Skip to main content
Immunology logoLink to Immunology
. 2011 Sep;134(1):1–7. doi: 10.1111/j.1365-2567.2011.03463.x

Control of early stages in invariant natural killer T-cell development

Taishan Hu 1, Idoia Gimferrer 1, José Alberola-Ila 1,2
PMCID: PMC3173689  PMID: 21718314

Abstract

Natural killer T (NKT) cells develop in the thymus from the same precursors as conventional CD4+ and CD8+αβ T cells, CD4+ CD8+ double-positive cells. In contrast to conventional αβT cells, which are selected by MHC–peptide complexes presented by thymic epithelial cells, invariant NKT cells are selected by lipid antigens presented by the non-polymorphic, MHC I-like molecule CD1d, present on the surface of other double-positive thymocytes, and require additional signals from the signalling lymphocytic–activation molecule (SLAM) family of receptors. In this review, we provide a discussion of recent findings that have modified our understanding of the NKT cell developmental programme, with an emphasis on events that affect the early stages of this process. This includes factors that control double-positive thymocyte lifespan, and therefore the ability to generate the canonical Vα rearrangements that characterize this lineage, as well as the signal transduction pathways engaged downstream of the T-cell receptor and SLAM molecules.

Keywords: cell fate, development, natural killer T cells, signal transduction, survival

Introduction

Natural Killer T (NKT) cells are a subset of αβ T cells that are not restricted by classical MHC molecules, and that respond to antigen recognition with a ‘cytokine storm’; the secretion, within hours, of large quantities of T helper type 1 and type 2 cytokines and chemokines, reminiscent of innate rather than adaptive functions. In fact, the rapid and dual production, at a single cell level, of interferon-γ and interleukin-4 (IL-4) in response to stimulation is a characteristic of many of these cells.1 Through this cytokine and chemokine production, NKT cells influence the behaviour of many other cells in the immune system, including NK cells, macrophages, other αβ T cells, dendritic cells and neutrophils (reviewed in ref. 2), and have been implicated in multiple processes, including microbial immunity and tumour rejection. Similarly, they play a role in the pathogenesis of autoimmune processes, atherosclerosis and allergy (reviewed in ref. 3).

Although there are different types of NKT cells (see ref. 4 for a review), the most common, and better studied are type I NKT cells, also called invariant NKT cells (iNKT). These cells are characterized by the expression of an invariant T-cell receptor (TCR) α chain (Vα14-Jα18 in mice or Vα24-Jα18 in humans) in combination with certain TCR-β chains (using Vβ8.2, Vβ7 or Vβ2 in mice, or Vβ11 in humans). The iNKT cells in mice can be CD4+ or double-negative (DN; CD4 CD8), generally have a ‘memory’ or ‘activated’ phenotype (CD69+ CD62L CD44hi IL-2Rβhi) and express markers characteristic of NK cells, including NK1.1, NKG2D and Ly49. They are found mainly in the liver and bone marrow, but also in the thymus, spleen and blood. The numbers of NKT cells are highly variable between individuals and mouse strains.59 Given their role in immunoregulation this may be relevant for the pathogenesis of autoimmune diseases, and deficiencies in NKT cell numbers have been identified in pathologies such as systemic lupus erythematosus.10

Invariant NKT cells recognize the glycosphingolipid α-galactosylceramide presented by CD1d, and are best identified using a CD1d tetramer bound to this antigen.11 Other ligands for NKT cells have been identified, both endogenous and derived from pathogens, but the nature of the ligand(s) that mediate positive selection of this lineage in the thymus remains elusive.12,13

The iNKT cells develop in the thymus from the same precursors as conventional CD4+ and CD8+αβ T cells, CD4+ CD8+ double-positive (DP) cells.14 In contrast to conventional αβ T cells, which are selected by MHC–peptide complexes presented by thymic epithelial cells, iNKT are selected by lipid antigens presented by the non-polymorphic, MHC I-like molecule CD1d, present on the surface of other DP thymocytes (reviewed in refs 3,15,16).

The differentiation of iNKT cells in the thymus has been divided into a series of stages, based on phenotypic markers, and sensitivity to different mutations (reviewed in refs 11,1618) (Fig. 1). Once a DP thymocyte expresses the invariant TCR and receives signals through the signalling lymphocytic–activation molecule (SLAM)/SLAM-associated protein (SAP) and TCR (Control Point 1) it starts down-regulating Heat Stable Antigen (HSA) (CD24) and up-regulating first CD44, and later DX5. These cells are small, NK1.1 and not cycling. Some of these cells can exit the thymus and mature to NK1.1+ iNKT cells in the periphery, while others mature in the thymus. The transition from NK1.1 immature iNKT cells to NK1.1+ iNKT is accompanied by a proliferative burst (Control Point 2). Mature iNKT cells can be CD4+ or CD4 (i.e. DN), and these subsets are functionally different, at least in humans. CD4+ iNKT cells have a T helper type 0 cytokine profile while DN iNKT cells are more T helper type 1 biased. The relationship between these two subsets is unclear, although some genetic manipulations, like the deletion of GATA-3 at the DP stage, affect them differentially.19 Recent data suggest that some of the NK1.1 iNKT cells found in the periphery are a distinct mature population.20 There is also some evidence21,22 for a process of negative selection, induced by high-affinity ligands, although it has not been well characterized. In the periphery, iNKT cells are long-lived, have a slow turnover, are dependent on IL-15, and do not seem to require low-affinity interactions of their TCR with CD1d for survival.23

Figure 1.

Figure 1

Stages of invariant natural killer T (iNKT) cell development. The NKT cells develop in the thymus from CD4+ CD8+ double-positive (DP) thymocytes. Thymocytes that express a T-cell receptor (TCR) that interacts with CD1d bound to self glycolipid, expressed by other DP thymocytes, enter the NKT-cell lineage. Signals from the signalling lymphocytic–activation molecule (SLAM) co-receptors are also required. Once selected, NKT-cell precursors undergo a series of differentiation steps that ultimately result in the NKT-cell pool. The initial selection event is called Control Point 1. Four distinct stages of differentiation have been defined through differential expression of CD24, CD44 and DX5. The NKT cells then undergo an actively regulated maturation step, which ultimately results in a range of functional and phenotypic changes, including expression of NK1.1. This maturation step is called Control Point 2. Many NKT emigrate from the thymus as immature cells and undergo final differentiation in the periphery. Some mature thymic NKT cells also migrate to the periphery, but many remain as long-term thymus-resident cells. Both immature and mature NKT cells include CD4+ and CD4 (DN) subsets.

Numerous recent reviews have addressed the mechanisms that control NKT cell development and function.4,11,16,18,2426 Here, we will focus on recent findings that impact our understanding of the early stages of iNKT development (Control Point 1).

Control of DP thymocyte lifespan

The first prerequisite for the generation of iNKT cells is the stochastic rearrangement of the invariant Vα14-Jα18 TCR-α chain. Given the orderly sequence of rearrangements in the TCR-α locus (proximal to distal), and the distal position of Jα18 in the Jα region, Vα14-Jα18 rearrangements are always secondary.27 Therefore an extended DP lifespan is necessary for the development of NKT cells, as first evidenced by the defects in NKT development observed in RAR-related orphan receptor γT (RORγT) -deficient and Bcl-xL-deficient mice.2830 In contrast with this model, it has also been proposed that some early αβ T-cell precursors are pre-committed to the iNKT lineage at the DN stage.31 It is unclear how these results can be reconciled with multiple lines of evidence that suggest that a long DP lifespan is a requirement for iNKT cell development.

Double-positive thymocytes have a short average lifespan (3–4 days).3234 The mechanisms that regulate this defined lifespan are not completely understood. Although DP thymocytes are exquisitely sensitive to elevations in glucocorticoid levels, as those induced by stress,35,36 the role of normal levels of glucocorticoids in determining DP thymocyte lifespan is controversial.3739 RORγT, a retinoid-related orphan nuclear receptor, was shown to control DP lifespan,40 and analysis of RORγT-deficient knockouts provided the first evidence for the requirement of long DP lifespans for iNKT generation. The RORγT affects DP survival through the regulation of Bcl-xL, an anti-apoptotic member of the Bcl-2 family. Although this basic relationship has been known for a number of years, a number of other factors that also contribute to DP survival have been recently identified.

HEB, a member of the E-box family of transcription factors, is required for iNKT cell development because of its effect on controlling expression of RORγT and, therefore, Bcl-xL.41 Similarly, DP-specific deletion of the transcription factor c-Myb results in a decrease in DP thymocyte lifespan,42 and a blockade of iNKT cell development.43 The alteration in DP lifespan is the result of its effect on Bcl-xL expression, but, in contrast with HEB, this effect is independent of RORγT expression. The effect on Bcl-xL does not seem to be direct, or, at least, c-Myb cannot be detected by ChiP on three predicted c-Myb binding sites present in the Bcl-xL promoter.42 Furthermore, although expression of Bcl-xL rescues DP lifespan, and distal Vα-Jα rearrangements, this is not sufficient to rescue NKT cell development in c-Myb-deficient thymocytes43 (see below). Other pathways that regulate of DP lifespan through control of Bcl-xL levels include the Wnt-β-catenin-TCF-1 pathway,4446 and the energy sensor LKB1, through activation of AMP-activated protein kinase.47

Another genetic alteration that results in decreased DP thymocyte survival, and biased TCR-α rearrangements, is the deletion of the transcriptional repressor histone deacetylase 7 (HDAC-7).48 Presumably this mutation should also block NKT development, although this was not tested. In this case the effect is apparently independent of alterations in RORγT or Bcl-xL, because their expression is not altered in HDAC-7-deficient thymocytes. The relationships between all these different pathways, and how they cooperate to maintain homeostasis of the DP population, are subjects of interest that will require additional experiments in the future (see Fig. 2).

Figure 2.

Figure 2

Control of double-positive (DP) thymocyte lifespan. CD4+ CD8+ DP thymocytes have a short lifespan, and in the absence of signals from the T-cell receptor (TCR) die in 3–4 days. Factors that shorten this lifespan dramatically affect development of invariant natural killer T cells, because the Vα14-Jα18 TCR-α rearrangement that characterizes them is always secondary. In this diagram we illustrate some of the known factors that influence DP lifespan, and their interactions.

Signals that regulate Control Point 1

It is thought that selection by DP cells imparts the unique developmental programme of iNKT cells, and other ‘innate-like’αβ T cells such as T-CD4 T cells,49,50 by the cooperative engagement of the TCR and at least two members of the SLAM family (SLAMF1 and SLAMF6),51 although it is unclear if this cooperative engagement alters the signal that a positively selected DP thymocyte receives from its TCR, or whether the SLAM/SAP-derived signals are required to block negative selection induced by strong TCR-derived signals.52 Below we discuss the known contributions of different signal transduction pathways downstream of the TCR and the SLAM axis. In some cases, most notably the nuclear factor-κB (NF-κB) pathway, both inputs are able to activate the same pathway (see Fig.3).

Figure 3.

Figure 3

Signals that drive Control Point 1. Positive selection of invariant natural killer T (iNKT) cells from double-positive (DP) thymocytes requires signals from the T-cell receptor (TCR) and the signalling lymphocytic–activation molecule (SLAM) co-receptors. This diagram shows some of the signal transduction pathways involved in this early stage of iNKT cell development, as well as some transcription factors that are required for further development.

Signals from the TCR

Selection of conventional αβ T cells from DP thymocytes requires TCR-mediated signals, which induce numerous intracellular events including activation of the Ras/mitogen-activated protein kinase (MAPK) cascade,53 increased intracellular calcium levels and subsequent activation of calcineurin54 and activation of itk family kinases.55,56 A number of recent papers has explored the contribution of these pathways to the selection of iNKT cells.

Activation of the Ca2+/calcineurin pathway is required for iNKT cell development. Deletion of calcineurin B1 in DP thymocytes produced a significant loss in both thymic and peripheral iNKT cell numbers.57 In the same work, the authors identified Egr2 as an important mediator of this pathway. Egr2-deficient fetal liver chimeras showed impaired development of iNKT cells, starting at the earliest stages of positive selection (Control Point 1). Although the evidence presented in this work suggested that neither Egr1, nor Egr3, played a role in iNKT cell development, more recent experiments, using competitive mixed bone marrow chimeras, and double Egr1-Egr2 knockouts, have shown that both Egr1 and Egr2 play a role in NKT cell development.58 Contrary to the situation in conventional αβ T-cell development, where Egr1 and Egr2 play a quantitatively similar role,59,60 during iNKT cell development Egr2 seems to play a much more central role. The mechanism that underlies these functional differences is not yet characterized. Another molecule induced by calcium signals, the high-mobility group box transcription factor TOX was also recently shown to be essential for iNKT cell development.61 The specific stage of iNKT development affected by TOX-deficiency was not determined, but the dramatic decrease in the percentage of CD1d-tet+ cells observed in these mice suggests an early block.

Although the Ras/MAPK cascade plays a central role in positive selection of conventional αβ T cells62,63 its involvement in iNKT cell development has not been studied in detail until recently, in part because an early report suggested that the number of NK1.1+ cells in the thymus of mice over-expressing dominant negative Ras and Mek-1 was normal.64 This result, together with data showing that cross-linking of SLAMF1 resulted in recruitment of RasGAP,65 resulted in a model where the role of the SLAM-derived signal would be to limit Ras/MAPK activation (see refs 4,24,25,66). However, a recent re-examination of iNKT populations in dnRas transgenic mice shows that inhibition of this pathway profoundly perturbs iNKT cell development, with a blockade at the early stages of positive selection (Control Point 1).58 Therefore, as with conventional αβ T cells the Ras/MAPK pathway plays a critical role during positive selection of iNKT cells. The Itk family kinases, which are important for the selection of conventional αβ T cells (reviewed in refs 55,56), also seem to play a role in iNKT cell development,67 but this effect seems to be evident first at later stages than those from mice defective in Ras or Egr.

The contribution of the NF-κB pathway to positive and negative selection of conventional αβ T cells is a controversial topic. In peripheral T cells, TCR triggering results in NF-κB activation mediated by protein kinase C-θ, Bcl10 CARMA-1 and Malt1, but gene knockout of these components does not alter positive selection of conventional αβ T cells.6870 However, other alterations further downstream from this pathway result in changes in T-cell development.71 In contrast, activation of the NF-κB pathway seems to be more important for the NKT cell lineage, because positive selection of NKT, but not conventional αβ T cells, is blocked in mice with a T-cell-specific deletion of IKK2.72,73 However, mice deficient in CARMA-1 and Bcl10 have normal NKT development.73,74 Protein kinase C-θ−/− mice show a partial defect in thymic development, with accumulation at immature stages, but almost normal populations in the periphery.73,75

SLAM/SAP and NKT cell development

The SLAM family consists of a number of related proteins including (Slamf1), CD48 (Slamf2), 2B4 (Slamf4), Ly9 (CD229, Slamf3), CD84 (Slamf5), NTB-A (Slamf6), Cracc (Slamf7), BLAME (Slamf8) and SF2001 (Slamf9). These receptors are expressed on the surface of a wide variety of haematopoietic cells, have diverse functions, including roles in regulating co-stimulation, T-cell cytokine production, adhesion between haematopoietic cells, the development of innate T lymphocytes (SLAM and Ly108) and others (see refs 76,77 for recent reviews on this family of receptors). Although DP thymocytes express high levels of SLAMF1, 2, 3, 5 and 6, and lower levels of SLAMF4, 7, 8 and 9,43,77,78 the only SLAM family members that seem to play a role in iNKT cell development are SLAMF1 and SLAMF6. Interestingly their function is redundant. Single knockouts have a partial phenotype,79,80 but a series of elegant mixed bone marrow chimeras clearly demonstrated that the combined lack of SLAMF1 and SLAMF6 causes a dramatic decrease in NKT cell numbers.51 This finding fits nicely with data obtained from NOD mice, a mouse strain that has been used as a model for autoimmune diabetes.The NOD mice have decreased levels of NKT cells.81 This phenotype was mapped to the SLAM locus,82 and a comparison of the expression pattern of SLAM family members in DP thymocytes showed a decrease in expression of SLAMF1 and SLAMF6, but no other SLAM family members.78 Recently, we found that the transcription factor c-Myb plays a role in NKT cell development in part by modulating expression of SLAMF1 and SLAMF6.43 Interestingly, the SLAM family expression patterns in DP thymocytes from mice carrying the NOD SLAM locus and in mice defective in c-Myb are very similar,78 suggesting that the defective expression of SLAM1 and SLAM6 may be the result of alterations in c-Myb binding.

It is well established that SLAM engagement recruits the adaptor SLAM-associated protein (SAP) and the Src kinase Fyn, both of which are essential for the selection of the iNKT cell lineage,8387 but the signal transduction pathways engaged downstream from the SLAM/SAP/FYN module that are relevant for NKT cell development are not well defined.

