Elsevier

Seminars in Immunology

Volume 20, Issue 4, August 2008, Pages 236-246
Seminars in Immunology

Review
Competition and collaboration: GATA-3, PU.1, and Notch signaling in early T-cell fate determination

https://doi.org/10.1016/j.smim.2008.07.006Get rights and content

Abstract

T-cell precursors remain developmentally plastic for multiple cell generations after entering the thymus, preserving access to developmental alternatives of macrophage, dendritic-cell, and even mast-cell fates. The underlying regulatory basis of this plasticity is that early T-cell differentiation depends on transcription factors which can also promote alternative developmental programs. Interfactor competition, together with environmental signals, keep these diversions under control. Here the pathways leading to several lineage alternatives for early pro-T-cells are reviewed, with close focus on the mechanisms of action of three vital factors, GATA-3, PU.1, and Notch–Delta signals, whose counterbalance appears to be essential for T-cell specification.

Section snippets

Models for hematopoietic lineage choice

Hematopoietic stem cells can develop into any of at least 10 different kinds of effector cells, and these can be grouped broadly into lymphoid, myeloid, erythroid, megakaryocytic, and other types. Stem cells become particular cell types by passing through sequences of partially restricted, but still pluripotent intermediates. The outcome of competing regulatory forces at work on the cells during the partially restricted intermediate stages ultimately forms the basis for the commitment decision

Landmarks for early T-cell development

Early-stage T-cell precursors are partially restricted cells that migrate to the thymus either just after or immediately before losing the ability to give rise to B lymphocytes, but before they shut off molecularly distinct developmental alternatives including macrophage or dendritic-cell fates. Differentiation of these cells occurs in the thymic cortex over a period of many days and is accompanied by a flexible degree of proliferation. Fig. 2 summarizes the cell-surface and molecular

Control of early T-cell development by external signals

T-lineage specification is driven and maintained by signals from the environment. The transition from each DN stage to the next depends on the existing internal transcription factor complement in the cell together with changes in transcription factor function induced by external environmental signals. The most important of these signals, on current evidence, are cytokine signals, signals through the Wnt-β-catenin-TCF pathway, and repeated signals from the Notch pathway. As for other

Context-dependent modulation of Notch signals

Notch signaling is not a Boolean operator that determines T-lineage fate at a stroke, but a quantitatively modulated participant in protracted lineage decisions. A sustained succession of interactions with Delta-class target ligands, through all the cell cycles from ETP throughout DN2 stages, is needed to elicit full commitment to the T-lineage [35], [36], [44], [77]. Notch–Delta interaction can drive many different starting populations of hematopoietic precursors into T-cell development [21],

Essential roles for GATA-3 and PU.1 in early T-cell development

Both GATA-3 and PU.1 are essential for T-cell development. GATA-3, a double zinc finger (C4-type) transcription factor, is expressed in an almost T-lineage-specific pattern among hematopoietic cells. It is turned on among the earliest genes activated by Notch–Delta signaling in hematopoietic precursors, and is used throughout T-cell development from the ETP stage all the way to post-thymic responses to antigen. The ETS-family transcription factor PU.1 is almost the reverse, with virtually no

GATA-3-driven lineage infidelity: dosage and Notch sensitivity

With its T-lineage specific expression and recurrent T-lineage roles, GATA-3 has been an appealing candidate for a factor conferring T-lineage identity in early thymocytes (Fig. 1B). However, gain of function of GATA-3, in the presence or absence of Notch signals, fails to enhance either appearance or developmental progression of the earliest T-lineage cells [36], [106], [111], [112]. Only around the time of lineage commitment, from the DN3 stage or its human equivalent onward, does high-level

PU.1 and myeloid alternatives to the T-lineage program

Although needed to start T-cell development, PU.1 also helps to maintain a progenitor-like developmental plasticity in the ETP and DN2 cells. During commitment, expression of PU.1 and SCL (Tal1) precipitously shuts off [33], [124]. Forced gain of PU.1 function, beyond the stage when it is normally shut off [51], [52], [53], [54], [55], [106], provides evidence that endogenous PU.1 expression in earlier T-cell precursors is probably rate-limiting for their developmentally regulated access to

GATA-3 as antagonist of myeloid lineage options

The functions mobilized by Notch to preserve T-lineage identity before lineage commitment need to be identified. GATA-3 is an obvious candidate for one of the T-lineage gatekeeper functions that antagonizes myeloid fates under normal conditions. Myeloid lineage conversion of thymocytes by either C/EBPα or PU.1 is inhibited if GATA-3 is co-expressed [52], [127]. Furthermore, GATA-3 may be suspected of a role in lineage commitment itself, as overexpression of GATA-3 in early fetal thymocytes

Transdifferentiation, not dedifferentiation, in response to regulatory perturbation

Neither myeloid nor mast-cell programs are thought to be closely related to lymphocyte developmental programs, and indeed, conversion of thymocytes to dendritic cells by PU.1, inflammatory macrophages by C/EBPα, and mast cells by excess GATA-3 all involve a repression of T-lineage specific gene expression. Their diversionary activity is even more remarkable in view of the positive roles that two of these diversion-inducing factors normally play in T-cell development, when expressed at different

Conclusions

The results reviewed here show that T-cell precursor specification is not only a protracted process but a remarkably precarious one at the regulatory level. This makes sense of much biology that could otherwise seem counterintuitive. An increasingly rich literature confirms that most T-cell precursors naturally retain access to non-T and non-lymphoid programs well beyond the stage when they have lost B-lineage potential [16], [17], [18], [19], [77], [133], [134], [135]. The myeloid

Acknowledgments

We are most grateful to the primary authors of the work cited in this review, including members and former members of this laboratory whose results were discussed in detail. We especially thank Thomas Graf for patient encouragement and valuable insights, and Howard Petrie, Cornelis Murre, Hiroshi Kawamoto and Avinash Bhandoola for exciting discussions of work before publication. The authors’ work was supported by NIH grants CA90233, CA98925, and HL089123, the Albert Billings Ruddock

References (152)

  • M.H. Sieweke et al.

    A transcription factor party during blood cell differentiation

    Curr Opin Genet Dev

    (1998)
  • A. Bhandoola et al.

    Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from

    Immunity

    (2007)
  • E.V. Rothenberg

    Negotiation of the T lineage fate decision by transcription-factor interplay and microenvironmental signals

    Immunity

    (2007)
  • S. Greenbaum et al.

    Regulation of early lymphocyte development by E2A family proteins

    Semin Immunol

    (2002)
  • M.K. Anderson et al.

    Constitutive expression of PU.1 in fetal hematopoietic progenitors blocks T-cell development at the pro-T stage

    Immunity

    (2002)
  • C.V. Laiosa et al.

    Reprogramming of committed T cell progenitors to macrophages and dendritic cells by C/EBPα and PU.1 transcription factors

    Immunity

    (2006)
  • C.J. Dionne et al.

    Subversion of T lineage commitment by PU.1 in a clonal cell line system

    Dev Biol

    (2005)
  • J. Kang et al.

    Cytokine functions in the formative stages of a lymphocyte's life

    Curr Opin Immunol

    (2004)
  • Y. Guo et al.

    Core binding factors are necessary for natural killer cell development, and cooperate with Notch signaling during T cell specification

    Blood

    (2008)
  • H. Wang et al.

    Distinct roles of IL-7 and stem cell factor in the OP9-DL1 T-cell differentiation culture system

    Exp Hematol

    (2006)
  • R.M. Okamura et al.

    Redundant regulation of T cell differentiation and TCRα gene expression by the transcription factors LEF-1 and TCF-1

    Immunity

    (1998)
  • Y. Baba et al.

    Constitutively active β-catenin confers multilineage differentiation potential on lymphoid and myeloid progenitors

    Immunity

    (2005)
  • G. Jeannet et al.

    Long-term, multilineage hematopoiesis occurs in the combined absence of β-catenin and γ-catenin

    Blood

    (2008)
  • A.I. Garbe et al.

    TCR and Notch synergize in αβ versus γδ lineage choice

    Trends Immunol

    (2007)
  • K. Lucas et al.

    A linkage between dendritic cell and T-cell development in the mouse thymus: the capacity of sequential T-cell precursors to form dendritic cells in culture

    Dev Comp Immunol

    (1998)
  • M. De Smedt et al.

    Different thresholds of Notch signaling bias human precursor cells toward B-, NK-, monocytic/dendritic-, or T-cell lineage in thymus microenvironment

    Blood

    (2005)
  • S.M. Lehar et al.

    Notch ligands Delta1 and Jagged1 transmit distinct signals to T cell precursors

    Blood

    (2005)
  • B.C. Harman et al.

    T/B lineage choice occurs prior to intrathymic Notch signalling

    Blood

    (2005)
  • T. Taghon et al.

    Developmental and molecular characterization of emerging β- and γδ-selected pre-T cells in the adult mouse thymus

    Immunity

    (2006)
  • J.W. Cave et al.

    A DNA transcription code for cell-specific gene activation by Notch signaling

    Curr Biol

    (2005)
  • D.J. Izon et al.

    Deltex1 redirects lymphoid progenitors to the B cell lineage by antagonizing Notch1

    Immunity

    (2002)
  • S.I. Samson et al.

    GATA-3 promotes maturation, IFN-γ production, and liver-specific homing of NK cells

    Immunity

    (2003)
  • C.V. Laiosa et al.

    Determinants of lymphoid–myeloid lineage diversification

    Annu Rev Immunol

    (2006)
  • T. Graf

    Transcription factors that induce commitment of multipotent hematopoietic progenitors: lessons from the MEP system

  • N. Rekhtman et al.

    Direct interaction of hematopoietic transcription factors PU.1 and GATA-1: functional antagonism in erythroid cells

    Genes Dev

    (1999)
  • P. Zhang et al.

    Negative cross-talk between hematopoietic regulators: GATA proteins repress PU.1

    Proc Natl Acad Sci USA

    (1999)
  • S.L. Nutt et al.

    Dynamic regulation of PU.1 expression in multipotent hematopoietic progenitors

    J Exp Med

    (2005)
  • K. Tanigaki et al.

    Regulation of lymphocyte development by Notch signaling

    Nat Immunol

    (2007)
  • I. Maillard et al.

    Regulation of lymphoid development, differentiation, and function by the Notch pathway

    Annu Rev Immunol

    (2005)
  • C. Cobaleda et al.

    Pax5: the guardian of B cell identity and function

    Nat Immunol

    (2007)
  • E.V. Rothenberg

    Cell lineage regulators in B and T cell development

    Nat Immunol

    (2007)
  • K. Akashi et al.

    A clonogenic common myeloid progenitor that gives rise to all myeloid lineages

    Nature

    (2000)
  • M. Lu et al.

    The earliest thymic progenitors in adults are restricted to T, NK, and dendritic cell lineage and have a potential to form more diverse TCRβ chains than fetal progenitors

    J Immunol

    (2005)
  • H.Q. Shen et al.

    T/NK bipotent progenitors in the thymus retain the potential to generate dendritic cells

    J Immunol

    (2003)
  • D. Allman et al.

    Thymopoiesis independent of common lymphoid progenitors

    Nat Immunol

    (2003)
  • A. Krueger et al.

    Phenotypic plasticity of T cell progenitors upon exposure to Notch ligands

    J Exp Med

    (2006)
  • J. Huang et al.

    Propensity of adult lymphoid progenitors to progress to DN2/3 stage thymocytes with Notch receptor ligation

    J Immunol

    (2005)
  • A.C. Hayday et al.

    Key factors in the organized chaos of early T cell development

    Nat Immunol

    (2007)
  • H.T. Petrie et al.

    Zoned out: functional mapping of stromal signaling microenvironments in the thymus

    Annu Rev Immunol

    (2007)
  • M.K. Anderson

    At the crossroads: diverse roles of early thymocyte transcriptional regulators

    Immunol Rev

    (2006)
  • Cited by (39)

    • ETV6-NCOA2 fusion induces T/myeloid mixed-phenotype leukemia through transformation of nonthymic hematopoietic progenitor cells

      2022, Blood
      Citation Excerpt :

      MEF2C is expressed in ETV6-NCOA2 PDX cells13 (supplemental Figure 9). The T-cell differentiation and the T/myeloid checkpoint are dependent on accurate regulation of NOTCH1 and PU.1/MEF2C pathways.32-35 For an early thymocyte to differentiate into a mature T cell, the NOTCH1 pathway should be activated whereas the PU.1/MEF2C pathway should be completely shut down (PU.1, MEF2C, and CSF1R).32-36

    • E2Atranscription factors limit expression of Gata3 to facilitate T lymphocyte lineage commitment

      2013, Blood
      Citation Excerpt :

      This hypothesis is consistent with previous studies that reported that GATA3 functions are concentration dependent and may be influenced by interactions with other transcription factors. For example, high concentrations of GATA3 antagonize PU.1 function in DN2s, whereas low concentrations of GATA3 synergize with PU.1 to produce a gene expression signature that allows for T-cell lineage commitment.6,45,46 Additional studies will be necessary to determine how the elevation of GATA3 influences the differentiation and commitment of T-lymphocyte progenitors.

    View all citing articles on Scopus
    View full text