CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife Assessment

    This study presents important findings on the role of CXXC-finger protein 1 in regulatory T cell gene regulation and function. The evidence supporting the authors' claims is solid, with mostly state-of-the-art technology, although the inclusion of more mechanistic insights would have strengthened the study. The work will be of relevance to immunologists interested in regulatory T cell biology and autoimmunity.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

FOXP3-expressing regulatory T (T reg ) cells play a pivotal role in maintaining immune homeostasis and tolerance, with their activation being crucial for preventing various inflammatory responses. However, the mechanisms governing the epigenetic program in T reg cells during their dynamic activation remain unclear. In this study, we demonstrate that CXXC finger protein 1 (CXXC1) interacts with the transcription factor FOXP3 and facilitates the regulation of target genes by modulating H3K4me3 deposition. Cxxc1 deletion in T reg cells leads to severe inflammatory disease and spontaneous T-cell activation, with impaired immunosuppressive function. As a transcriptional regulator, CXXC1 promotes the expression of key T reg functional markers under steady-state conditions, which are essential for the maintenance of T reg cell homeostasis and their suppressive functions. Epigenetically, CXXC1 binds to the genomic regulatory regions of T reg program genes in mouse T reg cells, overlapping with FOXP3 binding sites. Given its critical role in T reg cell homeostasis, CXXC1 presents itself as a promising therapeutic target for autoimmune diseases.

Article activity feed

  1. Author response:

    Reviewer #1 (Public review):

    Summary:

    This work investigated the role of CXXC-finger protein 1 (CXXC1) in regulatory T cells. CXXC1-bound genomic regions largely overlap with Foxp3-bound regions and regions with H3K4me3 histone modifications in Treg cells. CXXC1 and Foxp3 interact with each other, as shown by co-immunoprecipitation. Mice with Treg-specific CXXC1 knockout (KO) succumb to lymphoproliferative diseases between 3 to 4 weeks of age, similar to Foxp3 KO mice. Although the immune suppression function of CXXC1 KO Treg is comparable to WT Treg in an in vitro assay, these KO Tregs failed to suppress autoimmune diseases such as EAE and colitis in Treg transfer models in vivo. This is partly due to the diminished survival of the KO Tregs after transfer. CXXC1 KO Tregs do not have an altered DNA methylation pattern; instead, they display weakened H3K4me3 modifications within the broad H3K4me3 domains, which contain a set of Treg signature genes. These results suggest that CXXC1 and Foxp3 collaborate to regulate Treg homeostasis and function by promoting Treg signature gene expression through maintaining H3K4me3 modification.

    Strengths:

    Epigenetic regulation of Treg cells has been a constantly evolving area of research. The current study revealed CXXC1 as a previously unidentified epigenetic regulator of Tregs. The strong phenotype of the knockout mouse supports the critical role CXXC1 plays in Treg cells. Mechanistically, the link between CXXC1 and the maintenance of broad H3K4me3 domains is also a novel finding.

    Weaknesses:

    (1) It is not clear why the authors chose to compare H3K4me3 and H3K27me3 enriched genomic regions. There are other histone modifications associated with transcription activation or repression. Please provide justification.

    Thank you for highlighting this important point. We prioritized H3K4me3 and H3K27me3 because they are well-established markers of transcriptional activation and repression, respectively. These modifications provide a robust framework for investigating the dynamic interplay of chromatin states in Treg cells, particularly in regulating the balance between activation and suppression of key genes. While histone acetylation, such as H3K27ac, is linked to enhancer activity and transcriptional elongation, our focus was on promoter-level regulation, where H3K4me3 and H3K27me3 are most relevant. Although other histone modifications could provide additional insights, we chose to focus on these two to maintain clarity and feasibility in our analysis. We are happy to further elaborate on this rationale in the manuscript if necessary.

    (2) It is not clear what separates Clusters 1 and 3 in Figure 1C. It seems they share the same features.

    We apologize for not clarifying these clusters clearly. Cluster 1 and 3 are both H3K4me3 only group, with H3K4me3 enrichment and gene expression levels being higher in Cluster 1. At first, we divided the promoters into four categories because we wanted to try to classify them into four categories: H3K4me3 only, H3K27me3 only, H3K4me3-H3K27me3 co-occupied, and None. However, in actual classification, we could not distinguish H3K4me3-H3K27me3 co-occupied group. Instead, we had two categories of H3K4me3 only, with cluster 1 having a higher enrichment level for H3K4me3 and gene expression levels.

    (3) The claim, "These observations support the hypothesis that FOXP3 primarily functions as an activator by promoting H3K4me3 deposition in Treg cells." (line 344), seems to be a bit of an overstatement. Foxp3 certainly can promote transcription in ways other than promoting H3K3me3 deposition, and it also can repress gene transcription without affecting H3K27me3 deposition. Therefore, it is not justified to claim that promoting H3K4me3 deposition is Foxp3's primary function.

    We appreciate the reviewer’s thoughtful observation regarding our claim about FOXP3’s role in promoting H3K4me3 deposition. We acknowledge that FOXP3 is a multifunctional transcription factor with diverse mechanisms of action, including transcriptional activation independent of H3K4me3 deposition and transcriptional repression that does not necessarily involve H3K27me3 deposition.

    Our intention was not to imply that promoting H3K4me3 deposition is the exclusive or predominant function of FOXP3 but rather to highlight that this mechanism contributes significantly to its role in regulating Treg cell function. We agree that our wording may have overstated this point, and we will revise the text to provide a more nuanced interpretation. Specifically, we will clarify that our observations suggest FOXP3 can facilitate transcriptional activation, in part, by promoting H3K4me3 deposition, but this does not preclude its other regulatory mechanisms.

    (4) For the in vitro suppression assay in Figure S4C, and the Treg transfer EAE and colitis experiments in Figure 4, the Tregs should be isolated from Cxxc1 fl/fl x Foxp3 cre/wt female heterozygous mice instead of Cxxc1 fl/fl x Foxp3 cre/cre (or cre/Y) mice. Tregs from the homozygous KO mice are already activated by the lymphoproliferative environment and could have vastly different gene expression patterns and homeostatic features compared to resting Tregs. Therefore, it's not a fair comparison between these activated KO Tregs and resting WT Tregs.

    Thank you for this insightful comment and for pointing out the potential confounding effects associated with using Treg cells from homozygous Foxp3Cre/Cre (or Cre/Y) Cxxc1fl/fl mice. We agree that using Treg cells from _Foxp3_Cre/+ _Cxxc1_fl/fl (referred to as “het-KO”) and their littermate _Foxp3_Cre/+ _Cxxc1_fl/+ (referred to as “het-WT”) female mice would provide a more balanced comparison, as these Treg cells are less likely to be influenced by the activated lymphoproliferative environment present in homozygous KO mice.

    To address this concern, we will perform additional experiments using Treg cells isolated from _Foxp3_Cre/+ _Cxxc1_fl/fl (“het-KO”) and their littermate _Foxp3_Cre/+ _Cxxc1_fl/+ (“het-WT”) female mice. We will update the manuscript with these new data to provide a more accurate assessment of the impact of CXXC1 deficiency on Treg cell function.

    (5) The manuscript didn't provide a potential mechanism for how CXXC1 strengthens broad H3K4me3-modified genomic regions. The authors should perform Foxp3 ChIP-seq or Cut-n-Taq with WT and Cxxc1 cKO Tregs to determine whether CXXC1 deletion changes Foxp3's binding pattern in Treg cells.

    Thank you for your insightful comments and valuable suggestions. We greatly appreciate your recommendation to explore the potential mechanism by which CXXC1 enhances broad H3K4me3-modified genomic regions.

