10
E-protein regulatory network links TCR signaling to effector Treg cell differentiation Xiaojuan Han a,b , Huarong Huang a,b , Ping Gao a,b , Qi Zhang a , Xinyuan Liu a,b , Baoqian Jia a , Warren Strober c , Baidong Hou d , Xuyu Zhou a,e , George Fu Gao a,e,f,g , and Fuping Zhang a,e,1 a Chinese Academy of Sciences (CAS) Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101 Beijing, China; b College of Life Science, University of Chinese Academy of Sciences, 100049 Beijing, China; c Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892; d CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China; e Savaid Medical School, University of Chinese Academy of Sciences, 101408 Beijing, China; f National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 102206 Beijing, China; and g Research Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, 100101 Beijing, China Edited by Ananda W. Goldrath, University of California San Diego, La Jolla, CA, and accepted by Editorial Board Member Philippa Marrack January 17, 2019 (received for review January 11, 2018) T cell antigen receptor (TCR) signaling is essential for the differenti- ation and maintenance of effector regulatory T (Treg) cells. However, the contribution of individual TCR-dependent genes in Treg cells to the maintenance of immunotolerance remains largely unknown. Here we demonstrate that Treg cells lacking E protein undergo further differentiation into effector cells that exhibit high expression of effector Treg signature genes, including IRF4, ICOS, CD103, KLRG-1, and RORγt. E protein-deficient Treg cells displayed increased stability and enhanced suppressive capacity. Transcriptome and ChIP-seq anal- yses revealed that E protein directly regulates a large proportion of the genes that are specific to effector Treg cell activation, and impor- tantly, most of the up-regulated genes in E protein-deficient Treg cells are also TCR dependent; this indicates that E proteins comprise a critical gene regulatory network that links TCR signaling to the control of effector Treg cell differentiation and function. E protein | effector Treg cell | TCR signaling | gene regulatory network | differentiation and function I t is well established that regulatory T cells (Treg cells) play a pivotal role in maintaining immune tolerance and thus in preventing autoimmunity and chronic inflammation (1). These cells can be subdivided into different subsets with somewhat unique characteristics. One type of subset classification is based on site of development and consists of a natural Treg subset that develops in the thymus (called nTreg or tTreg cells) and a pe- ripheral Treg cell subset that develops in the periphery (called iTreg or pTreg cells) (2). Another type of subset classification is based on function. This classification recognizes a centralTreg population that is equivalent to naive CD4 + T cells with respect to various markers and circulatory patterns and that differs from one or more effectorTreg cell populations that exhibit en- hanced regulatory function and migration through nonlymphoid tissues (35). Such effector Treg cells make up a minor fraction of Treg cells in the circulation and secondary lymphoid or- gans and are referred to as activatedTreg cells in some studies because they share phenotypic features with activated conventional T cells. As such, they are variously defined as CD62L low CCR7 low CD44 hi KLRG1 + cells, ICOS + IRF4 + CD103 + cells, or CD45RA low CD25 hi cells, depending on the study. Importantly, Treg cells comprising the effector subpopulation defined above are thought to have encountered antigens and have undergone T cell antigen receptor (TCR) stimulation more recently than central Treg cells (69). This coincides with the fact that TCR signaling in differentiated Treg cells is essential to Treg cell homeostasis, suppressor function, and signature gene expression, especially to those signature genes that produce Treg cell effector molecules (1012). It should be noted, however, that the underlying molecular mechanisms downstream of TCR sig- naling that account for the above Treg cell characteristics are still poorly understood. In addition, the contributions of individual TCR-dependent genes in Treg cells to maintain immunotol- erance in the steady state and that restrain immune responses directed against commensal bacteria, environmental antigens, and pathogens, also require further elucidation. One important way in which effector Treg cell differentiation may be regulated is via the activity of E proteins, a family of transcriptional activators or repressors that bind to E-box sites at transcriptional sites and thus determine gene expression (13). Previous studies have shown that down-regulation of E-protein activity by TCR signaling accompanies the development of Foxp3 + regulatory T cells (14, 15), whereas increased E-protein activity accompanies Treg cell dysfunction (16). These observations sug- gested that recognition of self-antigen in matured Treg cells may further down-regulate E-protein activity, thus driving Treg cells to differentiate toward particular specificities that may potentiate their suppressive capacity during immunological challenges. Never- theless, whether E protein regulates matured Treg cell activation or whether E protein directly regulates certain activated Treg signature gene expression has never been demonstrated. To directly address the requirement of E-protein activity in the regulation of Treg cell homeostasis, we studied Treg cells in E2A fl/fl HEB fl/fl Foxp3-Cre-GFP mice with E-protein deletion spe- cific to Treg cells. Notably, we found that deficiency of E protein in differentiated Treg cells results in up-regulated expression of Significance Effector Treg cells comprise the subset of the Treg cell pop- ulation that exhibits enhanced regulatory function. Whereas the induction and maintenance of this subset are known to depend on TCR signaling, the underlying molecular mecha- nisms downstream of such signaling and the contributions of individual TCR-dependent genes to effector Treg cell genera- tion are still poorly understood. In the studies described here differentiated Treg cells in which E-protein (E2A/HEB) expression has been deleted were utilized to demonstrate that E proteins are transcriptional suppressors of a large number of genes as- sociated with effector Treg cell differentiation, localization, function, and proliferation. Thus, this finding indicates that continuous TCR signals modulating E-protein activity is a major mechanism underlying Treg cell acquisition of effector functions. Author contributions: F.Z. designed research; X.H., H.H., P.G., Q.Z., X.L., and B.J. performed research; B.H., X.Z., and G.F.G. contributed new reagents/analytic tools; X.H., B.H., X.Z., and G.F.G. analyzed data; and X.H., W.S., and F.Z. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. A.W.G. is a guest editor invited by the Editorial Board. Published under the PNAS license. 1 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1800494116/-/DCSupplemental. Published online February 15, 2019. www.pnas.org/cgi/doi/10.1073/pnas.1800494116 PNAS | March 5, 2019 | vol. 116 | no. 10 | 44714480 IMMUNOLOGY AND INFLAMMATION Downloaded by guest on June 4, 2021

E-protein regulatory network links TCR signaling to ... › content › pnas › 116 › 10 › 4471.full.pdfGFP-hCre mice (designated hereafter as E2Afl/flHEBfl/fl mice) exhibited

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • E-protein regulatory network links TCR signaling toeffector Treg cell differentiationXiaojuan Hana,b, Huarong Huanga,b, Ping Gaoa,b, Qi Zhanga, Xinyuan Liua,b, Baoqian Jiaa, Warren Stroberc,Baidong Houd, Xuyu Zhoua,e, George Fu Gaoa,e,f,g, and Fuping Zhanga,e,1

    aChinese Academy of Sciences (CAS) Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences,100101 Beijing, China; bCollege of Life Science, University of Chinese Academy of Sciences, 100049 Beijing, China; cMucosal Immunity Section, Laboratory ofHost Defenses, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892; dCAS Key Laboratory of Pathogenic Microbiology andImmunology, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China; eSavaid Medical School, University of Chinese Academy ofSciences, 101408 Beijing, China; fNational Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 102206Beijing, China; and gResearch Network of Immunity and Health, Beijing Institutes of Life Science, Chinese Academy of Sciences, 100101 Beijing, China

    Edited by Ananda W. Goldrath, University of California San Diego, La Jolla, CA, and accepted by Editorial Board Member Philippa Marrack January 17, 2019(received for review January 11, 2018)

    T cell antigen receptor (TCR) signaling is essential for the differenti-ation and maintenance of effector regulatory T (Treg) cells. However,the contribution of individual TCR-dependent genes in Treg cells tothe maintenance of immunotolerance remains largely unknown. Herewe demonstrate that Treg cells lacking E protein undergo furtherdifferentiation into effector cells that exhibit high expression ofeffector Treg signature genes, including IRF4, ICOS, CD103, KLRG-1,and RORγt. E protein-deficient Treg cells displayed increased stabilityand enhanced suppressive capacity. Transcriptome and ChIP-seq anal-yses revealed that E protein directly regulates a large proportion ofthe genes that are specific to effector Treg cell activation, and impor-tantly, most of the up-regulated genes in E protein-deficient Treg cellsare also TCR dependent; this indicates that E proteins comprise acritical gene regulatory network that links TCR signaling to the controlof effector Treg cell differentiation and function.

    E protein | effector Treg cell | TCR signaling | gene regulatory network |differentiation and function

    It is well established that regulatory T cells (Treg cells) play apivotal role in maintaining immune tolerance and thus inpreventing autoimmunity and chronic inflammation (1). Thesecells can be subdivided into different subsets with somewhatunique characteristics. One type of subset classification is basedon site of development and consists of a natural Treg subset thatdevelops in the thymus (called nTreg or tTreg cells) and a pe-ripheral Treg cell subset that develops in the periphery (callediTreg or pTreg cells) (2). Another type of subset classification isbased on function. This classification recognizes a “central” Tregpopulation that is equivalent to naive CD4+ T cells with respectto various markers and circulatory patterns and that differs fromone or more “effector” Treg cell populations that exhibit en-hanced regulatory function and migration through nonlymphoidtissues (3–5). Such effector Treg cells make up a minor fractionof Treg cells in the circulation and secondary lymphoid or-gans and are referred to as “activated” Treg cells in somestudies because they share phenotypic features with activatedconventional T cells. As such, they are variously defined asCD62LlowCCR7lowCD44hiKLRG1+ cells, ICOS+IRF4+CD103+

    cells, or CD45RAlowCD25hi cells, depending on the study.Importantly, Treg cells comprising the effector subpopulation

    defined above are thought to have encountered antigens andhave undergone T cell antigen receptor (TCR) stimulation morerecently than central Treg cells (6–9). This coincides with the factthat TCR signaling in differentiated Treg cells is essential toTreg cell homeostasis, suppressor function, and signature geneexpression, especially to those signature genes that produce Tregcell effector molecules (10–12). It should be noted, however, thatthe underlying molecular mechanisms downstream of TCR sig-naling that account for the above Treg cell characteristics are stillpoorly understood. In addition, the contributions of individualTCR-dependent genes in Treg cells to maintain immunotol-

    erance in the steady state and that restrain immune responsesdirected against commensal bacteria, environmental antigens,and pathogens, also require further elucidation.One important way in which effector Treg cell differentiation

    may be regulated is via the activity of E proteins, a family oftranscriptional activators or repressors that bind to E-box sites attranscriptional sites and thus determine gene expression (13).Previous studies have shown that down-regulation of E-proteinactivity by TCR signaling accompanies the development of Foxp3+

    regulatory T cells (14, 15), whereas increased E-protein activityaccompanies Treg cell dysfunction (16). These observations sug-gested that recognition of self-antigen in matured Treg cells mayfurther down-regulate E-protein activity, thus driving Treg cellsto differentiate toward particular specificities that may potentiatetheir suppressive capacity during immunological challenges. Never-theless, whether E protein regulates matured Treg cell activationor whether E protein directly regulates certain activated Tregsignature gene expression has never been demonstrated.To directly address the requirement of E-protein activity in the

    regulation of Treg cell homeostasis, we studied Treg cells inE2Afl/flHEBfl/flFoxp3-Cre-GFP mice with E-protein deletion spe-cific to Treg cells. Notably, we found that deficiency of E proteinin differentiated Treg cells results in up-regulated expression of

    Significance

    Effector Treg cells comprise the subset of the Treg cell pop-ulation that exhibits enhanced regulatory function. Whereasthe induction and maintenance of this subset are known todepend on TCR signaling, the underlying molecular mecha-nisms downstream of such signaling and the contributions ofindividual TCR-dependent genes to effector Treg cell genera-tion are still poorly understood. In the studies described heredifferentiated Treg cells in which E-protein (E2A/HEB) expressionhas been deleted were utilized to demonstrate that E proteinsare transcriptional suppressors of a large number of genes as-sociated with effector Treg cell differentiation, localization,function, and proliferation. Thus, this finding indicates thatcontinuous TCR signals modulating E-protein activity is a majormechanism underlying Treg cell acquisition of effector functions.

    Author contributions: F.Z. designed research; X.H., H.H., P.G., Q.Z., X.L., and B.J.performed research; B.H., X.Z., and G.F.G. contributed new reagents/analytic tools;X.H., B.H., X.Z., and G.F.G. analyzed data; and X.H., W.S., and F.Z. wrote the paper.

    The authors declare no conflict of interest.

    This article is a PNAS Direct Submission. A.W.G. is a guest editor invited by the EditorialBoard.

    Published under the PNAS license.1To whom correspondence should be addressed. Email: [email protected].

    This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplemental.

    Published online February 15, 2019.

    www.pnas.org/cgi/doi/10.1073/pnas.1800494116 PNAS | March 5, 2019 | vol. 116 | no. 10 | 4471–4480

    IMMUNOLO

    GYAND

    INFLAMMATION

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    http://crossmark.crossref.org/dialog/?doi=10.1073/pnas.1800494116&domain=pdfhttps://www.pnas.org/site/aboutpnas/licenses.xhtmlmailto:[email protected]://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/cgi/doi/10.1073/pnas.1800494116

  • various effector Treg cell markers such as CD103, ICOS, IRF4,KLRG1, RORγt, as well as enhanced Treg cell stability andsuppressive capacity. Further study demonstrated that E-proteinactivity was essential for Treg cells to maintain the expression of alarge proportion of genes found to be expressed almost exclusivelyin effector Treg cells. In addition, gene expression profile analysisindicated that a substantial amount of effector Treg signature genesregulated by E protein are also regulated by TCR. E proteins reg-ulate effector Treg cells by directly binding to a large number ofgenes that associated with effector Treg cell differentiation, locali-zation, function, and proliferation. Thus, our findings demonstratedthat downstream of TCR signaling, E-protein activity plays an es-sential role in the regulation of Treg cell activation.

    ResultsTreg Cell Homeostasis Is Regulated by Specific Deletion of E Proteinsin Mature Treg Cells. To explore the specific role of E proteins inmature Treg cells, we crossed E2Afl/flHEBfl/fl mice with mice thathave a bacterial artificial chromosome transgene encoding greenfluorescent protein (GFP) and humanized Cre recombinase(hCre) under the control of the Foxp3 promoter (Foxp3-GFP-hCre mice) (17, 18). Thus, the resultant E2Afl/flHEBfl/flFoxp3-GFP-hCre mice (designated hereafter as E2Afl/flHEBfl/fl mice)exhibited specific deletion of E2A and HEB in Treg cells but notin CD4+ conventional T cells (SI Appendix, Fig. S1A). We didnot observe changes in development of CD4+ and CD8+ T cellsor the proportion of Treg cells in the thymus (SI Appendix, Fig.S1 B and C). However, we did observe a small, but statisticallysignificant, increase in the proportion of Treg cells in the spleen,peripheral lymph nodes (pLN), and mesenteric lymph nodes(mLN) (SI Appendix, Fig. S1D), demonstrating a modest, butintegral role of E protein in the regulation of peripheral Treg cellhomeostasis. The lack of increase in Foxp3+ cells in thethymus of E2Af/fHEBf/f mice was likely due to the fact thatFoxp3+ cells in the thymus are newly developed Tregs in which aTCR-driven decline in E-protein levels determines the numberof Tregs rather than Foxp3(Cre)-dependent E-protein deletion.To investigate this possibility, we took advantage of Rosa26-loxP-Stop-loxP-YFP (R26YFP) mice, in which YFP is expressed only afterCre is expressed. Accordingly, we crossed E2Afl/flHEBfl/fl Foxp3Cre

    with R26YFP mice, in which Foxp3-expressing cells that areE-protein gene deleted are labeled with YFP, so that YFP+ cells areE-protein knockout (KO) Tregs. As expected, we found that mostof the Tregs in thymus of E2Afl/flHEBfl/flFoxp3CreR26YFP and WTFoxp3CreR26YFP mice were YFP-negative (SI Appendix, Fig. S1E),indicating that at this site TCR-driven decreases in E protein arelargely responsible for Treg development rather than Foxp3 (Cre)-dependent E-protein gene deletion. In contrast, most of the Tregcells in the periphery are YFP-positive, indicating that the cells haveundergone E-protein deletion as a result of Foxp3 (Cre) expression.Thus, the lack of increase of Tregs in the thymus of E2Af/fHEBf/f

    mice is probably due to the fact that the Tregs in thymus are subjectto a lesser degree of Foxp3-Cre mediated E-protein deletion thancells in the periphery.In related studies we found that the GI lamina propria, a

    mucosal lymphoid site that is exposed to an abundance of foreignantigens and that requires Treg cells for maintenance of intestinehomeostasis, exhibited a dramatic increase in Treg cell frequencyin E2Af/fHEBf/f mice and this increase was substantially greaterthan the increase in spleen and lymph nodes (Fig. 1A). Similarly,we found that Treg cell increases were also greater in liver andlung than in lymphoid organs (Fig. 1A). Recognizing that Tregcell development in the lamina propria is highly TGF-β de-pendent, we investigated the role of E protein on iTreg celldifferentiation in vitro and found that upon stimulation withTCR and TGF-β, cells lacking E protein gave rise to an increasedproportion of Foxp3+ cells (SI Appendix, Fig. S1F). Finally, weobserved an increase in the percentages of both Helios+Foxp3+cells and Helios−Foxp3+ cells in lamina propria of E2Afl/flHEBfl/flmice compared with that of WT mice (SI Appendix, Fig. S1G).

    Since Helios is a marker of nTreg cells (19), this suggests that Eprotein regulates both nTreg cells as well as iTreg cells.Collectively, these results indicate that the E-protein level in

    mature Treg cells plays a substantial role in regulating Treg cellhomeostasis particularly at nonlymphoid organs exposed to TCRsignaling by ambient antigens.

    E Proteins Regulate Treg Cell Homeostasis in a Cell-Intrinsic Manner.To determine if the effects of E protein on Treg cell homeostasisare cell-intrinsic, we generated mixed bone marrow (BM) chi-meric mice by mixing equal numbers of BM cells from CD45.1+

    CD45.2+ WT Foxp3Cre mice and CD45.1+ E2Afl/flHEBfl/fl

    Foxp3Cre mice and then transferred the cell mixtures into lethallyirradiated CD45.2+ mice. Analyses of the number of Treg cellsfrom WT (CD45.1+CD45.2+) or E protein-deficient (CD45.1+)cells in lymphoid organs 2 mo after cell transfer showed thatE protein-deficient Treg cells were significantly increased com-pared with WT Treg cells (Foxp3+CD45.1+/Foxp3+CD45.1+

    CD45.2+ ratio: 1.5) (Fig. 1B). In addition, in nonlymphoid or-gans of these mice, an even more pronounced increase inE protein-deficient Treg cells was observed (Foxp3+CD45.1+/Foxp3+CD45.1+CD45.2+ ratio: 2.5) (Fig. 1B). To investigateE-protein effects on Treg cell homeostasis under inflammatoryconditions, we immunized mixed bone marrow chimeric mice withmyelin oligodendrocyte glycoprotein (MOG) to induce experimentalallergic encephalomyelitis (EAE) or administered dextran sodiumsulfate (DSS) in drinking water to induce colitis. As expected,

    Foxp

    3

    lung liver

    CD4

    *SI-LP CLP

    05

    101520

    0

    5

    10

    155.9

    10.8

    8.2

    14.1

    WT

    WT WT

    SI-LP CLP***lung liver

    05

    101520

    05

    10

    20

    WT WT

    15*****

    0 103

    104

    105

    0

    103

    104

    105

    7.0

    12.6

    8.5

    16.3

    Foxp

    3+ C

    ells

    (%)

    Foxp

    3+ C

    ells

    (%)

    Foxp

    3+ C

    ells

    (%)

    Foxp

    3+ C

    ells

    (%)

    CD45.2

    CD

    45.1

    spleen SI-LP CLP

    spleen SI-LP CLP%

    of F

    oxp3

    + Cel

    lls

    020

    6080

    WT

    *** ***

    0-103

    103

    104

    105

    0

    -103

    103

    104

    105

    Gated Foxp3+ Cells

    40

    % o

    f Fox

    p3+ C

    ellls

    020

    6080

    WT

    40

    % o

    f Fox

    p3+ C

    ellls

    020

    6080

    WT

    40

    ***

    0-103

    103

    104

    105

    0

    -103

    103

    104

    105

    CD45.2

    CD

    45.1

    dLN spinal cord

    Gated Foxp3+ Cells

    spleen

    spleen dLN spinal cord

    % o

    f Fox

    p3+ C

    ellls

    020

    60

    100

    WT

    *** ***

    40

    % o

    f Fox

    p3+ C

    ellls

    020

    6080

    WT

    40

    % o

    f Fox

    p3+ C

    ellls

    020

    6080

    WT

    40

    ***

    0-103

    103

    104

    105

    0

    -103

    103

    104

    105

    CD45.2

    CD

    45.1

    Gated CLP Foxp3+ Cells

    CLP***

    % o

    f Fox

    p3+ C

    ellls

    020

    6080

    WT

    4080

    E2Afl/flHEBfl/fl

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    WT28

    KO72WT

    30

    KO70WT

    38

    KO62

    WT36

    KO64WT

    42

    KO58WT

    45

    KO55

    WT39

    KO61

    A

    B

    C D

    Fig. 1. E protein regulates Treg cell homeostasis in a cell-intrinsic manner.(A) Representative flow cytometric analysis (Left) and the percentages(Right) of Foxp3+ cells within CD4+ cells in indicated organs of WTFoxp3Cre

    and E2Afl/flHEBfl/flFoxp3Cre mice. (B–D) Mixed BM cells from WTFoxp3Cre mice(CD45.1+CD45.2+ cells) and E2Afl/flHEBfl/flFoxp3Cre mice (CD45.1+ cells) weretransferred into lethally irradiated recipient (CD45.2+) mice and were analyzed2 mo after reconstitution. (B) Data on the Left show representative flowcytometry analysis of WT and KO Treg cells in chimeric mice after gating onFoxp3+ cells and data on the Right show frequencies of WT (CD45.1+CD45.2+)and KO (CD45.1+) cells among the reconstituted Tregs (CD4+Foxp3+) in indicatedorgans. (C) Two months after reconstitution, EAE was induced in the chimericmice; 24 d after induction, Foxp3+ cells were analyzed by flow cytometry; dataon the Left show representative flow cytometry analysis of WT and KO Treg cellsin chimeric mice after gating on Foxp3+ cells and data on the Right show thefrequencies of WT (CD45.1+CD45.2+) and KO (CD45.1+) cells among the recon-stituted Tregs (CD4+Foxp3+) in indicated organs. (D) DSS-colitis was induced inthe chimeric mice; 10 d after induction Foxp3+ cells were analyzed by flowcytometry; representative flow cytometry analysis (Left) and frequencies (Right)of WT (CD45.1+CD45.2+) and KO (CD45.1+) cells in reconstituted Tregs(CD4+Foxp3+) in the CLP. Data in A–D, Left are representative of at leastthree independent experiments. Data in A–D, Right are pooled from at leastthree independent experiments. Each symbol represents data from onemouse. Graph shows mean ± SD (*P < 0.05, **P < 0.01, ***P < 0.001).

