou, D. G. et al. PD-1 pathway regulates ILC2 metabolism and PD-1 agonist treatment ameliorates airway hyperreactivity. Nat. Commun. 11, 3998 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Patsoukis, N. et al. PD-1 alters T-cell metabolic reprogramming by inhibiting and promoting lipolysis and fatty acid oxidation. Nat. Commun. 6, 6692 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Kamada, N., Seo, S.-U., Chen, G. Y. & Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 13, 321–335 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, T. et al. Probiotics Lactobacillus reuteri abrogates immune checkpoint blockade-associated colitis by inhibiting group 3 innate lymphoid cells. Front. Immunol. 10, 1235 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Song, M.-Y. et al. Protective effects of Fc-fused PD-L1 on two different animal models of colitis. Gut 64, 260–271 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Saleh, M. & Trinchieri, G. Innate immune mechanisms of colitis and colitis-associated colorectal cancer. Nat. Rev. Immunol. 11, 9–20 (2011).

    Article  CAS  PubMed  Google Scholar 

  39. Sugimoto, K. et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J. Clin. Invest. 118, 534–544 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sawa, S. et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat. Immunol. 12, 320–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Mao, K. et al. Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid metabolism. Nature 554, 255–259 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Rath, H. C. et al. Different subsets of enteric bacteria induce and perpetuate experimental colitis in rats and mice. Infect. Immun. 69, 2277–2285 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wlodarska, M. et al. Antibiotic treatment alters the colonic mucus layer and predisposes the host to exacerbated Citrobacter rodentium-induced colitis. Infect. Immun. 79, 1536–1545 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Di Luccia, B., Gilfillan, S., Cella, M., Colonna, M. & Huang, S. C.-C. ILC3s integrate glycolysis and mitochondrial production of reactive oxygen species to fulfill activation demands. J. Exp. Med. 216, 2231–2241 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, X. et al. Interleukin-22 alleviates metabolic disorders and restores mucosal immunity in diabetes. Nature 514, 237–241 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Guo, X. et al. Induction of innate lymphoid cell-derived interleukin-22 by the transcription factor STAT3 mediates protection against intestinal infection. Immunity 40, 25–39 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Routy, B. et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Seidel, J. A., Otsuka, A. & Kabashima, K. Treating tumors with immune checkpoint inhibitors: rationale and limitations. Trends Immunother. 1, 2–9 (2017).

    Article  Google Scholar 

  50. Zhong, C. et al. Group 3 innate lymphoid cells continuously require the transcription factor GATA-3 after commitment. Nat. Immunol. 17, 169–178 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Fuping You of Peking University (Beijing, China) for technique help. We thank Xiaohuan Guo of Tsinghua University (Beijing, China) for providing the C. rodentium strain and for assistance with the mixed bone marrow chimera experiments. We thank Huan Wang of Peking University for assistance with Seahorse assays. We thank all of the staff of the animal department for their help. This work is supported by the National Natural Science Foundation of China (no. 31770957, no. 91842102 and no. 32170896), and the Natural Science Foundation of Beijing (no. 18G10645).

Author information

Authors and Affiliations

Authors

Contributions

C.Z. conceived the project. D.W. performed all of the experiments. L.H. and Yime Zhang. helped with bioinformatic analyses. M.H. performed the scRNA-seq. Y.D. helped with cell sorting. G.R., X.Z., Z.L. P.L., Yinlian Zhang, S.C., J.L., Y.S. and P.W. helped in some experiments and data analysis. J.X. generated the critical mice tool. C.Z. and D.W. wrote the manuscript. C.Z. supervised the project.

Corresponding author

Correspondence to Chao Zhong.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Christoph Wilhelm and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary handling editor: Alfredo Gimenez-Cassina, in collaboration with the Nature Metabolism team.

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 PD-1 deficiency results in disturbed microbiota and aggravated colitis.

a, Differences in fecal taxa in Pdcd1+/+ and Pdcd1-/- mice were shown in taxonomic cladogram from linear discriminant analysis of effect size (LEfSe) (green, most abundant in Pdcd1+/+ mice; red, most abundant in Pdcd1-/- mice). b, Proportions of IgA-coating fecal bacteria from Pdcd1-/- mice and Pdcd1+/- littermates were examined by flow cytometry (n = 5 per group; *p = 0.0467). c, gut barrier integrity in Pdcd1+/+ and Pdcd1-/- mice were assessed by FITC-dextran 4 (FD4, molecular mass, 4 kDa) gavage at 10ml/kg. FD4 in mice serum was examined after 4 hours by fluorescence intensity (n = 5 per group; ns). d, Colitis was induced in Pdcd1-/- mice and Pdcd1+/- littermates by 3% DSS. Body weight change was examined every day (n = 5 per group; *p = 0.0484). e, On day 7 of DSS-induced colitis, mice were sacrificed and colon length was measured (n = 5 per group; **p = 0.0029). Data are representative of two independent experiment (a) or at least three independent experiments (b-e). Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (b, c, e) and two-sided paired t-test (d). Ns, not significant, *p < 0.05, **p < 0.01.

Source data

Extended Data Fig. 2 PD-1 expression on colonic LTi is upregulated during activation.

a, PD-1 expression on colonic ILCs, γδT, NKT, monocytes, and DC was examined by flow cytometry. Percentages of PD-1+ cells and their PD-1 MFI were calculated (n = 4 per group; ns, ns, ***p < 0.0001, ***p < 0.0001; ***p = 0.0008, ***p < 0.0001, ***p < 0.0001, ***p < 0.0001). b, Gating strategy for ILC subsets, including LTi, T-bet-(CCR6-) ILC3, T-bet+(NKp46-) ILC3, NKp46+ ILC3, ILC1/cNK, and ILC2. c, On day 7 of DSS-induced colitis, proportion of IL-22-producing lymphocytes in colon was analyzed (mean ± s.d.; n = 6). d, PD-1 expression between colonic LTi of B6 (Rag2+/+) and Rag2-/- mice were compared. Proportion of PD-1+ LTi and their PD-1 MFI were calculated (n = 5 per group; ***p < 0.0001, **p = 0.0027). e, IL-22 production by colonic LTi of mice in steady state and DSS-induced colitis (on day 7) was examined. Proportion of IL-22+ LTi and their IL-22 MFI were compared (n = 4 per group; **p = 0.0028, *p = 0.0189). f, On day 7 of DSS-induced colitis, forward and side scatters (FSC-A and SSC-A) of PD-1high and PD-1- colonic LTi were examined. Mean values of FSC-A and SSC-A were compared (n = 4 per group; ***p = 0.0001, ***p < 0.0001). g, On day 7 of DSS-induced colitis, IL-17A expression in PD-1high and PD-1- colonic LTi was examined. Proportion, IL-17A MFI and number of IL-17A+ LTi were compared (n = 5 per group; ***p = 0.0008, *p = 0.0454, **p = 0.0015). h, PD-1 expression on in vitro cultured LTi treated with heat-killed bacteria or PBS (left), or bacterial culture supernatant or culture medium (right) was examined. i, 7 days post C. rodentium infection, PD-1 expression on colonic LTi was examined. Proportion of PD-1+ LTi and their PD-1 MFI were calculated (n = 4 per group; ns, ns). Data are representative of at least three independent experiments. Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (a, d-g, i). Ns, not significant, *p < 0.05, **p < 0.01, ***p < 0.001.

Source data

Extended Data Fig. 3 Impact of PD-1 deficiency on colonic LTi activation.

a, Percentage and number of colonic LTi in Pdcd1+/+ and Pdcd1-/- mice were calculated (n = 4 per group; ns, ns). b, On day 7 of DSS-induced colitis, proportion, IL-22 MFI and number of IL-22+ colonic LTi in Pdcd1-/- and Pdcd1+/- (littermate) mice were analyzed (n = 5 per group; ***p = 0.0006, *p = 0.0489, *p = 0.0425). c, On day 3 of DSS-induced colitis, proportion, IL-17A MFI and number of IL-17A+ colonic LTi in Pdcd1+/+Rag2-/- and Pdcd1-/-Rag2-/- mice were analyzed (n = 5 per group; ns, ns, ns). (d-h) Pdcd1+/+Rag2-/- and Pdcd1-/-Rag2-/- mice were subjected to C. rodentium infection. d, Body weight was measured daily (n = 5 per group; ***p < 0.0001). Colon length (e) (n = 5 per group; *p = 0.0436) and H&E staining indicated tissue damage (f) was assessed on day 3, g, The bacteria burdens in feces, spleen, and liver were assessed on day 3 and 7 (n = 5 per group; *p = 0.0235, *p = 0.0389; *p = 0.0242, *p = 0.0142; *p = 0.0104, *p = 0.0492). CFU, colony-forming unit. h, On day 3, proportion, IL-22 MFI and number of IL-22+ colonic LTi were analyzed (n = 5 per group; *p = 0.0182, **p = 0.0029, *p = 0.0331). i, Under different stimulations, proportion and IL-22 MFI of IL-22+ siLP LTi in Pdcd1+/+Rag2-/- and Pdcd1-/-Rag2-/- mice were examined (n = 4 per group; ***p < 0.0001, ***p < 0.0001, ***p < 0.0001; *p = 0.0267, *p = 0.0423, ***p = 0.0001). P/I, PMA+ionomycin. j, Construction strategy for Pdcd1h/h mice. k, On day 7 of DSS-induced colitis, proportion and hPD-1 MFI of hPD-1+ colonic LTi in Pdcd1h/+ and Pdcd1h/- mice were assessed (n = 4 per group; ns, ns). l, PD-1 expression on lineage-RORγt+ siLP lymphocytes of Pdcd1fl/-Rorc-Cre and Pdcd1fl/- (littermate) mice were compared. Data are representative of at least three independent experiments. Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (a-c, e, g-i, k) and two-sided paired t-test (d). Ns, not significant, *p < 0.05, **p < 0.01, ***p < 0.001.

Source data

Extended Data Fig. 4 PD-1 signaling represses STAT3 phosphorylation in activated LTi.

a, Sorted LTi were stimulated with IL-23 (0.5 ng/ml) for 25 min, in presence or absence of rPD-L1 (5 µg/ml). Afterwards, STAT3 phosphorylation was examined by flow cytometry. b, On day 7 of DSS-induced colitis, colonic lymphocytes were collected from Pdcd1+/+ and Pdcd1-/- mice. STAT3 phosphorylation (pSTAT3) in LTi was compared by flow cytometry. Isotype antibody staining was used as negative control. MFI of pSTAT3 was calculated (n = 5 per group; ns). Data are representative of at least three independent experiments. Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (b). Ns, not significant.

Source data

Extended Data Fig. 5 scRNA-seq reveals metabolic reprogramming during ILC3 activation.

a, Lineage- CD127+ siLP ILCs sorted from B6 mice were subjected to single-cell RNA-sequencing. The cells were clustered into three subsets, ILC1, ILC2, and ILC3 (left), based on their Rorc, Gata3, and Ncr1 expression (right). b, Scorings of LTi (top) and Nkp46+ ILC3 (bottom) based on the ILC3 activation module was correlated with their Il22 expression in scatter plot. Pearson correlation was used to indicate the strength of linear relationship (95% confidence intervals by default). c, Differentially expressed genes in inactivated NKp46+ ILC3 and activated NKp46+ ILC3 clusters were shown with heatmap. d, Signature metabolic genes in inactivated NKp46+ ILC3 and activated NKp46+ ILC3 clusters were shown in violin plot. e, Enrichment of classical metabolic pathways (including carbohydrate metabolism, lipid metabolism, and respiration) in inactivated NKp46+ ILC3 and activated NKp46+ ILC3 was shown in radar chart. f, Respective enrichment of carbohydrate metabolism, lipid metabolism, and respiration pathways in inactivated NKp46+ ILC3 and activated NKp46+ ILC3, using the differentially expressed genes in (c), was compared in density plots. g, Enrichment of fatty acid oxidation and lipid biosynthesis in inactivated NKp46+ ILC3 and activated NKp46+ ILC3, using the differentially expressed genes in (c), was compared with density plot. ND, not detected, in the differentially expressed genes in (c).

Extended Data Fig. 6 Excessive FAO suppresses ILC3 activation.

a, Representative Seahorse palmitate-FAO assay result showed the impact of palmitate (PA) on FAO of LTi (n = 5 per group). b, Basal OCR, ATP-linked OCR, maximal OCR and SRC were quantified (n = 3 per group; **p = 0.0097, **p = 0.0082, *p = 0.0163, *p = 0.0462). c, After a 3-day PA (20 μM) or vehicle treatment, IL-22 expression in activated LTi was assessed . Proportion of IL-22+ LTi and their IL-22 MFI were calculated (n = 4 per group; **p = 0.001, *p = 0.0117). d, Viability of LTi after PA (100 μM) treatment was assessed (n = 5 per group; ns). e, CPT1A overexpression in LTi was measured by qRT-PCR (n = 5 per group; *p = 0.0297). f, Impact of CPT1A on IL-22 production in LTi were assessed. Proportion of IL-22+ LTi and their IL-22 MFI were calculated (n = 5 per group; *p = 0.0436, ***p < 0.0001). g, After a 3-day PA or vehicle treatment, IL-22 production in activated NKp46+ ILC3 was assessed. Proportion of IL-22+ NKp46+ ILC3 and their IL-22 MFI were calculated (n = 5 per group; ***p < 0.0001, ***p < 0.0001). (h and i) Rag2-/- mice were fed with HFD (high fat diet) or CD (chow diet) for 5 weeks. IL-22 expression in siLP NKp46+ ILC3 (h) and colonic LTi (i) was examined. Proportion of IL-22+ siLP NKp46+ ILC3 and their IL-22 MFI (n = 4 per group; *p = 0.0269, *p = 0.0151)(h), and proportion, IL-22 MFI and number of IL-22+ colonic LTi (n = 4 per group; *p = 0.0298, *p = 0.0477, *p = 0.0496)(i), were calculated. (j and k) Enriched features in PA untreated (j) and treated (k) LTi was analyzed using two-sided GSEA adopted permutation-test. Nominal p-value and NES were calculated. Data are representative of at least three independent experiments (a-i) or two independent experiments (j-k). Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (b-i). Ns, not significant, *p < 0.05, **p < 0.01, ***p < 0.001.

Source data

Extended Data Fig. 7 FAO inhibition restores IL-22 production in PD-1 deficient LTi.

a, Expression of CPT1A in colonic LTi from wild-type (Pdcd1+/+) and Pdcd1-/- mice in steady state was examined by flow cytometry. MFI of CPT1A was calculated (n = 4 per group; ns). b, FAO inhibitor Oxfenicine or vehicle (H2O) was added to siLP LTi sorted from Pdcd1+/+Rag2-/- and Pdcd1-/-Rag2-/- mice. After 4 days, the cells were harvested and stimulated by IL-23 (0.5 ng/ml) for 3 hours. Afterwards, IL-22 secretion was assessed by intracellular cytokine staining. c, Proportion and IL-22 MFI of IL-22+ LTi in (b) were calculated (n = 4 per group; **p = 0.0033, ns; **p = 0.0058, ns). d, Wild-type and Pdcd1-/- mice were injected (i.p.) with Oxfenicine or H2O during DSS-induced colitis. On day 7, mice were sacrificed, and IL-22 production in colonic LTi was examined by intracellular cytokine staining. Proportion, IL-22 MFI and cell number of IL-22+ LTi were calculated (n = 4 per group; **p = 0.0044, *p = 0.0119; **p = 0.0095, ***p = 0.0005; **p = 0.0033, ns). e, Cpt1a knockdown in LTi cells was assessed by qRT-PCR (n = 5 per group; ***p = 0.0009). f, IL-22 production by shScram (scramble) or shCpt1a lentivirus infected LTi was assessed by intracellular cytokine staining. Proportion and IL-22 MFI of IL-22+ LTi were calculated (n = 5 per group; **p = 0.0027, *p = 0.0213). Data are representative of at least three independent experiments. Data are presented as the mean ± s.d., and statistical significance was determined by two-sided unpaired t-test (a, c-f). Ns, not significant, *p < 0.05, **p < 0.01, ***p < 0.001.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

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. 6

Statistical Source Data

Source Data Fig. 7

Statistical Source Data

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. 6

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, D., Hu, L., Han, M. et al. PD-1 signaling facilitates activation of lymphoid tissue inducer cells by restraining fatty acid oxidation. Nat Metab 4, 867–882 (2022). https://doi.org/10.1038/s42255-022-00595-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00595-9

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