Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Newest Articles
    • Current Issue
    • Methods & Resources
    • Author Interviews
    • Archive
    • Subjects
  • Collections
  • Submit
    • Submit a Manuscript
    • Author Guidelines
    • License, Copyright, Fee
    • FAQ
    • Why submit
  • About
    • About Us
    • Editors & Staff
    • Board Members
    • Licensing and Reuse
    • Reviewer Guidelines
    • Privacy Policy
    • Advertise
    • Contact Us
    • LSA LLC
  • Alerts
  • Other Publications
    • EMBO Press
    • The EMBO Journal
    • EMBO reports
    • EMBO Molecular Medicine
    • Molecular Systems Biology
    • Rockefeller University Press
    • Journal of Cell Biology
    • Journal of Experimental Medicine
    • Journal of General Physiology
    • Journal of Human Immunity
    • Cold Spring Harbor Laboratory Press
    • Genes & Development
    • Genome Research

User menu

  • My alerts

Search

  • Advanced search
Life Science Alliance
  • Other Publications
    • EMBO Press
    • The EMBO Journal
    • EMBO reports
    • EMBO Molecular Medicine
    • Molecular Systems Biology
    • Rockefeller University Press
    • Journal of Cell Biology
    • Journal of Experimental Medicine
    • Journal of General Physiology
    • Journal of Human Immunity
    • Cold Spring Harbor Laboratory Press
    • Genes & Development
    • Genome Research
  • My alerts
Life Science Alliance

Advanced Search

  • Home
  • Articles
    • Newest Articles
    • Current Issue
    • Methods & Resources
    • Author Interviews
    • Archive
    • Subjects
  • Collections
  • Submit
    • Submit a Manuscript
    • Author Guidelines
    • License, Copyright, Fee
    • FAQ
    • Why submit
  • About
    • About Us
    • Editors & Staff
    • Board Members
    • Licensing and Reuse
    • Reviewer Guidelines
    • Privacy Policy
    • Advertise
    • Contact Us
    • LSA LLC
  • Alerts
  • Follow LSA on Bluesky
  • Follow lsa Template on Twitter
Research Article
Transparent Process
Open Access

Identification of a novel enhancer essential for Satb1 expression in TH2 cells and activated ILC2s

View ORCID ProfileAneela Nomura, Tetsuro Kobayashi, View ORCID ProfileWooseok Seo, View ORCID ProfileMichiko Ohno-Oishi, Kiyokazu Kakugawa, Sawako Muroi, Hideyuki Yoshida, Takaho A Endo, Kazuyo Moro, View ORCID ProfileIchiro Taniuchi  Correspondence email
Aneela Nomura
1Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Formal analysis, Investigation, Writing—original draft, Writing—review and editing
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Aneela Nomura
Tetsuro Kobayashi
2Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wooseok Seo
1Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Wooseok Seo
Michiko Ohno-Oishi
1Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Michiko Ohno-Oishi
Kiyokazu Kakugawa
3Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sawako Muroi
1Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Methodology
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hideyuki Yoshida
4Laboratory for YCI Laboratory for Immunological Transcriptomics, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Resources, Formal analysis, Methodology
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Takaho A Endo
5Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Resources, Software, Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kazuyo Moro
2Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
6Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School for Medicine, Osaka University, Osaka, Japan
Roles: Formal analysis
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ichiro Taniuchi
1Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences(IMS), Yokohama, Japan
Roles: Conceptualization, Supervision, Writing—original draft, Writing—review and editing
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Ichiro Taniuchi
  • For correspondence: ichiro.taniuchi@riken.jp
Published 16 May 2023. DOI: 10.26508/lsa.202301897
  • Article
  • Figures & Data
  • Info
  • Metrics
  • Reviewer Comments
  • PDF
Loading

Abstract

The genome organizer, special AT-rich binding protein-1 (SATB1), functions to globally regulate gene networks during primary T cell development and plays a pivotal role in lineage specification in CD4+ helper-, CD8+ cytotoxic-, and FOXP3+ regulatory-T cell subsets. However, it remains unclear how Satb1 gene expression is controlled, particularly in effector T cell function. Here, by using a novel reporter mouse strain expressing SATB1-Venus and genome editing, we have identified a cis-regulatory enhancer, essential for maintaining Satb1 expression specifically in TH2 cells. This enhancer is occupied by STAT6 and interacts with Satb1 promoters through chromatin looping in TH2 cells. Reduction of Satb1 expression, by the lack of this enhancer, resulted in elevated IL-5 expression in TH2 cells. In addition, we found that Satb1 is induced in activated group 2 innate lymphoid cells (ILC2s) through this enhancer. Collectively, these results provide novel insights into how Satb1 expression is regulated in TH2 cells and ILC2s during type 2 immune responses.

Introduction

The nuclear protein, special AT-rich binding protein 1 (SATB1), functions as a genome organizer and regulates highly ordered chromatin structures by tethering specialized AT-rich genomic regions, such as base unpairing regions (1, 2). SATB1 epigenetically regulates gene expression by recruiting various chromatin modifiers and nucleosome remodelling and deacetylase (NURD) complexes (3) and promote heterochromatin formation. Multiple studies have delineated the role of SATB1 for postnatal neuronal development and function (4, 5). In addition, SATB1 is highly expressed in the thymus and is deemed essential for the development of mature thymocytes (6). CD4+CD8+ double-positive (DP) immature thymocytes undergo a TCR-mediated selection process, known as positive selection, to become mature thymocytes that are committed to become either helper-(TH), cytotoxic-(TC) or regulatory-T (Treg) cells. In this context, SATB1 is required to control expression of genes encoding lineage specification transcription factors, Thpok, Runx3, and FoxP3 for TH, TC, and Treg cells, respectively. In the absence of SATB1, aberrant expression of these transcription factors occurs through their derepression. For instance, Thpok expression was induced in MHC-I-restricted TC cells and Foxp3 expression was induced in conventional CD4 T cells (7, 8).

The functions of SATB1 extend into the differentiation of effector T cells after encountering antigens in the periphery. Satb1 expression is thought to be under the control of TCR signalling (9), which is counteracted by TGF-β signalling (10). In these effector T cells, SATB1 functions to repress PD-1 expression and suppress T cell exhaustion (10). Lastly, Satb1 expression is increased upon IL-23 stimulation in pathogenic IL-17-producing TH17s and promotes their pathogenicity in experimental autoimmune encephalomyelitis via regulation of GM-CSF production and suppression of PD-1 (11). The role of SATB1 in CD4+ type 2 helper (TH2) differentiation has been characterized but has found very contradictory results. Based on the identification of SATB1-binding sites in the TH2 locus containing Il-5, Il-4, and Il-13 genes in a murine TH2 cell line in vitro, the first report claimed that SATB1 functioned as a positive regulator of TH2 cytokine expression (12). Other studies, however, demonstrated that SATB1 only represses IL-5 expression in human CD4 TH2 cultures (13). It was also reported that SATB1 cooperates with β-catenin to control the expression of Gata3, the key TH2 lineage transcription factor critical for TH2 differentiation and function (14). However, conditional loss of Satb1 in CD4+ T cells, using the ThPOK-Cre mouse strain, showed that loss of Satb1 expression had no detrimental effects on TH2 differentiation, at least in vitro. Furthermore, it has been shown that the expression of Satb1 is controlled by both IL-4 and NFκB signalling in vitro (15).

Currently, there is very little mechanistic insight to how Satb1 gene expression is transcriptionally induced in T cells. In mice, there are four alternative promoters, P1, P2, P3, and P4 generate Satb1-1a, Satb1-1b, Satb1-1c, and Satb1-1d transcripts, respectively. Usage of these four Satb1 promoters is differentially regulated in CD4 TH2 in vitro (15, 16). Interestingly, a GWAS study mapped a Psoriasis-associated single nucleotide polymorphism (SNP, rs73178598) around 240 kb upstream of the SATB1 locus, where an antisense noncoding RNA called SATB1-AS1 is transcribed (17). This study hinted the presence of cis-regulatory genomic elements that could control Satb1 expression in various effector T cells and contribute to tissue inflammation. Thus, it is quite important to define Satb1 expression during effector T cell differentiation and how it is controlled.

In this study, we used a novel Satb11-Venus reporter strain and a genome editing technology to identify a novel IL-4-responsive TH2-specific enhancer for Satb1, defined as Satb1-Eth2. Satb1-Eth2 is essential to maintain Satb1 expression, not only in CD4 TH2 cells but also in activated ILC2s. Furthermore, loss of Satb1-Eth2 resulted in elevated IL-5 expression in CD4 TH2 cells. Collectively, our study unravels mechanisms by which Satb1 expression is retained in immune cells mediating type 2 immune responses.

Results

Strict control of Satb1 expression revealed by a Satb1-Venus fusion reporter

To quantify SATB1 protein expression in various types of murine cells, particularly in T cells and at single cell level, we generated a Satb1Venus allele by the knock-in approach to insert the Venus open reading frame, downstream of the initiation codon (in exon2) of the Satb1 gene (Figs 1A and S1A and B). In Satb1+/Venus thymocytes, the Satb1Venus allele expressed the SATB1-Venus protein (Fig S1C) that showed a cage-like distribution in the nucleus, as previously reported (Fig S1D) (18). Phenotypic analyses in Satb1Venus/Venus mice did not show abnormalities in thymocyte development, that was observed in Satb1Flx/Flx Cd4-cre mice, and the numbers of mature CD4 and CD8 SP thymocytes were comparable with the wildtype (Fig S1E). Thus, the SATB1-Venus fusion protein is likely to retain endogenous SATB1 function to support, at least, primary T cell development. Using Satb1+/Venus mice, we observed that most of the CD4−CD8− DN thymocytes expressed low levels of SATB1-Venus (Figs 1B and S1F). Further analyses of DN sub-populations revealed that SATB1-Venus is initially lowly expressed in CD44+CD25− DN1 thymocytes, and it is then incrementally increased with DN1 transitioning into CD44+CD25+ DN2 and CD44−CD25+ DN3 thymocyte stages (Fig S1F). In CD25−CD44− DN4 thymocytes, there were SATB1-Venushi and SATB1-Venuslo populations, the latter of which is likely to be non-T lymphoid cells. There was also a significant increase in the number of SATB1-Venus+ DP thymocytes, during their transition from CD4−CD8− DN to CD4+CD8+ DP stage (Fig 1B). Notably, DP thymocytes undergoing positive selection (CD69+TCRβmid and CD69+TCRβhi) showed further increase in SATB1-Venus expression (Fig S1G), which may suggest that SATB1 expression, is positively controlled by TCR stimulation. In addition, there was a significant decline in SATB1-Venus expression from positively selected DPs to mature thymocytes (Figs 1B and S1G). Moreover, analyses of Satb1 transcript levels in DN3, DP, CD4 SP, and CD8 SP thymocytes from the immunological genome project (ImmGen, https://www.immgen.org/) (19) were highly consistent with the SATB1-Venus protein levels (Fig 1B). These data suggest that our Satb1-Venus reporter mouse model can be used to endogenously study the SATB1 protein function. In thymic Tregs, defined as CD24loTCRβhiCD4+CD8α−CD25+ cells, SATB1-Venus expression was lower than their conventional CD4 counterparts (Fig 1C). Further analyses of nonconventional T cell subsets, such as the invariant natural killer T (iNKTs) and γδ-T cells, showed that iNKTs cells expressed similar levels of SATB1-Venus to that in conventional CD4 SP cells but was significantly higher than in γδ-T cells. (Fig S1H).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1. Expression of SATB1-venus reporter during T cell development and activation.

(A) Schematic shows Satb1-Venus gene locus in Satb1Venus knockin mice. (B) Histograms, on the left, show expression of SATB1-Venus in CD4−CD8− DN, CD4+CD8+ DP, CD24loTCRβhiCD4+CD8− mature SP and CD24loTCRβhiCD4−CD8+ mature SP thymocytes of Satb1+/Venus mice. Numerical values indicate SATB1-VENUS MFI (mean fluorescence intensity) and data are a representative of three biological experiments and summarised in the middle graph. Statistics were calculated by two-way ANNOVA Tukey`s multiple comparisons: *p- < 0.05, **p- < 0.01, ***p- < 0.001. Graph, on the right, shows Satb1 mRNA expression in DN3, DP, CD4 SP and CD8 SP thymocytes, derived from IMMGEN RNA-seq database. (C) Histograms show SATB1-Venus expression in conventional CD4+CD25− versus regulatory CD4+CD25+ mature thymocytes of Satb1+/Venus mice. Numerical values indicate SATB1-Venus MFI. Data are representative of three biological experiments. (D) Histograms, on the left, show expression of SATB1-Venus in splenic B cells (B220+TCRβ−), T cells (B220−TCRβ+), CD4 T cells (TCRβ+CD4+CD8α−), and CD8 T cells (TCRβ+CD4−CD8α+) of Satb1+/Venus mice. Data are summarised in the middle graph from three biological experiments and statistics were calculated by two-way ANNOVA Tukey`s multiple comparisons: *P- < 0.05, **P- < 0.01, ***P- < 0.001. Graph on the right shows Satb1 expression in B cells, naïve CD4, and naïve CD8 T cells from IMMGEN RNA-seq database. (E) Histograms show SATB1-Venus expression in CD4 or CD8 T cell subpopulations: CD62LhiCD44lo naïve, CD62LhiCD44hi central memory, and CD62LloCD44hi effector memory T cells from Satb1+/Venus mice. Numerical values indicate SATB1-Venus MFI and data are a representative of four biological experiments. Data are summarised in the adjacent graph. (F) Contour plots show SATB1-Venus and CD62L expression in splenic versus intraepithelial TCRβ+ T cells of Satb1+/Venus mice, and the CD4 and CD8α expression patterns on CD62L− SATB1-Venuslo and CD62L+SATB1-Venushi T cell populations. Data are representative of three biological experiments. (G) Histograms show SATB1-Venus expression in naïve CD4 T cells undergoing activation in vitro for 0, 24, 48, and 72 h and summarised in the adjacent graph from three biological experiments. Statistics were calculated by unpaired t test; *P- < 0.05, **P- < 0.01, ***P- < 0.001. (H) Histograms show SATB1-Venus expression in naïve CD4 T cells versus in vitro differentiated CD4 TH0, TH1, TH2, TH17, and iTReg cells from Satb1+/Venus mice. Data are representative of three biological experiments and summarised in the adjacent graph. Statistics were calculated by two-way ANNOVA Tukey’s multiple comparisons: *P- < 0.05, **P- < 0.01, ***P- < 0.001.

Figure S1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure S1. Generation and characterisation of Satb1Venus knockin mice.

(A) Schematic shows the strategy to generate Satb1Venus allele by homologous recombination in mouse ES cells. Structures of Satb1 gene, a targeting vector, and targeted Satb1venusN are indicated. Neomycin resistance (neor) gene was removed in ES cells by transient transfection of expression vector-encoding Cre recombinase. (B) The result of genomic PCR to identify ES clone (11-7) that underwent homologous recombination event P; positive control, N; wildtype. (C) Immunoblot analysis shows the expression of SATB1-Venus protein in thymocytes of Satb1+/Venus mouse. (D) Representative image of fluorescent microscopy of thymocytes from Satb1+/Venus mice. (E) Dot plots show frequencies of mature thymocytes (CD24loTCRβhi, top panel) and their CD4/CD8 expression patterns (bottom panel) from Satb1+/+, Satb1Venus/Venus, and Satb1Flx/Flx CD4-Cre mice. Numbers of CD4-SP and CD8-SP thymocytes from Satb1+/+ and Satb1Venus/Venus mice are summarised in the adjacent graph and from at least three biological experiments. (F) Histograms show the expression of SATB1-VENUS in DN1 (CD44+CD25−), DN2 (CD44+CD25+), DN3 (CD44−CD25+), and DN4 (CD44−CD25−) thymocytes from Satb1+/Venus mice. Numerical values indicate SATB1-VENUS MFI and data are representative of three biological experiments. (G) Dot plots, on the left, show flow cytometry-gating strategies to define the following thymocyte populations: preselected CD24hiCD69−TCRβ− (1) and CD24hiCD69−TCRβlo (2) thymocytes; postselected CD24hiCD69+TCRβmid (3) and CD24hiCD69+TCRβhi (4) thymocytes; and mature CD24loTCRβhi (5) thymocytes of Satb1+/Venus mice. Histograms, in the middle, show SATB1-Venus expression in thymocyte populations 1,2,3,4, and 5 of Satb1+/Venus mice. Graph, on the right, shows quantification of SATB1-Venus MFI in DN, preselected DPs (populations 1 and 2), postselected DPs (populations 3 and 4), mature CD4 SP, and mature CD8 SPs thymocytes. Data are summarised from three biological experiments and statistics were calculated by two-way ANNOVA Tukey’s multiple comparisons: *P- < 0.05, **P- < 0.01, ***P- < 0.001. (H) Histograms show the expression of SATB1-Venus in thymic invariant NKT (aGalcer+TCRβmid) and γδ T cells (CD4−CD8α−TCRβ-TCRγδ+). Graph shows quantification of SATB1-Venus MFI in thymic iNKT and γδ T cells from Satb1+/Venus mice, summarised from three biological experiments. Statistics were calculated by paired t-test; *P- < 0.05, **P- < 0.01, ***P- < 0.001. (I) Graphs show the percentages of SATB1-VenusHi and SATB1-VenusLo populations from CD4+ or CD8+ CD62LhiCD44lo naïve, CD62LhiCD44hi central memory, and CD62LloCD44hi effector memory T cells from Satb1+/Venus mice. Data are summarised from four biological experiments.

We next analysed SATB1-Venus expression in peripheral lymphocytes in the spleen. Both CD4+ and CD8+ T cells in the spleen showed higher SATB1-Venus expression than B cells (Fig 1D), which again showed high consistency with the Satb1 transcript levels (https://www.immgen.org/) (19). When CD4+ T cells were further gated for naïve (CD62Lhi CD44lo), central memory (CD62LhiCD44hi), and effector memory (CD62LloCD44hi) populations, we noted a uniform and high SATB1-Venus expression in naïve CD4 T cells (Figs 1E and S1I). On the other hand, there were SATB1-Venushi and SATB1-Venuslo populations in both CD4 central memory and CD4 effector memory populations, with increased frequency of SATB1-Venuslo in effector memory cells. In the CD8 T cell pool, both naïve and central memory populations sustained a uniform and high SATB1-Venus expression, whereas the effector memory T cells consisted of both SATB1-Venushi and SATB1-Venuslo populations. These observations suggest that T cell memory differentiation in the periphery induces down-regulation of SATB1 expression.

Intestinal intraepithelial lymphocytes (IEL) are most likely to represent effector T cells, as they are continuously exposed to various antigens in the gut. In contrast to splenic T cells of Satb1+/Venus mice, 85% of which are SATB1-Venushi cells, only 2.5% of the intestinal TCRβ+ IEL were SATB1-Venushi (Fig 1F). This SATB1-Venushi IEL population expressed CD62L and showed similar CD4 and CD8α expression profiles to that in SATB1-Venushi splenic T cells. These data suggest that these CD62L+SATB1-VenushiTCRβ+ IEL are recent immigrants of circulating αβT cells. On the contrary, the CD62L−SATB1-Venuslo TCRβ+ IEL mainly represented a mixture of CD4+, CD4+CD8α+ and CD4−CD8α+ subpopulations and most of the CD4+CD8α+ cells were nonconventional CD8aα+ IELs (data not shown). This, therefore, suggests that SATB1-Venus could serve as a good marker for separating effector and naïve T cells.

To examine SATB1 expression during T cell activation, SATB1-Venus was quantified in Satb1+/Venus naïve CD4 T cells undergoing TCR activation in vitro. Indeed, SATB1-Venus levels significantly elevated after 24- and 48-h post activation and plateaued after 72 h post activation (Fig 1G). Furthermore, we traced SATB1-Venus expression in various effector CD4 helper T cell subsets that were differentiated in culture. Interestingly, SATB1-Venus expression is highest in CD4 TH1, TH2, and TH17 and lowest in Tregs (Fig 1H). Overall, SATB1 expression in T cells is dynamically regulated during thymocyte development and in their functional differentiation into effector cells, in a cell-context-dependent manner.

Identification of cis-regulatory genomic regions for Satb1 gene regulation

Our Satb1-Venus reporter mice revealed that the amount of SATB1 protein is significantly altered during T-cell development and transcriptome data generated by the ImmGen project (https://www.immgen.org/) (19) shows a nice correlation of SATB1-Venus expression with Satb1 mRNA levels. This indicates that amount of SATB1 protein is mainly regulated at the transcription level, rather than via post-translational mechanisms. Besides the usage of alternative gene promoters, cell-type specific gene expression is often regulated by cis-regulatory elements such as enhancer(s) (20), which are often located far from the transcription start site and shows higher chromatin accessibility (21). Therefore, we used publicly available ATAC-seq data (19) to search for open chromatin regions around the Satb1 locus. There are two genomic regions upstream of the Satb1 gene, which we referred them to as Satb1-a and Satb1-b, respectively, that showed significant ATAC-seq peaks in T cell subsets with activated/memory characteristics (Fig 2A, open chromatin regions highlighted in red dashed boxes). Sequences within these two genomic regions are evolutionally conserved, which imply that these regions may have some functions. To examine whether Satb1-a and Satb1-b genomic regions exhibit enhancer-like signatures, we also utilised publicly available H3K27 acetylation (H3K27Ac) ChIP-seq data (8), and indeed found that Satb1-a was much more highly enriched with H3K27Ac modifications than Satb1-b. This finding suggested that Satb1-a may function as an enhancer. Interestingly, the noncoding RNAs, Gm19585, and Gm20098, are transcribed near the Satb1-a and Satb1-b regions, respectively. RNA-seq data from ImmGen also showed that Gm19585 and Gm20098 are expressed in various T-cell subpopulations (Fig S2A). Specifically, Gm19585 and Satb1 transcripts were mostly co-expressed in various T cell populations, whereas Gm20098 co-expression with Satb1 is only limited to a small number of T cell subsets such as DP thymocytes. These observations prompted us to further study whether Satb1-a and Satb1-b function in Satb1 gene regulation.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2. Analyses of Satb1-a and Satb1-b function in regulating Satb1 expression.

(A) ATAC-seq and H3K27Ac ChIP-seq signals in Satb1, and in upstream genomic regions transcribing Gm20098 and Gm19585 in various murine T cell populations indicated. Red dashed boxes highlight genomic regions, Satb1-a and Satb1-b, that show ATAC-seq and/or H3K27Ac ChIP-seq signals in thymocytes and/or T cells. Satb1-a and Satb1-b are evolutionarily conserved and located near regions transcribing Gm19585 and Gm20098, respectively. (B) Histograms show SATB1-Venus expression in splenic CD4 or CD8 T cell subpopulations (1. CD62LhiCD44lo naïve, 2. CD62LhiCD44hi central memory, and 3.CD62LloCD44hi effector memory T cells) from Satb1+/Venus, Satb1+/Venus-Δa, and Satb1+/Venus-Δb mutant mice. Numerical values indicate SATB1-Venus MFI (mean fluorescence intensity). Graph shows quantification of SATB1-Venus MFI in splenic CD4 and CD8 T cell subpopulations. Data are summarised from four biological experiments. Statistics were calculated by two-way ANNOVA Tukey`s multiple comparisons; *P- < 0.05, **P- < 0.01, ***P- < 0.001. (C) Histograms show SATB1-Venus expression in in vitro differentiated CD4 TH0, TH1, TH2, TH17, and iTReg cells, generated from Satb1Venus-Δa (top, n = 3) and Satb1Venus-Δb (bottom, n = 2) mutant mice. Data are a representative of at least two biological experiments. Adjacent graphs show quantification of SATB1-Venus MFI. Data are summarised from two or three biological experiments. (D) qRT–PCR analysis of Satb1 and Gm19585 expression in Satb1+/+ and Satb1Δa/Δa CD4 TH0, TH1, TH2 cells. Data are summarised from four biological experiments. Statistics were calculated by unpaired t test; *P- < 0.05, **P- < 0.01, ***P- < 0.001, ****P- < 0.0001.

Figure S2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure S2. Analyses of Satb1-a and Satb1-b function in regulating Satb1 expression during T cell development.

(A) Histograms show comparative expression analyses of Satb1 versus Gm1958 (left) and Gm20098 (right) in various murine T cell subsets. Data are derived from IMMGEN RNA-seq database. (B) Sequence information of Satb1-a and Satb1-b regions. Conserved regions are shown in grey (shaded) and the sequences of sgRNAs used to induce their deletions in mice are highlighted in red font. Sequences flanked with asterisks (*) denote genomic regions deleted in Satb1Venus-Δa and Satb1Venus-Δb mutant mice. (C) Histograms show SATB1-Venus expression in CD4−CD8− DN thymocytes, CD4+CD8+ DP thymocytes, CD24loTCRβhiCD4+CD8− mature SP, and CD24loTCRβhiCD4−CD8+ mature SP thymocytes of Satb1+/Venus, Satb1+/Venus-Δa, and Satb1+/Venus-Δb mutant mice. Numerical values indicate SATB1-Venus MFI. Graph shows quantification of SATB1-Venus MFI in Satb1+/Venus, Satb1+/Venus-Δa, and Satb1+/Venus-Δb thymocytes, summarised from 3 biological experiments. Statistics were calculated by two-way ANNOVA Tukey’s multiple comparisons; *P- < 0.05, **P- < 0.01, ***P- < 0.001. (D) Immunoblot shows SATB1 expression in Satb1+/+and Satb1Δa/Δa TH0, TH1, TH2 cells differentiated in vitro. Data are summarised in the adjacent graphs from four biological experiments. (E) Schematic shows the experimental strategy to knockdown Gm19585 expression. After designing short hairpin (sh) RNA sequences to knockdown exon1-dervied (Gm19585-1-shRNA), exon2-derived (Gm19585-2-shRNA) or exon4-derived (Gm19585-4-shRNA-4) transcripts, pLKO-Gfp-Gm19585-1, pLKO-Gfp-Gm19585-2, or pLKO-Gfp-Gm19585-4 lentiviral vectors were generated and transduced into Satb1+/+ CD4 TH2 cells in vitro.

To determine whether the Satb1-a and Satb1-b regions have any roles in Satb1 gene regulation, we used CRISPR/Cas9-mediated genome editing technology to delete the core sequences of Satb1-a or Satb1-b, either on the Satb1+ or Satb1Venus alleles. Two single-guide RNAs (sgRNAs) for Satb1-a or Satb1-b were selected and co-injected with the Cas9-encoding mRNA into Satb1+/Venus murine zygotes. The F0 founder mice that had deleted Satb1-a or Satb1-b regions were crossed with C57/B6N mice, and F1 founders carrying the Satb1Venus allele, together with the deletion of Satb1-a or Satb1-b, were selected as heterozygous mice with Satb1+/Venus-Δa or Satb1+/Venus-Δb genotype. Among two and three F1 Satb1+/Venus-Δa and Satb1+/Venus-Δb founders, we chose one line as a representative for Satb1Venus-Δa and Satb1Venus-Δb (Sequence information shown in Fig S2B) and were examined for the expression of SATB1-Venus in various T cell subsets. In both Satb1+/Venus-Δa and Satb1+/Venus-Δb mice, there were no significant changes in SATB1-Venus expression in all DN, DP, CD4-SP, and CD8-SP thymocytes (Fig S2C). Analyses of spleens, however, revealed that SATB1-Venus expression levels were significantly reduced in naïve CD4 T cells of Satb1+/Venus-Δa, which were not observed in Satb1+/Venus-Δb mutant mice (Fig 2B). It was also noteworthy that loss of Satb1-a caused a significant reduction in SATB1-Venus expression in CD4 effector-memory populations but had no effects on CD8 naïve and memory T cell populations. These results suggest that Satb1-a is essential for maintaining Satb1 expression in peripheral CD4 T cells with activated/memory phenotypes.

To elucidate whether Satb1-a and Satb1-b have any roles in regulating SATB1 expression in effector CD4 T cell subsets, we activated naïve CD4 T cells of Satb1+/Venus-Δa and Satb1+/Venus-Δb mutant mice in vitro, under TH0, TH1, TH2, TH17 or iTreg polarising conditions. Interestingly, we observed a substantial reduction in SATB1-Venus expression in Satb1+/Venus-Δa CD4 TH0 and TH17 cells, but most strikingly, SATB1-Venus expression was almost lost in Satb1+/Venus-Δa TH2 cells (Fig 2C). Therefore, the Satb1-a enhancer may allow to maintain high levels of SATB1 expression in TH2 cells. On the contrary, we did not detect any decline in SATB1-Venus in Satb1+/Venus-Δb CD4 TH0, TH1, TH2, TH17 or iTreg cells (Fig 2C), thereby discarding any potential roles of Satb1-b in regulating Satb1 expression in T cells. We also confirmed that removal of the Satb1-a region from the Satb1 locus resulted in a significant reduction of SATB1 protein levels in in vitro-differentiated CD4 TH2 and TH0 cells (Fig S2D), which was accompanied with reduction of Satb1 transcripts (Fig 2D). These results confirm that Satb1-a is essential to maintain Satb1 expression under TH2 polarising conditions.

Satb1-a region functions as a genomic enhancer

In addition to the decline of Satb1 expression, we also observed a significant decline in the expression of Gm19585 transcripts in Satb1Δa/Δa CD4 TH2 cells (Fig 2D). Therefore, one would speculate whether the loss of Gm19585 transcripts, which may function as noncoding RNAs, were causative for impaired Satb1 expression. Hence, to mechanistically understand how Satb1-a controls cis expression of Satb1 in CD4 TH2 cells, we examined whether the noncoding Gm19585 RNAs are primarily involved in stabilising Satb1 expression. To this aim, we performed Gm19585 knockdown studies in TH2-polarised CD4 T cells in vitro. Analyses of Gm19585 transcripts deposited on UCSC genome browser showed that there are at least eight alternatively spliced transcripts, as some transcripts were either exon1- or exon2-depleted sequences. Therefore, three shRNA expression plasmids, pLKO-Gfp-Gm19585-1, pLKO-Gfp-Gm19585-2, and pLKO-Gfp-Gm19585-4, that target exon1, exon2, and exon4 of Gm19585 transcript respectively, were generated (Fig S2E). In vitro-activated CD4 T cells were transduced with these shRNAs expressing lentiviral particles and were maintained in TH2 polarising conditions for additional 6 d. shRNA-transduced CD4 TH2 cells defined as CD4+GFP+ were sorted and were subjected to Satb1 transcriptional analyses by qRT–PCR. pLKO-Gfp-Gm19585-2 or pLKO-Gfp-Gm19585-4 shRNAs were able to knockdown Gm19585 transcripts by twofold but had no significant effects on Satb1 transcript levels in CD4 TH2 cells (Fig 3A). These results suggest that the noncoding RNAs from Gm19585 are unlikely to be responsible for maintaining Satb1 expression under TH2 polarising conditions.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3. Satb1-a interacts with Satb1 promoter and is bound by STAT6.

(A) Graphs show qRT–PCR analyses of Gm19585 and Satb1 expression in CD4 TH2 cells transduced with pLKO-GFP-empty, pLKO-Gfp-Gm19585-1, pLKO-Gfp-Gm19585-2 or pLKO-Gfp-Gm19585-4 shRNA-expressing vectors. Data are representative of at least two biological experiments. (B) Heatmaps show contact matrices of chromosome 17 from murine embryonic stem cells (top) versus thymocytes (middle) and CD4 TH2 cells (bottom). The location of Satb1-exon1 and Satb1-a are pinpointed by black arrows. (C) Schematic shows the locations of Satb1-exon1 and Satb1-a loci and primer design for the 3C assay. The positions of primers S1 and S2 overlap Satb1-exon1b and Satb1-exon1a, respectively. The position of anchor primers AS1 and AS2, respectively, align outside and nearby the Satb1-a region. The primer pairs S1-AS1, S1-AS2, S2-AS1, and S2-AS2 were used in combination to capture interactions between Satb1-exon1 and Satb1-a. Data are summarised in the graph below, from two biological experiments. (D) Genome browser ChIP-seq tracks show binding of STAT6 to the Satb1-a in murine CD4 TH2 cells. ATAC-seq signals in colonic Tregs are shown as reference. Two putative STAT6-binding motifs within the Satb1-a region are indicated as grey ovals.

The above finding raised the possibility that Satb1-a functions as an enhancer to control Satb1 gene expression in cis. To investigate this possibility, we used a publicly available three-enzyme Hi-C (3e Hi-C) dataset from murine embryonic stem cells, thymocytes, and CD4 TH2 cells (22, 23), to identify genome wide interactions between Satb1-exon1 (which contain promoters) and Satb1-a region. Interestingly, we found that Satb1-exon1 and Satb1-a are closely located within a topologically associated domain, in both thymocytes and CD4 TH2 cells (Fig 3B). This indicates that a specific cis-genomic interaction between these regions is induced specifically in T cells, through chromatin looping. For further validation, we performed a chromatin capture conformation (3C) assay to confirm whether this chromatin interaction between Satb1-exon1 and Satb1-a specifically occurs in CD4 TH2 cells. For this study, we designed an anchor primer AS2 for capturing Satb1-a and one primer S1 to capture Satb1 exon1b, an alternative Satb1 promoter that has been reported to be highly expressed in CD4 TH2 cells (15) (Fig 3C); the primers AS1 and S2 were used as negative controls. As expected, the use of AS2-S1 primers strongly produced significant amplicon signals in CD4 TH2 cells than in CD4 TH0 and TH1 cells. Therefore, our 3C assay validated that the chromatin interaction between Satb1-exon1 and Satb1-a genomic regions is formed in CD4 TH2 cells, to regulate Satb1 gene expression.

Given that the regulation of chromatin looping is often mediated by a transcription factor, we next sought to identify which TH2-specific transcription factor could be responsible for promoting chromatin looping of Satb1-a to the Satb1 promoters in CD4 TH2 cells. It is already established that the signal transducer and activator of transcription 6 (STAT6) is activated downstream of IL-4 signalling in CD4 TH2 cells and directly regulates Satb1 expression (15). We noticed that there are STAT DNA-binding motifs within the Satb1-a region. Hence, we examined publicly available STAT6 ChIP-seq datasets in murine CD4 TH2 cells (24) and found that Satb1-a was indeed occupied by STAT6 in these cells, suggesting that STAT6 binds and activates the Satb1-a in TH2 cells (Fig 3D). Therefore, we have classified Satb1-a as an enhancer of Satb1 expression in the context of TH2 and henceforth shall be referred as Satb1-Eth2 (enhancer for TH2 cells).

Satb1-Eth2 is essential in repressing IL-5 expression in CD4 TH2 cells in vitro

Having established that Satb1-Eth2 is essential in maintaining Satb1 expression in CD4 TH2 cells, we then investigated the relevance of Satb1-Eth2 on the differentiation and function of CD4 TH2 cells. For this aim, we established Satb1ΔEth2/ΔEth2 mice on C57BL6 background by genome editing. First, it was previously reported that SATB1 functions to promote Gata3 expression in CD4 TH2 cells in vitro (14). Therefore, we first considered whether Satb1-Eth2 had any role in regulating Gata3 expression in CD4 TH2 cells in vitro. Interestingly, although Satb1ΔEth2/ΔEth2 CD4+ TH2 cells showed a significant loss of Satb1 mRNA, there was no significant reduction in Gata3 expression (Fig S3A). These data suggest that low levels of Satb1, via loss of Satb1-Eth2, did not impair Gata3 expression and primary CD4 TH2 differentiation in vitro. Next, it has been reported that SATB1 binds and regulates the expression of the TH2 cytokines IL-4, IL-5, and IL-13 (12), and hence, we examined whether Satb1-Eth2 is essential for the expression of IL-4, IL-5, and IL-13 in Satb1ΔEth2/ΔEth2 CD4 TH2 cells in vitro. PMA and ionomycin stimulation in Satb1ΔEth2/ΔEth2 CD4 TH2 cells revealed no significant differences in the intracellular expression of IL-4, which suggested that Satb1-Eth2 is not required for IL-4 induction in CD4 TH2 cells (Fig S3B). We, however, did find a significant increase in IL4+IL-5+ cells in our Satb1ΔEth2/ΔEth2 CD4 TH2 cultures (Fig 4A). The increase in IL4+IL-5+ cells in our Satb1ΔEth2/ΔEth2 CD4 TH2 cultures was also recaptured upon TCR stimulation (Fig S3C). Further analyses of Il-4, Il-5, and Il-13 transcripts in PMA and ionomycin-stimulated Satb1ΔEth2/ΔEth2 CD4 TH2 cells revealed that only Il-5 transcripts were significantly elevated in Satb1ΔEth2/ΔEth2 CD4 TH2 cells (Fig S3D). Moreover, the elevated expression of Il-5 in Satb1ΔEth2/ΔEth2 CD4 TH2 cells coincided with elevated IL-5 secretion in culture supernatants post PMA and ionomycin stimulation (Fig 4B). These results collectively show that Satb1-Eth2 is crucial to maintain SATB1 levels in CD4 TH2 cells, and to restrain IL-5 expression levels.

Figure S3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure S3. Function of Satb1-Eth2 (Satb1-a) in CD4 TH2 cells in vitro.

(A) Graph shows qRT–PCR analysis of Satb1 and Gata3 expression in in vitro-differentiated TH0 and TH2 cells of Satb1+/+and Satb1ΔEth2/ΔEth2 mice. Data are summarised from three biological experiments. (B) Dot plots show intracellular expression of IFN-γ and IL-4 expression in PMA and ionomycin-stimulated CD4 TH0, TH1, and TH2 cells from Satb1+/+and Satb1ΔEth2/ΔEth2 mice. Data are representative of at least four biological experiments. Graph shows the percentages of IFN-γ+ and IL-4+ cells in PMA- and ionomycin-stimulated Satb1+/+and Satb1ΔEth2/ΔEth2 CD4 TH0, TH1, and TH2 cells. Data are summarised from three biological experiments. (C) Intracellular expression of IL-4 and IL-5 in α-CD3-stimulated Satb1+/+and Satb1ΔEth2/ΔEth2 TH0 and TH2 cells in vitro. Data are representative of two biological experiments. (D) Graphs show qRT–PCR analysis of Il-4, Il-5, and Il-13 mRNA level in unstimulated, PMA, and ionomycin-stimulated CD4 TH0, TH1, and TH2 cells from Satb1+/+and Satb1ΔEth2 /ΔEth2 mice. Data are summarised from three biological experiments. Statistics were calculated by two-way ANNOVA Tukey`s multiple comparisons; *P- < 0.05, **P- < 0.01, ***P- < 0.001.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4. Analyses of Satb1-Eth2 (Satb1-a) in CD4 TH2 differentiation and function in vitro.

(A, B) Intracellular expression of IL-4 and IL-5 (A) and their concentrations in culture supernatants (B) from PMA- and -onomycin-stimulated Satb1+/+ and Satb1ΔEth2/ΔEth2 in vitro-differentiated TH0 and TH2 cells. Data are representative of at least three biological experiments and are summarised in the adjacent graph. Statistics were calculated by unpaired t test; *P- < 0.05, **P- < 0.01, ***P- < 0.001.

Satb1-Eth2 is required to maintain SATB1 expression in CD4 TH2 and ILC2s during TH2 immune responses in vivo

We have demonstrated that Satb1ΔEth2/ΔEth2 CD4 TH2 cells have elevated the expression of IL-5 in vitro. These results are consistent with former studies showing the requirement of SATB1 in suppressing IL-5 in human CD4 TH2 cells (13). IL-5 is the key soluble factor for eosinophil activation and recruitment. At the site of a TH2 immune response, eosinophils release granules containing proinflammatory proteins that cause inflammation and tissue damage. Dysregulation in IL-5 expression and eosinophil activity drive allergic reactions and have clinical implications in the progression of asthmatic responses. We therefore speculated that SATB1 functions to repress CD4 TH2 immune responses via IL-5 suppression and promotes immune resolution. Thus, we next explored the in vivo function of Satb1-Eth2 in controlling type 2 immune responses and used an extract of Alternaria alternata (A.A.) to induce experimental airway inflammation. First, we traced eosinophils infiltration in the bronchial alveolar lavage (BAL) after treating the Satb1+/Venus mice with A.A. on days 4, 7, and 10, and confirmed it to peak from day 7 to 10 (Fig S4A). On day 7, we found a small but significant induction of SATB1-Venus in CD4 TH2 cells (Fig S4B), which is further maintained at day 10. During this study, we also noticed that SATB1-Venus expression was induced in ILC2s (Fig S4B), a subset of lymphocytes responsible for TH2 responses at the early phase of lung inflammation. Previous studies failed to detect Satb1 expression in ILC2s of naïve mice, so we suspected whether ILC2s could only induce Satb1 expression during lung inflammation. Indeed, ex vivo analyses of lung resident ILC2s in naïve mice revealed very little expression of SATB1-Venus expression (Fig S4B). However, in vitro activation of lung-resident ILC2s with IL-33, IL-2, and IL-7 resulted in a significant induction of SATB1-Venus from Satb1Venus/Venus mice (Fig S4C). Importantly, this SATB1-Venus induction was completely absent in in vitro-activated ILC2s from Satb1Venus-ΔEth2/Venus-ΔEth2 mice (Fig S4C). These results indicate that Satb1-Eth2 is indispensable for Satb1 induction in activated ILC2s. We next examined SATB1-Venus expression in lung CD4 TH2 cells and ILC2s with or without the Satb1-Eth2 enhancer 7–10 d post A.A. extract injection. To our expectations, BAL-CD4 T cells and lung-derived CD4 TH2 cells and ILC2s from in A.A-injected Satb1+/Venus-ΔEth2 mice failed to up-regulate SATB1-Venus expression (Fig 5A). Crucially, these results demonstrate the strict requirement of Satb1-Eth2 in inducing SATB1 expression during in vivo TH2 immune responses. To determine whether loss of Satb1-Eth2 function exacerbated lung inflammation in response to A.A., we examined eosinophil infiltration in BAL of A.A.-treated Satb1ΔEth2/ΔEth2 mice versus Satb1+/+ but found no significant increase in eosinophil numbers in A.A.-treated Satb1 ΔEth2/ΔEth2 mice at 7 d post-injection (Fig 5B). In addition, quantification of IL-5 in the BAL supernatants of A.A.-treated Satb1ΔEth2/ΔEth2 mice also revealed no significant increase in IL-5 expression (Fig 5B). Alternatively, we also used the OVA induced lung inflammation model to induce TH2 immune responses in Satb1 ΔEth2/ΔEth2 mice (Fig S4D) but again found no significant increase in eosinophil numbers and in IL-5 concentrations in the BAL of OVA-challenged Satb1ΔEth2/ΔEth2 mice (Fig S4E). Lastly, sorted lung CD4 TH2 cells and ILC2s from A.A.-treated mice, at 10 d post-injection, showed similar frequencies of IL-4+IL-5+ or IL4−IL-5+ subpopulations between Satb1+/+ and Satb1 ΔEth2/ΔEth2 mice (Fig 5C). Hence, our in vivo models could not find a functional relevance of Satb1-Eth2 in restraining both TH2 immune responses and IL-5 expression in CD4 TH2 cells and ILC2s during A.A.-induced lung inflammation. Overall, our results from the A.A.-induced lung inflammation model confirmed that Satb1-Eth2 is required to maintain SATB1 expression in innate and adaptive TH2 lymphocytes during acute lung inflammation, but the physiological relevance of Satb1-Eth2 in regulating TH2 immune responses waits for further investigation.

Figure S4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure S4. Function of Satb1-Eth2 (Satb1-a) in lung-resident CD4 TH2 and ILC2s.

(A) Dot plots show the frequency of eosinophils (CD45+CD11C−SiglecF+) in bronchial alveolar lavage (BAL) of naïve (Day 0) and A.A.-treated Satb1+/Venus mice on days 4, 7, and 10. The graph summarises eosinophil frequencies in BAL of A.A.-treated Satb1+/Venus mice on days 4, 7, and 10. (B) Histograms show expression levels of SATB1-Venus in lung CD4 TH2 (CD45+CD4+GATA3+ST2+, top) and ILC2s (CD45+CD4−GATA3+ST2+, bottom) of naïve (day 0) and A.A.-treated Satb1+/Venus on days 4, 7 and 10. (right) Graphs summarise SATB1-Venus expression levels in lung CD4 TH2 and ILC2 of A.A.-treated Satb1+/Venus mice on days 4, 7, and 10. (C) Histograms show quantification of SATB1-Venus MFI in in vitro-activated ILC2s from Satb1Venus/Venus and Satb1Venus-ΔEth2/Venus-ΔEth2 mutant mice. Data are summarised in the adjacent graph from 3 biological experiments. (D) Schematic shows the experimental plan for OVA-induced TH2 immune responses. Satb1+/+ (n = 5) and Satb1 ΔEth2/ΔEth2 (n = 4) mice were first sensitized to OVA on days 0 and 13 via intraperitoneal injection and then challenged with OVA on days 25, 26, and 27 via intranasal injection. Lung-derived immune cells were all analysed 24 h post final challenge. (E) Graphs show quantification of eosinophils and IL-5 from BAL of OVA-treated mice. Statistics were calculated by unpaired t test; *P- < 0.05, **P- < 0.01, ***P- < 0.001.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5. Satb1-Eth2 functions to regulate SATB1 expression in CD4 TH2 cells and activated ILC2s in vivo.

(A) Histograms show SATB1-Venus expression in BAL CD4+ T cells (CD45+TCRβ+CD4+), in lung CD4 TH2 cells (CD45+TCRβ+CD4+GATA3+ST2+), and in lung ILC2s (CD45+TCRβ−CD4−GATA3+ST2+) of naïve and A.A. treated Satb1+/+, Satb1+/Venus and Satb1+/Venus−ΔEth2 mice on day 7. Data from A.A.-injected mice are a representative of three biological replicates. Graphs summarise SATB1-Venus expression in those cells from A.A. treated Satb1+/+, Satb1+/Venus and Satb1+/Venus−ΔEth2 mice on day 7. (B) Dot plots show frequencies of eosinophils in BAL of naïve (Satb1+/+) and A.A.-treated Satb1+/+ and Satb1ΔEth2/ΔEth2 mice on day 7. Graphs summarise eosinophil numbers and IL-5 concentration in BAL of naïve (Satb1+/+) and A.A.-treated Satb1+/+ and Satb1ΔEth2/ΔEth2 mice on day 7. Data are summarised from at least three biological replicates. (C) Contour plots show the intracellular expression of IL-4 and IL-5 in PMA and ionomycin-stimulated lung CD4 TH2 cells and ILC2s, from A.A.-treated Satb1+/+ and Satb1ΔEth2/ΔEth2 mice on day 10. Graphs show the frequencies of IL-4+IL-5−, IL-4+IL-5+, and IL-4−IL-5− subpopulations in PMA- and ionomycin-stimulated lung CD4 TH2 cells and ILC2s from A.A.-treated Satb1+/+ and Satb1ΔEth2/ΔEth2 mice on day 10. Data are summarised from three biological replicates. Statistics were calculated by unpaired t test; *P- < 0.05, **P- < 0.01, ***P- < 0.001.

Discussion

A recent GWAS study identified a Psoriasis-associated single nucleotide polymorphism in SATB1-AS1, an antisense RNA that is encoded 240 kb upstream of the SATB1 locus and showed a T-cell‒specific interaction with the SATB1 promoter (17). This study was not only the first to reveal the possibility of cis-regulatory transcriptional mechanisms that could regulate Satb1 expression in both human and murine T cells, but it was also the first-link SATB1 to allergy and inflammatory disease.

Previous studies have examined the function of SATB1 in TH2 cells. SATB1 was shown to bind to the murine TH2 locus and promoted Il-5, Il-4, and Il-13 gene expression (12). Another study showed the induction of Satb1 expression upon IL-4 signalling in in vitro-differentiated CD4 TH2 cells (15), in a STAT6-dependent manner. This up-regulation of Satb1 expression in CD4 TH2 cells is associated with altered Satb1 promoter usage from P1 to P2 and P3, but the biological relevance of these promoters in regulating Satb1 expression during T cell development were not fully addressed. Hence, it was unclear how Satb1 expression is specifically controlled during effector CD4 TH cell differentiation, particularly in CD4 TH2 cells. In addition, current studies have not examined whether Satb1 was induced during activation of ILC2s, in part because of low Satb1 expression in steady state ILC2s. Here, we have identified and shown a crucial function of Satb1-Eth2 in regulating Satb1 expression in TH2 cells and ILC2s in vitro and in vivo. Loss of Satb1-Eth2 function significantly impacted on the transcriptional levels of Satb1, specifically in TH2 cells and activated ILC2s. Interestingly, Satb1-Eth2 overlapped with exon 4 of the noncoding gene, Gm19585, and loss of Satb1-Eth2 function was accompanied with the reduction of Gm19585 transcript levels. However, knockdown of Gm19585 transcripts did not affect Satb1 expression in TH2 cells, which suggests that the Gm19585 transcripts have no role in regulating Satb1 gene expression. Rather, chromatin looping between Satb1-Eth2 with Satb1 promoters and STAT6 binding to the Satb1-Eth2 in TH2 cells support the notion that Satb1-Eth2 functioned as an IL-4 inducible TH2-specific enhancer for Satb1 gene expression.

Loss of Satb1-Eth2 expression significantly caused depression of IL-5 in TH2 cells in vitro, which therefore highlights a biological role of Satb1-Eth2 in suppressing IL-5 expression in TH2 cells. Our study is also consistent with what others have observed with Satb1 knockdown assays in human CD4 TH2 cells (13), and therefore indicates that IL-5 de-repression, caused by loss of the Satb1-Eth2 function, is mediated by the reduction of SATB1. Notably, IL-5 is known to play a major role in promoting eosinophil recruitment and promote the progression of allergy or asthma (25). A previous report presumed that SATB1 had a significant role in suppressing lung inflammation, but this was never examined in in vivo models (13). Indeed, we examined the role of Satb1-Eth2 in augmenting lung inflammation in vivo but found that loss of the Satb1-Eth2 had no additive effects on eosinophil infiltration or on IL-5 expression in the BAL of A.A.-treated mice, albeit of low Satb1 expression in both lung TH2 cells and ILC2s. Based on this key information, one would have to truly consider whether SATB1 has any function in controlling TH2 immune responses in vivo. However, these results do not formally exclude the involvement of SATB1 in controlling TH2 immune responses in vivo under different experimental settings, for instance in chronic lung inflammation and in atopic-dermatitis models (26), and thus would merit further investigation.

We also uncovered a partial but significant reduction of SATB1-Venus expression in naïve and effector memory CD4 T cells, caused by loss of Satb1-Eth2 function. Naïve CD4 T cells undergo homeostatic proliferation to maintain their survival in the periphery, which crucially requires both self-MHC ligands and the IL-7 cytokine (27). In addition, effector memory T cells require IL-15 for their survival. Both IL-7 and IL-15 signalling pathways activate the STAT5 transcription factors downstream. It should be noted that most of the STAT transcription factor members recognise the palindromic DNA sequence TTCNNNGAA. It is therefore possible that, in both naïve and effector memory CD4 T cells, STAT5 could bind to Satb1-Eth2 and regulate Satb1 expression. However, because loss of Satb1-Eth2 function had a partial effect on SATB1 expression in naïve CD4 T cells, we believe that Satb1-Eth2 is partially responsive to either IL-7 and/or IL-15. Alternatively, the regulation of Satb1 expression in naïve and memory CD4 T cells could be mediated by other cis- or trans- enhancers, which may require an in-depth analysis.

Overall, the present study identifies and demonstrates the essential role of the cis-regulatory enhancer, Satb1-Eth2, in regulating Satb1 expression, specifically in CD4 TH2 cells and ILC2s, which significantly contributes to our current understandings of how Satb1 expression is controlled during TH2-mediated immune responses. Our study signifies the beginnings of an era of identifying cis- and/or trans-regulatory elements that regulate Satb1 gene expression in T cells and thus provides insightful information on Satb1 function in a T cell context-dependent manner.

Materials and Methods

Mice

The Satb1Venus allele was generated by knock-in insertion of the Venus open reading frame downstream of transcription start site (exon2) of the Satb1 gene. For this aim, a BAC clone B6Ng01-312L13, which included the 5′ area of the Satb1 gene, was purchased from RIKEN BRC. A 13-kb genomic region harboring the ATG start codon was subcloned into the pBlueScript II vector (Stratagene). The DNA fragment harboring partial exon2 sequences and Venus cDNA sequences were generated by overlapping the PCR technique and was used to construct a targeting vector. The targeting vector was transfected into murine M1 ES cells as previously described (28). G418-resistant ES clones were screened for homologous recombination event between the target vector and the Satb1 gene. Appropriate ES clones were then used to generate chimera mice by ES aggregation. F1 founders from the chimera mice carrying the Satb1Venus allele were selected for establishing mouse line and were analysed. Satb1Flx CD4-cre mice were previously described in (7). Satb1+/Venus-Δa (or Satb1Venus-ΔEth2) and Satb1+/Venus-Δb mice were generated by co-injection of Cas9-mRNA with sgRNAs into Satb1+/Venus fertilised eggs that were generated by in vitro fertilisation with sperm from a Satb1Venus/Venus male mouse and Satb1+/+ oocytes. The F0 founders that had deleted Satb1-a or Satb1-b regions were then crossed with C57/B6N mice and F1 founders that deleted Satb1-a or Satb1-b on the Satb1Venus allele were selected as heterozygous mice with the Satb1+/Venus-Δa or Satb1+/Venus-Δb genotype. Among the two and three F1 Satb1+/Venus-Δa and Satb1+/Venus-Δb founders, we chose one line as a representative for Satb1Venus-Δa and Satb1 Venus-Δb and were examined with littermate controls. Similarly, C57/B6N Satb1Δa (Satb1ΔEth2) mice were generated by co-injection of Cas9-mRNA with Satb1-a gRNA into C57Bl/6 fertilised eggs. Sequences for sgRNA are as follows: Satb1Δa sgRNAs 5′-TCAACATCAGAATTTCT-3′ and 5′-CAGTCAACATCAGAATTTCT-3′; and Satb1Δb sgRNAs 5′-ACACACACTGTCTGTTGTGC-3′ and 5′-GCTGCCTGCTTTTACATATC-3′. All mice were maintained at the RIKEN Center for Integrative Medical Sciences. The animal protocol was approved by the Institutional Animal Care and Use Committee of RIKEN Yokohama Branch (2020-026). For all experimental procedures, at least two to three mice were used per experiment and are shown either as a representative or as a summary of these biological experiments.

Immunohistochemistry

50,000 of thymocytes from Satb1+/Venus mice were resuspended in 0.1 ml RPMI-1640 medium (10% FBS) and were mounted on a poly-L-lysine-coated glass slide (REF: 26414; Polysciences, Inc.) and incubated at 37°C for 2 h. The attached cells were gently washed three times in PBS. The cells were fixed with 4% PFA at room temperature for 10 min and then washed three times in PBS. The cells were permeabilized with 70% ethanol and kept at −20°C until analysis. On the day of the analysis, thymocytes were stained with 1/100 of DAPI (ab228549; Abcam) at room temperature, for 30 min, and washed three times in PBS. Cover slips were mounted on top of the thymocytes with Fluoromount-G mounting medium (00-4958-02; Thermo Fisher Scientific) and sealed with Biotium CoverGrip Coverslip Sealant (23005; REF). Confocal microscopic images were obtained with TCS-SP5 (Leica Microsystems) using 40x objective.

Ex vivo cell preparation for flow cytometry

Thymus, spleen, and peripheral lymph nodes (axillary, inguinal, and cervical) were removed from mice at 4–8 wk of age and were mashed through a 70-μm cell strainer in a Petri dish to make single-cell suspensions. For the elimination of red blood cells, splenocytes were treated with ACK lysis buffer (Gibco A1049201, 2.5 ml per spleen) for 3 min and pelleted by centrifugation at 300g at 4°C for 5 min. Supernatants were discarded and cell pellets were resuspended into RPMI-1640 (2% FBS). All cell suspensions were maintained at 4°C for flow cytometry analyses.

For the preparation of IEL, the small intestine was cut into three sections and Peyer Patches were removed. Each section was cut longitudinally and washed twice with HBSS (5% of FCS). Sections were further cut into 0.5-cm pieces and put into a 50 ml conical tube containing 25 ml HBSS (5% of FCS). The tissues were then placed in a bioshaker (TAITEC, BR-43FL) at 37°C, at 250 rpm for 15 min. The supernatant was transferred into a new 50 ml tube, through a metal mesh, and cells were pelleted down by centrifugation at 300g at 4°C for 5 min. The supernatant was discarded. The cell pellet was then resuspended in 8.5 ml of HBSS + 40% Percoll and layered onto 2 ml HBSS+ 70% Percoll in a 15 ml falcon tube. The cells were then centrifuged at 860g for 25 min at room temperature with brakes off. IEL were collected in the interphase between 40% and 70% Percoll and washed twice with HBSS (5% of FCS). IEL were then subjected to flow cytometric analyses.

For the preparation of lung cell suspensions, the lungs were first perfused with 20 ml of PBS through the left ventricle of the heart. Perfused lungs were dissected out and minced up before incubation in RPMI-1640 (2% FBS) + 0.3 mg/ml collagenase IV (C-5138; Sigma-Aldrich) + 0.3 mg/ml of DNase I (043-26773; Wako) at 37°C for 45 min with continuous shaking. To homogenize the samples, the digested lung tissues were mashed in a 70-μm cell strainer in a Petri dish and cell suspensions were then pelleted by centrifugation at 300g at 4°C for 5 min. For the elimination of debris, the cell pellets were resuspended in 10 ml RPMI (2% FBS) + 30% Percoll and separated by centrifugation at 860g at room temperature for 20 min with brakes off. Debris in the upper phase were aspirated off and cell pellets were resuspended in 5 ml RPMI-1640 (2% FBS). The cells were washed and pelleted by centrifugation at 300g at 4°C for 5 min and were then subjected for analysis.

Flow cytometry

Cells were stained with the following fluorophore-conjugated antibodies: B220 (RA3-6B2) CD4 (RM4-5), CD8a (53-6.7), CD11B (M1/70), CD11C (HL3), CD19 (1D3), CD24 (M1/69), CD25 (PC61.5), CD44 (IM7), CD45 (30-F11), CD62L (MEL-14), CD69 (H1.2F3), FOXP3 (FJK-16s), GATA3 (L50-823), Gr1 (RB6-8C5), IL-4 (11B11), IL-5 (TRFK5), IFN-γ (XMG1.2), Ly6C (AL-21), Ly6G (1A8), NK1.1 (PK136) Siglec-F (E13-161.7), ST2 (U29-93) TCRβ (H57-597), TCRγδ (GL3), and Ter119 (TER-119). Murine CD1d dimmer XI (557599; BD-Bioscience) loaded with α-GalCer was used to define iNKT cells. For flow cytometric analyses 1–2 × 106 cells were washed out of complete medium, by centrifugation at 300g at 4°C for 5 min and resuspended in FACS buffer (PBS + 1% FBS and 0.05% NaN3). For the analyses of extracellular marker on live cells, cell suspensions from spleen, pLNs, IEL, and lungs were treated with rat α-mouse CD16/CD32 (BD Biosciences) for 10 min at 4°C. Fluorophore antibodies for extracellular markers were then added and stained for 30 min at 4°C. After incubation, cells were then pelleted by centrifugation at 300g at 4°C for 5 min and resuspended in FACS buffer containing 7AAD/DAPI cell viability dye. For analyses of both extracellular and intracellular markers, cell suspensions of all tissues were stained with fixable live/dead cells (65-0866-18; Ebiosciences) for 10 min before rat α-mouse CD16/CD32 treatment and extracellular marker staining. Using the FOXP3 staining buffer kit (00-5523-00; Ebiosciences), stained cells were then fixed, permeabilised, and stained for intracellular transcription factors as per the manufacturer’s instructions. All cytometry analyses were performed using a FACS CANTO II (BD-Bioscience) and data were analysed using FlowJo (Tree Star) software.

In vitro CD4 T helper differentiation

Naïve CD4 T cells were pooled from spleen and peripheral lymph nodes. The spleen and peripheral lymph nodes (axillary, inguinal, and cervical) were removed from mice at 6–10 wk of age and were mashed through a 70-μm cell strainer in a Petri dish to make single-cell suspensions. Splenocytes were the treated with ACK lysis buffer (Gibco A1049201, 2.5 ml per spleen) for 3 min before pelleting (by centrifugation at 300g at 4°C for 5 min) and resuspension into RPMI-1640 (2% FBS). Naïve CD4 T cells from either sorted by FACS Aria (sorted for CD4+CD25−CD62LhiCD44+) or isolated by using EasySep mouse naïve CD4 T cell isolation kit (ST-19765). Naïve T cells were resuspended in complete medium (KOHJIN BIO DMEM-H [16003016] + 10% FBS) and activated in the presence of 2 μg/ml of anti-CD3 (REF: 553058; BD Pharmingen) and 2 μg/ml of anti-CD28 (REF: 553295; BD Pharmingen) (precoated in a rounded 96-well plate) in TH0 (5 μg/ml of anti-IFNγ [REF: 16-741185; Invitrogen], 5 μg/ml of anti-IL-12/23 [REF: 505308; BioLegend], 5 μg/ml of anti-IL-4 [REF: 554385; BD Pharmingen] and 10 ng/ml of IL-2 [404-ML-010; R&D systems]), TH1 (5 μg/ml of anti-IL-4, 10 ng/ml of IL-2 and 10 ng/ml of IL-12 [419-ML-010; R&D systems]), TH2 (5 μg/ml of anti-IFN- γ, 5 μg/ml of anti-IL-12/23,10 ng/ml of IL-2, and 10 ng/ml of IL-4 [402-ML-020; R&D systems]), TH17 (R and D systems, CDK017, as per the manual’s instructions), and iTreg (5 μg/ml of anti-IFNγ, 5 μg/ml of anti-IL-12/23, 5 μg/ml of anti-IL-4, 2 ng/ml of IL-2 and 3 ng/ml of TGFβ [7666-MB-055; R&D systems]) conditions. Naïve CD4 cells under TH0, TH1, and TH2 conditions were activated were 48 h and were further maintained in TH0/1/2 polarising conditions for another 3–5 d. Naïve CD4 cells under TH17 and iTreg conditions were activated for 72 h and were further maintained in TH17/Treg conditions for another 3–4 d. To confirm CD4 helper differentiation, 1 × 105 of TH0, TH1, TH2 and were stimulated either with 2 μg/ml of anti-CD3 (plate-bound) or with 100 ng/ml of PMA (P8139; Sigma-Aldrich) and 0.5 μg/ml of ionomycin (I0634-1MG; Sigma-Aldrich), in a rounded 96-well plate for 5 h in the presence of 2 μM of monesin (Cay16488-1; Biomol). TH17 cells were stimulated as per the manufacturer’s instructions (CDK017; R and D systems). After stimulation, cells were analysed for intracellular expression of IFN-γ, IL-4, and IL-17 by flow cytometry. The in vitro differentiated Tregs were analysed for the intracellular expression of FOXP3 by flow cytometry. After confirmation of CD4 helper differentiation, Satb1 expression levels were analysed by flow cytometry, immunoblotting, and qRT-PCR. Culture supernatants of PMA and ionomycin-treated CD4 TH0 and CD4 TH2 cells (in the absence of monesin) were also recovered, flash-frozen in N2, and stored in –80°C. The quantification of mIL-4 and mIL-5 cytokines in these supernatants was performed by the Laboratory for Immunogenomics (RIKEN, IMS) via Luminex analysis.

Western blotting

T cells in complete medium were pelleted down by centrifugation at 200g for 5 min at 4°C. Supernatants were aspirated, and the cell pellets were resuspended in 1 ml of ice-cold PBS. Cells were transferred into 1.5 ml Eppendorfs and pelleted down by centrifugation at 200g for 5 min at 4°C. The supernatants were aspirated and pellets were washed in 1 ml of ice-cold PBS. The cells were pelleted down by centrifugation at 200g for 5 min at 4°C and supernatants were aspirated. Cells were lysed in lysis buffer (2% SDS in 50 μM Tris–HCL pH.8, supplemented with EDTA-free protease inhibitor, Roche) at cell concentration of 20 × 106/ml and incubated at 95°C for 15 min. Debris were pelleted down by centrifugation at 13,000g for 10 min at room temperature and protein lysates were transferred into new 1.5 ml Eppendorf tubes. Protein lysates were adjusted with 2X laemil sample buffer (Bio-Rad) + 2-β-mercaptoethanol and boiled at 95°C for 10 min. Each lane was loaded with the equivalent of 100,000 T cells and separated by SDS–PAGE in 10% polyacrylamide gels (e-PAGEL, ATTO, EHR-T10L). Proteins were then transferred onto polyvinylidene difluoride membrane (REF:1704156; Bio-Rad) and blots were blocked with TBST (REF: 12749-21, 0.05% vol/vol; Nacalai tesque) + 5% (wt/vol) of milk for 1 h. All antibodies were used accordingly to the manufacturer`s instructions. Blots were then probed with the following primary antibodies in TBST (0.05% vol/vol) + 5% (wt/vol) of milk, overnight at 4°C: SATB1 (ab109122; Abcam), GAPDH (REF: 2275-PC-100; R and D systems) and SMC1 (ab9262; Abcam). The blots were then wash three times in 15–30 ml TBST (0.05% vol/vol) before incubation in HRP-conjugated secondary antibodies (REF:65-6120; Invitrogen) in TBST (0.05% vol/vol) + 5% (wt/vol) of milk. The blots were then wash three times in 15–30 ml TBST (0.05% vol/vol) and chemiluminescence were quantified on the Amersham Imager 680 (GE Healthcare). All immunoblots shown are representative of at least three biological replicates.

qRT–PCR

1 × 106 of TH0, TH1 or TH2 cells were pelleted down by centrifugation at 200g, at 4°C for 5 min. Supernatants were discarded and cell pellets were immediately resuspended in 750 μl of Trizol (REF: 15596026; Ambion). Samples were snap-frozen in liquid N2. For RNA isolation, cell lysates were thawed, and chloroform added (200 μl per 1 ml of Trizol). RNA samples were extracted as per the manufacturer’s instruction. The samples were vortexed for 20 s and incubated at room temperature for 3 min. The samples were pelleted down by centrifugation at 12,000g at 4°C for15 min. The top aqueous phase containing RNA was transferred into a new tube. RNA samples were purified by using Zymogen RNA clean and concentrator kit (R1017; Zymo research), as per the manufacturer’s instructions. RNA concentrations were obtained on a NanoDrop Spectrophotometer and 1.2 μg of RNA were subjected to cDNA synthesis by using the Super VILO cDNA synthesis kit (REF:11756050; Thermo Fisher Scientific). qRT–PCR was performed using Power up SYBR green master mix (REF: A25742; applied biosystems) on the QuantStudio three Real-Time PCR Systems, accordingly to the manufacturer’s instructions. The following gene expression levels analysed by qRT–PCR are listed in Table 1.

View this table:
  • View inline
  • View popup
Table 1.

List of cDNA primers used for qRT-PCR.

Gm19585 shRNA lentiviral production

The RNAi consortium was used to generate three shRNAs for the knockdown of Gm19585 derived transcripts (see Table 2). shRNA sequences were cloned into the PLKO.gfp cloning vector, which was kindly gifted by Dr. Jun Huh (Harvard University): cloning, transfection, and production of lentiviral particles were performed accordingly to Addgene’s protocol. After screening for shRNA inserts, lentiviruses for individual shRNAs were produced in HEK293T cells plated in six-well plates in antibiotic-free DMEM (Gibco, supplemented with glutamine and 10% FBS). For one transfection assay and using the Fugene HD transfection reagent (REF: E1312; Promega) accordingly to the manufacturer`s instructions, the HEK293T cells were cultured in 10% FBS-containing DMEM and were transfected with 1 μg of shRNA plasmid, 750 ng of psPAX2 packaging plasmid, and 250 ng of pMD2.G envelope plasmids. Packaging and envelope plasmids were obtained from Addgene. Viral supernatants were collected 3 d post transfection and filtered through 0.45 μm low protein-binding filters. Polybrene (Sigma-Aldrich) was added to the viral supernatants at a final concentration of 2 μg/ml, aliquoted, and stored at −80°C.

View this table:
  • View inline
  • View popup
Table 2.

List of shRNA primers used for Gm19585 knockdown study.

Gm19585 knock-down in in vitro TH2 CD4 T cells

CD4 T cells from wild-type mice were isolated from pLNs (axillary, inguinal, and cervical) by using EasySep mouse CD4 T cell isolation kit (ST-19752). Cells were stimulated with 2 μg/ml of immobilised anti-CD3 and anti-CD28 in TH2 polarising conditions (5 μg/ml of anti-IFN- γ, 5 μg/ml of anti-IL-12/23,10 ng/ml of IL-2, and 10 ng/ml of IL-4) for 24 h. After activation, cells were pelleted down by centrifugation at 200g, at room temperature for 5 min and resuspended in 20 μl of prewarmed PBS. The cells were added to 500 μl of thawed viral supernatants and put into 48-well plates. The cells were centrifuged at 660g at 32°C for 1.5 h and were rested in the tissue culture incubator for 1 h 1 volume of 2X TH2 cytokines (5 μg/ml of anti-IFN- γ, 5 μg/ml of anti-IL-12/23,10 ng/ml of IL-2, and 10 ng/ml of IL-4) were then added and the cells were incubated for another 48 h pLKO-Gfp (empty) and pLKO-Gfp-Gm19585 shRNA-transduced cells were monitored and TH2 cytokines were refreshed every second day. CD4+GFP+ cells were then sorted on 6 d post transduction and immediately lysed in Trizol for qRT–PCR analyses.

ATAC-seq analyses

ATAC-Bigwig files from Immgen database (19) were analysed and visualized on the IGV genome browser. Genomic regions of interest were then further analysed on the USCS genome browser for conservation.

3e HiC data analyses

3e HiC datasets from mESCs (GSM1620000) and CD4 TH2 cells (GSM1619998) were downloaded from the Gene Expression Omnibus (GEO) site with the accession numbers GSE66343 (22). HiC datasets from thymocytes (GSM5268043) were downloaded from GEO site with the accession number GSE173476 (23). To examine the chromosome structure and topologically associated domain structures surrounding Satb1 and Satb1-a (exon 4 of Gm19585) loci, reads on chromosome 17 were extracted and realigned on mouse genome mm10 using bowtie2 (version 2.4.6). Mapped reads were collected and converted into.hic format using Juicer tools (version 1.22.01, https://github.com/aidenlab/juicer) and visualized using Juicebox (version 1.6, https://github.com/aidenlab/Juicebox/).

H3K27 acetylation and STAT6 ChIP-seq analyses

Publicly available H3K27Ac ChIP-seq datasets from murine CD4+FOXP3- and CD4+FOXP3+ T cells (8) were used to analyse H3K27Ac peaks at the Satb1-a and Satb1-b loci. The Sequence Read Archive (SRA) files SR5385345 and SRR5385345 were downloaded from SRA website Similarly, STAT6 ChIP-seq in CD4 TH2 cells (24), was used to analyse STAT6 DNA-binding sites at the Satb1-a locus and the SRA file SRA054075 was downloaded from SRA website. Fastdump was used to convert all SRA files into fasta, which were processed for the removal of adaptor sequences and then realigned to mm10 reference genome using Bowtie2. Samtools was then used to sort and convert the SAM files (output files, generated from the genome alignment) into BAM files. BAM files were then indexed using samtools for the generation of bigwig files. The bigwig files were then visualized on the IGV genome browser.

3C assay

Procedures for 3C are described in our previously published article (29). The primer sequences used for the 3C assay qRT-PCR are provided in Table 3.

View this table:
  • View inline
  • View popup
Table 3.

List of primers used for 3C qRT-PCR.

A. alternata induced airway inflammation model

8–12 wk old female mice were anesthetized by isofluorane inhalation, followed by intra-nasal or intra-tracheal injection of A.A. extract (ITEA, 10117., 20 μg per head, in 40 μl of PBS) on Day 0, Day 3, Day 6, and/or Day 9. After 24 h post final challenge, naïve and A.A. treated female mice were euthanized by CO2 inhalation. For cytokine analyses, BAL fluid (BALF) was first collected by intratracheal insertion of a catheter and 1 lavage of 500 μl of HBSS (2% FBS) and transferred into new 1.5 ml Eppendorf tubes. The extracted BALF was immediately pelleted down by centrifugation (300g, 5 min at 4°C) and BALF supernatant were transferred into new 1.5 ml Eppendorf tubes. The pelleted cells were resuspended in 500 μl of HBSS (2% FBS). BALF supernatants were flash-frozen in liquid N2 and stored in −80°C. Additional two lavages of 500 μl of HBSS (2% FBS) were used to further collect the BALF of naïve and AA challenged mice and were combined with BALF cells from the first lavage, totalling to a final volume of 1.5 ml. BALF cells were kept on 4°C for flow cytometric analyses. Lung cell suspensions from mice were prepared as outlined above. After Percoll gradient centrifugation, CD4 TH2 cells (CD45+Thy1.2+CD4+ST2+) and ILC2s (CD45+Thy1.2+CD4−ST2+) were sorted using FACS Aria. 10,000 CD4 TH2 cells and ILC2s were activated with PMA (100 ng/ml) and ionomycin (0.5 μg/ml) in a rounded 96-well plate for 4 h in the presence of Monesin and were then subjected to flow cytometry analyses for intracellular cytokines levels. The quantification of the mIL-5 cytokine in BAL supernatants of naïve and A.A.-treated mice were also performed by the Laboratory for Immunogenomics (RIKEN, IMS) via Luminex analysis.

OVA-induced airway inflammation model

8–12-wk-old male and female mice were first immunized with intraperitoneal administration of 50 μg of OVA (in 100 μl PBS) + 100 μl of Alum (Imject Alum Adjuvant, Thermo Fisher Scientific), on Day 0 and Day 13. On days 27, 28, and 29, the mice were anesthetized by isoflurane inhalation, followed by intranasal injection of 50 μg of OVA (in 40 μl PBS). After 24 h post final challenge, BAL and lungs of naïve and OVA-challenged mice were processed and analysed using the same methods for A.A-induced airway inflammation model.

In vitro activation of ILC2s

Lung cell suspensions from mice were prepared as outlined above. After Percoll gradient centrifugation, 5,000 ILC2s (CD45+Lin−CD3ε−Thy1.2+) were sorted into each well of 96 well-plate. Sorted ILC2s were cultured in 100 μl of complete medium (KOHJIN BIO DMEM-H [16003016] + 10% FBS) in the presence of mIL-33 (10 ng/ml), mIL-2 (10 ng/ml) and mIL-7 (10 ng/ml) for 4 d.

Statistical analysis

Student T tests and two-way ANOVA with post hoc Bonferroni tests were performed using GraphPad Prism software: ***P < 0.001, **P < 0.01, and *P < 0.05.

Acknowledgements

We thank Noriko Yoza for her continuous help with cell sorting and Yusuke Iizuka and RIKEN IMS Animal group for ES aggregation and genome editing. We`d also like express our gratitude to Chizuko Miyamoto, Yuria Taniguchi, Miho Mochizuki, and Natsuki Takeno for their assistance with experiments and genotyping. This work was supported by the MITSUBISHI FOUNDATION (201910029) and Ministry of Education, Culture, Sports, Science and Technology Grants-in-Aid for Scientific Research on Innovative Areas “Replication of Non-Genomic Codes” (JP19H05747) (I Taniuchi) and by support funding from RIKEN’s Gender Equality Program (A Nomura).

Author Contributions

  • A Nomura: formal analysis, investigation, and writing—original draft, review, and editing.

  • T Kobayashi: formal analysis.

  • W Seo: formal analysis.

  • M Ohno-Oishi: formal analysis.

  • K Kakugawa: formal analysis.

  • S Muroi: methodology.

  • H Yoshida: resources, formal analysis, and methodology.

  • TA Endo: resources, software, and formal analysis.

  • K Moro: formal analysis.

  • I Taniuchi: conceptualization, supervision, and writing—original draft, review, and editing.

Conflict of Interest Statement

The authors declare that they have no conflict of interest.

  • Received January 3, 2023.
  • Revision received May 8, 2023.
  • Accepted May 8, 2023.
  • © 2023 Nomura et al.
Creative Commons logoCreative Commons logohttps://creativecommons.org/licenses/by/4.0/

This article is available under a Creative Commons License (Attribution 4.0 International, as described at https://creativecommons.org/licenses/by/4.0/).

