Summary
Commensal microbes induce cytokine-producing effector tissue-resident CD4 T cells, but the function of these T cells in mucosal homeostasis is not well understood. Here we report that commensal-specific intestinal TH17 cells possess an anti-inflammatory phenotype marked by expression of IL-10 and co-inhibitory receptors. The anti-inflammatory phenotype of gut-resident commensal-specific TH17 cells was driven by the transcription factor c-MAF. IL-10-producing commensal-specific TH17 cells were heterogeneous and derived from a TCF1+ gut-resident progenitor TH17 cell population. TH17 cells acquired IL-10 expression and anti-inflammatory phenotype in the small intestinal lamina propria. IL-10-production by CD4 T cells and IL-10 signaling in intestinal macrophages drove IL-10 expression by commensal-specific TH17 cells. Intestinal commensal-specific TH17 cells possessed immunoregulatory functions and curbed effector T cell activity in vitro and in vivo in an IL-10-dependent and c-MAF-dependent manner. Our results suggest that tissue-resident commensal-specific TH17 cells perform regulatory functions in mucosal homeostasis.
Introduction
Mucosal surfaces are colonized by a vast collection of resident microorganisms that shape tissue immune responses1,2. Intestinal tolerance towards commensals is promoted by induction of commensal-specific Foxp3+ regulatory T cells3–5. However, commensals can also induce effector CD4 T cells, such as TH17 cells6–9. The functions of commensal-specific TH17 cells (hereafter referred to as commensal TH17 cells) in mucosal immunity are incompletely understood. They can contribute to control of the inducing commensal10, but whether they perform additional functions in mucosal homeostasis is unclear.
IL-17-producing CD4 T cells (TH17 cells) are a functionally heterogeneous population and can acquire pathogenic and non-pathogenic phenotypes11–15. TH17 cells are known drivers of inflammation, including intestinal inflammation, and can promote the pathology of inflammatory bowel diseases (IBD)16,17. However, not all TH17 cells are inflammatory. For example, TH17 cell-derived cytokines participate in strengthening the epithelial barrier and, therefore, in protection from inflammation18–21. TH17 cells can also produce IL-10 and intestinal TH17 cells can convert to a regulatory phenotype under inflammatory conditions12,15,22. Although inflammatory TH17 cells have been well studied, the functions of non-pathogenic TH17 cells are incompletely understood23.
Gut-resident commensal TH17 cells are metabolically distinct from inflammatory TH17 cells24 and are generally considered non-pathogenic. However, whether commensal TH17 cells simply fail to participate in inflammatory responses or possess specific effector mechanisms to regulate inflammation that may direct additional functions in mucosal homeostasis is currently unknown.
Here, we examine in more detail the phenotype of various types of intestinal TH17 cells, including commensal TH17 cells induced by segmented filamentous bacteria (SFB). We find that SFB TH17 cells possess a unique anti-inflammatory phenotype characterized by expression of the transcription factor c-MAF and the cytokine IL-10. Establishment of this program occurs in the terminal ileum and requires the coordinated action of intestinal CD4 T cells and intestinal macrophages. We also find that SFB TH17 cells can curb effector T cell function both in vitro and in vivo. Our results describe anti-inflammatory functions of commensal TH17 cells and suggest that these cells may have important roles in maintaining intestinal homeostasis.
Results
Small-intestinal commensal TH17 cells have a regulatory transcriptional program
To identify unique features of commensal TH17 cells, we profiled their transcriptome by RNA-sequencing and compared it to the transcriptome of alternatively generated intestinal TH17 cells. Il17aGFP reporter animals were colonized with SFB to induce commensal TH17 cells (SFB TH17 cells). Intestinal TH17 cells were also induced by infecting Il17aGFP animals with Citrobacter rodentium (Crod) or by transferring naïve CD45RBhi CD4 T cells from Il17aGFP animals to RAG1-deficient animals in a classical model of intestinal inflammation. Lamina propria (LP) TH17 cells were isolated from small (SI) and large (LI) intestine at the peak of microbial colonization or colitis induction (Figure S1A–C). TH17 cells comprised significant percentage of CD4 T cells in intestinal tissues (Figure 1A). However, TH17 cell phenotype differed between the different conditions (Figure 1B). The transcriptional program of SFB-induced TH17 cells was distinct from the transcriptional programs of other intestinal TH17 cells (Figure 1B). Expression of 500–1,100 genes differed significantly between SFB TH17 cells and any other examined intestinal TH17 cells (Figure S1D). In contrast, DEG numbers were lower in pairwise comparisons between non-SFB TH17 cells (Figure S1E). We identified a core signature of 309 genes that differed significantly between SFB TH17 cells and at least three of the other TH17 cell datasets (Figure 1C and S1F). The core SFB TH17 cell program contained genes involved in inhibitory/regulatory (e.g., Ctla4, Lag3, Tigit), anti-inflammatory (e.g., Maf, Il10) and tissue-protective (e.g., Ahr, Areg) functions (Figure 1D, E and S1G). At the same time genes enriched in inflammatory TH17 cells were underrepresented in SFB TH17 cells (Figure S1H). Overall, SFB TH17 cells specifically expressed genes associated with decreased T cell responsiveness25. We, therefore, compared this program to the gene signatures of “non-responsive” T cells, such as exhausted and regulatory T cells. SFB TH17 cells resembled exhausted CD4 T cells generated following chronic infection (Figure 1F)26 and expressed classical markers of CD4 T cell exhaustion such as Ikzf2 and Tox27,28 (Figure 1E). At the same time, SFB TH17 cells closely resembled mouse and human IL-10 expressing immunoregulatory TH17 cells12,29 (Figure 1G and S1I). Moreover, commensal-induced TH17 cells were enriched in a subset of signature genes for IL-10+Foxp3neg TR1 cells (Figure 1H). Comparison of core leading edge genes in SFB TH17 cells to published mouse and human IL-10-expressing TH17 cells and non-pathogenic TH17 cells identified a set of 11 common genes, most notably genes encoding the prototypical anti-inflammatory cytokine IL-10, and the transcription factor c-MAF30–32 (Figure 1I). Among intestinal TH17 cells, expression of Il10 transcripts was restricted to commensal TH17 cells (Figure 1D and S1G) and Maf expression was significantly upregulated in SFB TH17 cells (Figure 1E and S1G). In addition to Maf, several other transcription factors, including Maf co-factors, involved in regulation of Il10 in CD4 T cells, such as Ikzf3, Ahr and Nfil3 33–35 were also expressed preferentially in commensal intestinal TH17 cells (Figure 1E). Collectively, these results suggest that commensal intestinal TH17 cells possess an anti-inflammatory transcriptional program that resembles that of IL-10-producing regulatory CD4 T cells.
Figure 1. SFB TH17 cells have an anti-inflammatory transcriptional program.

(A) Intestinal lamina propria (LP) TH17 cells induced by various mechanisms. SI, small intestine; LI, large intestine; Colitis, CD45RBhi colitis. Representative FACS plots gated on TCRβ+CD4+ LP lymphocytes.
(B) PCA plot of RNA-sequencing analysis of various intestinal LP TH17 cells. One experiment, N=2–3 mice/group.
(C) Heatmap of core SFB TH17 cells program genes in bulk RNA-seq samples from (B). c-MAF controlled genes36 are also marked on the right.
(D) Expression of selected cytokines and inhibitory receptors in LP TH17 cells in RNA-Seq data from (B).
(E) Expression of selected transcription factors in LP TH17 cells in RNA-Seq data from (B).
(F) Gene set enrichment analysis (GSEA) of genes upregulated in SFB TH17 cells compared to genes upregulated in exhausted CD4 T cells26.
(G) GSEA of genes upregulated in SFB TH17 cells compared to genes upregulated in mouse IL-10+ TH17 cells12.
(H) Expression of TR1 signature genes in various intestinal LP TH17 cells.
(I) Venn Diagram of leading-edge genes from GSEA of genes upregulated in SFB TH17 cells and published datasets12,13,29.
Small-intestinal commensal TH17 cells express IL-10 and co-inhibitory receptors
To confirm the RNA-Seq data we followed expression of IL-10 in commensal or non-commensal TH17 cells using Il10GFP/Il17aKatushka/Foxp3mRFP reporter mice. Non-commensal TH17 cells lacked expression of IL-10 in SI and LI LP (Figure 2A, B and S2A). In contrast, ~40% of SFB-induced TH17 cells in the small intestine co-expressed IL-17 and IL-10 (Figure 2A, B). Citrobacter-induced and colitogenic intestinal TH17 cells produced IFN-γ and GM-CSF (Figure S2B, C). In contrast, commensal TH17 cells lacked expression of these inflammatory cytokines (Figure S2B, C). We also examined expression of c-MAF in intestinal TH17 cells by flow cytometry. SFB colonization induced significant increase in the proportion of TH17 cells that co-expressed c-MAF and IL-17 (Figure 2C, D). On average, 50% of SI LP TH17 cells in SFB-positive animals expressed c-MAF, which was similar to that of intestinal Foxp3+ Tregs (Figure 2C, D). In contrast, other intestinal TH17 cells demonstrated either no change or decrease in the proportion of c-MAF+ cells (Figure 2C, D). The proportion of IL-10- and c-MAF-positive TH17 cells was not specifically increased in LI LP of SFB-positive animals (Figure S2A, D). SFB colonization generally did not significantly increase the proportion of Foxp3negIL-17negIL-10+ (TR1) cells, although slight increase was noted in the terminal ileum (Figure S2E). c-MAF induction in commensal TH17 cells preceded IL-10 expression and c-MAF was already significantly upregulated in IL-10GFPneg commensal TH17 cells (Figure 2E). However, c-MAF expression further increased in IL-10GFP+ SFB-induced TH17 cells (Figure 2F). Similarly to endogenous TH17 cells, naïve SFB-specific 7B8 Tg CD4 T cells adoptively transferred into SFB-positive WT C57BL/6 mice, differentiated into IL-10- and c-MAF-expressing TH17 cells (Figure 2G, H). c-MAF is a transcription factor known to promote IL-10 expression in T cells and non-pathogenic TH17 cells33,36. In addition, c-MAF has been shown, in several other T cell subsets, to imbue anti-inflammatory functions, even beyond IL-1025,36. In particular, c-MAF controls an inhibitory gene module in CD4 and CD8 T cells that contains a number of co-inhibitory receptors, e.g. CTLA4, LAG3, TIM3, TIGIT25. In agreement with a crucial role for c-MAF, SFB TH17 cells, but not other intestinal TH17 cells, contained a population that co-expressed IL-10 and co-inhibitory receptors (Figure 2I, J). Analysis of c-MAF target genes36 in our RNA-Seq datasets, showed that within the core SFB TH17 cell signature, genes upregulated in SFB TH17 cells were enriched in targets positively regulated by c-MAF and genes downregulated in SFB TH17 cell were enriched in targets negatively regulated by c-MAF (Figure 1C). To investigate whether the induction of IL-10+ TH17 cells was restricted to SFB, we induced TH17 cells in Il10GFP/Il17aKatushka/Foxp3mRFP reporter mice by oral gavage of Bifidobacterium adolescentis (Figure S2F) 37. B. adolescentis induced TH17 cells in the SI LP, which similarly to SFB TH17 cells, expressed IL-10 and c-MAF, as well as the co-inhibitory receptors CTLA-4 and LAG-3 (Figure 2K–M). Altogether our results suggest that c-MAF leads to expression of IL-10 and generally inhibitory T cell phenotype in commensal-specific SI LP TH17 cells.
Figure 2. SFB TH17 cells express IL-10 and co-inhibitory receptors.

(A, B) IL-10 expression in SI LP TH17 cells and Foxp3+ Tregs from Il10GFP/Il17aKatushka/Foxp3mRFP mice under various conditions. IL-17/IL-10 FACS plots in (A) gated on TCRβ+CD4+Foxp3mRFPneg lymphocytes. Foxp3/IL-10 FACS plot in (A) gated on TCRβ+CD4+IL-17Katushkaneg lymphocytes. (B) IL-10 (GFP) expression in TH17 (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+) or Treg (TCRβ+CD4+Foxp3mRFP+) cells. Three independent experiments, N=5–9 mice/group.
(C, D) c-MAF expression (intracellular staining) in SI LP TH17 cells and Foxp3+ Tregs. FACS plots in (C) gated on TCRβ+CD4+ LP lymphocytes. (D) c-MAF expression in TH17 (TCRβ+CD4+IL-17+) or Treg (TCRβ+CD4+Foxp3+) cells. Two independent experiments, N=5–6 mice/group.
(E) qPCR of Il10 and Maf transcripts in IL-10GFPneg and IL-10GFP+ SFB TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+) and IL-10GFPneg/IL-17Katushkaneg/Foxp3mRFPneg control (C) CD4 T cells FACS-purified from SI LP of Il10GFP/Il17aKatushka/Foxp3mRFP mice. Two independent experiments, N=2–5 mice/group.
(F) Representative histograms of c-MAF expression (intracellular staining) in IL-10GFPneg and IL-10GFP+ TH17 cells and control CD4 T cells FACS-purified from SI LP of SFB-colonized Il10GFP/Il17aKatushka/Foxp3mRFP mice. Two independent experiments, N=2–5 mice/group.
