Abstract
Although invariant Vα14+ natural killer T cells (NKT cells) are thought to be generated from CD4+CD8+ double-positive (DP) thymocytes, the developmental origin of CD4–CD8– double-negative (DN) NKT cells still remains unresolved. Here we provide definitive genetic evidence obtained, through studies of mice with DP-stage-specific ablation of expression of the gene encoding the recombinase component RAG-2 (Rag2) and by a fate-mapping approach, that supports the proposal of the existence of an alternative developmental pathway through which a fraction of DN NKT cells with strong T-helper-type-1 (TH1)-biased and cytotoxic characteristics develop from late DN-stage thymocytes, bypassing the DP stage. These findings provide new insight into understanding of the development of NKT cells and propose a role for timing of expression of the invariant T cell antigen receptor in determining the functional properties of NKT cells.
Similar content being viewed by others
Main
Natural killer T cells (NKT cells) are characterized by the expression of a unique T cell antigen receptor (TCR) α-chain composed of a single invariant α-chain variable region 14–α-chain joining region 18 (Vα14Jα18) in mice1 and Vα24Jα18 in humans2,3, both of which recognize almost identical glycolipid-binding sites contributed mainly by the proximal Jα18 region4. Thus, both human NKT cells and mouse NKT cells recognize the same glycolipid ligands, such as α-galactosylceramide (α-GalCer), in conjunction with the monomorphic major histocompatibility complex (MHC)-like antigen-presenting molecule CD1d5,6 and show cross-species reactivity7.
The majority of NKT cells are T-helper-type-1 (TH1)-type NKT cells, which have important protective roles in responses to tumors or pathogens due to their very rapid and powerful innate-like responses. In this context, both human CD4–CD8– (double-negative (DN)) NKT cells and mouse DN NKT cells, in contrast to CD4+ NKT cells, have been shown to elicit strong antitumor activity and TH1-type responses8,9,10. However, those functional differences between DN NKT cells and CD4+ NKT cells cannot be adequately explained by the currently accepted CD4+CD8+ (double-positive (DP)) pathway model for the development of NKT cells.
The DP pathway model is based on experiments that demonstrated the generation of NKT cells by intrathymic transfer of DP cells from Cd1d–/– mice into wild-type recipients11, as well as by fate-mapping experiments designed to trace cells that developed through the DP stage through the use of reporter mice that express Cre recombinase from the Cd4 promoter (Cd4-Cre) or the gene encoding the transcription factor RORγt (Rorc-Cre), together with a transgene with a loxP-flanked stop cassette expressed from the ubiquitous Rosa26 gene, for the expression of green fluorescent protein (GFP), in which GFP+ NKT cells were detected but GFP– NKT cells were not12. According to this DP pathway model, the DP thymic precursor cells that have rearranged an invariant Vα14Jα18 chain are positively selected on CD1d to give rise to DN NKT cells as well as CD4+ NKT cells13,14. However, it has also been shown that not all DN αβ T cells originate from DP-stage cells15 and also that the timing of TCR expression affects fate 'decisions'16. Therefore, questions about the influence of the timing of NKT cell TCR expression on the differentiation and function of DN NKT cells have remained unanswered.
To clarify the developmental origin and functional attributes of DN NKT cells, we reasoned that fate-mapping or conditional-gene-ablation approaches controlled by DP-stage-specific expression of Cre recombinase from the enhancer E8III (E8III-Cre) would be ideally suited for testing the possibility of generating NKT cells from the DN stage, because in this model, Cre is expressed specifically in DP-stage thymocytes without affecting cells in the DN stage17,18. Through the use of this approach, we were able to demonstrate the existence of an alternative NKT cell developmental pathway, distinct from the DP pathway, that originated at the DN stage of thymic ontogeny. This DN pathway 'preferentially' gave rise to TH1 NKT cells with strong cytotoxic potential and distinct distribution patterns in peripheral tissues. This adds new insight to the understanding of NKT cell development and suggests that the differentiation stage as well as the microenvironmental niche of precursor cells undergoing positive selection have an important role in determining their further developmental programs.
Results
Detection of NKT cells of DN-stage origin by fate mapping
To investigate whether NKT cells can be derived from DN-stage precursors through an alternative developmental pathway different from the DP pathway, we performed fate-mapping experiments by using mice bearing a reporter transgene with a loxP-flanked stop cassette expressed from the ubiquitous Rosa26 gene, for the expression of yellow fluorescent protein (YFP), controlled by the DP-stage-specific expression of E8III-Cre (E8III-Cre+Rosa26-YFP mice). In these E8III-Cre+Rosa26-YFP mice, the expression of E8III-Cre is regulated by the E8III enhancer and promoter elements of the gene encoding the co-receptor CD8α, which restricts its expression exclusively to DP thymocytes17,18. Therefore, the Cre-mediated deletion of the loxP-flanked stop cassette turns on the expression of the gene encoding the YFP reporter and thus all progeny of DP-stage thymocytes are labeled with YFP.
Flow cytometry of splenic TCRβ+ cells from E8III-Cre+Rosa26-YFP mice showed almost completely uniform expression of the YFP reporter by CD4+ T cells and CD8+ T cells (Fig. 1a), which indicated that conventional CD4+ T cells and CD8+ T cells were the progeny of DP thymocytes. Although the majority of DN TCRβ+ splenocytes were YFP+ cells of DP-stage-thymocyte origin, we observed that ∼14% of DN TCRβ+ cells were YFP– (Fig. 1a), which indicated that these YFP– TCRβ+ cells were likely to have differentiated from DN-stage thymocytes and to have bypassed the DP stage during thymic development. Further analysis of this population through the use of staining with α-GalCer-loaded CD1d dimers, in which unloaded CD1d dimers were used to exclude non-specific staining, revealed that ∼14% of YFP– DN TCRβ+ splenocytes were NKT cells (Fig. 1b). In comparison, the frequency of NKT cells among the YFP+ DN TCRβ+ fraction, which had developed through the DP stage, was ∼25% (Fig. 1b).
(a) Flow cytometry of cells from E8III-Cre+Rosa26-YFP mice (n = 6), showing the gating of viable TCRβ+B220– splenocytes by expression of CD4 and CD8 (far left), and expression of the YFP reporter by the electronically gated CD4+ TCRβ+ fraction (middle left), CD8+ TCRβ+ fraction (middle right) and DN TCRβ+ fraction (far right). Numbers in top corners of plots (middle and right) indicate percent YFP– cells (left corner) or YFP+ cells (right corner) in gates bracketed below, as follows (values, mean ± s.e.m.): YFP– cells, 5.3% ± 0.8% (CD4+), 4.0% ± 0.6% (CD8+) and 9.6% ± 1.0% (DN); YFP+ cells, 95.0% ± 0.8% (CD4+), 96.0% ± 0.6% (CD8+) and 90.0% ± 1.0% (DN). (b,c) Flow cytometry of DN thymocytes (n = 3 mice per genotype) (b) and splenocytes (n = 6 mice) (c) from C57BL/6 mice (B6) or E8III-Cre+Rosa26-YFP mice, purified by magnetic depletion with anti–mouse CD4 and CD8 microbeads and assessed with unloaded CD1d dimers (far left, b) or α-GalCer (GC)-loaded CD1d dimers. Numbers adjacent to outlined areas indicate percent CD1d-dimer+ NKT cells among DN TCRβ+ splenocytes (left) or among gated YFP– (middle) or YFP+ (right) DN TCRβ+ splenocytes (b), or percent YFP– cells (left) or YFP+ cells (right) among DN TCRβ+ splenocytes (c, top), or CD1d-dimer+ NKT cells among those gated DN TCRβ+ splenocytes (c, below). Data are from three independent experiments.
We then investigated the presence of YFP– DN NKT cells in the thymus of E8III-Cre+Rosa26-YFP mice (Fig. 1c). The results demonstrated the presence of YFP– TCRβ+ cells; ∼21% of these were mature NKT cells, as indicated by staining with α-GalCer–CD1d dimers (Fig. 1c). Although the majority of thymic DN NKT cells were of DP-stage-thymocyte origin (the absolute number of DN NKT cells was ∼2 × 105 YFP+ cells and ∼1 × 104 YFP– cells) (data not shown), we note that YFP– NKT cells represented a readily detectable population.
To confirm the results obtained with the E8III-Cre+Rosa26-YFP mice, we generated another, similar reporter model with Cd4-Cre (the widely used gold standard for fate-mapping experiments to trace cells that went through the DP stage) and determined whether the resultant Cd4-Cre+Rosa26-YFP mouse harbored YFP– NKT cells or not. Analysis of Cd4-Cre+Rosa26-YFP mice demonstrated the presence of YFP– cells among DN TCRβ+ splenocytes (Supplementary Fig. 1a) as well as YFP– NKT cells (Supplementary Fig. 1b). This confirmed the results obtained with the E8III-Cre+Rosa26-YFP mice. Together, the results of the fate-mapping experiments demonstrated that not all NKT cells were the progeny of DP thymocytes and further suggested the possibility that a fraction of the DN NKT cells developed directly from the thymic DN stage, which precedes the DP stage of thymic differentiation.
Conditional ablation of Rag2 in DP-stage thymocytes
To investigate the development of the NKT cells of DN-stage-thymocyte origin, we next used mice with conditional ablation of loxP-flanked Rag2 alleles (Rag2fl/fl) specifically at the DP stage. For this purpose, we introduced the E8III-Cre transgene into Rag2fl/fl mice to generate E8III-Cre+Rag2fl/fl mice in which the DP-stage-specific expression of Cre would excise Rag2fl/fl in DP thymocytes while leaving Rag2fl/fl in DN thymocytes intact.
To confirm the DP-stage-specific conditional deletion of Rag2fl/fl, we sorted DP and DN thymocyte fractions from E8III-Cre+Rag2fl/fl mice and their Rag2fl/fl (Cre–) littermates (which served as a control) (Fig. 2a), and assessed Rag2 expression by real-time quantitative RT-PCR (qPCR). Rag2 transcripts were selectively lower in abundance in DP thymocytes, while Rag2 expression in DN thymocytes was largely unaffected, in E8III-Cre+Rag2fl/fl mice relative to the expression of Rag2 in the control mice (Fig. 2b). Therefore, the E8III-Cre+Rag2fl/fl mouse was indeed a faithful experimental model of DN-stage-restricted Rag2 expression. Consequently, flow cytometry of thymocytes showed a major decrease in both the frequency and absolute number of CD4+ or CD8+ T cells in E8III-Cre+Rag2fl/fl mice relative to the abundance of these cells in the control mice, whereas cells in the DP fraction were slightly greater in abundance, by ∼10%, in E8III-Cre+Rag2fl/fl mice relative to their abundance in the control mice, but with no apparent change in the DN fraction (Fig. 2b,c), which suggested that the DP-stage-specific ablation of Rag2 resulted in a developmental block at the DP stage.
(a) Flow cytometry of thymocytes from E8III-Cre+Rag2fl/fl mice and their control littermates (Ctrl), showing the expression of CD4 and CD8 among viable (7-AAD–) cells. Numbers in quadrants indicate percent cells in each throughout. (b) qPCR analysis of Rag2 mRNA in sorted DP and DN thymic fractions obtained from E8III-Cre+Rag2fl/fl mice and their control littermates; results are presented relative to those of the internal-control gene Gapdh. (c) Absolute number of cells in thymocyte populations gated as in a. CD4SP, CD4+ single-positive; CD8SP, CD8+ single-positive. *P < 0.05 (unpaired t-test). Data are representative of three independent experiments (mean + s.e.m. of three biological replicates in b,c).
Detection of transcripts encoding Vα14Jα18 of DN-stage origin
To determine the effect of DP stage-specific RAG deletion on the rearrangement of an invariant Vα14Jα18 TCRα chain (encoded by Trav11 and Traj18, respectively), we sorted DP and DN thymocyte fractions from E8III-Cre+Rag2fl/fl mice, gating the cells as TCRβneg–loCD5loCD69– cells (Fig. 3a). We noted that the lower abundance of TCRβpos–hiCD5hiCD69hi thymocytes in E8III-Cre+Rag2fl/fl mice than in control mice (Fig. 3a and data not shown) seemed to suggest an efficient developmental block beyond the DP stage in E8III-Cre+Rag2fl/fl mice. qPCR analysis of these sorted thymocyte fractions revealed that DP-stage-specific ablation of Rag2 resulted in loss of Trav11Traj18 transcripts in the DP fraction, whereas these transcripts were readily detected in the DN thymocyte fraction (Fig. 3b).
(a) Flow cytometry of viable B220–CD1d-dimer–TCRβneg–lo DN and DP thymocyte fractions from E8III-Cre+Rag2fl/fl mice, showing the sorting strategy. Numbers adjacent to outlined areas indicate percent CD5loCD69– (pre-selection DP and DN) cells. (b) qPCR analysis of Trav11Traj18 mRNA in DP and DN thymic fractions from E8III-Cre+Rag2fl/fl mice sorted as in a (results presented as in Fig. 2b). ND, not detected. (c) Flow cytometry of DN thymocytes obtained from E8III-Cre+Rag2fl/fl mice and purified by depletion of CD4+, CD8+ and DP cells via magnetic microbeads and assessed by staining with unloaded CD1d dimers (staining control; left) or α-GalCer-loaded CD1d dimers (right). Numbers adjacent to outlined areas indicate percent CD1d-dimer+ NKT cells among gated DN TCRβ+ thymocytes. (d) Absolute number of CD1d-dimer+ DN NKT cells among DN thymocytes from control mice (n = 5) or E8III-Cre+Rag2fl/fl mice (n = 8). Data are representative of three independent experiments (mean + s.em. of n = 3 biological replicates in b; mean + s.em. in d).
As another definitive way to demonstrate ongoing gene rearrangement of Trav11Traj18 in the DN thymocyte fraction, we sorted bulk DN thymocytes from E8III-Cre+Rag2fl/fl mice and performed high-throughput next-generation sequencing of Trav11Trac transcripts (which encode Vα14 and the α-chain constant region (Cα)), regardless of the Jα-region-encoding gene being used. A total of 56,056 sequences encoding Vα14Jα chains were obtained, and among 3,002 total out-of-frame sequences, 293 (∼9.8%) were Trav11Traj18 (Table 1). Of particular note, analysis of the out-of-frame sequences encoding Vα14Jα chains revealed diverse use of Jα-encoding genes, such as Traj6, Traj9, Traj11, Traj18, Traj21, Traj26, Traj27, Traj32, Traj33, Traj34, Traj35, Traj38, Traj39, Traj42, Traj47, Traj48, Traj49, Traj53, Traj56 and Traj57 (Supplementary Fig. 2); this provided definitive evidence of ongoing random TCR α-chain rearrangement in the DN fraction of E8III-Cre+Rag2fl/fl mice. In addition, among 52,703 total productive sequences, 47,841 (∼90.8%) were in-frame Trav11Traj18 sequences (Table 1). The predominance of in-frame sequences encoding the invariant Vα14Jα18 chain (Trav11Traj18) among all the in-frame sequences encoding Vα14Jα chains would be best explained by the presence of positively selected mature NKT cells among the sorted bulk population of DN thymocytes. The presence of out-of-frame Trav11Traj18 rearrangements unambiguously demonstrated that Trav11Traj18 rearrangements were initiated, at least in part, at the DN stage.
NKT cells that developed through an alternative pathway
Next we investigated the presence of mature NKT cells positive for α-GalCer-loaded CD1d dimers in the DN TCRβ+ thymocyte fraction of E8III-Cre+Rag2fl/fl mice. Flow cytometry of these DN thymocytes demonstrated the presence of mature NKT cells of DN-stage origin, at a frequency of ∼67% among the gated DN TCRβ+ thymocytes (Fig. 3c), among which the absolute number of DN NKT cells from E8III-Cre+Rag2fl/fl mice was ∼50% of that from their control littermates (Fig. 3d). Next we analyzed use of TCR Vβ chains to investigate whether NKT cells of DN-stage-thymocyte origin were normally selected. The Vβ repertoire of NKT cells of DN-stage-thymocyte origin was grossly normal and similar to that of NKT cells of DP-stage-thymocyte origin (Supplementary Fig. 3).
To further verify the results obtained with E8III-Cre+Rag2fl/fl mice, we made another model of conditional ablation of Rag2 at the DP stage by generating Cd4-Cre+Rag2fl/fl mice. Analysis of sorted DP and DN thymic fractions showed that Trav11Traj18 transcripts were present in the DN thymocytes but not in DP thymocytes (Supplementary Fig. 4a), which suggested that the mature NKT cells detected in Cd4-Cre+Rag2fl/fl mice (Supplementary Fig. 4b,c) probably represented NKT cells of DN-stage-thymocyte origin. These data confirmed the results obtained with E8III-Cre+Rag2fl/fl mice.
To further confirm the results of Cre-recombinase-mediated approaches, such as E8III-Cre+Rosa26-YFP and E8III-Cre+Rag2fl/fl mice, we used mice with sequence encoding GFP knocked into Rorc (which encodes the transcription factor RORγt; called 'Rorc–/–' here), which is a characterized mouse model that lacks Trav11Traj18 rearrangements in DP stage thymocytes because of the shortened lifespan of DP thymocytes19,20. qPCR analysis of DN and DP thymocyte fractions sorted from Rorc–/– mice revealed that Trav11Traj18 transcripts were present only in the DN thymocyte fraction but not in the DP thymocyte fraction (Supplementary Fig. 5a) and, in agreement with the presence of Trav11Traj18 transcripts, analysis of the DN thymic population revealed the presence of mature CD1d-dimer+ TCRβ+ NKT cells in Rorc–/– mice (Supplementary Fig. 5b).
To further investigate the developmental time window for the initiation of Trav11Traj18 rearrangements in DN-stage thymocytes, we used mice deficient in the pre-TCRα-chain (Ptcra–/–), a well-known mouse model that lacks a functional β-selection process during the T cell development beyond DN stage 3 of thymic ontogeny21. We observed a complete lack of expression of Trav11Traj18 transcripts in both DN thymic fractions and DP thymic fractions in Ptcra–/– mice (Supplementary Fig. 6a), accompanied by the absence of mature NKT cells within the DN compartment (Supplementary Fig. 6b). These results suggested an absolute requirement for β-selection in the initiation of Trav11Traj18 rearrangements in the DN stage, which seemed to occur beyond DN stage 3 of development, presumably at DN stage 4, as shown previously22. Collectively, the results reported above provided evidence for the existence of an alternative developmental pathway of NKT cells from DN-stage precursor cells that was different from the DP developmental pathway.
TH1-biased function of NKT cells of DN-stage origin
To investigate the properties of the NKT cells of DN-stage-thymocyte origin, we sorted DN thymic NKT cells from E8III-Cre+Rag2fl/fl mice and stimulated the cells for 48 h with antibody to the TCR invariant chain CD3 (anti-CD3) and antibody to the co-receptor CD28 (anti-CD28), then analyzed their ex vivo cytokine-secretion profiles. For reference purposes, we used the well-characterized wild-type C57BL/6 thymic NKT cells sorted on the basis of differences the stages in their expression of the marker NK1.1, whereby the NK1.1– fraction consisted of CD44lo ('stage 1') and CD44hi ('stage 2') NKT cells, and the NK1.1+ fraction consisted of CD44hiNK1.1+ ('stage 3') NKT cells.
The results showed that NKT cells from E8III-Cre+Rag2fl/fl mice produced substantial amounts of interferon-γ (IFN-γ) and secreted a little interleukin 4 (IL-4), but no IL-17A or IL-10, a cytokine profile that mirrored that of wild-type NK1.1+ stage 3 NKT cells (Fig. 4a). As expected, the NK1.1– stage 1 and stage 2 NKT cells from wild-type mice had a robust capacity to secrete IL-4 and IL-17 after being activated (Fig. 4a). Of particular note, we observed that only wild-type NK1.1– NKT cells secreted IL-10 after stimulation with anti-CD3 and anti-CD28 (Fig. 4a). These results demonstrated that the mature NKT cells of DN-stage-thymocyte origin closely resembled wild-type NK1.1+ stage 3 NKT cells in terms of their cytokine-secretion profiles, as well as in their surface CD24loCD44hiNK1.1+ phenotype (Supplementary Fig. 7).
(a) Cytokine bead assay of IFN-γ, IL-4, IL-10 and IL-17A in supernatants of thymic NK1.1– NKT cells (stages 1 and 2 (st.1,2)) or NK1.1+ NKT cells (stage 3 (st.3)) from wild-type (WT) mice, sorted as CD1d-dimer+ cells (2 × 104), as well as DN NKT cells from E8III-Cre+Rag2fl/fl mice (key), activated for 48 h in vitro with anti-CD3 and anti-CD28. (b) Correlation of gene expression (from total transcriptome data) among thymic NK1.1–CD44– (stage 1), NK1.1–CD44+ (stage 2) and NK1.1+CD44+ (stage 3) NKT cells from wild-type mice, and DN NKT cells from E8III-Cre+Rag2fl/fl mice (above plot), evaluated by Pearson's correlation (key). (c) RNA-seq analysis of key TH1-cell-related genes (left margin) in cells as in b, presented as log10 FPKM values (fragments per kilobase per million mapped reads). Data are representative of three independent experiments (a; mean + s.e.m. of triplicate samples pooled from four to eight mice per genotype) or are from two experiments with five to six biological replicates per group (b,c).
TH1 gene-expression profile of NKT cells of DN-stage origin
To investigate the molecular basis of the TH1-cell-biased cytokine production by NKT cells of DN-stage-thymocyte origin, we performed next-generation sequencing (RNA-seq) analysis of NKT cells from E8III-Cre+Rag2fl/fl mice ('NKT cells of DN-stage origin'), with wild-type C57BL/6 thymic NKT cell subsets sorted on the basis of differences among in the stages in their expression of the markers NK1.1 and CD44. Clustering analysis of genes with differences in expression by NKT cells in the three stages showed that the NKT cells of DN-stage-thymocyte origin were closer to wild-type stage 3 NKT cells (with a correlation coefficient of 0.986 at the total transcriptome level) than to wild-type stage 1 or stage 2 NKT cells(for which the correlation coefficients were 0.856 and 0.875, respectively) (Fig. 4b).
Moreover, a correlation between the NKT cells of DN-stage origin and wild-type stage 3 NKT cells was also observed when genes encoding products related to the TH1 response were analyzed (Fig. 4c). For example, key TH1-cell-related genes, such as Ifng, Gzmb, Gzma and Prf1, the transcription-factor-encoding genes Stat4, Tbx21 and Runx3, and the surface-molecule-encoding genes Fasl, Cd40lg, Infgr1, Slamf7, Il18rap, Il12rb2, Il7r, Il2rb, Nkg7 and Klrb1c, were upregulated in both the NKT cells of DN-stage origin and wild-type stage 3 NKT cells relative to their expression in stage 1 or stage 2 NKT cells, while expression of Id3, which encodes the TH2-cell-related transcription factor Id3, was lower in the NKT cells of DN-stage origin and wild-type stage 3 NKT cells than in stage 1 or stage 2 NKT cells (Fig. 4c). Together these results demonstrated that NKT cells of DN-stage origin expressed key genes characteristic of TH1 cells and cytotoxic effector cells in a fashion very similar to that of wild-type stage 3 NKT cells.
Distinct tissue distribution of NKT cells of DN-stage origin
Next we investigated the peripheral distribution of DN NKT cells of DN-stage origin from E8III-Cre+Rag2fl/fl mice, compared with that of NKT cells of DP-stage origin (i.e., YFP+ DN NKT cells from E8III-Cre+Rosa26-YFP mice). For analysis of the DN NKT cell fraction in the spleen, we used a negative-selection approach via depletion of cells expressing the surface antigens CD4, CD8, CD19 and TER119. The results of subsequent flow cytometry revealed that while the DN NKT cells of DP-stage origin were readily detectable in the spleen, liver, lungs, mesenteric lymph nodes, gut lamina propria and visceral adipose tissue, the DN NKT cells of DN-stage origin were detected mainly in the liver, spleen and lungs but not in the mesenteric lymph nodes, lamina propria or adipose tissue (Fig. 5a,b). When we compared the absolute number of cells in these peripheral locations, we found that DN NKT cells of DP-stage origin were mainly in the spleen, while those of DN-stage origin were mainly in the liver (Fig. 5b), which suggested distinct tissue-distribution patterns for DN NKT cells of DN-stage origin and those of DP-stage origin.
(a) Flow cytometry of DN NKT cells of DN-stage origin (from E8III-Cre+Rag2fl/fl mice; DN origin (left)) and DN NKT cells of DP-stage origin (YFP+ cells from E8III-Cre+Rosa26-YFP mice; DP origin (right)), in the spleen, liver, lungs, mesenteric lymph nodes (MLN), gut lamina propria (LP) and visceral adipose tissue (Adipose) (left margin). Numbers adjacent to outlined areas indicate percent DN NKT cells among viable B220–CD4–CD8– lymphocytes gated as CD1d-dimer+TCRβ+ DN NKT cells. (b) Absolute number of NKT cells of DN-stage origin (left) or DP-stage origin (right) as in a. (c–e) RNA-seq analysis of genes encoding liver homing-related products in liver (c) or thymus (d,e) NKT cells of DN- or DP-stage origin (defined as in a; key), presented as FPKM values. Data are representative of at least three independent experiments per group, with three to eight mice per tissue (a,b; mean + s.e.m.), or one experiment (c–e; mean + s.e.m. of n = 3 biological replicates).
In an attempt to explain the enhanced liver-homing capacity of NKT cells of DN-stage origin relative to their poor homing into peripheral lymphoid locations, we performed RNA-seq analysis of genes encoding lymphocyte-homing-related factors, such as chemokine receptors and integrins expressed on thymic and liver NKT cells of DN- or DP-stage origin. Transcriptome analysis revealed that liver NKT cells of DN-stage origin had high expression of Ccr5 and Cxcr3, which encode well-known liver-homing factors23 (Fig. 5c). We also observed that liver NKT cells of DN-stage origin, in contrast to those of DP-stage origin, had high expression of the integrin-encoding genes Itga1 and Itga4 (Fig. 5c); the latter integrin is reported to have a role in the hepatic localization of lymphocytes via interaction with the adhesion molecule VCAM-1 (ref. 23). Of note, our results demonstrated that NKT cells of DN-stage origin, in contrast to those of DP-stage origin, had high expression of S1pr1 and S1pr5, which encode receptors for sphingosine 1-phosphate (Fig. 5c,d); the former receptor is involved in the egress of lymphocytes from the thymus and secondary lymphoid organs24 and might have a role in homing to the liver as a result of differences in the gradient of sphingosine 1-phosphate, while the latter is required for the trafficking of NK cells25.
In contrast to the results obtained for the expression of genes encoding liver-homing factors, NKT cells of DN-stage origin had lower expression of the chemokine-receptor-encoding genes Ccr6, Ccr7 and Ccr9 than that of their counterparts of DP-stage origin (Fig. 5e); these chemokine receptors are reported to have important roles in the trafficking of lymphocytes into lymph nodes and peripheral sites24. Together these observations could provide some explanation of not only the greater liver-homing capacity of NKT cells of DN-stage origin but also to their inferior ability to home to peripheral lymphoid organs.
Superior cytotoxicity of NKT cells of DN-stage origin
Because liver NKT cells with a CD4– phenotype are reported to elicit antitumor activity superior to that elicited by CD4+ NKT cells10, and also because of the substantial presence of NKT cells of DN-stage origin in the liver compartment, we compared the cytotoxic function of DN NKT cells of DN-stage origin with that of DN NKT cells of DP-stage origin. We first analyzed the expression of TH1-cell-related genes and genes encoding cytotoxicity-related products in steady-state ex vivo cell samples; we compared DN NKT cells of DN-stage origin (from E8III-Cre+Rag2fl/fl mice) with those of DP-stage origin (i.e., YFP+ DN NKT cells from E8III-Cre+Rosa26-YFP mice). qPCR analysis results showed higher expression of TH1-cell-related genes, such as Ifng, Tbx21, Il12rb2, Cd40lg and Runx3, and of genes encoding cytotoxicity-related factors, such as Gzmb (which encodes granzyme B), Prf1 (which encodes perforin) and Fasl (which encodes the ligand for the death receptor Fas), in NKT cells of DN-stage origin than in their counterparts of DP-stage origin (Fig. 6a).
(a) qPCR analysis of genes characteristic of the cytotoxic lineage (vertical axes) in liver DN NKT cells of DN- or DP-stage origin (defined as in Fig. 5a; key); results are presented relative to those of Traj18-Trac (internal control). (b) Flow cytometry of sorted liver DN NKT cells of DN- or DP-stage origin (as in a; effector cells) activated overnight with anti-CD3 and anti-CD28 and then cultured for 4 h together with YAC-1 target cells (labeled with the cell-division tracker dye CellTrace Violet) at an effector cell/target cell ratio of 5:1 and assessed with the apoptosis marker annexin V and the membrane-impermeable DNA-intercalating dye 7-aminoactinomycin D; results are presented as specific cytotoxicity, calculated as follows: percent cytotoxicity = (target cytotoxicity – spontaneous cytotoxicity / maximum cytotoxicity – spontaneous cytotoxicity) × 100. Data are representative of three independent experiments with six to nine mice per group (mean + s.e.m.).
Consistent with their gene-expression profiles, liver NKT cells of DN-stage origin that were activated with anti-CD3 and anti-CD28 showed greater cytotoxic activity in vitro against YAC-1 target cells (mouse lymphoma cells) than that of their counterparts of DP-stage origin (Fig. 6b). To further confirm the results obtained with E8III-Cre+Rag2fl/fl mice, we also assessed the cytotoxic activity of YFP– thymic NKT cells of DN-stage origin and YFP+ thymic NKT cells of DP-stage origin (purified from E8III-Cre+Rosa26-YFP mice). The results showed that the cytotoxic activity of YFP– NKT cells seemed to be stronger than that of YFP+ NKT cells (Supplementary Fig. 8). Together these results demonstrated that the DN pathway 'preferentially' generated NKT cells with superior cytotoxic effector function.
Discussion
Here we identified a previously unknown NKT cell developmental pathway that originates from DN thymocytes and bypasses the DP stage. We have designated this the 'DN pathway', a pathway distinct from the currently accepted DP pathway model, which states that all NKT cells are generated at the DP stage during thymic development5. The DP pathway model is based mainly on the results of two types of experiments: intrathymic transfer of Cd1d–/– DP thymocytes11 or fate-mapping approach using Cd4-Cre or Rorc-Cre12. However, despite the insights gained from such studies, there are several important points that need to be considered. First, the developmental origin of DN NKT cells cannot be formally addressed by the simple transfer of DP thymocytes alone. Second, it has been shown that Rorc expression26,27 as well as Cd4-Cre activity28,29,30 are detected as early as the thymic DN stage 3. Therefore, these fate-mapping experiments could have resulted in unanticipated labeling of the progeny of DN stage thymocytes, in addition to those of the DP stage.
To circumvent those problems, we chose to use E8III-Cre for our fate mapping and for conditional ablation of Rag2. The experiments using E8III-Cre+Rosa26-YFP mice and E8III-Cre+Rag2fl/fl mice unambiguously demonstrated that a fraction of DN NKT cells were able to develop directly from DN-stage thymocytes without entering into the DP stage. This suggested a scenario in which DN-stage precursor cells harboring rearranged Trav11 and Traj18 generated by random rearrangement events are positively selected on CD1d and develop into mature DN NKT cells.
TCR-sequencing experiments revealed the presence of diverse out-of-frame sequences encoding Vα14Jα chains, including out-of-frame Trav11Traj18 sequences, in the DN population of E8III-Cre+Rag2fl/fl mice, which provided compelling evidence of ongoing stochastic TCR α-chain rearrangements at the DN stage. Of note, we observed 'preferential' rearrangements of Trav11 in the DN thymocyte fraction with particular genes encoding Jα chains, such as Traj6, Traj18, Traj26, Traj32 and Traj57, on the basis of the frequency of use of Jα-encoding genes, whose patterns were quite similar to those observed in DP thymocytes31. Since about 10% (293 of 3,002) of Trav11-containing out-of-frame sequences were Trav11Traj18, these results indicated that the tertiary DNA structure between the Trav11 locus and loci encoding Jα during gene rearrangement events might contribute to form the Trav11Traj18 rearrangement at unexpectedly high frequency, as suggested previously32.
Despite the fact that both DN pathways and DP pathways contributed to the generation of DN NKT cells, the former pathway 'preferentially' gave rise to IFN-γ-producing TH1 NKT cells with augmented cytotoxic function, whereby NKT cells of DN-stage origin demonstrated much stronger cytotoxicity against YAC-1 target cells and had higher expression of genes encoding cytotoxicity-related products, such as granzyme B, perforin and the transcription factor Runx3, than that of their counterparts of DP-stage origin. Such 'preferential' development of TH1-type cells is probably a general attribute of unconventional T cells that develop as a result of early TCR rearrangements, as shown previously33. A potential mechanism for the 'preferential' development of TH1-biased NKT cells might be related to differences in the timing of cytokine–cytokine receptor interactions during positive selection mediated by the TCR–CD1d interaction. Consistent with that, it is known that the timing of TCR α-chain expression is critically important for the further functional fate determination of T cells16.
In addition, a published study has elegantly shown, through the use of MHC-null mice, that the developmental pathway for cytotoxic CD8+ T cells is not dependent on positive selection by cognate TCR–MHC interactions itself. Instead, signals emanating from the IL-7 receptor (IL-7R) can bypass TCR–MHC–mediated signaling and result in the differentiation of cytotoxic CD8+ T cells17. In this context, it has been reported that DN-stage thymocytes normally express IL-7R, downregulate its expression after differentiating into the DP stage and then re-express it as post-selection αβ T cells18. It has also been shown that IL-7R determines the fate of cytotoxic effector cells via induction of Runx3, which upregulates genes associated with cytotoxic cell lineages17. Although Runx3 acts synergistically with the transcription factor T-bet to enhance the transcription of TH1-cell-related genes34, it seems that T-bet, in contrast to Runx3, is induced mainly by TCR signaling. Concordantly, our gene-expression-profiling experiments revealed that the NKT cells of DN-stage origin had higher expression of genes encoding IL-7R (Il7r) and its downstream associated genes characteristic of cytotoxic cell lineages, such as Runx3, Gzmb, Prf1, Tbx21 and Il12rb2, than that of the NKT cells of DP-stage origin. Therefore, lineage commitment at the DN stage might have an important role in determining the functional characteristics of NKT cells.
In addition to their functional bias, NKT cells of DN-stage origin had a peripheral distribution pattern different from that of NKT cells of DP-stage origin. NKT cells of DN-stage origin were present mainly in the liver, a result that could be explained in part by their higher expression of genes encoding liver-homing factors than that of NKT cells of DP-stage origin. In contrast, the NKT cells of DP-stage origin were present mainly in the spleen but also in mesenteric lymph nodes, lamina propria and adipose tissues, and they had high expression of genes encoding the homing factors responsible for peripheral localization, such as CCR6, CCR7 and CCR9. These results raise questions about whether NKT cells generated by distinct pathways have specific physiological functions.
As for the biological importance of the DN pathway, we speculate that NKT cells of DN-stage origin, with their potent TH1-cell-skewed functions, might mediate a potent adjuvant activity that is essential for the clonal expansion of effector cells that mediate effective protection against pathogens35,36,37 and thus is important for survival of the species. In fact, when we investigated the rate of nonsynonymous mutations versus that of synonymous mutations in Trav11, relative to that of other genes encoding Vα regions (such as Trav1), in different species, we observed positive selection only in Trav11 (not in genes encoding other Vа regions) at a particular species divergence, such as Apodemus speciosus to Mus species or Rattus to Mesocricetus auratus. This would suggest a phylogenic importance for NKT cells in host protection38.
In conclusion, we have identified an alternative DN pathway of NKT cell development that bypasses the DP pathway. We propose that the acquisition of diverse functional characteristics by NKT cells might be dependent on the timing of TCR expression as well as on the cytokine receptor signaling in precursor cells undergoing positive selection. We also propose that the developmental environments or specific niches provide an appropriate cytokine milieu required for the differentiation of these cells.
Methods
Mice.
C57BL/6J mice were purchased from Charles River Laboratories or CLEA Japan. Rag2fl/fl mice39 were from The Jackson Laboratory. Ptcra–/–, Rorc–/–, Rosa26-YFP, Cd4-Cre and E8III-Cre mice (all of C57BL/6 background) have been described previously12,17,21,40. All mice were kept under specific-pathogen-free conditions and were used at 8–16 weeks of age unless indicated otherwise. All experiments were in accordance with protocols approved by the RIKEN Animal Care and Use Committee.
Cell preparation.
Single cells from designated organs were prepared as reported41,42. Anti–mouse CD4 and CD8 microbeads and AutoMACS were used according to the manufacturer's instructions (Miltenyi Biotec) to prepare DN thymocytes and splenocytes. Where indicated, anti–mouse CD19 and TER119 microbeads (Miltenyi Biotec) were also used in addition to anti–mouse CD4 and CD8 microbeads for negative selection by magnetic-activated cell separation.
Flow cytometry and cell sorting.
for flow cytometry, monoclonal antibodies (mAbs) to the following were conjugated to FITC, PE, PerCP-Cy5.5, PE-Cy7, APC, APC-eFluor780 and BV421 (all mAbs here used at a concentration of 1 μl per 1 million cells): CD24 (BioLegend, clone M1/69), CD25 (BD Biosciences, clone PC61), CD44 (eBioscience, clone IM7), CD117 (eBioscience, clone 2B8), CD45R/B220 (BioLegend, clone RA3-6B2), NK1.1 (BD Biosciences, clone PK136) and TCRβ (BD Biosciences, clone H57-597), as well as α-galactosylceramide-loaded CD1d dimer (BD Biosciences). The panel of FITC-labeled mAbs to mouse TCR Vβ was purchased from BD Biosciences (catalog number 557004; 20 μl used per test, as recommended by the manufacturer). Cell staining was performed after blockade with anti-FcR (BD Bioscience, clone 2.4G2). Forward light-scatter gating and staining with the membrane-impermeable DNA-intercalating dye 7-AAD were used to gate out doublets and dead cells. Samples were analyzed on a FACSCanto II or FACSAria III (BD Biosciences), and data were analyzed with FlowJo (Tree Star). Cells were sorted using a FACSAria III with post-sort purities above 99%.
Cytokine measurement.
NKT cells (2 × 104) sorted by flow cytometry were stimulated with plate-bound mAb to CD3 (BD Biosciences, clone 145-2C11) and soluble mAb to CD28 (BD Biosciences, clone 37.51) at 10 μg/ml. Supernatants were collected after 48 h of incubation. IFN-γ, IL-4 and IL-17A were measured by a LEGENDPlex-bead-based immunoassay (BioLegend) according to the manufacturer's instructions. Data were acquired using a FACSCanto II (BD Biosciences) and were analyzed with LEGENDPlex software (BioLegend).
Quantitative real-time PCR.
RNA was prepared from sorted cells using RNeasy Plus Micro or RNeasy Plus Mini Kit (Qiagen) with a gDNA eliminator column treatment step. cDNA was synthesized using Superscript VILO cDNA Synthesis Kit (Life Technologies). Quantitative real-time PCR (qPCR) was performed using the ABI PRISM 7900HT system (Applied Biosystems) with a FastStart Universal Probe Master mix (Roche), or with the LightCycler 480 (Roche Applied Science) with a LightCycler 480 Probes Master mix (Roche). Relative gene expression was calculated with the 2–ΔΔCt method. The detection of Trav11Traj18 transcripts was performed as described previously11. The following primer pairs and probes were used for Universal Probe Library (UPL)- or TaqMan-probe-based quantitative PCR assays: Rag2 (forward) 5′-TGCCAAAATAAGAAAGAGTATTTCAC-3′, (reverse) 5′-GGGACATTTTTGATTGTGAATAGG-3′, UPL probe 4; Traj18Trac (forward) 5′-GGACTCAGCTGATTGTCATACCT-3′, (reverse) 5′-AGACCGAGGATCTTTTAACTGGT-3′, UPL probe 62; Gzmb (forward) 5′-GATGAAGATCCTCCTGCTACTGC-3′, (reverse) 5′-GTAAGGCCATGTAGGGTCGAGA-3′, probe (FAM-TAMRA) 5′-CCCGATGATCTCCCCTGCCTTTGTCCT-3′; Prf1 (forward) 5′-TGAGAAGACCTATCAGGACCAGTA-3′, (reverse) 5′-AGTCAAGGTGGAGTGGAGGTT-3′, probe (FAM-TAMRA) 5′-ACCAGGCGAAAACTGTACATGCGACACTC-3′; Fasl (forward) 5′-TCACCAACCAAAGCCTTAAAGTATC-3′, (reverse) 5′-AGAGGGATGGACCTTGAGTGG-3′, probe (FAM-TAMRA) 5′-AAATGGGCCACACTCCTCGGCTCTTT-3′; Cd40lg (forward) 5′-GCACACGTTGTAAGCGAAGC-3′, (reverse) 5′-CCCATTTTCAAGCATTACCAAGTTG-3′, probe (FAM-TAMRA) 5′-TGCAGCATCCGTTCTACAGTGGGCCA-3′. The following gene-expression assays (all from Applied Biosystems) were used: Ifng (Mm01168134_m1), Tbx21 (Mm00450960_m1), Runx3 (Mm00490666_m1), Il12rb2 (Mm00434200_m1) and Gapdh (Mm99999915_g1).
Next-generation sequencing (RNA-seq).
RNA was prepared with the RNeasy Plus Micro Kit (Qiagen). RNA-seq library samples were prepared using a TruSeq DNA sample Prep Kit v2 (Illumina) or ThruPLEX DNA-seq Kit (Rubicon Genomics) and a SMARTer Ultra Low RNA Kit for Illumina (Clontech). The sequencing was carried out using HiSeq1500 equipment (Illumina). The sequence reads were mapped to mouse genome (NCBI version 37) using TopHat2 version 2.0.8 and botwie2 version 2.1.0 with default parameters, and gene annotation was provided by NCBI. The transcript abundances were estimated using Cufflinks (version 2.1.1). Cufflinks was run with the same reference annotation with TopHat2 to generate FPKM (fragments per kilobase per million mapped reads) values for known gene models.
TCR sequencing.
DN thymocytes obtained from E8III-Cre+Rag2fl/fl mice and sorted by flow cytometry were lysed and RNA was extracted using an RNeasy kit (Qiagen). cDNA was prepared with Superscript First-Strand SuperMix (Life Technologies). PCR to amplify the Trav11Trac transcripts was done as described previously43. PCR products were purified and sequenced on a MiSeq system with a MiSeq Reagent Kit v3-600 (Illumina). Mouse genes encoding Jα regions were analyzed using IMGT/HighV-QUEST from the IMGT (international ImMunoGeneTics information system) database44.
Cytotoxicity assay.
Cytotoxic killing of YAC-1 target cells was performed with purified NKT cells (2 × 104), which were activated overnight with anti-CD3 (BD Biosciences, clone 145-2C11) and anti-28 (BD Biosciences, clone 37.51) (10 μg/ml each) in complete RPMI-1640 medium supplemented with mouse IL-2 (10 ng/ml, R&D). Target cells before incubation with effector cells were labeled with CellTrace Violet (Thermo Fisher Scientific) and, after 4 h of co-culture, were analyzed using the Annexin V staining kit according to the manufacturer′s instructions (BioLegend). The percentage of specific cytotoxicity was calculated by the following formula: % cytotoxicity = (target-spontaneous / maximum-spontaneous) × 100.
Statistical analysis.
Two-tailed, unpaired t-tests were done using PRISM 6 software (GraphPad). P < 0.05 was considered statistically significant.
Data availability.
Raw and processed RNA-seq data that support the findings of this study have been deposited in the NCBI Gene Expression Omnibus (GEO) database with the accession codes GSE53150 and GSE89532.
References
Taniguchi, M., Harada, M., Kojo, S., Nakayama, T. & Wakao, H. The regulatory role of Vα14 NKT cells in innate and acquired immune response. Annu. Rev. Immunol. 21, 483–513 (2003).
Lantz, O. & Bendelac, A. An invariant T cell receptor α chain is used by a unique subset of major histocompatibility complex class I-specific CD4+ and CD4–8– T cells in mice and humans. J. Exp. Med. 180, 1097–1106 (1994).
Dellabona, P., Padovan, E., Casorati, G., Brockhaus, M. & Lanzavecchia, A. An invariant Vα24-αQ/Vβ11 T cell receptor is expressed in all individuals by clonally expanded CD4-8- T cells. J. Exp. Med. 180, 1171–1176 (1994).
Borg, N.A. et al. CD1d-lipid-antigen recognition by the semi-invariant NKT T-cell receptor. Nature 448, 44–49 (2007).
Bendelac, A., Savage, P.B. & Teyton, L. The biology of NKT cells. Annu. Rev. Immunol. 25, 297–336 (2007).
Salio, M., Silk, J.D., Jones, E.Y. & Cerundolo, V. Biology of CD1- and MR1-restricted T cells. Annu. Rev. Immunol. 32, 323–366 (2014).
Brossay, L. et al. CD1d-mediated recognition of an α-galactosylceramide by natural killer T cells is highly conserved through mammalian evolution. J. Exp. Med. 188, 1521–1528 (1998).
Lee, P.T., Benlagha, K., Teyton, L. & Bendelac, A. Distinct functional lineages of human Vα24 natural killer T cells. J. Exp. Med. 195, 637–641 (2002).
Gumperz, J.E., Miyake, S., Yamamura, T. & Brenner, M.B. Functionally distinct subsets of CD1d-restricted natural killer T cells revealed by CD1d tetramer staining. J. Exp. Med. 195, 625–636 (2002).
Crowe, N.Y. et al. Differential antitumor immunity mediated by NKT cell subsets in vivo. J. Exp. Med. 202, 1279–1288 (2005).
Gapin, L., Matsuda, J.L., Surh, C.D. & Kronenberg, M. NKT cells derive from double-positive thymocytes that are positively selected by CD1d. Nat. Immunol. 2, 971–978 (2001).
Egawa, T. et al. Genetic evidence supporting selection of the Vα14i NKT cell lineage from double-positive thymocyte precursors. Immunity 22, 705–716 (2005).
Pellicci, D.G. et al. A natural killer T (NKT) cell developmental pathway involving a thymus-dependent NK1.1–CD4+ CD1d-dependent precursor stage. J. Exp. Med. 195, 835–844 (2002).
Benlagha, K., Wei, D.G., Veiga, J., Teyton, L. & Bendelac, A. Characterization of the early stages of thymic NKT cell development. J. Exp. Med. 202, 485–492 (2005).
Aifantis, I. et al. The E delta enhancer controls the generation of CD4–CD8– αβTCR-expressing T cells that can give rise to different lineages of αβ T cells. J. Exp. Med. 203, 1543–1550 (2006).
Baldwin, T.A., Sandau, M.M., Jameson, S.C. & Hogquist, K.A. The timing of TCR α expression critically influences T cell development and selection. J. Exp. Med. 202, 111–121 (2005).
Park, J.H. et al. Signaling by intrathymic cytokines, not T cell antigen receptors, specifies CD8 lineage choice and promotes the differentiation of cytotoxic-lineage T cells. Nat. Immunol. 11, 257–264 (2010).
McCaughtry, T.M. et al. Conditional deletion of cytokine receptor chains reveals that IL-7 and IL-15 specify CD8 cytotoxic lineage fate in the thymus. J. Exp. Med. 209, 2263–2276 (2012).
Sun, Z. et al. Requirement for RORγ in thymocyte survival and lymphoid organ development. Science 288, 2369–2373 (2000).
Guo, J. et al. Regulation of the TCRα repertoire by the survival window of CD4+CD8+ thymocytes. Nat. Immunol. 3, 469–476 (2002).
Fehling, H.J., Krotkova, A., Saint-Ruf, C. & von Boehmer, H. Crucial role of the pre-T-cell receptor α gene in development of αβ but not γδ T cells. Nature 375, 795–798 (1995).
Dashtsoodol, N., Watarai, H., Sakata, S. & Taniguchi, M. Identification of CD4–CD8– double-negative natural killer T cell precursors in the thymus. PLoS One 3, e3688 (2008).
Crispe, I.N. Migration of lymphocytes into hepatic sinusoids. J. Hepatol. 57, 218–220 (2012).
Cyster, J.G. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu. Rev. Immunol. 23, 127–159 (2005).
Jenne, C.N. et al. T-bet-dependent S1P5 expression in NK cells promotes egress from lymph nodes and bone marrow. J. Exp. Med. 206, 2469–2481 (2009).
Villey, I., de Chasseval, R. & de Villartay, J.P. RORγT, a thymus-specific isoform of the orphan nuclear receptor RORγ / TOR, is up-regulated by signaling through the pre-T cell receptor and binds to the TEA promoter. Eur. J. Immunol. 29, 4072–4080 (1999).
He, Y.W., Deftos, M.L., Ojala, E.W. & Bevan, M.J. RORγt, a novel isoform of an orphan receptor, negatively regulates Fas ligand expression and IL-2 production in T cells. Immunity 9, 797–806 (1998).
Wolfer, A. et al. Inactivation of Notch 1 in immature thymocytes does not perturb CD4 or CD8 T cell development. Nat. Immunol. 2, 235–241 (2001).
Laky, K. & Fowlkes, B.J. Presenilins regulate αβ T cell development by modulating TCR signaling. J. Exp. Med. 204, 2115–2129 (2007).
Kastner, P. et al. Bcl11b represses a mature T-cell gene expression program in immature CD4+CD8+ thymocytes. Eur. J. Immunol. 40, 2143–2154 (2010).
Bedel, R. et al. Lower TCR repertoire diversity in Traj18-deficient mice. Nat. Immunol. 13, 705–706 (2012).
Genolet, R., Stevenson, B.J., Farinelli, L., Osterås, M. & Luescher, I.F. Highly diverse TCRα chain repertoire of pre-immune CD8+ T cells reveals new insights in gene recombination. EMBO J. 31, 1666–1678 (2012).
Hendricks, D.W. & Fink, P.J. Uneven colonization of the lymphoid periphery by T cells that undergo early TCRα rearrangements. J. Immunol. 182, 4267–4274 (2009).
Djuretic, I.M. et al. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat. Immunol. 8, 145–153 (2007).
Brigl, M., Bry, L., Kent, S.C., Gumperz, J.E. & Brenner, M.B. Mechanism of CD1d-restricted natural killer T cell activation during microbial infection. Nat. Immunol. 4, 1230–1237 (2003).
Kinjo, Y. et al. Recognition of bacterial glycosphingolipids by natural killer T cells. Nature 434, 520–525 (2005).
Shimizu, K. et al. Invariant NKT cells induce plasmacytoid dendritic cell (DC) cross-talk with conventional DCs for efficient memory CD8+ T cell induction. J. Immunol. 190, 5609–5619 (2013).
Kanno, R. et al. Evolution of Vα14 TCR gene family in mice. in Genetics in Wild Mice (eds. Moriwaki, K., Shiroishi, T. & Yonekawa, H.), 193–202 (Japan Scientific Societies Press, Karger, Tokyo, Basel, 1994).
Hao, Z. & Rajewsky, K. Homeostasis of peripheral B cells in the absence of B cell influx from the bone marrow. J. Exp. Med. 194, 1151–1164 (2001).
Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).
Watarai, H., Nakagawa, R., Omori-Miyake, M., Dashtsoodol, N. & Taniguchi, M. Methods for detection, isolation and culture of mouse and human invariant NKT cells. Nat. Protoc. 3, 70–78 (2008).
Moro, K., Ealey, K.N., Kabata, H. & Koyasu, S. Isolation and analysis of group 2 innate lymphoid cells in mice. Nat. Protoc. 10, 792–806 (2015).
Dashtsoodol, N. et al. Generation of novel Traj18-deficient mice lacking Vα14 natural killer T cells with an undisturbed T cell receptor α-chain repertoire. PLoS One 11, e0153347 (2016).
Lefranc, M.P. IMGT, the International ImMunoGeneTics Information System for Immunoinformatics: methods for querying IMGT databases, tools, and web resources in the context of immunoinformatics. Mol. Biotechnol. 40, 101–111 (2008).
Acknowledgements
We thank A. Singer (US National Institutes of Health) for E8III-Cre mice; F. Constantini (Columbia University) for Rosa26-YFP mice; D. Littman (New York University) for Rorc–/– mice; P. Burrows for critical reading of this manuscript; T. Hasegawa for support with animal facility services; and N. Takeuchi for secretarial assistance. Supported by the Japan Society for the Promotion of Science (KAKENHI grants 24790490 to N.D. and 23229005 to M.T.).
Author information
Authors and Affiliations
Contributions
N.D. conceived of the study, performed experiments, analyzed data and wrote the paper; T.S., M.A. and R.O. assisted with experiments; S.K. and M.H. discussed experiments; T.A.E. did bioinformatics analyses; T.W. and O.O. performed next-generation sequencing experiments; M.T. directed the study and wrote the paper.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing financial interests.
Integrated supplementary information
Supplementary Figure 1 Confirmation of the DN-stage origin of NKT cells with the Cd4-Cre+Rosa26-YFP mouse.
(a) YFP reporter expression within gated viable B220-TCRβ+ splenic CD4+, CD8+, and DN fractions obtained from Cd4-Cre+Rosa26-YFP mice. Numbers on flow cytometry plots show the frequency of cells within indicated gates. (b) Detection of CD1d dimer+ NKT cells within gated YFP-negative and YFP-positive TCRβ+ DN splenocytes from Cd4-Cre+Rosa26-YFP mice. Prior to the cell staining, DN splenocytes were purified by AutoMACS using anti-CD4 and anti-CD8 microbeads. Numbers on flow cytometry plots show the percentage and absolute number of CD1d dimer+ NKT cells within gated YFP-negative and YFP-positive DN TCRβ+ cells. Results shown are from two mice done in one experiment.
Supplementary Figure 2 TCR α-chain joining-region gene-segment usage among out-of-frame Trav11-containing sequences detected in DN thymocytes from E8III-Cre+Rag2fl/fl mice.
High-throughput sequencing of Trav11Trac transcripts was carried out using the MiSeq system, where cDNA was prepared from sorted CD4-CD8- DN thymocytes from E8III-Cre+Rag2fl/fl mice. Bars depict the frequency (mean ± SEM, n = 3 biological replicates) of Traj gene segment usage among 3,002 out-of-frame Trav11 (CDR3) sequences detected when a total of 55,056 Trav11+ sequences were analyzed as shown in Table 1. The data are from one sequencing experiment.
Supplementary Figure 3 The TCR Vβ repertoire of NKT cells of DN-stage-thymocyte origin.
Flow cytometry analysis of TCR Vβ chain usage analysis of thymic CD1d dimer+CD3ɛ+ DN NKT cells from E8III-Cre+Rag2fl/fl (black bars) and littermate control mice (gray bars). Bars represent mean percentages ± SEM of three independent experiments with at least two mice per group.
Supplementary Figure 4 Detection of Trav11Traj18 transcripts and DN NKT cells in the thymus of Cd4-Cre+Rag2fl/fl mice.
(a) Viable 7-AAD-B220-CD1d dimer-TCRβ-/lo DP and DN thymocyte fractions that were electronically gated as CD5loCD69- cells were sorted from Cd4-Cre+Rag2fl/fl mice, and Trav11Traj18 mRNA expression was determined by qPCR analysis. Relative gene expression (mean ± SEM, n = 3) with Gapdh used as internal control. Results are representative of two experiments, (n.d.) not detected. (b) Detection of CD1d dimer+ DN NKT cells in the thymus, and (c) in the liver from Cd4-Cre+Rag2fl/fl mice, where littermate mice were used as controls. MACS depletion of CD4+, CD8+ and DP cells with magnetic microbeads was done prior to the cell staining. Numbers on flow cytometry plots show the percentage and the absolute number of CD1d dimer+TCRβ+ NKT cells among electronically gated DN cells. Results shown are from one experiment with two mice per genotype analyzed.
Supplementary Figure 5 Detection of Trav11Traj18 transcripts and mature DN NKT cells in the DN thymocyte fraction of Rorc–/– mice.
(a) Trav11Traj18 mRNA expression in sorted CD4+CD8+ DP and CD4-CD8- DN thymic fractions from Rorc-/- mice, as determined by qPCR. Bars show relative expression (mean ± SEM), where Gapdh was used as an internal control. Results are representative from three independent experiments with combined total of three to six mice per group, (n.d.) not detected. (b) Detection of CD1d dimer+ mature NKT cells within DN thymocytes of Rorc-/- mice. Prior to the cell staining, DN thymocytes were purified with AutoMACS by using anti-CD4 and anti-CD8 microbeads. Numbers on plots indicate the percentage and absolute number (mean ± SEM, n=6 mice) of CD1d dimer+ NKT cells within gated DN TCRβ+ cells. Unloaded CD1d staining of DN thymocytes from B6 mice was used as a staining control. Results shown are representative from three independent experiments with two mice per group.
Supplementary Figure 6 Lack of Trav11Traj 18 transcripts as well as mature DN NKT cells in the thymus of Ptcra–/– mice.
(a) The qPCR analysis of Trav11Traj18 mRNA expression in sorted thymic CD4+CD8+ DP, and CD44+CD25- DN1, CD44+CD25+ DN2, CD44-CD25+ DN3, CD44-CD25- DN4 fractions from Ptcra-/- mice. Bars show relative expression (mean ± SEM, n = 3 biological replicates), where Gapdh was used as an internal control. Results are representative from three independent experiments, (n.d.) not detected. (b) Detection of CD1d dimer+ NKT cells within DN thymocytes from B6 and Ptcra-/- mice. Numbers on flow cytometry plots indicate the percentage of CD1d dimer+TCRβ+ NKT cells within electronically gated DN cells. Prior to the cell staining, DN thymocytes were purified with AutoMACS by using anti-CD4 and anti-CD8 microbeads. Results shown are representative from three independent experiments with two mice per group.
Supplementary Figure 7 Surface phenotype of NKT cells of DN-stage-thymus origin is similar to that of B6 wild-type stage 3 NKT cells.
Viable 7-AAD-B220-CD1d dimer+TCRβ+ DN thymic NKT cells from B6 and E8III-Cre+Rag2fl/fl mice were analyzed for the expression of CD24, CD44, and NK1.1 surface antigens by flow cytometry. Numbers on the histograms show the percentage of CD24lo DN NKT cells, and numbers on flow cytometry plots represent the percentage of DN NKT cells within the indicated quadrant gates. Results shown are representative from three independent experiments, n = 3 mice per genotype.
Supplementary Figure 8 Cytotoxic killing assay with YFP– and YFP+ NKT cells from E8III-Cre+Rosa26-YFP mice.
FACS-sorted YFP-negative and YFP-positive thymic DN NKT cells from E8III-Cre+Rosa26-YFP mice were activated with anti-CD3 and anti-CD28 overnight, and then were co-cultured with CellTraceTM Violet labeled YAC-1 target cells at a E/T ratio of 5:1 for 4 hrs. Samples then were FACS analyzed using annexin V and 7-aminoactinomycin D. Bars depict the mean specific cell lysis percentage ± SEM of triplicate samples done in one experiment, and were calculated by the following formula: % cytotoxicity = (target-spontaneous/maximum-spontaneous) ×100.
Supplementary information
Supplementary Text and Figures
Supplementary Figures 1–8 (PDF 622 kb)
Rights and permissions
About this article
Cite this article
Dashtsoodol, N., Shigeura, T., Aihara, M. et al. Alternative pathway for the development of Vα14+ NKT cells directly from CD4–CD8– thymocytes that bypasses the CD4+CD8+ stage. Nat Immunol 18, 274–282 (2017). https://doi.org/10.1038/ni.3668
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/ni.3668
This article is cited by
-
Loss of Zfp335 triggers cGAS/STING-dependent apoptosis of post-β selection thymocytes
Nature Communications (2022)
-
Modulation of TCR signalling components occurs prior to positive selection and lineage commitment in iNKT cells
Scientific Reports (2021)
-
Thymic iNKT cell differentiation at single-cell resolution
Cellular & Molecular Immunology (2021)
-
Liver X receptors regulate natural killer T cell population and antitumor activity in the liver of mice
Scientific Reports (2021)
-
Liver metastases
Nature Reviews Disease Primers (2021)