    4472 | www.pnas.org/cgi/doi/10.1073/pnas.1800494116 Han et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/cgi/doi/10.1073/pnas.1800494116

  • 24 d after induction, we observed a markedly increased number ofE protein-deficient Treg cells vs. WT Treg cells infiltrating thespinal cord as well as in spleen and draining lymph nodes (dLN)(Fig. 1C) of mice with EAE. A similar observation was obtainedin colon lamina propria (CLP) of chimeric mice 10 d after DSSadministration (Fig. 1D). It should be noted that the frequencyof Treg cells in KO mice without inflammation was comparableto that of KO mice with EAE or colitis (compare Fig. 1B withFig. 1 C and D). These studies thus indicated that the increasedpercentage of Foxp3+ cells among CD4+ cells due to E-proteindeletion is maintained in a cell-intrinsic manner under bothsteady-state and inflammatory conditions.

    E-Protein Deletion Stabilizes Foxp3 Expression in Matured Treg Cells.While Treg cells are generally stable, under lymphopenic or in-flammatory conditions a fraction of these cells lose Foxp3 expres-sion and undergo other phenotypic changes, such as acquisitionof ability to secrete cytokines that mediate diverse effector func-tions (18). To elucidate the effect of E proteins on Treg cell sta-bility, we used cells from E2Afl/flHEBfl/flFoxp3CreR26YFP miceand WT Foxp3CreR26YFP mice as described above to comparethe effect of E-protein deletion on the generation of YFP+Foxp3−

    (GFP−) “exTreg” cells. In initial studies, we compared the impactof inflammatory cytokines on the stability of highly purified Eprotein-deficient vs. WT spleen YFP+ cells after culture of thecells in the presence of IL-2 plus various proinflammatory cyto-kines (IL-12, IFN-γ, IL-4, or IL-6). We found that even in thepresence of optimal amounts of IL-2, the stability of WT Treg cellswas severely compromised compared with that of E protein-deficientTreg cells as indicated by the greater percentage of exTreg cells thatappeared in theWT cells (Fig. 2A). Intriguingly, the exTreg cells fromthe WT mice produced more IFN-γ (SI Appendix, Fig. S2A) and IL-17A (SI Appendix, Fig. S2B) than exTreg cells from E protein-deficient mice; this suggests that E proteins may promote exTregcells to become pathogenic T helper cells during inflammations suchas EAE. In addition, we observed a lower percentage of YFP+Foxp3−

    (exTreg) cells among YFP+ cells in the small intestine laminapropria and CLP of E2Afl/flHEBfl/flFoxp3CreR26YFP mice than inWT Foxp3CreR26YFP mice (Fig. 2B and SI Appendix, Fig. S2E).However, in this case the difference, while significant, was lessthan that observed in cultured cells. In contrast, there were no dif-ferences in the percentage of exTregs in the spleen and pLN cellsof E2Af/fHEBf/fR26YFP mice and WT mice perhaps because cells inthese tissues are less subject to antigen stimulation than those in themucosal tissues (SI Appendix, Fig. S2F).To further investigate the role of E protein on the stability of

    Treg cells, we transferred highly purified YFP+ Treg cells fromspleen and lymph nodes of E2Afl/flHEBfl/flFoxp3CreR26YFP

    or WTFoxp3CreR26YFP mice into Rag2−/− recipients and assessedFoxp3+ (GFP+) cell stability under lymphopenic conditions 4 wkafter cell transfer. Consistent with the findings above, fewer Eprotein-deficient Treg cells lose Foxp3 expression than WT Tregcells, indicating that the E protein- deficient Treg cells are morestable under lymphopenic conditions (Fig. 2C).To confirm the above findings concerning Treg cell stability,

    we determined the role of E-protein activity in maintaining Tregcell stability in cells from Id2fl/flId3fl/flFoxp3CreR26YFP mice, i.e.,mice in which E-protein effects are increased because they arenot blocked by Id protein. Recognizing that mice with deletion ofId2/Id3 in matured Treg cells exhibit spontaneous inflammationthat might have a secondary effect on Treg cell stability (16), weexamined the effect of Id proteins on Treg cell stability under thenoninflammatory conditions in mixed bone marrow chimericmice. Bone marrow cells from WT Foxp3CreR26YFP and Id2fl/fl

    Id3fl/flFoxp3CreR26YFP mice were mixed at 1:1 ratio and trans-ferred into lethally irradiated recipient mice; subsequently (i.e.,at 8 wk after cell transfer when the recipient mice were robustand thus apparently free of inflammation), we analyzed thefraction of exTreg cells. We found that the percentage of exTreg

    (Foxp3−) cells among YFP+ cells was greatly increased in spleen,pLN, and mLN cells derived from Id protein-deficient micecompared with cells derived from WT mice (Fig. 2D). In com-panion studies we cultured highly purified CD25+CD4+ Tregcells from WTFoxp3Cre and Id2fl/flId3fl/flFoxp3Cre mice in thepresence of IL-2 and proinflammatory cytokines as describedabove, and found that the inflammatory cytokines compromisedFoxp3 expression in Id protein-deficient Treg cells to a greaterextent than in WT Tregs (Fig. 2E); in addition, these exTreg cellsfrom Id2fl/flId3fl/flFoxp3Cre mice produced increased amountsof cytokines (IFN-γ, IL-4) compared with that of WT exTreg cells (SIAppendix, Fig. S2 C andD). Collectively, these studies provide strongevidence that lack of E-protein expression in E2Afl/flHEBfl/flFoxp3Cre

    mice and increased E-protein activity in Id2fl/flId3fl/flFoxp3Cre miceaffects Treg stability in reciprocal ways, most likely via un-derlying effects on survival or proliferation of Foxp3+ cells.

    E Protein-Deficient Treg Cells Exhibit Enhanced ImmunosuppressiveFunction both in Vitro and in Vivo. Previous studies have demon-strated that Treg cells have a prominent capacity to limit in-flammation during experimental autoimmune diseases such asEAE or cell-transfer colitis. Based on the above findings, we

    Foxp

    3- in

    YFP

    + Cel

    ls (%

    ) SI-LP CLP*

    0

    30

    10

    20

    *

    Foxp3

    FSC

    IL-2+IFN-γ IL-2+IL-4 IL-2+IL-6

    0-103

    103

    104

    105

    0

    50K

    100K

    150K

    200K

    250K

    IL-2 IL-2+IL-12

    cells

    GFP

    63.4

    34.7

    0-103

    103

    104

    105

    0

    50K

    100K

    150K

    200K

    250K

    Foxp3

    FSC

    IL-2+IL-4 IL-2+IL-6IL-2 IL-2+IL-12

    3.6 11.2 7.3 15.1 35.8

    3.3 6.3 3.9 6.8 25.4

    9.9 16.5 11.0 12.8 54.0

    26.2 26.0 26.4 26.2 72.3

    WT R26YFP

    E2Afl/flHEBfl/flR26YFP

    WT WT

    IL-2+IFN-γ

    WT

    Id2fl/flId3fl/fl

    GFP

    - in

    YFP

    + Cel

    ls (%

    ) spleen***

    020

    8060

    WT

    40

    WT R26YFP

    YFP

    Foxp

    3

    spleen pLN mLN

    WT R26YFP

    Id2fl/flId3fl/fl

    R26YFP

    11.31.1

    4.00.8

    10.10.9

    3.31.1

    9.31.0

    3.90.9

    Foxp

    3- in

    YFP

    + Cel

    ls (%

    )

    0

    50

    10203040

    **

    0-103

    103

    104

    105

    0

    103

    104

    105

    Foxp

    3- in

    YFP

    + Cel

    ls (%

    )

    05

    20

    1015

    E2Afl/flHEBfl/flR26YFP

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    0-103

    103

    104

    105

    0

    20

    40

    60

    80

    100

    ******

    splee

    npL

    NmL

    N

    Id2fl/flId3fl/flR26YFPWT R26YFP

    BA

    DC

    E

    Fig. 2. E-protein deletion stabilizes Foxp3 expression in matured Tregcells. (A) Highly purified YFP+ Treg cells from WT Foxp3CreR26YFP andE2Afl/flHEBfl/flFoxp3CreR26YFP mice were activated and cultured in the pres-ence of IL-2 plus indicated cytokine for 4 d; Foxp3 expression (among YFP+

    cells) was then analyzed by flow cytometry. (B) The frequency of Foxp3−

    in YFP+ cells (Right) in indicated organs of WT Foxp3CreR26YFP andE2Afl/flHEBfl/flFoxp3CreR26YFP mice. (C) Highly purified Treg (YFP+) cells fromWT Foxp3CreR26YFPFoxp3GFP or E2Afl/flHEBfl/flFoxp3CreR26YFPFoxp3GFPmice weresorted by FACS and were transferred into Rag2−/−-recipient mice. GFP expressionamong YFP+ cells was analyzed by flow cytometry 4 wk after cell transfer (Left)and the frequency of GFP− (Foxp3−) among YFP+ cells is shown (Right). (D)Mixed BM chimeric mice were generated by transferring BM cells fromWT Foxp3CreR26YFP and Id2fl/flId3fl/flFoxp3CreR26YFP mice into lethally irradiatedrecipient mice. Flow cytometry analysis of Foxp3 and YFP expression in CD4+

    cells (Left) and the frequency of Foxp3− cells among the YFP+ cells (Right) ofWT Foxp3CreR26YFP and Id2fl/flId3fl/flFoxp3CreR26YFP donors in indicated organs inthe chimeric mice are shown. (E) Highly purified CD25hi CD4+ Treg cells fromWTFoxp3Cre and Id2fl/flId3fl/flFoxp3Cre mice were activated and cultured in thepresence of IL-2 plus indicated cytokine for 4 d; Foxp3 expression was analyzedby flow cytometry. (A, E, and B–D, Left) Data are representative of at least threeindependent experiments. (B–D, Right) Data are pooled from at least three in-dependent experiments. Each symbol represents data from one mouse. Graphshows mean ± SD (*P < 0.05, **P < 0.01, ***P < 0.001).

