Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 10.
Published in final edited form as: Clin Cancer Res. 2010 Aug 5;16(19):4695–4701. doi: 10.1158/1078-0432.CCR-10-0356

Wnt/β-catenin signaling in T cell immunity and cancer immunotherapy

Luca Gattinoni 1, Yun Ji 1, Nicholas P Restifo 1
PMCID: PMC3393131  NIHMSID: NIHMS224016  PMID: 20688898

Abstract

Wnt ligands are lipid-modified secreted glycoproteins that regulate embryonic development, cell fate specification and the homeostasis of self-renewing adult tissues. In addition to its well established role in thymocyte development, recent studies have indicated that Wnt/β-catenin signaling is critical for the differentiation, polarization and survival of mature T lymphocytes. Here, we describe our current understanding of Wnt signaling in the biology of post-thymic T cells and discuss how harnessing the Wnt/β-catenin pathway might improve the efficacy of vaccines, T-cell based therapies and allogeneic stem cell transplantation for the treatment of patients with cancer.

Background

Wnt ligands are secreted lipid-modified glycoproteins that are essential in diverse cellular processes including stem cell maintenance, cell fate decision, cell proliferation, survival, migration, and polarity determination. These manifold functions are accomplished by a large number of possible ligand-receptor combinations between 19 Wnt proteins, 10 Frizzled (Fzd) receptors, two co-receptors, and non-Fzd receptors such as the Receptor Tyrosine Kinase-like Orphan Receptor and the Ryk Receptor-like Tyrosine Kinase (1). These ligand–receptor interactions trigger a diversity of downstream signaling pathways including the “canonical” Wnt/β-catenin pathway, the Wnt/Planar cell polarity pathway, and the Wnt/Ca2+ pathway (1).

Here, we will focus our discussion on the role of the Wnt/β-catenin pathway in T cell immunity and cancer immunotherapy. Following a detailed review of the specifics of this signaling pathway, we will describe its role in the differentiation, polarization, and memory formation of post-thymic T lymphocytes. We will conclude by envisioning how the Wnt/β-catenin pathway might be harnessed to develop potent new immunotherapeutic approaches for the treatment of patients with cancer.

Molecular aspects of the Wnt/β-catenin signaling pathway

In the absence of Wnt signal, the cytoplasmic levels of β-catenin are regulated by the “destruction complex”, which consists of the scaffold proteins, Adenomatosis Polyposis Coli (Apc) and Axin, and the serine/threonine kinases, Glycogen-Synthase Kinase 3β (Gsk-3β) and Casein Kinase 1 (Ck1) (Fig.1). After binding Axin and Apc, β-catenin is phosphorylated by Ck1 at S45 and sequentially by Gsk-3β at T41/ S37/S33 (Fig.1). Phosphorylated β-catenin is further polyubiquitinated by β-transducin-repeat-containing protein and Wilms Tumor Suppressor protein complex and targeted for proteasome-mediated degradation (Fig.1) (2-5).

Fig.1. Wnt/β-catenin signaling pathway.

Fig.1

a) In the absence of Wnt signals, the destruction complex Adenomatous Polyposis Coli (Apc)–Axin 1–Glycogen synthase kinase-3β (Gsk-3β)–Casein Kinase 1 (Ck1) phosphorylates cytoplasmic β-catenin. β-transducin-repeat-containing protein (β-Trcp) and Wilms Tumor Suppressor protein (Wtx) complex polyubiquitinate phosphorylated β-catenin, targeting it for proteasome-mediated degradation. In the nucleus, DNA-binding proteins of the T cell factor (Tcf) and lymphoid enhancer-binding factor 1 (Lef-1) family are bound by transcriptional repressors such as the transducin-like enhancer proteins (Tle) and C-terminal Binding Protein (CtBP). Special AT-rich Binding Protein 1 (Satb1) is acetylated and predominantly functions as repressor of target genes. b) Upon Wnt binding to its Frizzled (Fzd) receptor and Lipoprotein Receptor-related Protein 5 or 6 (Lpr5/6) co-receptor, Disheveled (Dvl) is recruited to the Wnt/receptor complex to promote phosporylation of the Lrp5/6 co-receptor by Ck1 and Gsk-3β, generating a high-affinity binding site for Axin and subsequent disruption of the destruction complex. β-catenin accumulates and translocates into the nucleus where it binds to diverse DNA-binding partners to regulate gene transcription. B-cell CLL/Lymphoma 9 (Bcl9), Pygorus (Pygo), CREB Binding Protein (Cbp), E1A Binding Protein p300 (Ep300), PROP paired-like homeobox 1 (Prop1), Histone deacetylase (Hdac).

Upon Wnt binding to Fzd and the Low Density Lipoprotein Receptor-related Protein 5 or 6 (Lrp5/6) co-receptor, Disheveled is recruited to the Wnt/receptor complex to promote phosporylation of the Lrp5/6 co-receptor by Ck1 and Gsk-3β (Fig.1) (6). Phosphorylated Lrp5/6 in turn attracts Axin to the intracellular domains of Wnt/receptor complex resulting in the disruption of the “destruction complex” and accumulation and nuclear translocation of β-catenin (Fig.1) (6).

In the nucleus, β-catenin interacts with diverse DNA binding partners to remodel chromatin and orchestrate transcriptional programs (Fig.1). The best characterized β-catenin–binding partners are transcription factors of the T Cell Factor (Tcf)/Lymphocyte-Enhancer-Binding Factor (Lef) family, including Tcf1 (encoded by Tcf7), Lef1, Tcf3, and Tcf4 (7). In the absence of β-catenin, Tcf/Lef inhibit gene expression by interacting with repressors such as Transducin-Like Enhancer proteins (Tle) and C-terminal Binding Protein (CtBP), which mediate histone deacetylation and chromatin compaction (Fig.1) (8-10). β-catenin replaces Tle to interact with Tcf/Lef and recruits several co-activators including B-cell CLL/Lymphoma 9, Pygorus and the histone acetyltransferases CREB Binding Protein and E1A Binding Protein p300 (Ep300) to promote specific gene expression (Fig.1) (11, 12). More recently, PROP paired-like homeobox 1 (Prop1) (13) and the Special AT-rich Binding Protein 1 (Satb1) (14), have been identified as DNA binding transcription factors that recruit β-catenin to mediate Wnt signaling (Fig.1). Prop1–β-catenin signaling has been implicated in the regulation of pituary gland development by simultaneously promoting the transcription of the cell-lineage-determining factor Pou Class 1 Homeobox 1, and suppressing the expression of the transcriptional repressor Hesx Homeobox 1 via Tle/Reptin/Histone deacetylase 1 corepressor complexes (13). Satb1 is a chromatin organizer enriched in the T cell lineage that is critical for the expression of a large number of genes involved in T cell proliferation and development (14, 15). Satb1 acts predominantly as a repressor on most of its target genes, however, upon Wnt/β-catenin signaling it undergoes deacetylation, which facilitates its association with β-catenin and conversion into a transcriptional activator (14). Thus, at least three sets of DNA binding proteins – Tcf/Lef, Prop1 and Satb1– have been found to interact with β–catenin to regulate gene transcription.

The Wnt/β–catenin pathway is subjected to extensive regulation at various levels of the signaling cascade. Wnt–Fzds–Lrp5/6 interactions are modulated by the Secreted Frizzled-Related Protein, Dickkopf (Dkk) and Wise/Sclerostin family members as well as Wnt inhibitory factor 1, and Apc Down-Regulated 1 (6, 16). Nuclear Chibby Homolog 1 and Inhibitor of β-catenin and Tcf (ICAT) inhibit the interaction of β–catenin with Tcf/Lef, repressing β-catenin–Tcf /Lef-mediated transactivation (17-19). β-catenin–Tcf/Lef binding can also be inhibited by naturally occurring Tcf and Lef short isoforms (25-40 kD), which lack the N-terminal β-catenin binding domain and act as dominant negatives (20, 21). In addition, post-translational modification such as phosphorylation, acetylation, sumoylation, and ubiquitination affect the binding affinity of Tcf/Lef to β-catenin or Tle, thus further regulating either transcriptional activation or repression (20). Finally, expression of Wnt signaling components such as Fzds, Lrp6, Axin2, Tcf, Lef, and Dkk1 is also regulated by β-catenin–Tcf in either positive or negative feed-back loops (1, 6). The high degree of complexity and the level of regulation involved in the Wnt/β-catenin signaling pathway reflect its centrality to cellular processes.

Wnt/β-catenin signaling in T cell immunity

Several studies using gain- and loss-of-function approaches indicate that Wnt/β-catenin signaling is a key regulator of T cell development at various stages of thymocyte differentiation (reviewed elsewhere by Staal et al. (22) and Yu et al. (23)). In vivo studies using either transgenesis (24) or recombinant retroviruses (25) to complement Tcf1 deficiency, perhaps provide the clearest evidence that Wnt/β-catenin signaling is critical for T cell development. Mice deficient in Tcf1 have impaired thymocyte maturation with defects at double negative and immature single positive stages of T cell development (26, 27). This defect could be rescued by the Tcf1 p45 isoform but not by the p33 isoform, which lacks the N-terminal β-catenin-binding domain (24, 25). Although the role of Wnt/β-catenin signaling in T cell development has been extensively scrutinized in the past, its function in the biology of mature T lymphocytes has just begun to be explored.

Modulation of CD8+ T cell differentiation and memory formation

Early studies investigating a possible role of Wnt/β-catenin signaling in mature CD8+ T cells revealed that Tcf1-deficient CD8+ T cells had no defects in proliferative responses to mitogens or allogeneic antigens and could acquire cytotoxic effector functions similar to wild type controls (27). These findings led to the surprising notion that Tcf1, which in adult mammals is exclusively expressed by T cells (Ref. 28 and Supplementary Fig. 1), might not have a role in post-thymic CD8+ T lymphocyte function. However, new findings clearly indicate that Wnt/β-catenin signaling is functionally important in mature T cells in vivo.

Tcf/Lef reporter systems (29) and gene expression analyses (30-32) indicate that Wnt signaling is functionally active and dynamically regulated in mature T cells in vivo. Tcf7 and Lef1 are highly expressed by naïve CD8+ T (TN) cells, but their levels rapidly drop following a productive encounter with antigen as they undergo massive expansion and differentiation into cytotoxic effector T cells (30-32). The minority of cells that persist as long-lived memory T cells after the effector phase expresses intermediate levels of these Wnt transcription factors (32), but heterogeneity occurs within the T cell memory compartment. High levels of Tcf7 and Lef1 expression are found in “central memory” T (TCM) cells (30, 31), which express the lymphoid-homing molecules CD62L and CCR7, have long teleomeres, high proliferative capacity, and possess the stem-like quality for plasticity and self-renewal (33-36). Conversely, low levels of Tcf7 and Lef1 are found in CD62Llow and CCR7low “effector memory” T (TEM) cells (30, 31), which have poor replicative potential and have acquired the capacity to produce high amounts of IFN-γ (33-36). The tightly regulated expression of Wnt signaling transducers indicate that this pathway is associated with the maintenance and function of less differentiated TN and TCM cells, but causality has only recently been tested.

To directly assess the impact of Wnt/β-catenin signaling on mature CD8+ T cell differentiation we activated TN in the presence of Wnt3A or inhibitors of Gsk-3β to stabilize β-catenin and mimic downstream events of the Wnt signaling cascade (31, 37). Activation of the Wnt/β-catenin pathway suppressed the antigen-induced expression of the master regulator of CD8+ T cell effector functions, Eomesoderin (38), inhibiting the acquisition of cytotoxic functions and the capacity to release IFN-γ. This arrested differentiation promoted the generation of TCM and a novel T cell memory population named CD8+ memory stem (TSCM) cells that possess enhanced self-renewal capability in serial transplant experiments, have the multipotent capacity to generate TCM, TEM and effector T cells, and a proliferative potential that exceeds other effector and memory T cell subsets (31, 39).

Our work provides the first evidence that enforced Wnt/β-catenin signaling can favor CD8+ T cell memory formation by suppressing their maturation into terminally differentiated effector T cells (Fig.2), but Jeannet et al. (40) have now shown that Wnt has similar effects in the physiologic setting. Consistent with previous findings (27), they found that Tcf1 deficiency did not impair the ability of CD8+ T cells to undergo effector differentiation and control lymphocytic choriomeningitis virus (LCMV) infection. Rather, effector differentiation was enhanced, as manifested by the increased numbers of T cells expressing the terminal differentiation and senescence marker Klrg1 at the peak of the immune response (40). This unrestrained effector differentiation prevented CD8+ T cells from forming TCM and severely impaired the ability of CD8+ T cells to mediate immune responses to a secondary LCMV infection (40). Regulation of CD8+ T cell differentiation and memory formation by Tcf1 was dependent on its binding to β-catenin because genetic complementation with the Tcf1 p45 but not the p33 isoform inhibited terminal effector differentiation and restored memory recall responses (40). Secondary responses were also depressed in CD8+ T cells lacking both β- and γ-catenin (40). Taken together, these findings indicate that the Wnt/β-catenin pathway is essential for CD8+ T cell memory formation under physiological conditions in vivo.

Fig.2. Effects of Wnt/β-catenin signaling on T cell differentiation, polarization and survival.

Fig.2

In CD8+ T cells, activation of Wnt/β-catenin signaling during T cell priming inhibits effector differentiation while promoting the generation of self-renewing memory T cells, stem cell memory (TSCM) and central memory (TCM) cells. In CD4+ T cells, Wnt/β-catenin signaling favors T helper (Th) 2 over Th1 polarization and enhances survival of naturally occurring regulatory T cells (nTreg). Effector memory T cells (TEM); Effector T cells (TEFF); Induced regulatory T cells (iTreg); T follicular helper cells (Tfh).

Zhao and colleagues came to the same conclusion that the Wnt/β-catenin pathway promotes CD8+ T cell memory formation using transgenic mice to constitutively activate Wnt signaling, rather than to ablate it (32). Echoing our findings, enforced expression of stabilized β-catenin and Tcf1 inhibited the expansion of antigen-specific CD8+ T cells in the effector phase of the immune response and enhanced CD8+ T cell memory formation and function in a variety of infection models, including Listeria monocytogenes, LCMV and vaccinia virus (32).

A number of interesting findings further support a role for Wnt/β-catenin signaling in CD8+ T cell differentiation and memory formation. Firstly, enforced expression of ICAT, which specifically disrupts β-catenin–Tcf interactions, impairs the survival of CD8+ T cells following in vitro activation (41). Secondly, increased CD8+ T memory formation was observed in mice after administration of metformin (42), an agonist of adenosine monophosphate kinase that can, among other substrates, phosphorylate and stabilize β-catenin, promoting downstream Tcf/Lef transcriptional activity (43). Finally, gene expression data reveals that high levels of Tcf7 and Lef1 were found in CD8+ T cells with increased potential to form memory in vivo such as in T cells lacking Blimp-1 (44) and IL-2Rα (45, 46), two key drivers of terminal effector differentiation. Tcf7 and Lef1 were also high in T cells programmed in the presence of IL-21, a cytokine that inhibits effector differentiation and enhances functional memory (47, 48).

Arrest of lymphocyte differentiation to maintain long-lived antigen-experienced T cells with stem cell-like properties was postulated by Fearon almost a decade ago as a way to continually generate effector T cells, but the molecular mechanisms regulating this process were poorly understood (35). Given the weight of the evidence described above, it is now clear that Wnt/β-catenin is a key signaling pathway governing arrested differentiation in CD8+ T cells. These findings have parallels in stem cell biology, where Wnt signaling has a pivotal role in promoting stem cell self-renewal while limiting proliferation and differentiation (22, 49). Many unanswered questions remains to be addressed: i.) What Wnt ligands if any are modulating the differentiation and memory formation of CD8+ T lymphocytes in vivo? ii.) What are the cellular sources and anatomical niches where Wnt ligands are provided to CD8+ T cells and when this signaling takes place relative to antigen priming? iii.) What are the molecular events orchestrated by Wnt/β-catenin signaling that restrain effector differentiation in order to enhance memory formation?

Regualtion of CD4+ T cell polarization and survival

Like their CD8+ T cell counterparts, mature CD4+ T cells can respond to Wnt signals (Fig.2) (14). Following antigen activation, naïve CD4+ T cells polarize into T helper (Th) cell populations secreting distinct sets of cytokines (50). Classically, CD4+ T cell polarization has been viewed as a result of diverse transcriptional programs triggered by the cytokine signals that T cells experience at the time of encounter with antigen (50). However, new findings indicate that Wnt proteins can also influence the lineage choice of naïve CD4+ T cells (Fig.2) (14). The predominance of data implicates an active role of Wnt/β-catenin signaling in the differentiation of Th2 cells (14, 51), which express IL-4 and IL-13 under the control of the master transcription factor Gata3 (52). Wnt-dependent accumulation of β-catenin has been observed in CD4+ T cells undergoing Th2 polarization (14). Blockade of Wnt/β-catenin signaling by the Wnt antagonist, Dkk1, or by siRNA-mediated silencing of β-catenin resulted in decreased expression of Gata3 and low levels of Th2 cytokine secretion (14). Conversely, enforced expression of stabilized β-catenin was found to enhance Gata3 transcription and IL-4 production (51). These results indicate that Wnt signaling critically regulates Th2 polarization by promoting Gata3 expression.

The mechanism whereby Wnt/β-catenin signaling modulates Gata3 transcription has been studied in considerable detail (14, 51). Notani et al. have shown that Wnt signaling enhances Th2 polarization by recruiting Satb1, β-catenin and the histone acetyltransferase Ep300 to the Gata3 promoter (14). Studies by Yu et al. (51) have instead focused on Tcf1-β-catenin interactions in the initiation of Th2 cell differentiation. Chromatin immunoprecipitation and luciferase reporter assays indicated that Tcf1 and β-catenin bind to the Tcf1-binding site upstream of Gata3 exon-1b to activate Gata3-1b transcription (51). Tcf1 deficiency diminished Gata3-1b expression and IL-4 production in CD4+ T cells and impaired in vivo Th2 responses in a mouse model of allergic asthma (51). Impairment in Gata3-1b expression and IL-4 production was also observed upon enforced expression of ICAT demonstrating a requirement for Tcf1 and β-catenin interaction for Th2 polarization (51). Interestingly, the authors found that Tcf1 overexpression also inhibited Th1 fate and the production of the paradigmatic type 1 cytokine IFN-γ, although in a β-catenin-independent fashion (51).

The role of β-catenin has also been studied in naturally occurring, CD4+CD25+ T regulatory (nTreg) cells. Enforced expression of β-catenin did not alter immunosuppressive functions in nTreg cells, but enhanced their survival (Fig.2), resulting in more effective protection against inflammatory bowel disease (53). In addition, stabilized β-catenin impaired the ability of CD4+CD25- T cells to produce IFN-β and mediate autoimmune colitis. The authors concluded that β-catenin stabilization caused T cell anergy, but in light of Yu and Notani's results, Wnt/β-catenin might have had simply skewed CD4+ T cell fate toward less aggressive Th2-type immune responses (14, 51, 54).

In summary, new studies indicate that the Wnt/β-catenin pathway is critical in controlling important aspects of CD4+ T cell biology, including T cell survival and lineage fate decisions. It is now clear that Wnt/β-catenin signaling can favor Th2 over Th1 polarization and can enhance the persistence of nTreg, but its impact on the function and differentiation of other CD4+ T cell subsets remains to be investigated.

Clinical-Translational Advances

Therapeutic strategies targeting Wnt/β-catenin have thus far focused on the suppression of the pathway because of its involvement in carcinogenesis (55, 56). However, the discovery that Wnt/β-catenin signaling is a key regulator of T cell immunity now raises the possibility that potentiating Wnt signaling could be used to improve cancer therapies through immune-based mechanisms.

The generation of a robust population of memory T cells is critical for effective vaccine and T cell-based therapies to prevent and treat cancer (34). Although only a minority of patients receiving current therapeutic cancer vaccines experience objective tumor regression, and virtually none are cured by existing vaccines (57), recent phase III randomized trials have shown benefits in overall or disease-free survival in patients with refractory prostate cancer (58), lymphomas (59) and melanoma (60). Some cancer vaccines might fail because they drive T cells to terminal differentiation and senescence rather than generating stem-like T memory populations (34) that have been shown to be effective in both viral (61) and tumor models (31, 62). Generation of these potent memory populations might be enhanced if vaccinations were performed in concert with the immunomodulatory influences of agonists of Wnt/β-catenin signaling. For example, inhibitors of Gsk-3β, which mimic Wnt signaling, have been shown to augment T cell memory formation in response to a self/tumor antigen (31). This strategy could be rapidly translated in the clinical setting because many Gsk-3β inhibitors are already approved for human use or are in clinical evaluation for other indications (63).

The use of agonists of Wnt/β-catenin signaling also has immediate implications for the improvement of immunotherapies based on ex vivo manipulation of lymphocytes prior to adoptive transfer to patients. Allogeneic stem cell transplantation can be highly effective for the treatment patients with refractory leukemias and lymphomas, but graft-versus-tumor (GVT) responses can come with the high price of graft-versus-host-disease (GVHD), which can be difficult to manage and even lethal (64). Treg and Th2 cells are being actively explored as a means of inhibiting GVHD while maintaining some anti-tumor effects (65) and the immunoregulatory effects of these CD4+ T cell subsets might be considerably potentiated by enhancing Wnt/β-catenin signaling with drugs or gene therapy. Our own focus has been to enhance immunotherapies based on the adoptive transfer of autologous tumor-specific CD8+ lymphocytes. Preclinical models have shown that Gsk-3β inhibitors enable the generation of stem-like memory CD8+ T cells, which can be highly effective in small numbers (31). This pharmacologic approach might be employed to efficiently prevent the detrimental terminal differentiation of anti-tumor T cell populations that can occur when using current methods for ex vivo expansion (66, 67). Finally, human CD8+ T cells with stem-like properties generated under the influence of Wnt signaling and genetically engineered to have tumor specificities (68, 69) might reduce the cost and complexity of the treatment and ultimately allow for the widespread application of adoptive immunotherapies.

The role of the Wnt/β-catenin pathway in the function of post-thymic T cells has just begun to be realized, but it is clear that the modulation of this molecular machinery can be exploited to enhance CD8+ T cell memory formation and alter CD4+ T cell fate decisions. A deeper understanding of the biology of Wnt/β-catenin signaling in mature T lymphocytes might create new strategies for therapeutic intervention not previously imagined.

Supplementary Material

1

Acknowledgements

This research was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research. The authors would like to thank C. Klebanoff, P. Muranski and M. Bachinski for critical review of this manuscript.

Footnotes

The authors have no conflicting financial interests.

Reference List

  • 1.van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205–14. doi: 10.1242/dev.033910. [DOI] [PubMed] [Google Scholar]
  • 2.Yanagawa S, Matsuda Y, Lee JS, et al. Casein kinase I phosphorylates the Armadillo protein and induces its degradation in Drosophila. EMBO J. 2002;21:1733–42. doi: 10.1093/emboj/21.7.1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Amit S, Hatzubai A, Birman Y, et al. Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt pathway. Genes Dev. 2002;16:1066–76. doi: 10.1101/gad.230302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Aberle H, Bauer A, Stappert J, Kispert A, Kemler R. beta-catenin is a target for the ubiquitin-proteasome pathway. EMBO J. 1997;16:3797–804. doi: 10.1093/emboj/16.13.3797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Major MB, Camp ND, Berndt JD, et al. Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science. 2007;316:1043–6. doi: 10.1126/science/1141515. [DOI] [PubMed] [Google Scholar]
  • 6.MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17:9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Graham TA, Weaver C, Mao F, Kimelman D, Xu W. Crystal structure of a beta-catenin/Tcf complex. Cell. 2000;103:885–96. doi: 10.1016/s0092-8674(00)00192-6. [DOI] [PubMed] [Google Scholar]
  • 8.Roose J, Molenaar M, Peterson J, et al. The Xenopus Wnt effector XTcf-3 interacts with Groucho-related transcriptional repressors. Nature. 1998;395:608–12. doi: 10.1038/26989. [DOI] [PubMed] [Google Scholar]
  • 9.Brannon M, Brown JD, Bates R, Kimelman D, Moon RT. XCtBP is a XTcf-3 co-repressor with roles throughout Xenopus development. Development. 1999;126:3159–70. doi: 10.1242/dev.126.14.3159. [DOI] [PubMed] [Google Scholar]
  • 10.Billin AN, Thirlwell H, Ayer DE. Beta-catenin-histone deacetylase interactions regulate the transition of LEF1 from a transcriptional repressor to an activator. Mol Cell Biol. 2000;20:6882–90. doi: 10.1128/mcb.20.18.6882-6890.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Townsley FM, Cliffe A, Bienz M. Pygopus and Legless target Armadillo/beta-catenin to the nucleus to enable its transcriptional co-activator function. Nat Cell Biol. 2004;6:626–33. doi: 10.1038/ncb1141. [DOI] [PubMed] [Google Scholar]
  • 12.Mosimann C, Hausmann G, Basler K. Beta-catenin hits chromatin: regulation of Wnt target gene activation. Nat Rev Mol Cell Biol. 2009;10:276–86. doi: 10.1038/nrm2654. [DOI] [PubMed] [Google Scholar]
  • 13.Olson LE, Tollkuhn J, Scafoglio C, et al. Homeodomain-mediated beta-catenin-dependent switching events dictate cell-lineage determination. Cell. 2006;125:593–605. doi: 10.1016/j.cell.2006.02.046. [DOI] [PubMed] [Google Scholar]
  • 14.Notani D, Gottimukkala KP, Jayani RS, et al. Global regulator SATB1 recruits beta-catenin and regulates T(H)2 differentiation in Wnt-dependent manner. PLoS Biol. 2010;8:e1000296. doi: 10.1371/journal.pbio.1000296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pavan KP, Purbey PK, Sinha CK, et al. Phosphorylation of SATB1, a global gene regulator, acts as a molecular switch regulating its transcriptional activity in vivo. Mol Cell. 2006;22:231–43. doi: 10.1016/j.molcel.2006.03.010. [DOI] [PubMed] [Google Scholar]
  • 16.Shimomura Y, Agalliu D, Vonica A, et al. APCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex. Nature. 2010;464:1043–7. doi: 10.1038/nature08875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Henderson BR, Fagotto F. The ins and outs of APC and beta-catenin nuclear transport. EMBO Rep. 2002;3:834–9. doi: 10.1093/embo-reports/kvf181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Takemaru K, Yamaguchi S, Lee YS, Zhang Y, Carthew RW, Moon RT. Chibby, a nuclear beta-catenin-associated antagonist of the Wnt/Wingless pathway. Nature. 2003;422:905–9. doi: 10.1038/nature01570. [DOI] [PubMed] [Google Scholar]
  • 19.Tago K, Nakamura T, Nishita M, et al. Inhibition of Wnt signaling by ICAT, a novel beta-catenin-interacting protein. Genes Dev. 2000;14:1741–9. [PMC free article] [PubMed] [Google Scholar]
  • 20.Arce L, Yokoyama NN, Waterman ML. Diversity of LEF/TCF action in development and disease. Oncogene. 2006;25:7492–504. doi: 10.1038/sj.onc.1210056. [DOI] [PubMed] [Google Scholar]
  • 21.Staal FJ, Clevers HC. WNT signalling and haematopoiesis: a WNT-WNT situation. Nat Rev Immunol. 2005;5:21–30. doi: 10.1038/nri1529. [DOI] [PubMed] [Google Scholar]
  • 22.Staal FJ, Luis TC, Tiemessen MM. WNT signalling in the immune system: WNT is spreading its wings. Nat Rev Immunol. 2008;8:581–93. doi: 10.1038/nri2360. [DOI] [PubMed] [Google Scholar]
  • 23.Yu Q, Sharma A, Sen JM. TCF1 and beta-catenin regulate T cell development and function. Immunol Res. 2010 doi: 10.1007/s12026-009-8137-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ioannidis V, Beermann F, Clevers H, Held W. The beta-catenin--TCF-1 pathway ensures CD4(+)CD8(+) thymocyte survival. Nat Immunol. 2001;2:691–7. doi: 10.1038/90623. [DOI] [PubMed] [Google Scholar]
  • 25.Staal FJ, Meeldijk J, Moerer P, et al. Wnt signaling is required for thymocyte development and activates Tcf-1 mediated transcription. Eur J Immunol. 2001;31:285–93. doi: 10.1002/1521-4141(200101)31:1<285::AID-IMMU285>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 26.Verbeek S, Izon D, Hofhuis F, et al. An HMG-box-containing T-cell factor required for thymocyte differentiation. Nature. 1995;374:70–4. doi: 10.1038/374070a0. [DOI] [PubMed] [Google Scholar]
  • 27.Schilham MW, Wilson A, Moerer P, Benaissa-Trouw BJ, Cumano A, Clevers HC. Critical involvement of Tcf-1 in expansion of thymocytes. J Immunol. 1998;161:3984–91. [PubMed] [Google Scholar]
  • 28.Castrop J, van WD, Koomans-Bitter M, et al. The human TCF-1 gene encodes a nuclear DNA-binding protein uniquely expressed in normal and neoplastic T-lineage lymphocytes. Blood. 1995;86:3050–9. [PubMed] [Google Scholar]
  • 29.Jeannet G, Scheller M, Scarpellino L, et al. Long-term, multilineage hematopoiesis occurs in the combined absence of beta-catenin and gamma-catenin. Blood. 2008;111:142–9. doi: 10.1182/blood-2007-07-102558. [DOI] [PubMed] [Google Scholar]
  • 30.Willinger T, Freeman T, Herbert M, Hasegawa H, McMichael AJ, Callan MF. Human naive CD8 T cells down-regulate expression of the WNT pathway transcription factors lymphoid enhancer binding factor 1 and transcription factor 7 (T cell factor-1) following antigen encounter in vitro and in vivo. J Immunol. 2006;176:1439–46. doi: 10.4049/jimmunol.176.3.1439. [DOI] [PubMed] [Google Scholar]
  • 31.Gattinoni L, Zhong XS, Palmer DC, et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med. 2009;15:808–13. doi: 10.1038/nm.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhao DM, Yu S, Zhou X, et al. Constitutive activation of Wnt signaling favors generation of memory CD8 T cells. J Immunol. 2010;184:1191–9. doi: 10.4049/jimmunol.0901199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–63. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
  • 34.Klebanoff CA, Gattinoni L, Restifo NP. CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev. 2006;211:214–24. doi: 10.1111/j.0105-2896.2006.00391.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fearon DT, Manders P, Wagner SD. Arrested differentiation, the self- renewing memory lymphocyte, and vaccination. Science. 2001;293:248–50. doi: 10.1126/science.1062589. [DOI] [PubMed] [Google Scholar]
  • 36.Stemberger C, Neuenhahn M, Gebhardt FE, Schiemann M, Buchholz VR, Busch DH. Stem cell-like plasticity of naive and distinct memory CD8+ T cell subsets. Semin Immunol. 2009;21:62–8. doi: 10.1016/j.smim.2009.02.004. [DOI] [PubMed] [Google Scholar]
  • 37.Gattinoni L, Ji Y, Restifo NP. beta-catenin does not regulate memory T cell phenotype Reply. Nat Med. 2010;16:514–5. doi: 10.1038/nm0510-513. [DOI] [PubMed] [Google Scholar]
  • 38.Pearce EL, Mullen AC, Martins GA, et al. Control of effector CD8+ T cell function by the transcription factor Eomesodermin. Science. 2003;302:1041–3. doi: 10.1126/science.1090148. [DOI] [PubMed] [Google Scholar]
  • 39.Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat Med. 2005;11:1299–305. doi: 10.1038/nm1326. [DOI] [PubMed] [Google Scholar]
  • 40.Jeannet G, Boudousquie C, Gardiol N, Kang J, Huelsken J, Held W. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory. Proc Natl Acad Sci U S A. 2010;107:9777–82. 1. doi: 10.1073/pnas.0914127107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hossain MZ, Yu Q, Xu M, Sen JM. ICAT expression disrupts beta-catenin-TCF interactions and impairs survival of thymocytes and activated mature T cells. Int Immunol. 2008;20:925–35. doi: 10.1093/intimm/dxn051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009;460:103–7. doi: 10.1038/nature08097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zhao J, Yue W, Zhu MJ, Sreejayan N, Du M. AMP-activated protein kinase (AMPK) cross-talks with canonical Wnt signaling via phosphorylation of beta-catenin at Ser 552. Biochem Biophys Res Commun. 2010;395:146–51. doi: 10.1016/j.bbrc.2010.03.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rutishauser RL, Martins GA, Kalachikov S, et al. Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties. Immunity. 2009;31:296–308. doi: 10.1016/j.immuni.2009.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kalia V, Sarkar S, Subramaniam S, Haining WN, Smith KA, Ahmed R. Prolonged interleukin-2Ralpha expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. Immunity. 2010;32:91–103. doi: 10.1016/j.immuni.2009.11.010. [DOI] [PubMed] [Google Scholar]
  • 46.Mitchell DM, Ravkov EV, Williams MA. Distinct Roles for IL-2 and IL-15 in the Differentiation and Survival of CD8+ Effector and Memory T Cells. J Immunol. 2010 doi: 10.4049/jimmunol.0904089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hinrichs CS, Spolski R, Paulos CM, et al. IL-2 and IL-21 confer opposing differentiation programs to CD8+ T cells for adoptive immunotherapy. Blood. 2008;111:5326–33. doi: 10.1182/blood-2007-09-113050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yi JS, Du M, Zajac AJ. A vital role for interleukin-21 in the control of a chronic viral infection. Science. 2009;324:1572–6. doi: 10.1126/science.1175194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fleming HE, Janzen V, Lo CC, et al. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell. 2008;2:274–83. doi: 10.1016/j.stem.2008.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.O'Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098–102. doi: 10.1126/science.1178334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yu Q, Sharma A, Oh SY, et al. T cell factor 1 initiates the T helper type 2 fate by inducing the transcription factor GATA-3 and repressing interferon-gamma. Nat Immunol. 2009;10:992–9. doi: 10.1038/ni.1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zheng WP, Flavell RA. The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell. 1997;89:587–96. doi: 10.1016/s0092-8674(00)80240-8. [DOI] [PubMed] [Google Scholar]
  • 53.Ding Y, Shen S, Lino AC, Curotto de Lafaille MA, Lafaille JJ. Beta-catenin stabilization extends regulatory T cell survival and induces anergy in nonregulatory T cells. Nat Med. 2008;14:162–9. doi: 10.1038/nm1707. [DOI] [PubMed] [Google Scholar]
  • 54.Jung U, Foley JE, Erdmann AA, Eckhaus MA, Fowler DH. CD3/CD28-costimulated T1 and T2 subsets: differential in vivo allosensitization generates distinct GVT and GVHD effects. Blood. 2003;102:3439–46. doi: 10.1182/blood-2002-12-3936. [DOI] [PubMed] [Google Scholar]
  • 55.Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 2006;5:997–1014. doi: 10.1038/nrd2154. [DOI] [PubMed] [Google Scholar]
  • 56.Takahashi-Yanaga F, Kahn M. Targeting Wnt Signaling: Can We Safely Eradicate Cancer Stem Cells? Clin Cancer Res. 2010 doi: 10.1158/1078-0432.CCR-09-2943. [DOI] [PubMed] [Google Scholar]
  • 57.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Schellhammer PF, Higano C, Berger ER. A randomized, double-blind, placebo-controlled, multi-center, phase III trial of Sipuleucel-T in men with metastatic, androgen independent prostatic adenocarcinoma.. Presented at the American Urological Association Annual Meeting.; Chicago, IL. April 25-30, 2009. [Google Scholar]
  • 59.Schuster SJ, Neelapu SS, Gause BL, et al. Idiotype vaccine therapy (BiovaxID) in follicular lymphoma in first complete remission: Phase III clinical trial results. Journal of Clinical Oncology. 2009;27:2. [Google Scholar]
  • 60.Schwartzentruber DJ, Lawson D, Richards J, et al. A phase III multi-institutional randomized study of immunization with the gp100: 209-217(210M) peptide followed by high-dose IL-2 compared with high-dose IL-2 alone in patients with metastatic melanoma. Journal of Clinical Oncology. 2009;27:CRA9011. [Google Scholar]
  • 61.Wherry EJ, Teichgraber V, Becker TC, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nature Immunology. 2003;4:225–34. doi: 10.1038/ni889. [DOI] [PubMed] [Google Scholar]
  • 62.Klebanoff CA, Gattinoni L, Torabi-Parizi P, et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci U S A. 2005;102:9571–6. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gattinoni L, Klebanoff CA, Restifo NP. Pharmacologic induction of CD8+ T cell memory: better living through chemistry. Sci Transl Med. 2009;1:11ps12. doi: 10.1126/scitranslmed.3000302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–83. doi: 10.1182/blood-2008-03-077974. [DOI] [PubMed] [Google Scholar]
  • 65.Fowler DH. Shared biology of GVHD and GVT effects: potential methods of separation. Crit Rev Oncol Hematol. 2006;57:225–44. doi: 10.1016/j.critrevonc.2005.07.001. [DOI] [PubMed] [Google Scholar]
  • 66.Gattinoni L, Powell DJ, Jr., Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6:383–93. doi: 10.1038/nri1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gattinoni L, Klebanoff CA, Palmer DC, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005;115:1616–26. doi: 10.1172/JCI24480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–9. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kershaw MH, Teng MW, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol. 2005;5:928–40. doi: 10.1038/nri1729. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES