Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Nov;26(11):2059-2073.
doi: 10.1038/s41590-025-02295-4. Epub 2025 Oct 8.

Temporal and context-dependent requirements for the transcription factor Foxp3 expression in regulatory T cells

Affiliations

Temporal and context-dependent requirements for the transcription factor Foxp3 expression in regulatory T cells

Wei Hu et al. Nat Immunol. 2025 Nov.

Abstract

Regulatory T (Treg) cells, expressing the transcription factor Foxp3, are obligatory gatekeepers of immune responsiveness, yet the mechanisms by which Foxp3 governs the Treg transcriptional network remain incompletely understood. Using a novel chemogenetic system of inducible Foxp3 protein degradation in vivo, we found that while Foxp3 was indispensable for the establishment of transcriptional and functional programs of newly generated Treg cells, Foxp3 loss in mature Treg cells resulted in minimal functional and transcriptional changes under steady state. This resilience of the Foxp3-dependent program in mature Treg cells was acquired over an unexpectedly long timescale; however, in settings of severe inflammation, Foxp3 loss led to a pronounced perturbation of Treg cell transcriptome and fitness. Furthermore, tumoral Treg cells were uniquely sensitive to Foxp3 degradation, which led to impairment in their suppressive function and tumor shrinkage in the absence of pronounced adverse effects. These studies demonstrate a context-dependent differential requirement for Foxp3 for Treg transcriptional and functional programs.

PubMed Disclaimer

Conflict of interest statement

Competing interests: A.Y.R. is a Scientific Advisory Board member and has equity in Sonoma Biotherapeutics, RAPT Therapeutics, Coherus BioSciences, Santa Ana Bio, Odyssey Therapeutics, Nilo Therapeutics and Vedanta Biosciences; he is also a Scientific Advisory Board member of BioInvent and Amgen and a co-inventor of a CCR8+ Treg cell depletion IP licensed to Takeda, which is unrelated to the content of this publication. Z.-M.W. is an employee of Genentech, which is unrelated to the content of this publication. The other authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Foxp3 degradation causes minimal immune activation in adult lymphoreplete mice.
a, Schematic of the inducible Foxp3 protein degradation model. SCF, Skp-1-Cullin-F-box complex. b, Schematic of the Foxp3AID and R26TIR alleles. c, Flow cytometry plot showing 5-ph-IAA-induced Foxp3 protein degradation after 24 h (left). Scatter-plot of Foxp3 protein expression median fluorescence intensity (MFI) as assessed by flow cytometric analysis in Foxp3AID mice after 7 days of daily 5-ph-IAA injection (right). d, Experimental design. e, Size of spleen and lymph nodes after 14 days of Treg cell ablation or Foxp3 degradation. f, Activation, proliferation and cytokine production of CD4+ (top) and CD8+ (bottom) T cells following Treg cell ablation or Foxp3 degradation. g, Number of eosinophils, neutrophils and monocytes and CD86 levels on dendritic cells following Treg cell ablation or Foxp3 degradation. h, Serum antibody levels following Treg cell ablation or Foxp3 degradation. i,j, Representative hematoxylin and eosin (H&E) stain (i) and histology scores (j) of the liver following Treg cell ablation or Foxp3 degradation. k, Liver damage measured by serum alanine aminotransferase, albumin and albumin:globulin ratio. Scatter-plots represent mean ± s.e.m. Each point represents a unique mouse. Data are pooled from two independent experiments. Statistical analysis was conducted by one-way analysis of variance (ANOVA). Source data
Fig. 2
Fig. 2. Foxp3 degradation induces minimal gene expression and functional changes in mature Treg cells.
a, Experimental design of scRNA-seq and functional assays. Each genotype and time point consisted of four independent biological replicates. b, UMAP visualization of scRNA-seq data from Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) Treg cells before and 3 or 7 days after 5-ph-IAA-induced Foxp3 degradation. c, UMAP visualization of the same scRNA-seq data, colored by identified clusters. d, Fraction of each cluster within the total pool of Foxp3AIDR26WT or Foxp3AIDR26TIR1(F74G) Treg cells separated by time point. Each point represents a unique mouse. e, In vitro suppression assay of Treg cells sorted from Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) mice after 7 days of in vivo 5-ph-IAA treatment. 5-ph-IAA was included in culture to sustain Foxp3 degradation. Line graph represents mean ± s.e.m. Data are pooled from two independent experiments and analyzed using two-sided multiple t-tests. NS, not significant. f, Experimental design of the bulk RNA-seq analysis. Each genotype and time point consisted of three independent biological replicates. g, Gating strategy for sorting resting and activated Treg cells. h, Schematic comparison of the Foxp3GFPKO reporter-null allele and the functional Foxp3GFP allele. i, Scatter-plots and bar graphs showing the number of DEGs in resting or activated Treg cells caused by Foxp3 protein degradation or genetic Foxp3 deficiency. Source data
Fig. 3
Fig. 3. Foxp3 degradation in mature Treg cells induces expression changes in a small set of genes.
a, Treg cells from the scRNA-seq dataset were classified as resting or activated based on exceeding the threshold for resting or activated gene signature scores and were subsequently analyzed. b, Scatter-plot showing the correlation of gene expression changes induced by Foxp3 degradation at day 3 and day 7 in resting and activated Treg cells. FC, fold change. c, UMAP visualization of resting and activated Treg cells colored by gene signature scores for the ‘TIR1-up’ and ‘TIR1-down’ gene sets, up- and downregulated upon Foxp3 degradation, respectively. d, Dot plot summarizing statistically significant DEGs in resting or activated Treg cells following 3 or 7 days of 5-ph-IAA-induced Foxp3 degradation. The color represents the log2 fold change of R26TIR1(F74G) versus R26WT and the size represents the Benjamini–Hochberg adjusted P value of the differential expression test. e, Flow cytometry analysis of Foxp3 protein and mRNA levels (reported by ZsGreen) in Foxp3AID Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. Scatter-plots represent mean ± s.e.m. Data are pooled from two independent experiments and analyzed using a two-way ANOVA. pLN, peripheral lymph nodes. f, Flow cytometry analysis of CD25, CD122, OX40, GITR and FR4 protein levels in Foxp3AID Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. g, Flow cytometry analysis of CD127 and TCF1 protein levels in Foxp3AID Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. Scatter-plots represent mean ± s.e.m. (f,g). Each point represents a unique mouse. Data are pooled from two independent experiments and analyzed with two-sided multiple t-tests. Source data
Fig. 4
Fig. 4. Foxp3 degradation-sensitive genes in mature Treg cells are enriched for Foxp3 binding.
a, Bar graphs showing the proportion of ATAC-seq peaks near Foxp3 degradation-induced DEGs bound by Foxp3 (ref. ). Genes are stratified by statistical significance (P values) in resting and activated Treg cells. Bars represent mean ± s.e.m. Data were analyzed using a Mann–Whitney U-test. b, Bar graphs showing the proportion of ATAC-seq peaks near Foxp3-dependent DEGs bound by Foxp3. Genes are stratified by P values in resting and activated Treg cells. Bars represent mean ± s.e.m. Data were analyzed using a Mann–Whitney U-test. c, H3K27Ac and H3K27me3 ChIP-seq signals at Foxp3-bound ATAC-seq peaks near Foxp3 degradation-induced DEGs. Line graph represents mean ± s.e.m. Data were analyzed using a Mann–Whitney U-test. d, Dot plot showing transcription factor motif enrichment within Foxp3-bound regions near Foxp3 degradation-induced DEGs. e, Schematic diagram illustrating the ‘on’ and ‘off’ states of the reversible reporter-null Foxp3LSL allele. f, Experimental design of the gain-of-function experiment to induce Foxp3 expression in Treg ‘wannabe’ cells. Each genotype and time point consisted of two independent biological replicates. g, Line graph depicting gene expression changes of TIR1-up or TIR1-down genes below a specific P value cutoff across different time points following Foxp3 induction in Treg ‘wannabe’ cells. The line and shading represent the mean ± s.e.m. Data were analyzed using a Mann–Whitney U-test.
Fig. 5
Fig. 5. Foxp3 is preferentially required for regulation of gene expression during early Treg cell differentiation.
a, Experimental design for transcriptional profiling of developing thymic Treg cells. Each genotype consisted of three independent biological replicates. b, Gating strategy used to sort CD73⁻ nascent thymic Treg cells. c, Bar graph comparing the number of Foxp3 degradation-induced DEGs in thymic, resting and activated Treg cells from Foxp3AID/WT mice. d, Pearson correlation between Foxp3 degradation-induced and Foxp3-dependent DEGs in thymic, resting and activated Treg cells. e, Scatter-plot and cumulative distribution function (CDF) plots comparing Foxp3 degradation-induced and Foxp3-dependent DEGs across the three Treg populations. Data were analyzed using a Mann–Whitney U-test. f, Metacell analysis of thymocyte scRNA-seq data correlating UMI-normalized Foxp3 expression levels in each metacell with the expression of TIR1-up and TIR1-down gene signatures identified in ac. UMAP plots are colored by scaled expression levels of TIR1-up, TIR1-down and UMI-normalized counts of Foxp3. Dashed red line depicts line of best fit. Correlations and corresponding P values were calculated with Pearson correlation over all genes. g, Experimental design for in vivo Foxp3 degradation in 1-day-old neonatal Foxp3AID mice and adult Foxp3AID mice. h, CD4+ and CD8+ T cell activation in adult and neonatal Foxp3AID mice following Foxp3 degradation. i, Expansion of eosinophils and neutrophils in adult and neonatal Foxp3AID mice after Foxp3 degradation. j,k, Representative H&E staining (j) and histology scores of liver inflammation (k) in neonatal Foxp3AID mice following Foxp3 degradation. l, In vitro suppression assay of Treg cells sorted from Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) neonatal mice after 7 days of in vivo 5-ph-IAA administration. 5-ph-IAA was also included in culture to maintain Foxp3 degradation. Each point represents a unique mouse (hl). Data are pooled from two independent experiments. Scatter-plots represent mean ± s.e.m. Data were analyzed using a one-way ANOVA. m, Bar graphs summarizing the number of Foxp3 degradation-induced DEGs in Treg cells from neonatal and adult Foxp3AID/y mice after 14 days of Foxp3 degradation. n, Scatter-plot correlating gene expression changes induced by Foxp3 degradation and Foxp3 gene deficiency in neonatal mice. o, Scatter-plots comparing gene expression changes induced by 7 days of Foxp3 degradation in neonatal Treg cells to those in adult thymic, resting and activated Treg cells from Foxp3AID/WT mice. Source data