References

  1. 1.↵
    1. Bode J,
    2. Kohwi Y,
    3. Dickinson L,
    4. Joh T,
    5. Klehr D,
    6. Mielke C,
    7. Kohwi-Shigematsu T
    (1992) Biological significance of unwinding capability of nuclear matrix-associating DNAs. Science 255: 195–197. doi:10.1126/science.1553545
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Kohwi-Shigematsu T,
    2. Kohwi Y
    (1990) Torsional stress stabilizes extended base unpairing in suppressor sites flanking immunoglobulin heavy chain enhancer. Biochemistry 29: 9551–9560. doi:10.1021/bi00493a009
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Yasui D,
    2. Miyano M,
    3. Cai S,
    4. Varga-Weisz P,
    5. Kohwi-Shigematsu T
    (2002) SATB1 targets chromatin remodelling to regulate genes over long distances. Nature 419: 641–645. doi:10.1038/nature01084
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Balamotis MA,
    2. Tamberg N,
    3. Woo YJ,
    4. Li J,
    5. Davy B,
    6. Kohwi-Shigematsu T,
    7. Kohwi Y
    (2012) Satb1 ablation alters temporal expression of immediate early genes and reduces dendritic spine density during postnatal brain development. Mol Cell Biol 32: 333–347. doi:10.1128/mcb.05917-11
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Riessland M,
    2. Kolisnyk B,
    3. Kim TW,
    4. Cheng J,
    5. Ni J,
    6. Pearson JA,
    7. Park EJ,
    8. Dam K,
    9. Acehan D,
    10. Ramos-Espiritu LS, et al.
    (2019) Loss of SATB1 induces p21-dependent cellular senescence in post-mitotic dopaminergic neurons. Cell Stem Cell 25: 514–530.e8. doi:10.1016/j.stem.2019.08.013
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Alvarez JD,
    2. Yasui DH,
    3. Niida H,
    4. Joh T,
    5. Loh DY,
    6. Kohwi-Shigematsu T
    (2000) The MAR-binding protein SATB1 orchestrates temporal and spatial expression of multiple genes during T-cell development. Genes Dev 14: 521–535. doi:10.1101/gad.14.5.521
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. Kakugawa K,
    2. Kojo S,
    3. Tanaka H,
    4. Seo W,
    5. Endo TA,
    6. Kitagawa Y,
    7. Muroi S,
    8. Tenno M,
    9. Yasmin N,
    10. Kohwi Y, et al.
    (2017) Essential roles of SATB1 in specifying T lymphocyte subsets. Cell Rep 19: 1176–1188. doi:10.1016/j.celrep.2017.04.038
    OpenUrlCrossRef
  8. 8.↵
    1. Kitagawa Y,
    2. Ohkura N,
    3. Kidani Y,
    4. Vandenbon A,
    5. Hirota K,
    6. Kawakami R,
    7. Yasuda K,
    8. Motooka D,
    9. Nakamura S,
    10. Kondo M, et al.
    (2017) Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat Immunol 18: 173–183. doi:10.1038/ni.3646
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Gottimukkala KP,
    2. Jangid R,
    3. Patta I,
    4. Sultana DA,
    5. Sharma A,
    6. Misra-Sen J,
    7. Galande S
    (2016) Regulation of SATB1 during thymocyte development by TCR signaling. Mol Immunol 77: 34–43. doi:10.1016/j.molimm.2016.07.005
    OpenUrlCrossRef
  10. 10.↵
    1. Stephen TL,
    2. Payne KK,
    3. Chaurio RA,
    4. Allegrezza MJ,
    5. Zhu H,
    6. Perez-Sanz J,
    7. Perales-Puchalt A,
    8. Nguyen JM,
    9. Vara-Ailor AE,
    10. Eruslanov EB, et al.
    (2017) SATB1 expression governs epigenetic repression of PD-1 in tumor-reactive T cells. Immunity 46: 51–64. doi:10.1016/j.immuni.2016.12.015
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Yasuda K,
    2. Kitagawa Y,
    3. Kawakami R,
    4. Isaka Y,
    5. Watanabe H,
    6. Kondoh G,
    7. Kohwi-Shigematsu T,
    8. Sakaguchi S,
    9. Hirota K
    (2019) Satb1 regulates the effector program of encephalitogenic tissue Th17 cells in chronic inflammation. Nat Commun 10: 549. doi:10.1038/s41467-019-08404-w
    OpenUrlCrossRef
  12. 12.↵
    1. Cai S,
    2. Lee CC,
    3. Kohwi-Shigematsu T
    (2006) SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nat Genet 38: 1278–1288. doi:10.1038/ng1913
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Ahlfors H,
    2. Limaye A,
    3. Elo LL,
    4. Tuomela S,
    5. Burute M,
    6. Gottimukkala KVP,
    7. Notani D,
    8. Rasool O,
    9. Galande S,
    10. Lahesmaa R
    (2010) SATB1 dictates expression of multiple genes including IL-5 involved in human T helper cell differentiation. Blood 116: 1443–1453. doi:10.1182/blood-2009-11-252205
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Notani D,
    2. Gottimukkala KP,
    3. Jayani RS,
    4. Limaye AS,
    5. Damle MV,
    6. Mehta S,
    7. Purbey PK,
    8. Joseph J,
    9. Galande S
    (2010) Global regulator SATB1 recruits β-catenin and regulates TH2 differentiation in wnt-dependent manner. PLoS Biol 8: e1000296. doi:10.1371/journal.pbio.1000296
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Khare SP,
    2. Shetty A,
    3. Biradar R,
    4. Patta I,
    5. Chen ZJ,
    6. Sathe AV,
    7. Reddy PC,
    8. Lahesmaa R,
    9. Galande S
    (2019) NF-κB signaling and IL-4 signaling regulate SATB1 expression via alternative promoter usage during Th2 differentiation. Front Immunol 10: 667. doi:10.3389/fimmu.2019.00667
    OpenUrlCrossRef
  16. 16.↵
    1. Patta I,
    2. Madhok A,
    3. Khare S,
    4. Gottimukkala KP,
    5. Verma A,
    6. Giri S,
    7. Dandewad V,
    8. Seshadri V,
    9. Lal G,
    10. Misra-Sen J, et al.
    (2020) Dynamic regulation of chromatin organizer SATB1 via TCR-induced alternative promoter switch during T-cell development. Nucleic Acids Res 48: 5873–5890. doi:10.1093/nar/gkaa321
    OpenUrlCrossRef
  17. 17.↵
    1. Shi C,
    2. Ray-Jones H,
    3. Ding J,
    4. Duffus K,
    5. Fu Y,
    6. Gaddi VP,
    7. Gough O,
    8. Hankinson J,
    9. Martin P,
    10. McGovern A, et al.
    (2021) Chromatin looping links target genes with genetic risk loci for dermatological traits. J Invest Dermatol 141: 1975–1984. doi:10.1016/j.jid.2021.01.015
    OpenUrlCrossRef
  18. 18.↵
    1. Cai S,
    2. Han H-J,
    3. Kohwi-Shigematsu T
    (2003) Tissue-specific nuclear architecture and gene expession regulated by SATB1. Nat Genet 34: 42–51. doi:10.1038/ng1146
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Yoshida H,
    2. Lareau CA,
    3. Ramirez RN,
    4. Rose SA,
    5. Maier B,
    6. Wroblewska A,
    7. Desland F,
    8. Chudnovskiy A,
    9. Mortha A,
    10. Dominguez C, et al.
    (2019) The cis-regulatory atlas of the mouse immune system. Cell 176: 897–912.e20. doi:10.1016/j.cell.2018.12.036
    OpenUrlCrossRef
  20. 20.↵
    1. Sorge S,
    2. Ha N,
    3. Polychronidou M,
    4. Friedrich J,
    5. Bezdan D,
    6. Kaspar P,
    7. Schaefer MH,
    8. Ossowski S,
    9. Henz SR,
    10. Mundorf J, et al.
    (2012) The cis-regulatory code of Hox function in Drosophila. EMBO J 31: 3323–3333. doi:10.1038/emboj.2012.179
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Gorkin DU,
    2. Barozzi I,
    3. Zhao Y,
    4. Zhang Y,
    5. Huang H,
    6. Lee AY,
    7. Li B,
    8. Chiou J,
    9. Wildberg A,
    10. Ding B, et al.
    (2020) An atlas of dynamic chromatin landscapes in mouse fetal development. Nature 583: 744–751. doi:10.1038/s41586-020-2093-3
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Ren G,
    2. Jin W,
    3. Cui K,
    4. Rodrigez J,
    5. Hu G,
    6. Zhang Z,
    7. Larson DR,
    8. Zhao K
    (2017) CTCF-mediated enhancer-promoter interaction is a critical regulator of cell-to-cell variation of gene expression. Mol Cell 67: 1049–1058.e6. doi:10.1016/j.molcel.2017.08.026
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Zelenka T,
    2. Klonizakis A,
    3. Tsoukatou D,
    4. Papamatheakis D-A,
    5. Franzenburg S,
    6. Tzerpos P,
    7. Tzonevrakis I-R,
    8. Papadogkonas G,
    9. Kapsetaki M,
    10. Nikolaou C, et al.
    (2022) The 3D enhancer network of the developing T cell genome is shaped by SATB1. Nat Commun 13: 6954. doi:10.1038/s41467-022-34345-y
    OpenUrlCrossRef
  24. 24.↵
    1. Wei L,
    2. Vahedi G,
    3. Sun H-W,
    4. Watford WT,
    5. Takatori H,
    6. Ramos HL,
    7. Takahashi H,
    8. Liang J,
    9. Gutierrez-Cruz G,
    10. Zang C, et al.
    (2010) Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity 32: 840–851. doi:10.1016/j.immuni.2010.06.003
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Rothenberg ME,
    2. Hogan SP
    (2006) The eosinophil. Annu Rev Immunol 24: 147–174. doi:10.1146/annurev.immunol.24.021605.090720
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Oyoshi MK,
    2. Larson RP,
    3. Ziegler SF,
    4. Geha RS
    (2010) Mechanical injury polarizes skin dendritic cells to elicit a TH2 response by inducing cutaneous thymic stromal lymphopoietin expression. J Allergy Clin Immunol 126: 976–984.e5. doi:10.1016/j.jaci.2010.08.041
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Boyman O,
    2. Purton JF,
    3. Surh CD,
    4. Sprent J
    (2007) Cytokines and T-cell homeostasis. Curr Opin Immunol 19: 320–326. doi:10.1016/j.coi.2007.04.015
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Muroi S,
    2. Naoe Y,
    3. Miyamoto C,
    4. Akiyama K,
    5. Ikawa T,
    6. Masuda K,
    7. Kawamoto H,
    8. Taniuchi I
    (2008) Cascading suppression of transcriptional silencers by ThPOK seals helper T cell fate. Nat Immunol 9: 1113–1121. doi:10.1038/ni.1650
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Seo W,
    2. Shimizu K,
    3. Kojo S,
    4. Okeke A,
    5. Kohwi-Shigematsu T,
    6. Fujii S-i,
    7. Taniuchi I
    (2020) Runx-mediated regulation of CCL5 via antagonizing two enhancers influences immune cell function and anti-tumor immunity. Nat Commun 11: 1562. doi:10.1038/s41467-020-15375-w
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Download PDF
Email Article

Thank you for your interest in spreading the word on Life Science Alliance.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Identification of a novel enhancer essential for Satb1 expression in TH2 cells and activated ILC2s
(Your Name) has sent you a message from Life Science Alliance
(Your Name) thought you would like to see the Life Science Alliance web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
A novel TH2 specific enhancer for Satb1 expression
Aneela Nomura, Tetsuro Kobayashi, Wooseok Seo, Michiko Ohno-Oishi, Kiyokazu Kakugawa, Sawako Muroi, Hideyuki Yoshida, Takaho A Endo, Kazuyo Moro, Ichiro Taniuchi
Life Science Alliance May 2023, 6 (8) e202301897; DOI: 10.26508/lsa.202301897

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
A novel TH2 specific enhancer for Satb1 expression
Aneela Nomura, Tetsuro Kobayashi, Wooseok Seo, Michiko Ohno-Oishi, Kiyokazu Kakugawa, Sawako Muroi, Hideyuki Yoshida, Takaho A Endo, Kazuyo Moro, Ichiro Taniuchi
Life Science Alliance May 2023, 6 (8) e202301897; DOI: 10.26508/lsa.202301897
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
Issue Cover

In this Issue

Volume 6, No. 8
August 2023
  • Table of Contents
  • Cover (PDF)
  • About the Cover
  • Masthead (PDF)
Advertisement

Jump to section

  • Article
    • Abstract
    • Introduction
    • Results
    • Discussion
    • Materials and Methods
    • Acknowledgements
    • References
  • Figures & Data
  • Info
  • Metrics
  • Reviewer Comments
  • PDF

Subjects

  • Immunology

Related Articles

  • No related articles found.

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • HU modulates thiol–disulfide homeostasis
  • GSK3A, a proviral host factor for HAdV-B7 replication
  • PSME3 regulates myogenesis
Show more Research Article

Similar Articles

EMBO Press LogoRockefeller University Press LogoCold Spring Harbor Logo

Content

  • Home
  • Newest Articles
  • Current Issue
  • Archive
  • Subject Collections

For Authors

  • Submit a Manuscript
  • Author Guidelines
  • License, copyright, Fee

Other Services

  • Alerts
  • Bluesky
  • X/Twitter
  • RSS Feeds

More Information

  • Editors & Staff
  • Reviewer Guidelines
  • Feedback
  • Licensing and Reuse
  • Privacy Policy

ISSN: 2575-1077
© 2025 Life Science Alliance LLC

Life Science Alliance is registered as a trademark in the U.S. Patent and Trade Mark Office and in the European Union Intellectual Property Office.