    Han et al. PNAS | March 5, 2019 | vol. 116 | no. 10 | 4473

    IMMUNOLO

    GYAND

    INFLAMMATION

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplemental

  • reasoned that E-protein deletion specific to Treg cells wouldlead to enhanced Treg cell function. To investigate this possi-bility, we induced EAE in WT and E2Afl/flHEBfl/flFoxp3Cre miceand found that E2Afl/flHEBfl/flFoxp3Cre mice were more resistantto EAE, as evidenced by lower disease scores, not only duringthe initial progressive phase of disease, but also during theremitting-relapsing phase of disease occurring after peak disease(Fig. 3A). This finding could be attributed to differences in Tregcell migration since significantly more Foxp3+ cells were pre-sent in the spinal cords of E2Afl/flHEBfl/flFoxp3Cre mice thanthose of WT Foxp3Cre mice (Fig. 3B). Of interest, there were moreMOG-specific Treg cells in the E2Afl/flHEBfl/flFoxp3Cre mice thanthat of WT Foxp3Cre mice (Fig. 3C); accordingly, MOG-specificCD4+ T cells were greatly decreased in E2Af/fHEBf/f mice com-pared with that of WT mice (Fig. 3C), suggesting that E protein-deficient Treg cells are more responsive to antigen stimulation, andantigen-specific E protein-deficient Treg cells exhibit enhancedsuppressive capacity. E2Afl/flHEBfl/flFoxp3Cre mice spinal cordsharbored significantly fewer IL-17– and IFN-γ–producing cells(Fig. 3 D and E), which also suggests that E2Afl/flHEBfl/flFoxp3Cre

    mice were more resistant to EAE.As E2Afl/flHEBfl/flFoxp3Cre mice have an increased percentage

    of Treg cells in the spinal cords, the increased suppressivefunction of Treg cells in E2Afl/flHEBfl/flFoxp3Cre mice with EAEmay simply be due to the presence of an increased Treg cell poolrather than because the Treg cells are more suppressive. Toclarify this issue, we performed in vitro suppressive assays inwhich titrated Treg cells from WT or E2Afl/flHEBfl/flFoxp3Cre

    were cocultured with an equal number of carboxyfluoresceindiacetate succinimidyl ester (CFSE)-labeled CD4+ T cells andthe CD4+ T cell proliferation was assessed. We found that Eprotein-deficient Treg cells had an increased capacity to suppressconventional CD4+ T cell proliferation compared with WT Tregcells. Thus, Treg cells with E-protein deletion do exhibit in-creased intrinsic suppressive activity (Fig. 3F).To further investigate the suppressive function of E protein-

    deficient Treg cells in vivo, we turned to the well-characterizedcell transfer-induced colitis model. Here, we compared colitisdeveloping in Rag2−/− mice following transfer of naïve CD45RBhi

    CD4+ T cells together with Treg cells from WT Foxp3CreR26YFP

    or E2Afl/flHEBfl/flFoxp3CreR26YFP mice. Recipient mice cotransferredE protein- deficient Treg cells exhibited less colitis than micecotransferred WT Treg cells as indicated by their increasedweight gain (Fig. 3G), decreased cell infiltration in the colon(SI Appendix, Fig. S2G), and by the dramatic decrease of bothtotal CD4+ T cells and total IL-17– and IFN-γ–producing CD4+T cells (Fig. 3 H–J and SI Appendix, Fig. S2H). Importantly,although the Treg cell percentages were increased in spleen, mLN,and colon of mice that were transferred E protein-deficient Tregs(Fig. 3H and K) due to decreased total CD4+ T cell numbers (Fig.3H) compared with mice transferred WT Treg cells, the absolutenumbers of Treg cells in spleen, mLN, and colon were comparablein the two groups (Fig. 3K). Thus, these data further support theview that decreased E-protein levels in Treg cells result in Tregcells with increased regulatory function.

    E Protein Globally Regulates Effector Treg Cell Signature GeneExpression. The above findings showing that E protein-deficientTreg cells exhibit increased homeostasis, enhanced suppressivefunction and increased stability prompted us to hypothesize thatE protein might control a distinct, lineage-specific, transcrip-tional program in Treg cells. To investigate this possibility, weused high-throughput RNA-sequencing (RNA-seq) analysis tocompare the global gene-expression patterns of E protein-deficient and -sufficient Treg cells. Recognizing that Treg cellsare constantly being stimulated by self-antigens and that E pro-teins are regulated by TCR stimulation, we compared E protein-deficient and -sufficient Treg cells without stimulation [KO vs.WT (no TCR hereafter)] or stimulated with anti-CD3/CD28 for48 h [KO vs. WT (TCR 48 h hereafter)] in the studies.

    The RNA-seq study showed that 801 genes were up- or down-regulated at least twofold in E protein-deficient Treg cells rela-tive to the genes in WT Treg cells (Fig. 4A, Top and Fig. 4B,Venn A). These genes were termed “E protein-dependent genes,”whereas 1,611 genes changed expression at least twofold in Eprotein-deficient Treg cells subjected to TCR stimulation relativeto genes in WT Treg cells also subjected to TCR stimulation(Fig. 4A, Bottom and Fig. 4B, Venn B). Meanwhile, 6,606 geneexpressions were changed in WT Treg cells after TCR stimula-tion (WT vs. WT-48 h), which were termed “TCR-dependentgenes” (Fig. 4B, Venn C). In further analysis, we found that asimilar proportion of genes regulated by E protein [WT vs. KOcells (Fig. 4B, Venn A) and WT-TCR 48-h vs. KO-TCR 48-hcells (Fig. 4B, Venn B)] were also TCR dependent (Fig. 4B, RightTop, Venn A overlap C, 433/801 genes = 54% and Fig. 4B, RightBottom, Venn B overlap C, 887/1,611 genes = 55%, respectively).This similarity in the proportion of genes regulated by E proteinalone and by E protein plus TCR stimulation suggests that theeffects of E protein on gene regulation are parallel to or slightlyamplified by TCR stimulation. In addition, comparison of E proteinand TCR-dependent genes showed that most of the genes up-regulated in E protein-deficient Treg cells were also TCR-dependent genes (Fig. 4C). This suggests that a large proportionof Treg cell-specific gene expression downstream of TCR signaling,is dependent on E protein and therefore, the E-protein regulatorynetwork ties TCR signaling to matured Treg cell homeostasis.Examination of the specific genes up-regulated in KO vs. WT

    (TCR 48-h) disclosed several genes encoding transcription fac-tors shown previously to be involved in the differentiation andfunction of effector Treg cells such as Nfat5, Ikzf3, and Irf4, aswell as genes encoding factors with a similar potential func-tion, Itgae, Icos, Cd44, and Lag3 (Fig. 4D). In addition, genesencoding secreted factors associated with Treg cell suppressivefunction, including Granzyme B, Il10, and Fgl2 were also signif-icantly increased in KO vs. WT (TCR 48-h) as were genes in-volved in the TGF-β signaling pathways, such as Tgfbr3 andSmad3 (Fig. 4D). In a related vein, the expression of RORγt wasincreased in E protein-deficient Treg cells following TCR stim-ulation. This is significant because an effector Treg cell lineagesubset that expresses both Foxp3+ and RORγt+ has been shownto have enhanced suppressive capacity and increased ability toregulate Th17-mediated immune responses (20–22). Finally,several chemokine and chemokine receptor or integrin-encodinggenes were up-regulated in the E protein-deficient cells sub-jected to TCR stimulation, suggesting that such Treg cells havean increased ability to migrate to inflammatory sites (Fig. 4D).The up-regulation of specific genes in E protein-deficient cells

    subjected to TCR stimulation noted above was accompanied bythe up-regulation of a large number of genes involved in moregeneral cell functions. This includes genes encoding ribosomeRNA-processing genes, indicating that protein translation wasgreatly up-regulated by E-protein deletion in these cells; in ad-dition, consistent with gene ontology (GO) analysis, genes corre-lating with cell proliferation, cell cycling, and cell apoptosis, suchas cdk6, cdkn1a, and Bcl2 (Fig. 4D), were also greatly up-regulatedin E protein-deficient Treg cells. Thus, loss of E protein in dif-ferentiated Treg cells affect the expression of a large proportion ofgenes underlying the multifaceted function of effector Treg cells.Interestingly, one set of genes not up-regulated by TCR stim-

    ulation in E protein-deleted cells are genes associated withIL-2 and IL-7 signaling, i.e., genes that have been shown to becritically involved in initial Treg cell induction and maintenance(23). In particular, neither CD25, CD122 (IL-2Rα and IL-2Rβchains, respectively), CD127 (IL-7Rα chain) expression wassignificantly altered upon E-protein depletion in matured Tregcells (SI Appendix, Fig. S4A) nor was IL-2 signaling as evidencedby phosphorylation of STAT5 (SI Appendix, Fig. S3A). In addi-tion, c-Rel, a pioneering transcription factor that initiates Tregcell development was also not modulated by E-protein depletion(SI Appendix, Fig. S3B). It is thus apparent that while decreasedE-protein activity is a key feature of the cell signaling pathways

    4474 | www.pnas.org/cgi/doi/10.1073/pnas.1800494116 Han et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/cgi/doi/10.1073/pnas.1800494116

  • 0-103

    103

    104

    105

    0

    -103

    103

    104

    105

    dise

    ase

    sco

    re

    WT

    ** *

    *

    spin

    al c

    ord

    Foxp

    3+ C

    ells

    (%)

    spin

    al c

    ord

    IFN

    -γ+ C

    ells

    (%)

    spin

    al c

    ord

    IL-1

    7A+ C

    ells

    (%)

    spin

    al c

    ord

    IFN

    γ+IL

    17A

    + Cel

    ls (%

    )

    IL-17A

    IFN

    15105 20 250Time (d)

    1

    3

    0

    2

    010

    4050

    2030

    05

    10

    20*

    0

    3040

    1020 4

    6

    02

    8

    22.5 2.4

    69.1 6.0

    15.0 1.7

    81.0 2.3

    0 103

    104

    105

    0

    103

    104

    105

    IL-17A

    IFN

    PBS WT Treg KO Treg

    20.2 9.9

    55.7 14.2

    15.1 8.5

    58.8 14.6

    8.3 3.3

    75.8 12.6

    sple

    en IF

    N-γ

    + C

    ells

    (×10

    5 )

    012

    4

    sple

    en IL

    17A

    + Cel

    ls (×

    106 )

    0.00.51.0

    2.0

    WT

    *** **

    **

    WT WT WT

    0-103

    103

    104

    105

    0

    -103

    103

    104

    105

    CD4

    Foxp

    3

    WT

    WT

    CD45RBhi+

    WT T

    reg

    KO T

    reg

    PBS

    CD45RBhi+

    WT T

    reg

    KO T

    reg

    PBS

    CD45RBhi+

    19.5

    33.0

    MO

    G35

    -55

    CD44

    3.7

    2.2*

    (%) M

    OG

    35-5

    5+ o

    f Fox

    p3+ C

    ells

    0

    4

    12

    WT

    3

    15

    3 1.5

    spinal cord Gated spleen Foxp3+ Cells

    spinal cord

    *

    ****

    *

    ****

    *

    0

    6

    2

    WT

    4

    (%) M

    OG

    35-5

    5+ o

    f CD

    4+ C

    ells

    1.28 4.07 7.49

    0.24 5.44 8.46

    0.46

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    2.09 6.67

    PBS WT Treg KO Treg

    CD45RBhi+

    CD4

    Foxp

    3

    spleen

    mLN

    colon

    CD45RBhi+PBS

    CD45RBhi+KO Treg

    wei

    ght (

    % in

    itial

    )

    Time after transfer (weeks)

    CD45RBhi+WT Treg

    0 4 62 890

    100

    140150

    130

    110120

    0

    151015

    0

    5

    0246

    1020304050

    splee

    nmL

    NCL

    P

    CD45RBhi+PBS CD45RBhi+KO TregCD45RBhi+WT Treg

    10

    15**

    ******

    **

    ******

    **

    ******

    ns***

    ***

    ns

    ******

    ns

    *******

    **

    *

    ***

    *

    **

    CD

    4+ C

    ells

    (×10

    6 )

    Fox

    p3+ C

    ells

    (%)

    Fox

    p3+ C

    ells

    (×10

    4 )

    39.6

    25.0

    0-103

    103

    104

    105

    0

    50

    100

    150

    58.4

    40.3

    70.1

    59.7

    80.2

    70.1

    84.9

    81.0

    85.1

    83.4

    88.9

    1:1

    CFSE

    cells

    WT

    Treg: Tconv 1:2 1:4 1:8 1:16 1:32 0:1

    no Treg

    KO TregWT Treg

    % D

    ivid

    ed

    020

    80100

    6040

    1:1 1:2 1:4 1:81:16

    1:32 0:1

    Treg: Tconv ratio

    ***

    ***

    *****

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    0 103

    104

    105

    0-102

    102

    103

    104

    105

    E2Afl/flHEBfl/fl

    *

    *

    splee

    nmL

    NCL

    Psp

    leen

    mLN

    CLP

    A

    D

    F

    G

    I J K

    H

    E

    B C

    Fig. 3. Treg cells deficient in E protein exhibit enhanced immunosuppressive function in vivo. (A–E) EAE was induced in WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre

    mice and the extent of disease assessed at day 24. (A) The disease score of WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice after EAE induction (WTFoxp3Cre n = 4,E2Afl/flHEBfl/flFoxp3Cre n = 4). (B) Representative flow cytometric analysis (Left) and the frequency (Right) of Foxp3+ cells among CD4+ cells infiltrating the spinalcord of WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice. (C) Flow cytometric analysis of MOG-specific Treg cells in spleens of WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre

    mice (Left), frequency of MOG35–55+ among Foxp3+ cells (Middle), and the frequency of MOG35–55

    + among CD4+ cells (Right). (D and E) Cells from spinal cordof WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice were stimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin. (D) Cytokine production of thecells analyzed by flow cytometry. (E) The frequency of IFN-γ+, IL-17A+, and IFN-γ+IL-17A+ cells. (B–E) Data are pooled from two independent experiments;each symbol represents data from one mouse, mean ± SD are shown (*P < 0.05, **P < 0.01). (F) CFSE-labeled Tconv cells (CD4+CD25−CD45RBhi) from WT micewere cultured with or without titrated numbers of YFP+ Treg cells from WT Foxp3CreR26YFP or E2Afl/flHEBfl/flFoxp3CreR26YFP mice for 3 d after which celldivision was analyzed by flow cytometry. Shown are representative cell proliferation dye dilution profiles of Tconv cells of four independent experiments(Left) and percentages of these cells, divided at different Tregs. Tconv cell ratios (Right); n = 4, mean ± SD are shown (**P < 0.01, ***P < 0.001). (G–K) Six- to 8-wk old RAG2-deficient mice were adoptively transferred with WT naive CD45RBhi CD4+ T cells together with either PBS (CD45RBhi+PBS) or sorted Treg cellsderived from WT Foxp3CreR26YFP mice (CD45RBhi+WT Treg) or from E2Afl/flHEBfl/flFoxp3CreR26YFP mice (CD45RBhi+KO Treg) (n = 6 in each group). (G) Bodyweight curve of mice in the various groups. One-way ANOVA with the Dunnett’s multiple comparisons test were used to compare the weight loss curve. *P <0.05. (H) The absolute number of CD4+ cells (Top), the frequency of Foxp3+ cells in CD4+ cells (Middle), and the absolute number of Foxp3+ cells (Bottom) inindicated organs. (I) Flow cytometry of IFN-γ+ and IL-17A+ cells in CD4+ cells in the spleen. (J) The absolute number of IFN-γ+, IL-17A+, and IFN-γ+IL-17A+ cells.(K) Flow cytometry analysis of Foxp3+ cells in indicated organs. (I and K) Data are representative of two independent experiments. (H and J) Data are pooledfrom two independent experiments; each symbol represents data from one mouse, mean ± SD are shown. (One-way ANOVA with the Dunnett’s multiplecomparisons test were used. ns, not significant, *P < 0.05, **P < 0.01, ***P < 0.001.)

    Han et al. PNAS | March 5, 2019 | vol. 116 | no. 10 | 4475

    IMMUNOLO

    GYAND

    INFLAMMATION

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

  • necessary for initial development of Foxp3+ Treg cells, its furtherdecreased expression in matured Treg cells does not regulatethese signaling pathways.In a final set of studies relating to the above RNA-seq studies,

    we validated the various findings with quantitative RT-PCRanalyses. Accordingly, YFP+ Treg cells sorted from spleens ofE2Afl/flHEBfl/flFoxp3CreR26YFP or WT Foxp3CreR26YFP mice werestimulated with anti-CD3/28 for 48 h after which mRNA extractedfrom cultured cells was subjected to quantitative RT-PCR. Con-sistent with the RNA-seq data, a host of molecules essential foreffector Treg cell differentiation and function were significantlyincreased in TCR-stimulated E protein-deficient Treg cells vs. WTTreg cells (SI Appendix, Fig. S3C). In confirmation, similar studiesconducted with Treg cells sorted from Id2fl/flId3fl/flFoxp3CreR26YFP

    mice vs. cells from WT Foxp3CreR26YFP mice showed that theexpression of most of these genes was decreased in the Id2/Id3-depleted Treg cells (SI Appendix, Fig. S3D).

    E-Protein Activity Regulates Effector Treg Cell Differentiation,Trafficking, Proliferation, and Survival. The observation that theratio of Foxp3+ cells of E2Afl/flHEBfl/flFoxp3Cre mice increased

    more remarkably in nonlymphoid tissues than in the spleen orlymph nodes, and that many E protein-dependent genes wereassociated with effector Treg cell signatures as indicated inRNA-seq analysis, suggested that E protein-deficient Treg cellsare prone to differentiate toward effector Treg cells. To confirmthe impact of E-protein deletion on effector Treg cell differ-entiation, we compared E2Afl/flHEBfl/flFoxp3Cre mice and WTFoxp3Cre control mice with respect to expression of moleculespreviously known to associate with Treg cell activation and/oreffector function. This analysis revealed that surface moleculesindicative of activated or effector Treg cells, such as CD103,ICOS, and KLRG1, were significantly increased in E protein-deficient Treg cells (SI Appendix, Fig. S4A). As expected, thesechanges in expression were more pronounced in the CLP andother nonlymphoid organs such as lung and liver, as indicated bythe fact that the proportion of Foxp3+ cells that expressCD103 and KLRG1 were significantly increased at these tissuesites (Fig. 5 A and B). Similarly, the expression of ICOS andIRF4, the latter a transcription factor important for effectorTreg differentiation and function (24), was also greatly increased

    -log 1

    0 (pv

    al) K

    O v

    s W

    T (T

    CR

    48h

    )

    1.3

    20

    40

    Up: 1187Down: 424

    -log 1

    0 (pv

    al) K

    O v

    s W

    T (n

    o TC

    R)

    1.3

    10

    20

    Up: 338Down: 463

    log2 (fold change)-11-4 4

    log2 (fold change)-1 1-4 4 0

    100

    150

    5054% TCR & E-protein depenent

    WT vs KO ( no TCR)

    E-protein dependent

    WT vs KO (TCR 48h)

    Cdkn1aCdk17

    Ki67

    Cnbd2

    Bcl2

    proliferation&apoptosis

    Ccr5

    Icos

    Lag3

    Cd44Il1r1

    Tnfsf9

    Cell surface proteins

    GzmbLta

    Fgl2

    Il10Function associated

    Ahr

    Maf

    Irf4Batf3

    Ikzf3

    Nfat5

    Tbx21

    TCR 48hKO/WT

    no TCRKO/WT

    Transcription factorsCcl20

    Ccr2

    Ccl3Ccl28Ccl4

    Cxcr6Ccr5

    Chemkine & receptor

    Itgb3ItgaeItgavItgb5

    Itga3integrin

    Cdk6

    Bcl11b

    Il12rb2

    Tgfbr3Il18r1

    Tgfb

    RorcSmad3

    Irf8

    Rrp15Rrp7aRrp8Dis3

    Lars2Rp1l2KhnynRrp1b

    Exosc5

    RNA-processing

    Expression (log2fold)

    Furin

    TCR 48hKO/WT

    no TCRKO/WT

    TCR 48hKO/WT

    no TCRKO/WT

    TCR 48hKO/WT

    no TCRKO/WT

    TCR 48hKO/WT

    no TCRKO/WT

    TCR 48hKO/WT

    no TCRKO/WT

    TCR 48hKO/WT

    no TCRKO/WT

    g

    0

    100

    150

    5055%

    ( g2222222 )))

    -2 20

    A: WT vs KOB: WT-48h vs KO-48hC: WT vs WT-48hD: KO vs KO-48h

    A BC D

    E-protein dependent

    0

    100

    150

    5055%G

    enes

    (%)

    TCR dependent

    B: KO-48h vs WT-48h (log2fold>1)A: WT-48h vs WT

    A B

    KO-48h vs WT-48h upregulated

    Exp

    ress

    ion

    abs

    log 2

    (fol

    d)

    WT-

    48h

    vs W

    T

    7.510

    12.5 significant: 647

    Expression log2 (fold)KO vs WT (TCR 48h)

    -1 1-5 5 10-10

    2.55.0

    1.0

    TCR & E-protein depenent

    %TC

    R &

    E-p

    rote

    in-d

    epen

    dent

    gen

    es

    amon

    g E

    -pro

    tein

    de

    pend

    ent g

    enes

    %TC

    R &

    E-p

    rote

    in-d

    epen

    dent

    gen

    es

    amon

    g E

    -pro

    tein

    de

    pend

    ent g

    enes

    A B

    C

    D

    Fig. 4. E protein globally regulates effector Treg cell signature gene expression. (A) Genes expressed differently in Treg cells from E2Afl/flHEBfl/flFoxp3Cre mice(KO) vs. fromWTFoxp3Cre mice (WT) (Top) or after anti-CD3/CD28 stimulated for 48 h (WT-48 h, KO-48 h, respectively) (Bottom) were plotted; numbers in plotsindicate genes up-regulated (red) or down-regulated (green) by twofold or more (P < 0.05). (B, Left) Venn diagram. (Right) Genes differently expressedtwofold or more in KO Treg cells vs. WT Treg cells (Venn A) that also changed expression upon TCR stimulation (Venn C) (Top), and genes differently expressedtwofold or more in KO-48 h Treg cells vs. WT-48 h Treg cells (Venn B) that were also TCR dependent (Venn C) (Bottom). (C, Left) Genes up-regulated inE-protein KO-48 h cells vs. WT-48 h plotted against those differently expressed in WT-48 h vs. WT cells; (C, Middle) Venn diagram of genes differently expressedtwofold or more in WT-48 h vs. WT Treg cells (TCR-dependent genes) (Venn A) relative to those up-regulated twofold or more in KO-48 h vs. WT-48 h Tregcells (up-regulated genes in E-protein KO Tregs (Venn B); (C, Right) gene% = TCR-dependent genes among those up-regulated twofold or more in E-proteinKO Treg cells. (D) Heat map of E protein-regulated genes.

    4476 | www.pnas.org/cgi/doi/10.1073/pnas.1800494116 Han et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/cgi/doi/10.1073/pnas.1800494116

  • in Foxp3+ cells from E2Afl/flHEBfl/flFoxp3Cre mice compared withWT mice (Fig. 5C).Treg cells with effector characteristics display an increased

    ability to accumulate in inflammatory sites to suppress in-flammation (25). In a functional study of the effect of E-proteindeletion on Treg cell effector characteristics, we determined theeffect of deletion of E protein on the ability of Treg cells toaccumulate in the spinal cords of mice with EAE, i.e., a site ofinflammation where they can exert suppressive function asdemonstrated in Fig. 3. We found that in parallel with cells in thespleen (SI Appendix, Fig. S4B) and draining lymph nodes (SIAppendix, Fig. S4C) of E2Afl/flHEBfl/flFoxp3Cre mice undergoingEAE, the spinal cords of E2Afl/flHEBfl/flFoxp3Cre mice containeda significantly increased proportion of Treg cells expressingCD103 and KLRG1 (SI Appendix, Fig. S4D) as well as in-creased amounts of ICOS and IRF4 (SI Appendix, Fig. S4E).In additional studies to determine whether the increased ex-

    pression of activation/effector markers in Treg cells above werecell intrinsic, we conducted studies of effector Treg cells de-veloping in mixed BM chimeric mice generated as describedabove and analyzed 8 wk after reconstitution. As expected, wefound a higher percentage of CD103+, KLRG-1+ Treg cells (SIAppendix, Fig. S4F) and up-regulated expression of ICOS andIRF4 in Treg cells (SI Appendix, Fig. S4G) derived from theE2Afl/flHEBfl/flFoxp3Cre mice donors. In parallel studies, cellsfrom chimeric mice in which EAE had been induced were ex-amined. Here again, the percentage of CD103+Foxp3+ cells orKLRG-1+Foxp3+ cells infiltrating in spinal cord (Fig. 5D) ordraining lymph nodes (SI Appendix, Fig. S4H) as well as theexpression of ICOS and IRF4 on Treg cells in spinal cord (Fig.5E) or draining lymph nodes (SI Appendix, Fig. S4I) was greatlyincreased from the E2Afl/flHEBfl/flFoxp3Cre mice donors thanfrom WT donors. In reciprocal studies to determine if the phe-notype of Treg cells from Id2fl/flId3fl/flFoxp3Cre mice was cellintrinsic, we conducted studies of Treg cells in mixed bonemarrow chimeras reconstituted with cells from WT Foxp3Cre andId2fl/flId3fl/flFoxp3Cre mice. We found that CD103, ICOS, KLRG1,and IRF4 expression in Treg cells from Id2fl/flId3fl/flFoxp3Cre

    donor mice were significantly decreased compared with cells fromWT Foxp3Cre donor mice (Fig. 5 F and G). These various studiesof chimeras thus provided firm evidence that E protein modulateseffector Treg cell differentiation in a cell-intrinsic manner.The RNA-seq analysis also indicated that E protein-deficient Treg

    cells were also marked by increased expression of genes associatedwith “apoptosis” and “cell cycle.” On the basis of these findings, wereasoned that the increased Treg cell number in peripheral tissue of Eprotein-deficient mice (Fig. 1) was a consequence of increased Tregcell proliferation or survival in response to continuous autoantigenstimulation. To investigate this possibility, YFP+ Treg cells were sortedfrom E2Afl/flHEBfl/flFoxp3CreR26YFP and WT Foxp3CreR26YFP mice,labeled with CFSE and cultured in the presence of anti-CD3/CD28 and IL-2 for various lengths of time. Treg cells fromE2Afl/flHEBfl/flFoxp3Cre mice exhibited a greater number of cell di-visions in each time period than WT Foxp3+ cells (Fig. 5H) as wellas increased numbers of Ki67+ cells (Fig. 5I); in addition, theyexhibited decreased numbers of AnnexinV+ (apoptotic cells)over 24 h (SI Appendix, Fig. S4J). Accordingly, this was accom-panied by an increased accumulation of Treg cells over the 4-dperiod of study (Fig. 5J).Collectively, these data provided strong evidence that E protein

    exerts broad regulation of the gene expression required for effectorTreg cell differentiation, function, survival, and proliferation.

    Foxp3+RORγt+ but Not Foxp3+Tbet+ or Foxp3+GATA3+ Effector TregCell Subsets Are Regulated by E Protein. Previous reports indicatethat Foxp3+ Treg cells can co-opt the expression of specifictranscription factors that are associated with the differentiationand function of effector CD4+ T cell lineages. For example, ithas been demonstrated that Foxp3+ T cells expressing RORγtrepresent a stable regulatory T cell effector lineage with an en-

    hanced suppressive capacity during intestinal inflamma-tion. Similarly, GATA3 and T-bet are expressed in activatedTreg cells and are required for Treg cell suppressive functionunder specific physiological circumstances for the maintenanceof Treg cell homeostasis (26–30). Given that RORγt as well asT-bet expression was greatly increased upon TCR stimulation inE protein-deficient Treg cells in the RNA-seq analysis, we inves-tigated whether E protein specifically regulated certain effectorTreg cell lineage differentiation. Foxp3+RORγt+, Foxp3+T-bet+,and Foxp3+GATA3+ effector Treg cell subsets were detected inspleen, lung, liver, and CLP of WT Foxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice. However, whereas the Foxp3+RORγt+ subset wassignificantly increased in E2Afl/flHEBfl/flFoxp3Cre mice relative toWT Foxp3Cre mice (Fig. 6 A and B), Foxp3+T-bet+ and Foxp3+GATA3+ Treg cell subsets were not comparably increased (SIAppendix, Fig. S5 A and B). In a series of further studies, weinvestigated different Treg subset cells under inflammatoryconditions, including EAE, DSS-induced colitis, and Listeriamonocytogenes infection; we reproducibly observed that onlyFoxp3+RORγt+ Treg cells were dramatically increased in all ofthese disease conditions (Fig. 6 C–E), whereas the Foxp3+T-bet+subset did not change significantly even in the Th1-type Listeriainfection (Fig. 6F). Thus, our findings indicated that E-protein ac-tivity specifically regulates Foxp3+RORγt+ Treg cell differentiation.Given that E-protein activity was down-regulated by TCR stimu-lation, this finding suggests that RORγt+ effector Treg cell differ-entiation requires relatively stronger TCR stimulation than T-bet+and GATA3+ Treg cell differentiation.

    E Protein Regulates Effector Treg Cell Signature Gene Expression byDirectly Binding to the Regulatory Element of These Genes. To fur-ther investigate the mechanism by which E protein influencesgene expression in matured Treg cells, we performed genome-wide ChIP-seq assays. These showed that E2A binds to putativepromoter or enhancer regions of a host of genes, including theIcos, Itgae, Klrg1, Irf4, Rorc, GranzymeB, Il10, and Cdk6, Bcl2genes (Fig. 7A and SI Appendix, Fig. S5C). These data were thensupported by ChIP-qPCR analysis that showed that E47 bindingto genomic fragments containing promoter or conserved en-hancer sequences specific for Itgae, Irf4, Icos, Klrg1, and Rorc(Fig. 7B). In related studies we determined patterns of histonemethylation of E-protein binding sites in various genes modifiedby E-protein deletion. We found that trimethylation of histoneH3 at Lys4, a histone change consistent with active transcription,was increased in the Itgae, Irf4, Icos, Klrg1, and Rorc genes ofeffector Treg cells compared with conventional CD4+ T cells,and the trimethylation of this region was significantly increasedin corresponding genes of E protein-deficient Treg cells (Fig.7C). Taken together, these data indicate that E protein directlyregulates the expression of genes that are specific for effectorTreg cells by directly binding to the regulatory element of thesegenes and by remodeling of the chromatin at regulatory elementsof key genes affecting effector Treg cell differentiation so as torender these sites less transcriptionally active; in contrast, re-duced E-protein activity causes these sites to be more tran-scriptionally active and gives rise to differentiation of effectorTreg cells with enhanced suppressor function.

    DiscussionDespite major progress in our understanding of TCR engage-ment in the differentiation of effector Treg cells, the downstreammolecular mechanisms underlying TCR regulation of Treg cellhomeostasis and suppressive capacity is still poorly defined. Herewe demonstrate that specific deletion of E protein in maturedTreg cells (in E2Afl/flHEBfl/flFoxp3-GFP-hCre mice) resulted inTreg cells with increased stability/survival and enhanced sup-pressive function. We also showed that a large proportion ofgenes regulated by E protein was also TCR dependent, stronglysuggesting that the effects of E protein on gene regulation areparallel to or slightly amplified by TCR stimulation. Finally, weshowed with ChiP-seq analysis that E protein binds to genes

    Han et al. PNAS | March 5, 2019 | vol. 116 | no. 10 | 4477

    IMMUNOLO

    GYAND

    INFLAMMATION

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplementalhttps://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1800494116/-/DCSupplemental

  • 0 103

    104

    105

    0-10

    210

    2

    103

    104

    105

    CD103

    Foxp

    3

    **

    0

    80100

    4060

    3.0 4.5

    90.4 2.14.0 12.6

    82.4 3.0

    KLRG-1

    6.1 0.5

    93.1 0.3

    11.7 2.2

    85.6 0.5

    WT

    0

    2025

    1015

    WT

    20

    5

    CLP

    5.3 6.1

    86.3 2.3

    7.7 6.6

    82.0 3.7

    4.7 1.4

    91.9 2.0

    5.9 3.3

    81.1 9.7

    0 103

    104

    105

    0-10

    210

    2

    103

    104

    105

    5.0 17.5

    74.2 3.3

    5.8 10.2

    78.2 5.8CD103

    Foxp

    3

    KLRG-1

    19.3 6.7

    73.9 1.1

    26.3 12.8

    61.2 0.7

    WT *

    0

    4020

    100

    60

    CD

    103+

    % in

    Fo

    xp3+

    Cel

    ls K

    LRG

    -1+ %

    in

    Foxp

    3+ C

    ells

    0

    50

    2030

    WT

    10

    0 103

    104

    105

    0

    20

    40

    60

    80

    100

    ICOS

    cells

    IRF4

    - - -WT **

    WT

    80

    40sp

    inal

    cor

    d IC

    OS

    MFI

    (×10

    3 )

    10

    3

    5

    2

    4

    spin

    al c

    ord

    IRF4

    MFI

    (×10

    4 )

    1.0

    0.0

    1.5

    2.5

    0.5

    2.0

    spinal cord CD4+ Cells

    spinal cord Foxp3+ cells

    ****

    **

    0

    40

    60

    WTCD

    103+

    % in

    Fox

    p3+ C

    ells

    20

    **

    WT

    WT Id2f/fId3f/f

    18 1.9

    79.4 0.7

    9.2 0.4

    89.9 0.5

    CD103

    KLRG-1

    15.4 4.1

    79.7 0.8

    8.3 0.8

    90.3 0.6

    spleen CD4+ Cells

    0 103

    104

    105

    0-10

    210

    2

    103

    104

    105 6.9 3.0

    87.8 2.3

    9.0 3.5

    86.4 1.2

    5.0 0.7

    93.0 1.3

    5.7 1.8

    90.7 1.8

    WT

    WT

    0 104

    105

    0

    20

    40

    60

    80

    100

    IRF4

    ICOS

    - - -WT Id2fl/flId3fl/fl

    cells

    spleen Foxp3+ Cells

    Foxp

    3

    WT

    0

    34

    12

    ICO

    S M

    FI (×

    103 )

    0

    34

    12

    ICO

    S M

    FI (×

    103 )

    0

    34

    12

    ICO

    S M

    FI (×

    103 )

    0

    3

    5

    12

    4

    IRF4

    MFI

    (×10

    3 )

    0

    3

    5

    12

    4

    IRF4

    MFI

    (×10

    3 )

    0

    3

    5

    12

    4

    IRF4

    MFI

    (×10

    3 )

    **** **

    0

    40

    60

    20

    0

    3040

    2010

    KLR

    G-1

    + %

    in F

    oxp3

    + Cel

    ls

    WT0

    3040

    2010

    * *

    * *

    **

    **

    WTWT

    WT WTWT

    lung liver CLP lung liver CLP lung liver

    Foxp

    3

    0-103

    103

    104

    105

    0

    100

    200

    300

    400

    500

    0-103

    103

    104

    105

    0

    100

    200

    300

    400

    Ki67

    cells

    24h 48h 72h66.5

    80.1

    77.9

    90.2

    83.0

    95.2

    CFSE

    cells

    day 3 day 4 day 6

    8.0

    17.2

    17.3

    25.4

    50.9

    69.4

    WT

    Time (days)0 2 31 4

    0

    4

    23

    5

    Cel

    l num

    ber (

    ×105

    )

    1

    *

    *

    WT

    020

    80

    4060

    0

    30

    10

    20

    **

    Dilu

    ted

    Cel

    ls (%

    )

    0

    3040 ***

    day 6day 3 day 4***

    WT

    **

    WT

    ***

    WT

    Ki6

    7+ C

    ells

    (%)

    72h24h 48h***

    1020

    Dilu

    ted

    Cel

    ls (%

    )

    Dilu

    ted

    Cel

    ls (%

    )

    020

    100

    406080

    Ki6

    7+ C

    ells

    (%)

    020

    100

    406080

    Ki6

    7+ C

    ells

    (%)

    020

    100

    406080

    *

    0

    105

    25

    15

    CD

    103+

    % in

    Fo

    xp3+

    Cel

    lsK

    LRG

    -1+ %

    in

    Foxp

    3+ C

    ells

    0

    1510

    WT

    Id2fl/f

    l Id3fl/f

    l

    5

    20**

    WTWT WT

    0 103

    104

    105

    0

    103

    104

    105

    WT

    CD

    103+

    % in

    Fox

    p3+ C

    ells

    CD

    103+

    % in

    Fox

    p3+ C

    ells

    KLR

    G-1

    + %

    in F

    oxp3

    + Cel

    ls

    KLR

    G-1

    + %

    in F

    oxp3

    + Cel

    ls

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/flE2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    WT

    A

    D

    G

    I J

    H

    E F

    B C

    Fig. 5. E-protein activity regulates effector Treg cell differentiation, trafficking, proliferation, and survival. (A) Cells of indicated organs from WTFoxp3Cre

    and E2Afl/flHEBfl/flFoxp3Cre mice were stained directly with CD4, Foxp3, CD103, and KLRG-1. Dot plots were gated on CD4+ cells. (B) Frequencies of CD103+

    (Top) and KLRG-1+ (Bottom) cells in Foxp3+ cells in indicated organs from WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice. (C) Mean fluorescence of ICOS (Top)and IRF4 (Bottom) on Foxp3+ cells in indicated organs of WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre mice. (D and E) Mixed BM chimeric mice were generated bytransferring BM cells from CD45.1+CD45.2+ WT Foxp3Cre and CD45.1+ E2Afl/flHEBfl/flFoxp3Cre mice into lethally irradiated recipient mice. (D) Flow cytometricanalysis of CD103, KLRG-1, and Foxp3 expression among CD4+ cells (Left) and the frequencies of CD103+ (Right Top) and KLRG-1+ (Right Bottom) cells amongCD45.1+CD45.2+ WT Foxp3Cre and CD45.1+ E2Afl/flHEBfl/flFoxp3Cre Foxp3+ cells in the spinal cord of BM chimeric mice 24 d after EAE induction. (E) Flowcytometric analysis of ICOS and IRF4 expression (Left) and the mean fluorescence of ICOS (Right Top) and IRF4 (Right Bottom) among CD45.1+CD45.2+ WTFoxp3Cre and CD45.1+ E2Afl/flHEBfl/flFoxp3Cre Treg cells in the spinal cord of BM chimeric mice 24 d after EAE induction. (F and G) Mixed BM chimeric mice weregenerated by transferring BM cells from WT Foxp3Cre and Id2fl/flId3fl/flFoxp3Cre mice into lethally irradiated recipient mice. (F ) Flow cytometric analysis ofCD103, KLRG-1, and Foxp3 expression among CD4+ cells (Left) and the frequencies of CD103+ (Right Top) and KLRG-1+ (Right Bottom) cells among WTFoxp3Cre and Id2fl/flId3fl/flFoxp3Cre Foxp3+ cells in the spleen of the mixed BM chimeric mice. (G) Flow cytometric analysis of ICOS and IRF4 expression ofWT Foxp3Cre and Id2fl/flId3fl/flFoxp3Cre Foxp3+ cells in the spleen of the mixed BM chimeric mice. (H–J) Highly purified Treg cells from WT Foxp3CreR26YFP

    and E2Afl/flHEBfl/flFoxp3CreR26YFP mice were (H) labeled with CFSE and stimulated with anti-CD3 plus anti-CD28 in the presence of human IL-2 for theindicated time. CFSE dilution in the Treg cells at the indicated time points were analyzed by flow cytometry (Left) and the frequencies of CFSE-diluted cells(Right); or (I) Ki67+ in Foxp3+ cells at the indicated time (Left) and the frequencies of Ki67+ in Foxp3+ cells (Right). (J) Total cell numbers at the indicatedtime points were counted separately. (A, G and D, E, F, H, and I, Left) Data are representative of at least three independent experiments. (B, C, J and D, E,F, H, and I, Right) Data are pooled from at least three independent experiments, each symbol represents data from one mouse, mean ± SD are shown(*P < 0.05, **P < 0.01, ***P < 0.001).

    4478 | www.pnas.org/cgi/doi/10.1073/pnas.1800494116 Han et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

    https://www.pnas.org/cgi/doi/10.1073/pnas.1800494116

  • involved in Treg cell effector function and that decreased bind-ing (resulting from down-regulation) leads to enhanced expressionof these genes. Thus, TCR signaling regulates Treg cell functionby releasing these genes from the negative effects of E protein.Regarding the specific effects of E proteins, the above-

    mentioned ChIP-seq studies showed that before down-regulationby TCR signaling, E proteins bind to a large number of the genesthat are associated with effector Treg cell differentiation (Irf4,Rorc), localization (Itgae and Ccr5, Ccr2, etc.), function (Icos,Lag3, Il10, and GranzymeB), survival (Bcl2), and proliferation(Cdk6); in addition, E proteins bind to the gene encoding Klrg1, amarker of mature or terminally differentiated cells, including Tregcells. In studies probing the effects of E-protein binding we foundthat histone changes indicative of active gene transcription (tri-methylation at Lys4 of H3) were present in many of the abovegenes following E-protein down-regulation. Thus, the picture thatemerges is that E proteins have a potent capacity to suppresstranscription of those genes necessary for effector Treg cellfunction. Of particular interest in this context is that IRF4 isdramatically up-regulated during inflammation in E protein-deficient Treg cells, indicating that E protein negatively regulatesthe expression IRF4. This observation is significant becauseIRF4 is a TCR-induced gene that has been shown to be criticalfor the optimal function of effector Treg cells and to be a mediatorof effector Treg cell differentiation; thus, this finding greatlystrengthens the link between Treg cell differentiation and TCR-regulated E-protein activity.

    E-protein regulation of Treg cells is pertinent to the genera-tion of RORγt+Foxp3+ cells, a functionally distinct Treg cellsubset that has been shown to suppress Th17 responses in EAE(22). In the present study we showed that RORγt+ Foxp3+ Tregcells were significantly increased in mice with E protein-deficientTreg cells, both in the steady state and in inflammatory states. Inaddition, we found that the gene expression patterns of Eprotein-deficient Treg cells are very similar to that of RORγt+Foxp3+ cells found in the intestine and in immunization-inducedRORγt+ Foxp3+cells (22) since in all cases one sees up-regulationof the Icos, Itgae, Rorc, Il1r1, Il23r, Havrc2, and Il10 genes. Finally,E protein-deficient Treg cells exhibited enhanced suppressive ac-tivity and stability, also in line with characteristics of previouslydescribed RORγt+ Treg cells. These findings suggest that E pro-tein is a key regulator of the RORγt+ Treg cell subset and that thissubset arises when Tregs are subjected to a sufficient and possiblyhigher level of TCR signaling and E-protein deletion that allowstranscription of the RORγt gene.Recent studies of mice with Id2/Id3 deficiency in Foxp3+ Treg

    cells provide data in some ways parallel to those obtained in thisstudy in that these mice also manifest E-protein abnormalities(16). However, in this case increased E-protein function resultsin decreased Treg cell stability and in vivo suppressive function,the latter manifesting as spontaneous inflammation of the lungs,eyelids, skin, and esophagus (16). These changes at first glancesuggest that Id2/Id3 deletion provides a mirror image ofE-protein deletions and thereby predicts the effects of E-proteindeletion. However, this is probably not the case for severalreasons: first, Id2/Id3 deletion impacts mainly on regulation of Bcell/Th2 responses and results in allergic manifestations; incontrast, E-protein deletion has a more general effect on Tregcell suppressive function and leads to enhanced suppression ofTh1/Th17-mediated inflammation in EAE and transfer colitis;

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    RORγt

    Foxp

    3

    CLP

    WT

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    RORγt

    Foxp

    3

    spleen

    WT

    CLP11.9 0.9

    82.4 4.8

    14.6 2.1

    76.8 4.5

    7.5 6.5

    77.9 8.1

    5.5 8.9

    76.6 9.0

    5.1 6.4

    79.5 9.0

    6.4 9.1

    75.3 9.20-10

    310

    310

    410

    5

    0-10

    210

    2

    103

    104

    105

    spleen lung liver9.1 0.6

    88.0 2.3

    12.9 1.2

    82.5 3.4

    4.2 0.5

    92.3 3.0

    7.4 1.5

    83.4 7.7

    4.8 1.1

    90.8 3.18.9 2.9

    82.9 5.3

    05

    10

    20

    0

    10

    20

    30

    0

    10

    20

    30

    0WT

    RORγt

    Foxp

    3

    dLN

    WT

    6080

    2040

    WT WT WT

    15

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    * ** *CLPspleen lung liver

    0

    101520

    0

    6080

    2040

    WT WT

    ***CLPspleen

    RORγt

    Foxp

    3

    WT

    lung liver4.2 0.4

    92.4 3.0

    7.4 1.5

    83.4 7.7

    4.4 0.8

    91.7 3.18.9 2.9

    84.2 5.30

    1020

    40

    0

    10

    20

    30

    WT WT

    ***lung liver

    0 103

    104

    105

    0

    -103

    103

    104

    105

    3.0

    6.505

    10

    2015

    WT

    dLN*

    Tbet

    Foxp

    3

    5.7 1.0

    74.5 18.813.2 1.5

    82.3 13.0

    6.1 2.2

    57.134.618.5 4.6

    49.5.28.4

    30

    05

    10

    2015

    T-be

    t+ %

    in F

    oxp3

    + Cel

    ls

    01020

    50

    3040

    T-be

    t+ %

    in F

    oxp3

    + Cel

    ls

    WT WT

    5

    lung liver lung livernsns

    *

    0 103

    104

    105

    100

    101

    102

    103

    104

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    RO

    Rγt

    + %

    in F

    oxp3

    + Cel

    ls

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/flHEBfl/fl

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/f

    l HEBfl/f

    l

    E2Afl/flHEBfl/fl

    A B

    C D

    E F

    Fig. 6. Foxp3+RORγt+ but not Foxp3+Tbet+ or Foxp3+GATA3+ effector Tregcell subset is regulated by E protein. (A and B) Flow cytometric analysis ofRORγt and Foxp3 expression in cells from indicated organs from WTFoxp3Cre

    and E2Afl/flHEBfl/flFoxp3Cre mice (A) and the frequencies of RORγt+ cellswithin Treg cells (B). (C) Flow cytometric analysis of RORγt expression in Foxp3+

    cells in the draining lymph node from WTFoxp3Cre and E2Afl/flHEBfl/flFoxp3Cre

    mice 21 d after EAE induction (Left) and the frequency of RORγt+ cellswithin these Treg cells (Right). (D) DSS-colitis was induced in WTFoxp3Cre

    and E2Afl/flHEBfl/flFoxp3Cre mice; flow cytometric analysis of RORγt andFoxp3 expression in indicated organs (Left) and the frequencies of RORγt+

    cells in Treg cells 10 d after colitis induction (Right). (E and F) WTFoxp3Cre andE2Afl/flHEBfl/flFoxp3Cre mice were infected by L. monocytogenes; (E) flow cyto-metric analysis of RORγt and Foxp3 expression and the frequencies of RORγt+

    cells in isolated lung and liver Treg cells (Right). (F) Flow cytometric analysis ofT-bet and Foxp3 expression (Left) and the frequencies of T-bet+ cells among Tregcells (Right). (A and C–F, Left) Data are representative of at least three in-dependent experiments. (B and C–F, Right) Data are pooled from at least threeindependent experiments; each symbol represents data from one mouse,mean ± SD were shown (ns, not significant, *P < 0.05, **P < 0.01).