(G, H) Naive 7B8 SFB-specific TCR Tg CD4 T cells from 7B8 Il10GFP/Il17aKatushka/Foxp3mRFP mice were adoptively transferred into SFB-colonized congenic wild type mice. IL-10 (GFP) and c-MAF (intracellular staining) expression in transferred CD4 T cells was examined one week later. FACS plots gated on Ly5.1+CD4+TCRβ+Foxp3neg transferred 7B8 cells. Bar plots further gated on IL-17+ TH17 cells. Two independent experiments, N=5 mice.
(I, J) tSNE analysis based on multi-parameter flow cytometry of IL-10 and co-inhibitory receptors (CIR) expression in SI LP TH17 cells from SFB-colonized (I, J) or Citrobacter rodentium (Crod) infected (J) Il10GFP/Il17aKatushka/Foxp3mRFP mice. Plots gated on Ly5.1+CD4+TCRβ+Foxp3mRFPnegIL-17Katushka+ cells. Two independent experiments, N=5 mice/group.
(K) TH17 cell induction and IL-10 expression in SI LP TH17 cells from Il10GFP/Il17aKatushka/Foxp3mRFP mice after oral gavage of E. coli (Ec) or B. adolescentis (Ba) every other day for two weeks. IL-17/IL-10 FACS plots gated on TCRβ+CD4+Foxp3mRFPneg lymphocytes. Two independent experiments, N=4–5 mice/group.
(L, M) c-MAF (intracellular staining) (L) and LAG-3 and CTLA-4 expression (M) in SI LP TH17 cells the experiments in (K). Two independent experiments, N=4–5 mice/group.
The anti-inflammatory phenotype of SFB TH17 cells is driven by c-MAF
To directly assess the role of c-MAF in the acquisition of the TH17 cell regulatory phenotype we conditionally deleted c-MAF in TH17 cells by generating Il10GFP/Il17aKatushka/Foxp3mRFP/Il17aCre/Mafflox/flox/R26STOP-YFP mice (MafΔIL17). IL-17 expressing cells are also permanently labeled with YFP in these animals (Figure S3A). We confirmed TH17 cell-specific deletion of c-MAF in SI LP TH17 cells of MafΔIL17 mice (Figure 3A). SFB TH17 cells were present in SI LP of MafΔIL17 mice, albeit at slightly decreased frequency compared to littermate controls (Figure 3B and S3B). Other T cell and IL-17-expressing subsets were unchanged with exception of a decrease in IL-17+ γδ T cells (Figure S3C, D) as reported elsewhere 38. c-Maf-deficiency in Foxp3+ Tregs can also indirectly affect TH17 cell function through loss of IL-10 expression on Tregs 39. However, frequency and IL-10 production by Foxp3+ Tregs were unaffected in MafΔIL17 mice (Figure 3C and S3C). In contrast, SFB TH17 cells lacked IL-10 expression in MafΔIL17 mice compared to littermate controls (Figure 3C, D and S3C). Moreover, c-MAF-deficient SI LP TH17 cells also lost, or downregulated, other signature genes of the SFB anti-inflammatory program (Figure 3D and S3E). Instead, SI LP SFB TH17 cells from MafΔIL17 mice showed increased frequency of IFN-γ (Figure 3E) and up-regulated other genes associated with inflammatory TH17 cells (Figure 3D and S3E)40. To examine changes in the overall transcriptional program, we performed single cell RNA-sequencing (scRNA-Seq) on purified YFP+ TH17 cells from SI LP of MafΔIL−17 mice and littermate controls following SFB colonization. c-MAF-deficient SFB TH17 cells showed general loss of the SFB TH17 signature anti-inflammatory program (Figure 3F). In contrast to WT TH17 cells, the transcriptional program of SFB TH17 cells from MafΔIL17 mice resembled that of Crod-induced and colitogenic TH17 cells in our bulk RNA-Seq datasets (Figure 3G), as well as that of published inflammatory EAE TH17 cells (Figure S3F). These results suggest that acquisition of an anti-inflammatory phenotype by commensal TH17 cells, including IL-10-expression, requires c-MAF.
Figure 3. c-MAF drives anti-inflammatory identity of intestinal commensal TH17 cells.

(A) Intracellular staining for c-MAF in CD4 T (TCRβ+CD4+Foxp3mRFPneg) and TH17 (TCRβ+CD4+IL-17+) cells from SI LP of Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox (MafΔIL17) mice and Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/+ (WT) littermates. Three independent experiments, N=5–7 mice/group.
(B) Frequency of TH17 cells (intracellular staining) in SI LP of WT and MafΔIL17 mice. Three independent experiments, N=7 mice/group.
(C) IL-10GFP expression in SI LP TH17 cells and Foxp3+ Tregs from Il10GFP/Il17aKatushka/Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox (MafΔIL17) and littermate control (WT) mice. FACS plots gated on TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+ (TH17) or TCRβ+CD4+Foxp3mRFP+ (Treg) lymphocytes. Two independent experiments, N=2–4 mice/group
(D) Quantitative PCR of Il10, Areg, Tox, Ccl5 and Gzma mRNA in FACS-purified SI LP TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+) from WT and MafΔIL17 (Il10GFP/Il17aKatushka/Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice. Two independent experiments, N=6–7 mice/group.
(E) Intracellular staining for IL-17 and IFN-γ in (Left) CD4 T (TCRβ+CD4+) and (Right) TH17 (TCRβ+CD4+IL-17+) cells from SI LP of WT and MafΔIL17 (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice. Two independent experiments, N=6 mice/group.
(F) Heatmap of selected SFB TH17 cell signature genes in scRNA-Seq of FACS-purified SI LP TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17YFP+) from WT and MafΔIL17 (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice. One experiment, N=2–3 mice/group.
(G) GSEA of top 200 upregulated genes in MafΔIL17 SI LP SFB TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17YFP+) compared to genes upregulated in LI Crod TH17 cells and colitis TH17 cells in bulk RNA-Seq datasets in Figure 1.
Small-intestinal commensal TH17 cells have immunoregulatory functions
The forgoing results demonstrate that SFB TH17 cells express IL-10 and share transcriptional and phenotypic characteristics with IL-10-expressing immunoregulatory CD4 T cells, such as Foxp3neg TR1 cells. We, therefore, investigated whether commensal TH17 cells can regulate the function of other CD4 T cells. To evaluate inhibitory effects on T cell proliferation, we compared the proliferation of responder CD4 T cells in vitro in the presence or absence of purified intestinal TH17 cells. Citrobacter-induced TH17 cells from small or large intestine did not significantly affect proliferation of responder T cells (Figure 4A). In contrast, co-culture with SFB-induced SI LP TH17 cells led to significant inhibition of responder T cell proliferation (Figure 4A). Inhibition of proliferation did not correlate with preferential expansion of intestinal TH17 cells in these assays, because despite showing higher inhibitory activity, SFB TH17 cells demonstrated lower proliferative capacity than Citrobacter TH17 cells (Figure 4B). Inhibition of proliferation by SFB TH17 cells required IL-10 signaling, because it was virtually abrogated by addition of IL-10R-blocking antibody to the co-cultures (Figure 4C). Moreover, SFB TH17 cells did not inhibit proliferation of responder CD4 T cells that lacked expression of IL-10R (Figure 4D). SFB TH17 cells also express co-inhibitory receptors (Figure 2I, J). Blocking antibodies against CTLA-4, but not LAG-3, partially reduced the inhibitory ability of SI LP SFB TH17 cells (Figure 4E, F). In addition, c-MAF-deficient SI LP SFB TH17 cells lost the ability to suppress responder T cell proliferation (Figure 4G). These results suggest that intestinal commensal TH17 cells can exert regulatory functions in vitro in an IL-10 and c-MAF-dependent manner.
Figure 4. Regulatory functions of commensal TH17 cells.

(A) In vitro suppression assay. FACS-purified SI LP TH17 cells (TCRβ+CD4+ Foxp3mRFPnegIL-17GFP+) from SFB-colonized or Citrobacter rodentium infected (Crod) mice or Treg cells (TCRβ+CD4+Foxp3mRFP+) were co-cultured with WT naïve responder CD4 T cells from spleen of untreated mice as described in Methods. (Left) Proliferation of CTV-stained responder T cells (R) on Day 4. (Right) Percent suppression calculated as described in Methods. Cumulative of at least four independent experiments, N=2–3 technical replicates/experiment. Each dot represents a technical replicate.
(B) Division index (see Methods) of CTV-labelled SFB and Crod LP TH17 cells in in vitro suppression assay. One experiment, N=3 mice/group and 2 technical replicates/mouse. Each dot represents a technical replicate.
(C) Proliferation of WT responder CD4 T cells (R) alone or co-cultured with FACS-purified SI LP SFB TH17 cells (TCRβ+CD4+ Foxp3mRFPnegIL-17GFP+) in the presence of blocking anti-IL-10R antibody or isotype control. Five independent experiments, N=2–3 technical replicates/experiment. Significance, paired t-test.
(D) Inhibition of proliferation of WT or Il10rb−/− responder CD4 T cells by purified WT SI LP SFB TH17 cells (TCRβ+CD4+ Foxp3mRFPnegIL-17GFP+). Three independent experiments, N=2–3 technical replicates/experiment. Significance, paired t-test.
(E) Proliferation of WT responder CD4 T cells (R) alone or co-cultured with FACS-purified SI LP SFB TH17 cells (TCRβ+CD4+ Foxp3mRFPnegIL-17GFP+) in the presence of blocking anti-CTLA-4 antibody or isotype control. Four independent experiments, N=2–3 technical replicates/experiment. Significance, paired t-test.
(F) Proliferation of WT responder CD4 T cells (R) alone or co-cultured with FACS-purified SI LP SFB TH17 cells (TCRβ+CD4+ Foxp3mRFPnegIL-17GFP+) in the presence of blocking anti-LAG3 antibody or isotype control. Three independent experiments, 2–3 technical replicates/experiment. Significance, paired t-test.
(G) Proliferation of WT responder CD4 T cells (R) alone or co-cultured with SI LP SFB TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17YFP+) from WT or MafΔIL17 mice. Cumulative of three independent experiments, N=2–3 technical replicates/experiment. Each datapoint represents a technical replicate.
(H) Experimental schematic of in vivo suppression assay.
(I-K) Expansion (I) and TH17 cell differentiation (J, K) of naïve 7B8 CD4 T cells (Ly5.1+) in SI LP 8 days after transfer into SFB colonized RAG1-deficient mice alone (C) or with co-transfer of SI LP Treg cells (TCRβ+CD4+Foxp3mRFP+) or SI LP SFB TH17 cells (TCRβ+CD4+IL-17GFP+) with and without neutralization of IL-10 signaling by intraperitoneal injection of an anti-IL-10R or isotype control antibody. (I, J, K) Plots gated on Ly5.1+TCRβ+CD4+ (7B8) SI LP lymphocytes. (I, K) Data was normalized to the average of the corresponding control group. Cumulative of six independent experiments, N=7–17 mice/group.
To evaluate immunoregulatory functions of SFB TH17 cells in vivo we considered their localization. SFB TH17 cells are exclusively present in SI LP6,41–43. Compared to other TH17 cells in our dataset, SFB TH17 cells express a number of chemokine receptors, e.g. Ccr9, Ccr5, Ccr1, associated with tissue residency or homing to SI (Figure S3A)44–46. In addition, SFB TH17 cells almost uniformly express the tissue retention factor CD69 (Figure S3B)47. Purified small-intestinal SFB TH17 cells homed exclusively to the SI LP, but not to other tissues, including other intestinal tissues (Figure S3C). Thus, SFB TH17 cells possess features of tissue-resident CD4 T cells. We, therefore, investigated whether SFB TH17 cells exert immunoregulatory functions locally in the small intestine. Adoptive transfer of purified intestinal SFB TH17 cells into RAG1-deficient animals (Figure 4H) significantly inhibited expansion (Figure 4I) and IL-17 production (Figure 4J, K) of co-transferred naïve 7B8 Tg CD4 T cells. The inhibition was similar to that exerted by SI LP Foxp3+ Tregs (Figure 4I–K). Moreover, this inhibition occurred only in the SI LP and was not observed in mLN (Figure S3D). Neutralization of IL-10-signaling in vivo, significantly reduced commensal TH17 cell-mediated inhibition of CD4 T cell expansion and cytokine production in SI LP (Figure 4I–K). The foregoing results suggest that commensal TH17 cells can exert immunoregulatory functions and curb effector T cell activity both in vitro and in vivo in an IL-10-dependent manner.
Commensal TH17 cells are heterogeneous and contain a progenitor TCF1+ subset
To further examine the heterogeneity of commensal TH17 cells, we purified TH17 cells from SI LP of SFB-colonized Il17aKatushka/Foxp3mRFP reporter mice and performed scRNA-Seq. Uniform Manifold Approximation and Projection for Dimensional Reduction (UMAP) analysis of 5721 recovered single SI LP TH17 cells showed several transcriptionally distinct clusters (Figure 5A). We annotated these clusters into functional sets based on the genes that were differentially expressed relative to all other clusters (Figure 5B, C). Apart from two small clusters enriched in proliferation and interferon stimulated (ISG) genes respectively, the majority (97%) of intestinal TH17 cells had terminally differentiated or progenitor/stem-like phenotypes (Figure 5B, C). Terminally differentiated TH17 cells belonged to two distinct types with activated (C1, C3) and inhibitory (C2, C6) phenotypes respectively (Figure 5B, C). Activated TH17 cells expressed genes associated with T cell activation and intestinal tissue residency, e.g. Cd69, Cd28, Jun, Ccr9, Ccr2, Ccr5, Ccl20. In contrast, TH17 cells with inhibitory phenotype expressed inhibitory and tissue-repair genes, e.g. Lag3, Tim3 (Havcr2), IL17f, Tgfb1, Areg (Figure 5B). Cells in C1 and C6 contained higher expression of the corresponding effector programs (Figure 5B). Both types of terminally differentiated TH17 cells contained cells expressing Il10 (Figure 5D). Pathway analysis of differentially expressed genes demonstrated differences not only in activation, but also in their metabolic profile (Figure 5E). Metabolism is an established regulator of T cell functionality. We, therefore, applied the COMPASS algorithm48 to compare metabolic states of the two most divergent IL-10-expressing clusters (C1 and C6). COMPASS predicted that cells in C1 had increased levels of glycolysis and those in C6 had increased fatty acid oxidation and amino acid metabolism (Figure 5F). These differences parallel those previously described between pathogenic TH17 cells vs non-pathogenic TH17 cells and Foxp3+ Treg cells. c-MAF targets were specifically enriched in the two types of IL-10+ TH17 cells (Figure 5G). We next examined the role of c-MAF by purifying YFP+ SI LP TH17 cells from MafΔIL−17 mice and WT littermate controls and performing sc-RNA-Seq. Analysis of more than 10,000 SI LP TH17 cells identified similar UMAP functional clustering (Figure S5A, B). Further analysis revealed that both types of IL-10+ TH17 cell subsets required c-MAF for IL-10 expression (Figure 5H). TH17-specific deletion of c-MAF led to a decrease in the most differentiated IL-10+ TH17 cell clusters (Figure 5I, J). In addition, TH17-specific deletion of c-MAF resulted in loss of the overall anti-inflammatory program of both activated and inhibitory effector IL-17+ TH17 cell subsets (Figure 5K and Figure S5C). In addition, conditional deletion of c-MAF resulted in a significant increase in the proportion of YFP+IL-17neg (ex-TH17) cells with an inflammatory phenotype (Figure 5I–L and S5D). Thus, c-MAF not only drives the anti-inflammatory SFB TH17 cell program, but also inhibits conversion into inflammatory ex-TH17 cells.
Figure 5. Commensal TH17 cells are heterogeneous and contain two IL-10+ populations.

(A) UMAP clustering following scRNA-sequencing of 5721 SFB SI LP TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+) sorted from Il10GFP/Il17aKatushka/Foxp3mRFP reporter mice.
(B) Functional grouping of SI LP TH17 cell clusters in (A) based on expression of select marker genes.
(C) UMAP with annotation of the functional groups in (B).
(D) Expression of Il10 in individual SFB SI LP TH17 cells overlayed over the UMAP clustering in (A).
(E) Pathway analysis of differentially expressed genes between activated and inhibitory IL-10+ expressing groups.
(F) COMPASS analysis for metabolic pathways in the two most differentiated IL-10+ UMAP clusters – C1 and C6.
(G) GSEA for c-MAF target genes36 in individual SFB SI LP TH17 cells overlayed over the UMAP clustering in (A).
(H) Expression of Il10 mRNA in indicated functional groups in SI LP SFB TH17 cells from WT (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/+) and MafΔIL17 (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice, based on scRNA-Seq of SI LP TH17 cells sorted based on YFP expression. Based on the functional clustering in Figure S5B. SI LP SFB TH17 cells from individual mice were identified by hash-tagging of scRNA-Seq samples. N=2–3 mice/group.
(I) Frequency of cells in clusters C7 (ex- TH17), C8 (inhibitory), and C10 (activated) based on the UMPA clustering in Figure S5A. Data from hash-tagged scRNA-Seq samples from WT and MafΔIL17 (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice. Data integrated from N=2–3 mice/group.
(J) Statistics of (I)
(K) Heatmap of z score of average expression of selected SFB TH17 signature genes and inflammatory genes in indicated functional groups based on UMAP in Figure S5B in hash-tagged scRNA-Seq samples from WT and MafΔIL17 (Foxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox) mice.
(L) IL-17Katushka and ROSAYFP expression in (Left) CD4 T (TCRβ+CD4+Foxp3mRFPneg) and (Right) ex-TH17 (TCRβ+CD4+IL-17YFP+IL-17Katushkaneg) cells from SI LP of WT and MafΔIL17 Il10GFP/Il17aKatushkaFoxp3mRFP/R26STOP-YFP/Il17aCre/Mafflox/flox mice. N= 4 mice/group.
Progenitor-like commensal TH17 cells were defined by expression of stem-like features, e.g. Tcf7, Il7r, and Slamf649,50, with cluster C4 (Figure 5A) showing the highest combined expression of these genes (Figure 5A, 6A and S6A). We confirmed co-expression of TCF1 and IL-7R on a subset of commensal SI LP TH17 cells by flow cytometry (Figure 6B). In agreement with the scRNA-Seq data, TCF1 expression was downregulated in IL-10+ SFB TH17 cells (Figure 6C). In contrast, a subset of TCF1+ SFB TH17 cells expressed low levels of c-MAF (Figure 6D). Progenitor-like SI LP TH17 cells had significantly decreased ability to suppress T cell proliferation in vitro, compared to TCF1neg IL-10+ TH17 cells (Figure 6E and S6B, C). Trajectory analysis of scRNA-Seq data from SI LP TH17 cells revealed three distinct trajectories for progenitor-like TCF1+ TH17 cells in cluster C4 leading respectively to the two effector IL-10+ populations, or back to the progenitor-like group (Figure 6F). This suggests that TCF1+ TH17 cells have the potential to self-renew and are progenitors of TCF1negIL-10+ TH17 cells. We also identified a similar TCF1+ TH17 cell subset in the LI LP of animals infected with Citrobacter rodentium (Figure S6D, E). To confirm experimentally the progenitor nature of TCF1+ TH17 cells, we generated Tcf7mCherry reporter mice and crossed them to Il17aGFP and Il10Venus reporter animals (Figure S6F, G). Analysis of SI LP TH17 cells confirmed that TCF1mCherry+ TH17 cells do not express IL-10 (Figure 6G, H). Next, we purified TCF1mCherry+ TH17 cells from SI LP of SFB-positive animals and adoptively transferred these cells into SFB-colonized wildtype mice. TCF1mCherry+ TH17 cells homed to SI LP immediately after transfer and gave rise to TCF1mCherryneg TH17 cells at later timepoints (Figure 6I). Purified SFB TCF1+ LP TH17 cells also differentiated into TCF1negIL-10+ TH17 cells upon TCR stimulation in vitro, in contrast to TCF1+ LP TH17 cells from Crod-infected mice (Figure 6J). In these experiments, we could not recover TCFneg TH17 cells following adoptive transfer or in vitro culture, suggesting that they lose the ability to self-renew and to propagate an immune response (data not shown). Altogether the foregoing results suggest that IL-10+ commensal TH17 cells in the SI LP are heterogeneous and differentiate from TCF1+ progenitor TH17 cells that upregulate c-MAF and downregulate TCF1 expression.
Figure 6. Commensal TH17 cells contain a progenitor TCF1+ population.

(A) Expression of Tcf7 and Il7r mRNA in individual SFB SI LP TH17 cells overlayed over the UMAP clustering in Figure 5A.
(B) TCF1 and IL-7R expression in SI LP SFB TH17 cells. Gated on TCRβ+CD4+IL-17+ lymphocytes.
(C) Intracellular staining for TCF1 in FACS-purified IL-10GFPneg and IL-10GFP+ SI LP SFB TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+).
(D) Intracellular staining for TCF1 and c-MAF in SI LP SFB TH17 cells. Gated on TCRβ+CD4+IL-17+ lymphocytes.
(E) FACS-purified SI LP SFB progenitor TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+IL-10GFPnegIL-7R+) and SI LP SFB inhibitory TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+IL-10GFP+LAG-3+) were co-cultured with WT naïve responder CD4 T cells. (Left) Proliferation of CTV-stained responder T cells (R) on Day 4. (Right) Percent suppression. Cumulative of three independent experiments. Each dot represents a technical replicate.
(F) Trajectory analysis of scRNA-Seq data in Figure 5A with a start node in C4. UMAP annotation as in Figure 5C.
(G) Quantitative PCR for Il10 mRNA in FACS-purified TCF1mCherry+ and TCF1mCherryneg SI LP SFB TH17 cells from Tcf7mCherry/Il17aGFP mice. Two independent experiments, N=4 mice/group.
(H) TCF1mCherry and IL-10Venus expression in SI LP SFB TH17 cells (TCRβ+CD4+IL-17GFP+) from Tcf7mCherry/Il17aGFP/Il10Venus mice. Two independent experiments, N=3 mice/group.
(I) TCF1mCherry+IL-17AeGFP+ CD4 T cells were FACS-purified from SI LP of Tcf7mCherry/Il17aGFP mice (Ly5.1) and adoptively transferred into SFB-colonized WT mice (Ly5.2). TCF1 and IL-17 expression in transferred cells in SI LP was analyzed on Day 2 and Day 14 after transfer. Cumulative from several independent experiments, N=5 mice/group.
(J) TCF1mCherry+IL-17GFP+IL-10Venusneg TH17 cells were FACS-purified from SI LP of SFB-colonized or LI LP of Citrobacter rodentium-infected mice and stimulated in vitro as described in Methods. (Left) IL-10Venus and IL-17GFP expression in CD4 T cells. (Right) Proportion of IL-10Venus+ cells in TCF1mCherrynegIL-17GFP+ TH17 cells on Day 4. Three independent experiments, N=2–7 mice/group.
(K) PCA plot of bulk RNA-sequencing analysis of FACS-sorted TCF1mCherry+IL-17GFP+ and TCF1mCherrynegIL-17GFP+ TH17 cells from SI LP of SFB-colonized or LI LP of Citrobacter rodentium-infected (Crod) mice. One experiment, N=2–4 mice/group.
(L) Number of differentially expressed genes (DEGs) in indicated pairwise comparisons of RNA-sequencing analysis in (K). One experiment, N=2–4 mice/group.
(M) Gene set-enrichment analysis of genes (Left) upregulated in TCF1+ SFB TH17 cells compared to genes upregulated in total SFB TH17 cells or (Right) upregulated in TCF1+ Crod TH17 cells compared to total Crod TH17 cells.
(N) Heatmap of DEGs arranged by the comparison between TCF1+ SFB and TCF1+ Crod TH17 cells. SFB core signature anti-inflammatory genes in blue and inflammatory genes in red are listed on the right.
(O) Quantitative PCR for selected SFB signature genes in samples in K
(P) TCF1mCherry+IL-17GFP+IL-10Venusneg TH17 cells were FACS-purified from SI LP of SFB-colonized mice and stimulated in vitro in with or without 10 ng/ml IL-1β and 10 ng/ml IL23. (Left) IL-10Venus and IL-17GFP expression in CD4 T cells. (Right) Proportion of IL-10Venus+ cells in TCF1mCherrynegIL-17GFP+ TH17 cells on Day 4. Two independent experiments, each dot represents a technical replicate.
(Q) IFN-γ ELISA from in vitro cultures in (O). Two independent experiments, each dot represents a technical replicate.
To further examine the phenotype of TCF1+ progenitor TH17 cells we performed bulk RNA-Seq analysis on purified TCF1+ and TCF1neg SI LP TH17 cells from SFB-colonized or Crod-infected Tcf7mCherry/Il17aGFP reporter mice. SFB TCF1+ TH17 progenitors differed significantly from Crod TH17 progenitors (Figure 6K, L). Gene set enrichment analysis showed that SFB TCF1+ progenitors were enriched in the core SFB anti-inflammatory signature compared to Crod TCF1+ progenitors, which resembled the general Crod TH17 program (Figure 6M). For both types of microbes, the transcriptional program of progenitor TH17 cells most closely resembled that of the corresponding TCF1neg TH17 cells (Figure 6K) with TCF1+ and TCF1neg SFB TH17 cells overlapping most closely and expressing the least number of differentially expressed genes (DEGs) (Figure 5K, L). SFB TH17 cell signature anti-inflammatory genes were enriched in TCF1+ and TCFneg TH17 cells compared to Crod TH17 cells and, vice versa, inflammatory markers were enriched in Crod TCF1+ TH17 cells (Figure 6M, N and S6H). Il10, Maf, Tox and other genes associated with the SFB TH17 program were already upregulated in TCF1+ SFB progenitors, and further increased in TCFneg SFB effectors (Figure 6N, O and S6H, I). We next investigated whether inflammatory cytokines could affect the transcriptional program of commensal TH17 progenitors. In vitro stimulation of purified SI LP TCF1+IL-17+IL-10neg TH17 cells in the presence of IL-1β and IL-23 resulted in significant decrease in their ability to differentiate into IL-10+ effector TH17 cells (Figure 6P) and instead induced production of IFNγ (Figure 6Q). Combined our results suggest that LP TCF1+ progenitor commensal TH17 cells are transcriptionally poised to differentiate to anti-inflammatory effectors but retain the ability to respond to inflammatory queues from the environment.