Fig. 6
Fig. 6. Proliferation and inflammation increase Treg cell sensitivity to Foxp3 degradation.
a, Experimental design of proliferating Treg analysis in vivo. b, Flow cytometry analysis of CD25, GITR and CTLA4 protein levels in dividing versus nondividing Treg cells following 7 days of in vivo Foxp3 degradation, in comparison to Foxp3-deficient Treg ‘wannabe’ cells. Scatter-plots represent mean ± s.e.m. Each point represents a unique mouse. Data are representative of two independent experiments and were analyzed using a one-way ANOVA. c, Experimental design of proliferating Treg cell analysis in vitro. d,e, Combined data (d) and representative plots (e) showing CD153, GARP, CD4 and Foxp3 protein levels in lowly and highly divided Treg cells. Similarly treated naive CD4 T cells serve as Foxp3 controls. Bar graphs represent mean ± s.e.m. Each point represents cells from a unique mouse. Data are representative of two independent experiments and were analyzed using a two-way ANOVA. f, IL-2, IL-4 and IL-13 concentrations in the supernatant of in vitro proliferating Treg assay. Bar graphs represent mean ± s.e.m. Each point represents cells from a unique mouse. Data are pooled from two independent experiments and were analyzed using a two-tailed t-test. g, Experimental design of proliferating Treg cell analysis in vitro. Experiment consisted of five technical replicates per genotype. h, Volcano plots of DEGs from g separated by their presence among all up- or downregulated genes in identified in Fig. 3a. Numbers of up- or downregulated genes are labeled in each plot. i, Experimental design of inflammatory Treg cell analysis in vivo. Experiment consisted of four biological replicates per condition (Tcrbd double knockout (dKO) or Foxp3DTR). j, Flow cytometric analysis of the transferred Treg cell mixture in i. k, Recovery of cells in each condition stratified by genotype (mCherry+ for Foxp3AIDR26TIR1(F74G) or mCherry for Foxp3AIDR26WT) as identified in the scRNA-seq data. Each point represents a unique mouse. Bar graphs represent mean ± s.d. Data were analyzed using a paired two-tailed t-test. l, Density contour plots of R26WT and R26TIR1(F74G) overlayed on UMAP embeddings of the scRNA-seq data. m, Leiden clustering of gene expression data visualized on UMAP embedding for Treg cells from each condition. Clustering was performed independently for each condition using the same resolution value. Fraction of each cluster within the total pool of R26WT and R26TIR1(F74G) Treg cells separated by condition. Within each genotype, each point represents a unique mouse. Data were analyzed using a paired two-tailed t-test. n, Number of DEGs in each condition, colored by up- or downregulation. o, Scatter-plot of log2 fold changes of DEGs between Tcrbd KO and Foxp3DTR conditions. Points are colored by their direction and populations in which they are altered. Source data
Fig. 7
Fig. 7. Foxp3 degradation leads to tumor shrinkage with minimal adverse effects.
a, Schematic of the tumor experiment design. s.c., subcutaneous. b, Tumor burden over time, shown as average (left) and individual (right) tumor growth curves. Line graph represents mean ± s.e.m. (left). Each line represents a unique mouse (right). Data are pooled from two independent experiments and were analyzed using a two-way ANOVA (mixed-effects model) with Geisser-Greenhouse correction. c, Representative tumor images on day 20. d, Representative flow cytometry plots (left) and combined data (right) of IFNγ production by tumor-infiltrating CD8+ T cells. Each point represents a unique mouse. Scatter-plot shows mean ± s.e.m. Data are pooled from two independent experiments and were analyzed using a two-tailed t-test. e, Representative flow cytometry plots (left) and quantification (right) of IL-4 production by tumor-infiltrating ZsGreen⁻ CD4+ T cells. Each point represents a unique mouse. Scatter-plot shows mean ± s.e.m. Data are pooled from two independent experiments and were analyzed using a two-tailed t-test. f, Body weight monitoring throughout the experiment. Line