In a T-cell line SLAMF1 cross-linking induces recruitment and phosphorylation of of SHIP, Dok1, Dok2 and Ras-GTPase-activating protein (Ras-GAP)65 and this evidence has been used to propose a model where the SLAM-derived signals would inhibit activation of the Ras/MAPK cascade induced by the TCR.66 As discussed above,58 recent results argue against this model. SLAM-SAP triggering induces other signal transduction pathways. SLAM cross-linking on CD4 T cells results in AKT activation88 and prolongs protein kinase C-θ recruitment to the site of antigen-presenting cell contact, as well as influences Bcl-10 phosphorylation and patterns of NF-κB activation.89 It is unclear whether this effect on NF-κB activation by SLAM/SAP is important for NKT cell development. Activation of this pathway is important, as shown by the almost complete block in NKT cell development observed in IKK2−/− mice,73 and in mice expressing a degradation-resistant Iκ-Bα,90,91 but the contribution of different upstream activators is not clear.

Whether SLAM/SAP and TCR-derived signals cooperate to activate NF-κB during positive selection of DP thymocytes is unclear, although they do in mature CD4 T cells.89 Over-expression of a constitutively active IKKβ kinase in SAP−/− mice fails to rescue NKT cell development,92 suggesting that additional pathways downstream of SLAM/SAP are required. In over-expression studies, SAP also bound to PAK-interacting exchange factor (PIX), leading to synergistic NFAT (nuclear factor of activated T cells) activation in conjunction with ionomycin in Jurkat T cells93 and to NCK1 (non-catalytic region of tyrosine kinase 1).94 The possible contribution of these pathways to NKT cell development has not been explored.

Many factors important for the initial stages of NKT cell development have been recently identified. However, we still do not understand the specific contribution of the SLAM/SAP signal to this process, and how it complements TCR-derived signals. The DP-specific knockouts of other transcription factors such as Runx1,29 Fra-195 or Elf-196 also result in alterations at early stages in NKT cell development, but it is unclear whether signals from the TCR or SLAM affect the activity or expression of these transcription factors in this process, or how all these signals cooperate to set in motion the NKT-specific developmental programme.

Acknowledgments

This work was supported by grant AI059302 (NIH/NIAID) to J.A.I.

Disclosures

The authors declare no competing interests.

References

  • 1.Matsuda JL, Naidenko OV, Gapin L, Nakayama T, Taniguchi M, Wang CR, Koezuka Y, Kronenberg M. Tracking the response of natural killer T cells to a glycolipid antigen using CD1d tetramers. J Exp Med. 2000;192:741–54. doi: 10.1084/jem.192.5.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kronenberg M, Gapin L. The unconventional lifestyle of NKT cells. Nat Rev Immunol. 2002;2:557–68. doi: 10.1038/nri854. [DOI] [PubMed] [Google Scholar]
  • 3.Matsuda JL, Mallevaey T, Scott-Browne J, Gapin L. CD1d-restricted iNKT cells, the ‘Swiss-Army knife’ of the immune system. Curr Opin Immunol. 2008;20:358–68. doi: 10.1016/j.coi.2008.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Das R, Sant'Angelo DB, Nichols KE. Transcriptional control of invariant NKT cell development. Immunol Rev. 2010;238:195–215. doi: 10.1111/j.1600-065X.2010.00962.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Berzins SP, Cochrane AD, Pellicci DG, Smyth MJ, Godfrey DI. Limited correlation between human thymus and blood NKT cell content revealed by an ontogeny study of paired tissue samples. Eur J Immunol. 2005;35:1399–407. doi: 10.1002/eji.200425958. [DOI] [PubMed] [Google Scholar]
  • 6.Lee PT, Putnam A, Benlagha K, Teyton L, Gottlieb PA, Bendelac A. Testing the NKT cell hypothesis of human IDDM pathogenesis. J Clin Invest. 2002;110:793–800. doi: 10.1172/JCI15832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Montoya CJ, Pollard D, Martinson J, et al. Characterization of human invariant natural killer T subsets in health and disease using a novel invariant natural killer T cell-clonotypic monoclonal antibody, 6B11. Immunology. 2007;122:1–14. doi: 10.1111/j.1365-2567.2007.02647.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hammond KJ, Pellicci DG, Poulton LD, Naidenko OV, Scalzo AA, Baxter AG, Godfrey DI. CD1d-restricted NKT cells: an interstrain comparison. J Immunol. 2001;167:1164–73. doi: 10.4049/jimmunol.167.3.1164. [DOI] [PubMed] [Google Scholar]
  • 9.Rymarchyk SL, Lowenstein H, Mayette J, et al. Widespread natural variation in murine natural killer T-cell number and function. Immunology. 2008;125:331–43. doi: 10.1111/j.1365-2567.2008.02846.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cho YN, Kee SJ, Lee SJ, et al. Numerical and functional deficiencies of natural killer T cells in systemic lupus erythematosus: their deficiency related to disease activity. Rheumatology (Oxford) 2011;50:1054–63. doi: 10.1093/rheumatology/keq457. [DOI] [PubMed] [Google Scholar]
  • 11.Godfrey DI, Berzins SP. Control points in NKT-cell development. Nat Rev Immunol. 2007;7:505–18. doi: 10.1038/nri2116. [DOI] [PubMed] [Google Scholar]
  • 12.Venkataswamy MM, Porcelli SA. Lipid and glycolipid antigens of CD1d-restricted natural killer T cells. Semin Immunol. 2010;22:68–78. doi: 10.1016/j.smim.2009.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pei B, Speak AO, Shepherd D, Butters T, Cerundolo V, Platt FM, Kronenberg M. Diverse endogenous antigens for mouse NKT cells: self-antigens that are not glycosphingolipids. J Immunol. 2011;186:1348–60. doi: 10.4049/jimmunol.1001008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gapin L, Matsuda JL, Surh CD, Kronenberg M. NKT cells derive from double-positive thymocytes that are positively selected by CD1d. Nat Immunol. 2001;2:971–8. doi: 10.1038/ni710. [DOI] [PubMed] [Google Scholar]
  • 15.Bendelac A, Savage PB, Teyton L. The biology of NKT cells. Annu Rev Immunol. 2007;25:297–336. doi: 10.1146/annurev.immunol.25.022106.141711. [DOI] [PubMed] [Google Scholar]
  • 16.Kronenberg M, Engel I. On the road: progress in finding the unique pathway of invariant NKT cell differentiation. Curr Opin Immunol. 2007;19:186–93. doi: 10.1016/j.coi.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 17.Benlagha K, Wei DG, Veiga J, Teyton L, Bendelac A. Characterization of the early stages of thymic NKT cell development. J Exp Med. 2005;202:485–92. doi: 10.1084/jem.20050456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gapin L. The making of NKT cells. Nat Immunol. 2008;9:1009–11. doi: 10.1038/ni0908-1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kim PJ, Pai SY, Brigl M, Besra GS, Gumperz J, Ho IC. GATA-3 regulates the development and function of invariant NKT cells. J Immunol. 2006;177:6650–9. doi: 10.4049/jimmunol.177.10.6650. [DOI] [PubMed] [Google Scholar]
  • 20.McNab FW, Pellicci DG, Field K, Besra G, Smyth MJ, Godfrey DI, Berzins SP. Peripheral NK1.1 NKT cells are mature and functionally distinct from their thymic counterparts. J Immunol. 2007;179:6630–7. doi: 10.4049/jimmunol.179.10.6630. [DOI] [PubMed] [Google Scholar]
  • 21.Chun T, Page MJ, Gapin L, et al. CD1d-expressing dendritic cells but not thymic epithelial cells can mediate negative selection of NKT cells. J Exp Med. 2003;197:907–18. doi: 10.1084/jem.20021366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Pellicci DG, Uldrich AP, Kyparissoudis K, et al. Intrathymic NKT cell development is blocked by the presence of α-galactosylceramide. Eur J Immunol. 2003;33:1816–23. doi: 10.1002/eji.200323894. [DOI] [PubMed] [Google Scholar]
  • 23.Matsuda JL, Gapin L, Sidobre S, Kieper WC, Tan JT, Ceredig R, Surh CD, Kronenberg M. Homeostasis of Vα14i NKT cells. Nat Immunol. 2002;3:966–74. doi: 10.1038/ni837. [DOI] [PubMed] [Google Scholar]
  • 24.Berzins SP, Smyth MJ, Baxter AG. Presumed guilty: natural killer T cell defects and human disease. Nat Rev Immunol. 2011;11:131–42. doi: 10.1038/nri2904. [DOI] [PubMed] [Google Scholar]
  • 25.Godfrey DI, Stankovic S, Baxter AG. Raising the NKT cell family. Nat Immunol. 2010;11:197–206. doi: 10.1038/ni.1841. [DOI] [PubMed] [Google Scholar]
  • 26.D'Cruz LM, Yang CY, Goldrath AW. Transcriptional regulation of NKT cell development and homeostasis. Curr Opin Immunol. 2010;22:199–205. doi: 10.1016/j.coi.2010.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Krangel MS, Carabana J, Abbarategui I, Schlimgen R, Hawwari A. Enforcing order within a complex locus: current perspectives on the control of V(D)J recombination at the murine T-cell receptor α/δ locus. Immunol Rev. 2004;200:224–32. doi: 10.1111/j.0105-2896.2004.00155.x. [DOI] [PubMed] [Google Scholar]
  • 28.Guo J, Hawwari A, Li H, Sun Z, Mahanta SK, Littman DR, Krangel MS, He YW. Regulation of the TCRα repertoire by the survival window of CD4+CD8+ thymocytes. Nat Immunol. 2002;3:469–76. doi: 10.1038/ni791. [DOI] [PubMed] [Google Scholar]
  • 29.Egawa T, Eberl G, Taniuchi I, Benlagha K, Geissmann F, Hennighausen L, Bendelac A, Littman DR. Genetic evidence supporting selection of the Vα14i NKT cell lineage from double-positive thymocyte precursors. Immunity. 2005;22:705–16. doi: 10.1016/j.immuni.2005.03.011. [DOI] [PubMed] [Google Scholar]
  • 30.Bezbradica JS, Hill T, Stanic AK, Van Kaer L, Joyce S. Commitment toward the natural T (iNKT) cell lineage occurs at the CD4+ 8+ stage of thymic ontogeny. Proc Natl Acad Sci U S A. 2005;102:5114–9. doi: 10.1073/pnas.0408449102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dashtsoodol N, Watarai H, Sakata S, Taniguchi M. Identification of CD4–CD8– double-negative natural killer T cell precursors in the thymus. PLoS ONE. 2008;3:e3688. doi: 10.1371/journal.pone.0003688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Shortman K, Jackson H. The differentiation of T lymphocytes. I. Proliferation kinetics and interrelationships of subpopulations of mouse thymus cells. Cell Immunol. 1974;12:230–46. doi: 10.1016/0008-8749(74)90075-6. [DOI] [PubMed] [Google Scholar]
  • 33.Shortman K, Egerton M, Spangrude GJ, Scollay R. The generation and fate of thymocytes. Semin Immunol. 1990;2:3–12. [PubMed] [Google Scholar]
  • 34.Huesmann M, Scott B, Kisielow P, von Boehmer H. Kinetics and efficacy of positive selection in the thymus of normal and T cell receptor transgenic mice. Cell. 1991;66:533–40. doi: 10.1016/0092-8674(81)90016-7. [DOI] [PubMed] [Google Scholar]
  • 35.Claman HN. Corticosteroids and lymphoid cells. N Engl J Med. 1972;287:388–97. doi: 10.1056/NEJM197208242870806. [DOI] [PubMed] [Google Scholar]
  • 36.Wyllie AH. Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature. 1980;284:555–6. doi: 10.1038/284555a0. [DOI] [PubMed] [Google Scholar]
  • 37.Godfrey DI, Purton JF, Boyd RL, Cole TJ. Stress-free T-cell development: glucocorticoids are not obligatory. Immunol Today. 2000;21:606–11. doi: 10.1016/s0167-5699(00)01741-2. [DOI] [PubMed] [Google Scholar]
  • 38.Purton JF, Boyd RL, Cole TJ, Godfrey DI. Intrathymic T cell development and selection proceeds normally in the absence of glucocorticoid receptor signaling. Immunity. 2000;13:179–86. doi: 10.1016/s1074-7613(00)00018-2. [DOI] [PubMed] [Google Scholar]
  • 39.Vacchio MS, Ashwell JD. Glucocorticoids and thymocyte development. Semin Immunol. 2000;12:475–85. doi: 10.1006/smim.2000.0265. [DOI] [PubMed] [Google Scholar]
  • 40.Sun Z, Unutmaz D, Zou YR, Sunshine MJ, Pierani A, Brenner-Morton S, Mebius RE, Littman DR. Requirement for RORγ in thymocyte survival and lymphoid organ development. Science. 2000;288:2369–73. doi: 10.1126/science.288.5475.2369. [DOI] [PubMed] [Google Scholar]
  • 41.D'Cruz LM, Knell J, Fujimoto JK, Goldrath AW. An essential role for the transcription factor HEB in thymocyte survival, TCRa rearrangement and the development of natural killer T cells. Nat Immunol. 2010;11:240–9. doi: 10.1038/ni.1845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yuan J, Crittenden RB, Bender TP. c-Myb promotes the survival of CD4+ CD8+ double-positive thymocytes through upregulation of Bcl-xL. J Immunol. 2010;184:2793–804. doi: 10.4049/jimmunol.0902846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hu T, Simmons A, Yuan J, Bender TP, Alberola-Ila J. The transcription factor c-Myb primes CD4+ CD8+ immature thymocytes for selection into the iNKT lineage. Nat Immunol. 2010;11:435–41. doi: 10.1038/ni.1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ioannidis V, Beermann F, Clevers H, Held W. The β-catenin – TCF-1 pathway ensures CD4+CD8+ thymocyte survival. Nat Immunol. 2001;2:691–7. doi: 10.1038/90623. [DOI] [PubMed] [Google Scholar]
  • 45.Xie H, Huang Z, Sadim MS, Sun Z. Stabilized β-catenin extends thymocyte survival by up-regulating Bcl-xL. J Immunol. 2005;175:7981–8. doi: 10.4049/jimmunol.175.12.7981. [DOI] [PubMed] [Google Scholar]
  • 46.Hossain MZ, Yu Q, Xu M, Sen JM. ICAT expression disrupts β-catenin–TCF interactions and impairs survival of thymocytes and activated mature T cells. Int Immunol. 2008;20:925–35. doi: 10.1093/intimm/dxn051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cao Y, Li H, Liu H, Zheng C, Ji H, Liu X. The serine/threonine kinase LKB1 controls thymocyte survival through regulation of AMPK activation and Bcl-XL expression. Cell Res. 2010;20:99–108. doi: 10.1038/cr.2009.141. [DOI] [PubMed] [Google Scholar]
  • 48.Kasler HG, Young BD, Mottet D, Lim HW, Collins AM, Olson EN, Verdin E. Histone deacetylase 7 regulates cell survival and TCR signaling in CD4/CD8 double-positive thymocytes. J Immunol. 2011;186:4782–93. doi: 10.4049/jimmunol.1001179. [DOI] [PubMed] [Google Scholar]
  • 49.Li W, Kim MG, Gourley TS, McCarthy BP, Sant'Angelo DB, Chang CH. An alternate pathway for CD4 T cell development: thymocyte-expressed MHC class II selects a distinct T cell population. Immunity. 2005;23:375–86. doi: 10.1016/j.immuni.2005.09.002. [DOI] [PubMed] [Google Scholar]
  • 50.Li W, Sofi MH, Rietdijk S, Wang N, Terhorst C, Chang CH. The SLAM-associated protein signaling pathway is required for development of CD4+ T cells selected by homotypic thymocyte interaction. Immunity. 2007;27:763–74. doi: 10.1016/j.immuni.2007.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Griewank K, Borowski C, Rietdijk S, et al. Homotypic interactions mediated by Slamf1 and Slamf6 receptors control NKT cell lineage development. Immunity. 2007;27:751–62. doi: 10.1016/j.immuni.2007.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Veillette A, Dong Z, Latour S. Consequence of the SLAM-SAP signaling pathway in innate-like and conventional lymphocytes. Immunity. 2007;27:698–710. doi: 10.1016/j.immuni.2007.11.005. [DOI] [PubMed] [Google Scholar]
  • 53.Alberola-Ila J, Hernandez-Hoyos G. The Ras/MAPK cascade and the control of positive selection. Immunol Rev. 2003;191:79–96. doi: 10.1034/j.1600-065x.2003.00012.x. [DOI] [PubMed] [Google Scholar]
  • 54.Neilson JR, Winslow MM, Hur EM, Crabtree GR. Calcineurin B1 is essential for positive but not negative selection during thymocyte development. Immunity. 2004;20:255–66. doi: 10.1016/s1074-7613(04)00052-4. [DOI] [PubMed] [Google Scholar]
  • 55.Prince AL, Yin CC, Enos ME, Felices M, Berg LJ. The Tec kinases Itk and Rlk regulate conventional versus innate T-cell development. Immunol Rev. 2009;228:115–31. doi: 10.1111/j.1600-065X.2008.00746.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Readinger JA, Mueller KL, Venegas AM, Horai R, Schwartzberg PL. Tec kinases regulate T-lymphocyte development and function: new insights into the roles of Itk and Rlk/Txk. Immunol Rev. 2009;228:93–114. doi: 10.1111/j.1600-065X.2008.00757.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lazarevic V, Zullo AJ, Schweitzer MN, Staton TL, Gallo EM, Crabtree GR, Glimcher LH. The gene encoding early growth response 2, a target of the transcription factor NFAT, is required for the development and maturation of natural killer T cells. Nat Immunol. 2009;10:306–13. doi: 10.1038/ni.1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hu T, Gimferrer I, Simmons A, Wiest D, Alberola-Ila J. The Ras/MAPK pathway is required for generation of iNKT cells. PLoS ONE. 2011;6:e19890. doi: 10.1371/journal.pone.0019890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bettini M, Xi H, Milbrandt J, Kersh GJ. Thymocyte development in early growth response gene 1-deficient mice. J Immunol. 2002;169:1713–20. doi: 10.4049/jimmunol.169.4.1713. [DOI] [PubMed] [Google Scholar]
  • 60.Lauritsen JP, Kurella S, Lee SY, Lefebvre JM, Rhodes M, Alberola-Ila J, Wiest DL. Egr2 is required for Bcl-2 induction during positive selection. J Immunol. 2008;181:7778–85. doi: 10.4049/jimmunol.181.11.7778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Aliahmad P, Kaye J. Development of all CD4 T lineages requires nuclear factor TOX. J Exp Med. 2008;205:245–56. doi: 10.1084/jem.20071944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Alberola-Ila J, Forbush KA, Seger R, Krebs EG, Perlmutter RM. Selective requirement for MAP kinase activation in thymocyte differentiation. Nature. 1995;373:620–63. doi: 10.1038/373620a0. [DOI] [PubMed] [Google Scholar]
  • 63.Swan KA, Alberola-Ila J, Gross JA, Appleby MW, Forbush KA, Thomas JF, Perlmutter RM. Involvement of p21ras distinguishes positive and negative selection in thymocytes. EMBO J. 1995;14:276–85. doi: 10.1002/j.1460-2075.1995.tb07001.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Alberola-Ila J, Hogquist KA, Swan KA, Bevan MJ, Perlmutter RM. Positive and negative selection invoke distinct signaling pathways. J Exp Med. 1996;184:9–18. doi: 10.1084/jem.184.1.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Latour S, Gish G, Helgason CD, Humphries RK, Pawson T, Veillette A. Regulation of SLAM-mediated signal transduction by SAP, the X-linked lymphoproliferative gene product. Nat Immunol. 2001;2:681–90. doi: 10.1038/90615. [DOI] [PubMed] [Google Scholar]
  • 66.Borowski C, Bendelac A. Signaling for NKT cell development: the SAP-FynT connection. J Exp Med. 2005;201:833–6. doi: 10.1084/jem.20050339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Felices M, Berg LJ. The Tec kinases Itk and Rlk regulate NKT cell maturation, cytokine production, and survival. J Immunol. 2008;180:3007–18. doi: 10.4049/jimmunol.180.5.3007. [DOI] [PubMed] [Google Scholar]
  • 68.Sun Z, Arendt CW, Ellmeier W, et al. PKC-theta is required for TCR-induced NF-κB activation in mature but not immature T lymphocytes. Nature. 2000;404:402–7. doi: 10.1038/35006090. [DOI] [PubMed] [Google Scholar]
  • 69.Jost PJ, Weiss S, Ferch U, Gross O, Mak TW, Peschel C, Ruland J. Bcl10/Malt1 signaling is essential for TCR-induced NF-κB activation in thymocytes but dispensable for positive or negative selection. J Immunol. 2007;178:953–60. doi: 10.4049/jimmunol.178.2.953. [DOI] [PubMed] [Google Scholar]
  • 70.Egawa T, Albrecht B, Favier B, Sunshine MJ, Mirchandani K, O'Brien W, Thome M, Littman DR. Requirement for CARMA1 in antigen receptor-induced NF-κB activation and lymphocyte proliferation. Curr Biol. 2003;13:1252–8. doi: 10.1016/s0960-9822(03)00491-3. [DOI] [PubMed] [Google Scholar]
  • 71.Jimi E, Strickland I, Voll RE, Long M, Ghosh S. Differential role of the transcription factor NF-κB in selection and survival of CD4+ and CD8+ thymocytes. Immunity. 2008;29:523–37. doi: 10.1016/j.immuni.2008.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schmidt-Supprian M, Courtois G, Tian J, Coyle AJ, Israel A, Rajewsky K, Pasparakis M. Mature T cells depend on signaling through the IKK complex. Immunity. 2003;19:377–89. doi: 10.1016/s1074-7613(03)00237-1. [DOI] [PubMed] [Google Scholar]
  • 73.Schmidt-Supprian M, Tian J, Grant EP, et al. Differential dependence of CD4+ CD25+ regulatory and natural killer-like T cells on signals leading to NF-κB activation. Proc Natl Acad Sci U S A. 2004;101:4566–71. doi: 10.1073/pnas.0400885101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Medoff BD, Sandall BP, Landry A, Nagahama K, Mizoguchi A, Luster AD, Xavier RJ. Differential requirement for CARMA1 in agonist-selected T-cell development. Eur J Immunol. 2009;39:78–84. doi: 10.1002/eji.200838734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stanic AK, Bezbradica JS, Park JJ, Van Kaer L, Boothby MR, Joyce S. Cutting edge: the ontogeny and function of Va14Ja18 natural T lymphocytes require signal processing by protein kinase C theta and NF-κB. J Immunol. 2004;172:4667–71. doi: 10.4049/jimmunol.172.8.4667. [DOI] [PubMed] [Google Scholar]
  • 76.Veillette A. SLAM-family receptors: immune regulators with or without SAP-family adaptors. Cold Spring Harb Perspect Biol. 2010;2:a002469. doi: 10.1101/cshperspect.a002469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cannons JL, Tangye SG, Schwartzberg PL. SLAM family receptors and SAP adaptors in immunity. Annu Rev Immunol. 2010;29:665–705. doi: 10.1146/annurev-immunol-030409-101302. [DOI] [PubMed] [Google Scholar]
  • 78.Jordan MA, Fletcher JM, Pellicci D, Baxter AG. Slamf1, the NKT cell control gene Nkt1. J Immunol. 2007;178:1618–27. doi: 10.4049/jimmunol.178.3.1618. [DOI] [PubMed] [Google Scholar]
  • 79.Wang N, Satoskar A, Faubion W, et al. The cell surface receptor SLAM controls T cell and macrophage functions. J Exp Med. 2004;199:1255–64. doi: 10.1084/jem.20031835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Howie D, Laroux FS, Morra M, et al. Cutting edge: the SLAM family receptor Ly108 controls T cell and neutrophil functions. J Immunol. 2005;174:5931–5. doi: 10.4049/jimmunol.174.10.5931. [DOI] [PubMed] [Google Scholar]
  • 81.Baxter AG, Kinder SJ, Hammond KJ, Scollay R, Godfrey DI. Association between αβTCR+CD4–CD8– T-cell deficiency and IDDM in NOD/Lt mice. Diabetes. 1997;46:572–82. doi: 10.2337/diab.46.4.572. [DOI] [PubMed] [Google Scholar]
  • 82.Esteban LM, Tsoutsman T, Jordan MA, et al. Genetic control of NKT cell numbers maps to major diabetes and lupus loci. J Immunol. 2003;171:2873–8. doi: 10.4049/jimmunol.171.6.2873. [DOI] [PubMed] [Google Scholar]
  • 83.Nichols KE, Hom J, Gong SY, et al. Regulation of NKT cell development by SAP, the protein defective in XLP. Nat Med. 2005;11:340–5. doi: 10.1038/nm1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pasquier B, Yin L, Fondaneche MC, et al. Defective NKT cell development in mice and humans lacking the adapter SAP, the X-linked lymphoproliferative syndrome gene product. J Exp Med. 2005;201:695–701. doi: 10.1084/jem.20042432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chung B, Aoukaty A, Dutz J, Terhorst C, Tan R. Signaling lymphocytic activation molecule-associated protein controls NKT cell functions. J Immunol. 2005;174:3153–7. doi: 10.4049/jimmunol.174.6.3153. [DOI] [PubMed] [Google Scholar]
  • 86.Eberl G, Lowin-Kropf B, MacDonald HR. Cutting edge: NKT cell development is selectively impaired in Fyn-deficient mice. J Immunol. 1999;163:4091–4. [PubMed] [Google Scholar]
  • 87.Gadue P, Morton N, Stein PL. The Src family tyrosine kinase Fyn regulates natural killer T cell development. J Exp Med. 1999;190:1189–96. doi: 10.1084/jem.190.8.1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Howie D, Okamoto S, Rietdijk S, et al. The role of SAP in murine CD150 (SLAM)-mediated T-cell proliferation and interferon γ production. Blood. 2002;100:2899–907. doi: 10.1182/blood-2002-02-0445. [DOI] [PubMed] [Google Scholar]
  • 89.Cannons JL, Yu LJ, Hill B, et al. SAP regulates TH2 differentiation and PKC-theta-mediated activation of NF-κB1. Immunity. 2004;21:693–706. doi: 10.1016/j.immuni.2004.09.012. [DOI] [PubMed] [Google Scholar]
  • 90.Sivakumar V, Hammond KJ, Howells N, Pfeffer K, Weih F. Differential requirement for Rel/nuclear factor κB family members in natural killer T cell development. J Exp Med. 2003;197:1613–21. doi: 10.1084/jem.20022234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Stanic AK, Bezbradica JS, Park JJ, Matsuki N, Mora AL, Van Kaer L, Boothby MR, Joyce S. NF-κB controls cell fate specification, survival, and molecular differentiation of immunoregulatory natural T lymphocytes. J Immunol. 2004;172:2265–73. doi: 10.4049/jimmunol.172.4.2265. [DOI] [PubMed] [Google Scholar]
  • 92.Cen O, Ueda A, Guzman L, Jain J, Bassiri H, Nichols KE, Stein PL. The adaptor molecule signaling lymphocytic activation molecule-associated protein (SAP) regulates IFN-γ and IL-4 production in Vα14 transgenic NKT cells via effects on GATA-3 and T-bet expression. J Immunol. 2009;182:1370–8. doi: 10.4049/jimmunol.182.3.1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gu C, Tangye SG, Sun X, Luo Y, Lin Z, Wu J. The X-linked lymphoproliferative disease gene product SAP associates with PAK-interacting exchange factor and participates in T cell activation. Proc Natl Acad Sci U S A. 2006;103:14447–52. doi: 10.1073/pnas.0606624103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Li C, Schibli D, Li SS. The XLP syndrome protein SAP interacts with SH3 proteins to regulate T cell signaling and proliferation. Cell Signal. 2009;21:111–9. doi: 10.1016/j.cellsig.2008.09.014. [DOI] [PubMed] [Google Scholar]
  • 95.Lawson VJ, Maurice D, Silk JD, Cerundolo V, Weston K. Aberrant selection and function of invariant NKT cells in the absence of AP-1 transcription factor Fra-2. J Immunol. 2009;183:2575–84. doi: 10.4049/jimmunol.0803577. [DOI] [PubMed] [Google Scholar]
  • 96.Choi HJ, Geng Y, Cho H, Li S, Giri PK, Felio K, Wang CR. Differential requirements for the Ets transcription factor Elf-1 in the development of NKT cells and NK cells. Blood. 2011;117:1880–7. doi: 10.1182/blood-2010-09-309468. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES