Central role of ILT3 in the T suppressor cell cascade

https://doi.org/10.1016/j.cellimm.2007.01.013Get rights and content

Abstract

CD8+ T suppressor cells differentiate both in vivo and in vitro upon chronic exposure of responding T cells to allogeneic APC. These Ts are allospecific and exhibit their function interacting directly with priming APC which they render tolerogenic. Tolerogenicity of professional and non-professional human APC, such as dendritic cells and endothelial cells, respectively is due to the upregulation of the inhibitory receptors ILT3 and ILT4. ILT3 signals both intracellularly, inhibiting NF-κB activation, and transcription of costimulatory molecules, and extracellularly, inducing anergy and regulatory function in T cells with cognate specificity. Both membrane and soluble ILT3 are proteins with potent immunosuppressive activity which are of importance for treatment of rejection, autoimmunity and cancer.

Introduction

The immune system has developed several checkpoints and regulatory mechanisms to discriminate between self and non-self antigens and avoid autoimmunity [1]. Included among them are: (1) negative selection of auto-reactive T cells in the thymus [2]; (2) elimination of auto-reactive T cells via activation-induced cell death (AICD) [3], [4], [5], [6], [7]; (3) induction of anergy upon TCR triggering in the absence of co-stimulation [8], [9], [10], [11], [12]; (4) inhibition of immune function by T suppressor cells [13], [14], [15], [16], [17], [18], [19], [20].

Research on T suppressor cells re-emerged in the late 1990s when several subsets of T cells were shown to inhibit the proliferation of other T cells. Two broad categories of regulatory T (Treg) cells have been recognized. The first consists of naturally occurring and the second, of induced Treg [13], [14], [15], [16], [17], [18], [19], [20].

The naturally occurring CD4+CD25+ Treg subset is generated in the thymus as a functionally distinct subpopulation of T cells [13], [14], [15], [16], [17], [18], [19], [20]. These natural Treg cells play a major role in regulating self-reactive T cells and preventing autoimmune diseases. The transfer of T cell populations, from which the CD25+ subset has been depleted, into T cell-deficient mice caused severe autoimmune disorders, such as thyroiditis, gastritis, insulin-dependent diabetes mellitus and colitis [16], [19], [20]. Conversely, infusion of the Treg cells in these animals strongly suppressed autoimmunity. Recent evidence indicates that the transcription factor FOXP3 acts as a “master control gene” for the development and function of Treg [21], [22], [23]. A mutation in the gene encoding FOXP3 was identified as the genetic defect underlying autoimmune and inflammatory disease in scurfy mice and in humans with IPEX (immune dysregulation, polyendocrinopathy, enteropathy X-linked syndrome) and XLAAD (X-linked autoimmunity allergic dysregulation syndrome), emphasizing the importance of CD4+CD25+ Treg in the maintenance of normal immune homeostasis [21], [22], [23].

FOXP3 is a member of the forkhead–winged helix family of transcription factors. Retroviral mediated expression of FOXP3 converts naive T cells into CD25+ Treg. Thus, FOXP3 induces either directly or indirectly the expression of Treg-associated molecules. While in mice FOXP3 is stably expressed by CD4+CD25+ Treg and cannot be induced by activation of CD4+CD25 T cells, in humans it can be induced by activation and exposure to TGF-beta [24].

CD4+CD25+ T cells suppress activation and proliferation of naive CD4+ T cells in vitro through T-cell-to-T-cell contact [20], [21]. Treg cells are in an anergic state and do not proliferate in response to antigens (Ag) presented by MHC class II molecules [14], [15], [16], [17], [18], [19], [20], [21]. Triggering of Treg cells by anti-CD3 plus anti-CD28 monoclonal antibodies (mAbs) or exposure to IL-4 breaks the anergic state and abrogates the suppressor function of Treg [24], [25]. Also, triggering by an agonistic Ab to the cell surface receptor glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18), which is constitutively expressed on the surface of Treg cells [26], [27] abrogates the suppressor activity of Treg. IL-2 is mandatory for the in vitro activation of CD4+CD25+ Treg cells [28].

The mechanisms by which natural CD4+CD25+ Treg cells act in different autoimmune disease models seem to involve a discretionary requirement for the cytokines IL-10 and TGF-beta [17], and may depend on signals through CTLA-4 [29], [30], as well as regulation coordinated by GITR [26], [27].

Induced Treg cells are divided into two categories, one consisting of non-antigen-specific CD4+ and CD8+ Treg and the other of antigen-specific CD8+ T suppressor cells (TS) and CD4+CD25+ Treg. Non-antigen-specific regulatory CD4+CD25+ T cells can be generated from naïve peripheral CD25 precursors under certain culture conditions, such as by exposure to IL-10 and IFN-alpha or TGF-beta, or by stimulation with immature myeloid dendritic cells (DC) or mature plasmacytoid DC [11], [31], [32], [33], [34]. Activation, in vitro or in vivo, of human or mouse CD4+ T cells in the presence of IL-10 results in the generation of T-cell clones which produce significant amounts of IL-10, IFN-alpha, TFG-beta and IL-5. These T-cell clones, named TR1, inhibit Ag-induced activation of naïve autologous T cells via a mechanism which is partially mediated by IL-10 and TGF-beta [31], [32]. T cells with a TR1 cytokine profile have been described in several models of autoimmune diseases and transplantation [35], [36], [37], [38], [39]. In most cases, TR1 cells arise following repeated Ag stimulation either in vitro or in vivo[35]. However, because they cause bystander suppression, their inhibitory effect on other T cells is not antigen-specific.

The extent to which cell contact or soluble factors are required for TR1-mediated suppression of TH reactivity is still unknown. Experiments in which cytokine production was excluded, first by activating and then fixing naturally occurring CD4+CD25+ Treg cells, showed that these cells maintain their capacity to suppress normally responding CD4+CD25 T cell populations rendering them anergic. The newly anergized population further suppressed syngeneic CD4+ T cells via the production of inhibitory cytokines [40], [41]. It was suggested that suppression occurs in two sequential steps: the first one is a cell contact-dependent ‘transmission’ of anergy from a regulatory T cell to another T lymphocyte, while the second is a cell contact-independent, cytokine mediated suppression of other T helper cells [40], [41], [42].

Other non-antigen-specific regulatory T cell subtypes have been described including gamma–delta T cells, NK1.1 T cells, CD8+CD25+ and CD4CD8 T cells, but they remain poorly characterized [14].

A distinct category of regulatory T cells characterized by their antigen-specific activity and mechanism of action are CD8+CD28 TS cells first described in humans by our group. We showed that CD8+CD28 TS specific for alloantigens, xenoantigens or nominal antigens could be generated in vitro by repeated antigenic stimulation [43], [44], [45], [46], [47], [48], [49], [50], [51]. Next, we provided in vivo evidence that CD8+CD28 T cells act as antigen-specific suppressors in patients with heart, kidney or liver allografts [52], [53], [54], [55], [56]. We further demonstrated that antigen-specific CD8+CD28 TS express FOXP3, derive from an oligoclonal population of CD8+ FOXP3 cells, are MHC class I restricted, have no killing capacity and do not produce cytokines [43], [55], [56]. Instead they act on professional (DC) and non-professional (endothelial cells) APC directly i.e., by cell-to-cell contact, inducing qualitative changes characteristic of an alternative pathway of maturation toward a tolerogenic rather than immunogenic phenotype. These changes include the downregulation of NF-κB-dependent costimulatory molecules such as CD40, CD58, CD80, CD86, and the upregulation of inhibitory receptors, immunoglobulin like transcript 3 (ILT3) and ILT4 [48], [49], [50], [55], [56]. CD4+ TH, which interact with tolerogenic APC, become anergic and acquire regulatory activity [13], [55]. These data support a model in which T cell mediated suppression results from the sequential interaction between first, CD8+ Ts and APC and next, “tolerized” APC and CD4+ TH. In turn, anergic TH acquire regulatory capacity, in conjunction with FOXP3 expression, further perpetuating tolerance [13], [56]. The central role of ILT3high, ILT4high APC in the induction of suppression was further demonstrated in experiments in which the upregulation of these inhibitory receptors in DC and endothelial cells (EC) was induced by treating the cells with IL-10 plus IFN-alpha or IL-10 plus vitamin D3 [13], [55], [56]. Such ILT3high ILT4high APC induced the in vitro generation of TS and Treg from unprimed populations of CD4 and CD8 T cells [13], [55], [56].

Progress in understanding the mechanisms of T-cell activation and inactivation is currently being translated into strategies, enabling induction of selective immunosuppression for treatment of autoimmune diseases, allergies and allograft rejection. There is an imperative need for antigen-specific immunosuppression, as systemic immunosuppression is associated with an increased risk of malignancies, infections and considerable toxicity. Progress in the generation and characterization of antigen-specific TS, Treg cells and tolerogenic APC may pave the way to the induction of immunologic tolerance.