graph shows mean ± s.e.m. Data are pooled from two independent experiments. g, H&E staining of liver and intestine on day 20. Images are representative of two independent experiments. h, Expression levels of Foxp3, GITR, CD39, ZsGreen, CTLA4 and TCF1 in ZsGreen⁺ CD4 T cells from the dLN, ndLN and tumor on day 20. Each point represents a unique mouse. Scatter-plots show mean ± s.e.m. Data are pooled from two independent experiments and were analyzed using multiple t-tests. i,j, UMAP visualization of scRNA-seq analysis of tumor Treg cells from Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) mice on day 14 after tumor implantation, colored by genotype (i) or cluster (j). k, Heatmap showing scaled mean UMI-normalized expression values for each cluster in j. l, Proportional distribution of Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) Treg cells within each cluster in j. Each point represents a unique mouse. Scatter-plot represents mean ± s.e.m. Data were analyzed using multiple log-normal t-tests. m, Number of DEGs between Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) Treg cells in each cluster shown in j. Source data
Extended Data Fig. 1
Extended Data Fig. 1. Generation of Foxp3AID mice.
(a) Gene targeting strategy. The auxin-inducible degron (AID) sequence was fused to the N-terminus of Foxp3 via a seven-amino-acid flexible linker. An IRES-ZsGreen-T2A-iCre-Frt-neo-Frt cassette was inserted into the 3’ UTR. Arrows indicate the locations of PCR primers used to distinguish Foxp3WT and Foxp3AID alleles. IRES: internal ribosome entry sequence. iCre: codon improved Cre recombinase. PGK: PGK promoter. Neo: neomycin-resistant gene. pA: bGH polyA sequence. (b) PCR validation of the knock-in allele. Data is representative of over 200 mice from each genotype. (c) Expression pattern of the ZsGreen reporter in Foxp3AID/WT heterozygous females. ZsGreen expression was restricted to CD25⁺ CD4 T cells, consistent with Foxp3 expression. (d) ZsGreen⁺ Foxp3AID Treg cells suppressed naïve CD4⁺ T cell proliferation comparably to Foxp3GFP Treg cells in vitro. Line graph represents mean ± SEM of two biological replicates. (e) Naïve CD4⁺ T cells from Foxp3AID mice were cultured under Treg inducing conditions and transduced with either a TIR1-encoding retrovirus or the empty vector control. AID-tagged Foxp3 protein was selectively degraded in TIR1-transduced induced Treg (iTreg) cells upon indole acetic acid (IAA) treatment. Scatter plots represents mean ± SEM. Each point represents cells from a unique mouse. Source data
Extended Data Fig. 2
Extended Data Fig. 2. Generation of ROSA26TIR1 and ROSA26TIR1(F74G) mice.
(a) Gene targeting strategy. WPRE: Woodchuck hepatitis virus post-transcriptional regulatory element; DTA: Diphtheria toxin fragment A). (b) Southern blot validation of heterozygous ROSA26TIR1/+ mice using the hybridization probe shown in (a). Data is representative of two independent F1 mice. (c) PCR validation of ROSA26TIR1/+ mice. Data are representative of over 100 mice from each genoype. (d–f) Naïve CD4⁺ T cells from ROSA26TIR1/+ mice were co-transduced with retroviruses expressing Cre and AID-Foxp3 (d). TIR1 expression was induced in a Cre-dependent manner (e), resulting in AID-Foxp3 degradation upon IAA treatment (f). Data are representative of two independent experiments. (g) Guide RNA (gRNA) design for CRISPR-mediated F74-to-G mutation in TIR1. The gRNA seed sequence is shown in gray; the PAM sequence is in pink. The F74G mutation creates a KasI restriction site. (h–i) Validation of the F74G mutation by KasI digestion (h) and Sanger sequencing (i). (j–k) The TIR1 F74G mutation enables in vivo protein degradation in response to 5-ph-IAA. Source data
Extended Data Fig. 3
Extended Data Fig. 3. Foxp3 protein degradation in adult lymphoreplete mice induces minimal immune activation.
(a) Size of the spleen and lymph nodes after 28 days of Foxp3 degradation. (b) Serum antibody levels following Treg ablation or Foxp3 degradation. (c,d) Representative H&E stain (c) and histology scores (d) of the skin following Treg ablation or Foxp3 degradation. Each point represents a unique mouse. Data are pooled from two independent experiments. Scatter blots represent mean ± SEM. One-way ANOVA. Source data
Extended Data Fig. 4
Extended Data Fig. 4. Inducible Foxp3 gene knockout causes minimal immune activation in adult lymphoreplete mice.
(a) Experimental design. (b-c) Representative plot (b) and combined data (c) showing the efficiency of Foxp3 gene knockout. (d) Size of the spleen and lymph nodes after 14 days of inducible Foxp3 gene knockout. (e-g) T cell activation (e), serum antibody levels (f), and myeloid cell expansion (g) following 14 days of inducible Foxp3 gene knockout. (h) Representative H&E stain (left) and histology scores (right) of the skin, liver, and lung following 14 days of Treg ablation or Foxp3 degradation. Each point represents a unique mouse. Data are pooled from two independent experiments. Scatter blots represent mean ± SEM. One-way ANOVA. Source data
Extended Data Fig. 5
Extended Data Fig. 5. Foxp3 degradation in developed Treg cells induces minimal gene expression and chromatin accessibility changes.
(a) UMAP visualization of scRNA-seq data from Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) Treg cells on days 0, 3 and 7 of 5-ph-IAA-induced Foxp3 degradation. (b) UMAP visualization of representative genes from the single-cell RNA-seq dataset colored by expression level. (c) MA plot showing differentially accessible ATAC-seq peaks induced by Foxp3 degradation (left) and Foxp3 gene deficiency (right). Red points represent differentially accessible regions.
Extended Data Fig. 6
Extended Data Fig. 6. Foxp3 degradation in developed Treg cells altered the expression of a small group of genes.
(a) The composition of Foxp3AIDR26WT and Foxp3AIDR26WT(F74G) Treg cells within resting and activated subsets from day 0, day 3, and day 7 of Foxp3 degradation. (b) Number of resting and activated Treg cells utilized for the differential gene expression analysis in Fig. 3. (c) Flow cytometry analysis of Foxp3 protein and mRNA levels (reported by ZsGreen) in Foxp3WT Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. Scatter plots represent mean ± SEM. Data are pooled from two independent experiments. Two-way ANOVA. (d) Bulk RNA-seq read counts of indicated genes in Foxp3AID Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. Each point represents a unique mouse. Bar graphs represent mean ± SEM. (e) Flow cytometry analysis of CD25, CD122, OX40, GITR, and FR4 protein levels in Foxp3WT Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. (f) Flow cytometry analysis of CD127 and TCF1 protein levels in Foxp3AID Treg cells from heterozygous Foxp3AID/WTR26WT and Foxp3AID/WTR26TIR1(F74G) females after 7 days of Foxp3 degradation. (e-f) Scatter plots represent mean ± SEM. Each point represents a unique mouse. Data are pooled from two independent experiments. Multiple t-tests. Source data
Extended Data Fig. 7
Extended Data Fig. 7. Genes sensitive to Foxp3 degradation in mature Tregs are potentially enriched for direct Foxp3 targets.
(a–b) Representative tracks showing Foxp3 ChIP, Treg H3K27Ac, Treg H3K27me3, and RNA-seq profiles in activated, resting, and nascent thymic Treg cells for candidate Foxp3-activated (a) and Foxp3-repressed (b) genes. (c) Representative flow cytometry plots and gating strategy for RNA-seq analysis of Treg “wannabe” cells with or without Foxp3 induction at the indicated time points following 4-OHT administration.
Extended Data Fig. 8
Extended Data Fig. 8. Conservation of Foxp3 degradation effects across different tissues.
(a) Experimental design of single-cell RNA-seq experiment. Each genotype consisted of four independent biological replicates. (b) Density contour plots of Foxp3AIDR26WT and Foxp3AIDR26TIR1(F74G) Treg cells from SLO, liver, lung and LILP overlayed on UMAP embeddings. (c) UMAP visualization of Treg cells from depicted tissues colored by “resting” and “activated” gene signature scores based on the scoring strategy from Fig. 3a. (d) Bar plots showing the number of DEGs between R26WT and R26TIR1(F74G) in resting and activated Treg cells from indicated tissues, colored by up- or downregulation. (e) Pairwise Pearson correlation coefficients for log2 fold changes of DEGs between activated and resting Treg cells from each tissue. Rows and columns are hierarchically clustered. (f) Heatmap of log2 fold changes in Foxp3 degradation DEGs separated by Treg cell activation status and tissue of origin. Genes are clustered by k-means clustering and cluster numbers are indicated by the color bars on the left. (g) Heatmap of average log2 fold changes for each k-means cluster in Treg cells separated by tissue and activation status. Rows and columns are hierarchically clustered. (h) Leiden clustering results visualized by UMAP for LILP Treg cells. Clusters are labeled as peripheral Treg cell (pTreg), dividing Treg cell, lymphoid tissue thymic Treg cell (LT-tTreg) and non-lymphoid tissue thymic Treg cell (NLT-tTreg) based on differences in their gene expression profile. (i) Heatmap of scaled mean UMI-normalized expression values of indicated genes in each cluster. (j) Number of DEGs between R26WT and R26TIR1(F74G) Treg cells in pTreg, LT-tTreg and NLT-tTreg cells, colored by up- or downregulation. (k) Log2 fold changes of “Foxp3-repressed” and “Foxp3-activated” genes in pTreg, LT-tTreg and NLT-tTreg cells. Each point represents a gene from the Foxp3-repressed or Foxp3-activated gene set identified in Fig. 2. Horizontal line marks the median and box marks the interquartile range. Mann-Whitney U test. (l) Scatter-plot of log2 fold changes of DEGs between pTreg and NLT-tTreg cell clusters (left-top), pTreg and LT-tTreg cell clusters (left-bottom) and LT-tTreg and NLT-tTreg cell clusters (right-top). Points are colored by their direction and populations in which they are altered. Correlations were calculated with Pearson correlation over all DEGs.
Extended Data Fig. 9
Extended Data Fig. 9. Foxp3 is preferentially required during Treg cell development.
(a) Pearson correlation between Foxp3 degradation-induced and Foxp3-dependent DEGs in thymic, resting, and activated Treg cells, limited to Foxp3-bound genes. (b) Meta-cell analysis of resting and activated Treg scRNA-seq data from secondary lymphoid organs following Foxp3 degradation, correlating Foxp3 expression levels with “TIR1-up” and “TIR1-down” gene signatures identified in Fig. 3. Dashed red line depicts line of best fit. Correlations and corresponding p-values were calculated with Pearson correlation over all genes. (c) Cytokine production by CD4⁺ T cells from neonatal Foxp3AID mice after 14 days of Foxp3 degradation, in comparison to age-matched Foxp3WT and Foxp3GFPKO mice. (d) Neutrophil expansion in adult and neonatal Foxp3AID mice following 14 days of Foxp3 degradation. Age-matched Foxp3WT and Foxp3GFPKO mice serve as controls for neonatal Foxp3AID mice. (e) Representative H&E staining and histology scores of skin inflammation in neonatal Foxp3AID mice after 14 days of Foxp3 degradation, in comparison to age-matched Foxp3WT and Foxp3GFPKO mice. (ce) Scatter plots represent mean ± SEM. Each point represents a unique mouse. Data are pooled from two independent experiments. One-way ANOVA. (f) Scatter plots of DESeq2 normalized counts of select DEGs from the neonatal Foxp3 degradation RNA-seq experiment (Fig. 5m). Each point represents a unique mouse. Two-sided t-test. (g-h) Representative plots (g) and combined data (h) showing Treg proliferation in 8-week-old adult versus 7-day-old neonatal mice measured by Ki67 positivity. Scatter-plot represents mean ± SEM. Data are combined from two independent experiments. Two-tailed t-test. Source data
Extended Data Fig. 10
Extended Data Fig. 10. Foxp3 degradation leads to tumor shrinkage with minimal adverse effects.
(a–b) Representative flow cytometry plots (a) and combined data (b) of Ki67 expression and EdU incorporation in Treg cells from the tumor and tumor-draining lymph node (dLN). Scatter-plot shows mean ± SEM. Each point represents a unique mouse. Data are representative of two independent experiments. Two-tailed t-test. (c) Representative flow cytometry plots (left) and combined data (right) of IFN-γ production by tumor-infiltrating NK cells. Scatter-plot shows mean ± SEM. Each point represents a unique mouse. Data are pooled from two independent experiments. Two-tailed t-test. (d) Representative flow cytometry plots (left) and quantification (right) of IFN-γ production by tumor-infiltrating ZsGreen CD4 T cells. Scatter-plot shows mean ± SEM. Each point represents a unique mouse. Data are pooled from two independent experiments. Two-tailed t-test. (e) Volcano plts showing the number of genes up- and downregulated in Foxp3AIDR26TIR1(F74G) tumor Treg cells within each clustered defined in Fig. 7(l). Source data

Update of

References

    1. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol.4, 330–336 (2003). - PubMed
    1. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science299, 1057–1061 (2003). - PubMed
    1. Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat. Immunol.4, 337–342 (2003). - PubMed
    1. Gavin, M. A. et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature445, 771–775 (2007). - PubMed
    1. Lin, W. et al. Regulatory T cell development in the absence of functional Foxp3. Nat. Immunol.8, 359–368 (2007). - PubMed

Substances

LinkOut - more resources