IL-10 signaling and intestinal macrophages in terminal ileum instruct acquisition of TH17 anti-inflammatory phenotype
We next investigated the signals and participating innate immune cells that facilitate the differentiation of TCF1+ progenitors into IL-10-expressing anti-inflammatory TH17 cells. TCF1+ progenitor TH17 cells were present exclusively in the SI LP and not in mLN, and adoptively transferred TCF1+ progenitor TH17 cells homed exclusively to the SI LP (Figure S7A). In addition, although TCF1+ TH17 cells were present in both duodenum and ileum, TCFnegIL-10+ TH17 cells were specifically present in the terminal ileum (Figure 7A, B). Purified ileal TCF1+ SFB TH17 cells had a significantly increased capacity to generate TCF1neg IL-10+ TH17 cells in vitro, compared to TCF1+ SFB TH17 cells from duodenum or SI LP TCF1+ TH17 cells from Citrobacter-infected mice (Figure S7B). The foregoing results suggest that commensal precursor TH17 cells acquire IL-10 expression locally in the terminal ileum under the guidance of signals from the gut microenvironment.
Figure 7. IL-10 signaling in intestinal Mϕs drives generation of anti-inflammatory commensal TH17 cells in the terminal ileum.

(A) Intracellular staining for TCF1 and IL-17 in duodenum (Duo) and terminal ileum (Ile) SI LP of SFB-colonized and SFB-negative WT mice. (Left) FACS plots from SFB-colonized mice, gated on TCRβ+CD4+ lymphocytes. (Right) Proportion of TCF1neg effector cells in TH17 cells, gated TCRβ+CD4+IL-17+ (Right). Two independent experiments, N=3 mice/group.
(B) Distribution of IL-10GFP+ TH17 cells in duodenum (Duo) and terminal ileum (Ile) of SFB-colonized and SFB-negative Il10eGFP/Il17aKatushka/Foxp3mRFP mice. (Left) FACS plots from SFB-colonized mice gated on TCRβ+CD4+lymphocytes. (Right) Proportion of IL-10GFP+ cells in TH17 cells (TCRβ+CD4+IL-17Katushka+). Two independent experiments, N=4 mice/group.
(C-E) Naïve SFB-specific 7B8 splenic CD4 T cells were purified from 7B8.Ly5.1 Il10GFP/Il17aKatushka/Foxp3mRFP mice and adoptively transferred into SFB-colonized Ly5.2 WT or Il10−/− mice (C). Expression of IL-10 (GFP) (D) and c-MAF (intracellular staining) (E) in SI LP one week after transfer. FACS plots in (D) gated on Ly5.1+TCRβ+CD4+Foxp3mRFPneg 7B8 CD4 T cells. Bar plots in (D) further gated on IL-17Katushka+ 7B8 TH17 cells. Bar plots in (E) further gated on IL-17+ transferred 7B8 TH17 cells. Cumulative of three independent experiments, N=5–6 mice/group.
(F, G) Naïve SFB-specific 7B8 splenic CD4 T cells were purified from 7B8.Ly5.1 Il10GFP/Il17aKatushka/Foxp3mRFP mice and adoptively transferred into SFB-colonized WT or Cd4Cre/Il10flox/flox (Il10ΔT) mice. Expression of IL-10 (GFP) (F) and c-MAF (G) in SI LP one week after transfer. FACS plots in (F) gated on Ly5.1+TCRβ+CD4+Foxp3mRFP-neg 7B8 CD4 T cells. Bar plots further gated on IL-17Katushka+ (F) or IL-17+ (G) transferred 7B8 TH17 cells. Cumulative of two independent experiments, N=6–7 mice/group.
(H) Experimental schematic. Naïve splenic CD4 T cells were purified from Il10GFP/Il17aKatushka/Foxp3mRFP (Ly5.2) WT or Il10rb−/− mice and adoptively transferred into SFB-colonized Ly5.1 WT mice.
(I) IL-10 and c-MAF expression in transferred TH17 cells in SI LP two weeks after transfer from the mice in (H). FACS plots and bar plots gated on Ly5.2+TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+ TH17 cells. Cumulative of four independent experiments, N=8 mice/group.
(J, K) Naïve SFB-specific 7B8 splenic CD4 T cells were purified from 7B8.Ly5.1 Il10GFP/Il17aKatushka/Foxp3mRFP mice and adoptively transferred into SFB-colonized Ly5.2 WT or Il10rb−/− mice. IL-10 (GFP) (J) and c-MAF (K) expression in SI LP one week after transfer. FACS plots gated on Ly5.1+TCRβ+CD4+Foxp3mRFPneg transferred 7B8 T cells. Bar plots further gated on IL-17Katushka+ (J) or IL-17+ (K) transferred 7B8 TH17 cells. Cumulative of two independent experiments, N=4 mice/group.
(L) Experimental schematic. Naïve SFB-specific 7B8 splenic CD4 T cells were purified from 7B8/Ly5.1 Il10GFP/Il17aKatushka/Foxp3mRFP mice and adoptively transferred into DT-treated SFB-colonized Ly5.2 WT BM chimeras, reconstituted with 1:1 mix of BM from Ccr2DTR mice and either WT or Il10rb−/− mice. DT treatment was performed to deplete Ccr2DTR macrophages as described in Methods.
(M) IL-10 (GFP) expression in SI LP one weeks after transfer from the mice in (L). FACS plots gated on Ly5.1+TCRβ+CD4+Foxp3mRFPneg transferred 7B8 CD4 T cells. Bar plots further gated on IL-17Katushka+ transferred 7B8 TH17 cells. Cumulative of two independent experiments, N=8–9 mice/group.
(N) Quantitative PCR of Maf transcripts in FACS-purified transferred SI LP 7B8 TH17 cells (Ly5.1+TCRβ+CD4+Foxp3mRFPnegIL-17GFP+) from the mice in (L). Cumulative of two independent experiments, N=3 mice/group.
IL-10 induces TR1 cell differentiation in vitro51 and is required for the maintenance of IL-10 expression in TR1 cells and Foxp3+ Tregs52,53. We, therefore, investigated the role of IL-10 in generation of IL-10-producing SFB TH17 cells. For this, we crossed Il10GFP/Il17aKatushka/Foxp3mRFP reporter mice to SFB-specific 7B8 TCR Tg mice on a Ly5.1 congenic background. We then adoptively transferred naïve 7B8.Ly5.1-triple reporter CD4 T cells into WT and IL-10-deficient animals and examined SFB-specific TH17 cell induction, as well as the phenotype of the induced TH17 cells (Figure 7C). 7B8 CD4 T cells differentiated into IL-10-expressing TH17 cells in SI LP of WT control animals (Figure 7D). In contrast, although WT 7B8 CD4 T cells downregulated TCF1 and became TH17 cells in Il10−/− mice, they had significantly decreased proportion of IL-10+ TH17 cells (Figure 7D and Figure S7C, D). Moreover, 7B8 CD4 TH17 cells had decreased expression of c-MAF in the absence of environmental IL-10 (Figure 7E). Therefore, IL-10 is required for the induction of c-MAF and IL-10 in commensal TH17 cells. To investigate the source of IL-10, we next transferred triple reporter 7B8 Tg CD4 T cells into recipients with conditional deletion of IL-10 in T cells (Cd4Cre/Il10flox/flox or Il10ΔT mice). Despite similar TH17 cell differentiation, 7B8 TH17 cells had decreased c-MAF and IL-10 expression in the absence of IL-10 production by T cells (Figure 7F, G and S7E). These results suggest that IL-10 production by CD4 T cells is required for induction of IL-10 expression by commensal TH17 cells. To investigate whether IL-10 acts directly on the differentiating commensal TH17 cells, we transferred control and Il10rb−/− triple reporter CD4 T cells into WT recipients (Figure 7H). IL-10Rβ-deficient CD4 T cells differentiated into TH17 cells similarly to controls (Figure S7F) and contained similar proportion of c-MAF+ and IL-10+ TH17 cells (Figure 7I and Figure S7G). In contrast, despite unimpeded TH17 cell differentiation, WT SFB-specific CD4 T cells did not become IL-10+ or c-MAF+ TH17 cells when transferred into IL-10Rβ-deficient recipients (Figure 7J, K and S7H). Combined, these results suggest that IL-10 does not directly act on differentiating commensal TH17 cells. We previously reported that intestinal macrophages (iMf) participate in the induction of SFB-specific TH17 cells54. Moreover, IL-10Rβ signaling in iMϕ is crucial for establishment of intestinal homeostasis55. We, therefore, investigated whether IL-10Rβ signaling in iMϕ is required for induction of IL-10 expression by SFB TH17 cells. To conditionally delete IL-10Rβ in iMϕ we generated mixed bone marrow (BM) chimeras in which lethally irradiated WT mice were reconstituted with a 1:1 BM mixture from CCR2-DTR and Il10rb−/− animals (Figure 7L). We previously showed that diphtheria toxin (DT) injection leads to specific loss of iMfϕ, but not intestinal dendritic cells (iDCs) in CCR2-DTR animals54. Administration of DT in the mixed chimeras leads to deletion of CCR2-DTR iMfϕ, but not Il10rb−/− iMfϕ, resulting in an iMfϕ population that specifically lacks IL-10Rb expression (Figure S7I). SFB-specific CD4 T cells differentiated into IL-10+ TH17 cells when transferred into DT-treated control CCR2-DTR:WT BM chimeras (Figure 7M). In contrast, although 7B8 CD4 T cells differentiated similarly to TH17 cells in recipients with conditional deletion of IL-10Rβ in iMfϕ (Figure S7J), these TH17 cells had significantly decreased IL-10 and c-MAF expression (Figure 7M, N). Altogether the foregoing results suggest that iMfϕ detect T cell-derived IL-10 to induce or maintain production of IL-10 by commensal TH17 cells.
Discussion
TH17 cells are defined by the expression of the signature cytokine IL-17A. Although TH17 cells were originally described as pro-inflammatory, it is now appreciated that there is a considerable range in TH17 cell functionality12,13,15,22,48,49. Homeostatic non-pathogenic TH17 cells have been described, but their functions have not been defined. In the gut, the role of commensal-induced TH17 cells is unclear. Although absence of SFB TH17 cells leads to slight SFB increase in the gut lumen10, control of SFB is mainly mediated by type 3 innate lymphoid cells56. SFB TH17 cells were originally considered pro-inflammatory. However, it was later shown that SFB TH17 cells possess a non-pathogenic transcriptional program24. Our results demonstrate that SFB TH17 cells have a regulatory anti-inflammatory program and can produce IL-10. We further found that Bifidobacterium adolescentis induces TH17 cells with a similar phenotype, which suggests that IL-10 production by TH17 cells is characteristic of multiple commensal species. Thus, commensal TH17 cells may also play role in maintaining mucosal homeostasis. Indeed, intestinal SFB TH17 cells prevent metabolic disease in the context of diet-induced obesity57. In addition, herein we report that SFB TH17 cells suppress intestinal effector T cell responses via IL-10.
Intestinal IL-10+ TH17 cells were previously reported in an experimental model of intestinal inflammation and shown to transdifferentiate to TR1 cells22. SFB-induced IL-10+ TH17 cells in the current study also express TR1-associated genes. However, using scRNA-Seq of YFP+ CD4 T cells from Il17aCre/R26STOP-YFP mice we found few YFP+ CD4 T cells without IL-17 transcripts (ex-TH17 cells) in WT animals, and therefore little evidence for trans-differentiation of SFB TH17 cells at steady state. This is also in agreement with a prior study that concluded that SI LP SFB TH17 cells possess little plasticity24. In contrast, after TH17-specific ablation of c-MAF we found a considerable population of ex-TH17 cells, which expressed pro-inflammatory genes. Therefore, c-MAF not only maintains IL-10 expression in commensal TH17 cells, but also prevents trans-differentiation into pro-inflammatory CD4 T cells. Whether commensal TH17 cells can become fully functional TR1 cells remains to be investigated.
In our study, SFB TH17 cells inhibited expansion and cytokine production of effector CD4 T cells in an IL-10 and c-MAF-dependent manner. However, they expressed several inhibitory receptors. Indeed, in our hands, blockade of CTLA-4, but not LAG-3, also partially inhibited suppression in the in vitro assay. Therefore, commensal TH17 cells may possess additional mechanisms for maintaining T cell homeostasis, besides IL-10. In addition, in our in vivo experiments SFB TH17 cells suppressed SFB-specific CD4 T cells. Therefore, whether commensal TH17 cells can regulate non-cognate CD4 T cells remains to be investigated.
c-MAF plays divergent roles in the specialization of IL-17 producing T cells. c-MAF is not essential for RORγ expression in TH17 cells and TH17 cell differentiation38. However, c-MAF is activated early during TH17 cell polarization together with TH17-defining transcription factors and can act as a negative regulator36. In contrast, c-MAF is required for the development of RORγt+ regulatory T cells and for the specialization of IL-17+ γδ T cells, where it acts as an activator38,58,59. Regardless of its overall role, c-MAF is universally linked to positive regulation of IL-10 expression in T cells, including TH17 cells36,58. Here, we find that c-MAF is required for the production of IL-10 by commensal TH17 cells. Moreover, c-MAF was required not only for IL-10 production, but in general for the maintenance of the anti-inflammatory program of commensal TH17 cells. This included the expression of tissue repair factors and co-inhibitory receptors. TH17 cell-specific ablation of c-MAF led to expansion of IFNγ+ TH17 cells and IL-17neg ex-TH17 cells with a pro-inflammatory TH1 phenotype. Thus c-MAF may also maintain the anti-inflammatory phenotype of commensal TH17 cells by restricting inflammatory cytokines and TH17 cell plasticity.
We find that IL-10+ TH17 cells were only present in the terminal ileum. Therefore, signals in this location likely mediate the induction or maintenance of their anti-inflammatory program. We previously showed that intestinal epithelial cells and intestinal macrophages play crucial roles in SFB TH17 cell induction54,60. Here, we find that ablation of IL-10Rβ in macrophages perturbs induction of anti-inflammatory TH17 cells but does not affect overall TH17 cell differentiation. Therefore, intestinal macrophages are required for the induction or maintenance of anti-inflammatory TH17 cells. IL-10Rβ signaling in macrophages is required for maintenance of intestinal homeostasis55. Our data suggest that maintenance of anti-inflammatory commensal TH17 cells may contribute to the mechanisms by which resident macrophages suppress gut inflammation. Although the exact source of IL-10 required for iMϕ activation remains to be ascertained, we find that IL-10 from CD4 T cells is required for the presence of IL-10+ TH17 cells. Both Foxp3+ Tregs and Foxp3neg TR1 cells can produce IL-10 thus establishing an interdependent network of IL-10 producing CD4 T cells in mucosal homeostasis.
Pathogens induce quantitatively different T cell responses in the context of acute versus chronic infection. Whether commensals engage adaptive immunity in an acute or chronic manner is not known. In agreement with the expression of IL-10 and generally inhibitory or non-responsive program, commensal TH17 cells closely resembled exhausted T cells induced during chronic infection. Therefore, in terms of T cell responses, presence of commensals resembles chronic infection. We also found significant heterogeneity of commensal TH17 cells and the existence of a precursor TCF1+ TH17 cell population in the SI LP that generates TCF1neg IL-10+ TH17 cells. Similar TCF1+ progenitor CD4 and CD8 T cells maintain TCFneg effector responses in the context of chronic viral infections61,62, further underscoring the similarities between homeostatic commensal and chronic infection T cell responses. SFB TCF1+ progenitor TH17 cells were transcriptionally distinct from TCF1+ TH17 cells during Citrobacter rodentium infection. Even though they retained the potential to generate inflammatory TH17 cells, intestinal TCF1+ TH17 cells closely resembled their TCF1neg counterparts. Therefore, they were already poised towards an anti-inflammatory program. Our results suggest that this happens in the SI LP under the control of the local microenvironment. The specific signals controlling this transition, as well as the earliest events leading to the establishment of the SFB TH17 cell differentiation program will be important to elucidate in future studies.
TCFneg commensal TH17 cells possessed inhibitory or activated phenotype and contained both IL-10+ and IL-10neg TH17 cells. Therefore, individual commensals generate heterogeneous T cell responses. We identified two unique subsets of IL-10+ TH17 cells, both of which required c-MAF for IL-10 production. Whether these two subsets perform different functions or whether commensal TH17 subsets with distinct functions co-exist, will be important to elucidate in future studies.
Our results describe an inherent heterogeneity of the TH17 cell response to commensal microbes and show that such response may function not only in antigen-specific control of the inducing commensal, but also in general regulation of intestinal T cells in maintaining anti-inflammatory tone of the intestinal mucosa.
STAR Methods text
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Ivaylo Ivanov (ii2137@cumc.columbia.edu).
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Animals
C57BL/6J, Ly5.1 (CD45.1), RAG1-deficient, Il17aGFP, Il17aCre, Il10GFP, Foxp3mRFP, Il10−/−, Il10rb−/− and 7B8 transgenic mice were purchased from the Jackson Laboratory. Animals were purchased only from SFB-negative maximum barrier rooms at Jackson. All animals were tested for SFB upon arrival and maintained in an SFB-negative high barrier room at Columbia University. 7B8 mice were bred to Ly5.1 and Il17aGFP mice at Columbia University. Il17aKatushka mice63,64 were provided by Dr. Samuel Huber, Medical Center Hamburg-Eppendorf (UKE) with permission from Dr. Richard Flavell, Yale and bred at Columbia University. 7B8 mice were bred to Ly5.1 and to Il10GFP/Il17aKatushka/Foxp3mRFP mice at Columbia University. Maffl/fl on C57BL/6 background were obtained from Dr. Nicola Gagliani, University Medical Center Hamburg-Eppendorf and Dr. Arnold Han, Columbia University with permission from Dr. Carmen Birchmeier and bred at Columbia University. Ccr2DTR mice65 were gifted by Dr. Eric Parmer, Memorial Sloan-Kettering Cancer Center. Il10flox mice66 were obtained from Dr. A. Roers, Technische Universitat Dresden and bred to Cd4Cre mice at Columbia University. Il10Venus mice4 were gifted by Dr. Kenya Honda, Keio University with permission from Dr. Kiyoshi Takeda, Osaka Unviersity and bred to Tcf7mCherry mice at Columbia University. Tcf7mCherry mice were generated using CRISPR/Cas9 based gene editing in C57BL/6J mice. The targeted vector contains an mCherry reporter sequence preceded by a splice acceptor site and a P2A self-cleaving sequence placed in intron 2 and surrounded by a pair of non-complementary LoxP sites (Figure S5). The targeting construct also contained an inversion of the Tcf7 genomic sequence containing Exons 3–4 surrounded by two pairs of LoxP and LoxP2272 sequences in opposite orientation in intron 2 and intron 5. The targeted reporter allele therefore expresses mCherry and is a functional knock-out for TCF1 that can be conditionally activated upon expression of Cre-recombinase (Figure S5). Cre-recombinase was not used in this study and all animals used were heterozygous or Tcf7mCherry/+. All mouse strains were bred and housed under specific pathogen-free conditions at Columbia University Medical Center under IACUC approved guidelines. To control for microbiota and cage effects, experiments were performed with gender matched littermate control animals that were housed in the same cage.
METHOD DETAILS
SFB colonization and quantification
SFB colonization was performed by single oral gavage of fecal suspension from SFB-enriched mice as previously described42. To control for variability in SFB levels in feces used for gavage, all gavages were performed with frozen stocks from a single batch of SFB-enriched feces. Fecal samples were tested for SFB by quantitative RT-PCR and frozen as batch aliquots at −80C. Control SFB-negative feces were collected in a similar manner. SFB colonization levels were confirmed by qPCR and normalized to levels of total bacteria (UNI) as previously described42.
Citrobacter rodentium infection
Mice where infected with 1 × 109 CFU of Citrobacter rodentium by oral gavage. Infection was confirmed by measuring CFU in fecal samples throughout the course of infection.
Bifidobacterium adolescentis and Escherichia coli gavage
Mice were gavaged with Bifidobacterium adolescentis (Ba) or Escherichia coli (Ec) every other day for 14 days. Ba was grown in Reinforced Clostridial Medium in an anaerobic chamber (5% H2, 10% CO2, 85% N2) at 37C for 48 hours. Ec was grown overnight in Luria-Bertani (LB) broth at 37C. Ba and Ec were gavaged in 200 μl PBS/mouse. Mice received 1×108 CFU/ml per gavage of either Ba or Ec.
Transfer colitis
FACS-sorted CD45RBhigh CD4 T cells from spleen and lymph nodes were injected i.v. (5×105 cells/mouse) into RAG1-deficient mice. Lipocalin-2 in fecal samples was measured by ELISA.
In vitro suppression assay
Responder naïve CD4 T cells (WT or Il10rb−/−) isolated from spleen were purified via FACS (CD4+TCRβ+CD62+CD44negCD25neg). Responder CD4 T cells were labeled with 5 μM CellTrace violet dye (proliferation dye, Invitrogen) and stimulated in the presence of irradiated splenic APCs (25 Grey) and 1 μg/ml soluble anti-CD3 (clone 2C11). Responder CD4 T cells were cultured in the absence or presence of indicated SI LP CD4 T cells in a 2:1 ratio for 4 days. SFB and Crod TH17 cells (CD4+TCRβ+IL-17GFP+) were isolated from SI LP or LI LP of Il17aGFP reporter mice two weeks after SFB gavage or Crod infection respectively. Foxp3+ Treg cells were isolated from SI LP of Foxp3mRFP reporter mice. To assess the role of IL-10 signaling and co-inhibitory receptors, blocking antibodies against IL-10R, CTLA-4 or LAG-3 were added to some of the cell culture (anti-mouse IL-10R (1B1.3A), 10 μg/ml, Bio X Cell; anti-mouse CTLA-4 (63828), 10 μg/ml, R&D Systems; anti-mouse LAG-3 (C9B7W), 10 μg/ml, Bio X Cell). Division Index (DI) was calculated with FlowJo based on the divisions of responder T cells. Percent suppression was calculated using the following formula:
In vivo suppression assay
Naïve 7B8 CD4 T cells were isolated from spleen of 7B8.Ly5.1 Il17aGFP transgenic mice (Ly5.1) by FACS (Ly5.1+Vβ14+CD4+TCRβ+CD62L+CD44negCD25neg). Intestinal SFB TH17 cells (CD4+TCRβ+IL-17GFP+) and Foxp3+ Treg (CD4+TCRβ+Foxp3mRFP+IL-17Katushkaneg) cells were isolated from SI LP of Il17aGFP or Foxp3mRFP Ly5.2 mice respectively by FACS two weeks after SFB colonization. 30,000 naïve 7B8 CD4 T cells and 30,000 SI LP cells were injected intravenously in a 1:1 ratio into SFB-colonized Rag1−/− mice. Expansion and TH17 cell differentiation of Ly5.1+ 7B8 CD4 T cells was analyzed in SI LP and mLN on day 8 post transfer. To assess the role of IL-10 signaling, anti-IL-10R antibody (200 μg/mouse clone 1B13A, Bio X Cell) or an isotype control antibody were injected on day 2, 4 and 6 post transfer.
Adoptive transfers
SFB-negative WT, Il10−/−, Il10rb−/−, or Il10ΔT mice were gavaged with SFB-containing fecal pellets as described above. Five days after gavage, MACS-purified (CD4 beads, Miltenyi) 7B8 or total CD4 T cells from spleen and LN of SFB-negative (naïve) 7B8.Ly5.1 Il10eGFP/Il17aKatushka/Foxp3mRFP or Ly5.1 Il10eGFP/Il17aKatushka/Foxp3mRFP mice were transferred intravenously (5×105 7B8 cells/recipient or 2×106 total CD4 T cells/recipient).
Migration assays
Il17aGFP reporter mice or Tcf7mCherry/Il17aGFP double reporter mice were gavaged with SFB-containing feces as described above. Two weeks after gavage, SI LP lymphocytes were isolated and SFB TH17 cells (Ly5.1+CD4+TCRβ+IL-17GFP+) or TCF1+ SFB TH17 cells (Ly5.1+CD4+TCRβ+IL-17GFP+TCF1mCherry+) were FACS-purified. 50,000 SI LP TH17 cells (combined from multiple mice) were injected intravenously into SFB-colonized WT mice (Ly5.2). Cells were isolated from mLN, LI LP, SI LP and liver at indicated timepoints to examine transferred Ly5.1+ CD4 T cell.
Mixed bone marrow chimeras
Total bone marrow cells were isolated from Ccr2DTR mice, Il10rb−/− and WT C57BL/6 mice (all Ly5.2). After removal of red blood cells, Ccr2DTR bone marrow cells were mixed in a 1:1 ratio with Il10rb−/− or WT bone marrow cells and five million total cells were transferred into lethally irradiated (11 Grey) recipient WT Ly5.1 mice. 12 weeks later, mice were colonized with SFB as described above. The mice were treated with 20 ng/g diphtheria toxin (DT) every other day starting on Day −1. Ly5.1/Ly5.2 7B8 triple reporter CD4 T cells were transferred on Day 0 as described earlier. TH17 cell differentiation in SI LP was analyzed on Day 7. For Q-PCR analysis, intestinal cells were FACS-purified and sorted into TRIZOL reagent (Life technology).
Lymphocyte isolation from intestine
Lamina propria lymphocytes isolation from intestine was performed as previously described42. In brief, Peyer’s patched (SI) were removed and intestines were opened longitudinally. After washing, the intestines were cut into 1 cm long pieces and incubated in 5 mM EDTA solution twice for 20 min at 37°C. Lamina propria lymphocytes were isolated by digesting the tissue with Collagenase D, DNAse and Dispase three times for 20 min at 37°C. Lymphocytes were further purified using 80:40 Percoll gradient.
In vitro culture
Tcf7mCherry/Il17aGFP/Il10Venus triple reporter mice were gavaged with SFB-containing feces or infected with Citrobacter rodentium as described above. Lymphocytes were isolated two weeks later from terminal ileum (distal quarter of SI) or duodenum (proximal quarter of SI) (SFB-gavaged) or LI LP (Crod-infected). TCF1+ TH17 cells (CD4+TCRβ+IL-17GFP+TCF1mCherry+IL-10Venusneg) were FACS-purified from individual mice and plated in 96-well plates coated with 5 μg/ml aCD3 antibody (clone 2C11) in the presence of 5 μg/ml soluble aCD28 antibody (clone 37.51) for four days. Additionally, FACS-sorted SFB TCF1+ TH17 cells (CD4+TCRβ+IL-17GFP+TCF1mCherry+IL-10Venusneg) were plated in 96-well plates coated with 5 μg/ml aCD3 antibody (clone 2C11) in the presence of 5 μg/ml soluble aCD28 antibody (clone 37.51), 10 ng/ml IL-23 and 10 ng/ml IL-1β for four days.
Lipocalin-2 ELISA
Lipocalin-2 was measured in fecal pellets from colitogenic mice. Fecal pellets were weight and disrupted in PBS containing cOmplete protease inhibitor (Roche). After centrifugation, supernatant was collected and stored in −80C until Lipocalin-2 ELISA was performed. ELISA was performed according to manufacturer protocol.
IFN-γ ELISA
Cell culture supernatants were collected from in vitro cultures of SFB TCF1+ TH17 cells after four days in the presence or absence of IL-23 and IL-1β. IFN-γ ELISA was performed according to the manufacturer protocol.
Flow Cytometry
After isolation cells were analyzed immediately by flow cytometry. For intracellular cytokine and transcription factor staining, the cells were re-stimulated with PMA/Ionomycin for 3 hours in the presence of Brefeldin A, followed by fixation and permeabilization using Foxp3/transcription factor staining buffer kit (Tonbo) according to manufacturer protocol. Dead cells were excluded with fixable viability dye (eFluor506, Invitrogen).
Quantitative PCR
mRNA from FACS-sorted cells was isolated using TRIZOL reagent (Life technology) according to the manufacturer protocol. Reverse transcription was performed with QScript cDNA SuperMix (QuantaBio). Q-PCR was performed using SYBR Green on LightCycler 480 (Roche). Samples were analyzed using the ΔΔCt method and normalization to Gapdh.
Bulk RNA-sequencing and analysis
LP TCRβ+CD4+IL-17GFP TH17 cells were purified via FACS from small or large intestine two weeks after gavage with SFB or infection with Citrobacter rodentium, or 10 weeks after colitis induction (CD45RBhi colitis). Total mRNA was isolated using TRIZOL (Life technology) as per the manufacturer protocol. RNA-sequencing (RNA-Seq) was performed at the JP Sulzberger Columbia Genome Center. RNA amplification and library preparation was performed using the CLONTECH kit by Takara Bio. Libraries were then sequenced using Illumina NovaSeq 6000 (~40M reads). RTA (Illumina) was used for base calling and bcl2fastq2 (version 2.20) for converting BCL to FASTQ format. Raw reads were then processed by Cutadapt v2.1 with the following parameters: ‘--minimum-length 30:30 -u 15 -u −5 -U 15 -U −5 -q 20 --max-n 0 --pair-filter=any’ to remove low-quality bases and Illumina adapters. Next, pseudoalignment was performed against the index created from mouse transcriptomes (GRCm38) using Kallisto (0.44.0). Differential gene expression analysis was performed by DESeq2 using reads count estimated by pseudoalignment, and the sets of significantly differentially expressed genes were identified using the following steps: First, genes that were not significantly changed were excluded (padj < 0.05). Next, genes with very low expression level (transcripts per million, TPM < 5 in at least 9 out of the 10 samples) were also excluded. Finally, an unusually high level of Ig gene transcripts was observed in a few samples and, therefore, Ig genes were excluded from the analysis.
Gene set enrichment analysis (GSEA)
To identified if curated signature gene sets or other specific gene sets are significantly up-regulated or down-regulated compared to published datasets, we performed gene set enrichment analysis. Briefly, normalized gene expression levels by microarray or RNA-seq were obtained from NCBI Gene Expression Omnibus or the original publication. Next, fold-changes of gene expression between comparisons were calculated in R v.4.1.0, and normalized enrichment scores as well as p-values of given gene sets were then estimated using the fgsea R package v.1.24.0 with the setting “nperm=1000”.
Identification of c-MAF target genes
To identify potential c-MAF target genes, we extracted results from a regulatory network analysis for TH17 cell36 that integrated ChIP-seq data and RNA-seq data. Briefly, the summed scores for KC network of c-MAF were extracted from the original publication and genes with a score greater than 2 were defined as c-MAF target genes.
Single cell RNA-sequencing and analysis
SI LP SFB TH17 cells (TCRβ+CD4+Foxp3mRFPnegIL-17Katushka+) were FACS-sorted from SFB-colonized Il10eGFP/Il17aKatushka/Foxp3mRFP mice. In a second set of experiments, TH17 cells (TCRβ+CD4+IL-17YFP+) were FACS-sorted from SI LP of Foxp3mRFP/Il17aCre/Mafflox/flox/R26STOP-YFP mice (MafΔIL17) (n=2) and Foxp3mRFP/Il17aCre/Mafflox/+/R26STOP-YFP (WT) mice (n=3) two weeks after SFB gavage. Prior to sorting, cells from individual animals were labelled using hashtag antibodies conjugated to nucleotide barcodes (BioLegend, #155831, #155833, #155835). scRNA-seq was performed at the JP Sulzberger Columbia Genome Center using the 10X Genomics platform with a target of 5,000 nuclei per sample and 130M reads. Next, reads alignment, filtering, and barcode counting were performed using Cell Ranger v.3.0.2. All single-cell analyses were performed using R v.4.1.0 and Python v.3.6. Briefly, Seurat v.4.0.5 was utilized for preprocessing, normalization, and clustering. Ggplots2 v.3.3.5 was used to generate UMAP and dot plots. Low quality cell profiles were excluded if they met one of the following criteria: (i) number of genes expressed 200 or 2500 or (ii) 5% of the total unique molecular identifiers (UMIs) were mitochondrial RNA. The data was then normalized using the NormalizeData function. Wild type and MafΔIL17 TH17 cells were integrated using the SCTransform and FindIntegrationMarkers. Next, the RunPCA function was applied followed by FindNeighbors and FindClusters functions on the number of PCs selected using the ElbowPlot function. Marker genes that were differentially expressed within each cluster were identified by the FindAllMarkers function with average log-transformed fold change cutoffs of 0.25 and pct cutoffs of 0.25. Gene set scoring was performed using the VISION R package v.3.0.067. Gene set enrichment scores and p-values were computed using the fgsea R package v.1.24.0, a fast algorithm for Gene Set Enrichment Analysis (GSEA). Genes were ranked utilizing the wilcoxauc function from the presto R package, which performs a Wilcoxon rank sum test. COMET Python package68 was applied to predict cell surface markers for clusters of interest. COMPASS Python package48 was applied to characterize cellular metabolic states for clusters of interest. Slingshot v.2.2.069 was used for trajectory analysis starting at the progenitor-like population (C4).
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical significance was determined by unpaired t test with Welch’s correction or other methods as noted on figure legends. P values are represented on figures as follows: ns, not significant, * p < 0.05, ** p < 0.01, *** p < 0.005, **** p < 0.001, ***** p < 0.0005. Error bars on all figures represent standard error of the mean. Statistical analysis was performed using GraphPad Prism version 9.1 for Windows (GraphPad Software).
Supplementary Material
KEY RESOURCES TABLE.
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| Rat anti-mouse CD4 antibody, RM4-5, BUV737 | BD | #612844 |
| TCR beta monoclonal antibody, H57-597, APC-eFluor780 | eBioscience | #47-5961-82 |
| Anti-mouse CD45.1, A20, PerCP-Cyanine5.5 | Tonbo | #50-210-3580 |
| Mouse anti-mouse CD45.2, 104, BV421 | BD | #562895 |
| Anti-Human/Mouse CD45R (B220), RA3-6B2, PerCP-Cyanine5.5 | Tonbo | #65-0452-U100 |
| CD103 Monoclonal antibody, 2E7, PE | eBioscience | #12-1031-82 |
| CD11b Monoclonal antibody, M1/70, APC-Cyanine7 | Invitrogen | #A15390 |
| CD11c Monoclonal antibody, N418, PE-Cyanine7 | eBioscience | #25-0114-82 |
| Anti-mouse CD24 antibody, M1/69, BV510 | BioLegend | #101831 |
| CD62L Monoclonal antibody, MEL-14, FITC | eBioscience | #11-0621-82 |
| Anti-mouse CD64, X54-5/7.1, APC | BioLegend | #139334 |
| Anti-mouse CD69, H1.2F3, PE-Cyanine7 | Tonbo | #60-0691-U025 |
| CD127 monoclonal antibody, A7R34, PE | eBioscience | #12-1271-82 |
| CD223 monoclonal antibody, eBioC9B7W, PerCP-eFluor710 | eBioscience | #46-2231-82 |
| Rat monoclonal anti mouse MHCII, M5/114.15.2, Alexa Fluor 710 | Tonbo | #80-5321-U100 |
| American Hamster monoclonal anti-gδTCR, GL-3,GL3 APC | eBioscience | #17-5711-82 |
| CD366 monoclonal antibody, F38-2E2, APC | eBioscience | #17-3109-42 |
| Mouse monoclonal anti FoxP3, FJK-16s, BV421 | eBioscience | #404-5773-82 |
| Fixable Viability Dye eFluor 506 (FVD) | Invitrogen | #65-0866-14 |
| Rat monoclonal anti-mouse IFNγ, XMG1.2, APC | eBioscience | #17-7311-82 |
| Rat monoclonal anti-mouse IL-17A, eBio17B7, FITC | eBioscience | #11-7177-81 |
| Goat monoclonal anti IL-22 antibody (POLY5164) | Biolegend | #516406 |
| Anti-mouse TIGIT, 1G9, BV421 | BD | #565270 |
| Anti-mouse CD152, UC10-4F10-11, PE-Cyanine7 | Tonbo | #60-1522-U025 |
| Anti-Human/Mouse CD44, IM7, APC | Tonbo | #50-210-2735 |
| Rat monoclonal anti-mouse NKp46, 29A1.4, PerCP-Cyanine5.5 | eBioscience | #46-3351-80 |
| Rat monoclonal anti-mouse RORγt, PE | eBioscience | #12-6988-82 |
| c-MAF monoclonal antibody, sym0F1, PE | eBioscience | #12-9855-42 |
| Rat Anti-mouse GM-CSF, MP1-22E9, BV421 | BD | #564747 |
| TCF1/TCF7 Rabbit mAB, C63D9, APC | Cell Signaling Technology | #37636 |
| Rat monoclonal anti-Vβ14 TCR, 14-2(RUO), Biotin | BD Bioscience | #553257 |
| Rat Anti-mouse vb 14 T-Cell receptor, 14-2, FITC | BD | #553258 |
| Rat monoclonal anti-mouse RORγt, PE | eBioscience | #12-6988-82 |
| TotalSeq-B0301 anti-mouse Hashtag 1 Antibody | BioLegend | #155831 |
| TotalSeq-B0302 anti-mouse Hashtag 2 Antibody | BioLegend | #155833 |
| TotalSeq-B0302 anti-mouse Hashtag 3 Antibody | BioLegend | #155835 |
| CD4 MicroBeads, mouse | Miltenyi Biotec | #130-117-043 |
| Bacterial strains | ||
| Segmented Filamentous Bacteria (SFB) | Kenya Honda | (Umesaki et al., 1995) |
| Citrobacter rodentium | ATCC | #51459 |
| Bifidobacterium adolescentis | ATCC | #15703 |
| Escherichia coli | ATCC | |
| Chemicals, cytokines, and recombinant proteins | ||
| In VivoMAb anti-mouse IL-10R antibody (clone 1B1.3A) | BioXcell, | #BE0050 |
| CD3e monoclonal antibody, functional grade (clone 2C11) | eBioscience | #16-0031-82 |
| CD28 monoclonal antibody, functional grade (clone 37.51) | eBioscience | #16-0281-38 |
| Mouse CTLA-4 Antibody (clone 63828) | R&D Systems | #MAB434-100 |
| InVivoMAb anti-mouse LAG-3 (clone C9B7W) | BioXcell | #BE0174 |
| InVivoMAB rat IgG1 isotyoe control (clone HRPN) | BioXcell | #BE0088 |
| Recombinant Mouse IL-23 Protein | R&D Systems | #1887-ML |
| Recombinant Murine IL-1β | PeproTech | #211-11B |
| Corning Dispase, 100 mL | Corning (Fisher) | #354235 |
| Roche Collagenase D 2.5g from C.histolyticum | Roche (Sigma) | #11088882001 |
| Collagenase, type 1, powder | Gibco | #17018209 |
| Deoxyribonuclease I from bovine pancreas, 1g | Sigma | DN-25 |
| Hanks’ Balanced Salt solution (HBSS), 10X | CORNING | #36320020 |
| HyClone™ RPMI 1640 Medium, Sterile, pH 7.0 - 7.4, With L-glutamine, Liquid | Cytiva | SH30028.LS |
| Penicillin-Streptomycin (5.000U/ml) | Gibco | #15070063 |
| β-mercaptoethanol | Sigma-Aldrich | #60-24-2 |
| Natriumpyruvate (100 mM) | Gibco | #11360070 |
| L-Glutamin (200 mM) | Gibco | #A2916801 |
| MEM Non-Essential Amino Acids (100X) | Gibco | #11140068 |
| Sodium Bicarbonate | SIGMA | #46H02825 |
| Percoll®, Sterile, pH 8.5 - 9.5, Liquid | Cytiva | 17-0891-09 |
| Fetal Bovine Serum, Qualified, USDA approved | Thermo Scientific | #10437028 |
| HEPES(1M) | Thermofisher | #15630-080 |
| Phenol/Chloroform/Isoamyl alcohol (25:24:1), stabilized | Fisher Scientific | 327115000 |
| Ambion TRIzol reagent | Fisher Scientific | 15-596-018 |
| 2-Propanol, ACS reagent, ≥99.5% | Sigma-Aldrich | #190764 |
| Proteinase K | Lucigen | #MPRK092 |
| Cell Trace Violet cell proliferation kit | Life Technologies | #34557 |
| Ionomycin calcium salt from Streptomyces | Sigma-Aldrich | #10634 |
| PMA, for use in molecular biology | Sigma-Aldrich | #P1585 |
| Brefeldin A,from Penicillium brefeldianum, ≥99% (HPLC and TLC) | Sigma Aldrich | #B7651-5MG |
| Foxp3 / Transcription Factor Fix/Perm Concentrate (4X) | TONBO Biosciences | #TNB-1020-L050 |
| Foxp3 / Transcription Factor Staining Buffer Kit | TONBO Biosciences | #TNB-0607-KIT |
| Flow Cytometry Perm Buffer (10X) | TONBO Biosciences | #TNB-1213-L150 |
| Reinforced Clostridia Medium (RCM) | ThermoFisher | #CM0149 |
| LB Broth | Gibco | #10855001 |
| Critical commercial assays | ||
| Qscript cDNA Super Mix, QuantaBio | VWR | #101414-108 |
| 2X Universal SYBR Green Fast qPCR Mix - 25 mL | ABclonal | #RK21203 |
| IFN gamma Mouse ELISA Kit | Invitrogen | #BMS606-2 |
| Lipocalin-2 (LCN2) Mouse ELISA Kit | Invitrogen | #EMLCN2 |
| cOmplete, EDTA-free protease-inhibitor | Roche | #11836170001 |
| Experimental models: Organisms/strains | ||
| C57BL/6J, Room RB15 | The Jackson Laboratory | #000664 |
| Ptprc (CD45.1) | The Jackson Laboratory | #002014 |
| Cd4 CRE | The Jackson Laboratory | #022071 |
| 7B8 TCR Tg | The Jackson Laboratory | #027230 |
| Rag1 -/- | The Jackson Laboratory | #002216 |
| Il17a GFP | The Jackson Laboratory | #018472 |
| Il10 -/- | The Jackson Laboratory | #002251 |
| Il10rb -/- | The Jackson Laboratory | #005027 |
| Foxp3mRFP | The Jackson Laboratory | #008374 |
| Il10 GFP | The Jackson Laboratory | #008379 |
| Rosa26 YFP | The Jackson Laboratory | #038215 |
| Il17a Katushka | R. Flavell, Yale U | N/A |
| Il10 flox/flox | A. Roers, TU Berlin | N/A |
| Il10 Venus | K. Takeda, Osaka U | N/A |
| Ccr2 DTR | E. Pamer, MSKCC | N/A |
| Tcf7-STOP mice | This Study | N/A |
| Oligonucleotides | ||
| Il10 Fwd 5’-TTGGGTTGCCAAGCCTTATCG-3’ | This Study | N/A |
| Il10 Rev 5’-AATCGATGACAGCGCCTCAG-3’ | This Study | N/A |
| Maf Fwd 5’-GCGAAAGGGACGCCTACAAG-3’ | This Study | N/A |
| Maf Rev 5’-AACAAGGTGGCTAGCTGGGA-3’ | This Study | N/A |
| Il10rb Fwd 5’-TCAGTGCGACTTCTCTCATCTTTC-3’ | This Study | N/A |
| Il10rb Rev 5’-AGGAGGTCCAATGATGGTGTCTT-3’ | This Study | N/A |
| Areg Fwd 5’-TACTTTGGTGAACGGTGTGGAG-3’ | This Study | N/A |
| Areg Rev 5’-GCGAGGATGATGGCAGAGAC-3’ | This Study | N/A |
| Tox Fwd 5’-GTGTGAGGATGCCTCCAAGATCAA-3’ | This Study | N/A |
| Tox Rev 5’-ACAAAGCATAGGCAGACACAGG-3’ | This Study | N/A |
| Tcf7 Fwd 5’-GCGCGGGATAACTACGGAAA-3’ | This study | N/A |
| Tcf7 Rev 5’-GCCTAGAGCACTGTCATCGG-3’ | This study | N/A |
| Gzma Fwd 5’-GACACGGTTGTTCCTCACTCA-3’ | This study | N/A |
| Gzma Rev 5’-CAATCAAAGCGCCAGCACAG-3’ | This study | N/A |
| Ccl5 Fwd 5’-TGCTGCTTTGCCCTACCTCTC-3’ | This Study | N/A |
| Ccl5 Rev5’-CCTTCGAGTGACAAACACGACT-3’ | This Study | N/A |
| Ifng F 5-CACGGCACAGTCATTGAAAG-3’ | (Kawano et al., 2022) | N/A |
| Ifng R-5-GCTGATGGCCTGATTGTCTT-3’ | (Kawano et al., 2022) | N/A |
| Gapdh F 5-CCTCGTCCCGTAGACAAAATG-3’ | (Atarashi et al., 2008) | N/A |
| Gapdh R-5-TCTCCACTTTGCCACTGCAA-3’ | (Atarashi et al., 2008) | N/A |
| SFB F 5-GACGCTGAGGCATGAGAGCAT-3’ | (Barman et al., 2008) | N/A |
| SFB R-5-GACGGCACGGATTGTTATTCA-3’ | (Barman et al., 2008) | N/A |
| UNI F 5-ACTCCTACGGGAGGCAGCAGT-3’ | (Barman et al., 2008) | N/A |
| UNI R-5-ATTACCGCGGCTGCTGGC-3’ | (Barman et al., 2008) | N/A |
| Software and algorithms | ||
| Flow jo_v10.6.2 | BD | N/A |
| Bowtie2 v2.3.4 | N/A | N/A |
| 10X Genomics Cellranger toolkit v1.0.1 | N/A | N/A |
| USEARCH v11.0.667 | N/A | N/A |
| GraphPad Prism version 9.1 | N/A | N/A |
| Other | ||
| BD LSR Fortessa Flow Cytometer | BD | N/A |
| BD Aria, Floy Cytometer | BD | N/A |
| Zirconia/Silica Beads 0.1mm | Fisher Scientific | #11079101z |
| Miltenyi Biotec, Inc. LS Columns 25/PK | Miltenyil Biotec | #130-042-401 |
| LightCycler® 480 System | Roche | N/A |
| Fisherbrand Razor Blades | Fisher Schientific | #12640 |
| Insulin Syringes with Permanently Attached Needles | BD | #329420 |
| Cell Strainer, Individual Package, 40 um, blue | VWR | #76327-098 |
| Bead beater | Biospec | #1001 |
| Beads cleanup | Beckman-Coulter | # A63881 |
Acknowledgements
We thank Samuel Huber and Nicola Gagliani (UKE) for providing key mouse lines. We thank members of the Ivanov lab for technical help. This work was supported by funding from NIH (DK098378, AI144808, AI163069, AI146817) and Burroughs Wellcome Fund (PATH1019125) to I.I.I.. L.B. was partially supported by a fellowship from the German Research Foundation (DFG) (BR 6094/1–1). H.H.W. acknowledges funding from NSF (MCB-2025515), NIH (R01AI132403, R01DK118044, R01EB031935), Burroughs Wellcome Fund (PATH1016691), and the Irma T. Hirschl Trust.
Footnotes
Declaration of interests
H.H.W. is a scientific advisor of SNIPR Biome, Kingdom Supercultures and Fitbiomics, who were not involved in the study.
References
- 1.Ivanov II, Tuganbaev T, Skelly AN, and Honda K (2022). T Cell Responses to the Microbiota. Annual review of immunology 40, 559–587. 10.1146/annurev-immunol-101320-011829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hooper LV, Littman DR, and Macpherson AJ (2012). Interactions between the microbiota and the immune system. Science 336, 1268–1273. 10.1126/science.1223490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, Peterson DA, Stappenbeck TS, and Hsieh CS (2011). Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254. 10.1038/nature10434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, et al. (2013). Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232–236. 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
- 5.Nutsch KM, and Hsieh CS (2012). T cell tolerance and immunity to commensal bacteria. Current opinion in immunology 24, 385–391. 10.1016/j.coi.2012.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498. 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, and Littman DR (2008). Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4, 337–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, Suda W, Imaoka A, Setoyama H, Nagamori T, et al. (2015). Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 163, 367–380. 10.1016/j.cell.2015.08.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, Bridonneau C, Rochet V, Pisi A, De Paepe M, Brandi G, et al. (2009). The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31, 677–689. [DOI] [PubMed] [Google Scholar]
- 10.Kumar P, Monin L, Castillo P, Elsegeiny W, Horne W, Eddens T, Vikram A, Good M, Schoenborn AA, Bibby K, et al. (2016). Intestinal Interleukin-17 Receptor Signaling Mediates Reciprocal Control of the Gut Microbiota and Autoimmune Inflammation. Immunity 44, 659–671. 10.1016/j.immuni.2016.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, Konkel JE, Ramos HL, Wei L, Davidson TS, Bouladoux N, et al. (2010). Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature 467, 967–971. 10.1038/nature09447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Esplugues E, Huber S, Gagliani N, Hauser AE, Town T, Wan YY, O’Connor W Jr., Rongvaux A, Van Rooijen N, Haberman AM, et al. (2011). Control of TH17 cells occurs in the small intestine. Nature 475, 514–518. 10.1038/nature10228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, Pandolfi PP, Mak T, Satija R, Shalek AK, et al. (2015). Single-Cell Genomics Unveils Critical Regulators of Th17 Cell Pathogenicity. Cell 163, 1400–1412. 10.1016/j.cell.2015.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, and Rostami A (2011). The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nature immunology 12, 568–575. 10.1038/ni.2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, and Cua DJ (2007). TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nature immunology 8, 1390–1397. 10.1038/ni1539. [DOI] [PubMed] [Google Scholar]
- 16.Leppkes M, Becker C, Ivanov II, Hirth S, Wirtz S, Neufert C, Pouly S, Murphy AJ, Valenzuela DM, Yancopoulos GD, et al. (2009). RORgamma-expressing Th17 cells induce murine chronic intestinal inflammation via redundant effects of IL-17A and IL-17F. Gastroenterology 136, 257–267. [DOI] [PubMed] [Google Scholar]
- 17.Alexander M, Ang QY, Nayak RR, Bustion AE, Sandy M, Zhang B, Upadhyay V, Pollard KS, Lynch SV, and Turnbaugh PJ (2022). Human gut bacterial metabolism drives Th17 activation and colitis. Cell host & microbe 30, 17–30 e19. 10.1016/j.chom.2021.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Basu R, O’Quinn DB, Silberger DJ, Schoeb TR, Fouser L, Ouyang W, Hatton RD, and Weaver CT (2012). Th22 cells are an important source of IL-22 for host protection against enteropathogenic bacteria. Immunity 37, 1061–1075. 10.1016/j.immuni.2012.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Maxwell JR, Zhang Y, Brown WA, Smith CL, Byrne FR, Fiorino M, Stevens E, Bigler J, Davis JA, Rottman JB, et al. (2015). Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity 43, 739–750. 10.1016/j.immuni.2015.08.019. [DOI] [PubMed] [Google Scholar]
- 20.Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, Blumenschein WM, Judo M, Ayanoglu G, McClanahan TK, et al. (2015). Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 43, 727–738. 10.1016/j.immuni.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Fauny M, Moulin D, D’Amico F, Netter P, Petitpain N, Arnone D, Jouzeau JY, Loeuille D, and Peyrin-Biroulet L (2020). Paradoxical gastrointestinal effects of interleukin-17 blockers. Annals of the rheumatic diseases 79, 1132–1138. 10.1136/annrheumdis-2020-217927. [DOI] [PubMed] [Google Scholar]
- 22.Gagliani N, Amezcua Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, de Zoete MR, Licona-Limon P, Paiva RS, Ching T, et al. (2015). Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature 523, 221–225. 10.1038/nature14452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Schnell A, Littman DR, and Kuchroo VK (2023). T(H)17 cell heterogeneity and its role in tissue inflammation. Nature immunology 24, 19–29. 10.1038/s41590-022-01387-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, Li Y, Kelly G, Chakravarty P, Shoaie S, et al. (2019). The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells. Immunity 51, 77–89 e76. 10.1016/j.immuni.2019.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chihara N, Madi A, Kondo T, Zhang H, Acharya N, Singer M, Nyman J, Marjanovic ND, Kowalczyk MS, Wang C, et al. (2018). Induction and transcriptional regulation of the co-inhibitory gene module in T cells. Nature 558, 454–459. 10.1038/s41586-018-0206-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, and Wherry EJ (2014). Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 40, 289–302. 10.1016/j.immuni.2014.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Martinez GJ, Pereira RM, Aijo T, Kim EY, Marangoni F, Pipkin ME, Togher S, Heissmeyer V, Zhang YC, Crotty S, et al. (2015). The transcription factor NFAT promotes exhaustion of activated CD8(+) T cells. Immunity 42, 265–278. 10.1016/j.immuni.2015.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Alfei F, Kanev K, Hofmann M, Wu M, Ghoneim HE, Roelli P, Utzschneider DT, von Hoesslin M, Cullen JG, Fan Y, et al. (2019). TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269. 10.1038/s41586-019-1326-9. [DOI] [PubMed] [Google Scholar]
- 29.Aschenbrenner D, Foglierini M, Jarrossay D, Hu D, Weiner HL, Kuchroo VK, Lanzavecchia A, Notarbartolo S, and Sallusto F (2018). An immunoregulatory and tissue-residency program modulated by c-MAF in human TH17 cells. Nature immunology 19, 1126–1136. 10.1038/s41590-018-0200-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cao S, Liu J, Song L, and Ma X (2005). The protooncogene c-Maf is an essential transcription factor for IL-10 gene expression in macrophages. Journal of immunology 174, 3484–3492. 10.4049/jimmunol.174.6.3484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Xu J, Yang Y, Qiu G, Lal G, Wu Z, Levy DE, Ochando JC, Bromberg JS, and Ding Y (2009). c-Maf regulates IL-10 expression during Th17 polarization. Journal of immunology 182, 6226–6236. 10.4049/jimmunol.0900123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pot C, Jin H, Awasthi A, Liu SM, Lai CY, Madan R, Sharpe AH, Karp CL, Miaw SC, Ho IC, and Kuchroo VK (2009). Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. Journal of immunology 183, 797–801. 10.4049/jimmunol.0901233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, Burns EJ, Sherr DH, Weiner HL, and Kuchroo VK (2010). The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nature immunology 11, 854–861. 10.1038/ni.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhu C, Sakuishi K, Xiao S, Sun Z, Zaghouani S, Gu G, Wang C, Tan DJ, Wu C, Rangachari M, et al. (2015). An IL-27/NFIL3 signalling axis drives Tim-3 and IL-10 expression and T-cell dysfunction. Nat Commun 6, 6072. 10.1038/ncomms7072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ridley ML, Fleskens V, Roberts CA, Lalnunhlimi S, Alnesf A, O’Byrne AM, Steel KJA, Povoleri GAM, Sumner J, Lavender P, and Taams LS (2020). IKZF3/Aiolos Is Associated with but Not Sufficient for the Expression of IL-10 by CD4(+) T Cells. Journal of immunology 204, 2940–2948. 10.4049/jimmunol.1901283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ciofani M, Madar A, Galan C, Sellars M, Mace K, Pauli F, Agarwal A, Huang W, Parkhurst CN, Muratet M, et al. (2012). A validated regulatory network for Th17 cell specification. Cell 151, 289–303. 10.1016/j.cell.2012.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tan TG, Sefik E, Geva-Zatorsky N, Kua L, Naskar D, Teng F, Pasman L, Ortiz-Lopez A, Jupp R, Wu HJ, et al. (2016). Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proceedings of the National Academy of Sciences of the United States of America 113, E8141–E8150. 10.1073/pnas.1617460113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zuberbuehler MK, Parker ME, Wheaton JD, Espinosa JR, Salzler HR, Park E, and Ciofani M (2019). The transcription factor c-Maf is essential for the commitment of IL-17-producing gammadelta T cells. Nature immunology 20, 73–85. 10.1038/s41590-018-0274-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Neumann C, Blume J, Roy U, Teh PP, Vasanthakumar A, Beller A, Liao Y, Heinrich F, Arenzana TL, Hackney JA, et al. (2019). c-Maf-dependent T(reg) cell control of intestinal T(H)17 cells and IgA establishes host-microbiota homeostasis. Nature immunology 20, 471–481. 10.1038/s41590-019-0316-2. [DOI] [PubMed] [Google Scholar]
- 40.Hu D, Notarbartolo S, Croonenborghs T, Patel B, Cialic R, Yang TH, Aschenbrenner D, Andersson KM, Gattorno M, Pham M, et al. (2017). Transcriptional signature of human pro-inflammatory T(H)17 cells identifies reduced IL10 gene expression in multiple sclerosis. Nat Commun 8, 1600. 10.1038/s41467-017-01571-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Goto Y, Panea C, Nakato G, Cebula A, Lee C, Diez MG, Laufer TM, Ignatowicz L, and Ivanov II (2014). Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity 40, 594–607. 10.1016/j.immuni.2014.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Farkas AM, Panea C, Goto Y, Nakato G, Galan-Diez M, Narushima S, Honda K, and Ivanov II (2015). Induction of Th17 cells by segmented filamentous bacteria in the murine intestine. J Immunol Methods 421, 104–111. 10.1016/j.jim.2015.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, Lee JY, Ziel JW, Miraldi ER, Domingos AI, et al. (2015). An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses. Cell 163, 381–393. 10.1016/j.cell.2015.08.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Stenstad H, Ericsson A, Johansson-Lindbom B, Svensson M, Marsal J, Mack M, Picarella D, Soler D, Marquez G, Briskin M, and Agace WW (2006). Gut-associated lymphoid tissue-primed CD4+ T cells display CCR9-dependent and -independent homing to the small intestine. Blood 107, 3447–3454. 10.1182/blood-2005-07-2860. [DOI] [PubMed] [Google Scholar]
- 45.Woodward Davis AS, Roozen HN, Dufort MJ, DeBerg HA, Delaney MA, Mair F, Erickson JR, Slichter CK, Berkson JD, Klock AM, et al. (2019). The human tissue-resident CCR5(+) T cell compartment maintains protective and functional properties during inflammation. Sci Transl Med 11. 10.1126/scitranslmed.aaw8718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Iijima N, and Iwasaki A (2014). T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98. 10.1126/science.1257530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Turner DL, and Farber DL (2014). Mucosal resident memory CD4 T cells in protection and immunopathology. Front Immunol 5, 331. 10.3389/fimmu.2014.00331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wagner A, Wang C, Fessler J, DeTomaso D, Avila-Pacheco J, Kaminski J, Zaghouani S, Christian E, Thakore P, Schellhaass B, et al. (2021). Metabolic modeling of single Th17 cells reveals regulators of autoimmunity. Cell 184, 4168–4185 e4121. 10.1016/j.cell.2021.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schnell A, Huang L, Singer M, Singaraju A, Barilla RM, Regan BML, Bollhagen A, Thakore PI, Dionne D, Delorey TM, et al. (2021). Stem-like intestinal Th17 cells give rise to pathogenic effector T cells during autoimmunity. Cell 184, 6281–6298 e6223. 10.1016/j.cell.2021.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Nish SA, Zens KD, Kratchmarov R, Lin WW, Adams WC, Chen YH, Yen B, Rothman NJ, Bhandoola A, Xue HH, et al. (2017). CD4+ T cell effector commitment coupled to self-renewal by asymmetric cell divisions. The Journal of experimental medicine 214, 39–47. 10.1084/jem.20161046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, and Roncarolo MG (1997). A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 389, 737–742. 10.1038/39614. [DOI] [PubMed] [Google Scholar]
- 52.Brockmann L, Gagliani N, Steglich B, Giannou AD, Kempski J, Pelczar P, Geffken M, Mfarrej B, Huber F, Herkel J, et al. (2017). IL-10 Receptor Signaling Is Essential for TR1 Cell Function In Vivo. Journal of immunology 198, 1130–1141. 10.4049/jimmunol.1601045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, Jack RS, Wunderlich FT, Bruning JC, Muller W, and Rudensky AY (2011). Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 34, 566–578. 10.1016/j.immuni.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Panea C, Farkas AM, Goto Y, Abdollahi-Roodsaz S, Lee C, Koscsó B, Gowda K, Hohl TM, Bogunovic M, and Ivanov II (2015). Intestinal Monocyte-Derived Macrophages Control Commensal-Specific Th17 Responses. Cell Reports 12, 1314–1324. 10.1016/j.celrep.2015.07.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zigmond E, Bernshtein B, Friedlander G, Walker CR, Yona S, Kim KW, Brenner O, Krauthgamer R, Varol C, Muller W, and Jung S (2014). Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 40, 720–733. 10.1016/j.immuni.2014.03.012. [DOI] [PubMed] [Google Scholar]
- 56.Shih VF, Cox J, Kljavin NM, Dengler HS, Reichelt M, Kumar P, Rangell L, Kolls JK, Diehl L, Ouyang W, and Ghilardi N (2014). Homeostatic IL-23 receptor signaling limits Th17 response through IL-22-mediated containment of commensal microbiota. Proceedings of the National Academy of Sciences of the United States of America 111, 13942–13947. 10.1073/pnas.1323852111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kawano Y, Edwards M, Huang Y, Bilate AM, Araujo LP, Tanoue T, Atarashi K, Ladinsky MS, Reiner SL, Wang HH, et al. (2022). Microbiota imbalance induced by dietary sugar disrupts immune-mediated protection from metabolic syndrome. Cell 185, 3501–3519 e3520. 10.1016/j.cell.2022.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Xu M, Pokrovskii M, Ding Y, Yi R, Au C, Harrison OJ, Galan C, Belkaid Y, Bonneau R, and Littman DR (2018). c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature 554, 373–377. 10.1038/nature25500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wheaton JD, Yeh CH, and Ciofani M (2017). Cutting Edge: c-Maf Is Required for Regulatory T Cells To Adopt RORgammat(+) and Follicular Phenotypes. Journal of immunology 199, 3931–3936. 10.4049/jimmunol.1701134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ladinsky MS, Araujo LP, Zhang X, Veltri J, Galan-Diez M, Soualhi S, Lee C, Irie K, Pinker EY, Narushima S, et al. (2019). Endocytosis of commensal antigens by intestinal epithelial cells regulates mucosal T cell homeostasis. Science 363. 10.1126/science.aat4042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xia Y, Sandor K, Pai JA, Daniel B, Raju S, Wu R, Hsiung S, Qi Y, Yangdon T, Okamoto M, et al. (2022). BCL6-dependent TCF-1(+) progenitor cells maintain effector and helper CD4(+) T cell responses to persistent antigen. Immunity 55, 1200–1215 e1206. 10.1016/j.immuni.2022.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo M, Alfei F, Hofmann M, Wieland D, et al. (2016). T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 45, 415–427. 10.1016/j.immuni.2016.07.021. [DOI] [PubMed] [Google Scholar]
- 63.Kamanaka M, Huber S, Zenewicz LA, Gagliani N, Rathinam C, O’Connor W Jr., Wan YY, Nakae S, Iwakura Y, Hao L, and Flavell RA (2011). Memory/effector (CD45RB(lo)) CD4 T cells are controlled directly by IL-10 and cause IL-22-dependent intestinal pathology. The Journal of experimental medicine 208, 1027–1040. 10.1084/jem.20102149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M, Galan JE, Harhaj E, and Flavell RA (2006). Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 reporter knockin tiger mouse. Immunity 25, 941–952. 10.1016/j.immuni.2006.09.013. [DOI] [PubMed] [Google Scholar]
- 65.Hohl TM, Rivera A, Lipuma L, Gallegos A, Shi C, Mack M, and Pamer EG (2009). Inflammatory monocytes facilitate adaptive CD4 T cell responses during respiratory fungal infection. Cell host & microbe 6, 470–481. 10.1016/j.chom.2009.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Roers A, Siewe L, Strittmatter E, Deckert M, Schluter D, Stenzel W, Gruber AD, Krieg T, Rajewsky K, and Muller W (2004). T cell-specific inactivation of the interleukin 10 gene in mice results in enhanced T cell responses but normal innate responses to lipopolysaccharide or skin irritation. The Journal of experimental medicine 200, 1289–1297. 10.1084/jem.20041789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.DeTomaso D, Jones MG, Subramaniam M, Ashuach T, Ye CJ, and Yosef N (2019). Functional interpretation of single cell similarity maps. Nat Commun 10, 4376. 10.1038/s41467-019-12235-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Delaney C, Schnell A, Cammarata LV, Yao-Smith A, Regev A, Kuchroo VK, and Singer M (2019). Combinatorial prediction of marker panels from single-cell transcriptomic data. Mol Syst Biol 15, e9005. 10.15252/msb.20199005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Street K, Risso D, Fletcher RB, Das D, Ngai J, Yosef N, Purdom E, and Dudoit S (2018). Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477. 10.1186/s12864-018-4772-0. [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.
