Báo cáo khoa học: Tec family kinases: Itk signaling and the development of NKT ab and cd T cells potx

10 454 0
Báo cáo khoa học: Tec family kinases: Itk signaling and the development of NKT ab and cd T cells potx

Đang tải... (xem toàn văn)

Thông tin tài liệu

MINIREVIEW Tec family kinases: Itk signaling and the development of NKT ab and cd T cells Qian Qi 1,2 , Arun Kumar Kannan 1,2,3 and Avery August 1,2 1 Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology & Infectious Disease, The Pennsylvania State University, University Park, PA, USA 2 Department of Microbiology & Immunology, Cornell University, Ithaca, NY, USA 3 Immunology & Infectious Disease Graduate Program, The Pennsylvania State University, University Park, PA, USA Introduction Interleukin-2-inducible T-cell kinase (Itk) is a member of the Tec family of nonreceptor protein tyrosine kinases which includes Rlk and Tec, and is important for effective signaling through the T-cell receptor (TCR) [1,2]. There are additional Tec family kinases that signal from other receptors and have essential functions in other cell types, and these are reviewed in the accompanying minireviews [3]. In the absence of Itk, there are severe defects in activation of key signaling components including phospholipase C (PLC)c, which results in reduced influx of Ca 2+ , and defective activation of extracellular signal-regulated kinase ⁄ mitogen-activated protein kinase (ERK ⁄ MAPK), with resultant reduction in the activation of the transcription factors nuclear factor for activated T cells (NFAT), nuclear factor kappa-light chain enhancer of activated B cells (NFjB) and activator protein-1 [4]. A number of studies have examined the Keywords development; ERK; Id3; Interleukin-4; PLC; PLZF; SLP-76; signaling; T-bet; T cell receptor Correspondence A. August, Department of Microbiology & Immunology, C5 171 VMC, Cornell University, Ithaca, NY 14853-6401, USA Fax: +1 607 253 3384 Tel: +1 607 253 3400 E-mail: averyaugust@cornell.edu Note Q. Qi and A. K. Kannan contributed equally to this work (Received 31 August 2010, revised 28 October 2010, accepted 25 February 2011) doi:10.1111/j.1742-4658.2011.08074.x The Tec family tyrosine kinase interleukin-2 inducible T-cell kinase (Itk) is predominantly expressed in T cells and has been shown to be critical for the development, function and differentiation of conventional ab T cells. However, less is known about its role in nonconventional T cells such as NKT and cd T cells. In this minireview, we discuss evidence for a role for Itk in the development of invariant NKT ab cells, as well as a smaller pop- ulation NKT-like cd T cells. We discuss how these cells take what could be the same signaling pathway regulated by Itk, and interpret it to give differ- ent outcomes with regards to development and function. Abbreviations DN, double negative; ERK, extracellular signal-regulated kinase; Id3, inhibitor of DNA binding 3; IFN, interferon; IL, interleukin; i NKT, invariant natural killer T cells; Itk, interlukin-2 inducible T-cell kinase; MAPK, mitogen-activated protein kinase; NFAT, nuclear factor for activated T cells; NFjB, nuclear factor kappa-light chain enhancer of activated B cells; NK, natural killer cells; PLC, phospholipase C; PLZF, promyelocytic leukemia zinc finger protein; SAP, signaling lymphocyte activating molecule-associated protein; SLP-76, Src homology 2-domain containing leukocyte protein of 76 kDa; TCR, T-cell receptor. 1970 FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS role of Itk in T-cell development. In the absence of Itk, there is a partial block in the development of ab T cells and a reduced ratio of CD4 to CD8 single positive thy- mocytes in both the thymus and periphery [5]. In addi- tion, the absence of Itk was also found to affect positive and negative selection of thymocytes using TCR trans- genic mouse models, suggesting that Itk regulates the strength of the signal emanating from the TCR during T-cell selection [5–7]. Furthermore, combined deletion of Itk and Rlk leads to a further reduction in the TCR signal strength, resulting in the conversion of negative to positive selection and a rescue of T-cell numbers in T cell receptor transgenic mice [6]. More recently, Itk-deficient mice were reported to have reduced development of naı ¨ ve or conventional CD4 + and CD8 + T cells, and normal or increased development of CD4 + and CD8 + T cells, which have an activated memory-cell-like phenotype ([8–12], reviewed in [13] and [14]). These cells have increased expression of CD44 and CD122, have preformed mes- sage for interferon (IFN)- c, and are able to rapidly produce cytokines upon stimulation [8–12]. These cells, also referred to as innate memory phenotype T cells or nonconventional T cells, may develop via an indepen- dent pathway, dependent on expression of major histo- compatability complex molecules on bone-marrow- derived cells. These nonconventional or innate memory phenotype T cells share characteristics with invariant natural killer T cells (iNKT) and cd T cells, including the ability to rapidly produce cytokines, as well as alternative modes of development. The data that are accumulating suggest that the role of Itk in the devel- opment and function of iNKT cells and cd T cells seems to be quite different from conventional ab T cells. This minireview focuses on this aspect of Itk, its role in the development and function of iNKT and NKT-like cd T cells. Itk and i NKT cell development NKT cells are a subset of innate T cells characterized by their expression of the NK1.1 marker along with the ab TCR. Although these cells carry an antigen-specific TCR, they are characterized by their shared functions with natural killer (NK) cells. Thus NKT cells can directly kill target cells in an antigen-nonspecific fash- ion, but can also respond to stimulation via their TCR in an antigen-specific fashion. Like NK cells, they have the ability to rapidly produce large amounts of cyto- kines upon stimulation by ligands that interact with either their NK receptors or their TCRs. These cells share portions of their developmental program with conventional T cells. iNKT are a subset of NKT cells that largely express an invariant ab TCR. Both iNKT and conventional T cells develop in the thymus from T-cell progenitors derived from bone marrow, and progress through the CD4 + CD8 + double-positive thy- mocytes stage. However, iNKT cells diverge during positive selection and, in sharp contrast to conventional T cells, are selected to express a restricted ab TCR rep- ertoire characterized by a semiinvariant TCR chain formed through VDJ recombination. Although the pro- cess is stochastic, a majority of NKT cells carry a TCR composed of Va14–Ja18 segments, combined with either Vb8.2 or Vb7. These cells recognize glycolipids, proto- typically a-galactosyl ceramide (although a number of other ligands have been identified), in the context of the nonclassical major histocompatibility complex molecule CD1d [15]. Because of current technical difficulties in the isolation and analysis of other NKT cell subsets and their comparatively lower numbers, iNKT cells repre- sent the most widely studied NKT cell lineage. Unlike conventional T cells, iNKT cells are selected by CD1d expression on immature double-positive thymocytes [15,16]. Efficient selection of iNKT cells also depends on TCR signaling in response to cognate antigen in the context of CD1d. Indeed, a number of signaling molecules that lie downstream of the TCR can affect the development of iNKT cells (for review see [15,17]). iNKT cells pass through at least four stages of maturation based on their surface phenotype and expression of cytokines (Fig. 1) (reviewed in [17]). The earliest characterized iNKT cell progenitor is CD24 + ⁄ NK1.1 ) ⁄ CD44 ) (stage 0), and these progeni- tors can respond to interleukin (IL)-7. These cells then downregulate the expression of CD24 as they progress through to stage 1 (CD24 ) ⁄ NK1.1 ) ⁄ CD44 ) ). As the cells progress to stage 2 they upregulate CD44 (CD24 ) ⁄ NK1.1 ) ⁄ CD44 + ). At stages 1 and 2, these cells undergo extensive proliferation thus expanding the positively selected iNKT cell pool. Stage 3 marks the final maturation that can occur in either the thy- mus or the periphery. At this stage, most cells are CD44 hi ⁄ NK1.1 + and can secrete large amounts IFN-c and IL-4 [17]. These fully mature iNKT express high levels of the IL-15 receptor CD122 and their homeo- stasis is regulated by IL-15. The final maturation step, most clearly defined by the upregulation of NK1.1, is an important checkpoint to ensure normal numbers and frequency of iNKT cells in the periphery. This maturation step is also clinically relevant, as it has been implicated in ontogeny of autoimmunity induc- tion in nonobese diabetic (NOD) mice [18,19]. The Tec family kinases that are expressed in conven- tional T cells are also expressed in iNKT cells. Itk is the most abundantly expressed, followed by Txk ⁄ Rlk Q. Qi et al. Itk and i NKT and cd T cells FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 1971 (referred to as Txk) then Tec. All are upregulated in the mature NK1.1 + fraction when compared with NK1.1 ) cells in the thymus, although the expression levels are similar in the fractions in the periphery [20]. In the absence of Itk, there are reduced numbers of iNKT cells in the thymus and periphery [21]. More detailed analysis of the development of Itk-null i NKT cells revealed that they upregulate CD44, but fail to upregulate CD69, CD122 and NK1.1, thus fail- ing to progress to stage 3. The absence of Txk along with Itk results in a more severe block at the stage 2 ⁄ stage 3 transition point, suggesting that Txk may play some compensatory role in this developmental pathway [20]. In the absence of Itk, the splenic iNKT- cell population is increased in the CD44 lo ⁄ NK1.1 ) ⁄ CD69 ) population [20,22], which is exaggerated in the Itk ⁄ Txk double-knockout mice [20]. In the absence of Itk, there are also increased levels of apoptosis in peripheral iNKT cells [20]. This is sug- gested to correlate with decreased expression of the IL-15 receptor beta chain, CD122, which affects the IL-15 responsiveness of these cells in the periphery. CD122 expression is regulated in part by the transcrip- tional factor T-bet, the absence of which also results in a block on iNKT cell development [23,24]. Indeed, T-bet expression is reduced in Itk-deficient iNKT cells, and Itk may regulate the expression of T-bet in these cells, thus regulating iNKT cell development [20]. Role of Itk in i NKT cell function Analysis of the remaining Itk-null i NKT cells for cyto- kine production revealed that although these cells possess preformed mRNA for IL-4, IL-5, IL-13 and IFN-c, they lack the capacity to translate and secrete these cytokines upon antigenic stimulation both in vitro and in vivo [20,22,25]. By contrast, bypassing the TCR with the addition of 4b-phorbol 12-myristate 13-acetate and the calcium ionophore ionomycin can rescue cytokine secretion, indicating that TCR signals are defective for cytokine secretion in these cells in the absence of Itk. Thus although iNKT cell development is reduced in the absence of Itk, cells that can make it through this pathway are functionally able to make and secrete cytokines if full TCR signals are applied. Thus the absence of Itk does not affect the capacity of these cells to generate preformed cytokine mRNA and become poised for cytokine secretion [20,22]. In conventional T cells, particularly Th1 cells, IFN-c is predominantly regulated by T-bet, whereas IL-4 is Fig. 1. Involvement of Itk in the development of i NKT and NKT-like cd T cells. During T-cell development in the thymus, cd T cells separate from ab T cells during the CD4 ) CD8 ) DN thymocyte stage, although the exact separation point is unclear. Itk and SLP-76 regulate the develop- ment of NKT like Vc1.1 + ⁄ Vd6.2 ⁄ 3 + cd T cells, likely due to its ability to mediate TCR signal strength, regulating MAPK signaling, thus affecting the expression of PLZF and Id3. i NKT cells arise from CD4 + CD8 + double-positive thymocyte precursors through positive selection and develop through a series of developmental stages that ultimately become mature i NKT cells. Itk and Txk are involved in the final maturation of i NKT cells, which may be through regulating of the same pathway of TCR signal strength affecting the expression of transcription factors T-bet and PLZF, which is interpreted differently by developing i NKT cells. Dashed arrows indicate hypothesized or indirect interactions. Itk and i NKT and cd T cells Q. Qi et al. 1972 FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS regulated by the transcription factor GATA-3 [26]. However, in the absence of Itk, iNKT cells that do develop express IFN-c mRNA, despite the reduced expression of T-bet (although GATA-3 expression is normal) [20,22]. These findings suggest that IFN-c expression in iNKT cells may have less dependence on T-bet. Recently, the transcription factor promyelocytic leukemia zinc finger protein (PLZF), has been sug- gested to be a major regulator of i NKT cell develop- ment and function, with PLZF primarily expressed in iNKT cells and other nonconventional T cells [27]. PLZF belongs to the BTB-zinc finger family of tran- scription factors and in the absence of PLZF, the num- bers of mature iNKT cells is greatly reduced [28]. The homeostasis of these cells is also affected as a majority of iNKT cells in PLZF-null mice accumulate to lymph nodes, whereas the majority of iNKT cells in wild-type mice are found in the liver [28]. These PLZF-null iNKT cells also lack preformed mRNA for cytokines and are defective in cytokine production following TCR stimulation [28]. Whether Itk regulates expression and ⁄ or function of PLZF is unclear at this time, although there are some interesting findings along these lines as discussed below. Signaling by Itk in i NKT cells Itk plays a critical role in the increase in intracellular calcium in T cells, in part by interacting with Src homology 2-domain containing leukocyte protein of 76 kDa (SLP-76) and regulating tyrosine phosphoryla- tion and activation of PLC-c1 (Fig. 2) [1,2]. SLP-76 is also critical for the development of iNKT cells, partic- ularly the Itk binding site Y145 [29]. Thus the absence of the Itk signaling pathway results in reduced NFAT activation and expression of NFAT-regulated genes [30]. NFATc1 ⁄ NFAT2 is selectively upregulated after TCR stimulation in CD4 + iNKT cells and this can lead to a substantial increase in IL-4 production by these cells [31]. This NFAT activation may be due to Fig. 2. Signaling pathway leading to i NKT and NKT-like cd T cells regulated by Itk. Depiction of the signaling pathway used by the TCR and modulated by Itk that results in the development of i NKT ab and cd T cells. Note that in the case of the TCR, these pathways seem to be shared (negative regulation of PLZF, positive regulation of ERK), but lead to different developmental outcomes. Other pathways depicted such as PKC-h, CARMA–MALT–Bcl10 and NFjB that are critical for the development of i NKT cells are depicted for comparison. Dashed lines indicate proposed but indirect interactions. Q. Qi et al. Itk and i NKT and cd T cells FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 1973 activation of the calcium calcineurin–NFAT–Erg2 axis [32]. Although, further experiments are needed, the well-documented regulation of the Ca 2+ response, NFATc1 activation by Itk following TCR ligation in conventional T cells could be conserved in iNKT cells [1]. If conserved, this Itk-regulated signaling pathway leading to NFAT activation could be severely compro- mised, resulting in the observed deficiencies. Indeed, in conventional T cells, Itk is important for the induction of the transcription factor Egr2 (as well as Egr1 and -3), which lies downstream of NFAT [33]. Egr2 is uniquely critical for the development of iNKT cells, with the block observed at a similar stage to that observed in the absence of Itk in mice lacking this factor [20,32]. i NKT cell development and function in NFAT-deficient mice have not yet been analyzed, although the calcium path- way and calcineurin is critical for the development of these cells [32,34]. As discussed above, Itk-deficient iNKT cells express low levels of T-bet, and Itk may regulate the expres- sion of this critical transcription factor. Indeed, it has been suggested that Itk regulates T-bet levels in iNKT cells in the thymus, and that thymic egress favors those cells that express T-bet and CD122 [20]. In addition, iNKT cell development is dependent on signaling lymphocyte-activating molecule (SLAM), SLAM-associated protein (SAP), Src family kinase Fyn, PKCh, Bcl-10 and NFjB [17]. NFjB and PKCh are dispensable for selection of conventional T cells but critical for iNKT cell development [17]. Itk has been shown to modulate the localization of PKCh,as well as the activation of NFjB, and it is likely that the signaling pathway that Itk regulates is conserved in both conventional and iNKT cells [35,36]. Recent reports suggest that there may be some func- tional interaction between Itk and PLZF. PLZF is selectively upregulated in the CD4 + CD44 hi memory phenotype T cells that are found in the absence of Itk, although none of the CD8 + subsets expressed PLZF. These CD4 + CD44 hi cells have features of innate mem- ory phenotype cells discussed above [27]. In addition, the absence of PLZF leads to a block iNKT cell devel- opment at the initial stages of maturation [28], and both mice deficient in Itk and those that express a transgene for PLZF have a developmental block in stage 2 of iNKT cell maturation, all of which results in a severe reduction in the number and frequency of mature and functional iNKT cells. These seemingly opposing results might be explained by the tight regu- lation of PLZF expression during iNKT cell matura- tion. Cells in stage 1 express high levels of PLZF, and expression is decreased as the cells progress to stage 2, with levels are greatly reduced during final maturation to stage 3 [28]. Thus, the phenotype of PLZF-null mice could be due to the reliance of iNKT cells on this tran- scriptional regulator during the early stages of their development. The constitutive expression of PLZF in PLZF transgenic mice may lead to defective regulation of iNKT cell development and function. The findings reported to date favor a view that Itk regulates TCR signals which, dependent on the T-cell type, will have differential outcomes. Itk signals are important for the development of naı ¨ ve or conven- tional T cells, and are less important or not important for the development of nonconventional of innate memory phenotype T cells. By contrast, Itk is critical for the development of the iNKT-cell population. It remains to be seen if the molecular signals regulated by Itk are conserved in all of these T cell types. We next discuss another type of T cell whose development is dependent on Itk below, a small subset of cd T cell that have properties of NKT cells and express the CD4 and NK1.1 markers. Itk and NKT cd T-cell development Compared with ab T cells, the cd T-cell population is minor, comprising  5–10% of the total T cells in the blood and lymphoid organs. Although the cell num- bers are low in the periphery, cd T cells are more abundant in the skin and reproductive tract (as reviewed previously [37]). In this section, we discuss the role of Itk in the development of peripheral cd T cells, although Itk also plays a role in the develop- ment of skin cd T cells [38]. The cd T-cell population contains many distinct sub- sets which reside in different tissues, including the sec- ondary lymphoid organs and the epithelial layers of tissue such as the skin, intestinal epithelium and lung. The different subsets of cd T cells express distinct cd TCRs and develop at different times in the thymus. Skin cd T cells, also called skin-resident intraepithelial T lymphocytes, uniquely express Vc3 ⁄ Vd1, arise from fetal thymic precursor at around day 13, and become mature and migrate to the skin before birth in mice [39]. Vc4 + cd T cells are generated later than Vc3 + cd T cells in the fetal thymus and migrate to epithelial layers of reproductive tract, lung and tongue [39,40]. By contrast, cd T cells in the secondary lymphoid organs are only produced in the adult thymus, and they predominantly express Vc2 and Vc1.1 along with diverse Vd chains [41–45]. Populations of cd T cells that can uniquely secrete specific cytokines, including IL-17 or IL-4 have been described previously [45,46]. The IL-4 secreting population has been described as having properties of NKT cells [47]. Itk and i NKT and cd T cells Q. Qi et al. 1974 FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS An increasing number of studies suggest that TCR signaling strength determines T-cell lineage commit- ment, with stronger signals favoring cd T-cell develop- ment, whereas weaker signals favor ab T-cell development [48–50]. Although Itk may modulate TCR signal strength, and one might expect that reduced sig- nal strength received by Itk-null developing T cells would lead to reduced cd T-cell development, previous analysis of these cells in Itk-null mice suggested that this population is not affected [5]. However, we and others have recently reported that in the absence of Itk, the percentage and numbers of cd T cells in the adult thy- mus and secondary lymphoid organs is dramatically increased [51,52]. Further analysis showed that this increase is mainly due to the accumulation of a Vc1.1 ⁄ Vd6.3 subset of cd T cells, which express high levels of CD4 and NK1.1 [51,52]. This Vc1.1 ⁄ Vd6.3 sub- set of cd T cells are the same cd T-cell population previ- ously shown to secrete IL-4 and exhibit properties of NKT cells [47,53]. It has been suggested that these NKT cell-like Vc1.1 ⁄ Vd6.3 cd T cells may receive stron- ger TCR signals than other cd T-cell subsets, which in wild-type animals could lead to negative selection during development [53]. Because Itk may act as an amplifier in the TCR signaling, Itk deficiency may affect the SLP-76 signaling complex and dampen the TCR- mediated Ca 2+ influx and activation of PLCc1, weaken- ing downstream signals, such as ERK ⁄ MAPK, NFAT and activator protein-1 [54]. Thus the Itk deficiency may decrease TCR signal strength and allow some Vc1.1 ⁄ Vd6.3 cd T cells to survive negative selection. SLP-76 is an adaptor protein that interacts