Abstract
CD4+ T cells differentiate into various subsets, including T helper 1 (Th1), Th2, Th9, Th17 and regulatory T (Treg) cells, which are essential for immune responses and cancer immunotherapy. However, the role of RNA N6-methyladenosine (m6A) modification in this differentiation is unclear. Here we show that YTHDF2, an important m6A reader protein known to destabilize m6A-modified mRNA, negatively regulates Th9 cell differentiation. Ablation of Ythdf2 in both mouse and human naive CD4+ T cells promotes Th9 differentiation by stabilizing Gata3 and Smad3 mRNA under interleukin-4 (IL-4) and transforming growth factor β (TGF-β) signaling, respectively. Ythdf2-deficient Th9 cells produce increased amounts of IL-9 and IL-21, leading to increased tumor infiltration and cytotoxicity by CD8+ T cells and natural killer (NK) cells, thereby improving antitumor activity compared with wild-type Th9 cells. Moreover, YTHDF2 depletion in CAR-Th9 cells enhances their immune activation, reduces their terminal differentiation and augments their antitumor efficacy. Targeting YTHDF2 is thereby a promising strategy to enhance Th9 and CAR-Th9 cell-based cancer immunotherapies.
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
The m6A-seq, RIP-seq and bulk RNA-seq data have been deposited in the Gene Expression Omnibus under accession code GSE279983. The m6A-seq and RIP-seq were mapped to the mouse (GRCm38) genomes. The bulk RNA-seq data were mapped to the human (GRCh39) or mouse (GRCm38) genomes. Source data are provided with this paper.
Code availability
Code is available at https://github.com/zerostwo/ythdf2-th9.
References
Gerlach, K. et al. TH9 cells that express the transcription factor PU.1 drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells. Nat. Immunol. 15, 676–686 (2014).
Végran, F. et al. The transcription factor IRF1 dictates the IL-21-dependent anticancer functions of TH9 cells. Nat. Immunol. 15, 758–766 (2014).
Xue, G. et al. Adoptive cell therapy with tumor-specific Th9 cells induces viral mimicry to eliminate antigen-loss-variant tumor cells. Cancer Cell 39, 1610–1622 (2021).
Dardalhon, V. et al. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3− effector T cells. Nat. Immunol. 9, 1347–1355 (2008).
Veldhoen, M. et al. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat. Immunol. 9, 1341–1346 (2008).
Lu, Y. et al. Th9 cells promote antitumor immune responses in vivo. J. Clin. Invest. 122, 4160–4171 (2012).
Lu, Y. et al. Tumor-specific IL-9-producing CD8+ Tc9 cells are superior effector than type-I cytotoxic Tc1 cells for adoptive immunotherapy of cancers. PNAS 111, 2265–2270 (2014).
Lu, Y. et al. Th9 cells represent a unique subset of CD4+ T cells endowed with the ability to eradicate advanced tumors. Cancer Cell 33, 1048–1060.e7 (2018).
Purwar, R. et al. Robust tumor immunity to melanoma mediated by interleukin-9-producing T cells. Nat. Med. 18, 1248–1253 (2012).
Liu, L. et al. Enhanced CAR-T activity against established tumors by polarizing human T cells to secrete interleukin-9. Nat. Commun. 11, 5902 (2020).
Ma, X. et al. Cholesterol negatively regulates IL-9-producing CD8+ T cell differentiation and antitumor activity. J. Exp. Med. 215, 1555–1569 (2018).
Liu, Y. et al. N6-methyladenosine-mediated gene regulation and therapeutic implications. Trends Mol. Med. 29, 454–467 (2023).
Li, X., Ma, S., Deng, Y., Yi, P. & Yu, J. Targeting the RNA m6A modification for cancer immunotherapy. Mol. Cancer 21, 76 (2022).
Xiao, S. et al. YTHDF2: a key RNA reader and antitumor target. Trends Immunol. 46, 485–498 (2025).
Wang, X. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).
Zhang, H. et al. YTHDF2 upregulation and subcellular localization dictate CD8 T cell polyfunctionality in anti-tumor immunity. Nat. Commun. 15, 9559 (2024).
Jing, H. et al. METTL3 governs thymocyte development and thymic involution by regulating ferroptosis. Nat. Aging 4, 1813–1827 (2024).
Guo, W. et al. Mettl3-dependent m6A modification is essential for effector differentiation and memory formation of CD8+ T cells. Sci. Bull. 69, 82–96 (2024).
Ito-Kureha, T. et al. The function of Wtap in N6-adenosine methylation of mRNAs controls T cell receptor signaling and survival of T cells. Nat. Immunol. 23, 1208–1221 (2022).
Zhou, J. et al. m6A demethylase ALKBH5 controls CD4+ T cell pathogenicity and promotes autoimmunity. Sci. Adv. 16, eabg0470 (2021).
Yao, Y. et al. METTL3-dependent m6A modification programs T follicular helper cell differentiation. Nat. Commun. 12, 1333 (2021).
Grenov, A., Hezroni, H., Lasman, L., Hanna, J. H. & Shulman, Z. YTHDF2 suppresses the plasmablast genetic program and promotes germinal center formation. Cell Rep. 39, 110778 (2022).
Huang, H. et al. Mettl14-mediated m6A modification is essential for germinal center B cell response. J. Immunol. 208, 1924–1936 (2022).
Grenov, A. C. et al. The germinal center reaction depends on RNA methylation and divergent functions of specific methyl readers. J. Exp. Med. 218, e20210360 (2021).
Song, H. et al. METTL3-mediated m6A RNA methylation promotes the anti-tumour immunity of natural killer cells. Nat. Commun. 12, 5522 (2021).
Ma, S. et al. The RNA m6A reader YTHDF2 controls NK cell antitumor and antiviral immunity. J. Exp. Med. 18, e20210279 (2021).
Han, D. et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature 566, 270–274 (2019).
Wen, C. et al. YTHDF1 loss in dendritic cells potentiates radiation-induced antitumor immunity via STING-dependent type I IFN production. J. Clin. Invest. 134, e181612 (2024).
Wang, H. et al. Mettl3-mediated mRNA m6A methylation promotes dendritic cell activation. Nat. Commun. 10, 1898 (2019).
Dong, L. et al. The loss of RNA N6-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8+ T cell dysfunction and tumor growth. Cancer Cell 39, 945–957.e10 (2021).
Yin, H. et al. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming. Nat. Commun. 12, 1394 (2021).
Zhuang, T. et al. ALKBH5-mediated m6A modification of IL-11 drives macrophage-to-myofibroblast transition and pathological cardiac fibrosis in mice. Nat. Commun. 15, 1995 (2024).
Wang, X. et al. The m6A reader IGF2BP2 regulates macrophage phenotypic activation and inflammatory diseases by stabilizing TSC1 and PPARγ. Adv. Sci. (Weinh., Baden.-Wurtt., Ger.) 8, 2100209 (2021).
Ge, X. et al. The loss of YTHDC1 in gut macrophages exacerbates inflammatory bowel disease. Adv. Sci. (Weinh., Baden.-Wurtt., Ger.) 10, e2205620 (2023).
Ma, S. et al. YTHDF2 orchestrates tumor-associated macrophage reprogramming and controls antitumor immunity through CD8+ T cells. Nat. Immunol. 24, 255–266 (2023).
Wang, L. et al. YTHDF2 inhibition potentiates radiotherapy antitumor efficacy. Cancer Cell 41, 1294–1308.e8 (2023).
Lyu, Z. et al. Suppression of YTHDF2 attenuates autoimmune hepatitis by expansion of myeloid-derived suppressor cells. J. Autoimmun. 135, 102993 (2023).
Nakatsukasa, H. et al. The DNA-binding inhibitor Id3 regulates IL-9 production in CD4+ T cells. Nat. Immunol. 16, 1077–1084 (2015).
Elyaman, W. et al. Notch receptors and Smad3 signaling cooperate in the induction of interleukin-9-producing T cells. Immunity 36, 623–634 (2012).
Jabeen, R. et al. Th9 cell development requires a BATF-regulated transcriptional network. J. Clin. Invest. 123, 4641–4653 (2013).
Bassil, R. et al. BCL6 controls Th9 cell development by repressing Il9 transcription. J. Immunol. 193, 198–207 (2014).
Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).
Sekine, T. TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells. Sci. Immunol. 5, eaba7918 (2020).
Tamiya, T. et al. Smad2/3 and IRF4 play a cooperative role in IL-9-producing T cell induction. J. Immunol. 191, 2360–2371 (2013).
Staudt, V. et al. Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells. Immunity 33, 192–202 (2010).
Jash, A. et al. Nuclear factor of activated T cells 1 (NFAT1)-induced permissive chromatin modification facilitates nuclear factor-κB (NF-κB)-mediated interleukin-9 (IL-9) transactivation. J. Biol. Chem. 287, 15445–15457 (2012).
Perumal, N. B. & Kaplan, M. H. Regulating Il9 transcription in T helper cells. Trends Immunol. 32, 146–150 (2011).
Gutierrez, P. S., Monteoliva, D. & Diambra, L. Cooperative binding of transcription factors promotes bimodal gene expression response. PLoS ONE 7, e44812 (2012).
Chang, H.-C. et al. The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation. Nat. Immunol. 11, 527–534 (2010).
Abdul-Wahid, A. et al. Induction of antigen-specific TH 9 immunity accompanied by mast cell activation blocks tumor cell engraftment. Int J. Cancer 139, 841–853 (2016).
You, F. P. et al. Th9 cells promote antitumor immunity via IL-9 and IL-21 and demonstrate atypical cytokine expression in breast cancer. Int. Immunopharmacol. 52, 163–167 (2017).
Chen, J. et al. Dectin-1-activated dendritic cells: a potent Th9 cell inducer for tumor immunotherapy. Oncoimmunology 5, e1238558 (2016).
Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723 (2017).
Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).
Ma, R. et al. An oncolytic virus expressing IL15/IL15Rα combined with off-the-shelf EGFR-CAR NK cells targets glioblastoma. Cancer Res. 81, 3635–3648 (2021).
Teng, K. Y. et al. Off-the-shelf prostate stem cell antigen-directed chimeric antigen receptor natural killer cell therapy to treat pancreatic cancer. Gastroenterology 162, 1319–1333 (2022).
Xue, G., Jin, G., Fang, J. & Lu, Y. IL-4 together with IL-1β induces antitumor Th9 cell differentiation in the absence of TGF-β signaling. Nat. Commun. 10, 1376 (2019).
Goswami, R. & Kaplan, M. H. A brief history of IL-9. J. Immunol. 186, 3283–3288 (2011).
Noelle, R. J. & Nowak, E. C. Cellular sources and immune functions of interleukin-9. Nat. Rev. Immunol. 10, 683–687 (2010).
Cui, A. et al. Dictionary of immune responses to cytokines at single-cell resolution. Nature 625, 377–384 (2024).
Skak, K., Frederiksen, K. S. & Lundsgaard, D. Interleukin-21 activates human natural killer cells and modulates their surface receptor expression. Immunology 123, 575–583 (2008).
Shanley, M. et al. Interleukin-21 engineering enhances NK cell activity against glioblastoma via CEBPD. Cancer Cell 42, 1450–1466 (2024).
Parrish-Novak, J., Foster, D. C., Holly, R. D. & Clegg, C. H. Interleukin-21 and the IL-21 receptor: novel effectors of NK and T cell responses. J. Leukoc. Biol. 72, 856–863 (2002).
Tian, Y. & Zajac, A. J. IL-21 and T cell differentiation: consider the context. Trends Immunol. 37, 557–568 (2016).
Vivier, E. et al. Innate or adaptive immunity? The example of natural killer cells. Science. 331, 44–49 (2011).
Huntington, N. D., Vosshenrich, C. A. J. & Di Santo, J. P. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat. Rev. Immunol. 7, 703–714 (2007).
Cappell, K. M. & Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).
Shulman, Z. & Stern-Ginossar, N. The RNA modification N6-methyladenosine as a novel regulator of the immune system. Nat. Immunol. 21, 501–512 (2020).
Zhang, L. et al. YTHDF2/m6A/NF‐κB axis controls anti‐tumor immunity by regulating intratumoral Tregs. EMBO J. 42, e113126 (2023).
Tone, Y. et al. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat. Immunol. 9, 194–202 (2008).
Verrecchia, F. & Mauviel, A. Transforming growth factor-beta and fibrosis. World J. Gastroenterol. 13, 3056–3062 (2007).
Lee, K. S. et al. Runx2 is a common target of transforming growth factor beta1 and bone morphogenetic protein 2, and cooperation between Runx2 and Smad5 induces osteoblast-specific gene expression in the pluripotent mesenchymal precursor cell line C2C12. Mol. Cell. Biol. 20, 8783–8792 (2000).
Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 13, 616–630 (2012).
Ross, S. & Hill, C. S. How the Smads regulate transcription. Int. J. Biochem. Cell Biol. 40, 383–408 (2008).
Feng, X. H. & Derynck, R. Specificity and versatility in TGF-beta signaling through Smads. Annu. Rev. Cell Dev. Biol. 21, 659–693 (2005).
Ma, S., Caligiuri, M. A. & Yu, J. Harnessing IL-15 signaling to potentiate NK cell-mediated cancer immunotherapy. Trends Immunol. 43, 833–847 (2022).
Shan, L., Flavell, R. A. & Herndler-Brandstetter, D. Development of humanized mouse models for studying human NK cells in health and disease. Methods Mol. Biol. 2463, 53–66 (2022).
Mansour, A. G. et al. Off-the-shelf CAR–engineered natural killer cells targeting FLT3 enhance killing of acute myeloid leukemia. Blood Adv. 7, 6225–6239 (2023).
Lu, T. et al. Off-the-shelf CAR natural killer cells secreting IL-15 target spike in treating COVID-19. Nat. Commun. 13, 2576 (2022).
Xiao, S. et al. The tumor-intrinsic role of the m6A reader YTHDF2 in regulating immune evasion. Sci. Immunol. 9, eadl2171 (2024).
Ma, S. et al. An XBP1s-PIM-2 positive feedback loop controls IL-15-mediated survival of natural killer cells. Sci. Immunol. 8, eabn7993 (2023).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Acknowledgements
We thank the City of Hope Center for Comparative Medicine, Pathology Core, Analytical Cytometry Core and Integrative Genomics Core for performing parts of the work. The research was supported by grants from the National Institutes of Health (CA210087, CA265095 and CA163205 to M.A.C.; NS106170, AI129582, CA247550, CA264512, CA266457 and CA223400 to J.Y.) and the California Institute for Regenerative Medicine (TRAN1-14716 to M.A.C.; TRAN1-14003 and DISC2-14190 to J.Y.).
Author information
Authors and Affiliations
Contributions
S.X., S.M., M.A.C. and J.Y. conceived and designed the project. S.X. and Y.H. contributed to most of the in vitro and in vivo experiments. S.D. contributed to the RNA-seq, m6A-seq and RIP-seq analysis. S.X., S.D. and J.Z. analyzed the data. S.X., S.M., M.A.C. and J.Y. wrote, reviewed and/or revised the paper. J.Y. and M.A.C. acquired funding and supervised the study. All authors discussed and approved the paper.
Corresponding authors
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: Nick Bernard, in collaboration with the Nature Immunology 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 YTHDF2 deficiency does not affect Th1, Th2, Th17, and Treg cell differentiation.
(a) YTHDF2 and β-actin expression in mouse wild-type (WT) and Ythdf2-knockout (KO) naive CD4+ T cells, determined by immunoblotting. Images are representatives of at least three independent experiments. (b-e) Representative plots of IFN-γ, IL-13, IL-17A, and FOXP3 expression in WT or Ythdf2-KO Th1 (b), Th2 (c), Th17 (d), and Treg (e) cells on the 5th day after differentiation. (f, g) Representative plots and percentage of IL-2 (f) and IL-21 (g) production from WT (Ythdf2+/+) or Ythdf2−/− Th9 cells on the 5th day after differentiation (n = 3 mice). Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (f right and g right).
Extended Data Fig. 2 Ythdf2cKO mice have normal T cell development.
(a) YTHDF2 and β-actin expression in mouse naive CD4+ T cells isolated from Ythdf2f/f and Ythdf2cKO mice, determined by immunoblotting (n = 4 mice). (b-k) Flow cytometric analysis of the frequencies of the indicated lymphoid immune cells in the thymus (b, c), spleen (d-g), and peripheral lymph nodes (h-k) of Ythdf2f/f and Ythdf2cKO mice. Data are presented as representative plots (a, c, e, g, and i) and summary bar graphs (b, d, f, h, and j) (n = 4 mice). Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (D and F) or the one-way ANOVA model (b, h, and j). DP, double positive; DN, double negative. Tn, naive T cells; Tcm, central memory T cells; Teff, effector T cells.
Extended Data Fig. 3 YTHDF2 deficiency in mice does not affect Th1, Th2, Th17, and Treg differentiation.
(a-h) Flow cytometric analysis of the frequencies of the indicated protein expression in Ythdf2f/f and Ythdf2cKO Th1 (a, b), Th2 (c, d), Th17 (e, f), and Treg cells (g, h). Data are presented as representative plots (a, c, e, and g) and summary bar graphs (b, d, f, and h) (n = 3 mice). (i-l) Representative plots and percentage of IL-2 (i, j) and IL-21 (k, l) production from Ythdf2f/f and Ythdf2cKO Th9 cells on the 5th day after differentiation (n = 3 mice). (m, n) Representative plots (m) and percentage (n) of WT (Ythdf2+/+) or Ythdf2−/− Tc9 cells on the 5th day after differentiation (n = 5 mice). Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (b, d, f, h, j, l, and n).
Extended Data Fig. 4 Transcriptome-wide RNA-seq, m6A-seq, and RIP-seq assays in murine Th9 cells.
(a) The m6A motif was detected by the HOMER motif discovery tool with m6A-seq data. (b) Density distribution of the m6A peaks across the mRNA transcriptome from m6A-seq data. (c) The proportion of m6A peak distribution in Th9 cells from Ythdf2f/f and Ythdf2cKO mice. (d) GO analysis of transcripts with m6A peaks. (e) Density distribution of the YTHDF2-binding sites across the mRNA transcriptome from RIP-seq data. (f) The proportion of YTHDF2-binding site distribution from RIP-seq data. (g) Top 10 GO clusters from GO analysis of YTHDF2 target genes from RIP-seq data. (h) Venn plot showing an overlapping analysis of genes identified by RNA-seq (upregulated genes), m6A-seq, and RIP-seq. (i) qPCR analysis of mRNA expression of Gata3 in Ythdf2f/f and Ythdf2cKO Th9 cells after intrasample normalization to the reference gene 18S (n = 4 biological replicates). (j and k) Immunoblot analysis of GATA3 in whole-cell lysates of naive Ythdf2f/f and Ythdf2cKO CD4+ T cells cultured under Th9 conditions (n = 3 biological replicates). (l) qPCR analysis of mRNA expression of Smad3 in Ythdf2f/f and Ythdf2cKO Th9 cells after intrasample normalization to the reference gene 18S (n = 3 biological replicates). (m and n) Immunoblot analysis of SMAD3 in whole-cell lysates of naive Ythdf2f/f and Ythdf2cKO CD4+ T cells cultured under Th9 conditions (n = 3 biological replicates). (o and p) GATA3, SMAD3 and β-actin expression in Gata3 (o) or Smad3 (p) knockout Ythdf2f/f Th9 cells, as well as in Ythdf2cKO Th9 cells, determined by immunoblotting. Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (i, k, l, and n). P values in a, d, e, and g were determined by a hypergeometric test.
Extended Data Fig. 5 Smad3 and Gata3 interaction promotes Th9 cell differentiation.
(a) Luciferase reporter assay showing that Gata3 alone, or Smad3 alone, or their combination did not activate Il9 gene transcription without p65 in 293T cells (n = 3 biological replicates). (b) Luciferase reporter assay showing that Irf4 alone, or Pu.1 alone, or their combination did not activate Il9 gene transcription without p65 in 293T cells (n = 3 biological replicates). (c) Luciferase reporter assay showing that Irf4 and/or Pu.1 activates Il9 gene transcription in the presence of p65 in 293T cells (n = 3 biological replicates). (d, e) Binding of Smad3 to the Il9 promoter in Smad3, Smad3 and Gata3 overexpressed 293T cells was determined by ChIP-qPCR (n = 3 biological replicates). Data are represented as the mean ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (d, e), or two-way ANOVA models with P values adjusted for multiple comparisons by Holm-Šídák method (a, b). For statistical analysis in (c), luciferase reporter assay results were log2-transformed after adding a pseudo count of 1 to each value to stabilize variance and account for zero values, and then two-way ANOVA models with P values adjusted for multiple comparisons by Holm-Šídák method.
Extended Data Fig. 6 Ythdf2cKO Th9 cells don’t affect mast cells, macrophages, and MDSCs.
(a) qPCR analysis of mRNA expression of Gzmb in Ythdf2f/f and Ythdf2cKO Th9 cells after intrasample normalization to the reference gene 18S (n = 4 mice). (b, c) Tumor growth in NSG (b; n = 3 mice) and Rag2−/−Il2gc−/−(c; n = 4 mice) mice that were s.c. inoculated with B16-OVA tumor cells and then adoptively transferred with Ythdf2f/f and Ythdf2cKO Th9 cells. (d-p) Flow cytometric analysis of tumor-infiltrating mast cells (d-g, n = 5 mice), macrophages (i-k: n = 4 mice; l: n = 5 mice), and MDSCs (n-p; n = 4 mice), and IHC staining analysis of tumor-infiltrating mast cells (h; n = 3 biological replicates), macrophages (m; n = 5 independent replicates) in B16-OVA tumor-bearing mice that received adoptive transfer of either OT-II Ythdf2f/f or Ythdf2cKO Th9 cells. Scale bar, 100 μm (m, n). Data are presented as representative plots, summary graphs, and the absolute number of per gram tumors. Data are represented as the mean ± SD. Statistical analysis was performed using unpaired two-tailed t-tests (a, e, f, h, i, k, and l), or two-way ANOVA models with P values adjusted for multiple comparisons by Holm-Šídák method (b, c).
Extended Data Fig. 7 Ythdf2cKO Th9 cells enhance cytotoxic CD8+ T cell infiltration.
(a) Representative plots (left), summary graphs (right) of tumor-infiltrating perforin+GZMB+ CD8+ T cells in B16-OVA tumor-bearing mice, by flow cytometric analysis (n = 5 mice). (b-g) Representative plots (b, e), summary graphs (c, n = 4 mice; f, n = 5 mice), and the absolute number of per gram tumors (d, n = 4 mice; g, n = 5 mice) of tumor-infiltrating CD8+ T cells, as indicated in different tumor-bearing mice, by flow cytometric analysis. (h-m) Representative plots (h, k), summary graphs (i, n = 4 mice; l, n = 5 mice), and the absolute number of per gram tumors (j, n = 4 mice; m, n = 5 mice) of tumor-infiltrating IFN-γ+CD8+ T cells, as indicated in different tumor-bearing mice, by flow cytometric analysis. Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests.
Extended Data Fig. 8 YTHDF2-deficient Th9 cells enhance the infiltration of NK cells.
(a, b) Flow cytometric analysis of tumor-infiltrating NK cells, as indicated in LLC1-OVA (a; n = 4 mice) and E0771-OVA (b; n = 5 mice) tumor-bearing mice that received adoptive transfer of either Ythdf2f/f or Ythdf2cKO Th9 cells. Data are presented as representative plots (left panels), summary graphs (middle panels), and the absolute number of per gram tumors (right panels). (c, d) Flow cytometric analysis of tumor-infiltrating IFN-γ+ NK cells, as indicated in different tumor-bearing mice (c, n = 4 mice; d, n = 5 mice). Data are presented as representative plots (left panels), summary graphs (middle panels), and the absolute number of per gram tumors (right panels). Data are represented as means ± SD. Statistical analysis was performed using unpaired two-tailed t-tests.
Extended Data Fig. 9 YTHDF2-deficient Th9 cells enhance the effector function of NK cells in Rag1−/− mice.
(a-d) (a, b) Flow cytometric analysis of tumor-infiltrating NK cells, as indicated in B16-OVA tumor-bearing Rag1−/− mice and (c, d) in EO771-OVA tumor-bearing mice. Data are presented as representative plots (left panels), summary graphs (middle panels), and the absolute number of per gram tumors (right panels) (a, c n = 4 mice; b, d, n = 5 mice). (e) The ability of NK cells to lyse B16F10 tumor cells in the presence of either Ythdf2f/f or Ythdf2cKO Th9 cells, measured by real-time cell analysis (n = 3 mice). Data are represented as the mean ± SD (a, b, c, and d) or the mean (e). Statistical analysis was performed using unpaired two-tailed t-tests (a, b, c, d).
Extended Data Fig. 10 YTHDF2 deficiency promotes the differentiation of Th9 cells in humans.
(a) Representative plots and percentage of human YTHDF2+/+ and YTHDF2−/− Tc9 cells on the 7th day after differentiation (n = 5 independent donors). (b, c) Flow cytometric analysis of the anti-PSCA (b; n = 5 independent donors) or anti-EGFR (c; n = 4 independent donors) CAR transduction efficacy of human YTHDF2+/+ and YTHDF2−/− CAR-Th9 cells. Data are presented as representative plots (left panels) and summary graphs (right panels). (d, e) Flow cytometric analysis of the IL-9 production in YTHDF2+/+ or YTHDF2−/− CAR-Th9 cells. Data are presented as representative plots (left panels) and summary graphs (right panels) (d, n = 5 independent donors; e, n = 4 independent donors). (f, g) The ability of human YTHDF2+/+ or YTHDF2−/− anti-EGFR CAR-Th9 to lyse A549 or Capan-1 tumor cells, measured by real-time cell analysis (representative of 2 experiments, showing the mean cell index). (h) Schematic of primary tumor growth assay involving NSCLC cells. (i) Tumor growth of established tumor cells, as described in f. Tumor volumes were measured every 4 days (one experiment was performed with n = 4 mice). (j) Schematic of primary tumor growth assay involving PDAC cells. (k) Tumor growth of established tumor cells, as described in H. Tumor volumes were measured every 4 days (one experiment was performed with n = 4 mice). Data in (i,k) show the mean plus SD. Statistical analysis was performed using paired two-tailed t-tests (a-e) and two-way ANOVA with a mixed-effects model with P values adjusted for multiple comparisons by Holm-Šídák method (i, k).
Supplementary information
Supplementary Information
Supplementary Fig. 1. Gating strategy of NK cells (CD45+CD3−NK1.1+) and CD8+ T cells (CD45+CD3+NK1.1−CD4−CD8+) in B16-OVA tumor-bearing mice that received adoptively transferred either OT-II-Ythdf2f/f or OT-II-Ythdf2cKO Th9 cells.
Supplementary Tables
Supplementary Table 1. A list of 117 genes that overlap among RNA-seq upregulated genes, m6A-seq data, and YTHDF2 RIP-seq data. Supplementary Table 2. List of immunoblotting antibodies used for this study. Supplementary Table 3. List of primers used for this study.
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. 3
Unprocessed western Blots.
Source Data Fig. 4
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Fig. 7
Statistical source data.
Source Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 1
Unprocessed western Blots.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 2
Unprocessed western Blots.
Source Data Extended Data Fig. 3
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 4
Unprocessed western Blots
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.
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
Xiao, S., Duan, S., Hong, Y. et al. Loss of YTHDF2 enhances Th9 programming and CAR-Th9 cell antitumor efficacy. Nat Immunol 26, 1501–1515 (2025). https://doi.org/10.1038/s41590-025-02235-2
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41590-025-02235-2
This article is cited by
-
A little less reading, a little more type 9 action
Nature Immunology (2025)