Trail+DX5Eomes natural killer (NK) cells arise in the mouse fetal liver and persist in the adult liver. Their relationships with TrailDX5+ NK cells remain controversial. We generated a novel Eomes-GFP reporter murine model to address this question. We found that Eomes NK cells are not precursors of classical Eomes+ NK cells but rather constitute a distinct lineage of innate lymphoid cells. Eomes NK cells are strictly dependent on both T-bet and IL-15, similarly to NKT cells. We observed that, in the liver, expression of T-bet in progenitors represses Eomes expression and the development of Eomes+ NK cells. Reciprocally, the bone marrow (BM) microenvironment restricts T-bet expression in developing NK cells. Ectopic expression of T-bet forces the development of Eomes NK cells, demonstrating that repression of T-bet is essential for the development of Eomes+ NK cells. Gene profile analyses show that Eomes NK cells share part of their transcriptional program with NKT cells, including genes involved in liver homing and NK cell receptors. Moreover, Eomes NK cells produce a broad range of cytokines, including IL-2 and TNF in vitro and in vivo, during immune responses against vaccinia virus. Thus, mutually exclusive expression of T-bet and Eomes drives the development of different NK cell lineages with complementary functions.

NK cells are innate lymphocytes that contribute to the early defense against intracellular pathogens and to the immunosurveillance of tumors. They have been recently reclassified as members of group 1 innate lymphoid cells (ILCs; Spits et al., 2013). They are defined by their perforin-dependent cytotoxic properties that can be enhanced upon activation by IL-15 (Verbist and Klonowski, 2012). Moreover, they produce large amounts of IFN-γ rapidly after pathogen infection, as well as other cytokines and chemokines that have important roles during the early steps of the immune reaction (Vivier et al., 2008). This property is shared with other innate lymphocytes such as NKT cells, γδ T cells, and adaptive lymphocytes such as memory CD8 T cells that behave like innate lymphocytes during the first phases of infections (Schoenborn and Wilson, 2007).

NK cells develop in the BM from pre-pro NK cells and NK cell precursors (Carotta et al., 2011; Fathman et al., 2011). Acquisition of the NK1.1 epitope marks their commitment to the NK cell lineage. Next, they undergo a sequential maturation program that includes four discrete steps marked by surface levels of CD27 and CD11b. The most immature NK cells do not express CD27 and CD11b and are found mainly in the liver (Chiossone et al., 2009). CD11b CD27+ NK cells express high levels of NKG2A and low levels of Ly49 receptors. They are found mainly in BM and LN. Upon acquisition of CD11b, NK cells massively proliferate in the BM (Kim et al., 2002b). CD11b+ CD27+ and CD11b+ CD27 correspond to mature NK cells mainly found at the periphery, display the full repertoire of Ly49 receptors, and have the highest cytotoxic potential (Hayakawa and Smyth, 2006). KLRG1 expression in CD11b+ CD27 NK cells marks cellular senescence (Huntington et al., 2007). At the CD11b+ CD27+ stage, NK cells acquire high expression of S1PR5 that induces their exit from the BM to the periphery (Walzer et al., 2007b). In parallel, they acquire expression of CX3CR1 (Grégoire et al., 2007) and progressively lose expression of CXCR3 and CXCR4 (Mayol et al., 2011), which have an impact on their trafficking. NK cells can also develop in the thymus (Vosshenrich et al., 2006) and NK cell precursors have been identified in human LNs (Freud et al., 2005), suggesting that NK cells may also develop at the periphery. Whether they develop through the same pathway as BM NK cells remains to be determined.

NK cell development is under the control of several transcription factors (TFs). The sequence of their respective actions is difficult to define as they often cross-regulate each other. E4BP4 (Gascoyne et al., 2009; Kamizono et al., 2009), Runx3 (Cruz-Guilloty et al., 2009; Lai and Mager, 2012), and ETS1 (Ramirez et al., 2012) act very early during NK cell development by inducing the expression of important downstream TFs, such as Id2 (Yokota et al., 1999) and Tox (Aliahmad et al., 2010), that repress many lymphoid genes and are also required for NK cell development. The T-box family TF T-bet and Eomesodermin (Eomes) are both expressed in mature NK cells (Gordon et al., 2012). They are believed to bind to the same DNA sequence but probably have both redundant and specific activities. Intlekofer et al. (2005) showed that mice with compound mutations of the genes encoding the TFs T-bet and Eomes were nearly devoid of several lineages dependent on IL-15, including memory CD8 T cells and mature NK cells, and that their cells had defective cytotoxic effector programming. They further showed that T-bet and Eomes cooperate to induce high expression of CD122, the β chain of IL-15. More recently they showed that Eomes-deficient mice lack all mature NK cells defined by high expression of integrin α2 (recognized by the DX5 antibody), whereas T-bet–deficient mice lack a population of liver NK cells with a Trail+ DX5 phenotype (Gordon et al., 2012). Trail+ DX5 NK cells had been previously described and shown to originate in the fetal liver and to progressively decrease after birth. They can secrete both IFN-γ and IL-13 but are less cytotoxic than DX5+ NK cells (Takeda et al., 2005). Their role in immunity is still unclear. Gordon et al. (2012) proposed that Trail+ DX5 NK cells correspond to immature NK cells that further differentiate into Trail DX5+ upon acquisition of Eomes expression. In this linear model of differentiation, Trail+ DX5 would depend on T-bet for developmental stability. However, several observations argue against this model. First, in the BM—the main site of NK cell development and therefore expected to contain a large pool of immature NK cells—there are very few Trail+ DX5 Eomes NK cells. Second, Peng et al. (2013) recently reported that liver DX5 NK cells did not convert into DX5+ NK cells upon adoptive transfer. Third, GATA3 has been shown to be important for the occurrence of liver but not BM NK cells, suggesting that liver and BM NK cells derive from different developmental pathways (Samson et al., 2003). The origin, developmental pathway, and functions of Trail+ DX5 Eomes NK cells thus remain unclear.

To clarify these issues, we generated a novel GFP reporter mouse model that allows the identification and tracing of Eomes-expressing cells. We used Eomes-GFP mice to analyze the relationship between Eomes-positive and -negative NK cells. Our results clearly demonstrate that Eomes NK cells develop in the liver and are not precursors of Eomes+ NK cells. Rather, Eomes NK cells constitute a novel subset of group 1 ILCs that resemble NKT cells in terms of trafficking machinery and cytokine expression, even though they are most closely related to Eomes+ NK cells in terms of global transcription. The development of Eomes NK cells is dependent on T-bet and IL-15. Their development is impaired in the BM because of an active repression of T-bet in the BM microenvironment that allows the development of classical Eomes+ NK cells instead. Reciprocally, the liver environment permits early expression of T-bet that represses Eomes and instructs the development of Trail+ DX5 Eomes NK cells. Eomes NK cells constitutively express mRNA encoding IL-2 and TNF and secrete these cytokines in the liver during antiviral immune responses, whereas Eomes+ NK cells only secrete IFN-γ. These data support complementary functions of NK cell subsets during immune responses.

A large fraction of liver NK cells does not express Eomes and displays an immature phenotype

To investigate the dynamics of Eomes expression in the immune system, we generated Eomes-GFP reporter mice by inserting an Ires-GFP cassette in the 3′ untranslated region of Eomes (Fig. 1, A–C). Unlike previous models (Arnold et al., 2009), the insertion of the reporter cassette did not alter endogenous Eomes protein expression and regulation, even when both Eomes alleles carried the GFP reporter (Fig. 1 D). Moreover, GFP expression faithfully reproduced the endogenous expression of Eomes (Fig. 1 E). The mean GFP fluorescence intensity was higher in spleen cells carrying two copies of Eomes-GFP alleles than in cells with only one copy, indicating a biallelic expression of Eomes (Fig. 1 D). Eomes-GFP was expressed in different spleen T cell subsets (unpublished data) and in NK cells (Fig. 2 A). The fraction of Eomes-GFP–negative NK cells was variable depending on the anatomical site. The liver contained the highest fraction (∼20–40%) of Eomes-GFP–negative NK cells, whereas the other organs contained 1–10% Eomes-GFP–negative cells (Fig. 2 A), which was in accordance with previously published results (Gordon et al., 2012). Eomes-GFP–negative NK cells displayed an immature phenotype on the basis of CD11b/CD27 staining (Fig. 2 B). Reciprocally, most CD11b CD27 NK cells were GFP in all organs. Previous articles reported the existence of Eomes-negative NK cells with a Trail+ DX5 phenotype in the liver (Takeda et al., 2005; Gordon et al., 2012). We confirmed that most Eomes-GFP–negative NK cells were Trail+ DX5 in the liver, but importantly a small fraction of Eomes-GFP+ is also Trail+ DX5, and conversely a fraction of Eomes-GFP–negative is Trail DX5+ (Fig. 2 C). The expression of Eomes is therefore not strictly correlated with that of Trail and DX5.

Eomes NK1.1+ CD3 cells are bona fide NK cells but resemble NKT cells

The localization of Eomes NK cells was highly skewed toward the liver (Figs. 2, A and D). This distribution was similar to that of NKT cells and different from that of classical Eomes+ NK cells (Fig. 2 D). To explore further the similarity between NKT cells and Eomes NK cells, we compared the expression of various cell surface or intracellular molecules between Eomes versus Eomes+ versus NKT cells. The lack of Eomes expression associated with low expression of both CD11b and CD49b (recognized by the DX5 antibody) was a feature of both Eomes NK cells and NKT cells (Fig. 2 E). Moreover, NKT cells and Eomes NK cells showed a striking similarity in terms of expression of cytokine receptors and receptors of the Ly49 family, with a high expression of CD127 (IL-7 receptor) and a lower expression of CD122 (IL-15 receptor β) and Ly49G2 and Ly49H compared with Eomes+ NK cells (Fig. 2 E). These data led us to hypothesize that Eomes NK cells could be NKT cells masquerading as NK cells because of low CD3/TCR expression, a phenomenon which we previously reported for γδ T cells (Stewart et al., 2007). However, a series of evidence argued against this possibility. First, unlike NKT cells, Eomes and Eomes+ NK cells expressed the NK cell marker NKp46 (Walzer et al., 2007a). Reciprocally, only NKT cells expressed intracellular CD3 (Fig. 2 E). Second, NKT cells expressed much higher levels of PLZF, the master NKT cell TF (Savage et al., 2008; Kovalovsky et al., 2008), than Eomes and Eomes+ NK cells (Fig. 2 E). Third, the fraction of Eomes cells within the NK cell gate was similar in WT and RAG2−/− mice (Fig. 2 F). Fourthly, both Eomes and Eomes+ NK cells were capable of killing YAC1 cells efficiently after overnight stimulation with IL-15 (Fig. 2 G). Thus, Eomes NK cells are bona fide NK cells that share several features with NKT cells.

Liver Eomes NK cells are not precursors of Eomes+ NK cells under physiological conditions

Previous studies have suggested that Eomes NK cells were immature precursors of Eomes+ NK cells on the basis of adoptive transfers of Trail+ DX5 (Gordon et al., 2012) NK cells or Trail+ (Takeda et al., 2005) NK cells into lymphopenic mice. To address this point more directly, we took advantage of Eomes-GFP reporter mice and FACS-sorted Eomes-GFP or Eomes-GFP+ NK cells. We first cultured these cells in vitro for 4 d in the presence of different cytokines. Eomes-GFP NK cells survived well but did not up-regulate Eomes expression upon culture with IL-15. IL-12 in the presence or absence of IL-18 did not induce Eomes expression by Eomes NK cells (Fig. 3, A and B; and unpublished data). Second, we adoptively transferred Eomes-GFP+ and Eomes-GFP into un-irradiated syngeneic CD45.1+ host mice. 2–3 wk after transfer, Eomes-GFP NK cells were found mainly in the liver, demonstrating the skewed tropism of these cells (Fig. 3 D, bottom). Moreover, no significant up-regulation of GFP was detected, even within cells homing to the spleen (Fig. 3, C and D). This was also true when donor Eomes-GFP NK cells were sorted from the spleen or when recipients were irradiated with a sublethal dose (unpublished data). Reciprocally, Eomes-GFP+ NK cells preferentially homed to the spleen and remained GFP+ (Fig. 3, C and D). Altogether, these results suggest that Eomes NK cells are not precursors of Eomes+ NK cells in homeostatic conditions and rather correspond to a distinct lineage of ILCs.

Liver Eomes NK cells arise in the liver and are dependent on T-bet and IL-15 but not IL-7 and TGF-β for their development

NK cells are thought to develop mainly in the BM in response to IL-15–dependent signals (Colucci et al., 2003). As Eomes NK cells are highly enriched in the liver, we sought to determine their site of development. Their high CD127 expression was reminiscent of thymic NK cells (Vosshenrich et al., 2006). However, the normal representation of Eomes NK cells in thymectomized mice (Fig. 4 A) excluded a thymic development of these cells. Unlike other NKp46+ innate lymphocytes, CD127 expression was also dispensable for their development (Fig. 4 B). Previous studies have shown that Trail+ DX5 NK cells are highly enriched in the fetal liver. Accordingly, we found that the livers of newborn mice were highly enriched in Eomes-GFP NK cells (Fig. 4 C, mean percentage of Eomes NK cells at birth: 90%). In contrast, in the BM and spleen of newborn mice, most NK cells were already Eomes+, which is another supportive piece of evidence of the dichotomy between Eomes and Eomes+ NK cell lineages (Fig. 4 C). In the BM, Eomes was not expressed in recently described NK cell progenitors (Fig. 4 D) but was induced upon acquisition of CD27 and NK1.1 (Fig. 2). The frequency of Eomes-GFP cells among liver NK cells progressively decreased after birth (Fig. 4 E). Altogether, these data strongly support the existence of a liver NK cell developmental pathway restricting Eomes expression.

The lack of Eomes expression in liver Eomes NK cells led us to test the role of T-bet in their development. We found that T-bet−/− mice lacked both NKT cells and Eomes NK cells, in accordance with previous observations (Townsend et al., 2004; Gordon et al., 2012), and further showing the similarity between Eomes NK cells and NKT cells (Fig. 4 F). NKT cells are dependent on both IL-15 (Ranson et al., 2003) and TGF-β (Doisne et al., 2009) for their development. To test the role of these cytokines in the development of Eomes NK cells, we used IL-15−/− mice and CD11c-dnTGFβRII mice. The latter mice express a dominant-negative form of the TGF-β receptor preventing TGF-β signaling in CD11c-positive cells (Laouar et al., 2005), including liver Eomes and Eomes+ NK cells (unpublished data). The number of liver Eomes NK cells was strongly reduced in IL-15−/− but not CD11c-dnTGFβRII mice (Fig. 4 G), showing that Eomes NK cells are dependent on IL-15 but not TGF-β for their development. Eomes+ NK cells were also absent from IL-15 KO mice but increased in CD11c-dnTGFβRII mice. Thus, Eomes NK cells and NKT cells share the dependence on T-bet and IL-15 for their development but only NKT cells are dependent on TGF-β.

T-bet expression is repressed in the BM allowing the development of Eomes+ NK cells

Having established the existence of two distinct pathways of NK cell development, we sought to identify the factors that instruct the commitment of precursor cells to each pathway. As T-bet is essential for the Eomes pathway, we hypothesized that T-bet could be differentially regulated in the BM and liver during early NK cell development. Remarkably, we found that T-bet expression was very low in BM NK cells compared with NK cells from all peripheral sites tested (Fig. 5 A), irrespective of the maturation status (Fig. 5 A). T-bet mRNA levels were similar between BM and spleen NK cells, suggesting that a posttranscriptional mechanism accounts for the difference in T-bet protein levels. The difference in T-bet levels between BM and spleen NK cells was true for different mouse strains (unpublished data) and for mice of different ages (Fig. 5 C). NK cell progenitors identified using the gating strategy shown in Fig. S1 (pre-pro NK cells and NKp) also expressed low levels of T-bet in the BM compared with those found in the liver (Fig. 5 D). Moreover, NK1.1+ T cells present in the BM also expressed lower levels of T-bet compared with those found in the periphery (Fig. 5 E). The low level of T-bet in BM NK cells was not due to an absence of T-bet induction in developing NK cells but rather to an active repression by the BM microenvironment. Indeed, when we adoptively transferred spleen or liver CD45.1+ NK cells into normal CD45.2 recipient mice, the level of T-bet in transferred NK cells that recirculated to the BM rapidly decreased to reach the same levels as that of recipient BM NK cells (Fig. 5 F). Reciprocally, when transferred, BM NK cells up-regulated T-bet expression upon migration to the spleen or to the liver while remaining T-betlow when homing to the BM (Fig. 5 F). Low T-bet expression in BM NK cells correlated with a modified pattern of expression of T-bet target genes. Indeed, WT BM NK cells were very similar to T-bet−/− spleen NK cells in terms of KLRG1, CD11b, and CD27 expression levels (Fig. 5 G).

To test the physiological relevance of BM-induced T-bet repression in NK cell development, we used transgenic mice expressing T-bet under the control of the CD2 promoter (Ishizaki et al., 2007). In these mice, T-bet is overexpressed both in NK and T cells in the BM and spleen (Fig. 6 A; Ishizaki et al., 2007). Results presented in Fig. 6 (B–D) showed that ectopic T-bet expression induced the neo development of a large population of Eomes NK cells in all organs, including the BM. T-bet transgenic Eomes NK cells expressed low levels of CD11b and DX5 and high levels of ITGA1 (Fig. 6 C), similar to WT Eomes NK cells (Fig. 2 and Fig. 7). Moreover, the size of the Eomes+ NK cell population decreased proportionally (Fig. 6 D). These results demonstrate that early induction of T-bet instructs the development of Eomes NK cells at the expense of Eomes+ NK cells.

One possible mechanism of T-bet action could be the repression of Eomes expression. To test this hypothesis, we compared the level of Eomes in Eomes+ NK cells isolated from mice expressing different doses of T-bet: T-bet transgenic, WT, T-bet+/−, and T-bet−/− mice. Remarkably, the level of Eomes in Eomes+ NK cells was inversely proportional to the level of T-bet, irrespective of the CD11b/CD27 NK cell subset analyzed (Fig. 6 E, compare top and bottom). In particular, T-bet−/− NK cells expressed very high level of Eomes whereas T-bet transgenic NK cells expressed very low levels of Eomes. Altogether, these results show that early induction of T-bet in liver NK cell progenitors instructs the development of Eomes NK cells via a mechanism that involves the repression of Eomes expression.

Global gene profile analyses reveal that NKT cells are more closely related to Eomes than to Eomes+ NK cells

Having shown the existence of two alternate NK cell developmental pathways, we sought to better understand their respective genetic program. We compared gene expression profile between liver Eomes, Eomes+ NK cells, and NKT cells. NKT cells were included in this analysis as we previously found similarities between Eomes and NKT cells. Globally, Eomes and Eomes+ NK cells were more related to each other than to NKT cells (Fig. 7 A). This segregation is due to the differential expression of T cell–specific (TCR–CD3 complex) transcripts and NK cell–specific (such as NCR1 encoding for NKp46 and KLRA8 encoding for Ly49H) transcripts by NKT and NK cell subsets, respectively (Fig. 7 B, red boxes). However, NKT cells were more closely related to Eomes than to Eomes+ NK cells in terms of global transcription (Euclidean distance 52,3 between NKT and Eomes+ vs. 43,2 between NKT and Eomes). When looking at differential gene expression between NKT versus Eomes+ NK cells (y axis) and Eomes versus Eomes+ NK cells (x axis), most genes were regulated similarly in Eomes NK cells and NKT cells (hence found on the diagonal of the figure), confirming the similarity between Eomes and NKT cells (Fig. 7 B). This included genes down-regulated compared with Eomes+ NK cells (Eomes and others) and genes up-regulated compared with Eomes+ NK cells (Fig. 7 B, green boxes; and Table S1). When looking at these genes, several interesting observations can be made. First, many of them were involved in cell trafficking. Eomes NK cells and NKT cells expressed high levels of CXCR6, ITGA1, ITGAV, and ITGB3 but did not express CD62L, CX3CR1, ITGB7, or S1PR5 (Fig. 7, C and D; and Table S1). Expression of CXCR6 and lack of CX3CR1, CD62L, ITGB7, S1PR1, and S1PR5 could explain their lack of recirculation and liver homing. Interestingly, Eomes NK cells express low levels of KLF2, which is known to induce CD62L, ITGB7, and S1PR1 (Carlson et al., 2006), which are required for lymphocyte recirculation (Arbonés et al., 1994; Wagner et al., 1998; Cyster and Schwab, 2012), suggesting that KLF2 is a target of Eomes. Second, Eomes NK cells expressed low levels of perforin and granzyme A/B (Table S1 and Fig. 8), but instead high levels of granzyme C and Trail, suggesting that both NK cell subsets kill target cells using different mechanisms. Eomes and Eomes+ NK cells could also recognize different types of target cells as they are clearly equipped with different sets of NK cell and other surface receptors (Table S1 and Fig. 8). Third, Eomes NK cells expressed constitutive levels of mRNA for different cytokines, including TNF and IL-2, and cytokine receptors (IL-7R, IL-17RD, IL-21R, and TGFBR), suggesting that they could be important in immune regulation. Fourthly, several genes involved in the Notch signaling pathway were enriched in Eomes NK cells (Table S1), suggesting that Notch could be important for their development as is the case for NKp46+ RORγt+ ILCs (Rankin et al., 2013). However, unlike the latter cells, Eomes NK cells do not express RORγt or constitutive levels of IL-22 (Table S1 and Fig. 8).

Eomes NK cells secrete a broad array of cytokines and contribute to immune responses against intracellular bacteria

A previous study showed that Trail+ DX5 NK cells secreted large amounts of IFN-γ and low amounts of IL-13 in response to different stimuli (Takeda et al., 2005). Based on the microarray results, we wanted to revisit this point and assessed the capacity of liver Eomes and Eomes+ NK cells to produce different cytokines at the single cell level in response to in vitro or in vivo stimuli. Upon stimulation with PMA/ionomycin, Eomes NK cells secreted a broad pattern of cytokines and chemokines including IL-2, IL-4, GM-CSF, TNF, CCL3, and IFN-γ (Fig. 9 A). IL-4 intracellular staining was weak but specific as shown using IL-4−/− NK cells (unpublished data). Their secretion of IL-2 that correlated with their constitutive expression of IL2 mRNA was particularly striking, even higher than that of NKT cells. Eomes NK cells did not express IL-17 or IL-10 (unpublished data). The cytokine secretion pattern of Eomes NK cells was very similar to that of NKT cells but in sharp contrast to that of Eomes+ NK cells which mainly secrete IFN-γ and CCL3. We then stimulated hepatic lymphocytes with cross-linking antibodies directed against NKp46, NK1.1, and NKG2D or with IL-12 combined with IL-18. Again, we found that Eomes NK cells secreted a broader pattern of cytokines than Eomes+ NK cells. In particular, they secreted TNF in response to activation via NKp46, NK1.1, or NKG2D (Fig. 9 B). Next, we treated mice with poly(I:C), a well-known activator of NK cells, or with α-GalCer, a glycolipid which strongly activates invariant NKT cells, and cytokine secretion was measured ex vivo. In these conditions, only IFN-γ was detected. Using both stimuli, Eomes NK cells expressed more IFN-γ than Eomes+ NK cells (Fig. 9 C). In particular, α-GalCer injection induced a coordinated secretion of IFN-γ by NKT cells and Eomes NK cells, suggesting that Eomes NK cells and NKT cells are located in the same hepatic niches. Finally, we assessed the cytokine response of hepatic lymphocytes after mouse infection with Francisella tularensis, a highly pathogenic intracellular bacterium, or with Vaccinia virus (VV). 48 h after infection with F. tularensis, hepatic NKT cells, Eomes NK cells, and Eomes+ NK cells were all activated and produced IFN-γ (Fig. 9 D). Upon VV infection, hepatic lymphocytes did not secrete much IFN-γ. Instead, Eomes NK cells and NKT cells, but not Eomes+ NK cells, secreted IL-2 and TNF early after infection, whereas Eomes+ NK cells secreted CCL3. Thus, Eomes NK cells contribute to the immune response against intracellular pathogens, such as F. tularensis or VV, by secreting a broad range of cytokines complementary to those produced by Eomes+ NK cells in the case of VV infection.

Research in the past few years has allowed the identification of several innate lymphoid subsets. They have been classified in three groups depending on the type of cytokines they produce (Spits et al., 2013). Group 1 ILCs express TH1 type cytokines such as IFN-γ and TNF. They include NK cells and ILC1, which are present in inflamed mucosal tissue (Vonarbourg et al., 2010) and differentiate from NCR+ ILC3s (Rankin et al., 2013) under the influence of IL-12 and IL-15. The data we present here classify liver Eomes NK cells as a novel subset of T-bet/IL-15–dependent group 1 ILC. These cells presumably derive from Id2+ common ILC precursors. They differentiate mostly in the liver and perhaps also in other organs. Our results show that all peripheral NK cells and precursors express high levels of T-bet, suggesting that Eomes+ NK cells only develop in the medullary environment whereas all other organs may rather drive the development of Eomes NK cells.

Using a novel model of Eomes-GFP reporter mice, we found that Eomes NK cells did not give rise to Eomes+ NK cells upon transfer to normal or irradiated recipient mice. This result strongly supports the existence of two alternative NK cell developmental pathways that segregate in time and space. Eomes NK cells appear earlier than Eomes+ NK cells, during fetal liver hematopoiesis, whereas Eomes+ NK cells constitute the main medullary subset, arising directly after birth and rapidly populating the periphery. Our results are in apparent contradiction with a previous study which showed that upon adoptive transfer into RAG−/− γc−/− immunoincompetent mice, Trail+ NK cells gave rise to Trail DX5+ Eomes+ NK cells (Gordon et al., 2012). One possible explanation for this discrepancy could be a minute contamination of the Trail+ population sorted by Gordon et al. (2012) with a few Eomes-positive cells. As Eomes+ NK cells express higher levels of CD122 and proliferate more than Eomes NK cells (unpublished data), they would rapidly outnumber Eomes NK cells in recipient mice. This emphasizes the fact that Eomes expression does not strictly correlate with the expression of any surface marker and underlies the superiority of the Eomes–GFP reporter system to tackle this question. How to discriminate between live Eomes and Eomes+ NK cells in the absence of GFP reporter or intracellular staining for Eomes? Our microarray analysis pointed to ITGA1 as a potential marker of Eomes NK cells, confirming a previous study (Peng et al., 2013). This is indeed the case in the liver, but not all Eomes NK cells are ITGA1+ in the spleen and vice versa (unpublished data). Thus, only an intracellular staining for Eomes unambiguously discriminates between Eomes and Eomes+ NK cells in all organs. The absence of a reliable marker for both NK cell subsets may be due to a redundant action of Eomes and T-bet on the expression of many of the surface markers commonly used to study NK cells. However, Eomes and T-bet may also cooperate to increase the likelihood of expression of other markers such as CD11b and DX5. Hence, we propose a model (Fig. 9 E) in which, depending on early expression of T-bet, Id2+ precursors may give rise either to Eomes NK cells with a CD11b CD27−/low Trail+/− DX5−/low phenotype or to Eomes+ NK cells. Eomes+ NK cells probably start their maturation process directly as CD11b CD27+ even though a small fraction of them may go through an earlier CD11b CD27 stage (Chiossone et al., 2009). They subsequently acquire CD11b expression before terminal maturation. T-bet expression, which is not influenced by the level of Eomes, is acquired upon migration of Eomes+ NK cells to the periphery and is involved in the completion of their differentiation process (Townsend et al., 2004; Soderquest et al., 2011). Importantly, our model of NK cell development does not exclude the possibility that Eomes NK cells may give rise to Eomes+ NK cells under certain conditions. Indeed, the plasticity of ILC subsets has already been appreciated (Rankin et al., 2013), and there may be special microenvironments that could induce the conversion of Eomes NK cells to Eomes+ NK cells. For example, IL-4 has been shown to strongly induce Eomes expression in CD8 T cells and could therefore induce Eomes expression in Eomes NK cells (Weinreich et al., 2010). A highly lymphopenic environment (e.g., RAG−/− γc−/− mice) could also artificially induce this conversion.

We found that T-bet was essential for the development of Eomes NK cells, confirming previous findings by Gordon et al. (2012). T-bet is therefore a master of development for various ILCs including gut ILC1, ILC3 (Powell et al., 2012; Sciumé et al., 2012; Klose et al., 2013; Rankin et al., 2013), and now liver Eomes NK cells. T-bet is also required for the development of NKT cells (Townsend et al., 2004). T-bet probably deeply imprints the genetic program of these various lymphoid subsets as illustrated by the important similarity between NKT cells and Eomes NK cells in terms of global gene expression. ChIP-Seq experiments will be required to understand which genes are directly regulated by T-bet or indirectly by other TFs modulated by T-bet. A previous study in CD4 T cells identified that T-bet induced a modification of the chromatin at the Eomes locus (Zhu et al., 2012). This correlates with our finding that T-bet expression inversely correlated with that of Eomes. Thus, early T-bet expression in liver NK cell progenitors could suppress Eomes expression and drive the differentiation of Eomes NK cells. Reciprocally, low T-bet expression in BM NK cells is probably not sufficient to repress Eomes and therefore allows the development of Eomes+ NK cells. Importantly, once Eomes is expressed, T-bet can probably no longer repress its expression. This may explain why mature peripheral NK cells that express high levels of T-bet express levels of Eomes similar to those expressed by BM NK cells.

The factors that repress T-bet expression in developing or recirculating NK cells in the BM remain to be determined. Our unpublished data exclude a role for major immune regulators such as IL-10, TGF-β, MHC-I, IL-4, MyD88, or type I IFN in this phenomenon. Similarly, the factors that induce T-bet at the periphery are unknown. Our microarray analysis showed that several members of the Notch signaling pathways were more expressed in Eomes NK cells compared with Eomes+ NK cells. It is therefore tempting to speculate that Notch could induce T-bet expression in peripheral NK cells, as previously suggested for NCR+ ILC3s (Rankin et al., 2013). Other good candidates include IL-12 and IFN-γ that are known to induce T-bet expression in T cells (Lazarevic and Glimcher, 2011).

Eomes and Eomes+ NK cells display important differences in terms of gene expression. As Eomes+ NK cells express both Eomes and T-bet whereas Eomes NK cells only express T-bet, this may reflect a nonredundant activity of Eomes compared with T-bet. In fact, mouse T-bet and Eomes have very similar DNA binding domains (90% identical). However, the other protein domains are totally different, which may explain the different functions of these TFs. In particular, they may recruit different co-activators or co-repressors of transcription. Eomes may be uniquely capable to induce the expression of Ly49 receptors and may also cooperate with T-bet to induce expression of perforin and IL2Rβ in NK cells. In contrast, Eomes could also repress a series of T cell genes, such as those encoding for IL7R or IL-2, and others such as Trail and ITGA1. Eomes and Eomes+ NK cells may therefore have different and perhaps complementary functions during immune responses. Eomes NK cells express high levels of CXCR3 and CXCR6 similarly to NKT cells (Kim et al., 2002a). These receptors are involved in the trafficking/homing of NK and NKT cells in the liver (Wald et al., 2006; Germanov et al., 2008). A previous study established that liver NK and NKT cells did not recirculate outside the liver (Thomas et al., 2011), which was later confirmed (Peng et al., 2013). Eomes NK cells may therefore be important for liver immune responses, especially in newborn mice. Our results show that they can secrete a wide variety of cytokines, including IL-2, TNF, GM-CSF, IFN-γ, and CCL3, whereas conventional NK cells only secrete IFN-γ and CCL3. This is not only true in vitro in response to different stimuli but also ex vivo after in vivo infection with VV. The receptors that trigger activation of Eomes NK cells remain to be identified, but our microarray analysis showed that Eomes NK cells express high levels of DNAM1 (CD226) and CD160, which are known co-stimulators of NK cells during antiviral responses. Cytokine secretion by Eomes NK cells may be important to amplify local immune responses and for the cross talk with other liver lymphocytes. In fact, we found that NKT cell activation with α-GalCer–induced Eomes+ NK cells, but especially Eomes NK cells, to produce IFN-γ, suggesting that Eomes NK cells and NKT cells are located in the same hepatic niches and thus exposed to the same inflammatory environment. Moreover, several articles reported that CXCR6+ (Paust et al., 2010) or DX5 (Peng et al., 2013) NK cells possess memory potential using models of contact hypersensitivity to haptens. Thus, Eomes NK cells may be important for recall responses in the liver.

Eomes NK cells could also be involved in the maintenance of the tolerogenic milieu in the liver under homeostatic conditions. Indeed, they express high levels of PD1-L, ICOSL, and IL-2, which may suppress T cell responses both directly (Terme et al., 2012) and indirectly via interaction with regulatory T cells (T reg cells). Interestingly, a recent study reported that T reg cells restrained the expansion of a population of spleen CD127+CD25+DX5Eomes NK cells (Gasteiger et al., 2013). Thus, there could be a preferential cross talk between T reg cells and Eomes NK cells. Consistent with this, RAG−/− T-bet−/− mice develop autoimmune-like syndromes, suggesting that T-bet–dependent innate lymphocytes may be important for limiting inflammation (Lazarevic and Glimcher, 2011).

Together, our findings establish the existence of two alternative pathways of NK cell development occurring in the BM and in the liver, respectively. Early T-bet levels are instrumental to determine the commitment of precursor cells to either lineage. The BM microenvironment restricts T-bet expression, thus allowing the development of classical Eomes+ NK cells. Reciprocally, the liver environment permits high expression of T-bet early on during NK cell development, which results in Eomes repression and the development of Eomes Trail+ DX5 Itga1+ NK cells that are specialized in cytokine secretion and may have immunoregulatory functions.

Mice, infections, and reagents

This study was performed in strict accordance with the French recommendations in the Guide for the ethical evaluation of experiments using laboratory animals and the European guidelines 86/609/CEE. All experimental studies were approved by the Animal Experimentation Ethics Committee CECCAPP. WT C57BL/6 and BALB/c mice were purchased from Charles River. CD127−/− (Peschon et al., 1994), RAG2−/− (Hao and Rajewsky, 2001), T-bet−/− (Finotto et al., 2002), IL-4−/−, CD11c-dnTGFβRII transgenic (Laouar et al., 2005), and CD2-T-bet transgenic (Ishizaki et al., 2007) mice have been previously described. IL-15−/− (Kennedy et al., 2000) and CX3CR1gfp/gfp (Jung et al., 2000) mice have been provided by S. Jung (Weizmann Institute, Rehovot, Israel) and J.P. Di Santo (Institut Pasteur, Paris, France). In some experiments, we also used C57BL/6 CD45.1 mice or C57BL/6 CD45.1 x CD45.2 mice that were bred in our animal house. Female mice 8–24 wk old were used unless specified. For infection, mice were injected intradermally with 5 × 103 cfu F. tularensis subspecies novicida strain U112 in 50 µl PBS or intraperitoneally with 2 × 105 PFU Vaccinia virus (Modified Vaccinia Virus Ankara). In some experiments, mice were injected with 150 µg poly(I:C) or 4 µg α-GalCer intraperitoneally. In some experiments, we used thymectomized mice. Thymectomies were performed as previously described (Pihlgren et al., 1996).

Generation of Eomes-GFP reporter/knockin mice

A 179 kb bacterial artificial chromosome containing Eomes 3′ region was selected (clone number RP23-448K1; CHORI). Using Red/ET cloning (Gene Bridges), an ires-GFP-loxP-tACE-Cre-PGK-gb2-neo-loxP cassette was inserted downstream the Eomes STOP codon. This cassette allows GFP expression under the control of Eomes regulatory regions. The rest of the cassette allows selection with neomycin in both bacteria and eukaryotic cells and is auto-excisable in male mice thanks to Cre expression under the control of the testis-specific Tace promoter and the loxP sites (Guia et al., 2011). The modified Eomes region and 3 kb on each side of the STOP codon were subcloned into PACYC177 using Red/ET cloning and included NotI sites allowing the release of the Eomes fragment. JM8.A3 C57BL/6N ES cells (Pettitt et al., 2009) were transfected with the NotI linearized targeting construct and cultured using standard culture conditions for ES cells. After G418 selection (150 µg/ml geneticin G418), ES clones were screened for homologous recombination by PCR analysis and Southern blotting. The occurrence of an appropriate homologous recombination event at the 5′ side was screened by PCR with the following primers: 5′-TTGACCTTGACCCTATGAGGGCTAT-3′ (forward primer); 5′-TAGGAATGCTCGTCAAGAAGACAG-3′ (reverse primer, 4.4 kb with knock-in allele). Next, a Southern blot was performed for some ES clones to further verify the recombination event using BamHI restriction enzyme digestion and probes amplified with the following primers: 5′ probe forward, 5′-CCAGGAGGAGGTTGCTACAG-3′; 5′ probe reverse, 5′-CAAAAGGATGAGCTGTCCAAG-3′; Neo Probe forward, 5′-GATCGGCCATTGAACAAGAT-3′; Neo probe reverse, 5′-ATACTTTCTCGGCAGGAGCA-3′.

Cells from each targeted ES clones were separately injected into C57BL/6 blastocysts to generate chimeric male mice that were mated with C57BL/6 females to obtain germline transmission of the mutated allele. Germline offspring of the GFP/KI genotype were routinely screened by PCR using the following primers in the gene encoding EGFP: 5′-AAGCTGACCCTGAAGTTCATCTGC-3′ (forward primer) and 5′-AAGTTCACCTTGATGCCGTTCTTCT-3′ (reverse primer). This pair of primers amplifies a 374 bp fragment. To generate homozygous mice, heterozygotes were mated and offspring screened using GFP PCR above and another set of primers amplifying the WT region across Exon6 and 3′UTR: 5′-ACTACCATGGACATCCAGAATGAGC-3′ (forward in Exon6) and 5′-CAAAGAACACAACAAAACACCACCA-3′ (reverse in 3′UTR). This pair of primers amplifies a 359 bp fragment in the case of a WT allele.

Generation of BM chimera

C57BL/6 CD45.1 x CD45.2 mice were irradiated twice at 450 rad within a 4-h interval. 4 h after the last irradiation, they received an intravenous injection of a 1:1 mixture of BM cells from WT CD45.1 and CD127−/− CD45.2 mice. BM chimeras were analyzed 6–12 wk after reconstitution.

Adoptive transfers

For assessing T-bet levels.

BM or spleen cells from CD45.1 mice were prepared and injected intravenously (2 × 107 cells of each genotype in PBS) into anesthetized CD45.2 C57BL/6 mice. 1 wk later, mice were sacrificed and transferred cells were analyzed in different organs by flow cytometry.

Fate mapping of Eomes-GFP NK cells.

We sorted Eomes-GFPNKp46+ and Eomes+NKp46+ from the liver and in some experiments from the spleen and BM. 20–50 × 104 cells were injected intravenously to recipient CD45.1 C57BL/6 congenic mice that were previously irradiated or not with a 500 rad dose. 2 wk later, mice were sacrificed and transferred NK cells were identified by their expression of CD45.2 in the spleen and liver. Their level of GFP was measured by flow cytometry.

Antibodies and flow cytometry

BM, blood, spleen, LN, and liver cells were isolated and stained as previously described. Cell counts were determined using an accuri C6 flow cytometer (BD). The following mAbs (eBioscience, BD, or BioLegend) were used: anti-CD19 (ebio1D3), anti-CD3 (145-2C11), anti-NK1.1 (PK136), anti NKp46 (29A1.4), anti-CD11b (M1/70), anti-CD27 (LG.7F9), anti-CD122 (5H4), anti-CD127 (A7R34), anti-CXCR3 (CXCR3-173), anti-CXCR4 (2B11) T-bet (ebio4B10), Eomes (Dan11mag), Ki67 (SolA15), KLRG1 (2F1), anti-CD49b (DX5), anti-ITGA1 (Ha31/8), anti-ITGAV (RMV7), anti-ITGA4 (9C10), anti-ITGB2 (C71/16), anti-ITGB3 (2C9), anti-ITGB7 (M293), anti-Trail (N2B2), anti-CD4 (GK1.5), anti-PLZF (Mags.21F7), anti-CD244 (ebio244f4), anti-Ly6D (49H4), anti-CD135 (A2F10), anti-CD45.1 (A20), anti-CD45.2 (104), anti-CD62L (Mel14), anti-RORγt (B2D), anti–granzyme A (3G8.5), anti–granzyme B (NGZB), anti-IL2 (JES6-5H4), anti-IL-4 (11b11), anti-GM-CSF (MP1-22E9), anti-CCL3 (DNT3CC), anti–TNF-α (MP6-XT22), anti–IFN-γ (XMG1), and relevant isotype controls. Intracellular stainings for T-bet, Eomes, RORγt, granzyme A, and granzyme B were performed using Foxp3 Fixation/Permeabilization Concentrate and Diluent. BrdU incorporation was measured using a kit (BD). For Eomes staining on Eomes-GFP spleen cells, the pH of all buffers was adjusted to 8 to preserve GFP fluorescence. To detect CXCR6 on NK cell surface, we used indirect staining with CXCL16-Fc-His tagged (R&D Systems), followed by anti-His (Miltenyi Biotec). Flow cytometry was performed on a FACSCanto, a FACSCanto II, or a FACS Fortessa (BD).

Quantitative RT-PCR

Lymphocyte subsets stained with the appropriate antibodies were sorted using a FACSAria cell sorter (BD). RNA was extracted using TRIzol reagent. We used High capacity RNA-to-cDNA kit (Applied Biosystems) to generate cDNA for RT-PCR. PCR was performed with a SybrGreen-based kit (FastStart Universal SYBR Green Master; Roche) on a StepOne plus instrument (Applied Biosystems). Primers were designed using the Roche software. The following primers were used: T-bet forward, 5′-CAACCAGCACCAGACAGAGA-3′; T-bet reverse, 5′-ACAAACATCCTGTAATGGCTTG-3′.

Cell culture and stimulation

For assessing differentiation of Eomes-NK cells.

We sorted Eomes-GFPNKp46+ and Eomes+NKp46+ from the liver of Eomes-GFP mice using flow cytometry. Cells were then cultured in complete medium in 24-well plates in the presence of 100 ng/ml IL-15 with or without 20 ng/ml IL-12. Cytokines were from R&D Systems.

For NK cell cytokine assays.

Hepatic lymphocytes were prepared and cultured with cytokines, coated antibodies (29A1.4, anti-NKp46; PK136, anti-NK1.1; CX5, anti-NKG2D), or 10 ng/ml PMA and 1 µg/ml ionomycin (Sigma-Aldrich) and Golgi-stop (BD). 25 ng/ml IL-12 and 5 ng/ml IL-18 (R&D Systems) were used. When NK cells were stimulated in vivo, hepatic lymphocytes were cultured in complete medium supplemented only with Golgi-stop. After stimulation/culture, intracellular stainings for cytokines were performed using the Cytofix/Cytoperm kit (BD) and cytokine production was measured by flow cytometry.

For cytotoxic assays.

Hepatic lymphocytes were prepared and cultured with 25 ng/ml IL-15 overnight. The next day, NK cells were sorted by flow cytometry and cultured 4 h with YAC1 cells previously labeled with CFSE at different E/T ratio. The percentage of TOPRO3+ YAC1 cells was then measured by flow cytometry.

Microarrays

Eomes-GFP+ NK1.1+ CD3, Eomes-GFPNK1.1+CD3, and NK1.1+CD3+ (NKT) hepatic cells were sorted by flow cytometry. Total RNA was extracted using TRIzol reagent according to the manufacturer’s instructions (Invitrogen). Quality and absence of genomic DNA contamination were assessed with a Bioanalyzer (Agilent Technologies). Total RNA from each sample was amplified, labeled, and hybridized to a mouse GeneChip HT MG-430 PM 16-Array Plate (Affymetrix). Affymetrix CEL files were analyzed in R using the Bioconductor suite of packages. Raw probe signals were background corrected using the maximum likelihood estimation of the normal-exponential mixture model (Silver et al., 2009) and normalized using the variance stabilization normalization (Huber et al., 2002), followed by a quantile normalization (Bolstad et al., 2003). Summarization was performed using the median-polish (Irizarry et al., 2003) using version 17.1 of the Entrez-Gene based reannotated chip description file (Dai et al., 2005). Non-informative genes were filtered using the I/NI algorithm (Talloen et al., 2007). Linear models were applied using the limma package to compute the mean expression level for each cell type corrected for the animal sex. Statistical contrasts were then applied to compute differential expression between the different cell types. The empirical Bayes method was used to compute moderated p-values that were then corrected for multiple comparisons using the Benjamini and Hochberg’s false discovery rate (FDR) controlling procedure. Microarray data were deposited into the Gene Expression Omnibus (accession no. GSE53486).

Statistical analyses

Statistical analyses were performed using two-tailed Student’s t tests or nonparametric tests when appropriate. These tests were run on the Prism (GraphPad Software). Levels of significance are expressed as p-values.

Online supplemental material

Fig. S1 shows the gating strategy used to identify pre-pro NK cells and NKp. Table S1 shows genes differentially expressed between Eomes+ and Eomes NK cells.

Authors thank the Plateau de Biologie Expérimentale de la Souris, and the flow cytometry facility of the SFR Biosciences Gerland. We also thank Steffen Jung and James P. Di Santo for providing the CX3CR1gfp/gfp and IL-15−/− mice.

The T. Walzer laboratory is supported by the FINOVI foundation, Agence Nationale de la Recherche (ANR JC sphinks), European Research council (ERC-Stg 281025), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université de Lyon, and Ecole Normale Supérieure de Lyon (ENS).

The authors declare no competing financial interests.

Aliahmad
P.
,
de la Torre
B.
,
Kaye
J.
.
2010
.
Shared dependence on the DNA-binding factor TOX for the development of lymphoid tissue-inducer cell and NK cell lineages
.
Nat. Immunol.
11
:
945
952
.
Arbonés
M.L.
,
Ord
D.C.
,
Ley
K.
,
Ratech
H.
,
Maynard-Curry
C.
,
Otten
G.
,
Capon
D.J.
,
Tedder
T.F.
.
1994
.
Lymphocyte homing and leukocyte rolling and migration are impaired in L-selectin-deficient mice
.
Immunity.
1
:
247
260
.
Arnold
S.J.
,
Sugnaseelan
J.
,
Groszer
M.
,
Srinivas
S.
,
Robertson
E.J.
.
2009
.
Generation and analysis of a mouse line harboring GFP in the Eomes/Tbr2 locus
.
Genesis.
47
:
775
781
.
Bolstad
B.M.
,
Irizarry
R.A.
,
Astrand
M.
,
Speed
T.P.
.
2003
.
A comparison of normalization methods for high density oligonucleotide array data based on variance and bias
.
Bioinformatics.
19
:
185
193
.
Carlson
C.M.
,
Endrizzi
B.T.
,
Wu
J.
,
Ding
X.
,
Weinreich
M.A.
,
Walsh
E.R.
,
Wani
M.A.
,
Lingrel
J.B.
,
Hogquist
K.A.
,
Jameson
S.C.
.
2006
.
Kruppel-like factor 2 regulates thymocyte and T-cell migration
.
Nature.
442
:
299
302
.
Carotta
S.
,
Pang
S.H.M.
,
Nutt
S.L.
,
Belz
G.T.
.
2011
.
Identification of the earliest NK-cell precursor in the mouse BM
.
Blood.
117
:
5449
5452
.
Chiossone
L.
,
Chaix
J.
,
Fuseri
N.
,
Roth
C.
,
Vivier
E.
,
Walzer
T.
.
2009
.
Maturation of mouse NK cells is a 4-stage developmental program
.
Blood.
113
:
5488
5496
.
Colucci
F.
,
Caligiuri
M.A.
,
Di Santo
J.P.
.
2003
.
What does it take to make a natural killer?
Nat. Rev. Immunol.
3
:
413
425
.
Cruz-Guilloty
F.
,
Pipkin
M.E.
,
Djuretic
I.M.
,
Levanon
D.
,
Lotem
J.
,
Lichtenheld
M.G.
,
Groner
Y.
,
Rao
A.
.
2009
.
Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs
.
J. Exp. Med.
206
:
51
59
.
Cyster
J.G.
,
Schwab
S.R.
.
2012
.
Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs
.
Annu. Rev. Immunol.
30
:
69
94
.
Dai
M.
,
Wang
P.
,
Boyd
A.D.
,
Kostov
G.
,
Athey
B.
,
Jones
E.G.
,
Bunney
W.E.
,
Myers
R.M.
,
Speed
T.P.
,
Akil
H.
et al
.
2005
.
Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data
.
Nucleic Acids Res.
33
:
e175
.
Doisne
J.-M.
,
Bartholin
L.
,
Yan
K.-P.
,
Garcia
C.N.
,
Duarte
N.
,
Le Luduec
J.-B.
,
Vincent
D.
,
Cyprian
F.
,
Horvat
B.
,
Martel
S.
et al
.
2009
.
iNKT cell development is orchestrated by different branches of TGF-β signaling
.
J. Exp. Med.
206
:
1365
1378
.
Fathman
J.W.
,
Bhattacharya
D.
,
Inlay
M.A.
,
Seita
J.
,
Karsunky
H.
,
Weissman
I.L.
.
2011
.
Identification of the earliest natural killer cell-committed progenitor in murine bone marrow
.
Blood.
118
:
5439
5447
.
Finotto
S.
,
Neurath
M.F.
,
Glickman
J.N.
,
Qin
S.
,
Lehr
H.A.
,
Green
F.H.Y.
,
Ackerman
K.
,
Haley
K.
,
Galle
P.R.
,
Szabo
S.J.
et al
.
2002
.
Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet
.
Science.
295
:
336
338
.
Freud
A.G.
,
Becknell
B.
,
Roychowdhury
S.
,
Mao
H.C.
,
Ferketich
A.K.
,
Nuovo
G.J.
,
Hughes
T.L.
,
Marburger
T.B.
,
Sung
J.
,
Baiocchi
R.A.
et al
.
2005
.
A human CD34(+) subset resides in lymph nodes and differentiates into CD56bright natural killer cells
.
Immunity.
22
:
295
304
.
Gascoyne
D.M.
,
Long
E.
,
Veiga-Fernandes
H.
,
de Boer
J.
,
Williams
O.
,
Seddon
B.
,
Coles
M.
,
Kioussis
D.
,
Brady
H.J.M.
.
2009
.
The basic leucine zipper transcription factor E4BP4 is essential for natural killer cell development
.
Nat. Immunol.
10
:
1118
1124
.
Gasteiger
G.
,
Hemmers
S.
,
Bos
P.D.
,
Sun
J.C.
,
Rudensky
A.Y.
.
2013
.
IL-2–dependent adaptive control of NK cell homeostasis
.
J. Exp. Med.
210
:
1179
1187
.
Germanov
E.
,
Veinotte
L.
,
Cullen
R.
,
Chamberlain
E.
,
Butcher
E.C.
,
Johnston
B.
.
2008
.
Critical role for the chemokine receptor CXCR6 in homeostasis and activation of CD1d-restricted NKT cells
.
J. Immunol.
181
:
81
91
.
Gordon
S.M.
,
Chaix
J.
,
Rupp
L.J.
,
Wu
J.
,
Madera
S.
,
Sun
J.C.
,
Lindsten
T.
,
Reiner
S.L.
.
2012
.
The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation
.
Immunity.
36
:
55
67
.
Grégoire
C.
,
Chasson
L.
,
Luci
C.
,
Tomasello
E.
,
Geissmann
F.
,
Vivier
E.
,
Walzer
T.
.
2007
.
The trafficking of natural killer cells
.
Immunol. Rev.
220
:
169
182
.
Guia
S.
,
Jaeger
B.N.
,
Piatek
S.
,
Mailfert
S.
,
Trombik
T.
,
Fenis
A.
,
Chevrier
N.
,
Walzer
T.
,
Kerdiles
Y.M.
,
Marguet
D.
et al
.
2011
.
Confinement of activating receptors at the plasma membrane controls natural killer cell tolerance
.
Sci. Signal.
4
:
ra21
.
Hao
Z.
,
Rajewsky
K.
.
2001
.
Homeostasis of peripheral B cells in the absence of B cell influx from the bone marrow
.
J. Exp. Med.
194
:
1151
1164
.
Hayakawa
Y.
,
Smyth
M.J.
.
2006
.
CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity
.
J. Immunol.
176
:
1517
1524
.
Huber
W.
,
von Heydebreck
A.
,
Sültmann
H.
,
Poustka
A.
,
Vingron
M.
.
2002
.
Variance stabilization applied to microarray data calibration and to the quantification of differential expression
.
Bioinformatics.
18
:
S96
S104
.
Huntington
N.D.
,
Tabarias
H.
,
Fairfax
K.
,
Brady
J.
,
Hayakawa
Y.
,
Degli-Esposti
M.A.
,
Smyth
M.J.
,
Tarlinton
D.M.
,
Nutt
S.L.
.
2007
.
NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation
.
J. Immunol.
178
:
4764
4770
.
Intlekofer
A.M.
,
Takemoto
N.
,
Wherry
E.J.
,
Longworth
S.A.
,
Northrup
J.T.
,
Palanivel
V.R.
,
Mullen
A.C.
,
Gasink
C.R.
,
Kaech
S.M.
,
Miller
J.D.
et al
.
2005
.
Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin
.
Nat. Immunol.
6
:
1236
1244
.
Irizarry
R.A.
,
Bolstad
B.M.
,
Collin
F.
,
Cope
L.M.
,
Hobbs
B.
,
Speed
T.P.
.
2003
.
Summaries of Affymetrix GeneChip probe level data
.
Nucleic Acids Res.
31
:
e15
.
Ishizaki
K.
,
Yamada
A.
,
Yoh
K.
,
Nakano
T.
,
Shimohata
H.
,
Maeda
A.
,
Fujioka
Y.
,
Morito
N.
,
Kawachi
Y.
,
Shibuya
K.
et al
.
2007
.
Th1 and type 1 cytotoxic T cells dominate responses in T-bet overexpression transgenic mice that develop contact dermatitis
.
J. Immunol.
178
:
605
612
.
Jung
S.
,
Aliberti
J.
,
Graemmel
P.
,
Sunshine
M.J.
,
Kreutzberg
G.W.
,
Sher
A.
,
Littman
D.R.
.
2000
.
Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion
.
Mol. Cell. Biol.
20
:
4106
4114
.
Kamizono
S.
,
Duncan
G.S.
,
Seidel
M.G.
,
Morimoto
A.
,
Hamada
K.
,
Grosveld
G.
,
Akashi
K.
,
Lind
E.F.
,
Haight
J.P.
,
Ohashi
P.S.
et al
.
2009
.
Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo
.
J. Exp. Med.
206
:
2977
2986
.
Kennedy
M.K.
,
Glaccum
M.
,
Brown
S.N.
,
Butz
E.A.
,
Viney
J.L.
,
Embers
M.
,
Matsuki
N.
,
Charrier
K.
,
Sedger
L.
,
Willis
C.R.
et al
.
2000
.
Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15–deficient mice
.
J. Exp. Med.
191
:
771
780
.
Kim
C.H.
,
Johnston
B.
,
Butcher
E.C.
.
2002a
.
Trafficking machinery of NKT cells: shared and differential chemokine receptor expression among Vα24+Vβ11+ NKT cell subsets with distinct cytokine-producing capacity
.
Blood.
100
:
11
16
.
Kim
S.
,
Iizuka
K.
,
Kang
H.-S.P.
,
Dokun
A.
,
French
A.R.
,
Greco
S.
,
Yokoyama
W.M.
.
2002b
.
In vivo developmental stages in murine natural killer cell maturation
.
Nat. Immunol.
3
:
523
528
.
Klose
C.S.N.
,
Kiss
E.A.
,
Schwierzeck
V.
,
Ebert
K.
,
Hoyler
T.
,
d’Hargues
Y.
,
Göppert
N.
,
Croxford
A.L.
,
Waisman
A.
,
Tanriver
Y.
,
Diefenbach
A.
.
2013
.
A T-bet gradient controls the fate and function of CCR6−RORγt+ innate lymphoid cells
.
Nature.
494
:
261
265
.
Kovalovsky
D.
,
Uche
O.U.
,
Eladad
S.
,
Hobbs
R.M.
,
Yi
W.
,
Alonzo
E.
,
Chua
K.
,
Eidson
M.
,
Kim
H.-J.
,
Im
J.S.
et al
.
2008
.
The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions
.
Nat. Immunol.
9
:
1055
1064
.
Lai
C.B.
,
Mager
D.L.
.
2012
.
Role of runt-related transcription factor 3 (RUNX3) in transcription regulation of natural cytotoxicity receptor 1 (NCR1/NKp46), an activating natural killer (NK) cell receptor
.
J. Biol. Chem.
287
:
7324
7334
.
Laouar
Y.
,
Sutterwala
F.S.
,
Gorelik
L.
,
Flavell
R.A.
.
2005
.
Transforming growth factor-β controls T helper type 1 cell development through regulation of natural killer cell interferon-γ
.
Nat. Immunol.
6
:
600
607
.
Lazarevic
V.
,
Glimcher
L.H.
.
2011
.
T-bet in disease
.
Nat. Immunol.
12
:
597
606
.
Mayol
K.
,
Biajoux
V.
,
Marvel
J.
,
Balabanian
K.
,
Walzer
T.
.
2011
.
Sequential desensitization of CXCR4 and S1P5 controls natural killer cell trafficking
.
Blood.
118
:
4863
4871
.
Paust
S.
,
Gill
H.S.
,
Wang
B.-Z.
,
Flynn
M.P.
,
Moseman
E.A.
,
Senman
B.
,
Szczepanik
M.
,
Telenti
A.
,
Askenase
P.W.
,
Compans
R.W.
,
von Andrian
U.H.
.
2010
.
Critical role for the chemokine receptor CXCR6 in NK cell–mediated antigen-specific memory of haptens and viruses
.
Nat. Immunol.
11
:
1127
1135
.
Peng
H.
,
Jiang
X.
,
Chen
Y.
,
Sojka
D.K.
,
Wei
H.
,
Gao
X.
,
Sun
R.
,
Yokoyama
W.M.
,
Tian
Z.
.
2013
.
Liver-resident NK cells confer adaptive immunity in skin-contact inflammation
.
J. Clin. Invest.
123
:
1444
1456
.
Peschon
J.J.
,
Morrissey
P.J.
,
Grabstein
K.H.
,
Ramsdell
F.J.
,
Maraskovsky
E.
,
Gliniak
B.C.
,
Park
L.S.
,
Ziegler
S.F.
,
Williams
D.E.
,
Ware
C.B.
et al
.
1994
.
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice
.
J. Exp. Med.
180
:
1955
1960
.
Pettitt
S.J.
,
Liang
Q.
,
Rairdan
X.Y.
,
Moran
J.L.
,
Prosser
H.M.
,
Beier
D.R.
,
Lloyd
K.C.
,
Bradley
A.
,
Skarnes
W.C.
.
2009
.
Agouti C57BL/6N embryonic stem cells for mouse genetic resources
.
Nat. Methods.
6
:
493
495
.
Pihlgren
M.
,
Dubois
P.M.
,
Tomkowiak
M.
,
Sjögren
T.
,
Marvel
J.
.
1996
.
Resting memory CD8+ T cells are hyperreactive to antigenic challenge in vitro
.
J. Exp. Med.
184
:
2141
2152
.
Powell
N.
,
Walker
A.W.
,
Stolarczyk
E.
,
Canavan
J.B.
,
Gökmen
M.R.
,
Marks
E.
,
Jackson
I.
,
Hashim
A.
,
Curtis
M.A.
,
Jenner
R.G.
et al
.
2012
.
The transcription factor T-bet regulates intestinal inflammation mediated by interleukin-7 receptor+ innate lymphoid cells
.
Immunity.
37
:
674
684
.
Ramirez
K.
,
Chandler
K.J.
,
Spaulding
C.
,
Zandi
S.
,
Sigvardsson
M.
,
Graves
B.J.
,
Kee
B.L.
.
2012
.
Gene deregulation and chronic activation in natural killer cells deficient in the transcription factor ETS1
.
Immunity.
36
:
921
932
.
Rankin
L.C.
,
Groom
J.R.
,
Chopin
M.
,
Herold
M.J.
,
Walker
J.A.
,
Mielke
L.A.
,
McKenzie
A.N.J.
,
Carotta
S.
,
Nutt
S.L.
,
Belz
G.T.
.
2013
.
The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway
.
Nat. Immunol.
14
:
389
395
.
Ranson
T.
,
Vosshenrich
C.A.J.
,
Corcuff
E.
,
Richard
O.
,
Laloux
V.
,
Lehuen
A.
,
Di Santo
J.P.
.
2003
.
IL-15 availability conditions homeostasis of peripheral natural killer T cells
.
Proc. Natl. Acad. Sci. USA.
100
:
2663
2668
.
Samson
S.I.
,
Richard
O.
,
Tavian
M.
,
Ranson
T.
,
Vosshenrich
C.A.J.
,
Colucci
F.
,
Buer
J.
,
Grosveld
F.
,
Godin
I.
,
Di Santo
J.P.
.
2003
.
GATA-3 promotes maturation, IFN-γ production, and liver-specific homing of NK cells
.
Immunity.
19
:
701
711
.
Savage
A.K.
,
Constantinides
M.G.
,
Han
J.
,
Picard
D.
,
Martin
E.
,
Li
B.
,
Lantz
O.
,
Bendelac
A.
.
2008
.
The transcription factor PLZF directs the effector program of the NKT cell lineage
.
Immunity.
29
:
391
403
.
Schoenborn
J.R.
,
Wilson
C.B.
.
2007
.
Regulation of interferon-γ during innate and adaptive immune responses
.
Adv. Immunol.
96
:
41
101
.
Sciumé
G.
,
Hirahara
K.
,
Takahashi
H.
,
Laurence
A.
,
Villarino
A.V.
,
Singleton
K.L.
,
Spencer
S.P.
,
Wilhelm
C.
,
Poholek
A.C.
,
Vahedi
G.
et al
.
2012
.
Distinct requirements for T-bet in gut innate lymphoid cells
.
J. Exp. Med.
209
:
2331
2338
.
Silver
J.D.
,
Ritchie
M.E.
,
Smyth
G.K.
.
2009
.
Microarray background correction: maximum likelihood estimation for the normal-exponential convolution
.
Biostatistics.
10
:
352
363
.
Soderquest
K.
,
Powell
N.
,
Luci
C.
,
van Rooijen
N.
,
Hidalgo
A.
,
Geissmann
F.
,
Walzer
T.
,
Lord
G.M.
,
Martín-Fontecha
A.
.
2011
.
Monocytes control natural killer cell differentiation to effector phenotypes
.
Blood.
117
:
4511
4518
.
Spits
H.
,
Artis
D.
,
Colonna
M.
,
Diefenbach
A.
,
Di Santo
J.P.
,
Eberl
G.
,
Koyasu
S.
,
Locksley
R.M.
,
McKenzie
A.N.J.
,
Mebius
R.E.
et al
.
2013
.
Innate lymphoid cells—a proposal for uniform nomenclature
.
Nat. Rev. Immunol.
13
:
145
149
.
Stewart
C.A.
,
Walzer
T.
,
Robbins
S.H.
,
Malissen
B.
,
Vivier
E.
,
Prinz
I.
.
2007
.
Germ-line and rearranged Tcrd transcription distinguish bona fide NK cells and NK-like γδ T cells
.
Eur. J. Immunol.
37
:
1442
1452
.
Takeda
K.
,
Cretney
E.
,
Hayakawa
Y.
,
Ota
T.
,
Akiba
H.
,
Ogasawara
K.
,
Yagita
H.
,
Kinoshita
K.
,
Okumura
K.
,
Smyth
M.J.
.
2005
.
TRAIL identifies immature natural killer cells in newborn mice and adult mouse liver
.
Blood.
105
:
2082
2089
.
Talloen
W.
,
Clevert
D.-A.
,
Hochreiter
S.
,
Amaratunga
D.
,
Bijnens
L.
,
Kass
S.
,
Göhlmann
H.W.H.
.
2007
.
I/NI-calls for the exclusion of non-informative genes: a highly effective filtering tool for microarray data
.
Bioinformatics.
23
:
2897
2902
.
Terme
M.
,
Ullrich
E.
,
Aymeric
L.
,
Meinhardt
K.
,
Coudert
J.D.
,
Desbois
M.
,
Ghiringhelli
F.
,
Viaud
S.
,
Ryffel
B.
,
Yagita
H.
et al
.
2012
.
Cancer-induced immunosuppression: IL-18–elicited immunoablative NK cells
.
Cancer Res.
72
:
2757
2767
.
Thomas
S.Y.
,
Scanlon
S.T.
,
Griewank
K.G.
,
Constantinides
M.G.
,
Savage
A.K.
,
Barr
K.A.
,
Meng
F.
,
Luster
A.D.
,
Bendelac
A.
.
2011
.
PLZF induces an intravascular surveillance program mediated by long-lived LFA-1–ICAM-1 interactions
.
J. Exp. Med.
208
:
1179
1188
.
Townsend
M.J.
,
Weinmann
A.S.
,
Matsuda
J.L.
,
Salomon
R.
,
Farnham
P.J.
,
Biron
C.A.
,
Gapin
L.
,
Glimcher
L.H.
.
2004
.
T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells
.
Immunity.
20
:
477
494
.
Verbist
K.C.
,
Klonowski
K.D.
.
2012
.
Functions of IL-15 in anti-viral immunity: multiplicity and variety
.
Cytokine.
59
:
467
478
.
Vivier
E.
,
Tomasello
E.
,
Baratin
M.
,
Walzer
T.
,
Ugolini
S.
.
2008
.
Functions of natural killer cells
.
Nat. Immunol.
9
:
503
510
.
Vonarbourg
C.
,
Mortha
A.
,
Bui
V.L.
,
Hernandez
P.P.
,
Kiss
E.A.
,
Hoyler
T.
,
Flach
M.
,
Bengsch
B.
,
Thimme
R.
,
Hölscher
C.
et al
.
2010
.
Regulated expression of nuclear receptor RORγt confers distinct functional fates to NK cell receptor-expressing RORγt+ innate lymphocytes
.
Immunity.
33
:
736
751
.
Vosshenrich
C.A.J.
,
García-Ojeda
M.E.
,
Samson-Villéger
S.I.
,
Pasqualetto
V.
,
Enault
L.
,
Richard-Le Goff
O.
,
Corcuff
E.
,
Guy-Grand
D.
,
Rocha
B.
,
Cumano
A.
et al
.
2006
.
A thymic pathway of mouse natural killer cell development characterized by expression of GATA-3 and CD127
.
Nat. Immunol.
7
:
1217
1224
.
Wagner
N.
,
Löhler
J.
,
Tedder
T.F.
,
Rajewsky
K.
,
Müller
W.
,
Steeber
D.A.
.
1998
.
L-selectin and β7 integrin synergistically mediate lymphocyte migration to mesenteric lymph nodes
.
Eur. J. Immunol.
28
:
3832
3839
.
Wald
O.
,
Weiss
I.D.
,
Wald
H.
,
Shoham
H.
,
Bar-Shavit
Y.
,
Beider
K.
,
Galun
E.
,
Weiss
L.
,
Flaishon
L.
,
Shachar
I.
et al
.
2006
.
IFN-γ acts on T cells to induce NK cell mobilization and accumulation in target organs
.
J. Immunol.
176
:
4716
4729
.
Walzer
T.
,
Bléry
M.
,
Chaix
J.
,
Fuseri
N.
,
Chasson
L.
,
Robbins
S.H.
,
Jaeger
S.
,
André
P.
,
Gauthier
L.
,
Daniel
L.
et al
.
2007a
.
Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46
.
Proc. Natl. Acad. Sci. USA.
104
:
3384
3389
.
Walzer
T.
,
Chiossone
L.
,
Chaix
J.
,
Calver
A.
,
Carozzo
C.
,
Garrigue-Antar
L.
,
Jacques
Y.
,
Baratin
M.
,
Tomasello
E.
,
Vivier
E.
.
2007b
.
Natural killer cell trafficking in vivo requires a dedicated sphingosine 1-phosphate receptor
.
Nat. Immunol.
8
:
1337
1344
.
Weinreich
M.A.
,
Odumade
O.A.
,
Jameson
S.C.
,
Hogquist
K.A.
.
2010
.
T cells expressing the transcription factor PLZF regulate the development of memory-like CD8+ T cells
.
Nat. Immunol.
11
:
709
716
.
Yokota
Y.
,
Mansouri
A.
,
Mori
S.
,
Sugawara
S.
,
Adachi
S.
,
Nishikawa
S.
,
Gruss
P.
.
1999
.
Development of peripheral lymphoid organs and natural killer cells depends on the helix-loop-helix inhibitor Id2
.
Nature.
397
:
702
706
.
Zhu
J.
,
Jankovic
D.
,
Oler
A.J.
,
Wei
G.
,
Sharma
S.
,
Hu
G.
,
Guo
L.
,
Yagi
R.
,
Yamane
H.
,
Punkosdy
G.
et al
.
2012
.
The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses
.
Immunity.
37
:
660
673
.

Abbreviations used:
ILC

innate lymphoid cell

TF

transcription factor

Author notes

C. Daussy, F. Faure, and K. Mayol contributed equally to this paper.

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/).

Supplementary data