Skip to main content
Journal of Leukocyte Biology logoLink to Journal of Leukocyte Biology
. 2014 Dec 18;97(3):477–485. doi: 10.1189/jlb.1RI0614-293R

The signaling symphony: T cell receptor tunes cytokine-mediated T cell differentiation

Weishan Huang 1,1, Avery August 1,1
PMCID: PMC4338847  PMID: 25525115

Review of the cross-talk between TCR and cytokine-mediated responses that control T cell behavior.

Keywords: pathway crosstalk, signal rheostat, proliferation, homeostasis, effector function

Abstract

T cell development, differentiation, and maintenance are orchestrated by 2 key signaling axes: the antigen-specific TCR and cytokine-mediated signals. The TCR signals the recognition of self- and foreign antigens to control T cell homeostasis for immune tolerance and immunity, which is regulated by a variety of cytokines to determine T cell subset homeostasis and differentiation. TCR signaling can synergize with or antagonize cytokine-mediated signaling to fine tune T cell fate; however, the latter is less investigated. Murine models with attenuated TCR signaling strength have revealed that TCR signaling can function as regulatory feedback machinery for T cell homeostasis and differentiation in differential cytokine milieus, such as IL-2-mediated Treg development; IL-7-mediated, naïve CD8+ T cell homeostasis; and IL-4-induced innate memory CD8+ T cell development. In this review, we discuss the symphonic cross-talk between TCR and cytokine-mediated responses that differentially control T cell behavior, with a focus on the negative tuning by TCR activation on the cytokine effects.

Introduction

T cell lineage differentiation relies on 2 major signals: 1, via the antigen-specific TCR and the other, via 1 or more cytokine receptors. The TCR is used by conventional T cells to recognize peptide antigens presented by MHC class I or II, expressed by APCs. This occurs over different developmental stages, from antigen-driven selection in the thymus and homeostasis post-thymic emigration to the response of naïve T cells to specific antigen in the periphery during an immune response to generate effector and memory T cells and the response of the latter cells to antigen re-exposure [1, 2]. Along with TCR signaling during T cell development, homeostasis, activation, and reactivation, cytokine signaling is critical for T cell proliferation, subset specialization, memory generation, maintenance, and recall [35]. The common γc cytokines, including IL-2, -4, -7, -9, -15, and -21, use IL-2Rγ as part of the receptor complex and signal through JAK/STAT pathways, acting in concert with TCR signals to drive normal T cell homeostasis, as well as immune responses [6]. The effects of these cytokines on regulating the differentiation of specific T cell subsets have been well investigated; however, whether and how TCR signals modulate these cytokine effects are less understood. Here, we summarize recent findings that suggest a critical regulatory role of the TCR and its proximal signalosome in cytokine-mediated T cell development or “TCR tuning.”

TCR SIGNALING AS NEGATIVE TUNER IN T CELL DEVELOPMENT AND HOMEOSTASIS

Activation of the TCR by peptide/MHC complexes triggers a downstream signaling cascade that can contribute to a variety of outcomes dependent on the stage of the T cell’s life [1, 7]. Upon TCR triggering, Src family kinase Lck is activated, leading to phosphorylation of ITAMs in the TCR/CD3 complex, an event that leads to the recruitment and activation of ZAP70, which phosphorylates further adaptor proteins LAT and SLP-76 [812] (see also review; ref. [13]). PI3K is also activated by Lck, catalyzing the generation of phosphatidylinositol (3,4,5)-trisphosphate lipids that interact with and recruit ITK onto the plasma membrane [14]. ITK can then interact with adaptor proteins LAT and SLP-76, which is critical for efficient activation of TCR signaling [15, 16]. Y145 in SLP-76 is involved in signaling downstream of ITK, and T cells expressing the Y145F mutant of SLP-76 exhibit similar developmental and functional defects to those lacking ITK [17, 18]. This ITK/SLP-76 clustering is part of a multiprotein complex that is able to regulate the actin cytoskeleton and other downstream signals (for review, see refs. [7, 1921]). This multiprotein complex further leads to phosphorylation of PLC-γ by ITK [22, 23]. PLC-γ catalyzes the generation of second messengers, which trigger calcium release [2427] and the subsequent activation and nuclear translocation of NFAT [13] and activation of PKCθ → Akt → NF-κB [2830] and RAS → MAPK [13] pathways. These pathways can control multiple events during the T cell’s life, including development and differentiation.

Although TCR signaling is necessary and a positive regulator in the differentiation of CD4+ naïve progenitors to Th1, Th2, and Th17 cells [3133], it has been shown to have a more complex role in the differentiation of CD4+ naïve progenitors to Tregs [3437]. In CD8+ T cells, TCR signals can contribute to regulatory feedback circuits that optimize CD8+ T cell homeostatic maintenance regulated by the cytokine IL-7 [38]. In addition, attenuated TCR signaling functionally enhances IL-4-induced development of IMP CD8+ T cells from CD8+ naïve thymic progenitors [39]. The latter findings reveal the versatility of TCR signaling in modulating cytokine-mediated T cell responses beyond previously held ideas about its role in this process. Therefore, intracellular signals triggered by the TCR may contribute to the tuning of cytokine-mediated signals. In the next sections, we discuss recent data indicating that TCR-triggered pathways negatively regulate or “tune” the response of T cells to cytokine-mediated signals, thus regulating T cell homeostasis and differentiation.

TCR SIGNALING NEGATIVELY TUNES IL-2-MEDIATED Treg DIFFERENTIATION

CD4+ Tregs are important immune regulators that promote self-tolerance in prevention of autoimmunity [40, 41] and act to restrain inflammatory responses to pathogens [42, 43]. tTregs develop from CD4+ SP T cells in the thymus when they receive high signals via the TCR (upon encounter with high levels or high affinity antigen/MHC complexes) and the costimulatory receptor CD28. These developing tTregs express the transcription factor Foxp3 and can further survive by up-regulating the IL-2R, stabilizing the Treg phenotype [4448]. Likewise, naïve CD4+ T cells in the periphery can be skewed toward the Treg fate when their TCR is triggered in the presence of the IL-2 and TGF-β, leading to the generation of iTregs [4953]. TCR signals that trigger the development of Tregs have been under intense study, and the strength of the TCR signal has been suggested to be a crucial parameter in the development of Tregs (e.g., tTregs [5457]). TCR signals are critical for the induction of Foxp3, as well as Foxp3-independent effects that lead to the development of Tregs (e.g., iTregs [58, 59]). The balance between IL-2 and TGF-β is critical for iTreg abundance and population size [60], and IL-2 signaling through STAT5 is indispensible for the survival of Foxp3-expressing cells during tTreg generation and homeostasis [45, 46]. The availability of IL-2 signaling can adjust the sensitivity of Treg to TCR signals during homeostatic proliferation, whereas TCR signals have been shown to be dispensable in the presence of elevated IL-2 [61]. Under pathogenic conditions, iTregs have been shown to be insensitive to activation-induced cell death but are very sensitive to IL-2 deprivation-induced death; TCR religation triggers an ERK and PI3K/mTOR-mediated loss of Foxp3 expression, resulting in the activation of an effector program in these cells, whereas the presence of TGF-β can attenuate the loss of Foxp3 [62]. TGF-β signaling activates the transcription factors Foxo1 and Foxo3a, which promote Foxp3 expression in iTregs [50, 53, 63]. This transcriptional activation of Foxp3 can be repressed by activation of the PI3K/Akt/mTOR pathway downstream of TCR [37] (Fig. 1). Intriguingly, Foxp3 negatively regulates TCR signaling circuits by directly suppressing components of the TCR proximal signalosome, including ZAP70 and ITK, as well as IL-2 [64], which may be a critical route for maintenance of tTregs. This cross-talk among TCR, IL-2, and TGF-β signaling pathways thus enables the TCR to act as a tuner of Treg differentiation (Fig. 1).