    A

    B

    C

    Fig. 7. E protein regulates effector Treg cell signature gene expression bydirectly binding to the regulatory element of these genes. (A) Thymocyteswere isolated from WT mice; E47 ChIP was performed and subjected to li-brary preparation and sequencing as described in Methods. Representativeof ChIP sequencing tracks showing composite binding of E2A to Itgae, Icos,Irf4, Klrg1, and Rorc genes. (B) Chromatin from thymocytes were subjectedto immunoprecipitation with anti-E47. Binding of E proteins to the indicatedgenes was analyzed by quantitative real-time PCR. Results are representativeof at least three independent experiments; mean ± SD are shown. (C) Chro-matin from purified naive CD4+ cells, WT Treg cells, and E protein-deficientTreg cells were subjected to immunoprecipitation with anti-H3K4. Bindingof H3K4 to the indicated genes was analyzed by quantitative real-time PCR.Results are representative of two independent experiments; mean ± SD areshown (*P < 0.05, **P < 0.01, ***P < 0.001).

    Han et al. PNAS | March 5, 2019 | vol. 116 | no. 10 | 4479

    IMMUNOLO

    GYAND

    INFLAMMATION

    Dow

    nloa

    ded

    by g

    uest

    on

    June

    4, 2

    021

  • second, because deletion of Id2/Id3 in matured Treg cells leadsto spontaneous autoimmune disease, the apparent effect of Id2/Id3 on Treg cell function derived from deletion studies may inreality be secondary to inflammatory effects of such deletion;third, TCR stimulation of Id protein-deleted cells results in bothdown-regulation of Id and E-protein expression rather than areciprocal change that one might expect in the case of opposingfunctions. These various differences between the effects of Id2/Id3 and E-protein deletion are likely to arise from the fact that atthe molecular level there does not exist a strict point/counter-point relation between these sets of transcriptional regulatoryfactors. For this reason, it is fair to say that direct deletion ofE protein provides a better picture of E-protein effects on Tregcells than the indirect effects of Id protein deletion.Prior studies have revealed that E-protein expression regulates

    cell proliferation and survival (apoptosis) (31). This correlateswith the results of RNA-seq analyses performed in this study thatshowed that genes involved in the “cell proliferation and cycling”such as Bcl2, Cdk6, and Cdk14 genes, were up-regulated in Eprotein-deficient Treg cells. In addition, the latter cells exhibitedincreased proliferative responses in vitro upon TCR stimulation,which most likely contributed to the increased percentage ofeffector Treg cells noted in E2Afl/flHEBfl/flFoxp3-GFP-hCre micein vivo. These findings correlated with the results of ChIP-seqanalysis that revealed that genes involved in cell cycling exhibiteda high level of E-protein binding, including the Bcl2 gene andcycle promoter gene, Cdk6; it is thus likely that the increasedproliferation and up-regulated expression of cell cycle geneswere also a major functional consequence of E-protein loss inTreg cells.In summary, these studies provide strong evidence that

    E protein regulates a large set of key effector Treg cell signaturegenes that govern the differentiation, effector function, proliferation,and survival of already established Treg cells. In as much as E-proteindown-regulation is largely under TCR control, these findings indicate

    that the molecular mechanism of TCR regulation of Treg cell dif-ferentiation is in fact its ability to control E-protein activity. Thus, Eprotein plays a stage-dependent role in Treg cells, whereby it is notonly indispensable for their development in the thymus as demon-strated previously (15), but also essential for their further differenti-ation into effector Treg cells in the periphery. On this basis, theunderlying regulatory circuits governing E-protein levels are likely toprovide insight into how Treg cells maintain their essential identityand at the same time exhibit plasticity in various tissue settings.

    MethodsMice. WT CD45.1 mice were obtained from The Jackson Laboratory. Tcf3(E2A)fl/flTcf12(HEB)fl/fl mice, Id2fl/flId3fl/fl mice, and Foxp3-GFPKI mice weregifts from Y. Zhuang, Duke University, Durham, NC, and from M. Oukka,Brigham and Women’s Hospital, Boston, MA, respectively. Foxp3Cre/Rosa26YFP

    mice were a gift from Xuyu Zhou, Chinese Academy of Sciences, Beijing, China.Strain-matched WT Foxp3Cre mice served as controls. E2Afl/flHEBfl/fl mice werecrossed with Foxp3-GFP-KI mice and Foxp3Cre/Rosa26YFP mice to generateE2Afl/flHEBfl/fl Foxp3-GFP-KI/Foxp3Cre/Rosa26YFP mice. The study was approvedby the Research Ethics Committee of the Institute of Microbiology, ChineseAcademy of Sciences (IMCAS), the permit number was APIMCAS2017015.

    Flow Cytometry and Cell Sorting. Flow cytometry was performed on aFACSCalibur or FACS CantoII (BD Biosciences) and the data were analyzed byFlowJo. Cell sorting was performed using a FACS AriaIII (BD). Fluorochrome-conjugated Abs are provided in SI Appendix.

    An extended methods section is provided in SI Appendix, SI Materialsand Methods.

    ACKNOWLEDGMENTS. We thank Prof. Y. Zhuang (Duke University MedicalCenter, Durham, NC) for providing E2Af/fHEBf/f and Id2f/fId3f/f mice. Thisstudy was supported by Strategic Priority Research Program of the ChineseAcademy of Sciences (Grant XDB29010000), the National Natural ScienceFoundation of China (Grant 31670894) and the National Key Research andDevelopment Project of China (Grant 2016YFC1200302).

    1. Sakaguchi S, Yamaguchi T, Nomura T, Ono M (2008) Regulatory T cells and immunetolerance. Cell 133:775–787.

    2. Curotto de Lafaille MA, Lafaille JJ (2009) Natural and adaptive foxp3+ regulatoryT cells: More of the same or a division of labor? Immunity 30:626–635.

    3. Campbell DJ, Koch MA (2011) Phenotypical and functional specialization of FOXP3+regulatory T cells. Nat Rev Immunol 11:119–130.

    4. Lee JH, Kang SG, Kim CH (2007) FoxP3+ T cells undergo conventional first switch tolymphoid tissue homing receptors in thymus but accelerated second switch to non-lymphoid tissue homing receptors in secondary lymphoid tissues. J Immunol 178:301–311.

    5. Panduro M, Benoist C, Mathis D (2016) Tissue Tregs. Annu Rev Immunol 34:609–633.6. Kleinewietfeld M, et al. (2005) CCR6 expression defines regulatory effector/memory-

    like cells within the CD25(+)CD4+ T-cell subset. Blood 105:2877–2886.7. Huehn J, et al. (2004) Developmental stage, phenotype, and migration distinguish

    naive- and effector/memory-like CD4+ regulatory T cells. J Exp Med 199:303–313.8. Suffia I, Reckling SK, Salay G, Belkaid Y (2005) A role for CD103 in the retention of

    CD4+CD25+ Treg and control of Leishmania major infection. J Immunol 174:5444–5455.9. Beyersdorf N, Ding X, Tietze JK, Hanke T (2007) Characterization of mouse CD4 T cell

    subsets defined by expression of KLRG1. Eur J Immunol 37:3445–3454.10. Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS (2012) A broad range of self-

    reactivity drives thymic regulatory T cell selection to limit responses to self. Immunity37:475–486.

    11. Vahl JC, et al. (2014) Continuous T cell receptor signals maintain a functional regu-latory T cell pool. Immunity 41:722–736.

    12. Levine AG, Arvey A, Jin W, Rudensky AY (2014) Continuous requirement for the TCRin regulatory T cell function. Nat Immunol 15:1070–1078.

    13. Murre C, McCaw PS, Baltimore D (1989) A new DNA binding and dimerization motif inimmunoglobulin enhancer binding, daughterless, MyoD, and myc proteins. Cell 56:777–783.

    14. Maruyama T, et al. (2011) Control of the differentiation of regulatory T cells andT(H)17 cells by the DNA-binding inhibitor Id3. Nat Immunol 12:86–95.

    15. Gao P, et al. (2014) Dynamic changes in E-protein activity regulate T reg cell devel-opment. J Exp Med 211:2651–2668.

    16. Miyazaki M, et al. (2014) Id2 and Id3 maintain the regulatory T cell pool to suppressinflammatory disease. Nat Immunol 15:767–776.

    17. Zhou X, et al. (2008) Selective miRNA disruption in T reg cells leads to uncontrolledautoimmunity. J Exp Med 205:1983–1991.

    18. Zhou X, et al. (2009) Instability of the transcription factor Foxp3 leads to the gener-ation of pathogenic memory T cells in vivo. Nat Immunol 10:1000–1007.

    19. Thornton AM, et al. (2010) Expression of Helios, an Ikaros transcription factor familymember, differentiates thymic-derived from peripherally induced Foxp3+ T regula-tory cells. J Immunol 184:3433–3441.

    20. Sefik E, et al. (2015) MUCOSAL IMMUNOLOGY. Individual intestinal symbionts inducea distinct population of RORγ+ regulatory T cells. Science 349:993–997.

    21. Yang BH, et al. (2016) Foxp3(+) T cells expressing RORγt represent a stable regulatoryT-cell effector lineage with enhanced suppressive capacity during intestinal inflam-mation. Mucosal Immunol 9:444–457.

    22. Kim BS, et al. (2017) Generation of RORγt+ antigen-specific T regulatory 17 cells fromFoxp3+ precursors in autoimmunity. Cell Rep 21:195–207.

    23. Setoguchi R, Hori S, Takahashi T, Sakaguchi S (2005) Homeostatic maintenance ofnatural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and inductionof autoimmune disease by IL-2 neutralization. J Exp Med 201:723–735.

    24. Zheng Y, et al. (2009) Regulatory T-cell suppressor program co-opts transcriptionfactor IRF4 to control T(H)2 responses. Nature 458:351–356.

    25. Liston A, Gray DH (2014) Homeostatic control of regulatory T cell diversity. Nat RevImmunol 14:154–165.

    26. Wohlfert EA, et al. (2011) GATA3 controls Foxp3+ regulatory T cell fate during in-flammation in mice. J Clin Invest 121:4503–4515.

    27. Wang Y, Su MA, Wan YY (2011) An essential role of the transcription factor GATA-3 for the function of regulatory T cells. Immunity 35:337–348.

    28. Yu F, Sharma S, Edwards J, Feigenbaum L, Zhu J (2015) Dynamic expression of tran-scription factors T-bet and GATA-3 by regulatory T cells maintains immunotolerance.Nat Immunol 16:197–206.

    29. Koch MA, et al. (2009) The transcription factor T-bet controls regulatory T cell ho-meostasis and function during type 1 inflammation. Nat Immunol 10:595–602.

    30. Levine AG, et al. (2017) Stability and function of regulatory T cell