Вы находитесь на странице: 1из 8

International Immunology, Vol. 23, No. 11, pp. 661668 doi:10.

1093/intimm/dxr078 Advance Access publication 23 September 2011

The Japanese Society for Immunology. 2011. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

Transcriptional control of T-cell development


Taku Naito1, Hirokazu Tanaka1, Yoshinori Naoe2 and Ichiro Taniuchi1
1

Laboratory of Transcriptional Regulation, RIKEN Research Institute for Allergy and Immunology, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan 2 Section of Immunology, Department of Mechanism of Aging, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka-machi, Obu, Aichi, 474-8511, Japan
Correspondence to: I. Taniuchi; E-mail: taniuchi@rcai.riken.jp Received 25 July 2011, accepted 29 August 2011

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

Abstract T lymphocytes, which are central players in orchestrating immune responses, consist of several subtypes with distinct functions. The thymus is an organ where hematopoietic progenitors undergo sequential developmental processes to give rise to this variety of T-cell subsets with diverse antigen specicity. In the periphery, naive T cells further differentiate into effector cells upon encountering antigens. There are several developmental checkpoints during T-cell development, where regulation by a combination of transcription factors imprints specic functional properties on precursors. The transcription factors E2A, GATA-binding protein 3 (Gata3) and RUNT-related transcription factor (Runx) are involved at various stages in the differentiation of double-negative thymocytes and in b-selection, as are transcription factors from the Notch signaling pathway; other transcription factors such as B-cell lymphoma/leukemia 11b (Bcl11b), myeloblastosis viral oncogene homolog (Myb) and inhibitor of DNA binding 3 (Id3) are involved at specic stages. Differentiation of T cells into helper versus cytotoxic cells involves not only antagonistic interplay between Runx and Th inducing POZ-Kruppel factor (ThPOK) but also complex interactions between MAZR, Gata3 and Myb in the activation and silencing of genes such as Cd4 and Cd8 as well as the gene that encodes ThPOK itself. A wide range of well-dened transcription factors, including signal transducer and activator of transcriptions (STATs), T-bet, Gata3, nuclear factor of activated T cell (NFAT), adaptor-related protein complex 1 (AP-1) and nuclear factor kB (NF-kB), are known to shape Th1/Th2 differentiation. Runx and Gata3 also operate in this process, as do c-Maf and recombining binding protein for immunoglobulin Jk region (RBP-J) and the chromatin-reorganizing protein special AT-rich sequence-binding protein 1 (SATB1). In this review, we briey discuss how T-cell characteristics are acquired and become divergent from the point of view of transcriptional regulation. Keywords: gene regulation, T-cell development, transcription factors

Introduction T lymphocytes, which are distinguished from other leukocytes by their expression of either the ab or the cd type of TCR, are essential for regulating immune responses in addition to their cytotoxic functions. Although all lymphocyte progenitors are initially generated in the bone marrow, the thymus is the sole organ that supports development of T lymphocytes, whereas development of other leukocytes continues mainly in bone marrow. There are several types of mature T cells, and the generation of distinct T-cell subsets takes place rst in thymus, which is followed by differentiation of naive T cells into effector T cells upon encountering antigens in the peripheral lymphoid organs. The thymus is thus a place where multipotent hematopoietic progenitors go through several cell-fate decision processes to prepare them to become T-cell subsets. Given that all T cells in any of the subsets retain identical genome sequences except at the Tcr loci [where gene sequences are modied by RAG (recombination-activated gene)-mediated site-specic recombination], the generation of cell subsets with distinct functional properties is regulated by spatiotemporal expression of a selected set of genes. Transcription factors are nuclear proteins that bind specic gene sequences and (alone or in complexes with other proteins) activate or repress transcription of DNA to mRNA. Studies in the last two decades have identied transcription factors that are involved in decision-making processes during T-cell differentiation in the thymus and in the periphery. In this review, we will briey discuss progress in our understanding of a transcriptional control of T-cell development with special focus on the early commitment to the ab T lineage, the helper versus cytotoxic lineage choice in the thymus and the differentiation of CD4+ T cells into Th1 and Th2 cells.

REVIEW

662 Transcription factors and T-cell development Early thymocyte differentiation T-cell development is initiated in the thymic subpopulation that lacks the cell surface expression of both CD4 and CD8 glycoproteins, thus called double-negative (DN) thymocytes. T-lineage thymocytes then become double-positive (DP; CD4+CD8+) and, subsequently, single-positive (SP; CD4+CD8 or CD4CD8+) thymocytes. Classically, the thymocyte differentiation process within the DN population is divided into four stages according to the expression of CD25 and CD44, starting from DN1 (CD44+CD25), followed by DN2 (CD44+CD25+), DN3 (CD44CD25+) and DN4 (CD44CD25) (Fig. 1). DN1 is a heterogeneous population, which is further divided into ve subsets, DN1a to DN1e, according to the expression of CD24 and c-kit (CD117), among which only DN1a and DN1b are canonical T-cell progenitors (1); that is, although all ve subsets can generate T cells in vitro, only DN1a and DN1b do so efciently in vivo. The earliest known T-cell progenitors that seed the thymus are called early T-cell progenitors (ETP), which are characterized by the lack of lineage-specic markers (these Lin cells lack lymphocyte markers such as CD3 as well as markers for other lineages) although they do express markers such as c-kit and Sca-1 (2); ETP are the equivalent of the DN1a subset plus the DN1b subset. ETP can be further divided by the expression of fms-like tyrosine kinase receptor 3 (Flt3). The Flt3+ ETP retain the potential to differentiate into lineages other than T cells, such as B cells, NK cells, dendritic cells (DCs) and macrophages, but the capability to produce B cells is lost in their Flt3 progeny. Flt3+ ETP thus phenotypically resemble lymphoid-primed multipotent progenitor (LMPP) (3), which are hematopoietic progenitors found in the bone marrow that are committed to develop into lympho-myeloid cells. Several transcription factors that are critical for the emergence of ETP have been reported. The Notch signaling pathway was the rst to be demonstrated as critical for the generation of ETP (4), and Mastermind-like is a key signaling protein in the Notch pathway. Inhibition of Notch signaling using dominant-negative Mastermind-like caused loss of ETP and its progeny but did not affect the LSK (LinSca-1+c-Kit+) cell population, which contain LMPP, the potential immediate progenitors of ETP. Recently, T-cell factor 1 (TCF-1, also known as TCF-7) was identied as a critical downstream factor of Notch1 (5). TCF-1-decient T-cell progenitors show marked reduction after ETP onward in competitive reconstitution assay. TCF-1 expression is low in LMPP or common lymphoid progenitor but induced in ETP. TCF-1 expression is induced by Notch signal presumably via direct regulation. Over-expression of TCF-1 elicits T lineage cells even in the absence of Notch signaling, inducing T cell-specic transcription factors including GATA-binding protein 3 (Gata3) and B-cell lymphoma/leukemia 11b (Bcl11b). Loss of function of the transcription factor Gata3 also caused severe inhibition of ETP development, but not that of the presumed immediate progenitors (6). In addition, Gata3 is known to be necessary for ETP to become DN2 thymocytes. Both E2A and HEB, which are members of the basic helixloophelix (bHLH) transcription factor family, also play an important role in early T-cell development. Analyses of E2Adecient mice indicated that E2A functions at multiple stages, but the earliest defect observed by a lack of E2A is a decrease of LMPP in the bone marrow; this decrease is carried over into the thymus as a decrease in ETP number (7). The amount of E47, an isoform of E2A, increases upon DN1-to-DN2 transition (8), and accumulation of DN1 cells is observed in E2A-decient mice (9). RUNT-related transcription factor (Runx) complexes, which are composed of a Runx protein and an obligate non-DNAbinding partner core-binding factor b (Cbfb), are also known to be essential for early thymocyte development. A hypomorphic mutation in the Cbfb gene resulted in a consecutive differentiation block within the DN population, starting with a decrease of ETP followed by an inefcient ETP-to-DN2 transition as well as DN2-to-DN3 transition (10). In addition, a complete developmental block at the DN2-to-DN3 transition was shown in recipient mice reconstituted with Runx1-decient bone marrow progenitors (11, 12). Cells at the DN2 stage still possess the potential to become DC, NK and macrophages in the beginning. They do,

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

Fig. 1. Regulation of early thymocyte development by transcription factors. The stages of early T-cell differentiation are shown in the middle. On the top, the expression of the markers dening each stage is shown. The stages during which each transcription factor functions are shown at the bottom. The black arrows indicate the points where transcription factors are required, whereas the black bar indicates inhibition by Id3.

Transcription factors and T-cell development 663 however, commit to the T-lineage at the late DN2 stage by losing this potential. Early and late DN2 stages can be distinguished by the expression of a pLckgreen uorescent protein (GFP) transgene (13) since GFP DN2 (early-DN2) cells can produce DC and macrophage, whereas GFP+ DN2 (late-DN2) cells produce only T cells in this culture system. Recently, it was shown that early-DN2 status can be maintained in vitro under high IL-7 concentrations. A shift to low IL-7 concentrations induced sequential differentiation through DN3 cells to DP thymocytes (14). These observations suggest that the transition from the DN2 stage to the DN3 stage is not an automatic process, and there is a developmental checkpoint before acquiring the DN3 phenotype. Recently, three individual papers (1315) identied a transcription factor, Bcl11b, which is essential to fully commit to the T-lineage, providing molecular evidence for an essential branching point at the DN2 stage. The expression of Bcl11b in hematopoietic cells is limited to the T-lineage and rises in DN2 thymocytes (15). Detailed analyses of Bcl11b germ line knockout mice revealed that thymocyte development had stopped at the c-Kit+CD25+ DN2 stage in the absence of Bcl11b (14, 16); furthermore, retroviral transduction of Bcl11b into self-proliferating early-DN2 cells unlocked the developmental block that occurs under high IL-7 concentrations (14). Conditional removal of Bcl11b from fully T-lineage-committed cells such as DN3 or even DP thymocytes results in the emergence of NK-like cells presumably via de-differentiation or transdifferentiation (15). These results demonstrated a crucial role of Bcl11b for T-lineage commitment that takes place at the early-to-late DN2 transition. Upon Bcl11b induction, expression of PU.1, an essential transcription factor for myeloid lineage development, decreased through an as-yet uncharacterized mechanism (14). Interestingly, as mentioned above, a developmental block at the DN2 stage was also observed by inactivation of the Runx1 gene (11, 12). We have thus started to get a list of transcription factors involved in the commitment toward the T-lineage. After acquiring T-lineage properties, abT-cell precursors at the DN3 stage have to pass another developmental checkpoint known as b-selection, where the expression of a functional TCRb chain is examined. Because of the necessity to generate TCR diversity, not only transcriptional activation of the Tcrb gene but also successful VDJ rearrangement are required to express a functional TCRb chain that forms preTCR complexes with the pre-Ta molecule (which is encoded by the Ptcra gene). Impairment of E2A and HEB function by expression of dominant-negative HEB protein causes a defect of VDJ rearrangement, thereby causing developmental arrest at DN3 (17). A direct requirement for E2A in activating VDJ recombination at the TCRb locus could be one reason for this rearrangement defect (18). The developmental block was not, however, rescued by the expression of a rearranged TCRb chain, indicating that bHLH proteins also play a crucial role after b-selection (17). E2A is, furthermore, required to maintain the integrity of the b-checkpoint (8, 19). The bHLH transcription factors are also involved in the regulation of expansion of thymocytes that have passed b-selection; in the absence of E2A or HEB, DN3 cells exhibited premature hyperproliferative activity (19, 20). Another player that gives a ticket to pass the DN3 stage is Notch. Knockout of either Notch1 or of recombining binding protein for immunoglobulin Jj region (RBP-J), which acts as an essential transcription factor downstream of the Notch pathway, causes accumulation of DN3 cells, with defective V-to-DJ rearrangement in the case of Notch1 inactivation (21, 22). Recent studies show the convergence of the E2A and Notch pathways at the DN3 stage. Activation of the Notch1 gene is downstream of E2A. In addition, E2A, together with Notch1, regulates many genes, some of which, such as hairy and enhancer of split 1 (Hes1) and Ptcra, were previously known as Notch1 targets (23). The amount of inhibitor of DNA binding 3 (Id3), an inhibitor of E2A, is up-regulated upon pre-TCR signaling and downmodulates E2A activity (8). At the DN3 stage, bHLH proteins are thus likely to cooperate with Notch1 to prevent uncontrolled proliferation and promote TCRb chain rearrangement. Once cells are signaled through their pre-TCR complexes, Id3 suppresses bHLH protein activity, a mechanism that supposedly functions to ensure allelic exclusion (18). Weakened bHLH activity and down-modulation of Notch1 releases the developmental block and allows cells to expand. Conditional deletion of myeloblastosis viral oncogene homolog (Myb) also causes a developmental block at the DN3 stage, with decreased V(D)J recombination at the TCRb locus (24, 25). Loss of Gata3 function results in accumulation of DN3 cells (26) with a defect in the expression of TCRb protein while rearrangement and transcription at the TCRb locus are intact. Runx1 is also known to be required for proliferative expansion after b-selection (27). At each step during early thymocyte differentiation, many transcription factors thus act cooperatively to imprint T-cell signatures. The helper versus cytotoxic lineage choice Thymocytes that have passed b-selection start to express the TCRa chain and the CD4 and CD8 co-receptors. These DP thymocytes comprise the largest cell population in the thymus. Each DP thymocyte is subjected to another selection process, known as positive selection, during which the quality of its ab TCR is examined according to its afnity for self-peptide presented on MHC molecules (28). Consequently, positive selection allows only a small percentage of DP thymocytes to further differentiate into one of two mature thymocytes subsets: CD4+CD8 or CD4CD8+ SP thymocytes. It is well documented that a DP thymocyte expressing an ab TCR that recognizes peptide on MHC class I differentiates into a CD4-CD8+ SP thymocyte that is committed to the cytotoxic lineage, whereas a DP thymocyte selected by MHC class II becomes a CD4+CD8 thymocyte committed to the helper lineage. To explain how MHC restriction of TCRs is linked with nal CD4/CD8 co-receptor expression and cell fate discrimination between helper versus cytotoxic lineages, several models have been proposed and challenged. Since another review article discussed these models in detail (29), we will not further discuss them here. Rather this review focuses on transcription factors regulating lineage choice and CD4/CD8 gene expression. Because of lineage-specic expression of genes encoding CD4 or CD8 co-receptors, it has been assumed that the

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

664 Transcription factors and T-cell development mechanism regulating Cd4 or Cd8 gene expression would share common molecules with the mechanism regulating the helper versus cytotoxic lineage choice (30). Studies in the 1990s had revealed that expression of the Cd4 gene is repressed in cytotoxic-lineage cells by an intronic transcriptional silencer, referred to as the Cd4 silencer (31, 32). Subsequently, Runx complexes were identied as proteins that bind to the Cd4 silencer (33) (Fig. 2). Interestingly, distinct Runx proteins regulate the Cd4 silencer activity at distinct developmental stages: Runx1 at the DN stage and Runx3 at the CD4CD8+ SP stage. Given redundant function between Runx1 and Runx3 for activating the Cd4 silencer, a stage-specic function of each Runx protein in Cd4 repression could be attributed to a stage-specic expression pattern of each Runx protein. In addition to the Cd4 gene regulation, Runx complexes were shown to bind to the enhancer regions in the Cd8 locus (34), suggesting that Runx complexes are involved in Cd8 gene activation during differentiation to CD4CD8+ SP thymocytes. Runx complexes were thus rst characterized as essential regulators for Cd4/Cd8 expression during cytotoxic-lineage differentiation. Recent work has identied other profound roles of Runx in repressing genetic programming toward the helper lineage via silencing the zinc nger and BTB domain containing 7b (Zbtb7b) gene, which encodes a central transcription factor for CD4+ Th cell development, namely Th inducing POZKruppel factor (ThPOK). The Zbtb7b gene was identied by Kappes and his colleagues as a responsive locus for the helper-decient (HD) phenotype in a natural mutant mouse strain that lacks the CD4+ T-cell subset in the periphery (35). In HD mice, although positive selection of thymocytes expressing MHC class II-restricted TCRs is intact, differentiation of those cells is re-directed into the alternative CD8+ cytotoxic lineage (36). This result indicates that the positive selection process is independent of lineage selection. Ectopic expression of ThPOK from the DP thymocyte stage not only rescued the HD phenotype but also induced re-directed differentiation of MHC class I-selected thymocytes into CD4+ T cells (35). Expression of a single transcription factor in postselected thymocytes is thus both essential and sufcient to acquire the CD4+CD8 phenotype with helper-related features. Consistent with the above observations, expression of the Zbtb7b gene during thymocyte development is rst detected in post-selection thymocytes and is up-regulated only during maturation of MHC class II-selected thymocytes (35). Understanding of how expression of the Zbtb7b is restricted to MHC class II-selected thymocytes therefore became a key to unravel how the CD4/CD8 linage choice is transcriptionally regulated. Interestingly, combined mutation in both Runx1 and Runx3 genes led to a similar phenotype to that of ThPOK transgenic mice, resulting in a re-direction of MHC class I-selected thymocytes into CD4+ T cells (37). In addition, premature expression of the Zbtb7b was observed in preselection (i.e. CD69) DP thymocytes by the ablation of the function of Runx complexes. Using ChIP-on-chip technology (which combines chromatin immunoprecipitation and microarray technologies to identify genomic regions bound by protein of interest), the Runx complex was found to associates with the Zbtb7b locus at two regions (37), which correspond to two regulatory regions, distal regulatory element (DRE) and proximal

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

Fig. 2. Regulation of ThPOK expression and ThPOK function during Th cell development. Expression of the Zbtb7b gene, encoding the transcription factor ThPOK, is negatively regulated by the ThPOK silencer, whose activity requires biding of the Runx complex and MAZR. TCR engagement by antigen on MHC class II initiates Zbtb7b expression in part by inactivation of the ThPOK silencer. ThPOK not only activates the helper program but also suppresses cytotoxic features such as Cd8 and Runx3 expression. Antagonistic interplay between ThPOK and Runx3 is thus central in the transcription factor network that governs the helper versus cytotoxic lineage choice. An antagonistic function of ThPOK for both the Cd4 silencer and the ThPOK silencer stabilizes ThPOK and CD4 expression.

Transcription factors and T-cell development 665 regulatory element (PRE), which were identied through the analysis of DNase I hypersensitive sites and a reporter transgene expression assay (38). DRE, which locates at 3.2kb upstream from the transcriptional start site in the Zbtb7b locus, possesses a transcriptional silencer activity, which is essential to confer helper lineage-specic expression of a reporter transgene driven by other regulatory regions from the Zbtb7b gene (38). On the other hand, PRE, locating at 7.5-kb downstream, possesses a transcriptional enhancer activity (38), which is essential to up-regulate, and maintains Zbtb7b expression during development of CD4+ Th cells (39). Taking these observations together, it seems that Runx complexes play an essential role in repressing Zbtb7b expression via direct regulation of a silencer activity in DRE (37). DRE was, however, shown to still function as a silencer without putative Runx recognition sites in a reporter transgene assay, raising a question about whether Runx complexes are essential for silencer activity (40). Targeted mutations in Runx sites at the endogenous DRE almost abrogated silencer activity, however (I.T. manuscript in preparation). Runx complexes are thus essential to limit ThPOK expression to MHC class II-selected cells. Conversely, ThPOK appeared to repress Runx3 expression during differentiation toward CD4+ T cells (39, 41, 42). Cross-antagonistic regulation between ThPOK and Runx3 is thus central in a transcription factor network that governs the helper versus cytotoxic lineage choice. Interestingly, ThPOK can antagonize the silencer activity of the DRE as well as the Cd4 silencer (39), indicating the presence of auto-feedforward loop that amplies ThPOK expression during Th cell development. MAZR has been identied as a negative regulator for CD8 expression (43). Recently, MAZR deciency was shown to cause a partial re-direction of MHC class I-selected cells (44). As observed in Runx-mutant mice, loss of MAZR function resulted in de-repression of the Zbtb7b gene, albeit to a lesser extent. Along with binding of MAZR to the DRE, MAZR is thus a functional unit in protein complexes that regulate DRE silencer activity (44). Another transcription factor involved in Zbtb7b gene regulation is Gata3. CD4 SP thymocyte development was markedly impaired by conditional inactivation of the Gata3 gene in DP thymocytes (26, 45). Although results differed among assay systems, MHC class II-selected thymocytes could be partially re-directed to CD8+ T cells in the absence of Gata3 function. Along with binding of Gata3 to two regions in the Zbtb7b locus, the lack of ThPOK expression in Gata3-decient thymus indicates that Gata3 functions as an upstream factor for Zbtb7b expression (46). Because transgenic expression of ThPOK failed to rescue CD4 SP thymocyte development caused by Gata3 deciency (46), however, a ThPOK-independent pathway also operates in parallel with a ThPOK-mediated pathway in the Gata3 axis for programming CD4+ T-cell development. Myb is another transcription factor involved in promoting CD4 lineage development (47). Although conditional knockout of Myb in DP thymocytes resulted in a decrease and an increase of CD4 SP cells and CD8 SP cells, respectively, redirection of MHC class II-restricted cells into CD8 SP cells was not observed. Binding of Myb to the Gata3 locus suggests that Myb might be a direct upstream molecule for Gata3 expression. TOX (thymocyte selection-associated high-mobility group box transcription factor) was originally identied as a molecule whose expression is up-regulated upon receiving positive selection signals. Over-expression of Tox by transgenesis promoted cytotoxic lineage development (48); however, Tox deciency resulted in a lack of CD4+ T cells and a lack of ThPOK expression (49), as was observed in Gata3-decient cells. Gata3 expression was detected in Tox-decient thymocytes, however, suggesting that Tox could function in activating the Zbtb7b gene in a Gata3independent manner. Differentiation of Th1 and Th2 cells The immune response to different pathogens is tailored by the differentiation of CD4+ Th cells into different effector types (50). After encountering antigen, and depending on the microenvironment (including the balance of cytokine stimulation), naive CD4+ T cells differentiate into distinct effector T cells. Currently, a variety of CD4+ T-cell subsets have been recognized, and transcription factors that involved in development of each subset are being identied (51); however, separation of effector CD4+ T-cell subsets was initiated with the identication of two subsets, Th1 and Th2 cells. Th1 cells are distinguished by predominant production of IFN-c and play a major role in protective immune responses to intracellular viral and bacterial infections. Th2 cells, by producing IL4, IL-5, IL-6, IL-9, IL-10, IL-13 and IL-25, are critical for expelling extracellular pathogens, such as bacteria and a variety of parasites, and are also involved in allergic reactions. Because of the long history of the studies, both transcriptional factors and cytokine signals leading to Th1 and Th2 differentiation are understood the best. We will therefore only briey review the developmental pathway of Th1 and Th2 subsets in this section. Development toward the Th1 subset is initiated by stimulation with IL-12 and IFN-c, which are secreted by DCs and macrophages in response to intracellular pathogen infection. Th1 differentiation is thus linked to activation of the transcription factors signal transducer and activator of transcription (STAT) 1 and STAT4 downstream of IFN-c and IL-12 signaling, respectively. Together with the transcription factors [such as nuclear factor of activated T cell (NFAT), adaptorrelated protein complex 1 (AP-1) and nuclear factor jB (NFjB)] that are activated by TCR engagement, STAT1 induces the expression of the master transcriptional factor of the Th1 subset, T-bet [also known as T-box transcription factor 21 (Tbx21)] (Fig. 3). Once expressed, T-bet induces IL-12R expression, resulting in enhanced IL-12STAT4 signaling (52). Subsequently, STAT4 and T-bet act coordinately to induce the expression of Runx3, which functions coordinately with T-bet to produce large amounts of IFN-c production in Th1 cells (53). Unlike T-bet, however, transduction of Runx3 did not potentiate IFN-c production in Th2 cells (53). It is likely that T-bet-dependent remodeling of chromatin structures at the Ifng locus early in Th1 development is important for

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

666 Transcription factors and T-cell development

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

Fig. 3. Key transcriptional factors in Th1 and Th2 differentiation. IFN-c stimulation leads to the activation of T-bet, the master regulator for Th1 cells, via activation of Stat1. IL-12 reinforces IFN-c production and Runx expression via Stat4 activation together with T-bet. Runx also represses IL-4 expression and antagonizes Gata3. In the presence of IL-4, the Stat6-mediated signaling pathway induces expression of Gata3, the master regulator for Th2 differentiation. Gata3 induces c-Maf expression, which cooperates with Gata3 to activate production of Th2 cytokines such as IL-4.

subsequent binding of Runx3 and other factors expressed at later stages. On the other hand, T-bet and Runx3 were also shown to inhibit Th2 programming both by antagonizing Gata3 activity (54, 55) and by direct repression of the Il4 gene through activating a silencer element in the Il4 locus (53, 56). Th2 development is promoted by the cytokine IL-4, which signals via activation of STAT6 after engagement of the IL4R. As is the case for IFN-c and IL-12 auto-regulation during Th1 differentiation, IL-4 signals enhance Il4 transcription in cooperation with NFAT, AP-1 and NF-jB (57, 58). Importantly, these signals also amplify expression level of a master regulator of Th2 differentiation, Gata3 (59). Gata3 is known to activate its own expression as well as to drive epigenetic changes at the Th2 cytokine cluster, which contains the Il4, Il5 and Il13 genes. Gata3 induces another transcription factor, c-Maf, which additionally helps to induce Il4 transcription. Forced expression of STAT6 in differentiated Th1 cells is sufcient to induce Gata3 expression as well as IL-4 and IL-5 cytokine production. In addition, ectopic expression of Gata3 in developing Th1 cells induces Th2 cytokine production (60). Unlike Gata3, however, c-Maf fails to induce IL-4 expression under Th1-skewing conditions (61), suggesting that c-Maf expression alone is not sufcient to induce a Th2 program in developing Th1 cells. On the other hand, Gata3 was shown to have antagonistic ability against the Th1 program by inhibiting responsiveness to IL-12 and IFN-c (62). STAT6, Gata3 and c-Maf thus cooperatively work not only to stabilize Th2 features in part by generating an auto-feedforward loop but also to extinguish Th1 features. Recent studies demonstrated that the Th2 cytokine loci undergo alterations of higher order chromatin structures

including changes in DNA methylation and histone modications. These epigenetic changes increase the accessibility of Th2 loci for recruitment of transcriptional factors, such as NFATs, STATs, Gata3 and c-Maf. NFAT1 is likely to play an important role in regulating chromatin structures during the initiation and re-activation processes as it binds to promoters of all Il4, Il5 and Il13 genes and enhancer elements in the Il4 locus (63); moreover, long-range interactions between the promoters of these genes have been proposed to function in bringing regulatory regions into close spatial proximity (64). Recently, it was shown that this process could be regulated by special AT-rich sequence-binding protein 1 (SATB1), a long-range chromatin-organizing protein, consistent with the essential requirement of SATB1 for Th2 cytokine expression (65). It has been demonstrated that Notch pathway is also involved in regulation of Th1/Th2 differentiation (66). Engagement of Notch with a Delta-like ligand (DLL)Fc fusion protein (which stimulates Notch) enhanced Th1 responses (67). In addition, ectopic expression of DLL1 and DLL4 on DCs stimulates Th1 differentiation (68). Given the association of Notch3 and RBP-J with the Tbx21 gene, Notch3 potentially activates the Tbx21gene (69). On the contrary, Notch1 and Notch2 were shown to enhance II4 expression via direct activation of the HS5 enhancer (68). In addition, Notch would activate the Gata3 gene, presumably via direct binding to the distal promoter in the Gata3 gene (69). Th2 cell responses are, furthermore, severely impaired by ablation of RBP-J or Notch1/Notch2 from T cells (22, 69). In addition to the cytokine environment, external stimuli signaled through Notch axis could thus be other factors in discriminating Th1/ Th2 differentiation.

Transcription factors and T-cell development 667 Conclusions Because of its key role in regulating acquired immune responses and its usefulness as a model system for studying cell differentiation, T-cell development has been of interest for immunologists. With advances in genetic and bioinformatics technology, transcription factors that play a pivotal role in regulating T-cell development have been identied; however, it remains elusive how these factors control genetic programming toward a particular lineage during the commitment process and how the given integrity is maintained. For instance, although ThPOK is important for preventing expression of cytotoxic-lineage related genes, such as Runx3 and Cd8, ThPOK target genes that are necessary to install helper function are poorly characterized. In addition, re-expression of Runx3 occurs during CD4+ T-cell differentiation to the Th1 subset while the Cd8 gene is still repressed. Accumulating evidence suggest that epigenetic mechanisms contribute to retain cell identity through imprinting gene expression patterns during the commitment process. Future studies will aim to understand how a genetic program activated by a combinatorial activity of transcription factors is stabilized by epigenetic machineries in a locusand stage-specic manner while leaving some developmental potency for further differentiation upon encounter to a novel microenvironment. References
1 Porritt, H. E., Rumfelt, L. L., Tabrizifard, S., Schmitt, T. M., ZunigaPucker, J. C. and Petrie, H. T. 2004. Heterogeneity among DN1 prothymocytes reveals multiple progenitors with different capacities to generate T cell and non-T cell lineages. Immunity 20:735. 2 Allman, D., Sambandam, A., Kim, S. et al. 2003. Thymopoiesis independent of common lymphoid progenitors. Nat. Immunol. 4:168. 3 Adolfsson, J., Mansson, R., Buza-Vidas, N. et al. 2005. Identication of Flt3+ lympho-myeloid stem cells lacking erythromegakaryocytic potential a revised road map for adult blood lineage commitment. Cell 121:295. 4 Sambandam, A., Maillard, I., Zediak, V. P. et al. 2005. Notch signaling controls the generation and differentiation of early T lineage progenitors. Nat. Immunol 6:663. 5 Weber, B. N., Chi, A. W., Chavez, A. et al. 2011. A critical role for TCF-1 in T-lineage specication and differentiation. Nature 476:63. 6 Hosoya, T., Kuroha, T., Moriguchi, T. et al. 2009. GATA-3 is required for early T lineage progenitor development. J. Exp. Med. 206:2987. 7 Dias, S., Mansson, R., Gurbuxani, S., Sigvardsson, M. and Kee, B. L. 2008. E2A proteins promote development of lymphoid-primed multipotent progenitors. Immunity 29:217. 8 Engel, I., Johns, C., Bain, G., Rivera, R. R. and Murre, C. 2001. Early thymocyte development is regulated by modulation of E2A protein activity. J. Exp. Med 194:733. 9 Bain, G., Engel, I., Robanus Maandag, E. C. et al. 1997. E2A deciency leads to abnormalities in abT-cell development and to rapid development of T-cell lymphomas. Mol. Cell. Biol 17:4782. 10 Talebian, L., Li, Z., Guo, Y. et al. 2007. T-lymphoid, megakaryocyte, and granulocyte development are sensitive to decreases in CBFb dosage. Blood 109:11. 11 Ichikawa, M., Asai, T., Saito, T. et al. 2004. AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis. Nat. Med. 10:299. 12 Growney, J. D., Shigematsu, H., Li, Z. et al. 2005. Loss of Runx1 perturbs adult hematopoiesis and is associated with a myeloproliferative phenotype. Blood 106:494. 13 Masuda, K., Kakugawa, K., Nakayama, T., Minato, N., Katsura, Y. and Kawamoto, H. 2007. T cell lineage determination precedes the initiation of TCRb gene rearrangement. J. Immunol. 179:3699. 14 Ikawa, T., Hirose, S., Masuda, K. et al. 2010. An essential developmental checkpoint for production of the T cell lineage. Science 329:93. 15 Li, P., Burke, S., Wang, J. et al. 2010. Reprogramming of T cells to natural killer-like cells upon Bcl11b deletion. Science 329:85. 16 Li, L., Leid, M. and Rothenberg, E. V. 2010. An early T cell lineage commitment checkpoint dependent on the transcription factor Bcl11b. Science 329:89. 17 Barndt, R. J., Dai, M. and Zhuang, Y. 2000. Functions of E2A-HEB heterodimers in T-cell development revealed by a dominant negative mutation of HEB. Mol. Cell. Biol 20:6677. 18 Agata, Y., Tamaki, N., Sakamoto, S. et al. 2007. Regulation of T cell receptor b gene rearrangements and allelic exclusion by the helixloop-helix protein, E47. Immunity 27:871. 19 Engel, I. and Murre, C. 2004. E2A proteins enforce a proliferation checkpoint in developing thymocytes. EMBO J 23:202. 20 Wojciechowski, J., Lai, A., Kondo, M. and Zhuang, Y. 2007. E2A and HEB are required to block thymocyte proliferation prior to preTCR expression. J. Immunol 178:5717. 21 Wolfer, A., Wilson, A., Nemir, M., MacDonald, H. R. and Radtke, F. 2002. Inactivation of Notch1 impairs VDJb rearrangement and allows pre-TCR-independent survival of early ab lineage Thymocytes. Immunity 16:869. 22 Tanigaki, K., Tsuji, M., Yamamoto, N. et al. 2004. Regulation of ab/cd T cell lineage commitment and peripheral T cell responses by Notch/RBP-J signaling. Immunity 20:611. 23 Ikawa, T., Kawamoto, H., Goldrath, A. W. and Murre, C. 2006. E proteins and Notch signaling cooperate to promote T cell lineage specication and commitment. J. Exp. Med 203:1329. 24 Lieu, Y. K., Kumar, A., Pajerowski, A. G., Rogers, T. J. and Reddy, E. P. 2004. Requirement of c-myb in T cell development and in mature T cell function. Proc. Natl. Acad. Sci. USA 101:14853. 25 Bender, T. P., Kremer, C. S., Kraus, M., Buch, T. and Rajewsky, K. 2004. Critical functions for c-Myb at three checkpoints during thymocyte development. Nat. Immunol. 5:721. 26 Pai, S. Y., Truitt, M. L., Ting, C. N., Leiden, J. M., Glimcher, L. H. and Ho, I. C. 2003. Critical roles for transcription factor GATA-3 in thymocyte development. Immunity 19:863. 27 Egawa, T., Tillman, R. E., Naoe, Y., Taniuchi, I. and Littman, D. R. 2007. The role of the Runx transcription factors in thymocyte differentiation and in homeostasis of naive T cells. J. Exp. Med 204:1945. 28 Germain, R. N. 2002. T-cell development and the CD4-CD8 lineage decision. Nat. Rev. Immunol 2:309. 29 Singer, A., Adoro, S. and Park, J. H. 2008. Lineage fate and intense debate: myths, models and mechanisms of CD4- versus CD8-lineage choice. Nat. Rev. Immunol. 8:788. 30 Ellmeier, W., Sawada, S. and Littman, D. R. 1999. The regulation of CD4 and CD8 coreceptor gene expression during T cell development. Annu. Rev. Immunol. 17:523. 31 Sawada, S., Scarborough, J. D., Killeen, N. and Littman, D. R. 1994. A lineage-specic transcriptional silencer regulates CD4 gene expression during T lymphocyte development. Cell 77:917. 32 Siu, G., Wurster, A. L., Duncan, D. D., Soliman, T. M. and Hedrick, S. M. 1994. A transcriptional silencer controls the developmental expression of the CD4 gene. EMBO J 13:3570. 33 Taniuchi, I., Osato, M., Egawa, T. et al. 2002. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell 111:621. 34 Sato, T., Ohno, S., Hayashi, T. et al. 2005. Dual functions of Runx proteins for reactivating CD8 and silencing CD4 at the commitment process into CD8 thymocytes. Immunity 22:317. 35 He, X., He, X., Dave, V. P. et al. 2005. The zinc nger transcription factor Th-POK regulates CD4 versus CD8 T-cell lineage commitment. Nature 433:826. 36 Keefe, R., Dave, V., Allman, D., Wiest, D. and Kappes, D. J. 1999. Regulation of lineage commitment distinct from positive selection. Science 286:1149. 37 Setoguchi, R., Tachibana, M., Naoe, Y. et al. 2008. Repression of the transcription factor Th-POK by Runx complexes in cytotoxic T cell development. Science 319:822.

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

668 Transcription factors and T-cell development


38 He, X., Park, K., Wang, H. et al. 2008. CD4-CD8 lineage commitment is regulated by a silencer element at the ThPOK transcription-factor locus. Immunity 28:346. 39 Muroi, S., Naoe, Y., Miyamoto, C. et al. 2008. Cascading suppression of transcriptional silencers by ThPOK seals helper T cell fate. Nat. Immunol. 9:1113. 40 Hedrick, S. M. 2008. Thymus lineage commitment: a single switch. Immunity 28:297. 41 Egawa, T. and Littman, D. R. 2008. ThPOK acts late in specication of the helper T cell lineage and suppresses Runxmediated commitment to the cytotoxic T cell lineage. Nat. Immunol 9:1131. 42 Wang, L., Wildt, K. F., Castro, E. et al. 2008. The zinc nger transcription factor Zbtb7b represses CD8-lineage gene expression in peripheral CD4+ T cells. Immunity 29:876. 43 Bilic, I., Koesters, C., Unger, B. et al. 2006. Negative regulation of CD8 expression via Cd8 enhancer-mediated recruitment of the zinc nger protein MAZR. Nat. Immunol 7:392. 44 Sakaguchi, S., Hombauer, M., Bilic, I. et al. 2010. The zinc-nger protein MAZR is part of the transcription factor network that controls the CD4 versus CD8 lineage fate of double-positive thymocytes. Nat. Immunol 11:442. 45 Hernandez-Hoyos, G., Anderson, M. K., Wang, C., Rothenberg, E. V. and Alberola-Ila, J. 2003. GATA-3 expression is controlled by TCR signals and regulates CD4/CD8 differentiation. Immunity 19:83. 46 Wang, L., Wildt, K. F., Zhu, J. et al. 2008. Distinct functions for the transcription factors GATA-3 and ThPOK during intrathymic differentiation of CD4+ T cells. Nat. Immunol 9:1122. 47 Maurice, D., Hooper, J., Lang, G. and Weston, K. 2007. c-Myb regulates lineage choice in developing thymocytes via its target gene Gata3. EMBO J 26:3629. 48 Wilkinson, B., Chen, J. Y., Han, P., Rufner, K. M., Goularte, O. D. and Kaye, J. 2002. TOX: an HMG box protein implicated in the regulation of thymocyte selection. Nat. Immunol 3:272. 49 Aliahmad, P. and Kaye, J. 2008. Development of all CD4 T lineages requires nuclear factor TOX. J. Exp. Med 205:245. 50 Weaver, C. T., Harrington, L. E., Mangan, P. R., Gavrieli, M. and Murphy, K. M. 2006. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity 24:677. 51 Zhou, L., Chong, M. M. and Littman, D. R. 2009. Plasticity of CD4+ T cell lineage differentiation. Immunity 30:646. 52 Afkarian, M., Sedy, J. R., Yang, J. et al. 2002. T-bet is a STAT1induced regulator of IL-12R expression in naive CD4+ T cells. Nat. Immunol. 3:549. 53 Djuretic, I. M., Levanon, D., Negreanu, V., Groner, Y., Rao, A. and Ansel, K. M. 2007. Transcription factors T-bet and Runx3 cooperate to activate IFNG and silence Il4 in T helper type 1 cells. Nat. Immunol. 8:145. 54 Usui, T., Preiss, J. C., Kanno, Y. et al. 2006. T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNc gene acetylation and transcription. J. Exp. Med 203:755. 55 Hwang, E. S., Szabo, S. J., Schwartzberg, P. L. and Glimcher, L. H. 2005. T helper cell fate specied by kinase-mediated interaction of T-bet with GATA-3. Science 307:430. 56 Naoe, Y., Setoguchi, R., Akiyama, K. et al. 2007. Repression of interleukin-4 in T helper type 1 cells by Runx/Cbfb binding to the Il4 silencer. J. Exp. Med 204:1749. 57 Macian, F., Garcia-Rodriguez, C. and Rao, A. 2000. Gene expression elicited by NFAT in the presence or absence of cooperative recruitment of Fos and Jun. EMBO J 19:4783. 58 Chuvpilo, S., Schomberg, C., Gerwig, R. et al. 1993. Multiple closely-linked NFAT/octamer and HMG I(Y) binding sites are part of the interleukin-4 promoter. Nucleic Acids Res. 21:5694. 59 Ouyang, W., Lohning, M., Gao, Z. et al. 2000. Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment. Immunity 12:27. 60 Zheng, W. and Flavell, R. A. 1997. The transcription factor GATA-3 is necessary and sufcient for Th2 cytokine gene expression in CD4 T cells. Cell 89:587. 61 Ho, I. C., Lo, D. and Glimcher, L. H. 1998. c-maf promotes T helper cell type 2 (Th2) and attenuates Th1 differentiation by both interleukin 4-dependent and -independent mechanisms. J. Exp. Med. 188:1859. 62 Zhang, D. H., Cohn, L., Ray, P., Bottomly, K. and Ray, A. 1997. Transcription factor GATA-3 is differentially expressed in murine Th1 and Th2 cells and controls Th2-specic expression of the interleukin-5 gene. J. Biol. Chem. 272:21597. 63 Ansel, K. M., Djuretic, I., Tanasa, B. and Rao, A. 2006. Regulation of Th2 differentiation and Il4 locus accessibility. Annu. Rev. Immunol. 24:607. 64 Spilianakis, C. G. and Flavell, R. A. 2004. Long-range intrachromosomal interactions in the T helper type 2 cytokine locus. Nat. Immunol. 5:1017. 65 Cai, S., Lee, C. C. and Kohwi-Shigematsu, T. 2006. SATB1 packages densely looped, transcriptionally active chromatin for coordinated expression of cytokine genes. Nat. Genet 38:1278. 66 Amsen, D., Antov, A. and Flavell, R. A. 2009. The different faces of Notch in T-helper-cell differentiation. Nat. Rev. Immunol. 9:116. 67 Maekawa, Y., Tsukumo, S., Chiba, S. et al. 2003. Delta1-Notch3 interactions bias the functional differentiation of activated CD4+ T cells. Immunity 19:549. 68 Amsen, D., Blander, J. M., Lee, G. R., Tanigaki, K., Honjo, T. and Flavell, R. A. 2004. Instruction of distinct CD4 T helper cell fates by different Notch ligands on antigen-presenting cells. Cell 117:515. 69 Amsen, D., Antov, A., Jankovic, D. et al. 2007. Direct regulation of Gata3 expression determines the T helper differentiation potential of Notch. Immunity 27:89.

Downloaded from http://intimm.oxfordjournals.org/ at UNIVERSIDAD NACIONAL AUTONOMA DE MEXICO on November 22, 2011

Вам также может понравиться