Abstract
Interleukin-17 (IL-17)-producing γδ T (Tγδ17) cells are innate-like mediators of intestinal barrier immunity. Although IL-17-producing helper T cell and group 3 innate lymphoid cell plasticity have been extensively studied, the mechanisms governing Tγδ17 cell effector flexibility remain undefined. Here, we combined type 3 fate mapping with single-cell ATAC-sequencing/RNA-sequencing multiome profiling to define the cellular features and regulatory networks underlying Tγδ17 cell plasticity. During homeostasis, Tγδ17 cell effector identity was stable across tissues, including for intestinal T-bet+ Tγδ17 cells that restrained interferon-γ production. However, Salmonella enterica subsp. enterica serovar Typhimurium infection induced intestinal Vγ6+ Tγδ17 cell conversion into type 1 effectors, with loss of IL-17A production and partial RORγt downregulation. Multiome analysis revealed a trajectory along Vγ6+ Tγδ17 cell effector conversion, with TIM-3 marking ex-Tγδ17 cells with enhanced type 1 functionality. Last, we characterized and validated a critical AP-1 regulatory axis centered around JUNB and FOSL2 that controls Vγ6+ Tγδ17 cell plasticity by stabilizing type 3 identity and restricting type 1 effector conversion.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
All sequencing data that support the findings of this study have been deposited in Gene Expression Omnibus under primary accession code GSE290581. All other data that support the findings of this study are available from the corresponding author upon request.
References
Vantourout, P. & Hayday, A. Six-of-the-best: unique contributions of γδ T cells to immunology. Nat. Rev. Immunol. 13, 88–100 (2013).
Hayday, A. C. γδ T cell update: adaptate orchestrators of immune surveillance. J. Immunol. 203, 311–320 (2019).
Parker, M. E. & Ciofani, M. Regulation of γδ T cell effector diversification in the thymus. Front. Immunol. 11, 42 (2020).
Muñoz-Ruiz, M., Sumaria, N., Pennington, D. J. & Silva-Santos, B. Thymic determinants of γδ T cell differentiation. Trends Immunol. 38, 336–344 (2017).
Heilig, J. S. & Tonegawa, S. Diversity of murine γ genes and expression in fetal and adult T lymphocytes. Nature 322, 836–840 (1986).
Haas, J. D. et al. Development of interleukin-17-producing γδ T cells is restricted to a functional embryonic wave. Immunity 37, 48–59 (2012).
Chien, Y. H., Zeng, X. & Prinz, I. The natural and the inducible: interleukin (IL)-17-producing γδ T cells. Trends Immunol. 34, 151–154 (2013).
Shih, H. Y. et al. Developmental acquisition of regulomes underlies innate lymphoid cell functionality. Cell 165, 1120–1133 (2016).
Lee, M. et al. Single-cell RNA sequencing identifies shared differentiation paths of mouse thymic innate T cells. Nat. Commun. 11, 4367 (2020).
Schmolka, N. et al. Epigenetic and transcriptional signatures of stable versus plastic differentiation of proinflammatory γδ T cell subsets. Nat. Immunol. 14, 1093–1100 (2013).
Mukasa, R. et al. Epigenetic instability of cytokine and transcription factor gene loci underlies plasticity of the T helper 17 cell lineage. Immunity 32, 616–627 (2010).
Morrison, P. J. et al. TH17-cell plasticity in Helicobacter hepaticus-induced intestinal inflammation. Mucosal Immunol. 6, 1143–1156 (2013).
Vonarbourg, C. et al. Regulated expression of nuclear receptor RORγt confers distinct functional fates to NK cell receptor-expressing RORγt+ innate lymphocytes. Immunity 33, 736–751 (2010).
Lee, Y. K. et al. Late developmental plasticity in the T helper 17 lineage. Immunity 30, 92–107 (2009).
Yen, H.-R. et al. Tc17 CD8 T cells: functional plasticity and subset diversity. J. Immunol. 183, 7161–7168 (2009).
Hirota, K. et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011).
Mills, K. H. G. IL-17 and IL-17-producing cells in protection versus pathology. Nat. Rev. Immunol. 23, 38–54 (2023).
Barros-Martins, J. et al. Effector γδ T cell differentiation relies on master but not auxiliary TH cell transcription factors. J. Immunol. 196, 3642–3652 (2016).
Sheridan, B. S. et al. γδT cells exhibit multifunctional and protective memory in intestinal tissues. Immunity 39, 184–195 (2013).
Kadekar, D. et al. The neonatal microenvironment programs innate γδ T cells through the transcription factor STAT5. J. Clin. Invest. 130, 2496–2508 (2020).
Schmolka, N. et al. MicroRNA-146a controls functional plasticity in γδ T cells by targeting NOD1. Sci. Immunol. 3, eaao1392 (2018).
Reis, B. S. et al. TCR-Vγδ usage distinguishes protumor from antitumor intestinal γδ T cell subsets. Science 377, 276–284 (2022).
Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).
Eberl, G. & Littman, D. R. Thymic origin of intestinal αβ T cells revealed by fate mapping of RORγt+ cells. Science 305, 248–251 (2004).
VanCott, J. L. et al. Regulation of host immune responses by modification of Salmonella virulence genes. Nat. Med. 4, 1247–1252 (1998).
Omenetti, S. et al. The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory TH17 cells. Immunity 51, 77–89 (2019).
Zhong, C. et al. Group 3 innate lymphoid cells continuously require the transcription factor GATA-3 after commitment. Nat. Immunol. 17, 169–178 (2016).
Tan, L. et al. Single-cell transcriptomics identifies the adaptation of Scart1+ Vγ6+ T cells to skin residency as activated effector cells. Cell Rep. 27, 3657–3671 (2019).
La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).
Gao, M., Qiao, C. & Huang, Y. UniTVelo: temporally unified RNA velocity reinforces single-cell trajectory inference. Nat. Commun. 13, 6586 (2022).
Edwards, S. C. et al. PD-1 and TIM-3 differentially regulate subsets of mouse IL-17A–producing γδ T cells. J. Exp. Med. 220, e20211431 (2023).
Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).
Grenningloh, R., Kang, B. Y. & Ho, I.-C. Ets-1, a functional cofactor of T-bet, is essential for TH1 inflammatory responses. J. Exp. Med. 201, 615–626 (2005).
Wahlen, S. et al. The transcription factor RUNX2 drives the generation of human NK cells and promotes tissue residency. eLife 11, e80320 (2022).
Thieu, V. T. et al. Signal transducer and activator of transcription 4 is required for the transcription factor T-bet to promote T helper 1 cell-fate determination. Immunity 29, 679–690 (2008).
Michel, M. L. et al. Interleukin 7 (IL-7) selectively promotes mouse and human IL-17-producing γδ cells. Proc. Natl Acad. Sci. USA 109, 17549–17554 (2012).
Agerholm, R., Rizk, J., Viñals, M. T. & Bekiaris, V. STAT3 but not STAT4 is critical for γδT17 cell responses and skin inflammation. EMBO Rep. 20, e48647 (2019).
Mikami, Y. et al. NCR+ ILC3 maintain larger STAT4 reservoir via T-BET to regulate type 1 features upon IL-23 stimulation in mice. Eur. J. Immunol. 48, 1174–1180 (2018).
Smith, M. et al. Tissue-specific transgenic knockdown of Fos-related antigen 2 (Fra-2) expression mediated by dominant negative Fra-2. Mol. Cell. Biol. 21, 3704–3713 (2001).
Carr, T. M., Wheaton, J. D., Houtz, G. M. & Ciofani, M. JUNB promotes TH17 cell identity and restrains alternative CD4+ T-cell programs during inflammation. Nat. Commun. 8, 301 (2017).
Ciofani, M. et al. A validated regulatory network for TH17 cell specification. Cell 151, 289–303 (2012).
Roychoudhuri, R. et al. BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 17, 851–860 (2016).
Thakore, P. I. et al. BACH2 regulates diversification of regulatory and proinflammatory chromatin states in TH17 cells. Nat. Immunol. 25, 1395–1410 (2024).
Shetty, A. et al. Interactome networks of FOSL1 and FOSL2 in human TH17 cells. ACS Omega 6, 24834–24847 (2021).
Roychoudhuri, R. et al. BACH2 represses effector programs to stabilize Treg-mediated immune homeostasis. Nature 498, 506–510 (2013).
Hasan, Z. et al. JUNB is essential for IL-23-dependent pathogenicity of TH17 cells. Nat. Commun. 8, 15628 (2017).
Klose, C. S. et al. A T-bet gradient controls the fate and function of CCR6–RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).
Parker, M. E. et al. c-MAF regulates the plasticity of group 3 innate lymphoid cells by restraining the type 1 program. J. Exp. Med. 217, e20191030 (2020).
Wang, Y. et al. The transcription factors T-bet and Runx are required for the ontogeny of pathogenic interferon-γ-producing T helper 17 cells. Immunity 40, 355–366 (2014).
Lazarevic, V. et al. T-bet represses TH17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt. Nat. Immunol. 12, 96–104 (2011).
Schraml, B. U. et al. The AP-1 transcription factor Batf controls TH17 differentiation. Nature 460, 405–409 (2009).
Glasmacher, E. et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1–IRF complexes. Science 338, 975–980 (2012).
Li, P. et al. BATF–JUN is critical for IRF4-mediated transcription in T cells. Nature 490, 543–546 (2012).
Aydintug, M. K., Roark, C. L., Chain, J. L., Born, W. K. & O’Brien, R. L. Macrophages express multiple ligands for γδ TCRs. Mol. Immunol. 45, 3253–3263 (2008).
Khairallah, C. et al. A blend of broadly-reactive and pathogen-selected Vγ4 Vδ1 T cell receptors confer broad bacterial reactivity of resident memory γδ T cells. Mucosal Immunol. 15, 176–187 (2021).
Kenner, L. et al. Mice lacking JUNB are osteopenic due to cell-autonomous osteoblast and osteoclast defects. J. Cell Biol. 164, 613–623 (2004).
Karreth, F., Hoebertz, A., Scheuch, H., Eferl, R. & Wagner, E. F. The AP1 transcription factor Fra2 is required for efficient cartilage development. Development 131, 5717–5725 (2004).
Wende, H. et al. The transcription factor c-MAF controls touch receptor development and function. Science 335, 1373–1376 (2012).
Kometani, K. et al. Repression of the transcription factor BACH2 contributes to predisposition of IgG1 memory B cells toward plasma cell differentiation. Immunity 39, 136–147 (2013).
Hatano, S. et al. Development of a new monoclonal antibody specific to mouse Vγ6 chain. Life Sci. Alliance 2, e201900363 (2019).
McKenzie, D. R. et al. IL-17-producing γδ T cells switch migratory patterns between resting and activated states. Nat. Commun. 8, 15632 (2017).
Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther. 7, 1063–1066 (2000).
Becattini, S. et al. Enhancing mucosal immunity by transient microbiota depletion. Nat. Commun. 11, 4475 (2020).
Schauer, D. B. et al. Genetic and biochemical characterization of Citrobacter rodentium sp. nov. J. Clin. Microbiol. 33, 2064–2068 (1995).
Satpathy, A. T. et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol. 37, 925–936 (2019).
Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).
Alquicira-Hernandez, J. & Powell, J. E. Nebulosa recovers single-cell gene expression signals by kernel density estimation. Bioinformatics 37, 2485–2487 (2021).
Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).
Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).
Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).
Ji, Z. & Ji, H. TSCAN: pseudo-time reconstruction and evaluation in single-cell RNA-seq analysis. Nucleic Acids Res. 44, e117 (2016).
Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. A comparison of single-cell trajectory inference methods. Nat. Biotechnol. 37, 547–554 (2019).
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
Chen, Z. et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity 51, 840–855 (2019).
Castro-Mondragon, J. A. et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 50, D165–D173 (2022).
Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).
Huynh-Thu, V. A., Irrthum, A., Wehenkel, L. & Geurts, P. Inferring regulatory networks from expression data using tree-based methods. PLoS ONE 5, e12776 (2010).
Skene, P. J., Henikoff, J. G. & Henikoff, S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat. Protoc. 13, 1006–1019 (2018).
Acknowledgements
We thank E. Park for assistance with gut preps. We also thank the Molecular Genomics Core for processing and running the 10x Genomics Multiome samples. We acknowledge the expert assistance of L. Martinek with flow cytometry sorting and S. Langdon at the Duke DNA Sequencing Facility. We also acknowledge the use of the Duke Compute Cluster. Last, we thank Y. Yoshikai (Kyushu University) for sharing the antibody to Vγ6. This work was funded by NIH R01 GM115474 and P01 AI102853 grants to M.C. M.E.P. was supported by F31 AI152457 and a Duke Training Grant in Digestive Diseases and Nutrition (5T32DK007568). N.U.M. was supported by F31 AI181082.
Author information
Authors and Affiliations
Contributions
M.E.P. and N.U.M. designed, performed and analyzed the experiments. T.-C.L. performed computational analysis for single-cell multiome experiments. W.H.T. and M.E.P. performed initial computational analyses for single-cell multiome experiments. T.-C.L. and J.B. performed the CUT&RUN analysis. J.B. and M.E.P performed the C. rodentium infection experiments. S.A.S. maintained the mouse colony and performed infections. M.E.P., N.U.M. and M.C. wrote the manuscript. M.C. conceived the study and designed, supervised and analyzed the experiments.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Venuprasad Poojary, Bruno Silva-Santos and the other, anonymous, reviewer(s) 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 Tγδ17 cells are stable at steady state and Vγ6+ Tγδ17 cells are plastic after S. typhimurium in mLN and coLP.
(a–i, k–n) Flow cytometric analysis across multiple tissues of fate-mapping mice. (a) RORγt, IL-17A, and ZsGreen expression in total γδ T cells from Rorc-Cre R26ZSG and Il17aCre R26ZSG mice. Representative of 3 experiments, n = 11. (b) Vγ6, Vγ4, and ZsGreen expression in total (left, top right) and ZsGreen+ (bottom right) γδ T cells from Rorc-Cre R26ZSG mice from 2 experiments, n = 10. (c) γδ T cells (CD3+γδTCR+TCRβ−) from tissues of Il17aCre R26TOM IFNγ-YFP mice at steady state; positive IFNγ-YFP control from coLP 4 days post-S. typhimurium (STm). Two experiments: n = 3 (FRT), 5 (siLP, coLP, lung, mLN, iLN). (d) RORγt and T-bet expression in γδ T cells from Il17aCre R26ZSG mice. 2 experiments, n = 6. (e) IL-17A and IFNγ in ZsGreen+ γδ T cells from coLP of naïve (n = 11) and STm (n = 12) infected Il17aCre R26ZSG mice; three experiments. (f) IL-17A−IFNγ+ ZsGreen− γδ T cells from coLP. n = 9 naïve, 10 STm; two experiments. (g) Frequency of IL-17A+IFNγ−, IL-17A+IFNγ+, IL-17A−IFNγ+ Vγ4+ ZsGreen+ γδ T cells from coLP. n = 9 naïve, 10 STm; two experiments. (h) Cytokine production by ZsGreen+ γδ T cells from mLN. Three experiments, n > 10. (i) Vγ4 or Vγ6 versus IFNγ-YFP in TOM+ γδ T cells from coLP of naïve and STm infected mice from 3 experiments, n = 10. (j) Splenic CFUs post-aroA− STm infection. n = 4/timepoint; one experiment. (k) IFNγ+ Vγ6+ ZsGreen+ γδ T cells after actA− L. monocytogenes (LmOva actA-) infection. n = 2-4/timepoint; one experiment. (l) IL-17A and IFNγ in CD4+ ZsGreen+ and Vγ6+ γδ T cells 15 days post-C. rodentium. Two experiments, summary from one, naïve (n = 4), Cr (n = 6). (m) Normalized RORγt in Vγ6+ ZsGreen+ γδ T cells from naïve (n = 11) and STm (n = 15) infected mice; three experiments. (n) IL-22 in CD4+ ZsGreen+ and Vγ6+ γδ T cells post-aroA− STm infection. n = 4/timepoint; one experiment. Cytokine expression in (a, e–h, k, l, n) measured after 4 h PMA/ionomycin stimulation. All results represent mean ± s.e.m. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant (two-tailed unpaired Student’s t-test). Numbers in flow plots represent percentages of cells in the gate.
Extended Data Fig. 2 Single cell multiome characterization of γδ T cells and Vγ6+ Tγδ17 cell trajectories.
(a) Bar plot of number of cells in each cluster from each condition for total γδ T cells. (b, c) Violin plots of type 3 and 1 genes for γδ T cells clusters. (d) Volcano plot for cells from naïve condition for C0 vs C6 Vγ6+ Tγδ17 cells, with blue dots indicating significance based on p-val adj < 0.05 and log2FC > 0.25; FC, fold change. (e) zFP506 (ZsGreen) Feature plot for γδ T cell clusters. (f) Plot of per cell unspliced versus spliced Rorc transcript, RNA velocity for Rorc, and Rorc expression feature plot all for Vγ6+ Tγδ17 clusters C0, C6, C9 and C7. (g) Monocle 3 trajectory of Vγ6+ Tγδ17 clusters C0, C6, C9 and C7. (h) CD8+ T cell gene signatures (GSE9650) projected onto UMAP space with Seurat’s AddModuleScore function. Nebulosa Density plot displaying enrichment of gene signature. (Left) Genes upregulated in naive vs effector CD8+ T cells. (Right) Genes downregulated in naive vs effector CD8+ T cells. (i) Euclidian distances between γδ T cell clusters based on WNN UMAP. Seurat MAST test (GLM framework) was used for differential expression in d.
Extended Data Fig. 3 Effector-converted Vγ6+ Tγδ17 cells have distinct transcriptional profiles compared to steady state.
(a) IFNγ-YFP expression in fate-mapped TIM-3− or TIM-3+ Vγ6+ TOM+ γδ T cells from naïve and S. typhimurium-infected Il17aCre R26TOM IFNγ-YFP mice. Summary plot pooled from two experiments, n = 8 mice. (b, c) Flow cytometric analysis of colonic Vγ6+ γδ T cells at 4 days post S. typhimurium infection, following daily intraperitoneal injections of either an isotype control or anti-TIM-3 antibody: (b) Fate-mapped Vγ6+ ZS+ γδ T cells from Rorc-Cre R26ZSG mice. Summary data include IFNγ expression after 4 h PMA/ionomycin stimulation and the absolute number of unstimulated Vγ6+ ZS+ γδ T cells (one experiment; n = 5 per condition). (c) Fate-mapped Vγ6+ TOM+ γδ T cells from Il17aCre R26TOM IFNγ-YFP mice. Data show IFNγ-YFP expression, Ki-67 expression, and the total number of Vγ6+ TOM+ γδ T cells (one experiment; n = 5 per condition). (d) Summary data for PD-1 gMFI in PD-1+TIM-3− or PD-1+TIM-3+ Vγ6+ ZS+ γδ T cells from three experiments, n = 13 mice. (e) Volcano plot for differentially expressed genes between C7 vs C9 Vγ6+ Tγδ17 cells with blue dots having p-val adj < 0.05 and log2FC > 0.25. (f) Volcano plot of differentially expressed transcription factors in type 1 converting Vγ6+ γδ T cell clusters (C7 + C9) compared to type 3 steady state clusters (C0 + C6) with red dots having p-val adj < 0.05 and log2FC > 0.25; FC, fold change. (g) Dot plot for Tγδ17 cell clusters for select transcriptional regulators downregulated (left) or upregulated (right) in Vγ6+ Tγδ17 cells with Vγ4+ Tγδ17 cells for comparison. All results represent mean ± s.e.m. Two-tailed paired t-test for a, d, two-tailed unpaired t-test for b, c, and Seurat MAST test (GLM framework) was used for differential expression in e, f. ***P < 0.001; ****P < 0.0001; ns, not significant (two-tailed paired Student’s t-test). Numbers in flow plots represent percentages of cells in the gate.
Extended Data Fig. 4 BACH2 and AP-1 TFs regulate Vγ6+ Tγδ17 plasticity in vitro.
(a) Schematic of Vγ6+ Tγδ17 cluster 0, 6, 9, and 7 for number of regions differentially accessible (DA) (p < 0.05) with regions increasing (UP, red) and decreasing (DOWN, blue) in accessibility. (b) Pseudobulk scATAC-seq CoveragePlots for Rorc and Ifng loci for Vγ6+ Tγδ17 cluster 0, 6, 9, and 7. Rectangle highlights regions with significant differential accessibility (p < 0.05) shown for decreasing (orange) or increasing (blue) in accessibility. Asterisks indicate JunB CUT&RUN binding in Vγ6+ Tγδ17 cells. (c) Motif activity dot plot of Vγ6+ Tγδ17 clusters using chromVAR with colored boxes highlighting specific TF families. (d) TFs in Vγ6+ Tγδ17 cell overexpression screen. X’s in RNA DEG column means the TF of interest is a DEG at some point along trajectory. X in Regulon column means the TF is significant in regulon analysis. X in Motif Activity column means TF has differential motif activity (chromVAR) during conversion. X in Literature column means TF is implicated in type 3 lymphocyte regulation. Blue TFs predicted to stabilize type 3 program and green TFs predicted to promote type 1 conversion. (e) Flow cytometric analysis of cytokine production from day 9 Tγδ17 mLN culture. Gated on transduced Vγ6+ (Vγ4−) Thy1.1+ ZS+ γδ T cells from steady state Il17aCreR26ZSG mice after 4 h PMA/Ionomycin stimulation. Summary graph pooled from two independent experiments. (f) Same as in e but from S. typhimurium infected Il17aCreR26ZSG mice. Summary graph from one independent experiment. For e, all conditions have n = 3 except SMAD3, LEF1, HIF1a, and ETV6, which have n = 2; for f, all conditions have n = 2. Statistical analyses included the LR framework test with Signac for differential accessibility calling in a, b, and chromVAR z-score based deviation test in c, and ordinary one-way ANOVA tests for e, f. Results represent mean ± s.e.m. *P < 0.05; **P < 0.01; ****P < 0.0001; DEG, differentially expressed gene; ns, not significant.
Extended Data Fig. 5 JunB plays a more prominent role than Fosl2 in Vγ6+ Tγδ17 cell plasticity.
(a, b) Flow cytometric analysis was performed on coLP ZsGreen+ Vγ6+ Tγδ17 cells from mice with compound Junb and Fosl2 conditional deletions on the Il17aCreR26ZSG deleter background at steady state (TF+/+, TFWT; TFfl/+, TFHET; TFfl/fl, TFKO): (a) Representative flow cytometric analysis of the frequency of IL-17A and IFNγ producing cells following 4 h PMA/ionomycin stimulation. (n = 8 WT, 5 JunB KO Fosl2 KO; three independent experiments) (b) Representative flow cytometric analysis and summary plots of the frequency of IL-17A and IFNγ producing colonic ZS+ Vγ6+ Tγδ17 cells at steady state following 20 h stimulation with IL-23 and IL-1β. (n = 6 Fosl2 KO, 11 JunB KO, 4 Fosl2 KO JunB HET, 4 Fosl2 HET JunB KO, 5 JunB KO Fosl2 KO; four independent experiments). (c) Flow cytometric analysis was performed on ZS+ Vγ6+ from mLN of naïve Bach2+/+Il17aCreR26ZSG (Bach2WT) and Bach2fl/flIl17aCreR26ZSG (Bach2KO) mice on day 9 of Tγδ17 mLN culture. Summary plots of the frequency of IL-17A and IFNγ producing cells following 4 h PMA/ionomycin stimulation (n = 5 mice/genotype; three independent experiments). (d) Gating strategy for fate-mapped Vγ6+ Tγδ17 cells (CD3ε+γδTCR+TCRβ−ZS+Vγ6+). (e) JunB gMFI measured by flow cytometry after 24 h culture with cytokines from coLP ZS+ Vγ6+ Tγδ17 cells sorted from Il17aCreR26ZSG mice. (f) Representative CUT&RUN tracks for JunB and Fosl2 in Vγ6+ Tγδ17 cells and pseudobulk ATAC track for C0 Vγ6+ Tγδ17 cells. Orange bar represents significant peak called over IgG control. Break in gene intron represented by // or \\. Visualized in IGV. (g) Barplot showing JunB-bound or not bound DEGs between C7 vs C0 Vγ6+ Tγδ17 cells (p-val adj < 0.05 and log2FC > |0.58|). (a–c) Gating was performed on fate-mapped Vγ6+ Tγδ17 cells (CD3ε+γδTCR+TCRβ−ZS+Vγ4−). Statistical analyses include Two-tailed unpaired Student’s t-tests for (a, c) and an Ordinary one-way ANOVA test for (b). Results represent mean ± s.e.m. *P < 0.05; ****P < 0.0001; ns, not significant. Numbers in flow plots represent percentages of cells in the gate. DEG, differentially expressed gene.
Supplementary information
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.
About this article
Cite this article
Parker, M.E., Mehta, N.U., Liao, TC. et al. Restriction of innate Tγδ17 cell plasticity by an AP-1 regulatory axis. Nat Immunol 26, 1299–1314 (2025). https://doi.org/10.1038/s41590-025-02206-7
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41590-025-02206-7