Cutting Edge: The Histone Methyltransferase G9a Is Required for Silencing of Helper T L ineage−Associated in Proliferating CD8 T Cells This information is current as of September 27, 2021. Daniel J. Verbaro, Nagisa Sakurai, Byungil Kim, Yoichi Shinkai and Takeshi Egawa J Immunol published online 2 May 2018 http://www.jimmunol.org/content/early/2018/05/02/jimmun ol.1701700 Downloaded from

Supplementary http://www.jimmunol.org/content/suppl/2018/05/02/jimmunol.170170

Material 0.DCSupplemental http://www.jimmunol.org/

Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication by guest on September 27, 2021

*average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2018 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published May 2, 2018, doi:10.4049/jimmunol.1701700 Th eJournal of Cutting Edge Immunology

Cutting Edge: The Histone Methyltransferase G9a Is Required for Silencing of Helper T Lineage–Associated Genes in Proliferating CD8 T Cells Daniel J. Verbaro,* Nagisa Sakurai,* Byungil Kim,*,1 Yoichi Shinkai,† and Takeshi Egawa* Helper versus cytotoxic T lineage decision in the thy- MHC class II or MHC class I and differentiate into cells in mus has been studied as a model for silencing of alter- the helper or cytotoxic T lineages, respectively. MHC class I– native lineage genes. Although the factor selected thymocytes express the RUNX3 RUNX3 is required for the initiation of Cd4 silencing that establishes the silencing of helper T lineage genes, in-

in developing CD8 T cells, it is unknown how silenc- cluding Cd4 and Zbtb7b (10–14). Downloaded from ing of Cd4 and other helper T lineage genes is main- However, it is poorly understood how helper T lineage– tained. We show that the histone methyltransferase associated genes are heritably silenced in mature cytotoxic T cells. During thymocyte development, Cd4 is transiently G9a is necessary for silencing helper T lineage genes 2 repressed by RUNX1, an ortholog of RUNX3, in CD4 in proliferating mouse CD8 T cells. Despite normal 2 initial Cd4 downregulation, G9a-deficient CD8 T cells CD8 double-negative thymocytes via direct binding to the derepress Cd4 and other helper lineage genes during silencer cis-element in the locus (11). This repression is sub- http://www.jimmunol.org/ repeated division in lymphopenia or in response to sequently reversed upon selection of a successfully rearranged tumor Ag. However, G9a was dispensable for contin- Tcrb locus (15). Cd4 is expressed uniformly in DP thymo- ued silencing of those genes in CD8 T cells that re- cytes that give rise to helper and cytotoxic T lineage cells after positive selection. Whereas Cd4 continues to be expressed in spond to infection by Listeria monocytogenes. These helper lineage T cells, CD8+ cytotoxic T cells terminate Cd4 results demonstrate that G9a facilitates maintenance transcription by upregulating RUNX3, which binds the of cellular identity of CD8 T cells during cell division, identical cis-element as RUNX1 (11). Deletion of the silencer which is further reinforced by inflammatory signals. The element or disrupting RUNX binding sites in the silencer by guest on September 27, 2021 Journal of Immunology, 2018, 200: 000–000. results in continued Cd4 expression in CD8 T cells (16, 17). However, deletion of the silencer or Runx3 in differentiated uring a binary fate decision, genes related to the CD8 T cells does not reactivate Cd4, indicating that the opposing lineage are heritably silenced (1, 2). This initial repression but not maintenance of Cd4 silencing re- D silencing is achieved through the constitutive activity quires RUNX3 (16, 18). RUNX3 may therefore recruit epi- of transcription factors involved in the lineage-determining genetic modifications to the Cd4 locus and loci encoding process or by recruitment of epigenetic machinery in a helper lineage genes, which are maintained independently of locus-specific manner, presumably by those lineage-specific RUNX3. Because the Cd4 locus is only reversibly repressed in transcription factors. The differentiation of the common double-negative thymocytes but irreversibly silenced in CD8 thymocyte precursor to the helper or the cytotoxic T cell T cells, we hypothesized that the irreversible silencing is lineage in the thymus has been studied to understand the mediated by epigenetic modifiers that specifically interact requirements for transcription factors and epigenetic with RUNX3 but not RUNX1. regulation for stable lineage decisions (1–9). CD4+CD8+ In this study, we identified the histone methyltransferase double-positive (DP) thymocytes are subjected to positive (HMT) G9a as an epigenetic modifier that preferentially in- selection of rearranged TCRab by self-peptides presented on teracts with RUNX3 to RUNX1 and is necessary for continued

*Department of Pathology and Immunology, Washington University School of Medi- the National Center for Biotechnology Information’s Omnibus (https://www. cine, St. Louis, MO 63110; and †Cellular Memory Laboratory, RIKEN, Wako, Saitama ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE101730) under accession number GSE101730. 351-0198, Japan Address correspondence and reprint requests to Dr. Takeshi Egawa, Washington Uni- 1Current address: Department of Biochemistry, Vanderbilt University, Nashville, TN. versity School of Medicine, 660 South Euclid Avenue, Box 8118, St. Louis, MO 63110. E-mail address: [email protected] ORCIDs: 0000-0001-5624-2488 (D.J.V.); 0000-0001-7489-1051 (T.E.). The online version of this article contains supplemental material. Received for publication December 11, 2017. Accepted for publication April 11, 2018. Abbreviations used in this article: DP, double-positive; FLAG-G9a, FLAG-tagged G9a This work was supported by National Institutes of Health Grants R01AI097244, retrovirus; H3K9me2, dimethylation of histone 3 lysine 9; H3K9me3, trimethylation R56AI114593 (to T.E.), and T32GM007200 (to D.J.V.), by the Leukemia and Lym- of histone 3 lysine 9; HMT, histone methyltransferase; Lm-OVA, L. monocytogenes phoma Society Scholar award (to T.E.), and by the Shawn Hu and Angela Zeng expressing OVA; RNA-seq, RNA sequencing. Fellowship (to D.J.V.). The mass spectrometry data presented in this article have been submitted to PRIDE (https:// Copyright Ó 2018 by The American Association of Immunologists, Inc. 0022-1767/18/$35.00 www.ebi.ac.uk/pride/) under accession number PXD007511. The RNA sequencing data and chromatin immunoprecipitation sequencing data presented in this article have been submitted to

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1701700 2 CUTTING EDGE: SILENCING OF HELPER LINEAGE GENES IN CD8 T CELLS BY G9a

2 2 2 2 silencing of helper lineage genes in dividing CD8 T cells under T cells purified from Tcrb / Tcrd / recipient mice 4 wk after transfer. noninflammatory conditions. G9a deficiency resulted in de- Sequenced tags were mapped to the mouse genome mm9 using TopHat (25) with default setting, followed by transcript assembly and estimation of ex- repression of several genes, which are otherwise expressed only pression levels using Cufflinks (26–29) on Galaxy (https://usegalaxy.org/). in CD4 T cells, although it was compensated for by the in- flammatory cytokine IL-12. These results suggest that G9a and Chromatin immunoprecipitation inflammatory cues cooperatively maintain the identity of CD8 Mononucleosomes were prepared from cultured CD8 T cells by micrococcal T cells during their division. nuclease digestion as described (30). Trimethylation of histone 3 lysine 9 (H3K9me3)–modified nucleosomes were immunoprecipitated using anti- H3K9me3 (8898; Abcam) conjugated with G magnetic beads (Life Materials and Methods Technologies). For genome-wide analysis, purified DNA from precipitated Mice nucleosomes was sequenced with a HiSeq 2500 sequencer (Illumina) with a 50-bp single end read option as described (23). C57BL/6N and B6-CD45.1 mice were purchased from Charles River Lab- oratories. OT-I mice (19) were purchased from Taconic. Cd4-cre (10) and Statistical analysis CD8-cre (E8I-cre) (20) mice were obtained from D. Littman (New York University). Ehmt2-flox mice were previously described (21). All mice were All statistics were performed using Graphpad Prism (version 7.0) using generated in or have been backcrossed more than eight times to C57BL/6 nonparametric two-tailed Student t tests for comparing two groups. Multiple 2 mice. Unless otherwise specified, littermate cre+ Ehmt2+/+ or cre Ehmt2F/F groups were analyzed using two-tailed ANOVA. All other statistics were were used as control. All mice were maintained in the specific pathogen-free performed as described in the manuscript. Statistical analyses are shown with facility at Washington University School of Medicine. All experiments were the mean 6 SD. A p value , 0.05 was considered significant. conducted following a protocol approved by the Washington University Animal Studies Committee. Results and Discussion Downloaded from Coimmunoprecipitation G9a forms a complex with RUNX3 1200M and AKR1 cell lines were transduced with MSCV-based retrovirus as To identify candidate epigenetic modifiers that are recruited described (22). For interactome analyses, 1200M cells in which endogenous by RUNX3, we analyzed RUNX1 and RUNX3 interactomes Runx1 expression had been knocked down (22) were transduced with in 1200M thymoma cells, which have active Cd4 silencing FLAG-hemagglutinin–tagged RUNX1 or RUNX3. RUNX1- and RUNX3- interacting in nuclear extract were immunoprecipitated with anti-FLAG machinery (15). Among 71 DNA-binding proteins and epi- http://www.jimmunol.org/ beads (M2; Sigma-Aldrich), eluted with 33 FLAG peptide (GenScript), genetic modifiers identified in RUNX1 or RUNX3 inter- andanalyzedbymassspectrometryattheTaplinMassSpectrometryFacility actomes, all three components of the G9a HMT complex, at Harvard University. For analytical immunoprecipitation, nuclear proteins G9a, GLP, and WIZ, as well as a known G9a-interacting were extracted from AKR1 cells that were transduced with RUNX1, RUNX3, RUNX1, and FLAG-tagged G9a retrovirus (FLAG-G9a), or RUNX3 and transcription factor CUX1 (31) were found predominantly FLAG-G9a. Immune complexes containing FLAG-tagged protein were pre- in the RUNX3 interactome (Fig. 1A, 1B). Preferential in- cipitated with anti-FLAG, followed by immunoblotting using anti-FLAG and teraction between G9a and RUNX3 compared with RUNX1 anti–pan-RUNX Abs (12). was confirmed by coimmunoprecipitation against FLAG-G9a Flow cytometry by guest on September 27, 2021 The following mAbs were purchased from BioLegend: allophycocyanin- conjugated anti-CD62L (MEL-14); allophycocyanin-Cy7–conjugated CD45.2 (104); FITC-conjugated anti-CD62L (MEL-14), anti-Va2 (B20.1); Pacific Blue–conjugated anti-CD44 (IM7); PE-conjugated anti-Vb5(MR9-4), anti–IFN-g (XMG1.2); PE-Cy7–conjugated anti-CD8a (53.6.7); PerCP- Cy5.5–conjugated anti-CD4 (GK1.5), anti-CD90.1 (OX-7). Cells were analyzed with an LSR II or an LSRFortessa or sorted with a FACSAria II (BD Biosciences). Dead cells were excluded by staining with DAPI (Sigma- Aldrich) or Aqua Live/Dead (Life Technologies). Data were analyzed on FlowJo software (Tree Star). T cell transfer, tumor inoculation, and Listeria monocytogenes infection Naive CD8 cells from OT-I TCR transgenic mice were sorted by flow 2 2 cytometry as Va2+Vb5+CD62L+CD44lo/ CD4 CD8+ cells. Cells (2 3 105) 2 2 2 2 were transferred i.v. to Tcrb / Tcrd / mice. PBMCs and splenocytes were isolated from the recipient mice 4 wk after transfer and analyzed for surface marker and gene expression. For experiments with transplanted tumors, 1 3 106 E.G7-OVA (no. CRL-2113; American Type Culture Collection) cells were injected s.c. in the flank of B6-CD45.1 mice. Five days later, 1 3 106 OT-I cells were transferred i.v. to the tumor-bearing mice followed by analysis of T cells collected from the draining lymph node 7 d later. For L. mono- cytogenes infection, 5 3 103 OT-I cells were transferred i.v. into B6-CD45.1 mice, which were infected i.v. with 2 3 104 CFU of L. monocytogenes expressing OVA (Lm-OVA) on the next day as described (23).

Quantitative RT-PCR FIGURE 1. G9a preferentially forms a complex with RUNX3 to RUNX1. A Total RNA was purified using TRIzol (Life Technologies) and reverse tran- ( ) Heat map showing the 71 DNA-interacting proteins that were coim- scribed using qScript (QuantaBio). Gene expression was quantitated using a munoprecipitated with only RUNX3 (cluster 1), RUNX1 and RUNX3 Luminaris SYBR Green reagent (Thermo Fisher) and a Roche LightCycler (cluster 2), or only RUNX1 (cluster 3) from transduced 1200M cells. (B) List 480. Primer sequences are listed in Supplemental Table I. of the transcription factors, DNA methyltransferases, and histone-modifying enzymes in (A). (C) Immunoblotting (IB) for RUNX proteins coimmuno- RNA sequencing precipitated with FLAG-G9a from lysates of AKR1 cells transduced RNA sequencing (RNA-seq) was done essentially as described previously (24) with RUNX1, RUNX3, or FLAG-G9a. Blots are representative of two 2 2 using total RNA extracted from 5 3 104 Ehmt2 / and Ehmt2+/+ Va2+CD8+ experiments. The Journal of Immunology 3 and immunoblotting with anti–pan-RUNX Ab that de- detectable in CD8+ mature thymocytes from Ehmt2F/F Cd4- 2 2 tects both RUNX1 and RUNX3 (Fig. 1C). These data cre mice (data not shown) (referred to as Ehmt2 / CD8 show that G9a and RUNX3 form a complex, which may T cells hereafter). As previously reported using pLck-cre (32), deliver the HMT activity to Cd4 and other helper lineage– numbers and frequencies of total thymocytes and CD4+ and 2 2 related genes repressed by RUNX3 in developing CD8 CD8+ splenic T cells were comparable between Ehmt2 / T cells. and control Ehmt2+/+ mice (data not shown). Different from 2 2 the reported phenotype in Runx3 / thymocytes (11, 14), G9a is required for silencing of helper lineage–associated genes in CD4 was normally downregulated in mature CD8 thymo- proliferating CD8 T cells in vivo under noninflammatory conditions cytes and was not expressed in splenic naive or memory CD8 To define the role of G9a in CD8 T cells, we conditionally T cells under steady-state conditions (data not shown). inactivated Ehmt2, encoding G9a, at the DP stage of thy- To determine whether G9a is required for maintaining Cd4 mocyte development using Cd4-cre. Ehmt2 mRNA was barely silencing during cell division, we adoptively transferred Downloaded from http://www.jimmunol.org/

FIGURE 2. G9a is required to maintain silencing of helper lineage genes in CD8 T cells during lymphopenia- or tumor Ag–driven proliferation. (A and B) CD4 expression and 2 2 2 2 CFSE dilution of CD8 T cells in PBMC of Tcrb / Tcrd / 2 2 mice that received Ehmt2 / or Ehmt2+/+ OT-I T cells 4 wk prior to the analysis. Data are pooled from three experiments by guest on September 27, 2021 in which one donor of each genotype was transferred into two to three recipients. (C and D) RNA-seq analysis of CD4+ 2 2 2 2 2 2 CD8+ Ehmt2 / , CD4 CD8+ Ehmt2 / , and CD4 CD8+ 2 2 2 2 Ehmt2+/+ OT-I T cells harvested from Tcrb / Tcrd / mice 4 wk after transfer. Quantification of genes with $1 frag- 2 2 ments per kb per million in Ehmt2 / or Ehmt2+/+ samples and .2-fold difference in expression is indicated for each genotype. Dashed red lines indicate 2-fold change between genotypes. (E) Heat maps showing genes differentially 2 2 2 expressed between CD4+CD8+ or CD4 CD8+ Ehmt2 / +/+ and control Ehmt2 CD8 T cells. Values represent the log2 fold change of the mean of two to four mice compared with Ehmt2+/+ CD8 T cells. Highlighted genes represent genes differentially expressed between CD4 and CD8 memory T cells from Immunological Genome Project datasets. (F and G) Expression of CD4 of OT-I T cells in the lymph node draining transplanted E.G7-OVA tumors. n = 6–8 in two experiments. 4 CUTTING EDGE: SILENCING OF HELPER LINEAGE GENES IN CD8 T CELLS BY G9a

2 2 2 Ehmt2 / or control Ehmt2+/+ naive CD4 CD8+ T cells in the Immunological Genome Project datasets (.1.8-fold 2 2 2 2 expressing the OT-I TCR transgene into Tcrb / Tcrd / difference), 92 genes that are more highly expressed in CD4 mice, in which donor-derived CD8 T cells divide under T cells, including Cd4, Foxp3, Cd40lg, Rorc, Rora, Zbtb7b, 2 2 noninflammatory conditions. By 4 wk after transfer, both and Il21, were derepressed in the Ehmt2 / CD4+CD8+ 2 2 Ehmt2 / CD8 T cells expanded at similar rates if not faster T cells (Fig. 2E). We also observed downregulation of 18 than control Ehmt2+/+ cells as determined by CFSE, and they genes that are more highly expressed in CD8 T cells in the repopulated in the recipients’ peripheral blood (Fig. 2A, data absence of G9a (Fig. 2E), suggesting that G9a also contributes 2 not shown). Although Ehmt2+/+ CD8 T cells remained CD4 , to turning on genes in CD8 T cells directly or indirectly. A 2 2 ∼30% of Ehmt2 / CD8 T cells that had diluted CFSE similar change in gene expression, albeit to lesser extent, was 2 2 2 beyond the limit of detection upregulated CD4 (Fig. 2A, 2B). also observed in Ehmt2 / CD4 CD8+ T cells in which we Because a similar result was observed with transferred Va2+ have confirmed Ehmt2 deletion was also nearly complete 2 2 CD8 T cells from Ehmt2F/F CD8-cre OT-I mice, in which (Fig. 2E, data not shown). In Ehmt2 / CD8 T cells, the Ehmt2 was deleted after positive selection (Fig. 2B), it is level of total dimethylation of histone 3 lysine 9 (H3K9me2) unlikely that Cd4 derepression is secondary to deregulated was substantially reduced whereas that of H3K9me3 was thymocyte selection in the absence of G9a. unchanged (Supplemental Fig. 1A). Additionally, we did not 2 2 To determine whether Ehmt2 / CD8 T cells derepressed observe a reduction in H3K9me3 deposition near transcrip- additional helper lineage genes, global gene expression in tional start sites of the genes that were upregulated in CD4+ + + 2 + 2/2 + 2/2 +/+ CD4 CD8 and CD4 CD8 Ehmt2 T cells as well as CD8 Ehmt2 T cells compared with Ehmt2 CD8 Downloaded from control Ehmt2+/+ CD8+ T cells 4 wk after transfer was profiled T cells (Supplemental Fig. 1B). Cd4 upregulation was also 2 2 by RNA-seq. Approximately 1100 genes were differentially observed in Ehmt2 / OT-I T cells in response to E.G7- expressed by .2-fold with most (637 genes) being upregu- OVA tumor cells transplanted to wild-type mice (Fig. 2F, 2 2 lated in Ehmt2 / CD8 T cells compared with Ehmt2+/+ 2G). These results suggest that derepression of helper lineage 2 2 CD8 T cells (Fig. 2C, 2D). Among the genes that were dif- genes in proliferating Ehmt2 / CD8 T cells occurs also ferentially expressed between CD4 and CD8 memory T cells in lymphocyte-replete mice although it may not continue http://www.jimmunol.org/ by guest on September 27, 2021

FIGURE 3. Increased TCR signaling compensates for G9a deficiency in silencing helper lineage genes in CD8 T cells. (A 2 2 and B) CD4 and CD8 expression of Ehmt2 / and Ehmt2+/+ OT-I T cells (Thy1.2/CD45.2) that were cotransferred as a 1:1 mixture with Thy1.1/CD45.2 OT-I T cells into CD45.1 mice 4 d after Lm-OVA infection. (B) Ratios of Thy1.2+ to Thy1.1+ donor cells 4 d postinfection. Data points represent individual recipient in three experiments, in which cells from one donor per genotype were transferred into three recipi- ents. (C and D) Expression of CD4 and CD8 on Ehmt2+/+ 2 2 (C)orEhmt2 / (D) T cells cultured in the presence of indicated concentrations of anti-CD3 and anti-CD28 Abs. Data are representative of three experiments (n = 2 per genotype per experiment). (E) Percentage of CD4+ cells in cultured 2 2 Ehmt2 / or Ehmt2+/+ CD8 T cells shown by mean 6 SD. 2 2 (F) Quantitative PCR analysis of gene expression in Ehmt2 / and Ehmt2+/+ CD8 T cells cultured in either the CD28hi or CD28lo condition. Ehmt2+/+ Th1 CD4 T cells were used as control (Th1). The Journal of Immunology 5 once they slow down or stop division, such as memory CD8 naive polyclonal CD8 T cells with varying concentrations of T cells under steady-state conditions. Collectively, these data anti-CD3 and anti-CD28 Abs and determined whether dis- indicate that G9a is required for continued silencing of a tinct intensities of signaling through TCR or CD28 alter Cd4 2 2 subset of helper lineage–associated genes in dividing CD8 derepression in Ehmt2 / CD8 T cells. As seen in the lym- 2 2 T cells, which appears independent of H3K9me3. Because the phopenic condition, a substantial fraction of Ehmt2 / CD8 G9a/GLP complex recruits PRC2 to its repressive target loci T cells derepressed CD4 when they were cultured with low in embryonic stem cells (33), these helper lineage–associated anti-CD3 and high anti-CD28 Ab concentrations (CD28hi genes may be kept silenced through the G9a-mediated re- condition), whereas CD4 was barely expressed in Ehmt2+/+ cruitment of PRC2 activity. 2/2 CD8 T cells (Fig. 3C–E). Ehmt2 CD8 T cells also up- G9a is dispensable for silencing helper lineage genes in the presence of regulated additional helper lineage–related genes, such as Il21 2 2 strong TCR or IL-12R signals and Rorc (Fig. 3F). In contrast, when Ehmt2 / CD8 T cells To determine whether G9a is required for continued Cd4 were stimulated with high anti-CD3 and low anti-CD28 Ab silencing in dividing CD8 T cells in response to infection, concentrations (CD28lo condition), derepression of the helper 2 2 Ehmt2 / or Ehmt2+/+ OT-I T cells (Thy1.2/CD45.2) mixed lineage genes was markedly reduced (Fig. 3D–F). 2 2 with internal control congenic OT-I T cells (Thy1.1/ Furthermore, Cd4 derepression in Ehmt2 / CD8 T cells CD45.2) were transferred into CD45.1 wild-type mice, cultured in the CD28hi condition was significantly inhibited

which were subsequently infected with Lm-OVA. In contrast by the cytokine IL-12 (Fig. 4A, 4B). We detected elevated Downloaded from 2 2 2 2 to lymphopenia- and tumor-driven proliferation, Ehmt2 / H3K9me2 in Ehmt2 / CD8 T cells cultured in the presence 2 OT-I T cells remained CD4 (Fig. 3A). Additionally, we of IL-12 compared with those without IL-12 (Fig. 4C, 4D). observed comparable expansion and the ability to produce The IL-12 treatment upregulated the H3K9me3 demethylase 2 2 IFN-g of Ehmt2 / and Ehmt2+/+ OT-I T cells relative to Kdm4c as well as GLP/Ehmt1 by 3-fold (Fig. 4E), thus pos- control OT-I T cells (Fig. 3B, data not shown). These results sibly maintaining H3K9me2-dependent gene regulation by

indicate that G9a is dispensable for CD8 T cells under in- increasing demethylation of H3K9me3 by KDM4C or by http://www.jimmunol.org/ flammatory conditions to maintain Cd4 silencing and express elevating residual HMT activity of GLP. These results suggest IFN-g, which is also RUNX3-dependent (34). that the inflammation-dependent compensation may rein- Distinct dependency of silencing of helper lineage genes on force stable lineage-specific gene expression signature in CD8 G9a expressed in CD8 T cells between inflammatory and T cells that proliferate in response to infection. noninflammatory conditions suggests that cell-extrinsic signals Our study has demonstrated that G9a is required for through TCR, costimulatory molecules, or cytokine receptors maintaining silencing of multiple helper lineage–associated engage compensatory pathways that reinforce gene silencing. genes, such as Cd4, in dividing CD8 T cells in response to To define such cell-extrinsic determinants, we first cultured lymphopenia or tumor Ag. In developing CD8 T cells, Cd4 is by guest on September 27, 2021

FIGURE 4. Signals through IL-12 compensate for G9a deficiency in repression of helper lineage genes in CD8 T cells. (A and B) 2 2 Percentages of CD4+ cells in Ehmt2 / or con- trol Ehmt2+/+ CD8 T cells cultured in the CD28lo condition with or without murine IL-12 (mIL-12; 10 ng/ml). Plots are representative of two independent experiments (two to three mice per experiment). (C) Immunoblotting for total 2 2 H3 and H3K9me2 of lysates from Ehmt2 / or Ehmt2+/+ CD8 T cells cultured in the CD28hi or CD28lo condition. Data are representative of two experiments. (D) Relative expression of Ehmt2 in 2 2 CD8 T cells from Ehmt2 / and Ehmt2+/+ mice cultured with the CD28hi condition with or without mIL-12. (E) Quantitative PCR analysis of expression of H3K9 HMTs and demethylases 2 2 in Ehmt2 / CD8 T cells cultured in the CD28hi condition with or without mIL-12. n =3. 6 CUTTING EDGE: SILENCING OF HELPER LINEAGE GENES IN CD8 T CELLS BY G9a shut off by RUNX3 in a G9a-independent manner. However, 11. Taniuchi, I., M. Osato, T. Egawa, M. J. Sunshine, S. C. Bae, T. Komori, Y. Ito, and D. R. Littman. 2002. Differential requirements for Runx proteins in CD4 repres- the continued silenced state, which is independent of sion and epigenetic silencing during T lymphocyte development. Cell 111: 621– RUNX3, is not maintained in the absence of G9a, suggesting 633. 12. Egawa, T., R. E. Tillman, Y. Naoe, I. Taniuchi, and D. R. Littman. 2007. The role that transient RUNX3-dependent recruitment of G9a estab- of the Runx transcription factors in thymocyte differentiation and in homeostasis of lishes the heritably silenced states of the locus in cooperation naive T cells. [Published erratum appears in 2008 J. Exp. Med. 205: 1939.] J. Exp. with other factors, such as additional methyltransferases. Al- Med. 204: 1945–1957. 13. Setoguchi, R., M. Tachibana, Y. Naoe, S. Muroi, K. Akiyama, C. Tezuka, ternatively, G9a is constitutively recruited to the Cd4 locus T. Okuda, and I. Taniuchi. 2008. Repression of the transcription factor Th-POK by initially by RUNX3 and subsequently by a RUNX3- Runx complexes in cytotoxic T cell development. Science 319: 822–825. 14. Woolf, E., C. Xiao, O. Fainaru, J. Lotem, D. Rosen, V. Negreanu, Y. Bernstein, independent mechanism. All the G9a-dependent repression 2 2 D. Goldenberg, O. Brenner, G. Berke, et al. 2003. Runx3 and Runx1 are required targets are not RUNX3 targets (18, 34), and Ehmt2 / CD8 for CD8 T cell development during thymopoiesis. Proc. Natl. Acad. Sci. USA 100: 7731–7736. T cells are also able to proliferate and express IFN-g, which is 15. Sawada, S., J. D. Scarborough, N. Killeen, and D. R. Littman. 1994. A lineage- dependent on RUNX3. Therefore, there are multiple distinct specific transcriptional silencer regulates CD4 gene expression during T lymphocyte development. Cell 77: 917–929. RUNX3- or G9a-containing complexes that regulate gene 16. Zou, Y. R., M. J. Sunshine, I. Taniuchi, F. Hatam, N. Killeen, and D. R. Littman. activation or repression in CD8 T cells, whereas Cd4 silencing 2001. Epigenetic silencing of CD4 in T cells committed to the cytotoxic lineage. and repression of some of TFH-signature genes (18), in- Nat. Genet. 29: 332–336. 17. Taniuchi, I., M. J. Sunshine, R. Festenstein, and D. R. Littman. 2002. Evidence for cluding Icos, Cxcr5, and Il21, appear to be dependent on a distinct CD4 silencer functions at different stages of thymocyte differentiation. Mol. complex containing both. Cell 10: 1083–1096. 18. Shan, Q., Z. Zeng, S. Xing, F. Li, S. M. Hartwig, J. A. Gullicksrud, S. P. Kurup, The absence of Cd4 derepression in CD8 T cells responding N. Van Braeckel-Budimir, Y. Su, M. D. Martin, et al. 2017. The transcription Downloaded from to Lm-OVA infection may be explained by high levels of Ag factor Runx3 guards cytotoxic CD8+ effector T cells against deviation towards follicular helper T cell lineage. Nat. Immunol. 18: 931–939. and IL-12, both of which compensate for G9a deficiency in 19. Hogquist, K. A., S. C. Jameson, W. R. Heath, J. L. Howard, M. J. Bevan, and CD8 T cells in vitro. Although the exact mechanism is un- F. R. Carbone. 1994. T cell receptor antagonist peptides induce positive selection. known, our data suggest that IL-12R signaling alters the Cell 76: 17–27. 20. Maekawa, Y., Y. Minato, C. Ishifune, T. Kurihara, A. Kitamura, H. Kojima, balance between histone methylation and demethylation to H. Yagita, M. Sakata-Yanagimoto, T. Saito, I. Taniuchi, et al. 2008. Notch2 in- increase the levels of H3K9me2 independent of G9a, possibly tegrates signaling by the transcription factors RBP-J and CREB1 to promote T cell http://www.jimmunol.org/ cytotoxicity. Nat. Immunol. 9: 1140–1147. facilitating heritable gene silencing in dividing CD8 T cells 21. Tachibana, M., M. Nozaki, N. Takeda, and Y. Shinkai. 2007. Functional dynamics through the compensatory pathways. CD8 T cells may thus of H3K9 methylation during meiotic prophase progression. EMBO J. 26: 3346– 3359. engage multiple epigenetic pathways in a context-dependent 22. Egawa, T., and D. R. Littman. 2011. Transcription factor AP4 modulates reversible manner to shape their gene expression signature. and epigenetic silencing of the Cd4 gene. Proc. Natl. Acad. Sci. USA 108: 14873– 14878. 23. Chou, C., A. K. Pinto, J. D. Curtis, S. P. Persaud, M. Cella, C. C. Lin, B. T. Edelson, P. M. Allen, M. Colonna, E. L. Pearce, et al. 2014. c--induced Acknowledgments + We thank D. Littman for cell lines, Cd4-cre mice, and CD8-cre mice, C.-S. transcription factor AP4 is required for host protection mediated by CD8 T cells. Nat. Immunol. 15: 884–893. Hsieh and E.M. Oltz for discussions, and J. Chaudhuri, P. Collins, 24. Chou, C., D. J. Verbaro, E. Tonc, M. Holmgren, M. Cella, M. Colonna, by guest on September 27, 2021 M. Holmgren, S. Hsiung, S. Raju, and E. Russler-Germain for technical D. Bhattacharya, and T. Egawa. 2016. The transcription factor AP4 mediates res- assistance. olution of chronic viral infection through amplification of germinal center B cell responses. Immunity 45: 570–582. 25. Trapnell, C., L. Pachter, and S. L. Salzberg. 2009. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111. Disclosures 26. Trapnell, C., B. A. Williams, G. Pertea, A. Mortazavi, G. Kwan, M. J. van Baren, The authors have no financial conflicts of interest. S. L. Salzberg, B. J. Wold, and L. Pachter. 2010. Transcript assembly and quan- tification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28: 511–515. 27. Roberts, A., H. Pimentel, C. Trapnell, and L. Pachter. 2011. Identification of novel References transcripts in annotated genomes using RNA-Seq. Bioinformatics 27: 2325–2329. 1. Vacchio, M. S., and R. Bosselut. 2016. What happens in the thymus does not stay 28. Roberts, A., C. Trapnell, J. Donaghey, J. L. Rinn, and L. Pachter. 2011. Improving 2 in the thymus: how T cells recycle the CD4+ CD8+ lineage commitment tran- RNA-Seq expression estimates by correcting for fragment bias. Genome Biol. 12: scriptional circuitry to control their function. J. Immunol. 196: 4848–4856. R22. 2. Wilson, C. B., K. W. Makar, and M. Pe´rez-Melgosa. 2002. Epigenetic regulation of 29. Trapnell, C., D. G. Hendrickson, M. Sauvageau, L. Goff, J. L. Rinn, and T cell fate and function. J. Infect. Dis. 185(Suppl. 1): S37–S45. L. Pachter. 2013. Differential analysis of gene regulation at transcript resolution 3. Bosselut, R. 2004. CD4/CD8-lineage differentiation in the thymus: from nuclear with RNA-seq. Nat. Biotechnol. 31: 46–53. effectors to membrane signals. Nat. Rev. Immunol. 4: 529–540. 30. Sellars, M., J. R. Huh, K. Day, P. D. Issuree, C. Galan, S. Gobeil, D. Absher, 4. He, X., and D. J. Kappes. 2006. CD4/CD8 lineage commitment: light at the end of M. R. Green, and D. R. Littman. 2015. Regulation of DNA methylation dictates the tunnel? Curr. Opin. Immunol. 18: 135–142. Cd4 expression during the development of helper and cytotoxic T cell lineages. Nat. 5. Singer, A., S. Adoro, and J. H. Park. 2008. Lineage fate and intense debate: myths, Immunol. 16: 746–754. models and mechanisms of CD4- versus CD8-lineage choice. Nat. Rev. Immunol. 8: 31. Nishio, H., and M. J. Walsh. 2004. CCAAT displacement protein/cut homolog 788–801. recruits G9a histone lysine methyltransferase to repress transcription. Proc. Natl. 6. Collins, A., D. R. Littman, and I. Taniuchi. 2009. RUNX proteins in transcription Acad. Sci. USA 101: 11257–11262. factor networks that regulate T-cell lineage choice. Nat. Rev. Immunol. 9: 106–115. 32. Thomas, L. R., H. Miyashita, R. M. Cobb, S. Pierce, M. Tachibana, E. Hobeika, 7. Taniuchi, I. 2009. Transcriptional regulation in helper versus cytotoxic-lineage M. Reth, Y. Shinkai, and E. M. Oltz. 2008. Functional analysis of histone meth- decision. Curr. Opin. Immunol. 21: 127–132. yltransferase G9a in B and T lymphocytes. J. Immunol. 181: 485–493. 8. Egawa, T. 2015. Regulation of CD4 and CD8 coreceptor expression and CD4 33. Mozzetta, C., J. Pontis, L. Fritsch, P. Robin, M. Portoso, C. Proux, R. Margueron, versus CD8 lineage decisions. Adv. Immunol. 125: 1–40. and S. Ait-Si-Ali. 2014. The histone H3 lysine 9 methyltransferases G9a and GLP 9. Issuree, P. D., C. P. Ng, and D. R. Littman. 2017. Heritable gene regulation in the regulate polycomb repressive complex 2-mediated gene silencing. Mol. Cell 53: 277– CD4:CD8 T cell lineage choice. Front. Immunol. 8: 291. 289. 10. Lee, P. P., D. R. Fitzpatrick, C. Beard, H. K. Jessup, S. Lehar, K. W. Makar, 34. Cruz-Guilloty, F., M. E. Pipkin, I. M. Djuretic, D. Levanon, J. Lotem, M. Pe´rez-Melgosa, M. T. Sweetser, M. S. Schlissel, S. Nguyen, et al. 2001. A critical M. G. Lichtenheld, Y. Groner, and A. Rao. 2009. Runx3 and T-box proteins co- role for Dnmt1 and DNA methylation in T cell development, function, and sur- operate to establish the transcriptional program of effector CTLs. J. Exp. Med. 206: vival. Immunity 15: 763–774. 51–59. Supplemental Data

Gene Forward Primer Reverse Primer Cd4 5'-GGGCTGTGGCAGTGTCTACT-3' 5'-GACACAGCAGAGGATGCAGA-3' Rorc 5'-TCTACGCTATGAGGAAGGAAGGC-3' 5'-GACTATGGAGGAGAAACAGGTCCC-3' Il21 5'-GCTCCACAAGATGTAAAGGGGC-3' 5'-CCACGAGGTCAATGATGAATGTC-3' Cd40lg 5'-GTGAGGAGATGAGAAGGCAA-3' 5'-CACTGTAGAACGGATGCTGC-3' Foxp3 5'-AGAAGCTGGGAGCTATGCAG-3' 5'-TACTGGTGGCTACGATGCAG-3' Ctla4 5'-GGATCCTTGTCGCAGTTAGC-3' 5'-TCACATTCTGGCTCTGTTGG-3' Aqp9 5'-TGGGGATTTGAGGTCTTCAC-3' 5'-GTTCGAGTGATGCATTTGGA-3' Zbtb7b 5'-TGCTTCCGCATGTGGATC-3' 5'-GTGAGAAGCCCTTTGCCTGT-3' Ehmt2 5'-TCATCCCTGTCCGGGTTTTC-3' 5'-TCACCGTAGTCAAAGCCCA-3' Ehmt1 5'-AAGCAAGAGACCAAGCAGGA-3' 5'-TGTGGAACCTTCATCAGCAG-3' Cd8a 5'-CACAGGAGCCGAAAGCGT-3' 5'-GGGCTTGCCTTCCTGTCTG-3' Rora 5'-CGCATTGATGGATTTATGGAG-3' 5'-TCGCATACTTCCCGTCAAAG-3' Kdm1a 5'-TGGGATGGATGTCACACTTC-3' 5'-CTCGTCCACCTACTCGATCC-3' Kdm1b 5'-GAGTATGCTTGTGGCAGCAG-3' 5'-GGGTATGGTCACCAGCAAAC-3' Kdm3a 5'-TGTCGACTATTGAGCCACACA-3' 5'-TCCTTTGACAGCTCGTTTCC-3' Kdm3b 5'-TGGAAGGCTCACTTGTTTGG-3' 5'-GGTATTCAACTGGCACCACAG-3' Kdm4c 5'-ATGGATTGACTACGGCAAGG-3' 5'-CCATGTCATTCCGACAAGTG-3' Kdm7a 5'-CAGCTCTACACGGCTCTTCC-3' 5'-ACAGGTTTGGAGCCATCATC-3' Phf8 5'-GTGTGACATGTGCCAGGACT-3' 5'-TCCTCCTCAACACCAACACA-3' Setdb1 5'-CACAAAGGCACCCTTATTGC-3' 5'-CGGGAGGGTGGTAATCATAG-3' Suv39h1 5'-AGGGGAGGAAGAAGTGGAAC-3' 5'-CAGGTCCTGCAGTTGATTCC-3' Prdm2 5'-CTCCACCTCTGCAAACATGA-3' 5'-ACTGCATCCTGGCTTACCAC-3' 18S rRNA 5'-CGGCTACCACATCCAAGGAA-3' 5'-GCTGGAATTACCGCGGCT-3'

Supplemental Table 1. Sequences of primers used for quantitative PCR.

1

Supplemental Figure 1. De-repression of the helper lineage-associated genes in Ehmt2–/– cells is independent of H3K9 trimethylation. (A) Western blots showing amounts of H3K9me2, H3K9me3 or total H3 in CD8 T cells from Ehmt2F/F Cd8-E8I-cre mice and control Ehmt2F/F cre– mice. Data are representative of two experiments. (B) A heat map displaying the distribution of H3K9me3 ChIP-seq tags at the 637 genes that were identified from the RNA-sequencing data with at least a 2-fold higher expression in + + F/F + F/F – the CD4 CD8 Ehmt2 Cd4-cre mice compared to CD8 cells from control Ehmt2 cre in Fig. 2C. Fold-changes of expression of those genes are plotted on the right panel.

2