SUMMARY
In mice, skin-resident type 2 innate lymphoid cells (ILC2s) exhibit some ILC3-like characteristics. However, the underlying mechanism remains elusive. Here, we observed lower expression of the ILC2 master regulator GATA3 specifically in cutaneous ILC2s (cILC2s) compared to canonical ILC2s, in line with its functionally divergent role in transcriptional control in cILC2s. Decreased levels of GATA3 enabled the expansion of RORγt fate-mapped (RORγtfm+) cILC2s after postnatal days, displaying certain similarities to ILC3s. Single-cell trajectory analysis showed a sequential promotion of the RORγtfm+ cILC2 divergency by RORγt and GATA3. Notably, during hair follicle recycling, these RORγtfm+ cILC2s accumulated around the hair follicle dermal papilla region to facilitate the process. Mechanistically, we found that GATA3-mediated integrin α3β1 upregulation on RORγtfm+ cILC2s was required for their positioning around the dermal papilla. Overall, our study demonstrates a distinct regulatory role of GATA3 in cILC2s, particularly in promoting RORγtfm+ cILC2 divergency to facilitate hair follicle recycling.
Graphical Abstract
In brief
Cutaneous ILC2s exhibit certain ILC3-like characteristics, yet the underlying mechanism and physiological significance remain elusive. Ren et al. demonstrate that decreased GATA3 levels in them lead to a divergent RORγt fate-mapped subgroup, which accumulates around hair follicle dermal papilla region through GATA3-controlled integrin α3β1 upregulation, thereby facilitating hair follicle recycling.
INTRODUCTION
Innate lymphoid cells (ILCs) are essential components of the immune system.1–4 Based on their effector functions, ILCs are categorized into type 1 ILCs (ILC1s) which mainly produce IFN-γ and TNF-α, ILC2s prominently secreting IL-5 and IL-13, and ILC3s that primarily releasing IL-22 and some IL-17. Additionally, there are lymphoid tissue inducer (LTi) cells that share most features with ILC3s but arise from a distinct developmental pathway.4–6 The development, maintenance and effector roles of these ILC subsets rely on specific master regulators.4 Particularly, T-bet is required for ILC1s, GATA3 is essential for ILC2s, and RORγt is necessary for both ILC3s and LTi cells.7–10
GATA3 is also a critical regulator for the development and functions of all ILCs at multiple stages.11 Gata3fl/flVavCre mice exhibit impaired development of all ILCs, highlighting the essential role of GATA3 during ILC development.9 Furthermore, elevated GATA3 expression determines the lineage specification of ILC1s, ILC2s and ILC3s from LTi cells.5,6,12 Once mature, GATA3 remains highly expressed in ILC2s, serving as their master regulator.8,9 Although the levels of GATA3 are low in ILC3s and LTi cells, In ILC3s and LTi cells, it plays a crucial role in promoting their expansion and effector functions.13
ILCs play critical roles in maintaining tissue homeostasis.14 Skin is the outermost and largest barrier tissue of mammals, anatomically consisting of two cutaneous layers, epidermis and dermis, along with a subcutaneous layer, subcutis.15 Immune cells play crucial roles in regulating the physiological and pathological processes in skin.16–18 For example, during hair follicle recycling, regulatory T (Treg) cells expressing Jagged-1 accumulate around the bulge region of hair follicles, promoting efficient activation of hair follicle stem cells and thereby inducing the anagen phase.19,20 Skin ILCs have attracted significant interest in recent years.21–25 They are typically categorized as ILC2s.21,26–29 Consistently, they display increased IL-5 production during atopic dermatitis, thereby exacerbating the disease.26,28,30,31 However, they also possess certain alternative characteristics. A subgroup of skin ILCs located near sebaceous glands constrain sebaceous hyperplasia through secreting TNF and lymphotoxin.22 Additionally, skin ILCs are found to adopt a pathogenic ILC3-like state during psoriasis.23 However, the mechanism underlying these unique traits of skin ILCs remains unclear.
RESULTS
Cutaneous ILC2s exhibit decreased GATA3 expression
Similar to other ILC subsets, skin ILCs lacked of markers for mature hematopoietic lineages, while expressed CD90, CD127 and Id2 (Figures S1A and S1B). They were absent in Rag2−/−Il2rg−/− mice (Figure S1C). In each mouse ear, they numbered approximately 2000, constituting ~5% of immune cells (Figure S1D). Skin ILCs were missing in Gata3fl/flVavCre mice, indicating their origin from common ILC progenitors (Figure S1E).6 Across various skin layers, they were primarily distributed in the dermis, with comparatively lower presence in the epidermis and subcutis in terms of both percentages and numbers (Figures S1F and S1G). Skin ILCs did not express the master regulators T-bet and RORγt associated with ILC1 and ILC3, and thus were typically categorized as ILC2s (Figure S1H).21,32,33 However, they showed a capacity to converge towards a pathogenic ILC3-like state during psoriasis.23
To elucidate the distinction of skin ILCs from canonical ILC2s, the ILC2 master regulator GATA3 was particularly examined utilizing a Gata3 reporter mice tool (Gata3ZsG-fl/fl) (Figures 1A and S1I).34 In epidermal and dermal layers, skin ILCs exhibited lower GATA3-ZsG levels than those in subcutaneous layer (Figures 1B and 1C). The subcutaneous ILCs and small intestine lamina propria (siLP) ILC2s showed equivalent GATA3-ZsG levels. Whereas, although GATA3-ZsG levels in epidermal and dermal ILCs decreased, they remained higher than in other ILC subsets (Figure 1D). Further, only ILCs from epidermis and dermis showed decreased GATA3-ZsG levels among ILC2s from different tissues, largely excluding the contribution of tissue environment to the change (Figure 1E). Consistently, the decrease in endogenous GATA3 expression in epidermal and dermal ILCs was verified by anti-GATA3 staining (Figures 1F and 1G). These findings suggested that ILCs in the two cutaneous layers, hereafter referred to as cutaneous ILC2s (cILC2s), differed from canonical ILC2s, including those in the subcutis. Based on an assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) analysis, cILC2s exhibited reduced chromatin accessibility at the Gata3 locus compared to canonical ILC2s, suggesting potential cell-intrinsic alterations contributing to the decrease in GATA3 expression (Figure 1H).35
Figure 1. GATA3 exhibits decreased expression in cutaneous ILC2s.
(A) Construction strategy for Gata3ZsG-fl/fl mice.
(B) Representative flow cytometry showing ILCs in epidermis (Epi), dermis (Der), subcutis (Sub) of dorsal skin. Lineage (Lin) markers include CD3e, CD5, CD19, B220, CD11b, CD11c, NK1.1, FcεRI, TER-119 and Gr-1.
(C) Representative histogram showing GATA3-ZsG levels in ILCs from epidermis, dermis, and subcutis.
(D) Comparison of GATA3-ZsG levels in ILC subsets of the indicated tissues (n = 5 per group).
(E) Representative flow cytometry showing GATA3-ZsG levels in ILC2s of the indicated tissues.
(F) Representative histogram showing GATA3 levels in ILCs from epidermis, dermis, and subcutis.
(G) Comparison of GATA3 expression in ILC subsets of the indicated tissues (n = 5 per group).
(H) Chromatin accessibility at Gata3 locus in cILC2s, canonical ILC2s and ILC3s (GSE77695). Numbers indicate the percentages in each gate. Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, ***P < 0.001. Data are representative of at least three independent experiments.
See also Figure S1.
We further conducted a single-cell RNA-sequencing (scRNA-seq) analysis on ILCs from skin and small intestine, distinguished by different Sample Tags (Figure S1J). These cells were classified into cILC2, ILC1, ILC2, ILC3, and LTi based on their signature genes (Figures S1K and S1L). ILCs from skin and small intestine was further confirmed using Sample Tags (Figure S1M). Consistent with the variations in GATA3 levels, cILC2s and siLP ILC2s were separated into distinct clusters, highlighting significant differences in their transcriptomes (Figure S1K). Thus, the top 20 differentially expressed genes between cILC2s and siLP ILC2s were profiled (Figure S1N). Interestingly, Il18r1 and Itgae (encoding CD103) were upregulated in cILC2s, while Klrg1 was specifically expressed by siLP ILC2s. A previous study on lung ILC2s categorized the IL-18R1+CD103+ subgroup with decreased GATA3 expression as immature ILC2s, while ST2+KLRG1+ ILC2s were considered as effector cells.33 However, cILC2s exhibited notable expression of IL-18R1 and CD103, along with considerable expression of ST2 and minimal expression of KLRG1 (Figure S1O). Additionally, around 20% of cILC2s had expressed IL-5 (IL-5fm+) according to IL-5 fate-mapping, indicating their classification as effector cells (Figure S1P). Nevertheless, both IL-5fm+ and IL-5fm- cILC2s showed similar expression of IL-18R1, CD103, ST2 and KLRG1 (Figure S1Q). Thus, further exploration of the relationship between cILC2s and immature ILC2s in the lung is still required. Finally, almost no cILC2s had expressed IL-22, eliminating the possibility of ILC3 contamination in our analysis (Figure S1R).
Together, these findings suggest a significant decrease in GATA3 expression in cILC2s compared to canonical ILC2s, indicating the potential presence of distinct characteristics.
GATA3 exerts a unique transcriptional regulatory role in cutaneous ILC2s
Given the dosage effect of GATA3-mediated transcriptional regulation,36 we wondered whether GATA3 played distinct roles in cILC2s compared to canonical ILC2s. Thus, a Gata3ZsG-fl/flCreERT2 mouse strain enabling transient Gata3 deletion (Gata3ZsG-KO) through tamoxifen administration was developed (Figures 2A and 2B). The Gata3 deletion efficiency was confirmed by absence of siLP ILC2s and subcutaneous ILC2s (Figure S2A and S2B). However, cILC2s remained unaffected (Figures 2B and 2C). Gata3 deficiency also led to a reduction in ILC3 and LTi cells due to decreased CD127 expression,13 but the CD127 levels and number of cILC2s remained stable following Gata3 deletion (Figure 2D). Moreover, the GATA3-ZsG levels in Gata3ZsG-KO and Gata3ZsG-fl/fl cILC2s were comparable, suggesting that GATA3 did not promote its own expression in cILC2s (Figure 2E). Further, Gata3 deletion was performed in vitro on canonical siLP ILC2s and cILC2s isolated from Gata3ZsG-fl/flCreERT2 mice through 4-hydroxytamoxifen (4-OHT) administration (Figures 2F and S2C). Consistently, siLP ILC2 number decreased progressively, accompanied by decreased GATA3-ZsG levels after two days (Figures S2D and S2E). In contrast, the number and GATA3-ZsG levels of cILC2 remained unchanged, indicating distinct regulatory roles of GATA3 in cILC2s (Figures 2G and 2H). We thus conducted an RNA-sequencing (RNA-seq) analysis on sort-purified cILC2s from tamoxifen-administered Gata3fl/fl and Gata3fl/flCreERT2 (Gata3KO) mice (Figure 2I). Consequently, 267 GATA3-upregulated and 169 GATA3-downregulated genes (TPM > 5, fold change > 2, and P value < 0.05) were identified (Figure 2J). Among the GATA3-upregulated genes were well-known effector genes of canonical ILC2s, like Il5, Il13, Areg, Calca and Il1rl1. However, GATA3 also enhanced Il17a expression in cILC2s and its deficiency resulted in upregulated Il1r1, Il23r and Il6ra, indicating that the decreased GATA3 levels might confer some ILC3-like features to cILC2s. Additionally, GATA3 upregulated several noncanonical effector genes in cILC2s, including S100a8. These findings underscore the distinct regulatory role of GATA3 in cILC2s compared to canonical ILC2s.
Figure 2. GATA3 exerts a unique regulatory role in cutaneous ILC2s.
(A) Construction strategy for Gata3ZsG-fl/flCreERT2 mice and Gata3ZsG-KO mice.
(B) Schematic diagram illustrating tamoxifen administration on Gata3ZsG-fl/fl and Gata3 ZsG-KO mice (left) and representative flow cytometry of cILC2s in the ears (right).
(C) Percentages and numbers of ear cILC2s in Gata3ZsG-fl/fl and Gata3ZsG-KO mice (n = 5 per group).
(D) Representative histogram showing CD127 expression on ear cILC2s from Gata3ZsG-fl/fl and Gata3ZsG-KO mice (left) and geometric MFI (GMI) of CD127 levels (n = 5 per group) (right).
(E) Representative histogram showing GATA3-ZsG expression in ear cILC2s from Gata3ZsG-fl/fl and Gata3 ZsG-KO mice (left) and GMI of GATA3-ZsG levels (n = 5 per group) (right). GATA3-ZsG expression in wild-type (WT) ear cILC2s is used as negative control.
(F) Schematics of deleting Gata3 in sort-purified ear cILC2s from Gata3ZsG-fl/fl or Gata3ZsG-fl/flCreERT2 (CD45.2) mice and congenic (CD45.1) mice.
(G) Dynamic ratio changes between CD45.2 and CD45.1 cILC2s in (F) (n = 4 per group).
(H) Representative histogram showing GATA3-ZsG expression in Gata3ZsG-fl/fl and Gata3 ZsG-KO cILC2s on day 0 and day 2 post 4-OHT treatment (left), and GMI of GATA3-ZsG levels (n = 4 per group) (right).
(I) Volcano plot depicting gene expression difference between ear cILC2s of tamoxifen treated Gata3fl/fl and Gata3fl/flCreERT2 (Gata3KO) mice (TPM > 5, fold change > 2, and P value < 0.05).
(J) Heatmap profiling the differential genes between Gata3fl/fl and Gata3KO cILC2s.
(K) Schematics showing comparison of GATA3-regulated genes in cILC2s and canonical ILC2s (GSE47851), ILC3s, or LTi cells (GSE71198).
(L) UpSet plot showing shared or unique GATA3-regulated genes across the indicated ILC subsets.
(M) Relative expression of the indicated genes in cILC2s and canonical ILC2s from Gata3fl/fl and Gata3KO mice (GSE47851).
Numbers indicate the percentages in each gate. Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant. Data are representative of at least three independent experiments.
See also Figure S2.
Further, the regulatory effects of GATA3 across all ILC subsets were examined leveraging publicly available RNA-seq data (GSE47851 and GSE71198) (Figure S2F).9,13 Comparative analysis revealed distinct sets of GATA3-regulated and -downregulated genes in cILC2s, ILC2s, ILC3s and LTi cells (Figures 2K and 2L, and S2G). Particularly, only a small fraction of the GATA3-regulated genes exhibited similar expression patterns between cILC2 and canonical ILC2s, ILC3s or LTi cells, emphasizing the distinct regulatory role of GATA3 in cILC2s (Figures 2L and S2H). In addition, while the GATA3 binding motif was identified in opening chromatin regions (OCRs) associated with ~60% of the GATA3-upregulated genes in canonical ILC2s, this percentage decreased to ~40% in cILC2s, as well as in ILC3s and LTi cells, further indicating a disparity in GATA3-mediated transcriptional regulation between cILC2s and canonical ILC2s (Figures S2I and S2J). Notably, while the levels of Il7r remained unaffected in Gata3KO cILC2s, they significantly reduced in Gata3KO canonical ILC2s (Figure 2M). Additionally, cILC2s displayed specific expression of anti-apoptotic gene Bcl2a1, including Bcl2a1a, Bcl2a1b and Bcl2a1d (Figure 2M and S2K).37,38 And, two of these isoforms, Bcl2a1b and Bcl2a1d, were upregulated in Gata3KO cILC2s, potentially contributing to the survival of cILC2s after Gata3 deletion (Figures 2M).
Collectively, GATA3 is not indispensable for the maintenance of cILC2s and exerts regulatory effects over a specific set of genes, underscoring its distinctive regulatory function in cILC2s.
Decreased GATA3 levels associate with the divergence of RORγtfm+ cutaneous ILC2s
Consistent with the Il17a expression, ~75% of cILC2s had undergone RORγt expression (RORγtfm+), as demonstrated by RORγt fate-mapping (Rosa26TdtomatoRorc-Cre, or Rorc-fm) (Figure 3A). In contrast, the proportions of RORγtfm+ canonical ILC2s in the subcutis, siLP or lung were all below 10% (Figure 3B). RORγtfm+ and RORγtfm- cILC2s displayed similar expression of IL-18R1, CD103, ST2 and KLRG1 (Figure S3A). However, RORγtfm+ cILC2s exhibited subtle RORγt expression compared to RORγtfm- cILC2s (Figure 3C). Additionally, RORγtfm- cILC2s did not acquire RORγt expression when traced in transferred Gata3fl/flVavCre recipients, eliminating the emergence of RORγtfm+ cILC2s from RORγtfm- cILC2s during adulthood (Figure 3D). Thus, RORγt expression in cILC2s was examined in newborn mice. The percentage of RORγtfm+ cILC2s was only 20% on postnatal day 10 (P10), but rapidly rose to ~55% by P14 (Figure 3E). Consistently, RORγt expression in cILC2s peaked on P14 compared to P10 and adulthood (Figure 3F). Thus, the postnatal emergence of RORγtfm+ cILC2s, along with increased RORγt expression, suggested a divergence process for cILC2s.
Figure 3. GATA3 negatively correlates with divergency of RORγtfm+ cutaneous ILC2s.
(A) Representative flow cytometry showing RORγt fate-mapping of cILC2s.
(B) Representative histogram showing RORγt fate-mapping of ILC2s from the indicated tissues (left), and percentages of the RORγtfm+ cells (n = 6 per group) (right).
(C) Representative histogram showing RORγt expression in RORγtfm+ and RORγtfm- cILC2s (left), and GMI of RORγt levels (n = 4 per group) (right).
(D) Schematics of transferring RORγtfm+ or RORγtfm- cILC2s to irradiated Gata3fl/flVavCre recipient mice (left), representative flow cytometry showing RORγt fate-mapping of the transferred cILC2s after 4–6 weeks (middle), and percentages of the RORγtfm+ cells (n = 5 per group) (right).
(E) Representative flow cytometry showing RORγt fate-mapping of cILC2s at postnatal day 10 (P10), P14 and adulthood (> 6 weeks) (left), and percentages of the RORγtfm+ cells (n = 3 per group) (right).
(F) Representative flow cytometry showing RORγt expression in cILC2s at P10, P14 and adulthood (> 6 weeks) (left), and percentages of the RORγt+ cells (n = 5 per group) (right).
(G) Volcano plot depicting gene expression difference between RORγtfm+ and RORγtfm- cILC2s (TPM >5, fold change > 2 and P value < 0.05).
(H) Representative flow cytometry showing IL-17A production in RORγtfm+ and RORγtfm- cILC2s after stimulated with PMA and ionomycin for 3 hours (left), and percentages of the IL-17A+ cells (n = 5 per group) (right).
(I) Representative histogram comparing GATA3 expression between RORγtfm+ and RORγtfm- cILC2s (left), and GMI of the GATA3 levels (n = 4 per group) (right).
(J) Empirical cumulative distribution function (ECDF) plot showing relative expression of genes upregulated in RORγtfm+ (yellow line) and RORγtfm- (red line) cILC2s in comparison to the gene expression difference between Gata3fl/fl and Gata3KO cILC2s. Whole genes were used as a reference (gray line).
(K) Heatmap showing expression difference of the indicated genes between RORγtfm+ and RORγtfm- cILC2s, as well as between Gata3fl/fl and Gata3KO cILC2s.
Numbers indicate the percentages in each gate. Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. Data are representative of at least three independent experiments (A, B, C ,E ,F, H, I) and two independent experiments (D, G, J, K).
See also Figure S3.
Through transcriptomic comparison between RORγtfm+ and RORγtfm- cILC2s using RNA-seq, we identified 241 and 324 upregulated genes (TPM > 5, fold change > 2, and P value < 0.05) in them, respectively (Figure 3G and Table S1). Among the upregulated genes in RORγtfm+ cILC2s were Rorc, Il17f and Lta, indicating that they might possess some ILC3-like characteristics. Correspondingly, upon stimulation, RORγtfm+ cILC2s, rather than RORγtfm- cILC2s, demonstrated IL-17A production (Figure 3H). In contrast, the upregulated genes in RORγtfm- cILC2s included Il4, Il5, Areg and Il1rl1, indicating a closer resemblance to canonical ILC2s (Figure 3G). Consistently, upon stimulation, they produced more IL-5 than RORγtfm+ cILC2s (Figure S3B). An empiric cumulative distribution function (ECDF) plot also illustrated the preferential expression of the RORγtfm+ cILC2-upregulated genes within ILC3s over canonical ILC2s (Figure S3C). Further, through ATAC-seq analysis, there were 1463 RORγtfm+ cILC2-specific OCRs shared with either canonical ILC2s or ILC3s displaying preferentially increased accessibility in ILC3s, while 1538 RORγtfm- cILC2-specific OCRs tending to show enhanced accessibility in canonical ILC2s (Figure S3D). Specifically, RORγtfm+ cILC2s showed increased accessibility to the Il17a and Il17f loci, while reduced accessibility to the Il5 locus (Figure S3E). Overall, these findings indicate that RORγtfm+ cILC2s share greater similarity with ILC3s in terms of both transcriptome and chromatin accessibility.
GATA3 expression further decreased in RORγtfm+ cILC2s, raising the possibility of its involvement in their divergency (Figure 3I). Thus, the transcriptomic differences between Gata3fl/fl and Gata3KO cILC2s were compared with those between RORγtfm+ and RORγtfm- cILC2s (Figures 2I and 3G). An ECDF plot showed that the RORγtfm- cILC2-upregulated genes were also preferentially expressed in Gata3fl/fl cILC2s (Figure 3J). Specifically, Areg, Il4, Il5, Il13 and Il1rl1 were downregulated in both RORγtfm+ cILC2s and Gata3KO cILC2s, suggesting that the decreased GATA3 expression might correlated with the divergence of RORγtfm+ cILC2s (Figure 3K). Additionally, the involvement of RORγt in this process was also evaluated. Rorc deficiency (Rorcfl/flVavCre, or RorcKO in brief) did not impact the development or maintenance of cILC2s, further excluding their origin from ILC3s (Figure S3F). Nevertheless, transcriptomic change in RorcKO cILC2s were observed, suggesting that RORγt indeed left certain imprints in RORγtfm+ cILC2s, despite being shortly expressed postnatally (Figure S3G). However, the ECDF plot indicated that RORγt was not involved in the divergence of RORγtfm+ cILC2s (Figure S3H).
Altogether, these data indicate that the transient postnatal RORγt expression leads to the emergence of a distinct RORγtfm+ cILC2 subgroup with certain ILC3-like characteristics, and the decreased GATA3 levels in cILC2s are associated with this divergency.
RORγt and GATA3 sequentially promote the divergence of RORγtfm+ cutaneous ILC2s
To further elucidate the regulatory roles of RORγt and GATA3 in cILC2s, a scRNA-seq analysis was conducted on cILC2s from wild-type (WT), RorcKO and Gata3KO mice in their adulthood (Figures 4A and S4A). After integrating with the previous WT cILC2 scRNA-seq data (Figure S1J and S1K), these cILC2s were further classified into 6 clusters, namely Tnf+, Cxcr4+, Jmy+, Ccr6+, Areg+ and Il1rl1+ based on their preferentially expressed genes (Figures 4B, and S4B and S4C). All 6 clusters exhibited significant expression of Gata3, and were not contaminated by ILC1s, ILC3s, or T cells (Figure S4D). Among them, the Areg+ and Il1rl1+ clusters demonstrated obvious RORγtfm- cILC2 features characterized by the expression of their upregulated genes, while the Jmy+ and Ccr6+ clusters exhibited typical RORγtfm+ cILC2 traits (Figures S4E and 4C). Additionally, the Tnf+ and Cxcr4+ clusters, while not clearly demonstrating RORγtfm+ cILC2 features, already showed diminished RORγtfm- cILC2 characteristics, indicating that they were in the early stages of RORγtfm+ cILC2 divergence. This observation was further supported by profiling the RORγtfm+ and RORγtfm- cILC2-upregulated genes across the 6 clusters (Figure S4F). Next, cILC2s from WT, Gata3KO and RorcKO mice were individually examined (Figures 4D and S4G). No significant alterations were found in Gata3KO or RorcKO cILC2s regarding the expression of effector cytokines for ILC3s and ILC2s, such as Il17a, Il5, Il13, Csf2, and Areg, as well as markers for immature lung ILC2s, like Il18r1 and Itgae (Figure S4H). However, using a Milo tool,39 we did observe that the percentage of RORγtfm- cILC2 clusters significantly decreased in Gata3KO mice, but almost remained unchanged in RorcKO mice, confirming the impact of GATA3 on the RORγtfm+ cILC2 divergency (Figures 4D and 4E, and S4I). Additionally, through a particular examination of the RORγtfm+ cILC2s, a notable increase in the percentage of the Tnf+ cluster was observed in RorcKO cILC2s, while a significant increase in the percentage of the Jmy+ cluster was identified in Gata3KO cILC2s, suggesting that RORγt and GATA3 sequentially promoted the further divergence of RORγtfm+ cILC2s towards the terminal stage (Figure 4F).
Figure 4. RORγt and GATA3 sequentially promote the divergency of RORγtfm+ cutaneous ILC2s.
(A) Schematics of scRNA-seq analysis on cILC2s from WT, RorcKO and Gata3KO mice.
(B) UMAP showing cILC2 classification.
(C) Violin plots showing the RORγtfm+ and RORγtfm- cILC2 features (related to Figure 3g) in each cILC2 cluster. P value were calculated by Mann-Whitney U test between each cluster and other clusters.
(D) Individual distribution of the cILC2 clusters in WT, RorcKO and Gata3KO mice.
(E) Pie chart showing percentages of cILC2 clusters referring to RORγtfm+ and RORγtfm- cILC2s in WT, RorcKO and Gata3KO mice. The percentages were calculated with the Milo tool, and P values were calculated by Mann-Whitney U test.
(F) Bar chart showing percentage changes of RORγtfm+ cILC2 clusters in RorcKO (green bar) and Gata3KO (red bar) mice compared to WT mice.
(G) Distribution of the four RORγtfm+ cILC2 clusters in WT, RorcKO and Gata3KO mice alongside the pseudotime trajectory (related to Figure S4J).
(H) Density of RORγtfm+ cILC2s along the pseudotime trajectory in WT, RorcKO, and Gata3KO mice.
(I) Division of RORγtfm+ cILC2s into four divergence stages based on gene expression dynamics alongside the pseudotime trajectory.
(J) ECDF plot showing relative expression of the genes annotated to Cxcr4+ (left) and Ccr6+ (right) cILC2 clusters in RorcKO (blue line), Gata3KO (red line) and WT (black line) mice, in comparison to the gene expression difference between stages I and II (left), and between stages III and IV (right).
(K) Representative flow cytometry showing CCR6 expression on RORγtfm+ cILC2s from Rorc-fm and Rorc-fmΔGata3 mice (left), and GMI of the CCR6 levels (n = 4 per group) (right).
(L) Representative flow cytometry showing CCR6 expression on cILC2s from Rorcfl/fl and RorcKO mice (left), and GMI of the CCR6 levels (n = 4 per group) (right).
Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, ***P < 0.001. Data are representative of two independent experiments (B, C, D, E, F, G, H, I, J) and at least three independent experiments (K, L).
See also Figure S4.
A pseudotime trajectory analysis was further performed. The RORγtfm+ cILC2s followed a divergency path starting from the Tnf+ cluster, passing through the Cxcr4+ and Jmy+ clusters, and ending at the Ccr6+ cluster, while the RORγtfm- cILC2s followed a progression from the Areg+ cluster to the Il1rl1+ cluster (Figure S4J). The influence of RORγt and GATA3 on the RORγtfm+ cILC2 divergency was further explored by assessing the difference between individual trajectories in WT, Gata3KO and RorcKO cILC2s (Figure 4G). Consistently, RORγt primarily dictated the divergence of RORγtfm+ cILC2s at an early stage, while GATA3 promoted the divergency in the later stage (Figure 4H). Based on cell density along the pseudotime trajectory, we divided the RORγtfm+ cILC2 into four divergence stages, roughly corresponding to the 4 clusters. Gene modules for the four divergence stages were defined validated by their expression dynamics along the trajectory (Figure 4I). We found that progression of the Cxcr4+ cluster from stage I to stage II was delayed by Rorc deficiency, while advancement of the Ccr6+ cluster from stage III to stage IV was impeded by Gata3 deficiency (Figure 4J). Further, using a mouse strain with specific Gata3 deficiency in RORγtfm+ cILC2s (Rosa26TdtomatoGata3fl/flRorc-Cre, or Rorc-fmΔGata3), the impaired terminal divergence of RORγtfm+ cILC2s was confirmed as indicated by their significantly reduced CCR6 expression (Figures S4K and S4L, and 4K). In contrast, the CCR6 levels in cILC2s of RorcKO mice remained unchanged (Figure 4L).
Collectively, the scRNA-seq analysis suggests that RORγt and GATA3 sequentially regulate the divergence of RORγtfm+ cILC2s, promoting them to a final stage with enhanced CCR6 expression.
RORγtfm+ cutaneous ILC2s is crucial in regulating hair follicle recycling
Despite the acknowledged role of cILC2s during pathology, their involvement in physiological processes in the skin remained largely unexplored.22–25 During postnatal days, cILC2s rapidly expanded from P7 to P10, coinciding with the onset of hair growth, which raised the possibility that cILC2s might participate in this process (Figures S5A and S5B). Thus, a depilation-induced synchronized hair regrowth model, previously used to elucidate the role of Treg cells in hair follicle recycling, was employed to evaluate the hair regrowth differences among WT mice, Rag2−/− mice lacking T cells, and Gata3fl/flVavCre mice with further cILC2s absence in the skin.19 Consistent with our expectation, Gata3fl/flVavCre mice displayed significantly delayed hair regrowth compared to both WT and Rag2−/− mice (Figures 5A and 5B). Further, reconstitution of Gata3fl/flVavCre mice with cILC2s and T cells led to notably accelerated hair regrowth compared to the reconstitution with T cells alone, suggesting a difference between the roles of cILC2s and Treg cells in this process (Figures 5C and 5D, and S5C–S5I). In addition, the reconstitution with cILC2s alone also accelerated hair regrowth in Gata3fl/flVavCre mice (Figure S5J). Together, these findings suggest that cILC2s facilitate hair follicle recycling.
Figure 5. RORγtfm+ cutaneous ILC2s facilitate hair follicle recycling.
(A) Schematics of depilation-induced hair regrowth (left), and the kinetics in WT, Rag2−/− and Gata3fl/fl VavCre mice (n = 4 per group) (right).
(B) Representative photographs showing hair regrowth in WT, Rag2−/− and Gata3fl/fl VavCre mice.
(C) Schematics of depilation-induced hair regrowth in Gata3fl/fl VavCre mice reconstituted by cILC2s and T cells or T cells alone (left), and the kinetics (n = 4 per group) (right).
(D) Representative flow cytometry showing replenish of cILC2s in dorsal skin of Gata3fl/fl VavCre mice reconstituted by cILC2s and T cells or T cells alone (left), and percentages of the reconstituted cILC2s in CD45+ immune cells (n = 5 per group) (right).
(E) Schematics of scRNA-seq on cILC2s across different time points of depilation-induced hair regrowth (left), and UMAP visualization of cILC2 clusters projected onto the previous plot (Figure 4B) (right).
(F) Proportion change of cILC2 clusters along depilation-induced hair regrowth.
(G) Representative histogram showing CCR6 expression on dorsal skin cILC2s at day 0 and day 7 post depilation (left), and GMI of the CCR6 levels (n = 5 per group) (right).
(H) Schematics of depilation-induced hair regrowth in Gata3fl/fl VavCre mice reconstituted by RORγtfm+ cILC2s and T cells, RORγtfm- cILC2s and T cells, or T cells alone (left), and the kinetics (n = 5 per group) (right).
(I) Cell-cell communication between cILC2s and hair follicle cells inferred using CellChat.
(J) Representative confocal image showing positioning of RORγtfm+ cILC2s around hair follicles on day 0 (left) and day 7 of depilation-induced hair regrowth (right).
(K) Numbers of RORγtfm+ cILC2s around infundibulum (IF) (n = 28 and 30) (top) and dermal papilla (DP) region of hair follicles (bottom) on day 0 and day 7 of depilation-induced hair regrowth (n = 27 and 30). Each dot represents the cell number around a single follicle (< 20μm).
Numbers indicate the percentages in each gate. Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. Data are representative of at least three independent experiments (A, B, C, D, G, H, J, K) and two independent experiments (E. F. I).
See also Figure S5.
The number of cILC2s remained unchanged throughout depilation-induced hair regrowth (Figure S5K). Thus, to further elucidate the alterations in cILC2 during this process, we conducted a scRNA-seq analysis on cILC2s from different time points of depilation-induced hair regrowth (Figures 5E and S5L). These cells were projected onto the previously obtained cILC2 UMAP plot, and the absence of contamination by T cells or other ILCs was validated (Figures S5M–S5O). The cILC2s at different time points were subsequently divided using their Sample Tags (Figure S5P). Proportional analysis of the cILC2 clusters revealed a notable increase in the Ccr6+ cluster on day 7 (Figure 5F). Consistently, upregulated CCR6 levels on cILC2s were observed then, indicated that RORγtfm+ cILC2s might contribute to hair follicle recycling (Figure 5G). Indeed, reconstitution of Gata3fl/flVavCre mice with RORγtfm+ cILC2s and T cells led to enhanced hair regrowth compared to the reconstitution with RORγtfm- cILC2s and T cells, or T cells alone (Figures 5H and S5Q). Additionally, the interaction between cILC2s and hair follicle cells was explored. Hair follicle cells, enriched via cell sorting, underwent scRNA-seq and classification analyses (Figure S5R). Using a CellChat algorithm, we discovered that Ccr6+ cILC2s exhibited preferential interaction with dermal papilla (DP) cells, which were known to provide essential signals for hair regrowth, as well as to a less extent with hair follicle cells (Figure 5I). Consistently, we observed a significant accumulation of RORγtfm+ cILC2s around the hair follicle DP region on day 7 post-depilation, accompanied by a decrease in their distribution around the infundibulum (IF) region (Figures 5J and 5K, and S5S and S5T). Therefore, our data suggests that RORγtfm+ cILC2s are essential in facilitating hair follicle recycling.
RORγtfm+ cutaneous ILC2s facilitate hair follicle recycling through GATA3
The Ccr6+ cILC2 cluster that showed a proportional increase on day 7 post-depilation displayed elevated expression of Ostf1, Fgl2, as well as several S100 molecules (S100a4, S100a6, S100a10, and S100a11), while the prior Jmy+ cluster exhibited relatively lower levels of these genes (Figures 6A and S6A). Whereas, in line with inhibitory roles of type 2 cytokines such as including IL-5 and IL-13 on hair follicle stem cell proliferation and hair regrowth,25 the expression of Il5 and Il13 decreased in the Ccr6+ cluster on day 7 (Figure 6A). Additionally, an intensified divergence of Ccr6+ cILC2s from stage III to stage IV was observed on day 7 post-depilation (Figure 6B). Consistent with the role of GATA3 in the late-stage divergency, both its expression and regulatory activity, as indicated by the upregulation of its target genes, increased on day 7, implicating that the role of RORγtfm+ cILC2s in hair follicle recycling was regulated by GATA3 (Figures S6B and 6C). Indeed, comparing the depilation-induced hair regrowth between Rorc-fm and Rorc-fmΔGata3 mice showed that Gata3 deficiency resulted in substantially impeded hair regrowth (Figures 6D and S6C). Moreover, although RORγtfm+ cILC2s showed sustained numbers in Rorc-fmΔGata3 mice, their CCR6 levels were markedly diminished, indicating that their terminal divergence was critical for hair follicle recycling (Figures S6D and 6E). Additionally, we observed disrupted accumulation of RORγtfm+ cILC2s surrounding the hair follicle DP region on day 7 post-depilation, highlighting the importance of GATA3 in directing the positioning process (Figures 6F and 6G). To further exclude potential interference by T cell defects in the Rorc-fmΔGata3 mice, we also performed the depilation-induced hair regrowth on Gata3fl/flVavCre mice reconstituted by RORγtfm+ cILC2s from either Rorc-fm or Rorc-fmΔGata3 mice and WT T cells (Figure 6H). Consistently, RORγtfm+ cILC2s from Rorc-fmΔGata3 mice could not effectively restore the hair regrowth (Figures 6H and S6E). And their CCR6 levels remained low after reconstitution (Figure S6F).
Figure 6. RORγtfm+ cutaneous ILC2s require GATA3 to facilitate hair follicle recycling.
(A) Bubble plot showing expression of the indicated effector genes in the Ccr6+ cILC2 cluster during depilation-induced hair regrowth.
(B) Density changes of RORγtfm+ cILC2s along their pseudotime trajectory (related to Figure S4J) during depilation-induced hair regrowth.
(C) Violin plot evaluating GATA3-upregulated genes (related to Figure 2I) in the Ccr6+ cILC2 cluster during depilation-induced hair regrowth.
(D) Schematics of depilation-induced hair regrowth in Rorc-fm and Rorc-fmΔGata3 mice (left), and the kinetics (n = 4 per group) (right).
(E) Representative histogram showing CCR6 expression on RORγtfm+ cILC2s from dorsal skin of Rorc-fm or Rorc -fmΔGata3 mice on day 7 post-depilation (left), and GMI of the CCR6 levels (n = 4 per group) (right).
(F) Representative confocal image showing positioning of RORγtfm+ cILC2s around hair follicles in Rorc-fm (left) or Rorc-fmΔGata3 (right) mice on day 7 of depilation-induced hair regrowth.
(G) Numbers of RORγtfm+ cILC2s around hair follicle DP region in Rorc-fm and Rorc-fmΔGata3 mice on day 7 post-depilation (n = 25 per group). Each dots represent the number of cells around a single follicle (< 20μm).
(H) Schematics of depilation-induced hair regrowth in Gata3fl/flVavCre mice reconstituted by T cells and RORγtfm+ cILC2s from Rorc-fm or Rorc-fmΔGata3 mice (top), and the kinetics (n = 4 per group) (bottom).
Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. Data are representative of two independent experiments (A, B, C, H) and at least three independent experiments (D, E, F, G).
See also Figure S6.
Altogether, these findings highlight the fundamental role of GATA3 during hair follicle recycling, which promotes the final divergence of RORγtfm+ cILC2s and their positioning around the hair follicle DP region.
GATA3 regulates RORγtfm+ cutaneous ILC2 positioning by promoting integrin α3β1 expression
Next, the underlying mechanism of GATA3-regulated RORγtfm+ cILC2 positioning during hair follicle recycling was explored, through a screening for genes upregulated by GATA3 both on day 7 post-depilation and in the Ccr6+ cILC2 cluster (Figures S7A and S7B). Ultimately, only Itga3 (encoding integrin α3), which associated with cell adhesion, was identified (Figure S7C). Notably, Itgb1 (encoding integrin β1) that formed a dimer with Itga3 was also found to be upregulated on day 7 post-depilation (Figure 7A). The increase in integrin α3β1 levels on RORγtfm+ cILC2s during hair follicle recycling was further validated by flow cytometry (Figure S7D). In consistent, the DP and hair follicle cells that interacted with Ccr6+ cILC2s also exhibited preferential expression of the laminin receptors for this integrin, including Lama3, Lamb3, Lamc2 (forming Laminin-332), Lama5, Lamb1 and Lamc1 (forming Laminin-511) (Figure S7E).40–42 Most importantly, the upregulation of integrin α3β1 was disrupted on RORγtfm+ cILC2s of Rorc-fmΔGata3 mice (Figure 7B). To further assess the direct regulation by GATA3, we revisited previous anti-GATA3 ChIP-seq data on ILC2s and identified a GATA3 binding site within the relevant OCRs of Itga3 in cILC2s (Figure 7C). Through CUT&RUN-qPCR, we confirmed that the GATA binding also existed in cILC2s specifically on day 7 post-depilation (Figure 7D). Thus, GATA3 directly regulates the expression of Itga3 in RORγtfm+ cILC2s during hair follicle recycling.
Figure 7. GATA3 directs the positioning of RORγtfm+ cutaneous ILC2s via upregulating integrin α3β1.
(A) Bubble plot showing the expression of Itga3 and Itgb1 in the Ccr6+ cluster during depilation-induced hair regrowth.
(B) Representative histogram showing integrin α3β1 expression on RORγtfm+ cILC2s from dorsal skin of Rorc-fm or Rorc-fmΔGata3 mice on day 7 post-depilation (left), and GMI of the integrin α3β1 levels (n = 4 per group) (right).
(C) Browser tracks displaying accessible chromatin regions in RORγtfm+ and RORγtfm- cILC2s and GATA3 binding sites in canonical ILC2s (GSE71198) at the Itga3 locus.
(D) CUT&RUN-qPCR illustrating binding of GATA3 to the Itga3 locus at the indicated site (C) on day 0 and day 7 of depilation-induced hair regrowth (n = 4 per group). IgG is utilized as a negative control.
(E) Schematics of depilation-induced hair regrowth in Rorc-fm mice administrated by PBS or LXY2 on day 4 (left), and the kinetics (n = 4 per group) (right).
(F) Representative confocal images showing the positioning of RORγtfm+ cILC2s around hair follicles of Rorc-fm mice administrated by PBS or LXY2 (E).
(G) Numbers of RORγtfm+ cILC2s around hair follicle DP region on day 7 post-depilation (n = 13 per group). Each dot represents the number of cells around a single follicle (< 20μm).
(H) Schematics of depilation-induced hair regrowth in Gata3fl/flVavCre mice reconstituted with cILC2s infected by lentivirus expressing shItga3 or shScram RNAs (left), and the kinetics (n = 4 per group) (right).
Data are shown as mean ± SEM. P values are calculated by unpaired t-test. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001. Data are representative of two independent experiments (A, C) and at least three independent experiments (B, D, E, F, G, H).
See also Figure S7.
To further elucidate the role of integrin α3β1 in positioning RORγtfm+ cILC2s, an integrin α3β1 inhibitor, LXY2, was administrated to mice on day 4 post-depilation.43 Comparing to the control group received PBS, LXY2 demonstrated a significant delay in hair regrowth (Figures 7E and S7F). Consistently, the accumulation of RORγtfm+ cILC2s around the hair follicle DP region was also compromised (Figure 7F and 7G). To exclude potential impacts of LXY2 on hair follicle cells, we also generated reconstituted Gata3fl/flVavCre mice with cILC2s experiencing interference in Itga3 expression. Briefly, sort-purified cILC2s were infected by lentivirus expressing short harpin RNAs for Itga3 or the corresponding scramble sequence. Subsequently, Gata3fl/flVavCre mice reconstituted with either of the lentivirally infected cILC2s, along with T cells, underwent depilation-induced hair regrowth after 4–6 weeks (Figure 7H). The reduced expression of integrin α3β1 on the cILC2s with shItga3 was validated, and the delayed hair regrowth in their reconstituted mice was recaptured (Figures 7H, and S7G, S7H).
Collectively, our data suggests a pivotal regulatory role of GATA3 in promoting integrin α3β1 expression on RORγtfm+ cILC2s, thereby positioning them around the hair follicle DP region to facilitate hair regrowth.
DISCUSSION
Skin ILCs exhibit varying cytokine profiles during atopic dermatitis and psoriasis, indicating their distinction from canonical ILC2s.21,44 And, their roles in physiology remain poorly defined. Here, we observe a notable decrease in GATA3 expression in cILC2s, with a significant proportion of them diverging into RORγtfm+ cells. GATA3 functions in a dose-dependent manner.36 Consistently, the decreased levels of GATA3 in cILC2s result in unique regulatory roles. Particularly, GATA3 is dispensable for the maintenance of cILC2s, as demonstrated in tamoxifen-treated Gata3fl/flCreERT2 mice and Rorc-fmΔGata3 mice. The elevated levels of Bcl2a1 in cILC2s compared to canonical ILC2s indicate a potential role of this anti-apoptotic molecule in promoting the survival of cILC2s. However, further investigation is still required. Additionally, Gata3 deficiency in cILC2s does not lead to reduced IL-7R expression as in ILC3s and LTi cells, which is also associated with their persistence in the skin. Moreover, while the ILC2 effector genes remains upregulated by GATA3 in cILC2s as in canonical ILC2s, the overall GATA3-upregulated genes in cILC2s are distinctive. Therefore, in line with the decreased GATA3 levels, its transcriptional regulatory roles in cILC2s differ significantly from those in canonical ILC2s.
GATA3 has been reported to suppress RORγt expression.13 In line with this, we find a correlation between decreased GATA3 levels in cILC2s and an expansion of RORγtfm+ cILC2s. Although the RORγt levels in RORγtfm+ cILC2s are modest, these cells exhibit some characteristics resembling ILC3s, such as IL-17A production upon stimulation. We have observed a notable increase in RORγt expression in cILC2s around P14, indicating their divergence at this phase. This postnatal upregulation of RORγt may imprint certain ILC3-like features on RORγtfm+ cILC2s. A previous study documented the transition of skin ILCs towards a pathogenic ILC3-like state during psoriasis.23 Our study, however, indicates that RORγtfm+ cILC2s already exhibit certain ILC3-like traits under steady-state conditions. It is important to note that the classification, trajectory, and characteristics of cILC2s in our study differ from those in the previous study, primarily due to differences in tissue environments. Furthermore, RORγtfm- cILC2s do not diverge into RORγtfm+ cILC2s in adulthood, suggesting that additional unidentified factors present around P14 are also necessary for initiating RORγt expression in cILC2s.
Through scRNA-seq analysis, we have identified six cILC2 clusters, with four of them displaying the RORγtfm+ cILC2 feature. The change in cell density along the RORγtfm+ cILC2 trajectory in Gata3KO and RorcKO mice suggests that their divergence is orchestrated sequentially by RORγt and GATA3. Further, Gata3 deficiency disrupts the divergency of RORγtfm+ cILC2s towards the terminal stage during hair follicle recycling, as indicated by their impaired CCR6 upregulation. RORγtfm+ cILC2s have been reported to localize around the sebaceous gland to restrict hyperplasia by producing TNF.22 Our study reveals an additional role of RORγtfm+ cILC2s in facilitating hair follicle recycling. They accumulate around the hair follicle DP region on day 7 of depilation-induced hair regrowth. This positioning is also regulated by GATA3, which directly promotes Itga3 upregulation. The enhanced terminal divergency and Itga3 levels regulated by GATA3 may confer crucial functions to the RORγtfm+ cILC2s during hair follicle recycling.
It has been previously reported that Treg cells are also involved in regulating hair follicle recycling. However, Treg cells primarily accumulate around the bulge region of hair follicles and exert their regulatory roles at a timepoint earlier than day 7.19 Therefore, it appears that cILC2s and Treg cells have distinct roles during hair follicle recycling. Indeed, Gata3fl/flVavCre mice constituted with T cells alone are unable to fully restore depilation-induced hair regrowth. Thus, these findings further expand our knowledge of immune regulation during hair follicle recycling.
Type 2 cytokines have been reported to impair hair follicle recycling.25 Consistently, we have observed decreased expression of Il5 and Il13 in the Ccr6+ cILC2s on day 7 of depilation-induced hair regrowth. In contrast, the regulatory role of GATA3 appears to be enhanced. Interestingly, we have detected increased expression of Ostf1, Fgl2, S100a4, S100a6, S100a10 and S100a11 in the Ccr6+ cILC2s on day 7 post-depilation, indicating that cILC2s may perform different functions compared to canonical ILC2s during this process. These differences imply that cILC2s represent a unique subgroup within ILC2s. Despite being overlooked previously, further investigations are still necessary to fully understand the significance of cILC2s in skin physiology and pathology.
Limitations of the study
It is important to acknowledge that the use of straight gene deletion strategies to elucidate the function of GATA3 in regulating the divergence of RORγtfm+ cILC2s and the subsequent roles of RORγt and GATA3 in sequentially promoting their further divergency towards the terminal stage may introduce potential extrinsic differences. Thus, further diligent validation may still be necessary to strengthen this conclusion.
STAR METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Chao Zhong (zhongc@pku.edu.cn).
Materials availability
This study did not generate new unique reagents.
Data and code availability
All data reported in this paper will be shared by the lead contact upon request.
RNA sequencing and ATAC sequencing experiments are deposited at GSA with accession number GSA: CRA010739.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
All animal study protocols used for experiments were approved by the ethics committee of Peking University Health Science Center. All animals were bred and maintained in a specific-pathogen-free facility with a 12 h light/12 h dark cycle, an ambient temperature of 20–24 °C, and humidity of 30–70%. All mice used in experiments were age and sex matched. Male and female mice aged 8–16 weeks if not specifically described were used for all studies. C57BL/6 and CD45.1+ congenic mice were purchased from the Department of Laboratory Animal Science, Peking University Health Science Center. Rorcfl/fl, Rag2−/− and Rag2−/−Il2rg−/− mice were purchased from the Jackson Laboratory (stock# 008771, 008449, 014593). Gata3fl/fl, Gata3fl/flVavCre, Gata3ZsG-fl/fl, Gata3fl/flCreERT2, Id2YFP/+, RORγt-fm (RorcCre-Rosa26TdTomato) and RORγt-fmΔGata3 (Gata3fl/flRorcCre-Rosa26TdTomato) mice on C57BL/6 background were previously described.6,9,13,34 Gata3ZsG-fl/fl mice were bred with CreERT2 mice (from the Jackson Laboratory, stock# 007001) to generate Gata3ZsG-fl/flCreERT2 mice. Rorcfl/fl mice were bred with VavCre mice to generate RorcKO (Rorcfl/flVavCre) mice.
METHOD DETAILS
Tamoxifen treatment
1 mg tamoxifen was dissolved in 150 ml corn oil and intraperitoneally injected into Gata3ZsG-fl/flCreERT2 and Gata3fl/flCreERT2 mice daily for five days to generate Gata3ZsG-KO and Gata3KO mice.
Tissue digestion and cell preparation
Skin cell suspensions were prepared from mouse dorsal skin or ear, as previously described with minor modifications.45 Briefly, for dorsal skin digestion, dorsal hairs were firstly shaved and depilated. Subcutis was teared directly, and epidermis and dermis were separated with forceps after incubating in PBS containing 25 mg/mL Dispase. The subcutis, epidermis, and dermis were cut into ~ 2 mm X 2 mm pieces and digested in RPMI 1640 containing 1 mg/mL Collagenase P, 0.5 mg/mL Hyaluronidase and 0.1 mg/mL DNase I. For ear digestion, the dorsal and ventral sides were teared with forceps, cut into small pieces, and digested in the same condition as dorsal skin.
Digestion of small intestine was described previously.46 Briefly, contents of small intestines were firstly emptied. Then, small intestines were opened longitudinally, cut into 1 cm length pieces, and incubated in RPMI 1640 containing 2% FBS, 5 mM EDTA and 1 mM DTT. Afterwards, the samples were vortexed three times in RPMI 1640 containing 2 mM EDTA to remove epithelial cells. The remained fragments were digested in RPMI 1640 containing 0.05 mg/ml Collagenase IV, 0.5 mg/ml Dispase and 0.1 mg/ml DNase I.
The digested cells were filtered by a 40 μm strainer, centrifugated and resuspended in PBS containing 2% FBS.
Cell staining, flow cytometry, and cell sorting
Digested cells were incubated with an anti-CD16/32 antibody first to block Fc receptors, and then stained with antibodies to surface molecules and fixable viability dye for 30 min at 4 °C. For transcription factor staining, the cells were fixed and permeabilized with the Foxp3/Transcription Factor Staining Buffer Set (eBioscience), and then transcription factors were stained for at least 4 hours at 4 °C. For intracellular cytokine staining, digested cells were resuspended in RPMI 1640 containing 10% FBS and 10 ng/mL recombinant murine IL-7, and stimulated with 50 ng/mL phorbol myristate acetate (PMA) and 500 ng/mL ionomycin for 3 hours at 37 °C, with monensin (eBioscience) added after the first 30 min. The cells were harvested and stained by antibodies to surface molecules. Then, they were fixed by 2% paraformaldehyde in PBS for 20 min at room temperature. The fixed cells were permeabilized with 0.1% triton X-100 for 10 min at room temperature, and subjected to cytokine staining for 30 min at 4 °C.
Flow cytometry were performed on LSR Fortessa (BD Biosciences), and results were analyzed by FlowJo software (FlowJo, LLC). Cell sorting was performed on Aria III cytometers (BD Biosciences) at high purity.
Antibodies specific to mouse CD3e (145-2C11), CD5 (53–7.3), CD19 (eBio1D3), B220 (RA3-6B2), Gr-1 (RB6-8C5) and CD45.2 (104) were purchased from eBioscience; antibodies specific to mouse CD127 (A7R34), CCR6 (29-2L17), CD90.2(30-H12), KLRG1 (2F1/KLRG1) and TCRγ/δ (GL3) were purchased from Biolegend; antibodies specific to mouse RORγt (Q31-378) and GATA-3 (L50-823) were purchased from BD Biosciences; antibody specific to integrin α3β1 (BS-1057R) was purchased from Bioss.
Cell culture
Sort-purified cutaneous ILC2s and siLP ILC2s were cultured in RPMI 1640 containing 10% FBS, 1X Penicillin-Streptomycin, 10 ng/mL recombinant murine IL-7 and 10 ng/mL recombinant human IL-2. To induce Gata3 deletion in vitro, cutaneous ILC2s or siLP ILC2s were sort-purified from Gata3ZsG-fl/fl, Gata3ZsG-fl/flCreERT2, and WT CD45.1 mice. Gata3ZsG-fl/fl or Gata3ZsG-fl/flCreERT2 cells were mixed with CD45.1 cells at 1:1 ratio, and cultured in RPMI 1640 containing 10% FBS, 1X Penicillin-Streptomycin, 10 ng/mL recombinant murine IL-7 and 10 ng/mL recombinant human IL-2. 100 nM 4-OHT was added into the culture medium to induce Gata3 deletion. Cells were harvested daily for flow cytometry analysis.
Cell transfer
5 X 103 cILC2s from freshly digested skin and 5 X 106 T cells from lymph nodes were sort-purified, and transferred to irradiated Gata3fl/flVavCre recipient mice through intravenous injection. After 4–6 weeks, depilation-induced hair follicle recycling was performed using the reconstituted mice.
LXY2 treatment
LXY2 was previously reported as a high-affinity binding ligand of integrin α3β1, inhibiting its interaction with laminin. Its structure can reference from the initial study.43 This compound was synthesized by the company ChinaPeptides Co., Ltd. (QYAOBIO), and its quality was confirmed through mass-spectrum. To prepare this integrin α3β1 inhibitor for use, it was dissolved in PBS. On day 4 post-depilation, it was intracutaneously injected into the mice at 100 nmol per mouse.
Short hairpin RNA lentiviral transduction
Itga3 shRNA sequence (CGGATGGACATTTCAGAGAAA) described previously was cloned to pLKO.1 for lentiviral packaging.47 Regarding the lentiviral packaging, lentiviruses containing either shScram or shItga3 were generated by transfecting 293T cells in a 10 cm dish with lentiviral packaging vectors. The supernatant containing the lentivirus was collected at 48 and 72 hours post-transfection. Subsequently, the lentiviral supernatant underwent ultracentrifugation, and the resulting lentiviral pellets were resuspended in 500 μl RPMI 1640. For the lentiviral infection of cILC2s, we sort-purified 50,000 cILC2s from 6~8 mice. These cells were then seeded into two single wells of a 96-well plate in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) and 10 ng/ml IL-7. At the same time, 100 μl of either shScram or shItga3 lentivirus was added to the well. The infection lasted for 12 hours. Subsequently, the lentivirus-infected cILC2s were harvested, washed, and resuspended in PBS. A total of 5000 cILC2s in 200 μl PBS were then intravenously injected into the recipient mice. The reconstituted mice were used for further analysis after 4–6 weeks.
Immunofluorescent staining
Skin sheets were fixed in 1% paraformaldehyde for 1 hour at room temperature, and dehydrated serially in 20% and 30% sucrose. The dehydrated samples were embedded in OCT (Tissue-Tek) and subjected to snap-freezing. Then, they were cryo-sectioned at 20 μm thickness for immunofluorescent staining. The sectioned sample was incubated in blocking buffer (0.5% BSA, 5% mouse serum and 0.1% Triton-X100) overnight at 4 °C, followed by staining with fluorescein-labeled anti-CD3 (145-2C11) and anti-45.2 (104) antibodies for 12 h at 4 °C. Afterwards, the sample was washed three times in PBS containing 0.1% tween-20. Tissue imaging was performed on Nikon N-STORM 4.0 confocal microscope (Nikon Instruments, Inc.) and analyzed using ImageJ software (NIH).
Bulk RNA-sequencing
RNA-seq library preparation was performed using Single Cell Full Length mRNA Amplification Kit (Vazyme Biotech Co., Ltd.) according to the manufacturer’s instruction. Briefly, cutaneous ILC2s were collected by sorting directly to the lysis buffer for cDNAs reverse transcription and amplification using Single Cell Full Length mRNA Amplification Kit (Vazyme Biotech Co., Ltd.). RNA-seq library was prepared using TruePrep DNA Library Prep Kit V2 for Illumina (Vazyme Biotech Co., Ltd.) according to the manufacturer’s instruction. Library sequencing was performed on Illumina NovaSeq platform with paired-end 150 bp reads.
The RNA-seq reads were aligned to the GRCm38/mm10 assembly of mouse genome using HISAT2, and quantified by featurecounts. Genes with confirmative expression (TPM > 5 in each repeat of any group) were used for further analysis. Differential gene expression analysis was performed by DESeq2 pipeline.
Single-cell RNA-sequencing
Sort-purified cILC2s with different genotype or at different depilation days were labelled with Sample Tags from BD™ Mouse Immune Single-Cell Multiplexing Kit (BD Biosciences), according to the manufacturer’s instructions. Briefly, the BD™ Mouse Immune Single-Cell Multiplexing Kit (Cat# 633793) including Sample Tags composed of unique combinations of nuclear oligos that are conjugated to the anti-mouse CD45 antibody (Clone 30-F11) was used. Each cell sample was labeled with a unique Sample Tag and combined together for subsequent single-cell capture and cDNA synthesis on BD Rhapsody Single-Cell Analysis System (BD). After library preparation, samples were sequenced on Illumina platform with paired-end 150 bp reads.
Single-cell RNA-sequencing data were aligned and quantified using BD Rhapsody WTA Local bioinformatics pipeline (version 1.9.1, BD) against the mouse GRCm38/mm10 reference genome. Quality of scRNA-seq data was assessed based on three metrics: (1) the UMI counts per cell should be less than 6000; (2) features per cell should be more than 500 and less than 2000; (3) proportion of mitochondrial genes should be less than 15%. Seurat R package was used for normalization, dimension reduction, clustering, cluster-specific marker genes identification and clusters annotation.
For demultiplexing the tagged cells and assign them to each samples, the BD Rhapsody Analysis Pipeline, as outlined in the BD® Single-Cell Multiomics Bioinformatics Handbook (Doc ID: 23–21713), was employed. This pipeline automatically integrated the Sample Tag sequences into the FASTA reference file. Consequently, reads that aligned to a particular Sample Tag were used to determine the corresponding sample.
Finally, Monocle2 was used for trajectory inference. AddModuleScore was used to score RORγtfm+ and RORγtfm- gene signatures, and GATA3 upregulated gene signature.
CUT&RUN qPCR
CUT&RUN were performed using the Hyperactive pG-MNase CUT&RUN Assay Kit for Illumina (HD102, Vazyme Biotech Co., Ltd.) following the manufacturer’s instruction. Briefly, cILC2s were sort-purified, and 1% of the cells were saved for extraction of the input genome DNA. The remaining cILC2s were then washed and permeabilized with digitonin. Next, the cells were incubated with either IgG or anti-GATA3 antibody (558686, BD Bioscience) overnight at 4°C, followed by incubation with protein G-MNase for 1 hour at 4°C.The CUT&RUN reaction was initiated by adding Ca2+ and lasted for 30 minutes at 0°C. Afterward, the DNA fragments released into the supernatant were extracted using phenol-chloroform. Subsequently, the DNA fragments, including the input DNA, were quantified by PCR. Primers for Itga3, forward 5’-TCATTCTGTGTCTGATTCCCTATCT-3’, reverse 5’-TGTCAGTAATTGTCATGAGCAAAAG-3’. The relative quantification of each sample was calculated by comparing the intensity of the gel bands obtained from the CUT&RUN and input samples.
ATAC-sequencing
Sort-purified cILC2s were resuspended in 50 μL lysis buffer containing10 mM Tris-HCl pH 7.4, 10 mM NaCl, 3 mM MgCl2 and 0.1% NP-40. After loading on ice for 3 minutes, the cells were centrifuged at 600 Xg for 10 min to collect nuclei. Nuclei were resuspended using transposition reaction buffer. DNA fragmentation and library preparation were performed using TruePrep DNA Library Prep Kit V2 for Illumina (Vazyme) according to the manufacturer’s instruction. Quantity and quality of libraries were assessed on Agilent 2100 Bioanalyzer system. Library sequencing was performed at paired-end 150 bp on Illumina NovaSeq platform.
QUANTIFICATION AND STATISTICAL ANALYSIS
Animals were randomly allocated into different groups. Descriptive statistics were provided in the figure legends. All samples were compared using Prism v7 software (GraphPad) by either a two-sided unpaired Student’s t-test. P value < 0.05 was considered as significant.
Supplementary Material
Document S1. Figures S1–S7
Table S1. List of differentially expressed gene between RORγtfm+ and RORγtfm- cILC2s, related to Figure 3
Key resources table
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
eFluor™ 450 anti-mouse CD3e (Clone 145-2C-11) | eBioscience | Cat#48-0031-82; RRID: AB_10735092 |
eFluor™ 450 anti-mouse CD19 (Clone eBio1D3) | eBioscience | Cat#48-0193; RRID: AB_2043815 |
eFluor™ 450 anti-mouse CD5 (Clone 53-7.3) | eBioscience | Cat#48-0051-82; RRID: AB_1603250 |
eFluor™ 450 anti-mouse Ly-6G/Ly-6C(Gr-1) (Clone RB6-8C5) | eBioscience | Cat#48-5931-82; RRID: AB_1548788 |
eFluor™ 450 anti-mouse CD11b (Clone M1/70) | eBioscience | Cat#48-0112-82; RRID: AB_1582236 |
eFluor™ 450 anti-mouse CD11c (Clone N418) | eBioscience | Cat#48-0114-82; RRID: AB_1548654 |
eFluor™ 450 anti-mouse NK1.1 (Clone PK136) | eBioscience | Cat#48-5941-82; RRID: AB_2043877 |
eFluor™ 450 anti-mouse TER119 (Clone TER-119) | eBioscience | Cat#48-5921-82; RRID: AB_1518808 |
eFluor™ 450 anti-mouse CD45R(B220) (Clone RA3-6B2) | eBioscience | Cat#48-0452-82; RRID: AB_1548761 |
PE-Cyanine7 anti-mouse CD127(Clone A7R34) | eBioscience | Cat#25-1271-82; RRID: AB_469649 |
APC-Cyanine7 anti-mouse CD90.2(Clone 30-H12) | Biolegend | Cat#105328; RRID: AB_10613280 |
Super Bright™ 645 anti-mouse CD4(Clone RM4-5) | eBioscience | Cat#64-0042-82; RRID: AB_2662401 |
Brilliant Violet 650™ anti-mouse CD45.1(Clone A20) | Biolegend | Cat#110736; RRID: AB_11124743 |
Brilliant Violet 650™ anti-mouse CD179(c-Kit) (Clone ACK2) | Biolegend | Cat#135125; RRID: AB_2562446 |
Brilliant Violet 785™ anti-mouse CD196(CCR6) (Clone 29-2L17) | Biolegend | Cat#129823; RRID: AB_2715923 |
Brilliant Violet 785™ anti-mouse CD127(IL-7Ra) (Clone A7R34) | Biolegend | Cat#135037; RRID: AB_2565269 |
Brilliant Violet 785™ anti-mouse CD25(Clone PC61) | Biolegend | Cat#102051; RRID: AB_2564131 |
Brilliant Violet 785™ anti-mouse CD45.2(Clone 104) | Biolegend | Cat#109839; RRID: AB_2562604 |
FITC anti-mouse CD3e (Clone 145-2C11) | Biolegend | Cat#100306; RRID: AB_312671 |
FITC anti-mouse NK1.1(Clone PK136) | Biolegend | Cat#108706; RRID: AB_313392 |
FITC anti-mouse TER119(Clone TER-119) | Biolegend | Cat#116206; RRID: AB_313707 |
FITC anti-mouse CD5(Clone 53-7.3) | Biolegend | Cat#100606; RRID: AB_312735 |
FITC anti-mouse CD19(Clone 6D5) | Biolegend | Cat#115506; RRID: AB_313640 |
FITC anti-mouse Ly-6G/Ly-6C(Gr-1) (Clone RB6-8C5) | Biolegend | Cat#108406; RRID: AB_313370 |
FITC anti-mouse/human CD11b (Clone M1/70) | Biolegend | Cat#101206; RRID: AB_312788 |
FITC anti-mouse CD11c (Clone N418) | Biolegend | Cat#117306; RRID: AB_313775 |
FITC anti-mouse CD45.2(Clone 104) | Biolegend | Cat#109806; RRID: AB_313442 |
FITC anti-mouse CD45RB(B220) (Clone RA3-6B2) | Biolegend | Cat#103206; RRID: AB_312991 |
BD Horizon™ BV650 anti-mouse T-bet (Clone 04-46) | BD Biosciences | Cat#564142; RRID: AB_2738616 |
BD Pharmingen™ Alexa Fluor® 488 anti-mouse GATA3(Clone L50-823) | BD Biosciences | Cat#560077; RRID: AB_1645303 |
BD Horizon™ PE-CF594 anti-mouse RORγt (Clone Q31-378) | BD Biosciences | Cat#562684; RRID: AB_2651150 |
BD Pharmingen™ PE-Cy™7 anti-mouse GATA3(L50-823) | BD Biosciences | Cat#560405; RRID: AB_1645544 |
BD Pharmingen™ Alexa Fluor® 647 anti-mouse T-bet (Clone O4-46) | BD Biosciences | Cat#561267; RRID: AB_10564093 |
PE FOXP3 Monoclonal antibody (Clone FJK-16s) | eBioscience | Cat#12-5773-82; RRID: AB_465936 |
PE-Cyanine7 FOXP3 Monoclonal antibody (Clone FJK-16s) | eBioscience | Cat#25-5773-82; RRID: AB_891552 |
PE anti-mouse CD45.2 | Biolegend | Cat#109808; RRID: AB_313445 |
Rat anti-mouse CD16/32 | Biolegend | Cat#101320; RRID: AB_1574973 |
APC Rat anti-Integrinα3+β1 polyclonal antibody (Clone 976-1025/35-47) | BIOSS | Cat#bs-1057R; RRID: AB_10856085 |
PE anti-mouse TCR γ/δ (Clone GL3) | Biolegend | Cat#118108; RRID: AB_313832 |
APC-eFluor™ 780 anti-mouse CD45(Clone 30-F11) | eBioscience | Cat#47-0451-82; RRID: AB_1548781 |
PE anti-mouse CD196 (CCR6) (Clone 29-2L17) | Biolegend | Cat#129804; RRID: AB_1279139 |
PE-Cyanine7 anti-mouse IL-17A (Clone TC11-18H10.1) | Biolegend | Cat#506922; RRID: AB_2125010 |
FITC anti-mouse CD3(Clone 17A2) | Biolegend | Cat#100203; RRID: AB_312660 |
PE anti-mouse CD3(Clone 17A2) | Biolegend | Cat#100205; RRID: AB_312662 |
APC anti-mouse T-bet (Clone 4B10) | eBioscience | Cat#17-5825-82; RRID: AB_2744712 |
FITC anti-mouse/human KLRG1(Clone 2F1/KLRG1) | Biolegend | Cat#138410; RRID: AB_10643998 |
APC-Cyanine7 anti-mouse/human KLRG1(Clone 2F1/KLRG1) | Biolegend | Cat#138425; RRID: AB_2566553 |
PE anti-mouse CD218a(IL-18Ra) (Clone P3TUNYA) | eBioscience | Cat#12-5183-82; RRID: AB_2572617 |
APC anti-mouse CD218a(IL-18Ra) (Clone A17071D) | Biolegend | Cat#157906; RRID: AB_2860735 |
PE/Cyanine7 anti-mouse IL-33Rα (IL1RL1, ST2) (Clone DIH9) | Biolegend | Cat#145315; RRID: AB_2687366 |
Biotin anti-mouse IL-33Rα (IL1RL1, ST2) (Clone DJ8) | MD Biosciences | Cat#101001B; RRID: AB_947551 |
Alexa Fluor™ 700 anti-mouse CD103(Clone 2E7) | eBioscience | Cat#56-1031-82; RRID: AB_2637111 |
Brilliant Violet 421™ anti-mouse/human IL-5(Clone TRFK5) | Biolegend | Cat#504311; RRID: AB_2563161 |
eFluor™ 660 anti-mouse IL-13(Clone eBio13A) | eBioscience | Cat#50-7133-82; RRID: AB_2574279 |
APC anti-mouse CD8b (Clone H35-17.2) | eBioscience | Cat#17-0083-81; RRID: AB_657760 |
PE anti-mouse/human CD44(Clone IM7) | Biolegend | Cat#103008; RRID: AB_493687 |
APC-Cyanine7 anti-mouse CD62L (Clone MEL-14) | Biolegend | Cat#104428; RRID: AB_830799 |
APC anti-mouse CD133(Clone 315-2C11) | Biolegend | Cat#141207; RRID: AB_10898121 |
BD Pharmingen™ Purified Mouse anti-GATA3(Clone L50-823) | BD Biosciences | Cat#558686; RRID: AB_2108590 |
BD Horizon™ BUV737 Streptavidin | BD Biosciences | Cat#564293; RRID: AB_2869560 |
Chemicals, peptides, and recombinant proteins | ||
Recombinant Human IL-2 | Peprotech | Cat#200-2 |
Recombinant Murine IL-7 | Peprotech | Cat#217-17 |
DNase I | Roche | Cat#10104159001 |
Collagenase P | Roche | Cat#11213865001 |
Collagenase IV | Sigma-Aldrich | Cat#C5138 |
Hyaluronidase | Harveybio | Cat#EZ2176 |
LXY2 | Chinapeptides | N/A |
Foxp3/Transcription Factor Staining Buffer Set | eBioscience | Cat#00-5523-00 |
Paraformaldehyde | Sigma-Aldrich | Cat#P6148 |
Tissue-Tek® O.C.T. Compound | Sakura | Cat#4583 |
Neutral Protease (Dispase) | BioRuler | Cat#RH73624-25G |
RPMI 1640 | Gibco | Cat#31800105 |
Bovine serum albumin, fraction V | Sangon | Cat#A500023-0100 |
Triton X-100 | VWR | Cat#0694-1L |
Penicillin-Streptomycin | Gibco | Cat#15140-122 |
4-Hydroxytamoxifen, 99% | Macklin | Cat#A872541-5mg |
Fetal Bovine Serum | Gibco | Cat#10099141C |
Tween-20 | VWR | Cat#0777-1L |
eBioscience™ Cell Stimulation Cocktail (500X) | Invitrogen | Cat#00-4970-93 |
eBioscience™ Monensin Solution (1000X) | Invitrogen | Cat#00-4505-51 |
DTT | BBI | Cat#A620058 |
DAPI Stain Solution | BBI | Cat#E607303 |
eBioscience™ Fixable Viability Dye eFluor™ 506 | Invitrogen | Cat#65-0866-14 |
Critical commercial assays | ||
Single Cell Full Length mRNA Amplification Kit | Vazyme | Cat#N712 |
TruePrep DNA Library Prep Kit V2 for Illumina | Vazyme | Cat#TD501 |
BD™ Mouse Immune Single-Cell Multiplexing Kit | BD Biosciences | Cat#633793 |
Hyperactive pG-MNase CUT&RUN Assay Kit for Illumina | Vazyme | Cat#HD102 |
Deposited data | ||
Raw data of RNA-seq and ATAC-seq | This paper | GSA: CRA010739 |
RNA-seq data of GATA3 sufficient and deficient ILC2 | Yagi et al.9 | GEO: GSE47851 |
RNA-seq data of GATA3 sufficient and deficient ILC3 | Zhong et al.13 | GEO: GSE71198 |
ATAC-seq data of canonical ILC2 and ILC3 | Shih et al.35 | GEO: GSE77695 |
Experimental models: Organisms/strains | ||
Mouse: Gata3 fl/fl | NIAID -Taconic repository | Line 355; PMID:15475959 |
Mouse: Gata3 ZsG-fl/fl | Dr. JInfang Zhu, NIAID, NIH | PMID: 36466899 |
Mouse: Gata3 fl/fl VavCre | NIAID -Taconic repository | Line 8446; PMID:15475959 |
Mouse: Id2-YFP | Dr. Jonathan R. Keller, NCI, NIH | PMID: 25051963 |
Mouse: Rorc-Cre | The Jackson Laboratory | Cat#JAX: 022791; RRID: IMSR_JAX:022791 |
Mouse: Rosa26 TdTamato | The Jackson Laboratory | Cat#JAX: 007914; RRID: IMSR_JAX:007914 |
Mouse: CD45.2 Rag2 −/−Il2rg −/− | The Jackson Laboratory | Cat#JAX: 014593; RRID: IMSR_JAX:014593 |
Mouse: Rag2 −/− | The Jackson Laboratory | Cat#JAX: 008449; RRID: IMSR_JAX:008449 |
Mouse: Il22 Cre | The Jackson Laboratory | Cat#JAX: 027524; RRID: IMSR_JAX:027524 |
Mouse: Il5 iCre | The Jackson Laboratory | Cat#JAX: 030926; RRID: IMSR_JAX:030926 |
Mouse: Rosa26 YFP | The Jackson Laboratory | Cat#JAX: 006148; RRID: IMSR_JAX:006148 |
Mouse: Rorc fl/fl | The Jackson Laboratory | Cat#JAX: 008771; RRID: IMSR_JAX:008771 |
Mouse: CreERT2 | The Jackson Laboratory | Cat#JAX: 007001; RRID: IMSR_JAX:007001 |
Mouse: C57BL/6 | Department of Laboratory Animal Science, PKUHSC | N/A |
Mouse: B6. CD45.1 | Department of Laboratory Animal Science, PKUHSC | N/A |
Software and algorithms | ||
FlowJo v10 | Tree Star | http://www.flowjo.com/ |
GraphPad Prism 7 | GraphPad | http://www.graphpad.com/ |
ImageJ | NIH | https://imagej.net/ |
Highlight.
Decreased GATA3 levels allow postnatal emergence of RORγt fate-mapped cutaneous ILC2s
RORγt and GATA3 sequentially promote the terminal divergence of RORγtfm+ cILC2s
RORγtfm+ cILC2s accumulate around dermal papilla to facilitate hair follicle recycling
GATA3-controlled integrin α3β1 expression locates RORγtfm+ cILC2s around dermal papilla
ACKNOWLEDGMENTS
We thank Drs. Yuxin Yin, Jianyuan Luo, and Xiaoyan Qiu for their helpful advices, Drs Luyang Sun and Lin He for their instructions in some experiments, Ms. Yichen Deng for her helps in cell sorting. This work was supported by the National Natural Science Foundation of China (32170896, U23A20167, 31770957, 91842102), the National Key Research & Development Program of China (2022YFA1103602, 2022YFA0806400), the Shenzhen Innovation Committee of Science and Technology (JCYJ20220818100401003), and the Natural Science Foundation of Beijing (18G10645).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
DECLARATION OF INTERESTS
The authors declare no competing interests.
REFERENCES
- 1.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, et al. (2013). Innate lymphoid cells — a proposal for uniform nomenclature. Nature Reviews Immunology 13, 145–149. 10.1038/nri3365. [DOI] [PubMed] [Google Scholar]
- 2.Artis D, and Spits H (2015). The biology of innate lymphoid cells. Nature 517, 293–301. 10.1038/nature14189. [DOI] [PubMed] [Google Scholar]
- 3.Eberl G, Colonna M, Di Santo JP, and McKenzie AN (2015). Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 348, aaa6566. 10.1126/science.aaa6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, et al. (2018). Innate Lymphoid Cells: 10 Years On. Cell 174, 1054–1066. 10.1016/j.cell.2018.07.017. [DOI] [PubMed] [Google Scholar]
- 5.Constantinides MG, McDonald BD, Verhoef PA, and Bendelac A (2014). A committed precursor to innate lymphoid cells. Nature 508, 397–401. 10.1038/nature13047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhong C, Zheng M, Cui K, Martins AJ, Hu G, Li D, Tessarollo L, Kozlov S, Keller JR, Tsang JS, et al. (2020). Differential Expression of the Transcription Factor GATA3 Specifies Lineage and Functions of Innate Lymphoid Cells. Immunity 52, 83–95 e84. 10.1016/j.immuni.2019.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sawa S, Cherrier M, Lochner M, Satoh-Takayama N, Fehling HJ, Langa F, Di Santo JP, and Eberl G (2010). Lineage relationship analysis of RORgammat+ innate lymphoid cells. Science 330, 665–669. 10.1126/science.1194597. [DOI] [PubMed] [Google Scholar]
- 8.Hoyler T, Klose CS, Souabni A, Turqueti-Neves A, Pfeifer D, Rawlins EL, Voehringer D, Busslinger M, and Diefenbach A (2012). The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity 37, 634–648. 10.1016/j.immuni.2012.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Yagi R, Zhong C, Northrup DL, Yu F, Bouladoux N, Spencer S, Hu G, Barron L, Sharma S, Nakayama T, et al. (2014). The transcription factor GATA3 is critical for the development of all IL-7Ralpha-expressing innate lymphoid cells. Immunity 40, 378–388. 10.1016/j.immuni.2014.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Klose CSN, Flach M, Mohle L, Rogell L, Hoyler T, Ebert K, Fabiunke C, Pfeifer D, Sexl V, Fonseca-Pereira D, et al. (2014). Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356. 10.1016/j.cell.2014.03.030. [DOI] [PubMed] [Google Scholar]
- 11.Zhu J (2017). GATA3 Regulates the Development and Functions of Innate Lymphoid Cell Subsets at Multiple Stages. Front Immunol 8, 1571. 10.3389/fimmu.2017.01571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yu Y, Tsang JC, Wang C, Clare S, Wang J, Chen X, Brandt C, Kane L, Campos LS, Lu L, et al. (2016). Single-cell RNA-seq identifies a PD-1(hi) ILC progenitor and defines its development pathway. Nature 539, 102–106. 10.1038/nature20105. [DOI] [PubMed] [Google Scholar]
- 13.Zhong C, Cui K, Wilhelm C, Hu G, Mao K, Belkaid Y, Zhao K, and Zhu J (2016). Group 3 innate lymphoid cells continuously require the transcription factor GATA-3 after commitment. Nat Immunol 17, 169–178. 10.1038/ni.3318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Klose CS, and Artis D (2016). Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat Immunol 17, 765–774. 10.1038/ni.3489. [DOI] [PubMed] [Google Scholar]
- 15.Kabashima K, Honda T, Ginhoux F, and Egawa G (2019). The immunological anatomy of the skin. Nat Rev Immunol 19, 19–30. 10.1038/s41577-018-0084-5. [DOI] [PubMed] [Google Scholar]
- 16.Eyerich S, Eyerich K, Traidl-Hoffmann C, and Biedermann T (2018). Cutaneous Barriers and Skin Immunity: Differentiating A Connected Network. Trends Immunol 39, 315–327. 10.1016/j.it.2018.02.004. [DOI] [PubMed] [Google Scholar]
- 17.Kobayashi T, Ricardo-Gonzalez RR, and Moro K (2020). Skin-Resident Innate Lymphoid Cells - Cutaneous Innate Guardians and Regulators. Trends Immunol 41, 100–112. 10.1016/j.it.2019.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liu Y, Wang H, Taylor M, Cook C, Martinez-Berdeja A, North JP, Harirchian P, Hailer AA, Zhao Z, Ghadially R, et al. (2022). Classification of human chronic inflammatory skin disease based on single-cell immune profiling. Sci Immunol 7, eabl9165. 10.1126/sciimmunol.abl9165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ali N, Zirak B, Rodriguez RS, Pauli ML, Truong HA, Lai K, Ahn R, Corbin K, Lowe MM, Scharschmidt TC, et al. (2017). Regulatory T Cells in Skin Facilitate Epithelial Stem Cell Differentiation. Cell 169, 1119–1129 e1111. 10.1016/j.cell.2017.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Liu Z, Hu X, Liang Y, Yu J, Li H, Shokhirev MN, and Zheng Y (2022). Glucocorticoid signaling and regulatory T cells cooperate to maintain the hair-follicle stem-cell niche. Nat Immunol 23, 1086–1097. 10.1038/s41590-022-01244-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ricardo-Gonzalez RR, Van Dyken SJ, Schneider C, Lee J, Nussbaum JC, Liang HE, Vaka D, Eckalbar WL, Molofsky AB, Erle DJ, and Locksley RM (2018). Tissue signals imprint ILC2 identity with anticipatory function. Nat Immunol 19, 1093–1099. 10.1038/s41590-018-0201-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kobayashi T, Voisin B, Kim DY, Kennedy EA, Jo JH, Shih HY, Truong A, Doebel T, Sakamoto K, Cui CY, et al. (2019). Homeostatic Control of Sebaceous Glands by Innate Lymphoid Cells Regulates Commensal Bacteria Equilibrium. Cell 176, 982–997 e916. 10.1016/j.cell.2018.12.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bielecki P, Riesenfeld SJ, Hutter JC, Torlai Triglia E, Kowalczyk MS, Ricardo-Gonzalez RR, Lian M, Amezcua Vesely MC, Kroehling L, Xu H, et al. (2021). Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature. 10.1038/s41586-021-03188-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sakamoto K, Jin SP, Goel S, Jo JH, Voisin B, Kim D, Nadella V, Liang H, Kobayashi T, Huang X, et al. (2021). Disruption of the endopeptidase ADAM10-Notch signaling axis leads to skin dysbiosis and innate lymphoid cell-mediated hair follicle destruction. Immunity 54, 2321–2337 e2310. 10.1016/j.immuni.2021.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ricardo-Gonzalez RR, Kotas ME, O’Leary CE, Singh K, Damsky W, Liao C, Arouge E, Tenvooren I, Marquez DM, Schroeder AW, et al. (2022). Innate type 2 immunity controls hair follicle commensalism by Demodex mites. Immunity 55, 1891–+. 10.1016/j.immuni.2022.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kim BS, Siracusa MC, Saenz SA, Noti M, Monticelli LA, Sonnenberg GF, Hepworth MR, Van Voorhees AS, Comeau MR, and Artis D (2013). TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation. Sci Transl Med 5, 170ra116. 10.1126/scitranslmed.3005374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Roediger B, Kyle R, Yip KH, Sumaria N, Guy TV, Kim BS, Mitchell AJ, Tay SS, Jain R, Forbes-Blom E, et al. (2013). Cutaneous immunosurveillance and regulation of inflammation by group 2 innate lymphoid cells. Nat Immunol 14, 564–573. 10.1038/ni.2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, Thornton EE, Krummel MF, Chawla A, Liang HE, and Locksley RM (2013). Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248. 10.1038/nature12526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Schneider C, Lee J, Koga S, Ricardo-Gonzalez RR, Nussbaum JC, Smith LK, Villeda SA, Liang HE, and Locksley RM (2019). Tissue-Resident Group 2 Innate Lymphoid Cells Differentiate by Layered Ontogeny and In Situ Perinatal Priming. Immunity 50, 1425–1438 e1425. 10.1016/j.immuni.2019.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Leyva-Castillo JM, Galand C, Mashiko S, Bissonnette R, McGurk A, Ziegler SF, Dong C, McKenzie ANJ, Sarfati M, and Geha RS (2020). ILC2 activation by keratinocyte-derived IL-25 drives IL-13 production at sites of allergic skin inflammation. J Allergy Clin Immunol 145, 1606–1614 e1604. 10.1016/j.jaci.2020.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Schwartz C, Moran T, Saunders SP, Kaszlikowska A, Floudas A, Bom J, Nunez G, Iwakura Y, O’Neill L, Irvine AD, et al. (2019). Spontaneous atopic dermatitis in mice with a defective skin barrier is independent of ILC2 and mediated by IL-1beta. Allergy 74, 1920–1933. 10.1111/all.13801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Salimi M, Barlow JL, Saunders SP, Xue L, Gutowska-Owsiak D, Wang X, Huang LC, Johnson D, Scanlon ST, McKenzie AN, et al. (2013). A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J Exp Med 210, 2939–2950. 10.1084/jem.20130351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zeis P, Lian M, Fan X, Herman JS, Hernandez DC, Gentek R, Elias S, Symowski C, Knopper K, Peltokangas N, et al. (2020). In Situ Maturation and Tissue Adaptation of Type 2 Innate Lymphoid Cell Progenitors. Immunity 53, 775–792 e779. 10.1016/j.immuni.2020.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gurram RK, Wei D, Yu Q, Kamenyeva O, Chung H, Zheng M, Butcher MJ, Kabat J, Liu C, Khillan JS, and Zhu J (2022). Gata3 (ZsG) and Gata3 (ZsG-fl): Novel murine Gata3 reporter alleles for identifying and studying Th2 cells and ILC2s in vivo. Front Immunol 13, 975958. 10.3389/fimmu.2022.975958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Shih HY, Sciume G, Mikami Y, Guo L, Sun HW, Brooks SR, Urban JF Jr., Davis FP, Kanno Y, and O’Shea JJ (2016). Developmental Acquisition of Regulomes Underlies Innate Lymphoid Cell Functionality. Cell 165, 1120–1133. 10.1016/j.cell.2016.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Klein Wolterink RG, Serafini N, van Nimwegen M, Vosshenrich CA, de Bruijn MJ, Fonseca Pereira D, Veiga Fernandes H, Hendriks RW, and Di Santo JP (2013). Essential, dose-dependent role for the transcription factor Gata3 in the development of IL-5+ and IL-13+ type 2 innate lymphoid cells. Proc Natl Acad Sci U S A 110, 10240–10245. 10.1073/pnas.1217158110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Vogler M (2012). BCL2A1: the underdog in the BCL2 family. Cell Death Differ 19, 67–74. 10.1038/cdd.2011.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lionnard L, Duc P, Brennan MS, Kueh AJ, Pal M, Guardia F, Mojsa B, Damiano MA, Mora S, Lassot I, et al. (2019). TRIM17 and TRIM28 antagonistically regulate the ubiquitination and anti-apoptotic activity of BCL2A1. Cell Death Differ 26, 902–917. 10.1038/s41418-018-0169-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dann E, Henderson NC, Teichmann SA, Morgan MD, and Marioni JC (2022). Differential abundance testing on single-cell data using k-nearest neighbor graphs. Nat Biotechnol 40, 245–253. 10.1038/s41587-021-01033-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arimori T, Miyazaki N, Mihara E, Takizawa M, Taniguchi Y, Cabanas C, Sekiguchi K, and Takagi J (2021). Structural mechanism of laminin recognition by integrin. Nat Commun 12, 4012. 10.1038/s41467-021-24184-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.DeRouen MC, Zhen H, Tan SH, Williams S, Marinkovich MP, and Oro AE (2010). Laminin-511 and integrin beta-1 in hair follicle development and basal cell carcinoma formation. BMC Dev Biol 10, 112. 10.1186/1471-213X-10-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tayem R, Niemann C, Pesch M, Morgner J, Niessen CM, Wickstrom SA, and Aumailley M (2021). Laminin 332 Is Indispensable for Homeostatic Epidermal Differentiation Programs. J Invest Dermatol 141, 2602–2610 e2603. 10.1016/j.jid.2021.04.008. [DOI] [PubMed] [Google Scholar]
- 43.Yao N, Xiao W, Wang X, Marik J, Park SH, Takada Y, and Lam KS (2009). Discovery of targeting ligands for breast cancer cells using the one-bead one-compound combinatorial method. J Med Chem 52, 126–133. 10.1021/jm801062d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bernink JH, Ohne Y, Teunissen MBM, Wang J, Wu J, Krabbendam L, Guntermann C, Volckmann R, Koster J, van Tol S, et al. (2019). c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute to IL-17-mediated pathologies. Nat Immunol 20, 992–1003. 10.1038/s41590-019-0423-0. [DOI] [PubMed] [Google Scholar]
- 45.Xu M, Lu H, Lee YH, Wu Y, Liu K, Shi Y, An H, Zhang J, Wang X, Lai Y, and Dong C (2018). An Interleukin-25-Mediated Autoregulatory Circuit in Keratinocytes Plays a Pivotal Role in Psoriatic Skin Inflammation. Immunity 48, 787–798 e784. 10.1016/j.immuni.2018.03.019. [DOI] [PubMed] [Google Scholar]
- 46.Wu D, Hu L, Han M, Deng Y, Zhang Y, Ren G, Zhao X, Li Z, Li P, Zhang Y, et al. (2022). PD-1 signaling facilitates activation of lymphoid tissue inducer cells by restraining fatty acid oxidation. Nat Metab 4, 867–882. 10.1038/s42255-022-00595-9. [DOI] [PubMed] [Google Scholar]
- 47.Sachs N, Secades P, van Hulst L, Kreft M, Song JY, and Sonnenberg A (2012). Loss of integrin alpha3 prevents skin tumor formation by promoting epidermal turnover and depletion of slow-cycling cells. Proc Natl Acad Sci U S A 109, 21468–21473. 10.1073/pnas.1204614110. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Document S1. Figures S1–S7
Table S1. List of differentially expressed gene between RORγtfm+ and RORγtfm- cILC2s, related to Figure 3
Data Availability Statement
All data reported in this paper will be shared by the lead contact upon request.
RNA sequencing and ATAC sequencing experiments are deposited at GSA with accession number GSA: CRA010739.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.