Abstract
RORγt is the lineage-specific transcription factor for T helper 17 (TH17) cells and an attractive drug target for treating TH17-associated diseases. Although the critical role of RORγt in early TH17 cell differentiation has been well recognized, its function in mature TH17 cell maintenance remains largely unknown. Here, we show that genetic deletion of Rorc in mature TH17 cells inhibited their pathogenic functions. Mechanistically, loss of RORγt led to a closed chromatin configuration at key TH17-specific gene loci, particularly at the “super-enhancer” regions. Unexpectedly, RORγt directly bound and inhibited Il4 transcription, whereas pharmaceutically or genetically targeting RORγt caused spontaneous conversion of TH17 cells to TH2-like cells in vitro and in vivo. Our results thus reveal dual crucial functions of RORγt in effector TH17 cells in maintaining TH17 cell program and constraining TH2 cell conversion, offering previously unidenified considerations in therapeutic targeting of RORγt.
RORγt maintains TH17 lineage identity by activating chromatins of super enhancers and constraining IL-4–dependent TH2 program.
INTRODUCTION
Upon activation, CD4+ T cells differentiate into different subsets in response to various innate signals, including T helper (TH) 1, TH2, TH17, regulatory T (Treg), and T follicular helper (Tfh) cells (1). TH17 cells are important in protecting against extracellular pathogens and maintaining mucosal homeostasis, while dysregulated TH17 cells contribute to chronic inflammation and autoimmune diseases. The development of TH17 cells is initiated by the combination of cytokines transforming growth factor–β (TGF-β) and interleukin-6 (IL-6), expanded by IL-21 at the middle stage, and further reinforced by IL-23 at the late stage, involving multiple layers of regulations by transcription factors, epigenetic factors, and environmental cues (2). Drugs targeting TH17 cell–associated factors, such as IL-23, IL-6R, IL-1, IL-17A, IL-17RA, IL-17F, and Janus kinase 1/3, have been proved for treating a number of diseases (3, 4).
Compared with TH1 and TH2 cells, TH17 cells, at least those generated in vitro, are regarded to be highly unstable and plastic, possibly because of their intrinsic stem-like gene expression features (5). Culturing mature TH17 cells under Treg-, TH1-, and TH2-polarizing conditions results in FOXP3, interferon-γ (IFN-γ), and IL-4 up-regulation, respectively (6). In vivo, TH17 cells were reported to convert into IFN-γ–producing T cells when transferred into lymphopenic mice or under inflammatory conditions such as experimental autoimmune encephalomyelitis (EAE), which was believed to be important for their pathogenicity (6–8). Meanwhile, TH17 cells were also reported to gain Tfh phenotype to drive immunoglobulin A (IgA)–producing germinal B cell production in Peyer’s patch (9), and an IL-17A–producing TH2 cell population was reported in patients with severe asthma (10).
TH lineage–specific transcription factors mediate T cell fate commitment and blockage of alternative cell fates. Retinoic acid related orphan receptor γt (RORγt), induced at the early stage of TH17 cell differentiation following IL-6 and TGF-β stimulation (11, 12), has a pivotal role at the lineage commitment stage of TH17 cells (11). RORγt specifically binds to and regulates TH17-associated genes, such as Il17a, Il17f, and Il23r, by activating their transcription in coordination with other transcription factors (13). Therefore, genetically or pharmacologically targeting RORγt by small molecules is effective in ameliorating TH17-related inflammatory diseases, including EAE, psoriasis, arthritis, colitis, and glomerulonephritis, particularly in preventative disease models (11, 14–16). However, none of the RORγt inhibitors has passed phase 2 studies in human clinical trials, raising concerns on the safety and efficiency of targeting RORγt (17).
Although the lineage-specific transcription factors are essential in T subset development, their function in mature T cells appears to be distinct. For instances, continuous FOXP3 expression is required for maintaining the lineage stability and suppressive function of mature Treg cells (18). However, T-box transcription factor (T-bet) has a limited function in maintaining IFN-γ expression in mature TH1 cells (19, 20). GATA3 maintains IL-5 and IL-13 but is less important for IL-4 expression in mature TH2 cells (20, 21). Compared with the well-determined function of RORγt in TH17 cell differentiation, its role in maintaining mature TH17 program is not fully understood.
In this study, we found that mature TH17 cells require RORγt expression to maintain their lineage identity and pathogenicity, at least partly via regulating chromatin accessibility of TH17-associated super enhancers (SEs). Unexpectedly, we found a critical role of RORγt in direct inhibition of IL-4 expression and thus IL-4–dependent TH2 program. These data offer important insights into TH17 cell biology with implications in treating TH17-related diseases.
RESULTS
RORγt is required for the maintenance and function of effector TH17 cells
To investigate the function of RORγt in mature TH17 cells, we first established a Rorc-inducible deletion system by crossing Rorcfl/fl mice wild type (WT) with Rosa26-CreERT2 mice (hereafter as RorcERT2). Naïve CD4+ T cells from these mice were isolated and then cultured under TH17-polarizing condition. Three days later, when TH17-signature cytokine IL-17A was fully induced, 4-hydroxytamoxifen (4-OHT) was added into the culture to delete the Rorc gene (fig. S1A). The deletion efficiency was confirmed by RORγt staining (fig. S1B). Similar to its deletion in naïve CD4+ T cells (11), loss of Rorc in mature TH17 cells resulted in a progressive loss of IL-17A protein expression and a marked reduction of the mRNA expression of Il17a, Il17f, and Il23r without affecting the transcription of other key TH17-associated transcription factors, including Rora, Irf4, and Batf (Fig. 1, A and B).
Fig. 1. Continuous RORγt expression maintains TH17 cell lineage stability and function.
(A and B) Naïve T cells isolated from Rorcfl/fl (WT) and Rosa26-CreERT2 Rorcfl/fl (RorcERT2) mice were cultured with TGF-β, IL-6, IL-1β, and IL-23 for 3 days, followed by 4-OHT (2 μM) treatment. (A) Representative flow cytometry plots of IL-17A and IFN-γ expression (left) and statistical analysis of IL-17A expression (right) before (0 hours), 48 hours, or 96 hours after 4-OHT treatment. Each line represents one independent experiment. (B) Relative mRNA expression of TH17-related genes 96 hours after 4-OHT treatment. The expressions were normalized to Actb. (C to E) 2d2-WT and 2d2-RorcERT2 naïve T cells were polarized under TH17 culture condition for 3 days. RFP+ mature TH17 cells were sorted and treated with 4-OHT and then transferred into Rag1−/− mice to induce EAE. n = 5 mice each group. (C) EAE disease scores following MOG35–55 immunization. (D) Total number of CD4+ T cells infiltrated in the central nervous system of the recipient mice. (E) Representative flow cytometry plots and statistic data of CCR6, IL-17A, and IFN-γ expression in the CNS-infiltrated T cells. This experiment was repeated twice. P values were determined by paired Student’s t test (A), one-way ANOVA with Tukey’s multiple comparisons test (C), or unpaired Student’s t test (D and E). Data represent means ± SEM (C) or SD. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001.
To examine whether RORγt is required for maintaining TH17 effector function in vivo, RorcERT2 or Rorcfl/fl mice were crossed with 2d2–T cell receptor (TCR) transgenic mice and Il17f–red fluorescent protein (RFP) reporter mice (hereafter as 2d2-RorcERT2 or 2d2-WT). 2d2-WT and 2d2-RorcERT2cre naïve T cells were cultured under TH17 polarization condition in vitro. RFP+ mature TH17 cells were then sorted, treated with 4-OHT to induce Rorc deletion, and then transferred into Rag1−/− recipient mice to induce EAE (fig. S1C). Deleting Rorc in effector TH17 cells significantly delayed the onset and reduced the severity of EAE disease (Fig. 1C). We further analyzed central nervous system (CNS)–infiltrating CD4+ T cells. RORγt deletion was validated in 2d2-RorcERT2 donor T cells in vivo (fig. S1D). Compared to 2d2-WT T cells, CNS-infiltrating 2d2-RorcERT2 T cell numbers were significantly decreased, with decreased expression of CCR6 and IL-17A, although similar low IL-17A+IFN-γ+ population was found (Fig. 1, D and E). In conclusion, RORγt is critically required for maintaining the lineage identity and pathogenic function of effector TH17 cells.
Loss of RORγt alters epigenetic landscapes in effector TH17 cells.
Epigenetic mechanisms are critical in TH cell lineage specification. To understand how RORγt maintains TH17 cell program, we examined histone modification markers at the Il17 gene locus in 4-OHT–treated mature WT and RorcERT2 TH17 cells. Unlike its deficiency before the initiation of TH17 differentiation (22), Rorc deletion in mature TH17 cells barely affected the active histone marker H3K4me3 and the silence marker H3K27me3 at both the promoter and CNS2 enhancer region within the Il17-Il17f gene locus (Fig. 2A) but notably decreased the active and poised enhancer markers H3K27Ac and H3K4me1 (Fig. 2B), suggesting a selective role of RORγt in maintaining TH17-specific enhancer activities. Consistently, 5-hydroxymethylcytosine (5hmc) modification, a marker enriched at active enhancer regions in effector T cells (23), was also reduced at both the Il17 promoter and CNS2 regions in RorcERT2 TH17 cells followed by 4-OHT treatment (Fig. 2C).
Fig. 2. RORγt regulates SE activities in mature TH17 cells.
Naïve T cells isolated from WT and RorcERT2 mice were cultured under TH17-polarizing condition for 3 days followed by 4-OHT treatment for 96 hours. SE and traditional enhancer (TE) regions in TH17 cells were defined from previous study (26). (A to C) ChIP assay was performed with antibodies targeting H3K4me3 and H3K27me3 (A), H3K27Ac and H3K4me1 (B), and MeDIP assay were performed with antibody targeting 5hmc (C). The enriched DNA fragments were detected using qPCR and normalized with input control at Il17a promoter and CNS2 region. The experiments were repeated three times. (D and E) WT and RorcERT2 TH17 cells were restimulated with anti-CD3 and collected for ATAC-seq. (D) Volcano plot of differentially opened chromatin regions (OCRs). (E) Representative ATAC-seq tracks from WT and RorcERT2 TH17 cells. (F) The Venn diagram of previous reported RORγt-binding peaks, TEs, and SEs (13, 26). (G) The Venn diagram of ATAC-seq OCRs, promoter regions, SEs, and TEs (26). (H) The Venn diagram of reported RORγt-regulated p300-binding peaks (defined by differential binding of p300 in WT and RORγt-deficient TH17 cells), TEs, and SEs (13, 26). KO, knockout.
To further understand the role of RORγt in mature TH17 cells, we examined the effect of RORγt deficiency on genome-wide chromatin accessibility, especially in enhancer regions. ATAC-seq (assay for transposase-accessible chromatin sequencing) results revealed a total of 1249 and 1871 regions with reduced (RORγt-activated chromatins) or increased chromatin accessibility (RORγt-inhibited chromatins), respectively, following Rorc deletion in effector TH17 cells (Fig. 2D). The chromatin accessibility of the promoter regions of TH17 cell signature genes Il17a, Il17f, Il23r, and Ccr6 were notably decreased and conserved noncoding DNA regulatory regions, including the Il17-Il17f enhancer CNS2 (24) and the CNS6, CNS9, and CNS + 10 regions at the Rorc gene locus (Fig. 2E) (12, 25).
SEs are a subset of enhancers important in regulating core cell identity gene expression compared with traditional enhancers (TEs) (26). A total of 774 TH17 cell–associated SEs have been defined on the basis of exceptionally high occupancy of p300 compared with TEs. Among them, 99.44% (731 of 774) of TH17 cell–related SEs were colocalized with RORγt-binding sites, in contrast to only 40.80% (8117 of 19,893) of TH17 cell–related TEs (Fig. 2F) (13). On the basis of our ATAC-seq data, after Rorc deletion, 55.6% (1736 of 3120) of the differentially opened chromatin regions fell in enhancer regions, among which there were 41.5% SEs (321 of 774, affected/total) and 7.2% TEs (1437 to 19,893) affected (Fig. 2G). In line with these findings, reanalysis of previously published chromatin immunoprecipitation sequencing (ChIP-seq) data in WT and Rorc-deficient TH17 cells revealed that deletion of Rorc reduced p300 binding to 54.5% (422 to 774) SEs but only 8.02% (1596 to 19,893) in TEs (Fig. 2H) (13). These findings together highlight a particular important role of RORγt in regulating TH17-specific enhancers, especially SEs.
Loss of RORγt results in conversion of TH17 cells to TH2-like cells
Compared with other helper T cells, TH17 cells are highly plastic and prone to be converted to IFN-γ–producing TH1-like cells under both in vitro and in vivo conditions (7). Deletion of Rorc in effector TH17 cells, however, caused significant up-regulation of IL-4 and GATA3 (Fig. 3A), but not IL-9 and IFN-γ expression, under continuous TH17-polarizing culture condition (Fig. 1A and fig. S2A), although the expression of IL-4 and GATA3 was lower than that under TH2 culture condition (Fig. 3A). The transcripts for TH2 signature genes Il4, Il13, and Gata3 were also significantly increased in RorcERT2 compared with WT TH17 cells, while Ifng transcription was only slightly affected (fig. S2B). The mRNA and protein expression of IL-5 was barely detectable in either WT or RorcERT2 cells, suggestive of a potentially different regulatory mechanism (fig. S2C). To prove that the increased IL-4–expressing cells indeed are derived from ex-TH17 cells, we sorted Il17f-RFP+ WT and RorcERT2 TH17 cells and then recultured these developed TH17 cells in the presence of 4-OHT under TH17-polarizing condition. Consistently, IL-4 expression increased on day 4 after reculture and further elevated to a comparable level in TH2 cells on day 7 (fig. S2D). Deletion of Rorc in naïve CD4+ T cells (Cd4CreRorcfl/fl, hereafter as RorcCd4) also led to increased differentiation toward to IL-4–expressing TH2 cells, but not IFN-γ–producing TH1 phenotype, under TH17-polarizing condition (Fig. 3, B and C).
Fig. 3. Genetic or pharmaceutical inhibition RORγt converts TH17 cells into TH2-like cell.
(A) Naïve T cells isolated from WT and RorcERT2 mice were cultured under TH17-polarizing condition. After 3 days, the developed TH17 cells were recultured with 4-OHT under TH17- or TH2-polarizing (TH17-TH2) conditions for 96 hours. Representative flow cytometry plots of IL-4 and GATA3 were shown. (B and C) Intracellular staining of IL-4 and GATA3 (B), IFN-γ, and IL-17A (C) in WT and RorcCd4 TH17 cells cultured in vitro for 6 days. (D) Naïve T cells from WT and RorcERT2 mice and cultured with TGFβ, IL-1β, IL-6, and IL-23 for 3 days. The effector TH17 cells were then treated with 4-OHT for 2 days to remove the Rorc gene and then recultured with IL-12 and IFN-γ, with or without IL-4–neutralizing antibody for 4 days. IFN-γ and IL-4 expression was detected by flow cytometry. (E) Expression of the Il4-GFP reporter in TH17 cells induced in vitro in the presence of dimethyl sulfoxide (DMSO) or RORγt inhibitors GSK805 (0.5 μM), TMP920 (10 μM), or ursolic acid (UA) (2 μM) for 7 or 14 days. (F) Expression of the Il4-GFP reporter in TH17 cells induced in vitro with the presence of control DMSO or RORγt inhibitors from days 7 to 14 in the culture. All these experiments were repeated at least two to three times with consistent results.
When placed under TH2 culture conditions, Rorc-deficient TH17 cells exhibited augmented conversion toward TH2 cells, although less notably than under TH17-polarizing conditions (fig. S3, A and B). Consistent with previous findings that forced expression of RORγt had no effect on TGF-β–induced FOXP3 expression (27), deletion of Rorc did not promote TH17 conversion to FOXP3+ Treg cells (fig. S3B). In contrast, Rorc-deficient TH17 cells were much more resistant to be converted to an IFN-γ–producing TH1 cell fate when recultured under various TH1- or TH1/TH17-polarizing conditions compared with WT TH17 cells (fig. S3B). Unexpectedly, TH17 cells produced large amounts of IL-4, but not IFN-γ, when recultured under TH1 culture condition, and blockage of IL-4 significantly enhanced IFN-γ production in both WT and Rorc-deficient cells (Fig. 3D), suggesting intrinsic TH2-prone feature of mature TH17 cells in vitro.
To further substantiate the above findings, we isolated naïve CD4+ T cells from Il4GFP reporter mice and cultured them under TH17 cell–polarizing conditions in the presence of different RORγt inhibitors, including GSK805, TMP920, and ursolic acid. Consistent with above results, treatment with RORγt inhibitors significantly increased IL-4–green fluorescent protein (GFP) reporter expression 1 week later. In the prolonged culture, treatment with GSK805 or TMP920 even transformed ~80% of the cells to IL-4–producing TH2-like cells (Fig. 3E). Similar to genetic ablation of Rorc in effector TH17 cells, culturing these cells in the presence of RORγt inhibitors also led to a notable up-regulation of IL-4 from days 7 to 14 of TH17 cultures (Fig. 3F). In conclusion, these findings reveal a previously unidentified function of RORγt in repressing TH2 cell program in TH17 cells; inhibiting the expression or function of RORγt leads to spontaneous conversion of TH17 cells to TH2-like cells.
Loss of RORγt reprograms TH17 cells toward TH2 transcriptional and epigenetic programs
To further understand the function of RORγt in mature TH17 cells, RNA sequencing (RNA-seq) analysis was performed using mature WT and RorcERT2 TH17 cells treated with 4-OHT for 48 or 96 hours. A total of 140 genes were up-regulated, 75 genes were down-regulated in RorcERT2 TH17 cells compared with WT group treated with 4-OHT for 48 hours [fold changes of >2, false discovery rate (FDR) < 0.05, FPKM (fragments per kilobase of transcript per million fragments mapped) > 1 in any group], and the numbers of genes with altered expression were increased to 283 and 202 at 96 hours after 4-OHT treatment, respectively (Fig. 4A). Most of the RORγt-regulated genes showed a progressively increased trend of changes from 48 to 96 hours after inducing Rorc deletion (Fig. 4B).
Fig. 4. Loss of RORγt reprograms TH17 cells to a TH2-like transcriptional and epigenetic landscape.
WT and RorcERT2 TH17 cells treated with 4-OHT for 48 or 96 hours were collected for RNA-seq. (A) The volcano plots and (B) heatmap of DEGs (FPKM > 1 in any group). (C) GSEA analysis of TH1, TH2, TH17, and Treg signature gene sets (28) enrichment in WT and RorcERT2 TH17 cells. NES, normalized enrichment score; FDR, false discovery rate. (D) Heatmap of selected T subset signature genes expressed in WT and RorcERT2 TH17 cells. (E) Venn diagram of the RORγt inhibited or activated genes in the ATAC-seq data in Fig. 2D versus TH2 signature genes. (F) ATAC-seq peaks at the Il4-Il13 and Gata3 gene loci. (G) Enrichment of TF binding motifs in the ATAC-seq peaks in Fig. 2D.
To understand the effects of Rorc deletion on TH17 cell plasticity, we first defined a core set of signature genes specifically highly expressed in TH1, TH2, TH17, or Treg cells based on previously published transcriptional datasets (Materials and Methods) (28). With these TH-specific gene lists, we did gene set enrichment analysis (GSEA) analysis on the RNA-seq data obtained from WT and RorcERT2 TH17 cells treated with 4-OHT (Fig. 4A). Deletion of Rorc in mature TH17 cells significantly decreased the expression of TH17 signature genes and caused a significant up-regulation of TH2 signature genes but with a limited effect on TH1 and Treg signature genes. The trend of TH17 to TH2 conversion was progressive following RORγt deletion (Fig. 4, C and D).
To understand the epigenetic regulation by RORγt, we overlapped TH2 signature genes with chromatin regions activated or inhibited by loss of RORγt in our ATAC-seq data (Fig. 2D) and found that RORγt deficiency promoted chromatin accessibilities in 93 of total 709 TH2 signature genes, including Il4 and Gata3 (Fig. 4E). The chromatin accessibility at multiple regulatory regions at the Il4-Il13 gene locus was significantly increased, including the Il4 promoter, CNS2, HS2, and RHS6 regions, as well as the Gata3 gene locus, but not key TH1 or Treg signature genes, such as Tbx21, Foxp3, and Il10 (Fig. 4F and fig. S4). Consistently, in the transcription factor binding motif analysis, deficiency of RORγt caused a significant decrease in the binding motifs enriched for TH17-specific transcription factors, including signal transducers and activators of transcription 3 (STAT3) and RORα, as well as RORγt, but a concomitant increase in binding motifs for TH2 signature transcription factors GATA3 and STAT6 (Fig. 4G). Together, these findings indicate a specific role for RORγt in constraining TH2 cell program via epigenetic mechanisms.
RORγt antagonizes TH2 cell program through inhibiting IL-4 expression
The above results reveal a critical function of RORγt in restricting the expression of IL-4, directly or indirectly. Of note, addition of IL-4–blocking antibodies in the culture medium of 4-OHT–treated TH17 cells effectively blocked conversion of Rorc-deficient TH17 cells into IL-4– and GATA3-producing TH2-like cells (Fig. 5A), highlighting IL-4 as an obligatory factor in TH2 cell conversion by RORγt-deficient TH17 cells.
Fig. 5. RORγt prevents TH17 to TH2 conversion through inhibition of IL-4 expression.
(A) Intracellular staining of IL-4 and GATA3 in WT and RorcERT2 TH17 cells treated with 4-OHT for 96 hours in the presence or absence of IL-4–blocking antibody. (B) The ChIP assay was performed with antibodies against H3K4me3, H3K27me3, and H3K27Ac in 4-OHT–treated WT and RorcERT2 TH17 cells. The enrichment of Il4 promoter in ChIPed DNA was examined by qPCR and normalized against total input DNA. (C) TH2 cells transfected with the RVKM or RVKM-RORγt plasmids were used for ChIP assays using antibodies targeting H3K4me3 and H3K27Ac (left) or relative mRNA expression of Gata3 and Il4 (right). (D) Anti-HA ChIP assay in TH17 cells transfected with empty or RORγt-HA–containing plasmid. (E) The luciferase activity of Il4 promoter in 293T or EL4 cells transfected with empty or RORγt-containing plasmid. All the experiments were repeated at least two to three times with consistent results. Data represent means ± SD. P values were determined by unpaired Student’s t test with n = 3 replicates. ****P < 0.0001.
Therefore, we focused on investigating how RORγt regulates IL-4 expression. We first tested whether RORγt deficiency could affect the epigenetic status of the Il4 gene locus. Consistent to its function in suppressing IL-4 expression, deletion of Rorc in mature TH17 cells significantly enhanced permissive histone markers H3K27Ac and H3K4me3 levels at the Il4 promoter locus without affecting the nonpermissive histone marker H3K27me3 (Fig. 5B). Conversely, overexpression of RORγt in TH2 cells not only inhibited active histone modifications, including H3K4me3 and H3K27Ac, but also led to a marked reduction of Il4 expression (Fig. 5C). However, expression of Gata3 in TH2 cells was not affected by RORγt overexpression (Fig. 5C), suggesting Il4 rather than Gata3 as a potential direct target for RORγt in suppressing TH2 cell program.
To test this possibility, a hemagglutinin (HA)–tagged RORγt was retrovirally overexpressed in TH17 cells, which were then subjected to ChIP–polymerase chain reaction (PCR) assays by using control IgG and anti-HA antibodies. Compared to control IgG, anti-HA antibody detected significant DNA binding by RORγt at the Il4 promoter, supporting a direct binding by RORγt (Fig. 5D). For further validation, a dual luciferase reporter gene assay was performed using the Il4p-PGL3 reporter plasmid in 293T, a human kidney cell line, and EL4, a mouse lymphoma–derived T cell line. In either cell line, overexpression of RORγt led to ~50% reduction of the Il4 promoter activity (Fig. 5E), suggesting a direct inhibitory effect of RORγt on Il4 gene transcription. Together, our findings demonstrated a critical role of RORγt in maintaining TH17 lineage stability through a direct inhibitory effect on Il4 transcription.
Loss of RORγt in effector TH17 cells leads to TH2 conversion in vivo
The above studies indicate that RORγt deficiency in effector TH17 cells resulted in TH17 to TH2 conversion in vitro. To examine this under in vivo settings, in vitro–induced RFP+ 2d2-WT and 2d2-RorcERT2 TH17 cells were sorted, treated with 4-OHT, and then transferred into CD45.1-recipient mice followed by EAE induction (procedure as in fig. S1C). The recipient mice were euthanized and analyzed when they developed obvious disease symptoms. Compared with 2d2-WT TH17 cells, the adoptively transferred 2d2-RorcERT2 TH17 cells in the CNS expressed significantly reduced levels of IL-17A but showed two to threefold increase in the percentages of IL-4– and IL-13–producing cells (Fig. 6A). When IL-4 was neutralized, EAE disease severity was significantly increased in Rag1−/−-recipient mice receiving 2d2-RorcERT2 TH17 cells compared with control treatment group or even the mice receiving 2d2-WT cells (Fig. 6B), suggesting a protective role of IL-4 in the EAE model, which is consistent with previous work (29).
Fig. 6. RORγt deficiency converts TH17 cells toward TH2-like cells under inflammatory conditions.
(A) CD45.1 mice were subjected to EAE induction after receiving 4-OHT–treated 2d2-WT or 2d2-RorcERT2 TH17 cells and euthanized after developing apparent disease symptoms. The expression of IL-17A, IL-4, and IL-13 were analyzed by flow cytometry in CNS-infiltrated donor cells. (B) Rag1−/− mice were subjected to EAE induction after receiving 4-OHT–treated 2d2-WT or 2d2-RorcERT2 TH17 cells. IL-4–neutralizing antibody or PBS was intraperitoneally injected every 2 days on day 3 after MOG35–55 immunization. The disease scores were monitored daily. (C and D) Naïve T cells from WT or RorcCd4 mice were transferred into Tcrbd−/− recipients for colitis induction and analyzed 6 weeks later when recipients developed severe symptoms. (C) The body weight (B.W.) was monitored weekly. (D) Indicated cytokines were analyzed in gut-infiltrated CD4+ T cells. FSC-H, forward scatter height. The above experiments were repeated twice. P values were determined by two-way ANOVA with Tukey’s multiple comparisons test (B and C) or unpaired Student’s t test (A and D). Data were shown as means ± SEM (B and C) or SD (A and D). *P < 0.05, **P < 0.01, and ***P < 0.001. (E to G) RNA-seq analysis of gut-infiltrated CD4+ T cells in the colitis model. (E) Volcano plot of the RNA-seq (FPKM > 1 in any group). (F) Venn diagram of DEGs in colitis and in vitro 96-hour RNA-seq data in Fig. 4A. (G) GSEA analysis of TH1, TH2, TH17, and Treg signature genes (28) enrichment in WT and RorcCd4 CD4+ T cells.
To substantiate the above findings, we isolated naïve T cells from WT or RorcCd4 mice and transferred them into Tcrbd−/−-recipient mice to induce colitis. Mice receiving RorcCd4 T cells were largely resistant to colitis-induced weight loss compared to those receiving WT T cells (Fig. 6C). Consistent with the in vitro findings, the expression level of IL-4 was significantly increased, while IL-17A expression was nearly abolished in Rorc-deficient T cells, but the expression of IL-13 was comparable among two groups of mice (Fig. 6D), suggesting a specific repression of IL-4 expression by RORγt.
We also isolated gut-infiltrating CD4+ T cells 6 weeks later for RNA-seq. A total of 146 protein coding genes were down-regulated after Rorc deletion, while 133 genes up-regulated (Fig. 6E). Consistently, the GSEA analysis showed that TH2 signature genes also were significantly enriched in RORγt-deficient group, while TH1 and Treg signatures enrichment was comparable among two groups (Fig. 6F). We further overlapped RORγt-regulated differentially expressed genes (DEGs) in vivo and in vitro and found that 17 genes were consistently suppressed by RORγt, including type 2 signatures Il4, Il13, and Il3, and 20 genes were up-regulated by RORγt, including TH17 signature genes Il17a, Il17f, Ccl20, Ccr6, and Il23r (Fig. 6G). In summary, these findings together support an essential role of RORγt in maintaining the in vivo stability and function of TH17 cells, partially through inhibiting TH2 cell differentiation program.
DISCUSSION
In this study, the function of RORγt in mature TH17 cells was systematically examined by genetic and pharmacological approaches. Deficiency of RORγt abolished IL-17A expression in effector TH17 cells and caused ameliorated EAE disease. However, targeting RORγt led to a significant and spontaneous conversion of TH17 cells to TH2-like cells under both in vitro and in vivo settings, dependent on IL-4.
In differentiating TH17 cells, a major function of RORγt is to bind and activate canonical TH17 signature genes, including Il17a, Il17f, Ccr6, and Il23r, which are also regulated by RORγt in mature TH17 cells. Previous reports suggest a function of RORγt in regulating chromatin structures possibly through regulating histone modifications as its deficiency significantly reduces H3K4me3 but alters H3K27me3 modification at the Il17-17f gene locus (22, 24). In mature TH17 cells, however, loss of RORγt specifically reduces active histone markers H3K4me1 and H3K27Ac at the Il17-Il17f gene locus without affecting H3K4me3 and H3K27me3 (Fig. 2, A and B), indicating a specific function of RORγt in activating SEs. Compared with STAT3, interferon regulatory factor 4, and BATF (basic leucine zipper ATF-like transcription factor), it is shown that RORγt has a limited function in recruiting p300 and therefore a limited role in activating enhancers (13). Consistently, less than 10% of TEs show altered chromatin accessibility caused by RORγt deficiency based on ATAC-seq data in this study. In contrast, more than 50% SEs are affected and regulated by RORγt (Fig. 2G), a function not yet reported. Considering the importance of enhancers especially SEs in cell fate determination, our findings suggest that the lineage stability of mature TH17 cells are at least partially sustained by a direct binding and regulation of SEs by RORγt.
Compared to other helper T cells, TH17 cells were regarded with a strong stem-like gene expression features, with significant plasticity (5, 30). Perhaps for this reason, TH17 cells had been miscategorized as a subset of TH1 cells for almost 20 years because these cells are destined to become IFN-γ+ TH1-like cells in most cases under in vivo settings (8). RORγt may affect (15) or not affect (11, 31–33) TH1 cell differentiation in vivo, but its overexpression (34) or loss of function (35) had little effect on IFN-γ production under in vitro TH17 culture conditions, suggesting that RORγt may not directly inhibit TH1 cell differentiation. This idea is also confirmed by our data (Fig. 1, A and C). Of note, our work has revealed a potent role of RORγt in restricting TH2 cell program under both in vitro and in vivo conditions through direct inhibition of Il4 expression. In mature TH17 cells, deletion of Rorc followed by four more days of culture under TH17-polarizing condition caused ~10- to 100-fold increase in Il4 and Gata3 mRNA expression, although their protein expression was increased less markedly. A possible reason for this difference is that TH2 cytokines are notorious to be stained in C57BL/6 mice, and it is also possible that the process of TH17 to TH2 conversion takes time. As an alternative strategy to confirm this finding, we repeated the experiment using T cells isolated from the Il4-GFP reporter mice and found that treatment with RORγt inhibitors for 7 days resulted in 13 to 28% GFP expression in mature TH17 cells (Fig. 3F), and treatment with RORγt inhibitors throughout the 14 days of TH17 culture led to more than 80% of cells converted to Il4-GFP+ cells (Fig. 3E). These data, combined with 48- and 96-hour RNA-seq data of 4-OHT–treated RorcERT2 TH17 cells shown in Fig. 4B, suggest that the conversion of TH17- to TH2-like cells is also a gradual process.
When recultured under TH1 culture condition (Fig. 3D), copious amounts of IL-4 were produced in both WT and Rorc-deficient TH17 cells, which, in cell percentages, were even more than IFN-γ expression. This was probably caused by a rapid loss of RORγt that consequently led to a derepression of IL-4 production in the absence of TH17-maintaining cytokines. In addition, genetic ablation of Rorc also significantly increased IL-4 expression in T cells in vivo in both EAE and colitis models. Considering that TH17 to TH2 conversion occurs at conditions that not only lack exogenous TH2-polarizing cytokines but are also unfavorable for TH2 responses, including TH17, TH1, and medium-only culture conditions, our results thus highlight an intrinsic TH2-prone property of TH17 cells, particularly at conditions with destabilized RORγt-directed TH17 program.
Because of its critical role in controlling TH17 cell differentiation and function and also its structural advantages as ligand-activated nuclear receptor, more than 10 small molecular inhibitors for RORγt have been developed and entered clinical trials for treatment of TH17-associated autoimmune diseases, but so far, none of these drugs have successfully passed phase 2 studies, although targeting IL-17 is highly successful in treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis. In this study, our results showed a specific function of RORγt in suppressing IL-4 production in TH17 cells, and targeting RORγt, either genetically or pharmacologically, elicited a strong TH2-biased immune response, which was proved to be beneficial in the EAE model in this study. However, IL-4 blockage had no effect in the T cell transfer colitis model, suggesting that the consequence of TH17 to TH2 conversion in different TH17-associated diseases can be variable. Considering the critical pathogenic role of IL-4 in type II immune response–associated diseases, it is possible that clinical application of RORγt inhibitors may potentially convert the TH17-associated inflammation toward TH2-biased immunopathology under certain circumstances, such as in treatment of psoriasis, which needs to be further cautioned.
In summary, our data demonstrate a necessary and essential role of RORγt in maintaining the lineage stability and pathogenicity of mature TH17 cells, whereas loss of RORγt not only destabilizes TH17 cell program but also causes a spontaneous conversion of TH17 cells to IL-4–producing TH2-like cells. The findings in this study are not only useful for understanding the role of RORγt in TH17 cells but also helpful for development of RORγt inhibitors in the clinic.
MATERIALS AND METHODS
Mice
Rorcfl/fl (36) mice were crossed Il17fRFP (37), Rosa26-CreERT2, or Cd4cre (38) mice to generate conditional knockout mice as indicated. The C57BL/6J, CD45.1, Rag1−/−, Tcrbd−/−, Il4GFP, Rosa26-CreERT2, and 2d2 TCR transgenic mice were obtained from the Jackson Laboratory. All the mice were bred and maintained at specific pathogen–free condition in the animal facility of Tsinghua University. Six- to eight-week-old and sex-matched mice were used for all experiments. Animal experiments were performed according to the protocols approved by the Institutional Animal Care and Use Committee.
In vitro T cell culture
CD4+CD25−CD62LhiCD44low naïve T cells were isolated from the spleen and peripheral lymph nodes of indicated mice and activated by plate-bound anti-CD3 and anti-CD28 (5 μg/ml). TH17 cells were induced with the combination of human TGF-β (1 ng/ml), mouse interleukin 6 (mIL-6) (20 ng/ml), mIL-1β (10 ng/ml), and IL-23 (25 ng/ml). To analyze the maintenance of TH17 cells, 3 days later, the developed TH17 cells were restimulated with anti-CD3/28 and treated with 2 μM 4-OHT (Sigma-Aldrich) to induce Rorc deletion under TH17-polarizing condition. About 48 or 96 hours later, the cells were collected for further analysis. To analyze the plasticity, the developed TH17 cells were treated with 2 μM 4-OHT for 2 days, collected, and restimulated with anti-CD3/28 and cultured under various conditions. TH1 or TH1/17 cells were induced by the combination of mIL-12 (20 ng/ml), mouse interferon gamma (mIFN-γ) (10 ng/ml), and IL-23 (25 ng/ml) as indicated. TH2 cells were induced by IL-4 (20 ng/ml). Treg cells were induced by mIL-2 (10 U/ml) and human TGF-β (2 ng/ml). Four days later, the cells were collected for further analysis. Anti-CD3 and anti-CD28 antibodies were purchased from Bio X Cell, IL-23 and human TGF-β were from R&D System, and other cytokines were from PeproTech.
T cell transfer colitis induction
CD4+CD25−CD62LhiCD44low naïve T cells were isolated from the spleen and peripheral lymph nodes of Rorcfl/fl and Cd4creRorcfl/fl mice and were intravenously injected into age-matched male Tcrbd−/−-recipient mice. The body weight of the recipient mice was monitored weekly. The mice were euthanized about 6 weeks later when WT group developed obvious clinical symptoms, and the lamina propria lymphocytes from large intestines were isolated for further analysis.
EAE induction
2d2 Rorcfl/fl Il17fRFP (2d2-WT) or 2d2 Rosa26-CreERT2 Rorcfl/fl Il17fRFP (2d2-RorcERT2cre) naïve T cells were isolated and cultured under TH17 cell–polarizing condition for 3 days. RFP+ mature TH17 cells were sorted and then treated with 4-OHT for 2 days. Treated TH17 cells (~1 million) were intravenously injected into female Rag1−/−-recipient mice followed by EAE induction. The mice were immunized (subcutaneous injection on both dorsal sides) with 150 μg of myelin oligodendrocyte glycoprotein (MOG)35–55 peptide in complete Freund’s adjuvant (Sigma-Aldrich) containing Mycobacterium tuberculosis (5 mg/ml; BD Biosciences) following intraperitoneal injection of 500 ng of pertussis toxin in 1× phosphate-buffered saline (PBS) on the second day. IL-4 neutralizing antibody (1.5 mg) or control 1× PBS was intraperitoneally injected every 2 days from the third day after MOG immunization. The disease scores were measured daily: 0, none; 1, limp tail or waddling gait with tail tonicity; 2, wobbly gait; 3, hindlimb paralysis; 4, hindlimb and forelimb paralysis; 5, death. The mice were euthanized after developing symptoms of EAE diseases, and the brain and spinal cord were collected for further analysis.
Flow cytometry
Single-cell suspensions were first stained with viability and surface marker antibodies. For intracellular staining of cytokines, both in vitro–cultured or in vivo–isolated cells were restimulated with phorbol 12-myristate 13-acetate (PMA; 50 ng/ml; Sigma-Aldrich) and ionomycin (500 ng/ml; Sigma-Aldrich) in the presence of GolgiStop (BD Biosciences) for 5 hours before surface staining and followed by fixation and permeabilization according to the manufacturer’s protocol. Cells were analyzed using LSRFortessa (BD Biosciences) flow cytometer and FlowJo X software. Dead cells were excluded on the basis of viability dye staining (Fixable Viability Dye eF506, eBioscience).
ChIP–quantitative PCR
In vitro–cultured cells were cross-linked by 1% paraformaldehyde for 10 min at room temperature with gently shaking and were stopped by 100 mM glycine. The ChIP experiment was performed following the instructions of ChIP-IT Express ChIP kits (Active Motif). Antibodies, including anti-rabbit IgG [2729, Cell Signaling Technology (CST)], anti-HA (3724, CST), anti-H3K4me3 (07-473, Millipore), anti-H3K27me3 (07-499, Millipore), anti-H3K27Ac (39133, Active Motif), anti-H3K4me1 (39297, Active Motif), and Dynabeads protein A and G (Life Technologies) were used for IPs. The precipitated DNA was quantified by real-time PCR and normalized on the basis of total input DNA.
Methylated DNA immunoprecipitation sequencing (MeDIP)-qPCR
The MeDIP experiment was performed as previously described (23). Briefly, genomic DNA was extracted and sonicated into 200– to 500–base pair fragments. The DNA fragments were then denatured and incubated with antibody against 5hmC (39791, Active Motif) or rabbit IgG for IPs. The precipitated DNA was quantified by real-time PCR and normalized on the basis of input DNA.
Dual-luciferase reporter assay
The Il4 promoter region (2 kb from transcription start site) was cloned into pGL3 luciferase reporter plasmid as previously described methods (24). The RORγt expression plasmids were cloned previously (39). The 293T or EL4 cells were transfected with indicated vectors together with a Renilla luciferase reporter plasmid. The promoter activity was further measured by Dual-Luciferase Reporter Assay System (Promega). The transfection efficiency and luciferase activity were normalized based on Renilla luciferase.
RNA-seq and analysis
In vitro–cultured cells were restimulated with plate bound anti-CD3 for 4 hours before RNA extraction. The gut-isolated CD4+ T cells were restimulated by PMA and ionomycin for 2 hours before RNA extraction. Total RNA was extracted with TRIzol (Life Technologies) according to the manufacturer’s instructions, and the RNA-seq library was constructed and sequenced by BGI Genomics. Low-quality reads and adaptor sequences were removed by Trim Galore v0.4.4. The clean reads were aligned to mm10 by Bowtie 2 with default parameter, and the uniquely mapping reads were summarized by featureCounts (from Subread package). DEGs are identified by DESeq2 using at least two fold change and FDR-adjusted P value of 0.05.
Gene set enrichment analysis
The TH17/TH1/TH2/Treg signature gene lists were generated on the basis of published microarray datasets GSE14308 (28). The most highly expressed genes in TH17 cells compared with TH1, TH2, Treg, and naïve T cells were calculated (fold change of >2, FDR < 0.05, up-regulated highest in TH17 cells), and the top 200 genes were defined as TH17 signature genes and were used for further analysis. The TH1, TH2, and Treg signature genes were defined by similar approaches. The GSEA analysis was used to determine enrichment of T subset signatures in WT and Rorc-deficient T cells based on the defined TH signature genes.
ATAC-seq and analysis
In vitro–cultured cells were restimulated with plate bound anti-CD3 for 4 hours. The ATAC-seq DNA library was constructed using the TruePrep DNA Library Prep Kit V2 (Vazyme) and TruePrep Index Kit V2 for Illumina (Vazyme) according to the manufacturer’s instructions. Briefly, the nucleus was extracted, and the genome was broken into DNA fragments and linked with adaptors by the TruePrep Tagment Enzyme Mix. Fragmented DNA was further purified by AMPure beads and amplified with dual indexes by PCR. Sequencing was performed using HiSeq PE150 (Illumina) by Novogene.
Low-quality reads and adaptor sequences were removed by Trim Galore v0.4.4 with parameters “-q 10 –length 30 –stringency 50.” Paired-end reads were mapped to the mm10 reference genome using Bowtie 2 with –x2000. Reads that aligned to the mitochondrial genome were filtered, and PCR duplicates were removed with picard MarkDuplicates. To identify peaks, the bam files containing unique, non-chromatin reads were used to call peaks with MACS2 using parameters “--nomodel --keep-dup all -q 0.01-g mm.” For differential coverage, corresponding bam files were merged to call peaks to get a union peak set. For each peak in the peak set, raw ATAC-seq reads were counted by featureCounts from Subread package. Differentially expressed peaks were identified by at least two fold change and a P value of 0.001 using DEGseq. HOMER (findMotifsGenome) was used to find transcription factor (TF) motifs using parameters -len 8,10,12 -size −300,100.
SEs and TEs were defined by p300 loading in TH17 cells according to published dataset (26). Rank ordering of SEs (ROSE) (40) was used to identify SE domains, command as: python ROSE_main.py -g mm10 -i. /data/ TH17_peaks.gff -r. /data/ p300WTTh17.sort.bam -o TH17_rose -s 12500 -t 2500. BEDTools intersect function was used to identified overlaps between different sets of peaks.
Statistical analysis
The statistical significance was determined by Student’s t test or two-way analysis of variance (ANOVA) as indicated (two-tailed, *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001) using Prism software. All the experiments were repeated at least two to three times with consistent results.
Acknowledgments
We thank the Immunology Core Facility and the animal facility at Tsinghua University and all Dong Laboratory members for help and support.
Funding: This work was supported by grants from Beijing Natural Science Foundation (5222011 to X.W.), National Natural Science Foundation of China (31630022, 31821003, and 31991170 to C.D. and 32070889 to X.W.), and Shanghai Science and Technology Commission (21JC1404200 to C.D.).
Author contributions: X.C., X.W., and C.D. designed the project and prepared the manuscript. X.C. performed most experiments. W.J. identified the role of RORgt in maintaining Th17 cell identity in vitro. X.C. and X.Z. analyzed the bioinformatic data. J.S., X.B., T.X., and Y.J. helped with the experiments.
Competing interests: C.D. is a FViL investigator. The authors declare that they have no other competing interests.
Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. The RNA-seq and ATAC-seq data have been deposited in the GEO database under the accession code: GSE190539.
Supplementary Materials
This PDF file includes:
Figs. S1 to S4
REFERENCES AND NOTES
- 1.Dong C., Cytokine regulation and function in T cells. Annu. Rev. Immunol. 39, 51–76 (2021). [DOI] [PubMed] [Google Scholar]
- 2.Jiang Y., Wang X., Dong C., Molecular mechanisms of T helper 17 cell differentiation: Emerging roles for transcription cofactors. Adv. Immunol. 144, 121–153 (2019). [DOI] [PubMed] [Google Scholar]
- 3.Fragoulis G. E., Siebert S., Mcinnes I. B., Therapeutic targeting of IL-17 and IL-23 cytokines in immune-mediated diseases. Annu. Rev. Med. 67, 337–353 (2016). [DOI] [PubMed] [Google Scholar]
- 4.Garbers C., Heink S., Korn T., Rose-John S., Interleukin-6: Designing specific therapeutics for a complex cytokine. Nat. Rev. Drug Discov. 17, 395–412 (2018). [DOI] [PubMed] [Google Scholar]
- 5.Muranski P., Borman Z. A., Kerkar S. P., Klebanoff C. A., Ji Y., Sanchez-Perez L., Sukumar M., Reger R. N., Yu Z., Kern S. J., Roychoudhuri R., Ferreyra G. A., Shen W., Durum S. K., Feigenbaum L., Palmer D. C., Antony P. A., Chan C. C., Laurence A., Danner R. L., Gattinoni L., Restifo N. P., Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity 35, 972–985 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nurieva R., Yang X. O., Chung Y., Dong C., Cutting edge: In vitro generated Th17 cells maintain their cytokine expression program in normal but not lymphopenic hosts. J. Immunol. 182, 2565–2568 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hirota K., Duarte J. H., Veldhoen M., Hornsby E., Li Y., Cua D. J., Ahlfors H., Wilhelm C., Tolaini M., Menzel U., Garefalaki A., Potocnik A. J., Stockinger B., Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kamali A. N., Noorbakhsh S. M., Hamedifar H., Jadidi-Niaragh F., Yazdani R., Bautista J. M., Azizi G., A role for Th1-like Th17 cells in the pathogenesis of inflammatory and autoimmune disorders. Mol. Immunol. 105, 107–115 (2019). [DOI] [PubMed] [Google Scholar]
- 9.Hirota K., Turner J.-E., Villa M., Duarte J. H., Demengeot J., Steinmetz O. M., Stockinger B., Plasticity of TH17 cells in Peyer’s patches is responsible for the induction of T cell–dependent IgA responses. Nat. Immunol. 14, 372–379 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wang Y.-H., Voo K. S., Liu B., Chen C.-Y., Uygungil B., Spoede W., Bernstein J. A., Huston D. P., Liu Y.-J., A novel subset of CD4+ TH2 memory/ effector cells that produce inflammatory IL-17 cytokine and promote the exacerbation of chronic allergic asthma. J. Exp. Med. 207, 2479–2491 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ivanov I. I., McKenzie B. S., Zhou L., Tadokoro C. E., Lepelley A., Lafaille J. J., Cua D. J., Littman D. R., The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133 (2006). [DOI] [PubMed] [Google Scholar]
- 12.Chang D., Xing Q., Su Y., Zhao X., Xu W., Wang X., Dong C., The conserved non-coding sequences CNS6 and CNS9 control cytokine-induced Rorc transcription during T helper 17 cell differentiation. Immunity 53, 614–626.e4 (2020). [DOI] [PubMed] [Google Scholar]
- 13.Ciofani M., Madar A., Galan C., Sellars M., MacE K., Pauli F., Agarwal A., Huang W., Parkurst C. N., Muratet M., Newberry K. M., Meadows S., Greenfield A., Yang Y., Jain P., Kirigin F. K., Birchmeier C., Wagner E. F., Murphy K. M., Myers R. M., Bonneau R., Littman D. R., A validated regulatory network for Th17 cell specification. Cell 151, 289–303 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Leppkes M., Becker C., Ivanov I. I., Hirth S., Wirtz S., Neufert C., Pouly S., Murphy A. J., Valenzuela D. M., Yancopoulos G. D., Becher B., Littman D. R., Neurath M. F., RORγ-expressing Th17 cells induce murine chronic intestinal inflammation via redundant effects of IL-17A and IL-17F. Gastroenterology 136, 257–267 (2009). [DOI] [PubMed] [Google Scholar]
- 15.Steinmetz O. M., Summers S. A., Gan P.-Y., Semple T., Holdsworth S. R., Kitching A. R., The Th17-defining transcription factor RORγt promotes glomerulonephritis. J. Am. Soc. Nephrol. 22, 472–483 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Xue X., Soroosh P., De Leon-Tabaldo A., Luna-Roman R., Sablad M., Rozenkrants N., Yu J., Castro G., Banie H., Fung-Leung W.-P., Santamaria-Babi L., Schlueter T., Albers M., Leonard K., Budelsky A. L., Fourie A. M., Pharmacologic modulation of RORγt translates to efficacy in preclinical and translational models of psoriasis and inflammatory arthritis. Sci. Rep. 6, 37977 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sun N., Guo H., Wang Y., Retinoic acid receptor-related orphan receptor gamma-t (RORγt) inhibitors in clinical development for the treatment of autoimmune diseases: A patent review (2016-present). Expert Opin. Ther. Pat. 29, 663–674 (2019). [DOI] [PubMed] [Google Scholar]
- 18.Williams L. M., Rudensky A. Y., Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat. Immunol. 8, 277–284 (2007). [DOI] [PubMed] [Google Scholar]
- 19.Mullen A. C., Hutchins A. S., High F. A., Lee H. W., Sykes K. J., Chodosh L. A., Reiner S. L., Hlx is induced by and genetically interacts with T-bet to promote heritable T(H)1 gene induction. Nat. Immunol. 3, 652–658 (2002). [DOI] [PubMed] [Google Scholar]
- 20.Martins G. A., Hutchins A. S., Reiner S. L., Transcriptional activators of helper T cell fate are required for establishment but not maintenance of signature cytokine expression. J. Immunol. 175, 5981–5985 (2005). [DOI] [PubMed] [Google Scholar]
- 21.Zhu J., Min B., Hu-Li J., Watson C. J., Grinberg A., Wang Q., Killeen N., Urban J. F. Jr., Guo L., Paul W. E., Conditional deletion of Gata3 shows its essential function in TH1-TH2 responses. Nat. Immunol. 5, 1157–1165 (2004). [DOI] [PubMed] [Google Scholar]
- 22.Lee S., Kim J., Min H., Seong R. H., RORγt-driven TH17 cell differentiation requires epigenetic control by the Swi/Snf chromatin remodeling complex. iScience 23, 101106 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ichiyama K., Chen T., Wang X., Yan X., Kim B. S., Tanaka S., Ndiaye-Lobry D., Deng Y., Zou Y., Zheng P., Tian Q., Aifantis I., Wei L., Dong C., The methylcytosine dioxygenase Tet2 promotes DNA demethylation and activation of cytokine gene expression in T cells. Immunity 42, 613–626 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wang X., Zhang Y., Yang X. O., Nurieva R. I., Chang S. H., Ojeda S. S., Kang H. S., Schluns K. S., Gui J., Jetten A. M., Dong C., Transcription of Il17 and Il17f Is controlled by conserved noncoding sequence 2. Immunity 36, 23–31 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zuberbuehler M. K., Parker M. E., Wheaton J. D., Espinosa J. R., Salzler H. R., Park E., Ciofani M., The transcription factor c-Maf is essential for the commitment of IL-17-producing γδ T cells. Nat. Immunol. 20, 73–85 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Vahedi G., Kanno Y., Furumoto Y., Jiang K., Parker S. C. J., Erdos M. R., Davis S. R., Roychoudhuri R., Restifo N. P., Gadina M., Tang Z., Ruan Y., Collins F. S., Sartorelli V., O’Shea J. J., Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhou L., Lopes J. E., Chong M. M. W., Ivanov I. I., Min R., Victora G. D., Shen Y., Du J., Rubtsov Y. P., Rudensky A. Y., Ziegler S. F., Littman D. R., TGF-B-induced Foxp3 inhibits TH17 cell differentiation by antagonizing RORγt function. Nature 453, 236–240 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wei G., Wei L., Zhu J., Zang C., Hu-Li J., Yao Z., Cui K., Kanno Y., Roh T.-Y., Watford W. T., Schones D. E., Peng W., Sun H.-W., Paul W. E., O’Shea J. J., Zhao K., Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30, 155–167 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bettelli E., Das M. P., Howard E. D., Weiner H. L., Sobel R. A., Kuchroo V. K., IL-10 is critical in the regulation of autoimmune encephalomyelitis as demonstrated by studies of IL-10- and IL-4-deficient and transgenic mice. J. Immunol. 161, 3299–3306 (1998). [PubMed] [Google Scholar]
- 30.Basu R., Hatton R. D., Weaver C. T., The Th17 family: Flexibility follows function. Immunol. Rev. 252, 89–103 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Withers D. R., Hepworth M. R., Wang X., Mackley E. C., Halford E. E., Dutton E. E., Marriott C. L., Brucklacher-Waldert V., Veldhoen M., Kelsen J., Baldassano R. N., Sonnenberg G. F., Transient inhibition of ROR-γt therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat. Med. 22, 319–323 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Garidou L., Pomié C., Klopp P., Waget A., Charpentier J., Aloulou M., Giry A., Serino M., Stenman L., Lahtinen S., Dray C., Iacovoni J. S., Courtney M., Collet X., Amar J., Servant F., Lelouvier B., Valet P., Eberl G., Fazilleau N., Douin-Echinard V., Heymes C., Burcelin R., The gut microbiota regulates intestinal CD4 T cells expressing RORγt and controls metabolic disease. Cell Metab. 22, 100–112 (2015). [DOI] [PubMed] [Google Scholar]
- 33.Xiao S., Yosef N., Yang J., Wang Y., Zhou L., Zhu C., Wu C., Baloglu E., Schmidt D., Ramesh R., Lobera M., Sundrud M. S., Tsai P. Y., Xiang Z., Wang J., Xu Y., Lin X., Kretschmer K., Rahl P. B., Young R. A., Zhong Z., Hafler D. A., Regev A., Ghosh S., Marson A., Kuchroo V. K., Small-molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity 40, 477–489 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yang X. O., Nurieva R., Martinez G. J., Kang H. S., Chung Y., Pappu B. P., Shah B., Chang S. H., Schluns K. S., Watowich S. S., Feng X. H., Jetten A. M., Dong C., Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 29, 44–56 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Huh J. R., Leung M. W. L., Huang P., Ryan D. A., Krout M. R., Malapaka R. R. V., Chow J., Manel N., Ciofani M., Kim S. V., Cuesta A., Santori F. R., Lafaille J. J., Xu H. E., Gin D. Y., Rastinejad F., Littman D. R., Digoxin and its derivatives suppress Th17 cell differentiation by antagonizing RORγt activity. Nature 472, 486–490 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kurebayashi S., Ueda E., Sakaue M., Patel D. D., Medvedev A., Zhang F., Jetten A. M., Retinoid-related orphan receptor gamma (RORγ) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc. Natl. Acad. Sci. U.S.A. 97, 10132–10137 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yang X. O., Chang S. H., Park H., Nurieva R., Shah B., Acero L., Wang Y.-H., Schluns K. S., Broaddus R. R., Zhu Z., Dong C., Regulation of inflammatory responses by IL-17F. J. Exp. Med. 205, 1063–1075 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lee P. P., Fitzpatrick D. R., Beard C., Jessup H. K., Lehar S., Makar K. W., Pérez-Melgosa M., Sweetser M. T., Schlissel M. S., Nguyen S., Cherry S. R., Tsai J. H., Tucker S. M., Weaver W. M., Kelso A., Jaenisch R., Wilson C. B., A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001). [DOI] [PubMed] [Google Scholar]
- 39.Yang X. O., Pappu B. P., Nurieva R., Akimzhanov A., Kang H. S., Chung Y., Ma L., Shah B., Panopoulos A. D., Schluns K. S., Watowich S. S., Tian Q., Jetten A. M., Dong C., T helper 17 lineage differentiation is programmed by orphan nuclear receptors RORα and RORγ. Immunity 28, 29–39 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Whyte W. A., Orlando D. A., Hnisz D., Abraham B. J., Lin C. Y., Kagey M. H., Rahl P. B., Lee T. I., Young R. A., Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figs. S1 to S4