    In response, we plan to conduct CUT&Tag experiments for Foxp3 in both WT and Cxxc1 cKO Treg cells.

    Reviewer #2 (Public review):

    FOXP3 has been known to form diverse complexes with different transcription factors and enzymes responsible for epigenetic modifications, but how extracellular signals timely regulate FOXP3 complex dynamics remains to be fully understood. Histone H3K4 tri-methylation (H3K4me3) and CXXC finger protein 1 (CXXC1), which is required to regulate H3K4me3, also remain to be fully investigated in Treg cells. Here, Meng et al. performed a comprehensive analysis of H3K4me3 CUT&Tag assay on Treg cells and a comparison of the dataset with the FOXP3 ChIP-seq dataset revealed that FOXP3 could facilitate the regulation of target genes by promoting H3K4me3 deposition.

    Moreover, CXXC1-FOXP3 interaction is required for this regulation. They found that specific knockdown of Cxxc1 in Treg leads to spontaneous severe multi-organ inflammation in mice and that Cxxc1-deficient Treg exhibits enhanced activation and impaired suppression activity. In addition, they have also found that CXXC1 shares several binding sites with FOXP3 especially on Treg signature gene loci, which are necessary for maintaining homeostasis and identity of Treg cells.

    The findings of the current study are pretty intriguing, and it would be great if the authors could fully address the following comments to support these interesting findings.

    Major points:

    (1) There is insufficient evidence in the first part of the Results to support the conclusion that "FOXP3 functions as an activator by promoting H3K4Me3 deposition in Treg cells". The authors should compare the results for H3K4Me3 in FOXP3-negative conventional T cells to demonstrate that at these promoter loci, FOXP3 promotes H3K4Me3 deposition.

    We appreciate the reviewer’s critical observation regarding our claim about FOXP3’s role in promoting H3K4me3 deposition. We acknowledge that FOXP3 is a multifunctional transcription factor with diverse mechanisms of action, including transcriptional activation independent of H3K4me3 deposition and transcriptional repression that does not necessarily involve H3K27me3 deposition.

    Our intention was not to imply that promoting H3K4me3 deposition is the exclusive or predominant function of FOXP3 but rather to highlight that this mechanism contributes significantly to its role in regulating Treg cell function. We agree that our wording may have overstated this point, and we will revise the text to provide a more nuanced interpretation. Specifically, we will clarify that our observations suggest FOXP3 can facilitate transcriptional activation, in part, by promoting H3K4me3 deposition, but this does not preclude its other regulatory mechanisms.

    We will compare H3K4me3 levels at the promoter loci of interest between FOXP3-negative conventional T cells and FOXP3-positive regulatory T cells. This comparison will help elucidate whether FOXP3 directly promotes H3K4me3 deposition at these loci.

    (2) In Figure 3 F&G, the activation status and IFNγ production should be analyzed in Treg cells and Tconv cells separately rather than in total CD4+ T cells. Moreover, are there changes in autoantibodies and IgG and IgE levels in the serum of cKO mice?

    We appreciate the reviewer’s constructive feedback on the analyses presented in Figures 3F and 3G and the additional suggestion to investigate autoantibodies and serum immunoglobulin levels.

    Regarding Figures 3F and 3G, we agree that separating Treg cells and Tconv cells for analysis of activation status and IFN-γ production would provide a more precise understanding of the cellular dynamics in Cxxc1 cKO mice.

    To address this, we will reanalyze the data to examine Treg and Tconv cells independently and include these results in the revised manuscript.

    As for the changes in autoantibodies and serum IgG and IgE levels, we acknowledge that these parameters are important indicators of systemic immune dysregulation.

    We will now measure serum autoantibodies and immunoglobulin levels in Cxxc1 cKO mice and WT controls.

    (3) Why did Cxxc1-deficient Treg cells not show impaired suppression than WT Treg during in vitro suppression assay, despite the reduced expression of Treg cell suppression assay -associated markers at the transcriptional level demonstrated in both scRNA-seq and bulk RNA-seq?

    Thank you for your thoughtful question. We appreciate your interest in understanding the apparent discrepancy between the reduced expression of Treg-associated suppression markers at the transcriptional level and the lack of impaired suppression observed in the in vitro suppression assay.

    There are several potential explanations for this observation:

    (1) Functional Redundancy: Treg cell suppression is a complex, multi-faceted process involving various effector mechanisms such as cytokine production (e.g., IL-10, TGF-β), cell-cell contact, and metabolic regulation. Thus, even though the transcriptional signature of suppression-associated genes is altered, compensatory mechanisms may still allow Cxxc1-deficient Treg cells to retain functional suppression capacity under these specific in vitro conditions.

    (2) In Vitro Assay Limitations: The in vitro suppression assay is a simplified model of Treg function that may not capture all the complexities of Treg-mediated suppression in vivo. While we observed altered gene expression in Cxxc1-deficient Treg cells, this might not directly translate to a functional defect under the specific conditions of the assay. In vivo, additional factors such as cytokine milieu, cell-cell interactions, and tissue-specific environments may be required for full suppression, which could be missing in the in vitro assay.

    (4) Is there a disease in which Cxxc1 is expressed at low levels or absent in Treg cells? Is the same immunodeficiency phenotype present in patients as in mice?

    Thank you for your insightful question regarding the role of CXXC1 in Treg cells and its potential link to human disease. To our knowledge, no specific human disease has been identified where CXXC1 is expressed at low levels or absent specifically in Treg cells. There is currently no direct evidence of an immunodeficiency phenotype in human patients that parallels the one observed in Cxxc1-deficient mice.

    Reviewer #3 (Public review):

    In the report entitled "CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells", the authors demonstrated that Cxxc1-deletion in Treg cells leads to the development of severe inflammatory disease with impaired suppressive function. Mechanistically, CXXC1 interacts with Foxp3 and regulates the expression of key Treg signature genes by modulating H3K4me3 deposition. Their findings are interesting and significant. However, there are several concerns regarding their analysis and conclusions.

    Major concerns:

    (1) Despite cKO mice showing an increase in Treg cells in the lymph nodes and Cxxc1-deficient Treg cells having normal suppressive function, the majority of cKO mice died within a month. What causes cKO mice to die from severe inflammation?

    Considering the results of Figures 4 and 5, a decrease in Treg cell population due to their reduced proliferative capacity may be one of the causes. It would be informative to analyze the population of tissue Treg cells.

    We thank the reviewer for this insightful comment and acknowledge the importance of understanding the causes of severe inflammation and early mortality in cKO mice. Based on our data and previous studies, we propose the following explanations:

    (1) Reduced Treg Proliferative Capacity: As shown in Figure 5I, the decreased proportion of FOXP3+Ki67+ Treg cells in cKO mice likely reflects impaired proliferative capacity, which may limit the expansion of functional Treg cells in response to inflammatory cues, particularly in peripheral tissues where active suppression is required.

    (2) Altered Treg Function and Activation: Cxxc1-deficient Treg cells exhibit increased expression of activation markers (Il2ra, Cd69) and pro-inflammatory genes (Ifng, Tbx21). This suggests a functional dysregulation that may impair their ability to suppress inflammation effectively, despite their presence in lymphoid organs.

    (3) Tissue Treg Populations: Although our study focuses on lymph node-resident Treg cells, tissue-resident Treg cells play a crucial role in maintaining local immune homeostasis. It is plausible that Cxxc1 deficiency compromises the accumulation or functionality of tissue Treg cells, contributing to uncontrolled inflammation in non-lymphoid organs. Unfortunately, we currently lack data on tissue Treg populations, which limits our ability to directly address this hypothesis.

    Regarding the suggestion to analyze tissue Treg populations, we agree that this would be an important next step in understanding the cause of the severe inflammation and early mortality in Cxxc1-deficient mice.

    We plan to perform detailed analyses of Treg cell populations in various tissues, including the gut, lung, and liver, to determine if there are specific defects in tissue-resident Treg cells that could contribute to the observed phenotype.

    (2) In Figure 5B, scRNA-seq analysis indicated that Mki67+ Treg subset are comparable between WT and Cxxc1-deficient Treg cells. On the other hand, FACS analysis demonstrated that Cxxc1-deficient Treg shows less Ki-67 expression compared to WT in Figure 5I. The authors should explain this discrepancy.

    Thank you for pointing out the apparent discrepancy between the scRNA-seq and FACS analyses regarding Ki-67 expression in Cxxc1-deficient Treg cells.

    In Figure 5B, the scRNA-seq analysis identified the Mki67+ Treg subset as comparable between WT and Cxxc1-deficient Treg cells. This finding reflects the overall proportion of cells expressing Mki67 transcripts within the Treg population. In contrast, the FACS analysis in Figure 5I specifically measures Ki-67 protein levels, revealing reduced expression in Cxxc1-deficient Treg cells compared to WT.

    To address this discrepancy more comprehensively, we will further analyze the scRNA-seq data to directly compare Mki67 mRNA expression levels between WT and Cxxc1-deficient Treg cells.

    In addition, the authors concluded on line 441 that CXXC1 plays a crucial role in maintaining Treg cell stability. However, there appears to be no data on Treg stability. Which data represent the Treg stability?

    We appreciate the reviewer’s observation and recognize that our wording may have been overly conclusive. Our data primarily highlight the impact of Cxxc1 deficiency on Treg cell homeostasis and transcriptional regulation, rather than providing direct evidence for Treg cell stability. Specifically, the downregulation of Treg-specific suppressive genes (Nt5e, Il10, Pdcd1) and the upregulation of pro-inflammatory markers (Gzmb, Ifng, Tbx21) indicate a shift in functional states. While these findings may suggest an indirect disruption in the maintenance of suppressive phenotypes, they do not constitute a direct measure of Treg cell stability.

    To address the reviewer’s concern, we will revise our conclusion to more accurately state that our data support a role for CXXC1 in maintaining Treg cell homeostasis and functional balance, without overextending claims about Treg cell stability. Thank you for bringing this to our attention, as it will help us improve the clarity and precision of our manuscript.

    (3) The authors found that Cxxc1-deficient Treg cells exhibit weaker H3K4me3 signals compared to WT in Figure 7. This result suggests that Cxxc1 regulates H3K4me3 modification via H3K4 methyltransferases in Treg cells. The authors should clarify which H3K4 methyltransferases contribute to the modulation of H3K4me3 deposition by Cxxc1 in Treg cells.

    Thank you for pointing out the need to clarify the role of H3K4 methyltransferases in the modulation of H3K4me3 deposition by CXXC1 in Treg cells.

    In our study, we found that Cxxc1-deficient Treg cells exhibit reduced H3K4me3 levels, as shown in Figure 7. CXXC1 has been previously reported to function as a non-catalytic component of the Set1/COMPASS complex, which contains H3K4 methyltransferases such as SETD1A and SETD1B. These methyltransferases are the primary enzymes responsible for H3K4 trimethylation.

    References:

    (1) Lee J.H., Skalnik D.G. CpG-binding protein (CXXC finger protein 1) is a component of the mammalian Set1 histone H3-Lys4 methyltransferase complex, the analogue of the yeast Set1/COMPASS complex. J. Biol. Chem. 2005; 280:41725–41731.

    (2). J. P. Thomson, P. J. Skene, J. Selfridge, T. Clouaire, J. Guy, S. Webb, A. R. W. Kerr, A. Deaton, R. Andrews, K. D. James, D. J. Turner, R. Illingworth, A. Bird, CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464, 1082–1086 (2010).

    (3) Shilatifard, A. 2012. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu. Rev. Biochem. 81:65–95.

    (4) Brown D.A., Di Cerbo V., Feldmann A., Ahn J., Ito S., Blackledge N.P., Nakayama M., McClellan M., Dimitrova E., Turberfield A.H. et al. The SET1 complex selects actively transcribed target genes via multivalent interaction with CpG Island chromatin. Cell Rep. 2017; 20:2313–2327.

    Furthermore, it would be important to investigate whether Cxxc1-deletion alters Foxp3 binding to target genes.

    Thank you for this important suggestion regarding the impact of Cxxc1 deletion on FOXP3 binding to target genes. We agree that understanding whether Cxxc1 deficiency affects FOXP3’s ability to bind to its target genes would provide valuable insight into the regulatory role of CXXC1 in Treg cell function.

    To address this, we plan to perform CUT&Tag experiments to assess FOXP3 binding profiles in Cxxc1-deficient versus wild-type Treg cells. These experiments will allow us to determine if Cxxc1 loss disrupts FOXP3’s occupancy at key regulatory sites, which may contribute to the observed functional impairments in Treg cells.

    (4) In Figure 7, the authors concluded that CXXC1 promotes Treg cell homeostasis and function by preserving the H3K4me3 modification since Cxxc1-deficient Treg cells show lower H3K4me3 densities at the key Treg signature genes. Are these Cxxc1-deficient Treg cells derived from mosaic mice? If Cxxc1-deficient Treg cells are derived from cKO mice, the gene expression and H3K4me3 modification status are inconsistent because scRNA-seq analysis indicated that expression of these Treg signature genes was increased in Cxxc1-deficient Treg cells compared to WT (Figure 5F and G).

    Thank you for the insightful comment. To clarify, the Cxxc1-deficient Treg cells analyzed for H3K4me3 modification in Figure 7 were indeed derived from Cxxc1 conditional knockout (cKO) mice, not mosaic mice.

    The scRNA-seq analysis presented in Figures 5F and G revealed an upregulation of Treg signature genes in Cxxc1-deficient Treg cells. This finding suggests that the loss of Cxxc1 drives these cells toward a pro-inflammatory, activated state, underscoring the pivotal role of CXXC1 in maintaining Treg cell homeostasis and suppressive function.

    Regarding the apparent discrepancy between the reduced H3K4me3 levels and the increased expression of these genes, it is important to note that H3K4me3 primarily functions as an epigenetic mark that facilitates chromatin accessibility and transcriptional regulation, acting as an upstream modulator of gene expression. However, gene expression levels are also influenced by downstream compensatory mechanisms and complex inflammatory environments. In this context, the reduction in H3K4me3 likely reflects the direct role of CXXC1 in epigenetic regulation, whereas the upregulation of gene expression in Cxxc1-deficient Treg cells may result as a side effect of the inflammatory environment.

    To further substantiate our findings, we performed RNA-seq analysis on Treg cells from _Foxp3_Cre/+ _Cxxc1_fl/fl (“het-KO”) and their littermate _Foxp3_Cre/+ _Cxxc1_fl/+ (“het-WT”) female mice, as presented in Figure S6C. This analysis revealed a notable reduction in the expression of key Treg signature genes, including Icos, Ctla4, Tnfrsf18, and Nt5e, in het-KO Treg cells. Importantly, the observed changes in gene expression were consistent with the altered H3K4me3 modification status, further supporting the epigenetic regulatory role of CXXC1. These results further emphasize the critical role of CXXC1 promotes Treg cell homeostasis and function by preserving the H3K4me3 modification.

  2. eLife Assessment

    This study presents important findings on the role of CXXC-finger protein 1 in regulatory T cell gene regulation and function. The evidence supporting the authors' claims is solid, with mostly state-of-the-art technology, although the inclusion of more mechanistic insights would have strengthened the study. The work will be of relevance to immunologists interested in regulatory T cell biology and autoimmunity.

  3. Reviewer #1 (Public review):

    Summary:

    This work investigated the role of CXXC-finger protein 1 (CXXC1) in regulatory T cells. CXXC1-bound genomic regions largely overlap with Foxp3-bound regions and regions with H3K4me3 histone modifications in Treg cells. CXXC1 and Foxp3 interact with each other, as shown by co-immunoprecipitation. Mice with Treg-specific CXXC1 knockout (KO) succumb to lymphoproliferative diseases between 3 to 4 weeks of age, similar to Foxp3 KO mice. Although the immune suppression function of CXXC1 KO Treg is comparable to WT Treg in an in vitro assay, these KO Tregs failed to suppress autoimmune diseases such as EAE and colitis in Treg transfer models in vivo. This is partly due to the diminished survival of the KO Tregs after transfer. CXXC1 KO Tregs do not have an altered DNA methylation pattern; instead, they display weakened H3K4me3 modifications within the broad H3K4me3 domains, which contain a set of Treg signature genes. These results suggest that CXXC1 and Foxp3 collaborate to regulate Treg homeostasis and function by promoting Treg signature gene expression through maintaining H3K4me3 modification.

    Strengths:

    Epigenetic regulation of Treg cells has been a constantly evolving area of research. The current study revealed CXXC1 as a previously unidentified epigenetic regulator of Tregs. The strong phenotype of the knockout mouse supports the critical role CXXC1 plays in Treg cells. Mechanistically, the link between CXXC1 and the maintenance of broad H3K4me3 domains is also a novel finding.

    Weaknesses:

    (1) It is not clear why the authors chose to compare H3K4me3 and H3K27me3 enriched genomic regions. There are other histone modifications associated with transcription activation or repression. Please provide justification.

    (2) It is not clear what separates Clusters 1 and 3 in Figure 1C. It seems they share the same features.

    (3) The claim, "These observations support the hypothesis that FOXP3 primarily functions as an activator by promoting H3K4me3 deposition in Treg cells." (line 344), seems to be a bit of an overstatement. Foxp3 certainly can promote transcription in ways other than promoting H3K3me3 deposition, and it also can repress gene transcription without affecting H3K27me3 deposition. Therefore, it is not justified to claim that promoting H3K4me3 deposition is Foxp3's primary function.

    (4) For the in vitro suppression assay in Figure S4C, and the Treg transfer EAE and colitis experiments in Figure 4, the Tregs should be isolated from Cxxc1 fl/fl x Foxp3 cre/wt female heterozygous mice instead of Cxxc1 fl/fl x Foxp3 cre/cre (or cre/Y) mice. Tregs from the homozygous KO mice are already activated by the lymphoproliferative environment and could have vastly different gene expression patterns and homeostatic features compared to resting Tregs. Therefore, it's not a fair comparison between these activated KO Tregs and resting WT Tregs.

    (5) The manuscript didn't provide a potential mechanism for how CXXC1 strengthens broad H3K4me3-modified genomic regions. The authors should perform Foxp3 ChIP-seq or Cut-n-Taq with WT and Cxxc1 cKO Tregs to determine whether CXXC1 deletion changes Foxp3's binding pattern in Treg cells.

  4. Reviewer #2 (Public review):

    FOXP3 has been known to form diverse complexes with different transcription factors and enzymes responsible for epigenetic modifications, but how extracellular signals timely regulate FOXP3 complex dynamics remains to be fully understood. Histone H3K4 tri-methylation (H3K4me3) and CXXC finger protein 1 (CXXC1), which is required to regulate H3K4me3, also remain to be fully investigated in Treg cells. Here, Meng et al. performed a comprehensive analysis of H3K4me3 CUT&Tag assay on Treg cells and a comparison of the dataset with the FOXP3 ChIP-seq dataset revealed that FOXP3 could facilitate the regulation of target genes by promoting H3K4me3 deposition.

    Moreover, CXXC1-FOXP3 interaction is required for this regulation. They found that specific knockdown of Cxxc1 in Treg leads to spontaneous severe multi-organ inflammation in mice and that Cxxc1-deficient Treg exhibits enhanced activation and impaired suppression activity. In addition, they have also found that CXXC1 shares several binding sites with FOXP3 especially on Treg signature gene loci, which are necessary for maintaining homeostasis and identity of Treg cells.

    The findings of the current study are pretty intriguing, and it would be great if the authors could fully address the following comments to support these interesting findings.

    Major points:

    (1) There is insufficient evidence in the first part of the Results to support the conclusion that "FOXP3 functions as an activator by promoting H3K4Me3 deposition in Treg cells". The authors should compare the results for H3K4Me3 in FOXP3-negative conventional T cells to demonstrate that at these promoter loci, FOXP3 promotes H3K4Me3 deposition.

    (2) In Figure 3 F&G, the activation status and IFNγ production should be analyzed in Treg cells and Tconv cells separately rather than in total CD4+ T cells. Moreover, are there changes in autoantibodies and IgG and IgE levels in the serum of cKO mice?

    (3) Why did Cxxc1-deficient Treg cells not show impaired suppression than WT Treg during in vitro suppression assay, despite the reduced expression of Treg cell suppression assay -associated markers at the transcriptional level demonstrated in both scRNA-seq and bulk RNA-seq?

    (4) Is there a disease in which Cxxc1 is expressed at low levels or absent in Treg cells? Is the same immunodeficiency phenotype present in patients as in mice?

  5. Reviewer #3 (Public review):

    In the report entitled "CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells", the authors demonstrated that Cxxc1-deletion in Treg cells leads to the development of severe inflammatory disease with impaired suppressive function. Mechanistically, CXXC1 interacts with Foxp3 and regulates the expression of key Treg signature genes by modulating H3K4me3 deposition. Their findings are interesting and significant. However, there are several concerns regarding their analysis and conclusions.

    Major concerns:

    (1) Despite cKO mice showing an increase in Treg cells in the lymph nodes and Cxxc1-deficient Treg cells having normal suppressive function, the majority of cKO mice died within a month. What causes cKO mice to die from severe inflammation?

    Considering the results of Figures 4 and 5, a decrease in Treg cell population due to their reduced proliferative capacity may be one of the causes. It would be informative to analyze the population of tissue Treg cells.

    (2) In Figure 5B, scRNA-seq analysis indicated that Mki67+ Treg subset are comparable between WT and Cxxc1-deficient Treg cells. On the other hand, FACS analysis demonstrated that Cxxc1-deficient Treg shows less Ki-67 expression compared to WT in Figure 5I. The authors should explain this discrepancy.

    In addition, the authors concluded on line 441 that CXXC1 plays a crucial role in maintaining Treg cell stability. However, there appears to be no data on Treg stability. Which data represent the Treg stability?

    (3) The authors found that Cxxc1-deficient Treg cells exhibit weaker H3K4me3 signals compared to WT in Figure 7. This result suggests that Cxxc1 regulates H3K4me3 modification via H3K4 methyltransferases in Treg cells. The authors should clarify which H3K4 methyltransferases contribute to the modulation of H3K4me3 deposition by Cxxc1 in Treg cells.

    Furthermore, it would be important to investigate whether Cxxc1-deletion alters Foxp3 binding to target genes.

    (4) In Figure 7, the authors concluded that CXXC1 promotes Treg cell homeostasis and function by preserving the H3K4me3 modification since Cxxc1-deficient Treg cells show lower H3K4me3 densities at the key Treg signature genes. Are these Cxxc1-deficient Treg cells derived from mosaic mice? If Cxxc1-deficient Treg cells are derived from cKO mice, the gene expression and H3K4me3 modification status are inconsistent because scRNA-seq analysis indicated that expression of these Treg signature genes was increased in Cxxc1-deficient Treg cells compared to WT (Figure 5F and G).