Section snippets

Molecular characterization of CD8+CD28 Ts

To gain insight into the common denominators of antigen-specific and non-specific Treg cells we have analyzed some of their characteristics at the molecular level. We compared by RT-PCR the expression of genes, known to be upregulated in natural CD4+CD25+ Treg from fresh peripheral blood [21], [26], [27], [56], [57], [58], [59], [60], [61], [62], [63] with their expression in allospecific CD8+CD28 TS from T cell lines (TCL) [64], [65]. The following genes were studied: CD25, GITR, CTLA-4,

Molecular and functional events resulting from TS mediated suppression

Molecular changes induced by CD8+ TS in DC were analyzed using Affymetrix gene chips. The overall picture that emerged was that tolerogenic DC differ from both immature and mature DC with respect to molecules involved in signal transduction, chemokines, cytokines, transcription factors, apoptosis-related proteins and cell growth regulators [49], [50]. Most importantly, mRNA expression profiling showed that tolerogenic DC exhibit a high cell surface expression of the inhibitory molecules ILT3

Evaluation of the clinical significance and therapeutic potential of allospecific TS

We have explored the in vivo relevance of CD8+CD28 TS and of CD4+CD25+ Treg in recipients of heart, kidney or liver transplants [52], [53], [54], [55], [56]. Serial studies of the phenotype displayed by T cells from heart allograft recipients demonstrated a significant increase of the CD8+CD28CD27+ perforin negative T-cell population in rejection-free patients [52], [53], [54], [55], [56]. This phenotype is characteristic of in vitro generated TS as demonstrated by flow cytometry and cDNA

CD8+ FOXP3+ T suppressor cells transfer allogeneic tolerance in rodents and induce PIR-B in donor APC

Recently, evidence has been provided that the paired immunoglobulin like receptor (PIR)-B expressed on B cells and myeloid cells of rodents shares structural and functional characteristics with ILT4 [72]. PIR-B, which belongs to a MHC class I recognition system, contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in its cytoplasmic domain and can inhibit receptor-mediated activation signaling similar to its human orthologue ILT4 [73]. Transfer of allogeneic splenocytes into PIR-B

Immunoregulatory activity of membrane and soluble ILT3

Effective priming of CD4+ T lymphocytes requires cell-to-cell interaction between APC and T cells. This interaction allows bi-directional costimulatory signals which activate both the APC and T cells with cognate-specific TCR. Triggering of TCR induces upregulation of CD40L expression on activated CD4+ T cells. The interaction of CD40L with CD40 expressed by APC induced the upregulation of costimulatory molecules that interact with their counter receptor on CD4+ Th cells, eliciting their

Acknowledgments

This work was supported by a grant from the NIH RO1 AI55234-04 and the Interuniversitary Organ Transplantation Consortium, Rome, Italy

References (102)

  • A.I. Colovai et al.

    Regulatory CD8+ CD28− T cells in heart transplant recipients

    Hum. Immunol.

    (2003)
  • R. Ciubotariu et al.

    Detection of T suppressor cells in patients with organ allografts

    Hum. Immunol.

    (2001)
  • R. Cortesini et al.

    Tailoring of immunosuppression in renal and liver allograft recipients displaying donor specific T-suppressor cells

    Hum. Immunol.

    (2002)
  • J.S. Manavalan et al.

    High expression of ILT3 and ILT4 is a general feature of tolerogenic dendritic cells

    Transpl. Immunol.

    (2003)
  • S. Fu et al.

    CD4+ CD25+ CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential

    Am. J. Transplant.

    (2004)
  • N. Suciu-Foca et al.

    Molecular characterization of allospecific T suppressor and tolerogenic dendritic cells: review

    Int. Immunopharmacol.

    (2005)
  • L. Scotto et al.

    Overlap between molecular markers expressed by naturally occurring CD4+ CD25+ regulatory T cells and antigen specific CD4+ CD25+ and CD8+ CD28− T suppressor cells

    Hum. Immunol.

    (2004)
  • M. Colonna et al.

    A family of inhibitory and activating Ig-like receptors that modulate function of lymphoid and myeloid cells

    Semin. Immunol.

    (2000)
  • H. Waldmann et al.

    Regulating the immune response to transplants. A role for CD4+ regulatory cells?

    Immunity

    (2001)
  • S. Kim-Schulze et al.

    Regulation of ILT3 gene expression by processing of precursor transcripts in human endothelial cells

    Am. J. Transplant.

    (2006)
  • J. Liu et al.

    Rat CD8+ FOXP3+ T suppressor cells mediate tolerance inducing to allogeneic heart transplants PIR-B in APC and rendering the graft invulnerable to rejection

    Transpl. Immunol.

    (2004)
  • S. Cobbold et al.

    Infectious tolerance

    Curr. Opin. Immunol.

    (1998)
  • L. Adorini et al.

    Pharmacological induction of tolerogenic dendritic cells and regulatory T cells

    Semin. Immunol.

    (2004)
  • E.H. Preston et al.

    IDEC-131 (anti-CD154), sirolimus and donor-specific transfusion facilitate operational tolerance in non-human primates

    Am. J. Transplant.

    (2005)
  • E. Gonzalez-Rey et al.

    Regulation of immune tolerance by anti-inflammatory neuropeptides

    Nat. Rev. Immunol.

    (2007)
  • M.W. Hoffmann et al.

    Deletion of high-avidity T cells by thymic epithelium

    Proc. Natl. Acad. Sci. USA

    (1995)
  • O. Janssen et al.

    Regulation of activation-induced cell death of mature T-lymphocyte populations

    Cell Tissue Res.

    (2000)
  • T. Nguyen et al.

    The regulation of FasL expression during activation-induced cell death (AICD)

    Immunology

    (2001)
  • L.A. Conroy et al.

    The role of intracellular signalling pathways regulating thymocyte and leukemic T cell apoptosis

    Leukemia

    (1996)
  • H.H. Oberg et al.

    Ligation of cell surface CD4 inhibits activation-induced death of human T lymphocytes at the level of Fas ligand expression

    J. Immunol.

    (1997)
  • K.M. Lee et al.

    Molecular basis of T cell inactivation by CTLA-4

    Science

    (1998)
  • A.D. Wells et al.

    Signaling through CD28 and CTLA-4 controls two distinct forms of T cell anergy

    J. Clin. Invest.

    (2001)
  • N. Shirasugi et al.

    Prevention of chronic rejection in murine cardiac allografts: a comparison of chimerism- and nonchimerism-inducing costimulation blockade-based tolerance induction regimens

    J. Immunol.

    (2002)
  • C. Nagler-Anderson et al.

    Control freaks: immune regulatory cells

    Nat. Immunol.

    (2004)
  • S. Sakaguchi et al.

    Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases

    J. Immunol.

    (1995)
  • E.M. Shevach et al.

    Control of T-cell activation by CD4+ CD25+ suppressor T cells

    Immunol. Rev.

    (2001)
  • S. Sakaguchi et al.

    Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance

    Immunol. Rev.

    (2001)
  • M. Itoh et al.

    Thymus and autoimmunity: production of CD25+CD4+ naturally anergic and suppressive T cells as a key function of the thymus in maintaining immunologic self-tolerance

    J. Immunol.

    (1999)
  • E.M. Shevach

    Regulatory T cells in autoimmmunity

    Annu. Rev. Immunol.

    (2000)
  • S. Hori et al.

    Control of regulatory T cell development by the transcription factor Foxp3

    Science

    (2003)
  • R. Khattri et al.

    An essential role for Scurfin in CD4+CD25+ T regulatory cells

    Nat. Immunol.

    (2003)
  • J.D. Fontenot et al.

    Foxp3 programs the development and function of CD4+CD25+ regulatory T cells

    Nat. Immunol.

    (2003)
  • T. Takahashi et al.

    Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state

    Int. Immunol.

    (1998)
  • J. Shimizu et al.

    Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance

    Nat. Immunol.

    (2002)
  • A.M. Thornton et al.

    Cutting edge: IL-2 is critically required for the in vitro activation of CD4+CD25+ T cell suppressor function

    J. Immunol.

    (2004)
  • S. Read et al.

    Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation

    J. Exp. Med.

    (2000)
  • C.I. Kingsley et al.

    CD25+CD4+ regulatory T cells prevent graft rejection: CTLA-4- and IL-10-dependent immunoregulation of alloresponses

    J. Immunol.

    (2002)
  • M.K. Levings et al.

    The role of IL-10 and TGF-beta in the differentiation and effector function of T regulatory cells

    Int. Arch. Allergy Immunol.

    (2002)
  • M.K. Levings et al.

    IFN-alpha and IL-10 induce the differentiation of human type 1 T regulatory cells

    J. Immunol.

    (2001)
  • M.G. Roncarolo et al.

    Differentiation of T regulatory cells by immature dendritic cells

    J. Exp. Med.

    (2001)
  • Cited by (53)

    • Role of regulatory T cells in cancer

      2022, Role of Tumor Microenvironment in Breast Cancer and Targeted Therapies
    • Induction of CD4 <sup>+</sup> CD25 <sup>+</sup> Foxp3 <sup>+</sup> T regulatory cells by dendritic cells derived from ILT3 lentivirus-transduced human CD34 <sup>+</sup> Cells

      2012, Transplant Immunology
      Citation Excerpt :

      Ideal tolerogenic DCs can be applied to induce the acceptance of organ/tissue allogeneic transplants or to prevent autoimmune diseases. Numerous studies have reported that gene-modified DCs can control cellular functions, alter immune responses [18,21], and that high expression of ILT3 and ILT4 is a general feature of tolerogenic DCs [22]. ILT3 and ILT4 contain ITIMs in their cytoplasmic tail [23], leading to downstream events and gene modulation.

    View all citing articles on Scopus
    View full text