Figure 1. TCR tuning of IL-2-mediated Treg differentiation. Under Treg differentiation conditions, TGF-β activates transcriptional factors Foxo1/3a to enforce Foxp3 expression, whereas IL-2 activates STAT5, PI3K/Akt/mTOR, and ERK pathways to regulate cell proliferation and metabolism. TCR engagement activates the proximal signalosome involving ITAM/ZAP70/SLP-76/ITK to activate further ERK and PI3K/Akt/mTOR signaling, triggering PTEN turnover and Myc/miR19b-mediated targeting of PTEN to release PI3K/Akt/mTOR signaling from PTEN suppression. Active PI3K/Akt/mTOR is essential for glucose metabolism and can suppress Foxo-mediated Foxp3 expression. Foxp3, in turn, directly suppresses expression of IL-2, ITK, and ZAP70, further regulating PI3K/Akt/mTOR-mediated suppression of Foxp3 expression. Of note, the TCR proximal signalosome can negatively tune IL-2/STAT5 signaling strength, although the details are currently unclear.

Figure 1.

The intensity of TCR signaling has been suggested be an important factor in regulating Treg development, but its definitive role is unclear. Whereas it is reported that development of tTregs require high TCR signals [57, 65], it has also been suggested that TCR signals may need to be attenuated early after activation for optimal iTreg development [59]. Other data also suggest that low antigen dosage or impaired TCR signaling favors tTreg and iTreg differentiation [3437, 60, 66]. Although TCR activation is required to initiate Treg differentiation, high TCR signaling triggered by high antigen dose or high concentration of anti-CD3ε antibody induces strong activation of Akt/mTOR signaling that favors an effector CD4+ T cell fate and diminished iTreg development [35, 36] (Fig. 1). Interestingly, however, high affinity antigen given at a low dose or in disrupted periods increases the abundance of iTregs, suggesting complex regulation of Treg development by antigen potency, concentration, and duration of TCR signals [35].

Genetically modified mice that have impaired TCR signaling also exhibit altered Treg development, with enhanced tTreg abundance in vivo and iTreg differentiation in vitro. Mice carrying mutated TCR ζ chains that disrupt all 6 ITAMs and thus, have attenuated TCR activity show increased frequency of Tregs [67]. Likewise, mice that carry the SLP-76 Y145F mutation, which affects its interaction with ITK [66], or those that lack ITK [34, 68] exhibit increased frequency of tTreg in the thymus and/or periphery. Furthermore, naïve CD4+ T cell precursors inversely respond to incremental TCR signals under iTreg-differentiating conditions in vitro, with a reduced proportion of iTreg in the culture, and those that have reduced TCR signals as a result of the absence of ITK are unresponsive to the gradient of TCR signaling [34]. These findings support the view that CD4+ T cell differentiation into Tregs occurs at a higher frequency in the face of attenuated TCR signaling. Most interestingly, the reduction in TCR signals in the absence of ITK is accompanied by an enhanced responsiveness of the IL-2/STAT5 signaling pathway [34] and enhanced expansion in response to IL-2 in vivo [68], suggestive of a regulatory role for TCR signals in regulating Treg differentiation by tuning the signals they receive from IL-2.

The work of Gomez-Rodriguez et al. [34] recently reveals a potential mechanism for this TCR tuning of IL-2-mediated Treg differentiation. TCR activation results in down-regulation of the phosphatase PTEN in naïve CD4+ T cells and Tregs [69]. PTEN turnover alters the T cell response to IL-2, with resultant enhanced PI3K/Akt pathway activation, in addition to STAT5 phosphorylation [69] (Fig. 1). In the face of impaired TCR signal in Itk−/− T cells, PTEN degradation is attenuated, coupled with inefficient activation of the Akt/mTOR pathway and hyperactive responsiveness to IL-2 [34]. In support of the proposal that PTEN degradation is impaired, Itk−/− T cells exhibit impaired Myc and miR19b up-regulation, which normally represses PTEN expression (Fig. 1). The weakened mTOR and Myc pathways in Itk−/− cells are also likely to be the leading reasons for a decreased expression of the transcription factor HIF-1α and accompanying reduction in glucose metabolism, thus affecting energy production and proliferation in these cells [34]. Thus, TCR signals mediate regulation of PTEN, which is regulated by signals coming from ITK. PTEN then regulates IL-2 distal signaling and impacts the Treg differentiation. However, it is yet unclear whether the TCR proximal signalosome acts directly on IL-2 proximal signaling pathways to modulate signaling sensitivity.

“CORECEPTOR TUNING”: TCR SIGNALS ACT IN A NEGATIVE-FEEDBACK LOOP TO FINE TUNE IL-7-MEDIATED, NAïVE CD8+ T CELL HOMEOSTASIS

T cell homeostasis in the periphery is critical for maintenance of immunocompetence, and the survival and homeostasis of naïve T cells require IL-7 signaling [2, 7074]. The level of IL-7R expression is tightly controlled to optimize IL-7 consumption in support of T cell homeostasis [75]. The Singer group [76] has shown that naïve CD8+ T cell homeostasis is regulated by a negative-feedback loop, in which the IL-7R is transcriptionally repressed via signals induced by γc cytokines, including IL-7 itself. These naïve CD8+ T cells require a GFI-1-dependent pathway to dampen IL-7R expression in response to IL-7 or other γc cytokines [76]. In addition, naïve CD8+ T cells are subjected to modulation by a second regulatory feedback circuit: CD8 coreceptor-assisted, TCR-mediated, negative tuning of IL-7/IL-7R signaling [77].

In response to γc cytokines, including IL-2, IL-4, IL-7, and IL-15, CD8+ T cells up-regulate CD8 expression, which does not occur in response to non-γc cytokines, such as IL-6 and TNF-α [38, 78]. However, the engagement of TCR signals, with assistance of the CD8 coreceptor, during IL-7 stimulation can down-regulate IL-7 signaling. This reduced IL-7/STAT5 signaling activity, in turn, down-regulates CD8 expression, which reduces TCR/CD8 signaling and alleviates the TCR-mediated suppression of IL-7R expression and signals [38]. The CD8-mediated TCR signaling that suppresses IL-7R expression is the driving force for the oscillation of IL-7 and TCR signaling and is termed coreceptor tuning [77] (Fig. 2).

Figure 2. TCR/CD8 coreceptor tuning of IL-7-mediated CD8+ T cell homeostasis. IL-7/IL-7R signaling is critical for naïve CD8+ T cell homeostasis. IL-7R induces high levels of IFN-γ that induce CICD through auto- and paracrine mechanisms, which counteract the homeostatic proliferation. To prevent CICD, IL-7R activation induces GFI-1-dependent CD8 expression, which potentiates TCR-mediated negative tuning of IL-7R expression and thus, IFN-γ-induced CICD. The IL-7R → CD8 → TCR ⊣ IL-7R negative-feedback loop drives cell-intrinsic IL-7R and TCR oscillatory signaling.

Figure 2.

When released from this coreceptor tuning constraint, IL-7 can trigger signals necessary for CD8+ T cell proliferation under normal homeostasis. However, prolonged IL-7 signaling paradoxically induces CICD [77]. When the IL-7R is constitutively expressed on CD8+ T cells, the intrinsic oscillation driven by TCR/CD8-mediated, negative feedback or coreceptor tuning is disrupted, and IL-7-driven CD8+ T cell proliferation is elevated, accompanied by significant secretion of cytotoxic cytokine IFN-γ, which leads to CICD through auto- and paracrine effects (Fig. 2) [77]. This negative-feedback loop, IL-7R → CD8 → TCR ⊣ IL-7R (Fig. 2), thus forms a circuit that acts as a cell-intrinsic rheostat for tuning naïve CD8+ T cell homeostasis through TCR/CD8 and IL-7-mediated signaling oscillations.

TCR SIGNALING NEGATIVELY TUNES IL-4-INDUCED IMP CD8+ T CELL DEVELOPMENT

Innate memory T cells were discovered recently during the characterization of T cell phenotypes in Itk−/− mice, in which CD8 SP thymocytes were first found to be increased significantly [7981]. These cells were later shown to express memory T cell markers CD44 and CD122 and the transcription factor Eomes and are endowed with rapid effector cytokine production capacity upon stimulation [8286]. Although these phenotypes are typical characteristics of memory T cells derived from conventional T cell activation in the periphery, Itk−/− CD8 SP thymocytes gain them in the thymus during development, independently of peripheral stimulation, and have thus been termed memory-like or IMP T cells. The development of IMP T cells shares the early stages of conventional T cell differentiation and likely diverges from the double-positive stage. However, whereas IMP T cell development is dependent on hematopoietic cell–MHC expression, it can be independent of the thymic MHC and even the entire thymus, regardless of the presence of ITK [84, 86, 87].

Cells with similar phenotypes to the Itk−/− IMP CD8+ T cells have been observed in mice expressing the SLP-76 Y145F mutant (the mutant that disrupts ITK/SLP-76 coupling as described in earlier section) and others [18, 88, 89]. Ablation of the IL-4R blocks the elevation of the IMP CD8+ T cells in Itk−/− mice, supporting a critical role for IL-4 in development of IMP CD8+ T cells in the absence of ITK [39, 88]. In WT mice, iNKT cells are able to produce IL-4 in a PLZF-dependent manner and thus, were originally proposed to be the source of IL-4 for the development of IMP T cells [88]. However, Itk−/− iNKT cells are severely impaired in number as well as in production of IL-4 [9092], and so, the proposed candidates for the source of IL-4 have been suggested to be a subset of NKT-like γδ T cells [93, 94] and/or a CD4+ PLZFhi population of thymocytes [88] that are both capable of IL-4 production and are expanded in the absence of ITK. It is shown recently that iNKT and γδ T cells are dispensable for development of IMP CD8+ T cells in the absence of ITK [39]; thus, it is likely that Itk−/− CD4+ PLZFhi thymocytes produce sufficient IL-4 to drive development of Itk−/− IMP CD8+ T cells [88, 95]. Furthermore, Itk−/− CD8+ T cells exhibited better responsiveness to IL-4 than WT cells [39]. Intriguingly, similar to the case with Treg differentiation, the reduced TCR signaling manifest in the absence of ITK results in enhanced, IL-4-induced IMP development, suggesting that TCR signaling functions during development of IMP CD8+ T cells to tune IL-4 signals negatively [39]. Indeed, provision of exogenous IL-4 to OTI-Rag−/− mice in vivo results in the up-regulation of the Eomes protein and conversion of a significant population of naïve CD8+ T cells to the IMP, which was enhanced in the absence of ITK. When cultured with IL-4 in vitro, naïve OTI-Rag−/− CD8 SP thymocytes preferentially develop an IMP-like phenotype [39] (Fig. 3), and the frequency of these IL-4-induced, IMP-like CD8+ T cells is inversely correlated to the amount of TCR signals provided [39]. Of further interest is the finding that naïve, peripheral CD8+ T cells lacking ITK express elevated Eomes mRNA but lower Eomes protein, and provision of exogenous IL-4 induced significantly higher expression of Eomes protein in Itk−/− cells compared with WT cells, likely, in part, through translation of the premade Eomes mRNA [39]. These data suggest that Itk−/− CD8+ T cells receive weak TCR signals during development and may be primed to respond to IL-4 signals to become IMP cells.

Figure 3. TCR tuning of IL-4-induced IMP CD8+ T cell development. IL-4 drives STAT6-dependent Eomes expression in naïve CD8 SP thymic progenitors, leading to development of the IMP. IL-4 activates PI3K/Akt pathways and drives Eomes translation, likely involving mTOR-mediated translational machinery. TCR signals also activate PI3K/Akt but suppress IL-4R signaling.

Figure 3.

A role for the ITK-containing signalosome in the IL-4-induced generation of IMP CD8+ T cells is supported by findings from Carty and colleagues [96, 97], who have reported in conference abstracts that IL-4 induced enhanced STAT6 and Akt activation in SLP-76 Y145F innate-like CD8 SP thymocytes compared with conventional CD8 SP thymocytes. This negative tuning of IL-4 signals by the TCR may be facilitated or modulated by the reciprocal interaction between downstream STAT6 and ERK [98] (Fig. 3). PI3K activity has been shown to be important for IL-4-induced expression of IFN-γ and Eomes in CD8+ T cells in vitro [99], and it is possible that as seen for Treg differentiation, TCR/ITK regulation of PTEN may influence the level of IL-4 signaling. Given the fact that peripheral, naïve Itk−/− CD8+ T cells carry higher levels of preformed Eomes mRNA without efficient translation until IL-4 is provided [39], it is likely that enhanced Akt activity downstream of IL-4 is coupled with mTOR activity to regulate protein synthesis [100] (Fig. 3). Overall, these results reveal a suppressive function of the TCR proximal signalosome on STAT6 and Akt signaling, tuning IL-4-mediated IMP CD8+ T cell differentiation, in part, via regulation of expression of Eomes. Under conditions of reduced TCR signaling, there may be enhanced IL-4-induced signaling, contributing to enhanced IMP CD8+ T cell development (Fig. 3).

Similarly to IL-7-induced up-regulation of CD8, discussed in coreceptor tuning, IL-4/STAT6 stimulation of CD8+ T cells induces a significant increase in CD8 expression [38]. Thus, it is likely that coreceptor tuning may also be involved in IL-4-mediated IMP CD8+ T cell differentiation and homeostasis, as IMP CD8+ T cells accumulate under conditions of high levels of IL-4 and attenuated TCR signal strength. One interesting difference is the cytokine-modulated expression of the cytokine receptor: IL-7 signaling leads to IL-7R down-regulation, whereas IL-4 stimulation induces IL-4R expression [38], which may further complicate the outcome of TCR tuning on cytokine signaling. Nevertheless, IL-4/TCR and IL-7/TCR interplay shares similarities in cytokine-induced coreceptor expression and TCR tuning of a cytokine-mediated cell response, suggestive of a general mechanism used by the TCR to tune cytokine-induced T cell differentiation.

TCR CONDUCTS THE TUBA PLAYERS TO TUNE THE CYTOKINE PITCH

Although it is well accepted that Th2 cell differentiation requires effective TCR triggering, negative tuning of IL-4-mediated signaling by TCR ligation has also been reported in Th2 cells [32]. Strikingly, during the first 12 h following TCR triggering, naïve CD4+ T cells exhibit potent but transient suppression of IL-4-induced tyrosine phosphorylation of IL-4Rα, JAK1/3, STAT6, and insulin receptor substrate 2 [101]. This suppressive effect of TCR triggering on naïve CD4+ T cells also occurs following IL-2-induced STAT5 and IL-6-induced STAT3 activation, suggestive of a general phenomenon of negative tuning by TCR for cytokine-mediated signaling in CD4+ T cells [101]. As full cytokine signaling activity returns ∼20 h post-TCR ligation, this transient desensitization of cytokine signaling by TCR ligation may be a mechanism to tune preferentially specific programming to enhance T cell effector function before enrichment of the resultant population by cytokine-mediated T cell expansion and/or differentiation. CD4+ T cells defective in proximal TCR signalosome, such as in the absence of ITK, also have defects in IL-4-mediated Th2 [25, 102, 103] and IL-6/TGF-β-mediated Th17 [33] differentiation, and we speculate that TCR tuning of cytokine responses may play a role in this process as well.

WHO ARE THE TUBA PLAYERS?

Among all of the examples depicted above, there is a missing link between TCR activation and the suppression of cytokine signaling, in which the early engagement of the regulatory machinery may be the tuner during transient and/or long-term suppression. Downstream of γc cytokine/cytokine receptor triggering, the JAK/STAT signaling pathways are the most prominent players [3, 104106]. There are data that support a regulatory role for TCR signaling in tuning the γc expression/sensitivity and the downstream JAK/STAT6 activation.

TCR stimulation can lead to activation and increased expression of calpain [107], which has been demonstrated to be able to catalyze the proteolysis of γc [108] in a calcium-dependent fashion. The levels of intracellular calcium and calpain activity are inversely correlated with responsiveness of the IL-2/γc/STAT5 pathway [108, 109]. Furthermore, the inhibition of calpain can delay the progression of skin-graft rejection and multiple sclerosis in murine disease models, with delayed development of T cell effectors and/or enhanced Treg function [109, 110], similar to what has been observed in cases of impaired TCR proximal signalosome. Given the critical role of TCR proximal signaling in regulating calcium influx into T cells (see reviews; refs. [7, 21]), calcium-activated calpain may serve as the tuba player conducted by TCR in tuning down the γc expression and thus, the downstream cytokine signaling activity. Alternatively, it has been shown recently that activated T cells produce an alternatively spliced γc mRNA, encoding the sγc, which is secreted and competes with membrane-bound, full-length γc to alter T cell responses to IL-2 and IL-7, shown to lead to impaired survival and enhanced Th17 effector function in T cells [111]. sγc acts to tune down the immune-regulatory effects mediated by IL-2 and IL-7 during TCR activation, which may occur in CD8+ T cells as well, but it is unclear how activated TCR signaling triggers γc mRNA alternative splicing, the latter being pervasive in activated T cells [112]. Ca2+-independent TCR proximal signaling, mediated by PKC and Ras, was shown to be critical for alternative splicing of PTP CD45 during T cell activation [113]. It is likely that T cells have evolved Ca2+-dependent and -independent pathways downstream of TCR to modulate the intensity of γc interaction with cytokines.

JAK/STAT activation is regulated by multiple inhibitory mechanisms [114], among which are some components known to be induced by TCR signaling. These include proximal as well as distal downstream modulators, such as PTP (including CD45, SHP-1, and SHIP-1), SOCS (CIS1 and SOCS1–7), and PIAS that can suppress cytokine responses [114117]. CD45, expressed on the surface of T cells, can regulate an essential axis of the negative feedback downstream of TCR by recruiting an adaptor protein (Dok-1), suppressing IL-2-induced signaling [118]. TCR ligation induces the assembly of signaling complexes that include Dok-1/2, SHIP-1, and growth factor receptor-bound protein 2, which negatively tune LAT/ZAP70 phosphorylation and IL-2 production [119]. IL-4-induced STAT6 activation exhibits transient hypersensitivity in Dok-1−/− splenocytes [120]. Of interest, given the findings with TCR tuning of IL-4 to induce CD8+ IMP cells, is the finding that overexpression of Dok-1 suppresses STAT6 activation and GATA3 expression in CD4+ T cells [121]. Another PTP, SHP-1, can be recruited to lipid rafts in a TCR signaling-mediated manner [122], and its expression is gradually enhanced in CD8+ T cells exhibiting increased TCR affinity over time [123], suggesting that the TCR may be able to orchestrate cytokine signaling through activation and/or expression of SHP-1. In support of the idea that SOCS can tune T cell development and homeostasis, SOCS1−/− CD8+ T cells exhibit IL-7/IL-15-dependent hyperproliferation in lymphopenic hosts [124]. Furthermore, SOCS3, a well-known, cytokine-induced regulatory gene, can be up-regulated by TCR triggering as well [125]. TCR triggering also induces the expression of CIS1, which can attenuate IL-2-induced STAT5 activation [126]. Although a modest change in STAT1-related cytokine signaling has been observed in Pias4−/− mice, no overt difference in lymphocytes has been determined [127, 128]. However, given the high homology of the 5 PIAS family members [129], this may be a result of compensation as a result of functional redundancy. Thus, the role of PIAS in T cells and their functional behavior downstream of TCR are of considerable interest but remain to be elucidated.

Whereas it is unclear whether calpain, PTP/Dok, SOCS, and/or PIAS are downstream participants in the TCR-mediated negative tuning described above, TCR triggering can indeed activate and/or induce expression of some members of these groups, making them promising candidates to bridge the TCR in tuning γc cytokine expression and signaling during T cell differentiation and homeostasis (Fig. 4).

Figure 4. Generalized model for TCR tuning of cytokine-mediated T cell differentiation and homeostasis. γc cytokines (IL-2/IL-4/IL-7) activate the receptor complex and the downstream JAK/STAT and PI3K/Akt signaling pathways. Active STAT can enhance cell proliferation and directly or indirectly modulate expression of effector components, such as cytokines, coreceptor, or TCR signalosome components (e.g., Eomes → IFN-γ, GFI-1 → CD8, Foxp3 ⊣ IL-2/ITK). TCR triggering (with assistance from the coreceptor) suppresses cytokine receptor signaling, likely through modulating receptor complex expression or receptor/JAK/STAT signaling cascade via alternative splicing of RNA, calcium → calpain, PTPs, Dok, SOCS, and/or PIAS. TF, transcription factor.

Figure 4.

CONCLUDING REMARKS

The examples of TCR signals tuning γc cytokine (IL-2/IL-4/IL-7) signaling, discovered so far, suggest that the TCR is not just a receptor for activation of T cells but is also a rheostat that can tune cytokine responses to control diverse, effective outcomes. Depending on the situation, TCR tuning of cytokine effects may create a window of time that allows T cell effector programming before the cells go on with cytokine-driven population expansion. This may be an essential mechanism for T cell memory formation to potentiate antigenic specificity. In the cytokine milieu, cytokine-driven T cell homeostasis and differentiation are thus modulated by TCR signals through the cell-extrinsic antigenic stimulation or cell-intrinsic alteration in TCR signaling strength. We suggest that this property of the TCR may be exploited to optimize the development of specific T cell lineages and/or memory responses.

ACKNOWLEDGMENTS

This work was supported, in part, by grants from the U.S. National Institutes of Health (AI073955 and AI108958; to A.A.). The authors thank members of the Department of Microbiology & Immunology at Cornell University for discussion and feedback and Dr. Jean K. Millet for reading the manuscript.

Glossary

−/−

deficient

γc

cytokine receptor common γ chain

Akt

protein kinase B

CICD

cytokine-induced cell death

CIS1

cytokine-induced Src homology 2 protein 1

Dok-1

downstream of tyrosine kinase 1

Eomes

eomesodermin

Foxp3

forkhead box p3

GFI-1

growth factor independence 1

HIF-1α

hypoxia-inducible factor 1-α

IMP

innate memory phenotype

iNKT

invariant NK T cell

ITK

IL-2-inducible T cell kinase

iTreg

induced Foxp3-expressing conventional regulatory T cell

LAT

linker for activation of T cells

miR19b

microRNA 19b

mTOR

mammalian target of rapamycin

OTI

transgenic TCR recognizing OVA peptides 257–264 presented by MHCI

PIAS

protein inhibitor of activated STAT

PKC

protein kinase C

PLC

phospholipase C

PLZF

promyelocytic leukemia zinc finger

PTEN

phosphatase and tensin homolog

PTP

protein tyrosine phosphatase

sγc

soluble extracellular domain of cytokine receptor common γ chain

SHIP-1

Src homology 2-containing inositol phosphatase-1

Src

homology 2-containing phosphatase-1

SLP-76

Src homology 2-containing phosphatase-1

SLP-76

Src homology 2 domain-containing leukocyte protein of 76 kDa

SOCS

suppressor of cytokine signaling

SP

single-positive

Treg

Foxp3-expressing conventional regulatory T cell

tTreg

thymic-derived Foxp3-expressing conventional regulatory T cell

WT

wild-type

Y145

tyrosine 145

AUTHORSHIP

W.H. and A.A. wrote the manuscript.

DISCLOSURES

The authors declare no competing financial interests.

REFERENCES

  • 1.Anderson G., Moore N. C., Owen J. J., Jenkinson E. J. (1996) Cellular interactions in thymocyte development. Annu. Rev. Immunol. 14, 73–99. [DOI] [PubMed] [Google Scholar]
  • 2.Surh C. D., Sprent J. (2008) Homeostasis of naive and memory T cells. Immunity 29, 848–862. [DOI] [PubMed] [Google Scholar]
  • 3.Schluns K. S., Lefrançois L. (2003) Cytokine control of memory T-cell development and survival. Nat. Rev. Immunol. 3, 269–279. [DOI] [PubMed] [Google Scholar]
  • 4.Zhu J., Paul W. E. (2010) Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol. Rev. 238, 247–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.O’Garra A. (1998) Cytokines induce the development of functionally heterogeneous T helper cell subsets. Immunity 8, 275–283. [DOI] [PubMed] [Google Scholar]
  • 6.Rochman Y., Spolski R., Leonard W. J. (2009) New insights into the regulation of T cells by gamma(c) family cytokines. Nat. Rev. Immunol. 9, 480–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kannan A., Huang W., Huang F., August A. (2012) Signal transduction via the T cell antigen receptor in naïve and effector/memory T cells. Int. J. Biochem. Cell Biol. 44, 2129–2134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Irving B. A., Weiss A. (1991) The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways. Cell 64, 891–901. [DOI] [PubMed] [Google Scholar]
  • 9.Letourneur F., Klausner R. D. (1992) Activation of T cells by a tyrosine kinase activation domain in the cytoplasmic tail of CD3 epsilon. Science 255, 79–82. [DOI] [PubMed] [Google Scholar]
  • 10.Chan A. C., Iwashima M., Turck C. W., Weiss A. (1992) ZAP-70: a 70 kd protein-tyrosine kinase that associates with the TCR zeta chain. Cell 71, 649–662. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang W., Sloan-Lancaster J., Kitchen J., Trible R. P., Samelson L. E. (1998) LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular activation. Cell 92, 83–92. [DOI] [PubMed] [Google Scholar]
  • 12.Bubeck Wardenburg J., Fu C., Jackman J. K., Flotow H., Wilkinson S. E., Williams D. H., Johnson R., Kong G., Chan A. C., Findell P. R. (1996) Phosphorylation of SLP-76 by the ZAP-70 protein-tyrosine kinase is required for T-cell receptor function. J. Biol. Chem. 271, 19641–19644. [DOI] [PubMed] [Google Scholar]
  • 13.Smith-Garvin J. E., Koretzky G. A., Jordan M. S. (2009) T cell activation. Annu. Rev. Immunol. 27, 591–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.August A., Sadra A., Dupont B., Hanafusa H. (1997) Src-induced activation of inducible T cell kinase (ITK) requires phosphatidylinositol 3-kinase activity and the Pleckstrin homology domain of inducible T cell kinase. Proc. Natl. Acad. Sci. USA 94, 11227–11232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ching K. A., Grasis J. A., Tailor P., Kawakami Y., Kawakami T., Tsoukas C. D. (2000) TCR/CD3-induced activation and binding of Emt/Itk to linker of activated T cell complexes: requirement for the Src homology 2 domain. J. Immunol. 165, 256–262. [DOI] [PubMed] [Google Scholar]
  • 16.Bunnell S. C., Diehn M., Yaffe M. B., Findell P. R., Cantley L. C., Berg L. J. (2000) Biochemical interactions integrating Itk with the T cell receptor-initiated signaling cascade. J. Biol. Chem. 275, 2219–2230. [DOI] [PubMed] [Google Scholar]
  • 17.Jordan M. S., Smith J. E., Burns J. C., Austin J. E., Nichols K. E., Aschenbrenner A. C., Koretzky G. A. (2008) Complementation in trans of altered thymocyte development in mice expressing mutant forms of the adaptor molecule SLP76. Immunity 28, 359–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gordon S. M., Carty S. A., Kim J. S., Zou T., Smith-Garvin J., Alonzo E. S., Haimm E., Sant’Angelo D. B., Koretzky G. A., Reiner S. L., Jordan M. S. (2011) Requirements for eomesodermin and promyelocytic leukemia zinc finger in the development of innate-like CD8+ T cells. J. Immunol. 186, 4573–4578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.August A., Ragin M. J. (2012) Regulation of T-cell responses and disease by tec kinase Itk. Int. Rev. Immunol. 31, 155–165. [DOI] [PubMed] [Google Scholar]
  • 20.Berg L. J., Finkelstein L. D., Lucas J. A., Schwartzberg P. L. (2005) Tec family kinases in T lymphocyte development and function. Annu. Rev. Immunol. 23, 549–600. [DOI] [PubMed] [Google Scholar]
  • 21.Andreotti A. H., Schwartzberg P. L., Joseph R. E., Berg L. J. (2010) T-Cell signaling regulated by the Tec family kinase, Itk. Cold Spring Harb. Perspect. Biol. 2, a002287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Joseph R. E., Min L., Xu R., Musselman E. D., Andreotti A. H. (2007) A remote substrate docking mechanism for the tec family tyrosine kinases. Biochemistry 46, 5595–5603. [DOI] [PubMed] [Google Scholar]
  • 23.Perez-Villar J. J., Kanner S. B. (1999) Regulated association between the tyrosine kinase Emt/Itk/Tsk and phospholipase-C gamma 1 in human T lymphocytes. J. Immunol. 163, 6435–6441. [PubMed] [Google Scholar]
  • 24.Hogan P. G., Lewis R. S., Rao A. (2010) Molecular basis of calcium signaling in lymphocytes: STIM and ORAI. Annu. Rev. Immunol. 28, 491–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fowell D. J., Shinkai K., Liao X. C., Beebe A. M., Coffman R. L., Littman D. R., Locksley R. M. (1999) Impaired NFATc translocation and failure of Th2 development in Itk-deficient CD4+ T cells. Immunity 11, 399–409. [DOI] [PubMed] [Google Scholar]
  • 26.Schaeffer E. M., Debnath J., Yap G., McVicar D., Liao X. C., Littman D. R., Sher A., Varmus H. E., Lenardo M. J., Schwartzberg P. L. (1999) Requirement for Tec kinases Rlk and Itk in T cell receptor signaling and immunity. Science 284, 638–641. [DOI] [PubMed] [Google Scholar]
  • 27.Liu K. Q., Bunnell S. C., Gurniak C. B., Berg L. J. (1998) T Cell receptor-initiated calcium release is uncoupled from capacitative calcium entry in Itk-deficient T cells. J. Exp. Med. 187, 1721–1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wegener E., Oeckinghaus A., Papadopoulou N., Lavitas L., Schmidt-Supprian M., Ferch U., Mak T. W., Ruland J., Heissmeyer V., Krappmann D. (2006) Essential role for IkappaB kinase beta in remodeling Carma1-Bcl10-Malt1 complexes upon T cell activation. Mol. Cell 23, 13–23. [DOI] [PubMed] [Google Scholar]
  • 29.Narayan P., Holt B., Tosti R., Kane L. P. (2006) CARMA1 is required for Akt-mediated NF-kappaB activation in T cells. Mol. Cell. Biol. 26, 2327–2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Blonska M., Lin X. (2011) NF-κB signaling pathways regulated by CARMA family of scaffold proteins. Cell Res. 21, 55–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yamashita M., Kimura M., Kubo M., Shimizu C., Tada T., Perlmutter R. M., Nakayama T. (1999) T Cell antigen receptor-mediated activation of the Ras/mitogen-activated protein kinase pathway controls interleukin 4 receptor function and type-2 helper T cell differentiation. Proc. Natl. Acad. Sci. USA 96, 1024–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yamashita M., Katsumata M., Iwashima M., Kimura M., Shimizu C., Kamata T., Shin T., Seki N., Suzuki S., Taniguchi M., Nakayama T. (2000) T cell receptor-induced calcineurin activation regulates T helper type 2 cell development by modifying the interleukin 4 receptor signaling complex. J. Exp. Med. 191, 1869–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gomez-Rodriguez J., Sahu N., Handon R., Davidson T. S., Anderson S. M., Kirby M. R., August A., Schwartzberg P. L. (2009) Differential expression of interleukin-17A and -17F is coupled to T cell receptor signaling via inducible T cell kinase. Immunity 31, 587–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gomez-Rodriguez J., Wohlfert E. A., Handon R., Meylan F., Wu J. Z., Anderson S. M., Kirby M. R., Belkaid Y., Schwartzberg P. L. (2014) Itk-mediated integration of T cell receptor and cytokine signaling regulates the balance between Th17 and regulatory T cells. J. Exp. Med. 211, 529–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gottschalk R. A., Corse E., Allison J. P. (2010) TCR ligand density and affinity determine peripheral induction of Foxp3 in vivo. J. Exp. Med. 207, 1701–1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Turner M. S., Kane L. P., Morel P. A. (2009) Dominant role of antigen dose in CD4+Foxp3+ regulatory T cell induction and expansion. J. Immunol. 183, 4895–4903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sauer S., Bruno L., Hertweck A., Finlay D., Leleu M., Spivakov M., Knight Z. A., Cobb B. S., Cantrell D., O’Connor E., Shokat K. M., Fisher A. G., Merkenschlager M. (2008) T Cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc. Natl. Acad. Sci. USA 105, 7797–7802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Park J. H., Adoro S., Lucas P. J., Sarafova S. D., Alag A. S., Doan L. L., Erman B., Liu X., Ellmeier W., Bosselut R., Feigenbaum L., Singer A. (2007) ‘Coreceptor tuning’: cytokine signals transcriptionally tailor CD8 coreceptor expression to the self-specificity of the TCR. Nat. Immunol. 8, 1049–1059. [DOI] [PubMed] [Google Scholar]
  • 39.Huang W., Huang F., Kannan A. K., Hu J., August A. (2014) ITK tunes IL-4-induced development of innate memory CD8+ T cells in a γδ T and invariant NKT cell-independent manner. J. Leukoc. Biol. 96, 55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sakaguchi S., Yamaguchi T., Nomura T., Ono M. (2008) Regulatory T cells and immune tolerance. Cell 133, 775–787. [DOI] [PubMed] [Google Scholar]
  • 41.Josefowicz S. Z., Lu L. F., Rudensky A. Y. (2012) Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Belkaid Y., Tarbell K. (2009) Regulatory T cells in the control of host-microorganism interactions (*). Annu. Rev. Immunol. 27, 551–589. [DOI] [PubMed] [Google Scholar]
  • 43.Curotto de Lafaille M. A., Lafaille J. J. (2009) Natural and adaptive Foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 30, 626–635. [DOI] [PubMed] [Google Scholar]
  • 44.Tai X., Erman B., Alag A., Mu J., Kimura M., Katz G., Guinter T., McCaughtry T., Etzensperger R., Feigenbaum L., Singer D. S., Singer A. (2013) Foxp3 transcription factor is proapoptotic and lethal to developing regulatory T cells unless counterbalanced by cytokine survival signals. Immunity 38, 1116–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Burchill M. A., Yang J., Vogtenhuber C., Blazar B. R., Farrar M. A. (2007) IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J. Immunol. 178, 280–290. [DOI] [PubMed] [Google Scholar]
  • 46.Yao Z., Kanno Y., Kerenyi M., Stephens G., Durant L., Watford W. T., Laurence A., Robinson G. W., Shevach E. M., Moriggl R., Hennighausen L., Wu C., O’Shea J. J. (2007) Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109, 4368–4375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zorn E., Nelson E. A., Mohseni M., Porcheray F., Kim H., Litsa D., Bellucci R., Raderschall E., Canning C., Soiffer R. J., Frank D. A., Ritz J. (2006) IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 108, 1571–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Burchill M. A., Yang J., Vang K. B., Moon J. J., Chu H. H., Lio C. W., Vegoe A. L., Hsieh C. S., Jenkins M. K., Farrar M. A. (2008) Linked T cell receptor and cytokine signaling govern the development of the regulatory T cell repertoire. Immunity 28, 112–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fu S., Zhang N., Yopp A. C., Chen D., Mao M., Chen D., Zhang H., Ding Y., Bromberg J. S. (2004) TGF-beta induces Foxp3 + T-regulatory cells from CD4 + CD25 − precursors. Am. J. Transplant. 4, 1614–1627. [DOI] [PubMed] [Google Scholar]
  • 50.Tone Y., Furuuchi K., Kojima Y., Tykocinski M. L., Greene M. I., Tone M. (2008) Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat. Immunol. 9, 194–202. [DOI] [PubMed] [Google Scholar]
  • 51.Davidson T. S., DiPaolo R. J., Andersson J., Shevach E. M. (2007) Cutting edge: IL-2 is essential for TGF-beta-mediated induction of Foxp3+ T regulatory cells. J. Immunol. 178, 4022–4026. [DOI] [PubMed] [Google Scholar]
  • 52.Zheng S. G., Gray J. D., Ohtsuka K., Yamagiwa S., Horwitz D. A. (2002) Generation ex vivo of TGF-beta-producing regulatory T cells from CD4+CD25− precursors. J. Immunol. 169, 4183–4189. [DOI] [PubMed] [Google Scholar]
  • 53.Chen W., Jin W., Hardegen N., Lei K. J., Li L., Marinos N., McGrady G., Wahl S. M. (2003) Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 198, 1875–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lerman M. A., Larkin J. III, Cozzo C., Jordan M. S., Caton A. J. (2004) CD4+ CD25+ regulatory T cell repertoire formation in response to varying expression of a neo-self-antigen. J. Immunol. 173, 236–244. [DOI] [PubMed] [Google Scholar]
  • 55.Cozzo Picca C., Simons D. M., Oh S., Aitken M., Perng O. A., Mergenthaler C., Kropf E., Erikson J., Caton A. J. (2011) CD4⁺CD25⁺Foxp3⁺ regulatory T cell formation requires more specific recognition of a self-peptide than thymocyte deletion. Proc. Natl. Acad. Sci. USA 108, 14890–14895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hsieh C. S., Liang Y., Tyznik A. J., Self S. G., Liggitt D., Rudensky A. Y. (2004) Recognition of the peripheral self by naturally arising CD25+ CD4+ T cell receptors. Immunity 21, 267–277. [DOI] [PubMed] [Google Scholar]
  • 57.Moran A. E., Holzapfel K. L., Xing Y., Cunningham N. R., Maltzman J. S., Punt J., Hogquist K. A. (2011) T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J. Exp. Med. 208, 1279–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ohkura N., Hamaguchi M., Morikawa H., Sugimura K., Tanaka A., Ito Y., Osaki M., Tanaka Y., Yamashita R., Nakano N., Huehn J., Fehling H. J., Sparwasser T., Nakai K., Sakaguchi S. (2012) T Cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 37, 785–799. [DOI] [PubMed] [Google Scholar]
  • 59.Morikawa H., Ohkura N., Vandenbon A., Itoh M., Nagao-Sato S., Kawaji H., Lassmann T., Carninci P., Hayashizaki Y., Forrest A. R., Standley D. M., Date H., Sakaguchi S.; FANTOM Consortium (2014) Differential roles of epigenetic changes and Foxp3 expression in regulatory T cell-specific transcriptional regulation. Proc. Natl. Acad. Sci. USA 111, 5289–5294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kretschmer K., Apostolou I., Hawiger D., Khazaie K., Nussenzweig M. C., von Boehmer H. (2005) Inducing and expanding regulatory T cell populations by foreign antigen. Nat. Immunol. 6, 1219–1227. [DOI] [PubMed] [Google Scholar]
  • 61.Zou T., Satake A., Corbo-Rodgers E., Schmidt A. M., Farrar M. A., Maltzman J. S., Kambayashi T. (2012) Cutting edge: IL-2 signals determine the degree of TCR signaling necessary to support regulatory T cell proliferation in vivo. J. Immunol. 189, 28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tischner D., Wiegers G. J., Fiegl H., Drach M., Villunger A. (2012) Mutual antagonism of TGF-beta and interleukin-2 in cell survival and lineage commitment of induced regulatory T cells. Cell Death Differ. 19, 1277–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Harada Y., Harada Y., Elly C., Ying G., Paik J. H., DePinho R. A., Liu Y. C. (2010) Transcription factors Foxo3a and Foxo1 couple the E3 ligase Cbl-b to the induction of Foxp3 expression in induced regulatory T cells. J. Exp. Med. 207, 1381–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Marson A., Kretschmer K., Frampton G. M., Jacobsen E. S., Polansky J. K., MacIsaac K. D., Levine S. S., Fraenkel E., von Boehmer H., Young R. A. (2007) Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature 445, 931–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mahmud S. A., Manlove L. S., Schmitz H. M., Xing Y., Wang Y., Owen D. L., Schenkel J. M., Boomer J. S., Green J. M., Yagita H., Chi H., Hogquist K. A., Farrar M. A. (2014) Costimulation via the tumor-necrosis factor receptor superfamily couples TCR signal strength to the thymic differentiation of regulatory T cells. Nat. Immunol. 15, 473–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Caton A. J., Kropf E., Simons D. M., Aitken M., Weissler K. A., Jordan M. S. (2014) Strength of TCR signal from self-peptide modulates autoreactive thymocyte deletion and Foxp3(+) Treg-cell formation. Eur. J. Immunol. 44, 785–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hwang S., Song K. D., Lesourne R., Lee J., Pinkhasov J., Li L., El-Khoury D., Love P. E. (2012) Reduced TCR signaling potential impairs negative selection but does not result in autoimmune disease. J. Exp. Med. 209, 1781–1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Huang W., Jeong A. R., Kannan A. K., Huang L., August A. (2014) IL-2-inducible T cell kinase tunes T regulatory cell development and is required for suppressive function. J. Immunol. 193, 2267–2272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bensinger S. J., Walsh P. T., Zhang J., Carroll M., Parsons R., Rathmell J. C., Thompson C. B., Burchill M. A., Farrar M. A., Turka L. A. (2004) Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J. Immunol. 172, 5287–5296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Schluns K. S., Kieper W. C., Jameson S. C., Lefrançois L. (2000) Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat. Immunol. 1, 426–432. [DOI] [PubMed] [Google Scholar]
  • 71.Sprent J., Surh C. D. (2011) Normal T cell homeostasis: the conversion of naive cells into memory-phenotype cells. Nat. Immunol. 12, 478–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fry T. J., Mackall C. L. (2001) Interleukin-7: master regulator of peripheral T-cell homeostasis? Trends Immunol. 22, 564–571. [DOI] [PubMed] [Google Scholar]
  • 73.Khaled A. R., Durum S. K. (2002) The role of cytokines in lymphocyte homeostasis. Biotechniques 33, (Suppl), S40–S45. [PubMed] [Google Scholar]
  • 74.Tan J. T., Dudl E., LeRoy E., Murray R., Sprent J., Weinberg K. I., Surh C. D. (2001) IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc. Natl. Acad. Sci. USA 98, 8732–8737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mazzucchelli R., Durum S. K. (2007) Interleukin-7 receptor expression: intelligent design. Nat. Rev. Immunol. 7, 144–154. [DOI] [PubMed] [Google Scholar]
  • 76.Park J. H., Yu Q., Erman B., Appelbaum J. S., Montoya-Durango D., Grimes H. L., Singer A. (2004) Suppression of IL7Ralpha transcription by IL-7 and other prosurvival cytokines: a novel mechanism for maximizing IL-7-dependent T cell survival. Immunity 21, 289–302. [DOI] [PubMed] [Google Scholar]
  • 77.Kimura M. Y., Pobezinsky L. A., Guinter T. I., Thomas J., Adams A., Park J. H., Tai X., Singer A. (2013) IL-7 signaling must be intermittent, not continuous, during CD8⁺ T cell homeostasis to promote cell survival instead of cell death. Nat. Immunol. 14, 143–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Takada K., Jameson S. C. (2009) Self-class I MHC molecules support survival of naive CD8 T cells, but depress their functional sensitivity through regulation of CD8 expression levels. J. Exp. Med. 206, 2253–2269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Liao X. C., Littman D. R. (1995) Altered T cell receptor signaling and disrupted T cell development in mice lacking Itk. Immunity 3, 757–769. [DOI] [PubMed] [Google Scholar]
  • 80.Schaeffer E. M., Broussard C., Debnath J., Anderson S., McVicar D. W., Schwartzberg P. L. (2000) Tec family kinases modulate thresholds for thymocyte development and selection. J. Exp. Med. 192, 987–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lucas J. A., Atherly L. O., Berg L. J. (2002) The absence of Itk inhibits positive selection without changing lineage commitment. J. Immunol. 168, 6142–6151. [DOI] [PubMed] [Google Scholar]
  • 82.Atherly L. O., Brehm M. A., Welsh R. M., Berg L. J. (2006) Tec kinases Itk and Rlk are required for CD8+ T cell responses to virus infection independent of their role in CD4+ T cell help. J. Immunol. 176, 1571–1581. [DOI] [PubMed] [Google Scholar]
  • 83.Dubois S., Waldmann T. A., Müller J. R. (2006) ITK and IL-15 support two distinct subsets of CD8+ T cells. Proc. Natl. Acad. Sci. USA 103, 12075–12080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Broussard C., Fleischacker C., Horai R., Chetana M., Venegas A. M., Sharp L. L., Hedrick S. M., Fowlkes B. J., Schwartzberg P. L. (2006) Altered development of CD8+ T cell lineages in mice deficient for the Tec kinases Itk and Rlk. Immunity 25, 93–104. [DOI] [PubMed] [Google Scholar]
  • 85.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Huang W., Hu J., August A. (2013) Cutting edge: innate memory CD8+ T cells are distinct from homeostatic expanded CD8+ T cells and rapidly respond to primary antigenic stimuli. J. Immunol. 190, 2490–2494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Huang W., Qi Q., Hu J., Huang F., Laufer T. M., August A. (2014) Dendritic cell-MHC class II and Itk regulate functional development of regulatory innate memory CD4+ T cells in bone marrow transplantation. J. Immunol. 192, 3435–3441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Weinreich M. A., Odumade O. A., Jameson S. C., Hogquist K. A. (2010) T Cells expressing the transcription factor PLZF regulate the development of memory-like CD8+ T cells. Nat. Immunol. 11, 709–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lai D., Zhu J., Wang T., Hu-Li J., Terabe M., Berzofsky J. A., Clayberger C., Krensky A. M. (2011) KLF13 sustains thymic memory-like CD8(+) T cells in BALB/c mice by regulating IL-4-generating invariant natural killer T cells. J. Exp. Med. 208, 1093–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Felices M., Berg L. J. (2008) The Tec kinases Itk and Rlk regulate NKT cell maturation, cytokine production, and survival. J. Immunol. 180, 3007–3018. [DOI] [PubMed] [Google Scholar]
  • 91.Gadue P., Stein P. L. (2002) NK T cell precursors exhibit differential cytokine regulation and require Itk for efficient maturation. J. Immunol. 169, 2397–2406. [DOI] [PubMed] [Google Scholar]
  • 92.Au-Yeung B. B., Fowell D. J. (2007) A key role for Itk in both IFN gamma and IL-4 production by NKT cells. J. Immunol. 179, 111–119. [DOI] [PubMed] [Google Scholar]
  • 93.Qi Q., Xia M., Hu J., Hicks E., Iyer A., Xiong N., August A. (2009) Enhanced development of CD4+ gammadelta T cells in the absence of Itk results in elevated IgE production. Blood 114, 564–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Felices M., Yin C. C., Kosaka Y., Kang J., Berg L. J. (2009) Tec kinase Itk in gammadeltaT cells is pivotal for controlling IgE production in vivo. Proc. Natl. Acad. Sci. USA 106, 8308–8313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Prince A. L., Watkin L. B., Yin C. C., Selin L. K., Kang J., Schwartzberg P. L., Berg L. J. (2014) Innate PLZF+CD4+ αβ T cells develop and expand in the absence of Itk. J. Immunol. 193, 673–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Carty, S. A., Koretzky, G. A., Jordan, M. S. (2012) The role of IL-4 signaling in CD8+ innate-like lymphocyte development. 54th American Society of Hematology Annual Meeting and Exposition (abs.). [Google Scholar]
  • 97.Carty S. A., Koretzky G. A., Jordan M. S. (2014) Interleukin-4 regulates eomesodermin in CD8+ T cell development and differentiation. PLoS One 9, e106659 .doi:10.1371/journal.pone.0106659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.So E. Y., Oh J., Jang J. Y., Kim J. H., Lee C. E. (2007) Ras/Erk pathway positively regulates Jak1/STAT6 activity and IL-4 gene expression in Jurkat T cells. Mol. Immunol. 44, 3416–3426. [DOI] [PubMed] [Google Scholar]
  • 99.Oliver J. A., Stolberg V. R., Chensue S. W., King P. D. (2012) IL-4 acts as a potent stimulator of IFN-γ expression in CD8+ T cells through STAT6-dependent and independent induction of Eomesodermin and T-bet. Cytokine 57, 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang X., Proud C. G. (2006) The mTOR pathway in the control of protein synthesis. Physiology (Bethesda) 21, 362–369. [DOI] [PubMed] [Google Scholar]
  • 101.Zhu J., Huang H., Guo L., Stonehouse T., Watson C. J., Hu-Li J., Paul W. E. (2000) Transient inhibition of interleukin 4 signaling by T cell receptor ligation. J. Exp. Med. 192, 1125–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Au-Yeung B. B., Katzman S. D., Fowell D. J. (2006) Cutting edge: Itk-dependent signals required for CD4+ T cells to exert, but not gain, Th2 effector function. J. Immunol. 176, 3895–3899. [DOI] [PubMed] [Google Scholar]
  • 103.Kosaka Y., Felices M., Berg L. J. (2006) Itk and Th2 responses: action but no reaction. Trends Immunol. 27, 453–460. [DOI] [PubMed] [Google Scholar]
  • 104.O’Shea J. J., Murray P. J. (2008) Cytokine signaling modules in inflammatory responses. Immunity 28, 477–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.O’Shea J. J., Gadina M., Kanno Y. (2011) Cytokine signaling: birth of a pathway. J. Immunol. 187, 5475–5478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Stark G. R., Darnell J. E. Jr., (2012) The JAK-STAT pathway at twenty. Immunity 36, 503–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Deshpande R. V., Goust J. M., Chakrabarti A. K., Barbosa E., Hogan E. L., Banik N. L. (1995) Calpain expression in lymphoid cells. Increased mRNA and protein levels after cell activation. J. Biol. Chem. 270, 2497–2505. [DOI] [PubMed] [Google Scholar]
  • 108.Noguchi M., Sarin A., Aman M. J., Nakajima H., Shores E. W., Henkart P. A., Leonard W. J. (1997) Functional cleavage of the common cytokine receptor gamma chain (gammac) by calpain. Proc. Natl. Acad. Sci. USA 94, 11534–11539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Letavernier E., Dansou B., Lochner M., Perez J., Bellocq A., Lindenmeyer M. T., Cohen C. D., Haymann J. P., Eberl G., Baud L. (2011) Critical role of the calpain/calpastatin balance in acute allograft rejection. Eur. J. Immunol. 41, 473–484. [DOI] [PubMed] [Google Scholar]
  • 110.Trager N., Smith A., Wallace Iv G., Azuma M., Inoue J., Beeson C., Haque A., Banik N. L. (2014) Effects of a novel orally administered calpain inhibitor SNJ-1945 on immunomodulation and neurodegeneration in a murine model of multiple sclerosis. J. Neurochem. 130, 268–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hong C., Luckey M. A., Ligons D. L., Waickman A. T., Park J. Y., Kim G. Y., Keller H. R., Etzensperger R., Tai X., Lazarevic V., Feigenbaum L., Catalfamo M., Walsh S. T., Park J. H. (2014) Activated T cells secrete an alternatively spliced form of common γ-chain that inhibits cytokine signaling and exacerbates inflammation. Immunity 40, 910–923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ip J. Y., Tong A., Pan Q., Topp J. D., Blencowe B. J., Lynch K. W. (2007) Global analysis of alternative splicing during T-cell activation. RNA 13, 563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lynch K. W., Weiss A. (2000) A model system for activation-induced alternative splicing of CD45 pre-mRNA in T cells implicates protein kinase C and Ras. Mol. Cell. Biol. 20, 70–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wormald S., Hilton D. J. (2004) Inhibitors of cytokine signal transduction. J. Biol. Chem. 279, 821–824. [DOI] [PubMed] [Google Scholar]
  • 115.Yoshimura A. (1998) The CIS family: negative regulators of JAK-STAT signaling. Cytokine Growth Factor Rev. 9, 197–204. [DOI] [PubMed] [Google Scholar]
  • 116.Yoshimura A. (1998) The CIS/JAB family: novel negative regulators of JAK signaling pathways. Leukemia 12, 1851–1857. [DOI] [PubMed] [Google Scholar]
  • 117.Rawlings J. S., Rosler K. M., Harrison D. A. (2004) The JAK/STAT signaling pathway. J. Cell Sci. 117, 1281–1283. [DOI] [PubMed] [Google Scholar]
  • 118.Wu L., Bijian K., Shen S. H. (2009) CD45 recruits adapter protein DOK-1 and negatively regulates JAK-STAT signaling in hematopoietic cells. Mol. Immunol. 46, 2167–2177. [DOI] [PubMed] [Google Scholar]
  • 119.Dong S., Corre B., Foulon E., Dufour E., Veillette A., Acuto O., Michel F. (2006) T Cell receptor for antigen induces linker for activation of T cell-dependent activation of a negative signaling complex involving Dok-2, SHIP-1, and Grb-2. J. Exp. Med. 203, 2509–2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Inoue A., Yasuda T., Yamamoto T., Yamanashi Y. (2007) Dok-1 is a positive regulator of IL-4 signalling and IgE response. J. Biochem. 142, 257–263. [DOI] [PubMed] [Google Scholar]
  • 121.Lee C. M., Jung I. D., Noh K. T., Lee J. S., Park J. W., Heo D. R., Park J. H., Chang J. H., Choi I. W., Kim J. S., Shin Y. K., Park S. J., Han M. K., Lee C. G., Cho W. K., Park Y. M. (2012) An essential regulatory role of downstream of kinase-1 in the ovalbumin-induced murine model of asthma. PLoS ONE 7, e34554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kosugi A., Sakakura J., Yasuda K., Ogata M., Hamaoka T. (2001) Involvement of SHP-1 tyrosine phosphatase in TCR-mediated signaling pathways in lipid rafts. Immunity 14, 669–680. [DOI] [PubMed] [Google Scholar]
  • 123.Hebeisen M., Baitsch L., Presotto D., Baumgaertner P., Romero P., Michielin O., Speiser D. E., Rufer N. (2013) SHP-1 phosphatase activity counteracts increased T cell receptor affinity. J. Clin. Invest. 123, 1044–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Ramanathan S., Gagnon J., Leblanc C., Rottapel R., Ilangumaran S. (2006) Suppressor of cytokine signaling 1 stringently regulates distinct functions of IL-7 and IL-15 in vivo during T lymphocyte development and homeostasis. J. Immunol. 176, 4029–4041. [DOI] [PubMed] [Google Scholar]
  • 125.Banerjee A., Banks A. S., Nawijn M. C., Chen X. P., Rothman P. B. (2002) Cutting edge: suppressor of cytokine signaling 3 inhibits activation of NFATp. J. Immunol. 168, 4277–4281. [DOI] [PubMed] [Google Scholar]
  • 126.Li S., Chen S., Xu X., Sundstedt A., Paulsson K. M., Anderson P., Karlsson S., Sjögren H. O., Wang P. (2000) Cytokine-induced Src homology 2 protein (CIS) promotes T cell receptor-mediated proliferation and prolongs survival of activated T cells. J. Exp. Med. 191, 985–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Wong K. A., Kim R., Christofk H., Gao J., Lawson G., Wu H. (2004) Protein inhibitor of activated STAT Y (PIASy) and a splice variant lacking exon 6 enhance sumoylation but are not essential for embryogenesis and adult life. Mol. Cell. Biol. 24, 5577–5586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Roth W., Sustmann C., Kieslinger M., Gilmozzi A., Irmer D., Kremmer E., Turck C., Grosschedl R. (2004) PIASy-deficient mice display modest defects in IFN and Wnt signaling. J. Immunol. 173, 6189–6199. [DOI] [PubMed] [Google Scholar]
  • 129.Palvimo J. J. (2007) PIAS proteins as regulators of small ubiquitin-related modifier (SUMO) modifications and transcription. Biochem. Soc. Trans. 35, 1405–1408. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Leukocyte Biology are provided here courtesy of The Society for Leukocyte Biology

RESOURCES