with, and is important for the activation of Itk and other signaling proteins during TCR signaling [55–57]. It is therefore of considerable interest that transgenic mice expressing two SLP-76 mutants including one carry- ing a mutant of the Itk binding site (Y145F, Y112- 128F) also exhibit significantly increased numbers of Vc1.1 ⁄ Vd6.3 cd T cells [29,58]. Thymocytes expressing these SLP-76 mutants have defects in TCR mediated PLC-c1 activation, Ca 2+ influx and Erk activation, demonstrating that TCR signal strength is weakened in these T cells [29]. These data suggest that Itk regu- lates the development of Vc1.1 ⁄ Vd6.3 cd T cells through altered TCR signaling strength via SLP-76. As discussed above, SAP is important in iNKT cell development. SAP deficiency in these SLP-76 trans- genic mice results in normalization (i.e. reduced numbers compared to the SLP-76 mutants) of the altered numbers of Vc1.1 ⁄ Vd6.3 cd T cells, suggesting that SAP is also involved in the developmental pathway of these Vc1.1 ⁄ Vd6.3 cd T cells [58]. Because SLP-76 and Itk interact during TCR signaling (via Y145), it is possible that SAP also modulates the pathway regu- lated by Itk in the development of Vc1.1 ⁄ Vd6.3 cd T cells. Inhibitor of DNA binding 3 (Id3) is an E-pro- tein inhibitor that is downstream of MAPK signaling pathway. Similar to Itk-null mice, mice lacking Id3 have alterations in cd T-cell development, and also show increased numbers of Vc1.1 ⁄ Vd6.3 cd T cells [58–61]. PLZF, shown to regulate iNKT cell develop- ment, has also been shown to regulate the development and function of this Vc1.1 ⁄ Vd6.2 ⁄ 3 cd T-cell popula- tion [58]. Itk may thus modulate TCR signals that regulate expression of PLZF and Id3, thus affecting development of this unique cd T-cell population. Because the microenvironments in the fetal thymus and adult thymus are different, the production of dis- tinct subsets of cd T cells during different stages of ontogeny suggests that they have distinct developmen- tal mechanisms. We have also found that that mice lacking Itk have significantly reduced numbers of another unique population of cd T cells that carry the Vc3 ⁄ Vd1 cd TCR and home to the skin, skin-resident intraepithelial T lymphocytes, suggesting that Itk is important for their development [38,62]. Further analy- sis indicates that Itk regulates the migration and hom- ing, but not maturation and homeostasis, of these cd skin-resident intraepithelial T lymphocytes. Thus Itk plays distinct roles in the development of different cd T-cell subsets. Role of Itk in NKT-cell-like function of Vc1.1/Vd6.3 cd T cells It has long been observed that naı ¨ ve Itk-null mice have high levels of serum IgE despite the observed defects in Th2 cytokine secretion from conventional and iNKT cells. IgE production is highly dependent on IL-4, and the CD4 + NKT-like Vc1.1 ⁄ Vd6.3 cd T cells can rapidly secrete IL-4 in vitro and in vivo [53,58,63]. Several published studies suggest that IL-4-secreting cd T cells contribute to helping B cells class switch to pro- duce IgE. Mice lacking ab T cells have normal B-cell phenotypes, germinal center formation and production of antibodies, particularly IgG1 and IgE, which was suggested to be due to the IL-4 production by cd T cells [64,65]. Human cd T cells can also induce class switch- ing in B cells to produce IgE [66]. In an allergic asthma model, mice lacking cd T cells had decreased production of IgE, which were rescued by adding IL-4, suggesting that cd T cells are important for IL-4 production and help the production of IgE and IgG1 [67]. The CD4 + NKT-like cd T cells observed in the Itk- null mice largely carry the Vc1.1 ⁄ Vd6.2 ⁄ 3 TCR, and stimulation of these cells purified from either wild-type Q. Qi et al. Itk and i NKT and cd T cells FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 1975 or Itk-null mice via the TCR induces large amounts of IL-4 [51,52]. The finding that these IL-4 producing CD4 + NKT-cell-like cd T cells accumulate in Itk-null mice suggests that these cells may be responsible for this paradoxical finding. Indeed, removing cd T cells from the Itk-null mice results in significantly reduced serum IgE [51,52], and transfer of these cells along with wild-type B cells induced class switch and IgE production in RAG-null mice [52]. These Itk-null NKT-like cd T cells express CD40L and OX40, costimulatory molecules that provide B-cell help, in response to anti-TCR d stimulation [51]. Interestingly, LAT mutant mice (Y175 ⁄ 195⁄ 235F), which have no ab T cells but accumulate high numbers of cd T cells in peripheral lymphoid organs, have high levels of serum IgE and IgG1, suggesting that LAT may also play a role in the development of similar if not the same population of NKT-like cd T cells that secrete IL-4 and can induce B-cell class switch [68]. One signal, many outcomes The combination of studies on conventional ab T-cell development, iNKT cell and cd T-cell development, including NKT-like cd T cells, suggests that signaling pathways regulated by Itk may be interpreted quite dif- ferently dependent on the cell type. Itk is critical for effective development of conventional ab T cells, the major T-cell population that participates in the immune response [1,13]. Similarly, signals regulated by Itk are important for effective development of iNKT cells, but interestingly not their primed state with preformed cytokine message, although Itk is required for cytokine secretion. By contrast, Itk seems to play a negative reg- ulatory role in the development of NKT-like cd T cells (carrying the Vc1.1 ⁄ Vd6.2 ⁄ 3 TCR), and does not affect their ability to secrete IL-4, but does affect the ability of other cd T-cell populations to secrete IFN-c. What can we surmise from these findings about the signals regulated by Itk downstream of the TCR dur- ing the development of these various subsets of T cells? Based on the studies to date, it is clear that the cal- cium pathway and SLP-76 are critical mediators of Itk. SLP-76, and particularly the Itk-binding site within SLP-76, is critical for the development of iNKT cells and plays a role in restraining the develop- ment of NKT-like Vc1.1 ⁄ Vd6.2 ⁄ 3 cd T cells, perhaps due to negative selection. Similarly, the Ras ⁄ Erk ⁄ MAPK pathway, also downstream of Itk, was previ- ously described as not being critical for the develop- ment of iNKT or cd T cells [69]. However, given the more recent studies, a re-examination of the role of Ras in the development of these cells seems to be war- ranted. With regards to transcriptional targets of the Itk pathway, although the spotlight has been on NFAT, other factors are coming into focus, in particu- lar, Egr family members (Egr1, -2 and -3), Id3 and PLZF. However, these pathways have different effects on iNKT cells versus NKT-like Vc1.1 ⁄ Vd6.2 ⁄ 3 cd T cells. In iNKT cells, the pathway is required, whereas in NKT-like Vc1.1 ⁄ Vd6.2 ⁄ 3 cd T cells, the pathway restrains. Given that both cell types can secrete IL-4, it is likely that the production of this cytokine, and the T-cell types that can produce it, need to be tightly controlled. Like iNKT cells, NKT-like Vc1.1 ⁄ Vd6.2 ⁄ 3 cd T cells seem to have a conserved ligand. Although yet to be identified, this ligand (or related ligands), may be involved in negative selection of these cells, likely via the Itk–SLP-76–ERK ⁄ MAPK module. Manipulating these pathways may result in differential manipulation of these cells and thus the immune response. Future experiments determining whether the same signaling pathway is used differen- tially by these different T-cell populations will be very informative. Nevertheless, these findings have impor- tant implications for the potential use of Itk inhibitors in various inflammatory diseases [2]. We have recently reported that iNKT cell develop- ment can be partially rescued by a kinase deleted mutant of Itk, suggesting that kinase activity is only partially required for the development of these cells. This partial rescue correlated with rescued expression of CD122 and T-bet, and suppression of Eomeso- dermin [70]. Acknowledgements This work was supported by National Institutes of Health Grants AI51626, AI065566, and AI073955 (to A.A.). References 1 Readinger J, Mueller K, Venegas A, Horai R & Schwartzberg P (2009) Tec kinases regulate T-lymphocyte development and function: new insights into the roles of Itk and Rlk ⁄ Txk. Immunol Rev 228, 93–114. 2 Sahu N & August A (2009) ITK inhibitors in inflamma- tion and immune-mediated disorders. Curr Top Med Chem 9, 690–703. 3 Ellmeier W, Abramova A & Schebesta A (2011) Tec family kinases: regulation of FceRI-mediated mast-cell activation. FEBS J 278, 1990–2000. 4 Schwartzberg P, Finkelstein L & Readinger J (2005) Tec-family kinases: regulators of T-helper-cell differenti- ation. Nat Rev Immunol 5, 284–295. Itk and i NKT and cd T cells Q. Qi et al. 1976 FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 5 Liao X & Littman D (1995) Altered T cell receptor sig- naling and disrupted T cell development in mice lacking Itk. Immunity 3, 757–769. 6 Schaeffer E, Broussard C, Debnath J, Anderson S, McVicar D & Schwartzberg P (2000) Tec family kinases modulate thresholds for thymocyte development and selection. J Exp Med 192, 987–1000. 7 Schaeffer E et al. (1999) Requirement for Tec kinases Rlk and Itk in T cell receptor signaling and immunity. Science 284, 638–641. 8 Broussard C, Fleischacker C, Horai R, Chetana M, Venegas AM, Sharp LL, Hedrick SM, Fowlkes BJ & Schwartzberg PL (2006) Altered development of CD8 + T cell lineages in mice deficient for the tec kinases Itk and Rlk. Immunity 25, 93–104. 9 Horai R, Mueller K, Handon R, Cannons J, Anderson S, Kirby M & Schwartzberg P (2007) Requirements for selection of conventional and innate T lymphocyte lin- eages. Immunity 27, 775–785. 10 Hu J & August A (2008) Naive and innate memory phenotype CD4 + T cells have different requirements for active Itk for their development. J Immunol 180, 6544–6552. 11 Hu J, Sahu N, Walsh E & August A (2007) Memory phenotype CD8 + T cells with innate function selectively develop in the absence of active Itk. Eur J Immunol 37, 2892–2899. 12 Atherly L, Lucas J, Felices M, Yin C, Reiner S & Berg L (2006) The Tec family tyrosine kinases Itk and Rlk regulate the development of conventional CD8 + T cells. Immunity 25, 79–91. 13 Berg LJ (2007) Signalling through TEC kinases regu- lates conventional versus innate CD8(+) T-cell develop- ment. Nat Rev Immunol 7, 479–485. 14 Gomez-Rodriguez J, Kraus Z & Schwartzberg P(2010) Tec family kinases Itk and Rlk ⁄ Txk in T lymphocytes: cross-regulation of cytokine production and T-cell fates. FEBS J 278, 1980–1989. 15 Bendelac A, Savage P & Teyton L (2007) The biology of NKT cells. Annu Rev Immunol 25, 297–336. 16 Wei D, Lee H, Park S, Beaudoin L, Teyton L, Lehuen A & Bendelac A (2005) Expansion and long-range dif- ferentiation of the NKT cell lineage in mice expressing CD1d exclusively on cortical thymocytes. J Exp Med 202, 239–248. 17 Godfrey D, Stankovic S & Baxter A (2010) Raising the NKT cell family. Nat Immunol 11, 197–206. 18 Hammond K, Poulton L, Palmisano L, Silveira P, Godfrey D & Baxter A (1998) T cell receptor (TCR)CD4CD8 (NKT) thymocytes prevent insulin- dependent diabetes mellitus in nonobese diabetic (NOD) ⁄ Lt mice by the influence of interleukin (IL)-4 and ⁄ or IL-10. J Exp Med 187, 1047–1056. 19 Lehuen A, Lantz O, Beaudoin L, Laloux V, Carnaud C, Bendelac A, Bach J & Monteiro R (1998) Overexpres- sion of natural killer T cells protects V14–J281 transgenic nonobese diabetic mice against diabetes. J Exp Med 188 , 1831–1839. 20 Felices M & Berg L (2008) The Tec kinases Itk and Rlk regulate NKT cell maturation, cytokine production, and survival. J Immunol 180, 3007–3018. 21 Gadue P & Stein P (2002) NK T cell precursors exhibit differential cytokine regulation and require Itk for efficient maturation. J Immunol 169, 2397–2406. 22 Au-Yeung B & Fowell D (2007) A key role for Itk in both IFN gamma and IL-4 production by NKT cells. J Immunol 179, 111–119. 23 Townsend M, Weinmann A, Matsuda J, Salomon R, Farnham P, Biron C, Gapin L & Glimcher L (2004) T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells. Immunity 20, 477–494. 24 Matsuda J, Zhang Q, Ndonye R, Richardson S, Howell A & Gapin L (2006) T-bet concomitantly controls migration, survival, and effector functions during the development of Valpha14i NKT cells. Blood 107, 2797– 2805. 25 Stetson D, Mohrs M, Reinhardt R, Baron J, Wang Z, Gapin L, Kronenberg M & Locksley R (2003) Constitu- tive cytokine mRNAs mark natural killer (NK) and NK T cells poised for rapid effector function. J Exp Med 198, 1069–1076. 26 Zhu J, Yamane H & Paul W (2010) Differentiation of effector CD4 T cell populations. Annu Rev Immunol 28, 445–489. 27 Raberger J et al. (2008) The transcriptional regulator PLZF induces the development of CD44 high memory phenotype T cells. Proc Natl Acad Sci USA 105, 17919– 17924. Epub 2008 Nov 12. 28 Kovalovsky D, Uche OU, Eladad S, Hobbs RM, Yi W, Alonzo E, Chua K, Eidson M, Kim HJ, Im JS et al. (2008) The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions. Nat Immunol 9 , 1055–1064. 29 Jordan M, Smith J, Burns J, Austin J, Nichols K, Aschenbrenner A & Koretzky G (2008) Complementation in trans of altered thymocyte development in mice express- ing mutant forms of the adaptor molecule SLP76. Immu- nity 28, 359–369. 30 Fowell D, Shinkai K, Liao X, Beebe A, Coffman R, Littman D & Locksley R (1999) Impaired NFATc translocation and failure of Th2 development in Itk deficient CD4 T cells. Immunity 11, 399–409. 31 Wang Z, Kusam S, Munugalavadla V, Kapur R, Brut- kiewicz R & Dent A (2006) Regulation of Th2 cytokine expression in NKT cells: unconventional use of Stat6, GATA-3, and NFAT2. J Immunol 176, 880–888. 32 Lazarevic V, Zullo A, Schweitzer M, Staton T, Gallo E, Crabtree G & Glimcher L (2009) The gene encoding early Q. Qi et al. Itk and i NKT and cd T cells FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 1977 growth response 2, a target of the transcription factor NFAT, is required for the development and maturation of natural killer T cells. Nat Immunol 10, 306–313. 33 Miller AT & Berg LJ (2002) Defective Fas ligand expression and activation-induced cell death in the absence of IL-2-inducible T cell kinase. J Immunol 168, 2163–2172. 34 Oh-hora M (2009) Calcium signaling in the develop- ment and function of T-lineage cells. Immunol Rev 231, 210–224. 35 Thuille N, Lutz-Nicoladoni C, Letschka T, Hermann- Kleiter N, Heit I & Baier G (2009) PKCtheta and Itk functionally interact during primary mouse CD3 + T cell activation. Immunol Lett 126, 54–59. 36 Schaeffer E, Yap G, Lewi C, Czar M, McVicar D, Cheever A, Sher A & Schwartzberg P (2001) Mutation of Tec family kinases alters T helper cell differentiation. Nat Immunol 2, 1183–1188. 37 Xiong N & Raulet D (2007) Development and selection of gammadelta T cells. Immunol Rev 215, 15–31. 38 Xia M, Qi Q, Jin Y, Wiest D, August A & Xiong N (2010) Differential roles of IL-2-inducible T cell kinase- mediated TCR signals in tissue-specific localization and maintenance of skin intraepithelial T cells. J Immunol 184, 6807–6814. 39 Jameson J, Witherden D & Havran WL (2003) T-cell effector mechanisms: gammadelta and CD1d-restricted subsets. Curr Opin Immunol 15, 349–353. 40 Bonneville M, Ito K, Krecko EG, Itohara S, Kappes D, Ishida I, Kanagawa O, Janeway CA, Murphy DB & Tonegawa S (1989) Recognition of a self major histocom- patibility complex TL region product by gamma delta T-cell receptors. Proc Natl Acad Sci USA 86, 5928–5932. 41 Heilig JS & Tonegawa S (1986) Diversity of murine gamma genes and expression in fetal and adult T lym- phocytes. Nature 322, 836–840. 42 Korman AJ, Marusic-Galesic S, Spencer D, Kruisbeek AM & Raulet DH (1988) Predominant variable region gene usage by gamma ⁄ delta T cell receptor-bearing cells in the adult thymus. J Exp Med 168, 1021–1040. 43 Takagaki Y, Nakanishi N, Ishida I, Kanagawa O & Tonegawa S (1989) T cell receptor-gamma and -delta genes preferentially utilized by adult thymocytes for the surface expression. J Immunol 142, 2112–2121. 44 Pereira P, Hermitte V, Lembezat MP, Boucontet L, Azuara V & Grigoriadou K (2000) Developmentally regulated and lineage-specific rearrangement of T cell receptor Valpha ⁄ delta gene segments. Eur J Immunol 30, 1988–1997. 45 Jensen KD, Su X, Shin S, Li L, Youssef S, Yamasaki S, Steinman L, Saito T, Locksley RM, Davis MM et al. (2008) Thymic selection determines gammadelta T cell effector fate: antigen-naive cells make interleukin-17 and antigen-experienced cells make interferon gamma. Immunity 29, 90–100. 46 Gerber D, Azuara V, Levraud J, Huang S, Lembezat M & Pereira P (1999) IL-4-producing gamma delta T cells that express a very restricted TCR repertoire are preferentially localized in liver and spleen. J Immunol 163, 3076–3082. 47 Azuara V, Levraud J, Lembezat M & Pereira P (1997) A novel subset of adult gamma delta thymocytes that secretes a distinct pattern of cytokines and expresses a very restricted T cell receptor repertoire. Eur J Immunol 27, 544–553. 48 Hayes SM, Li L & Love PE (2005) TCR signal strength influences alphabeta ⁄ gammadelta lineage fate. Immunity 22, 583–593. 49 Haks MC, Lefebvre JM, Lauritsen JP, Carleton M, Rhodes M, Miyazaki T, Kappes DJ & Wiest DL (2005) Attenuation of gammadeltaTCR signaling efficiently diverts thymocytes to the alphabeta lineage. Immunity 22, 595–606. 50 Kreslavsky T, Garbe AI, Krueger A & von Boehmer H (2008) T cell receptor-instructed alphabeta versus gam- madelta lineage commitment revealed by single-cell analysis. J Exp Med 205, 1173–1186. 51 Felices M, Yin CC, Kosaka Y, Kang J & Berg LJ (2009) Tec kinase Itk in gammadeltaT cells is pivotal for controlling IgE production in vivo. Proc Natl Acad Sci USA 106, 8308–8313. 52 Qi Q, Xia M, Hu J, Hicks E, Iyer A, Xiong N & August A (2009) Enhanced development of CD4 + gam- madelta T cells in the absence of Itk results in elevated IgE production. Blood 114, 564–571. 53 Kreslavsky T et al. (2009) TCR-inducible PLZF tran- scription factor required for innate phenotype of a sub- set of gammadelta T cells with restricted TCR diversity. Proc Natl Acad Sci USA 106, 12453–12458. 54 Berg LJ, Finkelstein LD, Lucas JA & Schwartzberg PL (2005) Tec family kinases in T lymphocyte development and function. Annu Rev Immunol 23, 549–600. 55 Bunnell SC, Diehn M, Yaffe MB, Findell PR, Cantley LC & Berg LJ (2000) Biochemical interactions integrat- ing Itk with the T cell receptor-initiated signaling cascade. J Biol Chem 275, 2219–2230. 56 Qi Q & August A (2007) Keeping the (kinase) party going: SLP-76 and ITK dance to the beat. Sci STKE 2007, pe39. 57 Bogin Y, Ainey C, Beach D & Yablonski D (2007) SLP-76 mediates and maintains activation of the Tec family kinase ITK via the T cell antigen receptor- induced association between SLP-76 and ITK. Proc Natl Acad Sci USA 104 , 6638–6643. 58 Alonzo ES, Gottschalk RA, Das J, Egawa T, Hobbs RM, Pandolfi PP, Pereira P, Nichols KE, Koretzky GA, Jordan MS et al. (2010) Development of promyelocytic zinc finger and ThPOK-expressing innate gamma delta T cells is controlled by strength of TCR signaling and Id3. J Immunol 184, 1268–1279. Itk and i NKT and cd T cells Q. Qi et al. 1978 FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 59 Verykokakis M, Boos M, Bendelac A, Adams E, Pere- ira P & Kee B (2010) Inhibitor of DNA binding 3 limits development of murine slam-associated adaptor pro- tein-dependent ‘innate’ gammadelta T cells. PLoS ONE 5, e9303. 60 Lauritsen JP, Wong GW, Lee SY, Lefebvre JM, Ciofani M, Rhodes M, Kappes DJ, Zu´ n ˜ iga-Pflu ¨ cker JC & Wiest DL (2009) Marked induction of the helix-loop- helix protein Id3 promotes the gammadelta T cell fate and renders their functional maturation Notch independent. Immunity 31, 565–575. 61 Ueda-Hayakawa I, Mahlios J & Zhuang Y (2009) Id3 restricts the developmental potential of gamma delta lineage during thymopoiesis. J Immunol 182, 5306– 5316. 62 Xiong N, Kang C & Raulet D (2004) Positive selection of dendritic epidermal gammadelta T cell precursors in the fetal thymus determines expression of skin-homing receptors. Immunity 21, 121–131. 63 Wen L, Barber DF, Pao W, Wong FS, Owen MJ & Hayday A (1998) Primary gamma delta cell clones can be defined phenotypically and functionally as Th1 ⁄ Th2 cells and illustrate the association of CD4 with Th2 differentiation. J Immunol 160, 1965–1974. 64 Wen L et al. (1994) Immunoglobulin synthesis and generalized autoimmunity in mice congenitally deficient in alpha beta(+) T cells. Nature 369, 654–658. 65 Wen L et al. (1996) Germinal center formation, immu- noglobulin class switching, and autoantibody produc- tion driven by ‘non alpha ⁄ beta’ T cells. J Exp Med 183, 2271–2282. 66 Horner AA, Jabara H, Ramesh N & Geha RS (1995) gamma ⁄ delta T lymphocytes express CD40 ligand and induce isotype switching in B lymphocytes. J Exp Med 181, 1239–1244. 67 Zuany-Amorim C, Ruffie C, Haile S, Vargaftig BB, Pereira P & Pretolani M (1998) Requirement for gam- madelta T cells in allergic airway inflammation. Science 280, 1265–1267. 68 Nun ˜ ez-Cruz S, Aguado E, Richelme S, Chetaille B, Mura AM, Richelme M, Pouyet L, Jouvin-Marche E, Xerri L, Malissen B et al. (2003) LAT regulates gammadelta T cell homeostasis and differentiation. Nat Immunol 4, 999–1008. 69 Alberola-Ila J, Hogquist K, Swan K, Bevan M & Perlmutter R (1996) Positive and negative selection invoke distinct signaling pathways. J Exp Med 184, 9–18. 70 Qi Q, Xia M, Bai Y, Yu S, Cantorna M & August A (2011) Interleukin-2-inducible T cell kinase (Itk) network edge dependence for the maturation of iNKT cell. J Biol Chem 286, 138–146. Q. Qi et al. Itk and i NKT and cd T cells FEBS Journal 278 (2011) 1970–1979 ª 2011 The Authors Journal compilation ª 2011 FEBS 1979 . regulated by Itk. Depiction of the signaling pathway used by the TCR and modulated by Itk that results in the development of i NKT ab and cd T cells. Note that in the case of the TCR, these pathways. of Itk in the development of i NKT and NKT- like cd T cells. During T- cell development in the thymus, cd T cells separate from ab T cells during the CD4 ) CD8 ) DN thymocyte stage, although the. conventional ab T cells. This minireview focuses on this aspect of Itk, its role in the development and function of iNKT and NKT- like cd T cells. Itk and i NKT cell development NKT cells are a subset of

Ngày đăng: 28/03/2014, 22:21

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan