Medullary thymic epithelial cells (mTECs) expressing the autoimmune regulator AIRE and various tissue-specific antigens (TSAs) are critical for preventing the onset of autoimmunity and may attenuate tumor immunity. However, molecular mechanisms controlling mTEC development remain elusive. Here, we describe the roles of the transcription factor Spi-B in mTEC development. Spi-B is rapidly up-regulated by receptor activator of NF-κB ligand (RANKL) cytokine signaling, which triggers mTEC differentiation, and in turn up-regulates CD80, CD86, some TSAs, and the natural inhibitor of RANKL signaling, osteoprotegerin (OPG). Spi-B–mediated OPG expression limits mTEC development in neonates but not in embryos, suggesting developmental stage–specific negative feedback regulation. OPG-mediated negative regulation attenuates cellularity of thymic regulatory T cells and tumor development in vivo. Hence, these data suggest that this negative RANKL–Spi-B–OPG feedback mechanism finely tunes mTEC development and function and may optimize the trade-off between prevention of autoimmunity and induction of antitumor immunity.

Of the various T cell types that develop in the thymus, medullary thymic epithelial cells (TECs [mTECs]) are crucial to the induction of T cell tolerance to endogenous tissues during thymic T cell development (Anderson and Takahama, 2012). Mature mTECs highly express MHC class II (MHC II) and co-stimulatory molecules, such as CD80 and CD86, and function as self-antigen–presenting cells in the thymus (Kyewski and Klein, 2006; Klein et al., 2009; Hinterberger et al., 2010). Uniquely, mature mTECs promiscuously express a wide variety of endogenous tissue-specific antigens (TSAs), including insulin, C-reactive protein, and caseins (Kyewski and Klein, 2006; Klein et al., 2009). The autoimmune regulator Aire, mutations in which cause human autoimmune diseases, is a transcription factor that is highly expressed in mature mTECs and that enhances TSA diversity (Abramson et al., 2010). Consequently, mature mTECs promote clonal deletion and regulatory T cell (T reg cell) conversion of potentially TSA-reactive T cells; these are critical for preventing the onset of autoimmunity. Furthermore, recent studies have shown that Aire deficiency inhibits tumor growth and T reg cell accumulation in tumors (Träger et al., 2012; Malchow et al., 2013; Zhu et al., 2013), suggesting that mTECs induce immunological tolerance in tumor and normal tissues. This implies that precise regulation of mTEC-mediated tolerance may be critical for balancing prevention of autoimmunity with induction of tumor immunity, but the molecular mechanisms underlying development and function of mTECs are poorly understood.

We and others previously reported that the receptor activator of NF-κB (RANK) ligand (RANKL) promotes development of mature mTECs (Rossi et al., 2007; Akiyama et al., 2008, 2012b; Hikosaka et al., 2008). Moreover, several signal transducers regulating NF-κB activation pathways, such as TNF receptor–activated factor 6 (TRAF6), NF-κB–inducing kinase (NIK), and the NF-κB family member RelB, are required for mTEC development (Burkly et al., 1995; Weih et al., 1995; Kajiura et al., 2004; Akiyama et al., 2005). Thus, RANKL probably triggers mTEC differentiation by activating NF-κB pathways (Akiyama et al., 2012b), but the molecular events involved remain unknown.

The Ets transcription factor family member Spi-B (Ray et al., 1992) regulates plasmacytoid dendritic cell development and function, B cell antigen receptor signaling, early T cell lineage decisions, and intestinal M cell development (Garrett-Sinha et al., 1999; Schotte et al., 2004; Dontje et al., 2006; Kanaya et al., 2012; Sasaki et al., 2012). The locus of human SPIB has also been associated with autoimmune primary biliary cirrhosis (Liu et al., 2010), implicating it in prevention of autoimmunity.

Here, we demonstrate that Spi-B links RANKL–NF-κB signaling with up-regulation of several molecules expressed in mature mTECs, including CD80, CD86, some TSAs, and osteoprotegerin (OPG), the natural inhibitor of RANKL. Moreover, we show that Spi-B–mediated OPG expression in the thymus limits the development of mature mTECs via a negative feedback regulatory circuit that may facilitate immune responses to tumors.

RANKL signaling up-regulates Spi-B expression in mTECs through an NIK-dependent pathway

We recently identified candidate transcriptional regulators of mTEC development by microarray analysis (Ohshima et al., 2011). Spi-B was selected for further analysis because of its possible involvement in autoimmune disease (Liu et al., 2010). We first investigated whether RANKL signaling induces the expression of Spi-B in mTECs. RANKL stimulation is known to induce differentiation of mature mTECs expressing Aire, TSAs, and an mTEC marker, UEA-1 lectin ligand (Fig. 1 A) in in vitro organ culture of fetal thymic stroma (2′-deoxyguanosine [2DG]–fetal thymus organ culture [FTOC]; Rossi et al., 2007; Akiyama et al., 2008), which is prepared by depleting cells of hematopoietic origin from fetal thymus (Aichinger et al., 2012). Quantitative RT-PCR (qPCR) revealed that Spib mRNA was significantly up-regulated by RANKL stimulation in 2DG-FTOCs (Fig. 1 B). Addition of RANK-Fc blocked RANKL-dependent Spib expression (Fig. 1 B), confirming the requirement of RANKL–RANK interactions. RANKL-dependent up-regulation of Spib preceded that of Aire, the TSA Salivary protein 1 (Spt1), and UEA-1 (Figs. 1, A and C), suggesting that Spib is an early gene that responds to RANKL signaling.

We next investigated whether RANKL signaling induces Spib expression in an established mTEC line because 2DG-FTOCs contain several types of stromal cells, including TECs. This mTEC line was retrovirally transduced with RANK (RANK-TEC) because endogenous RANK expression was undetectable in the currently available mTEC lines. Stimulation with RANKL rapidly and continuously up-regulated Spib expression in the RANK-TECs (Fig. 1 D). These findings suggest that Spib expression is rapidly induced by RANKL signaling in mTECs.

Subsequently, we determined whether Spi-B is expressed in mTECs in vivo. Adult mouse thymic stroma cells (CD45TER119) were sorted by staining with anti-EpCAM (an epithelial cell marker) and UEA-1 and were subsequently processed for qPCR gene expression assays. The mTEC fraction (UEA-1+EpCAM+), which exhibits high RANK expression levels (Fig. 1 E), showed significantly higher levels of Spi-B expression than did the cTEC-rich fraction (UEA-1EpCAM+) and the nonepithelial stromal cell fraction (EpCAM; Fig. 1 E). These data were consistent with increased Spi-B expression after RANKL signaling in thymic RANK-expressing mTECs.

Next, we investigated the mechanisms regulating Spi-B expression that occur downstream of RANKL signaling. Previous studies showed that murine Spib is transcribed from two different promoters (Chen et al., 1998), which can be discriminated using qPCR (Bartholdy et al., 2006). RANKL stimulation selectively induced 1 of the Spib transcripts (Spib1) in 2DG-FTOC (Fig. 1 F). Moreover, Spib1 expression was higher in mTECs than in other stromal cell fractions (Fig. 1 G). Thus, increased Spib expression in mTECs (Fig. 1 E) follows expression of Spib1 but not that of Spib2. Because RANKL signaling selectively activates the promoter that drives Spib1 transcription, the genomic region containing the conserved noncoding sequence upstream of the Spib1 initiation site (Fig. 1 H) was linked to a luciferase gene for reporter assays (Spib-P1-luc). As expected, Spib-P1-luc was responsive to RANKL stimulation in the RANK-mTEC line (Fig. 1 H), suggesting that this region contains the promoter that drives Spib transcription.

Previous studies showed that the signal transducers TRAF6 and NIK, which mediate classical and nonclassical NF-κB activation pathways, respectively, are required during RANKL-mediated mTEC differentiation (Kajiura et al., 2004; Akiyama et al., 2005, 2008). Thus, we investigated whether overexpression of these two signal transducers activated Spib-P1 in cells. Interestingly, Spib-P1 was activated only in NIK-overexpressing cells (Fig. 1 I), whereas an artificial NF-κB promoter (3κB-luc) was effectively activated by NIK or TRAF6 overexpression (Fig. 1 I). Induction of NIK activates the RelB complex (Thu and Richmond, 2010; Sun, 2012). Overexpression of the latter effectively activated the Spib-P1 promoter (Fig. 1 I). Moreover, a dysfunctional point mutation in Nik (aly/aly mutation) markedly diminished RANKL-mediated Spib expression in 2DG-FTOCs (Fig. 1 J; Shinkura et al., 1999). Thus, these data suggested that Spib is a target gene of RANKL–NIK–NF-κB signaling that promotes mTEC development (Kajiura et al., 2004; Akiyama et al., 2008, 2012b). Therefore, we investigated the roles of Spi-B in mature mTEC development in Spi-B–deficient (Spib−/−) mice.

Spi-B limits development of mature mTECs but promotes CD80 expression in mature mTECs

Adult murine mTECs are classified into subsets according to the expression levels of MHC II. mTECs expressing high MHC II levels (mTEChi) express a wide variety of TSAs (Kyewski and Klein, 2006; Anderson and Takahama, 2012) and are considered more mature than those expressing low MHC II (mTEClo) levels (Gray et al., 2007; Anderson et al., 2009). Flow cytometry revealed that the numbers and frequencies of total mTECs and mTEChi were significantly increased in the absence of Spi-B (Fig. 2 A), indicating that Spi-B is a negative regulator of mature mTEC development. To confirm that expression of Spi-B in TECs, but not in thymocytes, is responsible for this phenotype, we transplanted 2DG-FTOCs from Spib−/− and WT fetal thymus into the kidney capsules of athymic nude mice. In these chimeric mice, the thymus comprises Spi-B–deficient thymic stroma and thymocytes derived from nude mice. mTEChi numbers were significantly increased in the developed thymus of mice receiving Spib−/− 2DG-FTOCs (Fig. 2 B), indicating that Spi-B in thymic stromal cells limits the development of mTEChi cells.

We next investigated the expression of other mTEC marker proteins in Spib−/− thymuses using flow cytometry. The mTEChi subset includes Aire-expressing mTECs (Gray et al., 2007). Moreover, mTEChi cells express high levels of co-stimulatory molecules, such as CD80, CD86, PD-L1, and CD40 (Kyewski and Klein, 2006; Klein et al., 2009; Hinterberger et al., 2010). As expected, Aire expression levels in whole thymus were increased by the lack of Spi-B (Fig. 2 C). Consistently, the number and frequency of Aire-expressing mTECs were increased in Spib−/− thymuses (Fig. 2 D), whereas Aire expression levels in Aire-expressing mTECs were slightly reduced by the absence of Spi-B (Fig. 2 E). Remarkably, CD80 expression was diminished in Spib−/− mTEChi, including Aire-expressing mTECs (Fig. 2, F and G). Moreover, mTECs expressing high levels of CD86 were significantly reduced in Spib−/− mice (Fig. 2 H). In contrast, expression of the other co-stimulatory molecules, PD-L1 and CD40, was not affected by the absence of Spi-B (Fig. 2 I), suggesting that Spi-B selectively up-regulates CD80 and CD86 among co-stimulatory molecules in mTEChi cells. Altogether, these data demonstrated two distinct novel functions of Spi-B during mTEC development: Spi-B limits mature mTEC development and promotes expression of co-stimulatory molecules CD80 and CD86 in mTECs.

Spi-B facilitates expression of OPG and some TSAs in mTECs

Because Spi-B may selectively regulate gene expression in mTECs, we subsequently identified other genes that are regulated by Spi-B in mTECs. We sorted mTEC fractions (EpCAM+UEA-1+) from Spib−/− and WT mice and investigated gene expression using qPCR. Indeed, expression of the TSAs Spt1 and Col2 was significantly reduced in Spib−/− mTEC fractions compared with those in WT mTEC fractions (Fig. 3 A). Thus, Spi-B may facilitate the expression of some TSA members by an Aire-independent mechanism. Notably, the expression of OPG was significantly reduced in Spib−/− mTECs (Fig. 3 A), and reduced OPG protein expression was confirmed by immunohistochemical staining of thymic sections (Fig. 3 B). OPG competitively inhibits RANKL–RANK interactions as a decoy receptor of RANKL (Leibbrandt and Penninger, 2008), thereby inhibiting mTEC development (Hikosaka et al., 2008). Because this inhibitory effect (Fig. 2) may relate to reduced OPG expression, we further characterized Spi-B–dependent OPG expression in mTECs.

As expected, Opg mRNA expression was induced by RANKL signaling in 2DG-FTOCs (Fig. 3 C). RANKL stimulation consecutively induced Spib and Opg expression (compare Fig. 3 C with Fig. 1 C). RANKL-dependent Opg expression was significantly reduced in the absence of Spi-B in 2DG-FTOCs (Fig. 3 D). These data suggested that RANKL signaling up-regulates Spib expression and promotes expression of its own inhibitor, OPG, in mTECs.

Subsequently, we investigated the mechanism regulating Opg expression via Spi-B in mTECs. Because Spi-B overexpression did not activate the putative proximal promoter of Opg in reporter assays (not depicted), Spi-B was expected to control Opg transcription without directly binding to the proximal promoter of the gene. Thus, we analyzed CpG methylation of Opg in sorted mTEC fractions. Initially, we observed two CpG-rich regions that were hypomethylated in mTECs compared with EpCAM thymic stromal cells (Fig. 3 E), suggesting that Opg is tissue-dependently and differentially methylated (Yagi et al., 2008) in these regions (referred to as tissue-dependently and differentially methylated region [T-DMR] #1 and T-DMR#2). T-DMRs were hypermethylated in Spib−/− mTECs compared with control mTECs (Fig. 3 E). Thus, Spi-B expression in mTECs leads to either demethylation of CpG sites or maintenance of their hypomethylated states in Opg, which may enhance Opg expression.

Moreover, we genetically verified that enhancement of mTEC development in Spib−/− mice was dependent on OPG expression. As reported earlier (Hikosaka et al., 2008), OPG-deficient (Opg−/−) mice showed enhanced development of mTECs (Fig. 3 F) compared with WT controls (Figs. 2 A and 3 F [green lines]). Indeed, mTEC development was similar between Opg−/− mice and Spi-B and OPG double-deficient (Spib−/−Opg−/−) mice (Fig. 3 F). Thus, Spi-B does not attenuate mTEC development in the absence of OPG, strongly suggesting that the difference in mTEC development between WT and Spib−/− mice was caused by differential expression of OPG.

Spi-B–mediated negative feedback regulation of RANKL signaling initiates perinatally, limiting mTEC development

Aire-expressing mTECs in the perinatal period are essential and sufficient for establishment of long-lasting self-tolerance (Guerau-de-Arellano et al., 2009). Therefore, we investigated mTEC development in embryonic and neonatal Spib−/− mice. Flow cytometric analyses of embryonic day (E) 15.5, E17.5, postnatal day (PN) 1, and PN8 thymuses revealed incremental development of mTECs (MHC II+UEA-1+) in Spib−/− PN1 and PN8 mice, but not in E15.5 and E17.5 mice (Fig. 4 A). Immunohistochemical analysis confirmed enhanced development of mTECs in postnatal Spib−/− thymuses (Fig. 4 B). Moreover, Aire expression levels in mature mTECs were significantly increased only in postnatal Spib−/− thymuses (Fig. 4 C). Thus, negative regulation of mTEC development by Spi-B characteristically starts perinatally. Although an increment in mTEC number in response to a lack of OPG has been reported only in adult mice (Hikosaka et al., 2008), OPG function in embryonic and neonatal mTEC development remained unknown. We therefore investigated whether OPG attenuates mTEC development in a developmental stage–dependent manner, similar to Spi-B. Indeed, mTEC development was not influenced by OPG deficiency in E15.5 thymuses and minimally enhanced in E17.5 Opg−/− thymuses (Fig. 4 D). In contrast, mTEC development was markedly enhanced in PN1 and PN8 Opg−/− mice (Fig. 4 D). In embryonic and neonatal thymus, CD80 is a more expedient marker for mature mTECs than MHC II. Consistently, CD80hi mature mTECs were increased in Opg−/− PN1 and PN8, but not in E15.5 and E17.5 mice (Fig. 4 E). Overall, these data suggested that the negative regulation of mature mTEC development by the Spi-B–OPG system is initiated perinatally. We subsequently identified the stromal cell type that expresses Opg using qPCR. We found that Opg is expressed in CD80hi mature mTECs in PN1 mice (Fig. 4 F). Interestingly, Opg expression levels in mature mTECs were significantly up-regulated perinatally and plateaued in neonatal thymuses (Fig. 4 G). This may explain the stage-dependent negative regulation of mTEC development by OPG and implies a qualitative change in mature mTECs during the perinatal period.

We confirmed the RANKL–RANK–Spi-B–OPG–RANKL feedback loop in the perinatal period. Given that such a negative feedback loop exists, the absence of OPG would enhance Spi-B expression in mTECs by increasing RANKL signaling. Indeed, Spi-B expression was significantly enhanced in Opg−/− mTECs at PN1 (Fig. 4 H). Interestingly, Spi-B was expressed in both CD80lo immature mTECs and CD80hi mature mTECs (Fig. 4 H) and was also up-regulated in the absence of OPG in both mTEC types, suggesting that RANKL signaling rapidly induces Spi-B in immature mTECs before promoting differentiation into mature mTECs. Cumulatively, these data strongly suggested that Spi-B–mediated Opg expression attenuates the development of mTECs by a negative feedback regulation of RANKL signaling.

Thymic stromal OPG attenuates cellularity of T reg cells in the thymus

We subsequently determined whether this negative regulation via RANKL–Spi-B–OPG influences mTEC-mediated thymic functions. We investigated the effects of abolishing this negative feedback loop in Opg−/− mice rather than Spib−/− mice because Spi-B reduces expression of CD80, CD86, and some TSAs in mTECs (Figs. 3 and 4). As reported earlier (Khan et al., 2014), expression levels of TSAs and Aire were significantly increased in Opg−/− thymuses (Fig. 5 A). Because mature mTECs promote T reg cell development as self-antigen–presenting cells (Hinterberger et al., 2010; Lei et al., 2011; Cowan et al., 2013; Malchow et al., 2013) and an increase in mTEC cellularity enhances thymic T reg cell development (Hauri-Hohl et al., 2014), we investigated whether OPG deficiency facilitates T reg cell development in the thymus. Indeed, numbers and frequency of Foxp3+ T reg cells were significantly increased in thymuses of neonatal and young Opg−/− mice (Fig. 5 B). Moreover, the frequency of thymic T reg cell precursors (CD4+CD8Foxp3GITR+CD25+; Lio and Hsieh, 2008) was increased in Opg−/− thymuses (Fig. 5 C). In vitro FTOC experiments in which mTECs and T reg cells develop by endogenous signaling suggested that the increase in T reg cells by OPG deficiency is caused in a thymus-intrinsic manner (Fig. 5 D). Fetal thymic stroma transplantation experiments indicated that OPG deficiency in thymic stromal cells caused an increment of T reg cell frequency in thymuses, whereas the increase in splenic T reg cells was not significant (Fig. 5 E). Overall, these data supported the idea that negative feedback regulation via RANKL–Spi-B–OPG attenuates mTEC-mediated thymic generation of T reg cells, although it is possible that more T reg cells are retained in the Opg−/− thymus.

Thymic stromal OPG attenuates tumor incidence and growth and facilitates humoral immune responses to tumors

Because negative feedback regulation of mTEC development via RANKL–Spi-B–OPG is initiated perinatally (Figs. 4 and 5), establishing long-term self-tolerance by Aire-expressing mTECs (Guerau-de-Arellano et al., 2009), we investigated the role of this negative regulation in modulation of immunological tolerance in vivo. Recent studies suggested that Aire-expressing mTECs impede antitumor immunity (Träger et al., 2012; Malchow et al., 2013; Zhu et al., 2013). Therefore, we speculated that this feedback regulation may contribute to antitumor immunity by attenuating mTEC-dependent tolerance to tumor-derived antigens. We tested this hypothesis in mouse carcinogen-induced tumor and tumor transplantation models. To exclude any other effects caused by increased RANKL activity, we initially adoptively transplanted Opg−/− 2DG-FTOCs (BALB/c) into kidney capsules of BALB/c nude mice (Leibbrandt and Penninger, 2008; Akiyama et al., 2012a). A limited dose of the carcinogenic polycyclic aromatic hydrocarbon, methylcholanthrene (MCA), was injected into chimeric nude mice that had received Opg−/− 2DG-FTOCs (Opg−/−/nude) and control mice that had received WT 2DG-FTOCs. Tumor incidence was significantly increased in Opg−/−/nude mice compared with control mice (Fig. 6 A), indicating that thymic stromal OPG limits MCA-dependent tumor incidence in vivo.

After congenic mouse tumor cells were subcutaneously transferred into nude recipients, tumor growth was significantly increased in Opg−/−/nude compared with WT/nude mice (Fig. 6 B). Tumor-infiltrating T cells were reduced in tumors of Opg−/−/nude mice (Fig. 6 C). In addition, T reg cell numbers were significantly increased in the draining lymph nodes of these mice (Fig. 6 D). Titers of antitumor cell antibodies evoked by tumor cell transplantation (determined by immunochemistry) were decreased in the serum of tumor-transplanted Opg−/−/nude mice compared with those in WT/nude control mice (Fig. 6 E), suggesting weaker humoral immune responses to tumors in the former. Notably, the kinds of tumor-derived proteins recognized by serum antibodies upon Western blotting were also reduced in tumor-transplanted Opg−/−/nude mice (Fig. 6 E, arrows), implying that TCR repertoires against tumor-derived proteins were decreased by the absence of OPG in thymic stroma. Thus, OPG expression in thymic stromal cells enhances immune responses to and limits the growth of tumors. These data further indicated that negative regulation via RANKL–Spi-B–OPG fine-tunes induction of mTEC-mediated tolerance and may ensure immune responses to tumors.

Although RANKL signaling is known to trigger differentiation of mature mTECs, molecular events linking RANKL signaling to the expression of molecules regulating mTEC functions have been largely unexplored. Here, we demonstrated that Spi-B expression links RANKL–NF-κB signaling with the expression of CD80, CD86, some TSAs, and OPG, implying a key regulatory role for Spi-B in mTECs. Interestingly, Spi-B deficiency did not affect the expression of Aire, MHC II, PDL1, or CD40, which are up-regulated in mature mTECs. This suggests that other unidentified mechanisms must regulate the expression of these molecules downstream of RANKL–NF-κB signaling.

Spi-B regulates high expression of CD80 and CD86 in mature mTECs, suggesting its positive role in mTEC-mediated function. Interestingly, Spi-B, CD80, and NF-κB1 have been associated with primary biliary cirrhosis (Liu et al., 2010; Mells et al., 2011), implying that Spi-B–mediated CD80 expression is correlated to the onset of autoimmunity.

Regulation of gene expression by Spi-B in mTECs remains unclear. However, the nearest homologue of Spi-B, PU.1, reportedly induces nucleosome remodeling and H3K4 monomethylation (Heinz et al., 2010). These epigenetic events are usually associated with changes in DNA methylation (Jones and Baylin, 2007). Given that Spi-B promotes demethylation of T-DMRs in Opg, Spi-B may exert epigenetic controls over gene expression in mTECs. Genome-wide epigenetic and expression studies of Spib−/− mTECs may verify this hypothesis in future.

The mechanisms that activate self-tolerance by mTECs require stochastic encounters between self-reactive T cells and mature mTECs or dendritic cells that receive TSAs from mTECs. This mechanism may be inherently imperfect (Bouneaud et al., 2000) because of its stochasticity. The negative feedback regulation described herein ensures inadequacy of mTEC-dependent tolerance and could mediate detrimental effects by increasing the prevalence of autoreactive T cells. However, our data suggest that this negative regulation may contribute to tumor immunity and may optimize the trade-off between prevention of autoimmunity and induction of tumor immunity. Future analyses of this regulatory mechanism will promote understanding of tumor immunosurveillance and may lead to the development of new therapeutic approaches for cancer.

Mice.

Spib−/− mice were established on a C57BL/6 background (Sasaki et al., 2012). The C57BL/6, BALB/cA, BALB/cA nude/nude, aly/+, and aly/aly mice were obtained from CLEA Japan. The Opg−/− mice have been described previously (Mizuno et al., 1998). The Opg−/− mice on a BALB/cA background were prepared by backcrossing with BALB/cA mice for 10 generations. Littermates or age-matched WT mice from the same colonies as the mutant mice were used as controls. All mice were maintained under specific pathogen–free conditions and were handled in accordance with the Guidelines for Animal Experiments of the Institute of Medical Science, The University of Tokyo (Tokyo, Japan). E0.5 was defined as the first morning a vaginal plug was observed. Pregnant female mice were checked every morning and evening, and discovery of new pups was marked as day 1.

Antibodies and reagents.

Rabbit anti–mouse keratin-5 antibody was purchased from Covance. Biotinylated UEA-1 was obtained from Vector Laboratories. The APC-Cy7–conjugated rat anti–mouse CD45, PE-Cy7–conjugated TER119, and PE-conjugated anti–mouse EpCAM (G8.8) antibodies were obtained from BD or BioLegend. The PE-conjugated rat anti-CD45, PE-conjugated mouse TER-119, FITC-conjugated mouse MHC II (M5/114.15.1), and APC-conjugated Aire antibodies were obtained from eBioscience. PE-conjugated CD80, CD86, CD40, and PD-L1 antibodies, purified anti–mouse CD16/32, FITC-conjugated ant–mouse EpCAM (G8.8), APC-Cy7–conjugated CD4, PE–Cy-7–conjugated CD8, and APC-conjugated GITR antibodies were obtained from BioLegend. PE-conjugated CD8, FITC-conjugated CD4, and FITC-conjugated CD3e antibodies were obtained from BD. PE-conjugated anti-Foxp3 (FJK-16s) was purchased from eBioscience. 7-aminoactinomycin D and recombinant mouse RANKL were purchased from Wako Pure Chemical Industries. Anti–mouse OPG antibody was purchased from R&D Systems. A RANK-Fc chimera was obtained from Sigma-Aldrich. Alexa Fluor 546–conjugated streptavidin and Alexa Fluor 488–conjugated anti–rabbit IgG were obtained from Invitrogen.

Flow cytometric analysis and cell sorting.

Whole thymus was used to prepare a single-cell suspension by digestion in RPMI 1640 medium containing collagenase/disperse (Roche) and DNase I (Sigma-Aldrich). Dead cells were excluded from the analysis after 7-aminoactinomycin D staining. After the Fc-receptor was blocked with CD16/32 antibody, the cells were stained with the indicated antibodies. For the analysis of T reg cells, thymocytes were prepared and used for nuclear staining according to the supplier’s protocol (Foxp3 staining buffer set; eBioscience) and analyzed using a fluorescence-activated cell sorter (Epics XL [Beckmann Coulter] or Canto II [BD]). The single-cell suspension was separated using a Percoll gradient to obtain a TEC-rich fraction, which was stained with anti-CD45, anti–TER-119, anti-EpCAM, and UEA-1 antibodies and sorted using a cell sorter (Aria; BD).

FTOC.

Thymic lobes were isolated from E15 embryos and were cultured on Nucleopore filters (Whatman) placed in R10 medium containing RPMI 1640 (Invitrogen) supplemented with 10% FBS (Equitech-Bio), 2 mM l-glutamine (Wako Pure Chemical Industries), 100 U/ml penicillin (Banyu Pharmaceutical), 100 µg/ml streptomycin (Meiji Seika Kaisha), and 50 µM 2-mercaptoethanol (Wako Pure Chemical Industries). FTOCs proceeded for 7 d and were subsequently analyzed by flow cytometry to investigate mTEC and T reg cell development in vitro. To isolate fetal thymic stromal (2DG-FTOC) cells, thymic lobes were cultured in the presence of 1.35 mM 2DG (Sigma-Aldrich) for 4 d. In some experiments, the 2DG-FTOC cells were further cultured in R10 with 1 µg/ml recombinant RANKL. For the blocking experiments, a RANK-Fc chimera (5 µg/ml) was added to the medium. For each day indicated after ligation, 2DG-FTOC was recovered and used for the analyses. For the transplantation experiments, the 2DG-FTOC was further cultured for 1 d without 2DG to eliminate 2DG before transplantation.

Transplantation of 2DG-FTOC into nude mice and tumor development.

2DG-FTOCs (six lobes each) were transplanted to the kidney capsule of 6-wk-old female nude mice. For tumor transfer experiments, Meth A (5 × 106 cells) was injected subcutaneously into the nude mice receiving 2DG-FTOC at 6 wk after transplantation. Tumor sizes were measured on days 10, 12, and 15 after transplantation. At day 15, the recipient mice were killed, and the inguinal lymph nodes were prepared for flow cytometric analysis. For the carcinogen-induced model, 50 µg MCA was injected subcutaneously into nude mice receiving 2DG-FTOC.

Histopathology and immunohistochemistry.

For immunohistochemistry, tissues were embedded in OCT compound (Sakura) and frozen in liquid nitrogen. Cryostat sections (6-µm thick) were fixed with ice-cold acetone and incubated with primary antibody for 1 h at room temperature. The slides were subsequently incubated with secondary antibody for 40 min at room temperature. For immunostaining of Meth A cells, cells were fixed with methanol and incubated with serum (100× dilution) for 1 h at room temperature and then incubated with secondary antibody (anti–mouse IgG–Alexa Fluor 488) and 1 µg/ml propidium iodide for 40 min at room temperature. Confocal color images were obtained using an FV1000D (Olympus) at 20 or 60 (Fig. 6 E) magnification and Fluoview software (Olympus).

qRT-PCR.

Total RNA was extracted using an RNeasy micro kit (QIAGEN) or TRIzol (Invitrogen) and was subjected to random-primed reverse transcription using a Primescript II first strand cDNA synthesis kit (Takara Bio Inc.). Real-time qPCR was performed using an ABI PRISM 7300 Sequence Detection System (Applied Biosystems) and SYBR Green Master Mix (Roche or TOYOBO). The primers used were 36B4: 5′-TCCAGGCTTTGGGCATCA-3′ and 5′-CTTTATCAGCTGCACATCACT-3′; Gapdh: 5′-ACCATGTAGTTGAGGTCAATGAAGG-3′ and 5′-GGTGAAGGTCGGTGTGAACG-3′; Spib: 5′-CTGCAAGCCCTTCAGTTACC-3′ and 5′-AAAGGCAGCAGTAGCAGGAT-3′; Aire: 5′-GGTTCTGTTGGACTCTGCCCTG-3′ and 5′-TGTGCCACGACGGAGGTGAG-3′; Spt1: 5′-GTGTTGCTTGGTGTTTCCAC-3′ and 5′-GTGTTGCTTGGTGTTTCCAC-3′; Rank: 5′-GCTGGCTACCACTGGAACTC-3′ and 5′-GTGCAGTTGGTCCAAGGTTT-3′; Spib1: 5′-CTCTGAACCACCATGCTTGCT-3′ and 5′-TCCTTCTGGGTACAAACAGCTTAA-3′; Spib2: 5′-AGGGCGGCCCTGACAT-3′ and 5′-TCCTTCTGGGTACAAACAGCTTAA-3′; Opg: 5′-GGGCGTTACCTGGAGATCG-3′ and 5′-GAGAAGAACCCATCTGGACATTT-3′; Ccl19: 5′-GCTAATGATGCGGAAGACTG-3′ and 5′-ACTCACATCGACTCTCTAGG-3′; Col2: 5′-AGAACAGCATCGCCTACCTG-3′ and 5′-CTTGCCCCACTTACCAGTGT-3′; and Csnb: 5′-GGCACAGGTTGTTCAGGCTT-3′ and 5′-AAGGAAGGGTGCTACTTGCTG-3′.

CpG methylation analysis.

Cell pellets were subjected to bisulfite treatment using the EZ DNA methylation-Direct kit (Zymo Research) according to the manufacturer’s instructions. PCR was performed using treated DNA obtained from 200–300 cells. For bisulfite sequencing, PCR products were cloned into a pGEM-T easy vector (Promega), and >16 clones were sequenced using the ABI 3130 sequencer and Big-Dye terminator kit (Applied Biosystems).

Preparation of tumor-infiltrating lymphocytes.

Tumors were minced with razor blades and digested in RPMI 1640 (Wako Pure Chemical Industries) containing 1 mg/ml collagenase D (Roche), 0.1 mg/ml DNase I, and 10% FBS for 1 h, with shaking. The remaining undigested fragments were passed through a needle. After addition of EDTA (final concentration: 10 mM), the cell suspension was filtered through a nylon mesh. The cells were collected and resuspended in 5 ml RPMI 1640 containing 45% Percoll and 10% FBS, and the mixture was layered on top of the same volume of 67.5% Percoll. After the cells were centrifuged at 800 g for 20 min at room temperature, the layer between 45% and 67.5% Percoll was collected.

Statistical analysis.

P-values were calculated using Student’s t test, Mann–Whitney U test with two-tailed distribution, and two-sample equivalent variance parameters. For statistical analysis of the tumor-free rate, the log-rank test was performed.

This work was supported by a Grant-in-Aid for Scientific Research from the Precursory Research for Embryonic Science and Technology (PRESTO) program of the Japan Science and Technology Agency (to T. Akiyama), grants from the Japanese Society for the Promotion of Science (to T. Akiyama), Takeda Science Foundation (to T. Akiyama), Princess Takamatsu Cancer Research Fund (to T. Akiyama), grants for Priority Area Research (to T. Akiyama and J.-i. Inoue), and a Grant-in-Aid for Challenging Exploratory Research (to T. Akiyama).

The authors declare no competing financial interests.

Abramson
,
J.
,
M.
Giraud
,
C.
Benoist
, and
D.
Mathis
.
2010
.
Aire’s partners in the molecular control of immunological tolerance
.
Cell.
140
:
123
135
.
Aichinger
,
M.
,
M.
Hinterberger
, and
L.
Klein
.
2012
.
Probing gene function in thymic epithelial cells
.
Eur. J. Cell Biol.
91
:
24
30
.
Akiyama
,
T.
,
S.
Maeda
,
S.
Yamane
,
K.
Ogino
,
M.
Kasai
,
F.
Kajiura
,
M.
Matsumoto
, and
J.
Inoue
.
2005
.
Dependence of self-tolerance on TRAF6-directed development of thymic stroma
.
Science.
308
:
248
251
.
Akiyama
,
T.
,
Y.
Shimo
,
H.
Yanai
,
J.
Qin
,
D.
Ohshima
,
Y.
Maruyama
,
Y.
Asaumi
,
J.
Kitazawa
,
H.
Takayanagi
,
J.M.
Penninger
, et al
.
2008
.
The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymic medullary microenvironment and self-tolerance
.
Immunity.
29
:
423
437
.
Akiyama
,
T.
,
M.
Shinzawa
, and
N.
Akiyama
.
2012a
.
RANKL-RANK interaction in immune regulatory systems
.
World J. Orthop.
3
:
142
150
.
Akiyama
,
T.
,
M.
Shinzawa
, and
N.
Akiyama
.
2012b
.
TNF receptor family signaling in the development and functions of medullary thymic epithelial cells
.
Front. Immunol.
3
:
278
.
Anderson
,
G.
, and
Y.
Takahama
.
2012
.
Thymic epithelial cells: working class heroes for T cell development and repertoire selection
.
Trends Immunol.
33
:
256
263
.
Anderson
,
G.
,
E.J.
Jenkinson
, and
H.R.
Rodewald
.
2009
.
A roadmap for thymic epithelial cell development
.
Eur. J. Immunol.
39
:
1694
1699
.
Bartholdy
,
B.
,
C.
Du Roure
,
A.
Bordon
,
D.
Emslie
,
L.M.
Corcoran
, and
P.
Matthias
.
2006
.
The Ets factor Spi-B is a direct critical target of the coactivator OBF-1
.
Proc. Natl. Acad. Sci. USA.
103
:
11665
11670
.
Bouneaud
,
C.
,
P.
Kourilsky
, and
P.
Bousso
.
2000
.
Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion
.
Immunity.
13
:
829
840
.
Burkly
,
L.
,
C.
Hession
,
L.
Ogata
,
C.
Reilly
,
L.A.
Marconi
,
D.
Olson
,
R.
Tizard
,
R.
Cate
, and
D.
Lo
.
1995
.
Expression of relB is required for the development of thymic medulla and dendritic cells
.
Nature.
373
:
531
536
.
Chen
,
H.M.
,
D.A.
Gonzalez
,
H.S.
Radomska
,
M.T.
Voso
,
Z.
Sun
,
P.
Zhang
,
D.E.
Zhang
, and
D.G.
Tenen
.
1998
.
Two promoters direct expression of the murine Spi-B gene, an Ets family transcription factor
.
Gene.
207
:
209
218
.
Cowan
,
J.E.
,
S.M.
Parnell
,
K.
Nakamura
,
J.H.
Caamano
,
P.J.
Lane
,
E.J.
Jenkinson
,
W.E.
Jenkinson
, and
G.
Anderson
.
2013
.
The thymic medulla is required for Foxp3+ regulatory but not conventional CD4+ thymocyte development
.
J. Exp. Med.
210
:
675
681
.
Dontje
,
W.
,
R.
Schotte
,
T.
Cupedo
,
M.
Nagasawa
,
F.
Scheeren
,
R.
Gimeno
,
H.
Spits
, and
B.
Blom
.
2006
.
Delta-like1-induced Notch1 signaling regulates the human plasmacytoid dendritic cell versus T-cell lineage decision through control of GATA-3 and Spi-B
.
Blood.
107
:
2446
2452
.
Garrett-Sinha
,
L.A.
,
G.H.
Su
,
S.
Rao
,
S.
Kabak
,
Z.
Hao
,
M.R.
Clark
, and
M.C.
Simon
.
1999
.
PU.1 and Spi-B are required for normal B cell receptor-mediated signal transduction
.
Immunity.
10
:
399
408
.
Gray
,
D.
,
J.
Abramson
,
C.
Benoist
, and
D.
Mathis
.
2007
.
Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire
.
J. Exp. Med.
204
:
2521
2528
.
Guerau-de-Arellano
,
M.
,
M.
Martinic
,
C.
Benoist
, and
D.
Mathis
.
2009
.
Neonatal tolerance revisited: a perinatal window for Aire control of autoimmunity
.
J. Exp. Med.
206
:
1245
1252
.
Hauri-Hohl
,
M.
,
S.
Zuklys
,
G.A.
Holländer
, and
S.F.
Ziegler
.
2014
.
A regulatory role for TGF-β signaling in the establishment and function of the thymic medulla
.
Nat. Immunol.
15
:
554
561
.
Heinz
,
S.
,
C.
Benner
,
N.
Spann
,
E.
Bertolino
,
Y.C.
Lin
,
P.
Laslo
,
J.X.
Cheng
,
C.
Murre
,
H.
Singh
, and
C.K.
Glass
.
2010
.
Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities
.
Mol. Cell.
38
:
576
589
.
Hikosaka
,
Y.
,
T.
Nitta
,
I.
Ohigashi
,
K.
Yano
,
N.
Ishimaru
,
Y.
Hayashi
,
M.
Matsumoto
,
K.
Matsuo
,
J.M.
Penninger
,
H.
Takayanagi
, et al
.
2008
.
The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator
.
Immunity.
29
:
438
450
.
Hinterberger
,
M.
,
M.
Aichinger
,
O.
Prazeres da Costa
,
D.
Voehringer
,
R.
Hoffmann
, and
L.
Klein
.
2010
.
Autonomous role of medullary thymic epithelial cells in central CD4+ T cell tolerance
.
Nat. Immunol.
11
:
512
519
.
Jones
,
P.A.
, and
S.B.
Baylin
.
2007
.
The epigenomics of cancer
.
Cell.
128
:
683
692
.
Kajiura
,
F.
,
S.
Sun
,
T.
Nomura
,
K.
Izumi
,
T.
Ueno
,
Y.
Bando
,
N.
Kuroda
,
H.
Han
,
Y.
Li
,
A.
Matsushima
, et al
.
2004
.
NF-κB-inducing kinase establishes self-tolerance in a thymic stroma-dependent manner
.
J. Immunol.
172
:
2067
2075
.
Kanaya
,
T.
,
K.
Hase
,
D.
Takahashi
,
S.
Fukuda
,
K.
Hoshino
,
I.
Sasaki
,
H.
Hemmi
,
K.A.
Knoop
,
N.
Kumar
,
M.
Sato
, et al
.
2012
.
The Ets transcription factor Spi-B is essential for the differentiation of intestinal microfold cells
.
Nat. Immunol.
13
:
729
736
.
Khan
,
I.S.
,
M.L.
Mouchess
,
M.L.
Zhu
,
B.
Conley
,
K.J.
Fasano
,
Y.
Hou
,
L.
Fong
,
M.A.
Su
, and
M.S.
Anderson
.
2014
.
Enhancement of an anti-tumor immune response by transient blockade of central T cell tolerance
.
J. Exp. Med.
211
:
761
768
.
Klein
,
L.
,
M.
Hinterberger
,
G.
Wirnsberger
, and
B.
Kyewski
.
2009
.
Antigen presentation in the thymus for positive selection and central tolerance induction
.
Nat. Rev. Immunol.
9
:
833
844
.
Kyewski
,
B.
, and
L.
Klein
.
2006
.
A central role for central tolerance
.
Annu. Rev. Immunol.
24
:
571
606
.
Lei
,
Y.
,
A.M.
Ripen
,
N.
Ishimaru
,
I.
Ohigashi
,
T.
Nagasawa
,
L.T.
Jeker
,
M.R.
Bösl
,
G.A.
Holländer
,
Y.
Hayashi
,
R.W.
Malefyt
, et al
.
2011
.
Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development
.
J. Exp. Med.
208
:
383
394
.
Leibbrandt
,
A.
, and
J.M.
Penninger
.
2008
.
RANK/RANKL: regulators of immune responses and bone physiology
.
Ann. N. Y. Acad. Sci.
1143
:
123
150
.
Lio
,
C.W.
, and
C.S.
Hsieh
.
2008
.
A two-step process for thymic regulatory T cell development
.
Immunity.
28
:
100
111
.
Liu
,
X.
,
P.
Invernizzi
,
Y.
Lu
,
R.
Kosoy
,
Y.
Lu
,
I.
Bianchi
,
M.
Podda
,
C.
Xu
,
G.
Xie
,
F.
Macciardi
, et al
.
2010
.
Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis
.
Nat. Genet.
42
:
658
660
.
Malchow
,
S.
,
D.S.
Leventhal
,
S.
Nishi
,
B.I.
Fischer
,
L.
Shen
,
G.P.
Paner
,
A.S.
Amit
,
C.
Kang
,
J.E.
Geddes
,
J.P.
Allison
, et al
.
2013
.
Aire-dependent thymic development of tumor-associated regulatory T cells
.
Science.
339
:
1219
1224
.
Mells
,
G.F.
,
J.A.
Floyd
,
K.I.
Morley
,
H.J.
Cordell
,
C.S.
Franklin
,
S.Y.
Shin
,
M.A.
Heneghan
,
J.M.
Neuberger
,
P.T.
Donaldson
,
D.B.
Day
, et al
Wellcome Trust Case Control Consortium 3
.
2011
.
Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis
.
Nat. Genet.
43
:
329
332
. (published erratum appears in Nat. Genet. 2011. 43:1164)
Mizuno
,
A.
,
N.
Amizuka
,
K.
Irie
,
A.
Murakami
,
N.
Fujise
,
T.
Kanno
,
Y.
Sato
,
N.
Nakagawa
,
H.
Yasuda
,
S.
Mochizuki
, et al
.
1998
.
Severe osteoporosis in mice lacking osteoclastogenesis inhibitory factor/osteoprotegerin
.
Biochem. Biophys. Res. Commun.
247
:
610
615
.
Ohshima
,
D.
,
J.
Qin
,
H.
Konno
,
A.
Hirosawa
,
T.
Shiraishi
,
H.
Yanai
,
Y.
Shimo
,
M.
Shinzawa
,
N.
Akiyama
,
R.
Yamashita
, et al
.
2011
.
RANK signaling induces interferon-stimulated genes in the fetal thymic stroma
.
Biochem. Biophys. Res. Commun.
408
:
530
536
.
Ray
,
D.
,
R.
Bosselut
,
J.
Ghysdael
,
M.G.
Mattei
,
A.
Tavitian
, and
F.
Moreau-Gachelin
.
1992
.
Characterization of Spi-B, a transcription factor related to the putative oncoprotein Spi-1/PU.1
.
Mol. Cell. Biol.
12
:
4297
4304
.
Rossi
,
S.W.
,
M.Y.
Kim
,
A.
Leibbrandt
,
S.M.
Parnell
,
W.E.
Jenkinson
,
S.H.
Glanville
,
F.M.
McConnell
,
H.S.
Scott
,
J.M.
Penninger
,
E.J.
Jenkinson
, et al
.
2007
.
RANK signals from CD4+3− inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla
.
J. Exp. Med.
204
:
1267
1272
.
Sasaki
,
I.
,
K.
Hoshino
,
T.
Sugiyama
,
C.
Yamazaki
,
T.
Yano
,
A.
Iizuka
,
H.
Hemmi
,
T.
Tanaka
,
M.
Saito
,
M.
Sugiyama
, et al
.
2012
.
Spi-B is critical for plasmacytoid dendritic cell function and development
.
Blood.
120
:
4733
4743
.
Schotte
,
R.
,
M.
Nagasawa
,
K.
Weijer
,
H.
Spits
, and
B.
Blom
.
2004
.
The ETS transcription factor Spi-B is required for human plasmacytoid dendritic cell development
.
J. Exp. Med.
200
:
1503
1509
.
Shinkura
,
R.
,
K.
Kitada
,
F.
Matsuda
,
K.
Tashiro
,
K.
Ikuta
,
M.
Suzuki
,
K.
Kogishi
,
T.
Serikawa
, and
T.
Honjo
.
1999
.
Alymphoplasia is caused by a point mutation in the mouse gene encoding Nf-κb-inducing kinase
.
Nat. Genet.
22
:
74
77
.
Sun
,
S.C.
2012
.
The noncanonical NF-κB pathway
.
Immunol. Rev.
246
:
125
140
.
Thu
,
Y.M.
, and
A.
Richmond
.
2010
.
NF-κB inducing kinase: a key regulator in the immune system and in cancer
.
Cytokine Growth Factor Rev.
21
:
213
226
.
Träger
,
U.
,
S.
Sierro
,
G.
Djordjevic
,
B.
Bouzo
,
S.
Khandwala
,
A.
Meloni
,
M.
Mortensen
, and
A.K.
Simon
.
2012
.
The immune response to melanoma is limited by thymic selection of self-antigens
.
PLoS ONE.
7
:
e35005
.
Weih
,
F.
,
D.
Carrasco
,
S.K.
Durham
,
D.S.
Barton
,
C.A.
Rizzo
,
R.P.
Ryseck
,
S.A.
Lira
, and
R.
Bravo
.
1995
.
Multiorgan inflammation and hematopoietic abnormalities in mice with a targeted disruption of RelB, a member of the NF-κB/Rel family
.
Cell.
80
:
331
340
.
Yagi
,
S.
,
K.
Hirabayashi
,
S.
Sato
,
W.
Li
,
Y.
Takahashi
,
T.
Hirakawa
,
G.
Wu
,
N.
Hattori
,
N.
Hattori
,
J.
Ohgane
, et al
.
2008
.
DNA methylation profile of tissue-dependent and differentially methylated regions (T-DMRs) in mouse promoter regions demonstrating tissue-specific gene expression
.
Genome Res.
18
:
1969
1978
.
Zhu
,
M.L.
,
A.
Nagavalli
, and
M.A.
Su
.
2013
.
Aire deficiency promotes TRP-1-specific immune rejection of melanoma
.
Cancer Res.
73
:
2104
2116
.

Abbreviations used:
2DG

2′-deoxyguanosine

FTOC

fetal thymus organ culture

MCA

methylcholanthrene

mTEC

medullary TEC

NIK

NF-κB–inducing kinase

OPG

osteoprotegerin

qPCR

quantitative RT-PCR

RANK

receptor activator of NF-κB

RANKL

RANK ligand

T-DMR

tissue-dependently and differentially methylated region

TEC

thymic epithelial cell

TSA

tissue-specific antigen

This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).