Single-cell RNA-sequencing identifies clearly distinct effector-like subpopulations in steady-state MP conventional CD4+ T cells.
Steady-state, unprimed specific-pathogen-free (SPF)-housed mice have a significant proportion of MP CD4+ T cells (TCRβ+CD1d tetramer−CD8−CD4+CD25-CD44highCD62Llow) in their spleen, thymus, inguinal lymph nodes (iLNs), mesenteric lymph nodes (mLNs), Peyer’s patches (PPs), and lung tissues (Fig. 1a and Fig. S1a, Supporting information). The proportion and number of CD4+ T cells increased with age in all tissues (Fig. S1, Supporting information). To identify and focus on the characteristics of MP CD4+ T cells, we used fluorescence-activated cell sorting (FACS) to sort MP CD4+ T cells from the spleens of 10-week-old C57BL/6 mice and performed scRNA-seq with paired V(D)J sequencing of the T cell receptor (Fig. S2a-d, Supporting information). Also, we generated a pipeline to filter out PLZF and TCR Vα14-Jα18 (TRAV11-TRAJ18) expressing cells, a well-known key transcription factor for the development of NKT and MAIT cells 33–35. An unbiased clustering analysis revealed clearly distinct effector T cell–like subpopulations (Fig. 1b) and differentially expressed genes (DEGs) defining each cluster (Fig. 1c and d). A Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of each cluster indicated that clusters 3 and 5 were Th1- and Th17-like populations, respectively, with significance in “chemokine-mediated signaling pathway” and “cellular response to interleukin-1” (Fig. S3, Supporting information). In addition, lineage-specific transcription factors and chemokine receptors were localized in each cluster (Fig. 1e and Fig. S4, Supporting information), suggesting that MP CD4+ T cells are composed of Th1-, Th17-, Tfh-, and Treg-like subpopulations. To examine whether the subpopulations of steady-state MP CD4+ T cells depended on germ or food antigens, we analyzed the proportion of CD44highCD62Llow MP CD4+ T cells in the tissues of SPF-, GF-, and AF-housed mice using flow cytometry. The proportions of total MP CD4+ T cells were almost identical in the spleen and other tissues except the mLN and PP (Fig. 1f, Fig. S5a and S5b, Supporting information), indicating that the generation of splenic MP CD4+ T cells is not affected by the microbiome or food antigen stimulation. Our scRNA-seq analysis further confirmed that the SPF- and GF-housed mice had identical proportions of distinct effector-like subpopulations of splenic MP CD4+ T cells (Fig. 1g and h), but the proportion of Th17-like MP cells in the mLNs of GF-housed mice was much lower than that in the mLNs of SPF-housed mice (Fig. 1i and j). In support, the proportion of CCR6high or RORγt+ MP CD4+ T cells in the mLNs of GF-housed mice was lower than that in SPF-housed mice, suggesting that the generation of gut MP CD4+ T cells is specifically dependent on germs (Fig. S6, Supporting information). The TCR clonal diversity of MP CD4+ T cells in GF-housed mice, especially the Th17-like population in the mLNs, was reduced compared with SPF-housed mice, though comparable diversity was observed in the spleen (Fig. 1k and l). There seemed no significant clonal expansion observed in MP subpopulations. Together, these data indicate that steady-state splenic MP CD4+ T cells contain heterogeneous subpopulations of Th1-, Th17-, Tfh-, and Treg-like cells that express effector molecules and exist independently of the gut microbiome and food antigens.
Differential innate-like effector functions of MP CD4+ T cells after exposure to IL-1 family and STAT activating cytokines.
To determine the effector functions of MP CD4+ T cells to respond to various cytokines, we performed scRNA-seq of MP CD4+ T cells cultured in conditioned medium with IL-12/IL-18, IL-25/IL-33, and IL-1β/IL-23 cytokines (Fig. 2a and Fig. S2e, Supporting information). Additionally, IL-7 was added to the culture medium for T-cell survival and maintenance 36. The merged Uniform Manifold Approximation and Projection (UMAP) plot shows phenotypic characteristics of Th1-, Th2-, Th17-, and Treg-like subpopulations (Fig. 2b and c), which express lineage specific genes (Fig. 2d). Each cytokine set induced distinct subpopulations localized exclusively in the plot (Fig. 2e). We further confirmed the expression of selected gene sets related to the Th1, Th2, and Th17 lineages in each bystander-activated condition, which shows that type 1, 2, and 3 cytokines can upregulate lineage-specific genes in MP CD4+ T cells (Fig. 2f and g). A single-cell regulatory network inference and clustering (SCENIC) analysis in each cytokine condition revealed a significant level of common or specific transcription factor activity (Fig. 2h). In addition, an Ingenuity pathway analysis (IPA) showed the predicted transcriptional regulators in each condition and speculated that the target transcription factors included Bhlhe40, which can be a potent regulator of bystander activation in MP CD4+ T cells (Fig. 2i). To evaluate the effector functions of MP CD4+ T cells, we treated MP CD4+ T cells with those cytokines and/or anti-CD3/anti-CD28 (Fig. 2j and k). Consistently, the MP CD4+ T cells responded to IL-12/IL-18, IL-25/IL-33, and IL-1β/IL-23 cytokines without TCR stimulation, and T-bet+IFN-γ+, GATA3+IL-13+, and RORγt+IL-17+ cells increased compared to TCR-stimulated condition (Fig. 2j). However, TNF-α was produced only in the presence of TCR stimulation and IFN-γ in the culture supernatant from IL-12/IL-18-conditioned MP CD4+ T cells, and IL-4 was also significantly detected with TCR stimulation but not in the bystander condition with IL-25/IL-33. Similarly, Granulocyte-macrophage colony-stimulating factor (GM-CSF) was secreted more efficiently with IL-1β/IL-23 and TCR stimulation than with the cytokines alone (Fig. 2k). Collectively, these results indicate that steady state MP CD4+ T cells have functional heterogeneity of innate-like responses to various sets of IL-1 family and STAT activating cytokines even in the absence of T cell receptor stimulation and suggest possible potent transcriptional regulators that control MP CD4+ T cell effector functions.
Potential responder MP CD4+ T cells express distinct chemokine receptors upon IL-12/IL-18 and IL-1β/IL-23 stimulation.
To determine which MP CD4+ T cells are potential responders to IL-1 family and STAT activating cytokines, we analyzed the subpopulation of expanding or responding clusters and performed trajectory analyses. Control MP CD4+ T cells cultured with IL-7 produced a significantly separate population of CXCR3high cells. In IL-18/IL-12-conditioned MP CD4+ T cells the CXCR3high cell population was reduced, and the IFN-γ- or IL-13highexpressing Th1 and proliferating Th1 populations were greatly increased (Fig. 3a and b). Th1 signature genes, Tbx21, Ifng, Cxcr3, Il2rb1, Il18r1, and CCR5 were highly expressed by IL-18/IL-12-conditioned MP CD4+ T cells (Fig. 3c). GO/KEGG analysis indicated that the related gene sets in those cells had increased including “Cellular response to interferon-gamma”, “Cytokine cytokine receptor interaction”, “Alzheimer’s disease”, and “Parkinson’s disease” (Fig. 3d). The IPA returned terminologies related to cytokines and inflammation such as “JAK/STAT signaling” and “neuroinflammation signaling pathway” and related to cytotoxic response including “Granzyme B signaling” (Fig. 3e). In a pseudo-time trajectory analysis, the MP CD4+ T cells formed a continuous progression that started in CXCR3high cells and gradually progressed toward Fate 1, which expressed Ifng, Stat5a, Bhlhe40, Batf3, Irf4 and Irf8 (Fig. 3f and g). Similarly, a CCR6high cluster was present in IL-7-conditioned MP CD4+ T cells, and its Th17-like cluster was specifically increased by IL-1β and IL-23 (Fig. 3h and i). Th17 signature genes, Rorc, Ccr6, Il17a Il1r1, Il23r, and proliferating marker Mki67 were expressed by those cells (Fig. 3j). GO/KEGG analysis predicted that the related gene sets in IL-1β /IL-23-cultured MP CD4+ T cells had increased “Alzheimer’s disease,” “Parkinson’s disease,” and “Huntington’s disease” which are neurological diseases and increased “cytokine signaling pathway” (Fig. 3k). The IPA more clearly explained the related pathways, “STAT3 pathway,” “leukocyte extravasation signaling,” “neuroinflammation signaling pathway,” “chemokine signaling,” and “Th17 activation pathway” (Fig. 3l). In the trajectory analysis, CCR6high cells seemed to be the starting point, and then the cells gradually differentiated toward Fate 2 (Fig. 3m), which expresses pathogenic Th17-related genes such as Rorc, Il17a, Csf2, Il22, Bhlhe40, Rora and Cebpb with increased activities (Fig. 3n) and expression level (Fig. 3o) of related transcriptomes. These results collectively reveal that Th1-like and Th17-like MP CD4+ T cells expressing different chemokine receptors respond specifically to IL-12/IL-18 and IL-1β/IL-23 cytokines with the effector functions.
Steady state CCR6high memory phenotype CD4+ T cells are bystander-activated by IL-1β and IL-23 to become pathogenic Th17-like cells.
Since scRNA-seq predicted that CCR6high cells were the major cells responding to IL-1β and IL-23, we sorted splenic CCR6high and CCR6low MP CD4+ T cells (Fig. 4a and b) and then determined their proportions in steady-state SPF- and GF-housed mice (Fig. 4c). We found that splenic CCR6high MP CD4+ T cells were independent of the gut. CCR6high MP CD4+ T cells expressed RORγt more highly than CCR6low cells and had comparable expression of T-bet (Fig. 4d). Steady-state CCR6high MP CD4+ T cells, but not CCR6low, expressed IL-17A (Fig. 4e), suggesting that CCR6high MP CD4+ T cells are Th17-like cells. Further stimulation by IL-1β and IL-23 induced IL-17A and GM-CSF expression (Fig. 4f and g), which confirms that CCR6high MP CD4+ T cells produce pathogenic cytokines in the bystander manner. The amount of cytokine secreted, as determined by ELISA, consistently showed that CCR6high MP CD4+ T cells, but not CCR6low, significantly produced IL-17A, GM-CSF, and IFN-γ and that IL-1β and IL-23 had important synergy for pathogenicity (Fig. 4h). In addition, IL-1β greatly expanded RORγt expression in CCR6high MP CD4+ T cells but not CCR6low cells, whereas IL-23 somewhat inhibited the proportion of Ki67-expressing cells, suggesting that IL-1β is important for the proliferation of CCR6high MP CD4+ T cells (Fig. 4i). To further confirm the functions of IL-1β and IL-23 in MP CD4+ T cells, we performed bulk-RNA seq with bystander-activated MP CD4+ T cells exposed to IL-1β and IL-23. In the heatmap DEG analysis, IL-1β increased the expression of proliferation-related gene such as Mki67 and cdk2, whereas IL-23 alone did not have any significant effects on gene expression (Fig. 4j). IL-23 and IL-1β together significantly induced the expression of pathogenic genes such as Bhlhe40, Il1r1, Csf2, Ifng, Il22 and Il17a. In support, Gene set enrichment analysis (GSEA) pathway enrichment plot of IL-1β vs. IL-1β and IL-23 demonstrated that the enrichment score for cell proliferation was higher with IL-1β, and the score of the pathogenic Th17 signature with IL-1β and IL-23 was higher than in the control group (Fig. 4k). Through these results, we understand that steady-state CCR6high MP CD4+ T cells, which show Th17-like characteristics, are the major bystander-activated cells responding to IL-1β and IL-23, which potentiate the cells’ pathogenic character.
CCR6high MP CD4+ T cells exacerbate autoimmune neuroinflammation in bystander manner.
To reveal the importance of splenic MP CD4+ T cells during an autoimmune disease, we first induced active EAE in 5-week-old mice, who have a lower proportion of MP CD4+ T cells than 10-week-old mice. In this mouse model, we compared the disease severity of control mice with that of those who received an additional adoptive transfer of Treg-deleted (Foxp3−) MP CD4+ T cells from 10-week-old Foxp3-GFP mice. EAE was rapidly induced and progressed by transferring additional MP CD4+ T cells from 10-week-old mice, suggesting that MP CD4+ T cells could be an important contributor to MOG35 − 55-induced EAE pathogenesis (Fig. S7a, Supporting information). In support, Rag−/− mice that adoptively transferred MOG-TCR transgenic (2D2) naïve CD45.1−Vβ11+CD4+ T cells with Treg-deleted MP CD4+ T cells (CD45.1+CD4+) showed a more severe phenotype of EAE than the mice that received only 2D2 T cells (Fig. S7b, Supporting information), suggesting that MP CD4+ T cells contribute to the pathogenesis of EAE. Based on our previous results, we hypothesized that CCR6high MP CD4+ T cells are the major pathogenic subpopulation contributing to EAE disease progression. To test that hypothesis, we transferred CCR6high and CCR6low MP CD4+ T cells (CD45.1+CD4+, gating strategy and purity is shown in Fig. S8, Supporting information) and 2D2 naïve CD4+ T cells into Rag−/− mice. The additional transfer of CCR6high MP CD4+ T cells exacerbated EAE development compared with 2D2 transfer alone or the transfer of CCR6low MP CD4+ T cells (Fig. 5a). Interestingly, the number of transferred MP CD4+ T cells that appeared in the spinal cord and brain tissue did not differ between conditions (Fig. 5b). However, CCR6high MP CD4+ T cells produced significantly more cytokines, particularly IL-17A and GM-CSF, in the spinal cord and brain tissue than CCR6low MP CD4+ T cells (Fig. 5c and d). Therefore, CCR6high MP CD4+ T cells, along with antigen-specific T cells, contribute to the pathogenicity of autoimmune neuroinflammation by expressing pathogenic cytokines such as IL-17A and GM-CSF. To clarify the antigen-independent activation of CCR6high MP CD4+ T cells in an EAE mouse model, we detected 2D2 TCR (Vα3.2+ and Vβ11+), which are predominantly expressed in 2D2 naïve CD4+ T cells. Indeed, CCR6high MP CD4+ T cells barely expressed Vα3.2+ and Vβ11+ (Fig. 5e and f). In support, we confirmed that steady-state CCR6high and CCR6low MP CD4+ T cells did not respond to the MOG35 − 55 antigen (Fig. 5g). Collectively, these results suggest that CCR6high MP CD4+ T cells infiltrate in CNS tissue and exacerbate autoimmune neuroinflammation in a bystander manner.
Innate-like effector functions of CCR6high MP CD4+ T cells are conferred by Bhlhe40/GM-CSF axis.
Among the candidate genes involved in bystander activation of CCR6high MP CD4+ T cells, we identified that Bhlhe40/GM-CSF axis could potentially give rise to the pathogenic function of CCR6high MP CD4+ T cells induced by IL-1β and IL-23 without TCR stimulation (Fig. 6a). By using Bhlhe40GFP mice, we found that CCR6high MP CD4+ T cells activated by IL-1β and IL-23 showed significantly increased level of Bhlhe40 (Fig. 6b). Interestingly, Bhlhe40GFP positive T cells majorly produced effector cytokines including IL-17A and GM-CSF compared to Bhlhe40GFP negative T cells (Fig. 6c). In support, Bhlhe40−/− CCR6high MP CD4+ T cells showed markedly reduced IL-17A and GM-CSF production compared to WT (Fig. 6d and e), suggesting that Bhlhe40 is an important transcriptional regulator for the pathogenic functions of bystander CCR6high MP CD4+ T cells. To confirm the in vivo relevance, we transferred WT CCR6high or Bhlhe40−/− CCR6high MP CD4+ T cells along with 2D2 naïve CD4+ T cells into Rag−/− mice. Bhlhe40−/− CCR6high MP CD4+ T cells showed abrogated functions for exacerbating EAE disease compared by WT CCR6high MP CD4+ T cells (Fig. 6f). Interestingly, the number of Bhlhe40−/− CCR6high MP CD4+ T cells expressing IL-17A and GM-CSF in the spinal cord and brain tissue was significantly reduced compared to WT CCR6high MP CD4+ T cells (Fig. 6g and h). In addition, transfer of GM-CSF-deficient CCR6high MP CD4+ T cells showed abrogated function of contributing EAE pathogenesis (Fig. 6i), suggesting that disease aggravation by bystander CCR6high MP CD4+ T cells is committed by IL-1β and IL-23 via Bhlhe40/GM-CSF axis. Collectively, these results indicate that Bhlhe40 confers innate-like pathogenic functions of CCR6high MP CD4+ T cells with GM-CSF production.