Elsevier

Molecular Immunology

Volume 44, Issue 11, April 2007, Pages 2884-2892
Molecular Immunology

A novel multimeric form of FasL modulates the ability of diabetogenic T cells to mediate type 1 diabetes in an adoptive transfer model

https://doi.org/10.1016/j.molimm.2007.01.014Get rights and content

Abstract

Activation induced cell death (AICD) via Fas/FasL is the primary homeostatic molecular mechanism employed by the immune system to control activated T-cell responses and promote tolerance to self-antigens. We herein investigated the ability of a novel multimeric form of FasL chimeric with streptavidin (SA-FasL) having potent apoptotic activity to induce apoptosis in diabetogenic T cells and modulate insulin-dependent type 1 diabetes (IDDM) in an adoptive transfer model. Diabetogenic splenocytes from NOD/Lt females were co-cultured in vitro with SA-FasL, SA control protein, or alone without protein, and adoptively transferred into NOD/Lt-Rag1null recipients for diabetes development. All animals receiving control (Alone: n = 16 or SA: n = 17) cells developed diabetes on average by 6 weeks, whereas animals receiving SA-FasL-treated (n = 25) cells exhibited significantly delayed progression (p < .001) and decreased incidence (70%). This effect was associated with an increase in CD4+CD25+ T cells and correlated with FoxP3 expression in pancreatic lymph nodes. Extracorporeal treatment of peripheral blood lymphocytes using SA-FasL during disease onset represents a novel approach that may alter the ability of pathogenic T cells to mediate diabetes and have therapeutic utility in clinical management of IDDM.

Introduction

Activation-induced cell death (AICD) via Fas/FasL is an important homeostatic molecular mechanism by which the immune system controls activated T-cell responses, promotes tolerance to self antigens, and prevents autoimmunity. T cells upregulate both Fas and FasL upon activation and become sensitive to autocrine and paracrine apoptosis following repeated reengagement with the challenge antigen (Krammer, 2000, Van Parijs and Abbas, 1998, Ju et al., 1995). The importance of Fas/FasL-mediated apoptosis to tolerance to self antigens is demonstrated in the lpr and gld mice wherein the lack of or low expression of Fas and FasL leads to lymphoproliferative disorders and autoimmunity (Nagata and Suda, 1995). Transgenic replacement of Fas expression in mice restores both AICD and a disease-free state (Wu et al., 1994).

The NOD mouse is a widely used spontaneous model for the study of insulin-dependent type 1 diabetes (IDDM). Although molecular and cellular mechanisms responsible for the development of IDDM in NOD are complex and not fully characterized, activation of autoreactive CD4+ and CD8+ T cells and their recruitment into pancreatic islets play an important role in the initiation of the disease (Bendelac et al., 1987, Yagi et al., 1992). A series of studies reported resistance to AICD in NOD mice which may contribute to the disease process (Decallonne et al., 2003, Leijon et al., 1994, Arreaza et al., 2003). Although the nature of these defects are not fully understood, over 20 susceptibility loci, termed insulin-dependent diabetes (idd), have been identified (Aoki et al., 2005). Some of these defects translate to dysregulated anti-apoptotic gene expression including Bcl-2 (Garchon et al., 1994) and Bcl-x (Lamhamedi-Cherradi et al., 1998), as well as expression of a less apoptotic allele of FasL (Kayagaki et al., 1997).

Despite these claims, a number of studies have reported that selective induction of apoptosis via AICD in diabetogenic lymphocytes can prevent autoimmune diabetes in NOD mice. In vivo administration of Jo-2, an agonistic antibody to Fas, resulted in diabetes remission. Immunohistochemical analysis of pancreata from treated animals demonstrated apoptosis of infiltrating cells, but not insulin and/or glucagon-producing cells, suggesting that remission was due to the induction of AICD in autoreactive lymphocytes (Dharnidharka et al., 2002). In a second study, lipopolysaccharide (LPS)-activated B cells have been shown to express FasL and induce diabetogenic T cells to undergo apoptosis in vitro (Tian et al., 2001). Transfusion of these LPS-activated B cells into prediabetic mice inhibited spontaneous development of diabetes in greater than 80% of the B-cell treated NOD mice at 52 weeks of age. Furthermore, co-transfer of LPS-activated B cells and diabetogenic splenic T cells prevented the disease in an adoptive transfer model. In addition, both membranous as well as soluble forms of human FasL were shown to engage Fas on naïve and diabetogenic T cells, induce apoptosis, and modulate development of diabetes by eliminating autoreactive diabetogenic lymphocytes and pre-activated CD4+CD45RBlow memory T cells (Kim et al., 2000). Of further interest, human soluble FasL was shown to function in vivo to delete potentially autoreactive memory lymphocytes in peripheral blood lymphocytes from mice as well as IDDM patients, complementing membrane FasL in promoting AICD (Kim et al., 2002). Altogether, these studies demonstrate that diabetogenic T cells are sensitive to FasL-mediated apoptosis and as such FasL-based immune intervention may have therapeutic utility for the prevention/treatment of IDDM.

FasL, a type II membranous protein, is expressed on antigen-activated T cells in response to TCR signaling and costimulation (Askenasy et al., 2005). This protein is cleaved off from the cell surface in response to various physiologic stimuli by matrix metalloproteinases (Ju et al., 1995, Tanaka et al., 1998). Traditionally, the membranous form is noted for its ability to induce apoptosis in autoreactive and alloreactive T cells and promote tolerance; in contrast, the soluble form may inhibit apoptosis, initiate inflammatory responses, and promote the active recruitment of neutrophils, thereby accelerating disease or allograft rejection (Ottonello et al., 1999, Seino et al., 1998, Suda et al., 1997). However, this functional difference between membranous and soluble forms of FasL applies to murine FasL more than the human molecule, which appears to have apoptotic function in both forms (Kim et al., 2002, Tanaka et al., 1995).

These diverse functions of soluble and membraneous forms of FasL may be a manifestation of their differential ability to crosslink the Fas receptor upon engagement and the quality/quantity of the ensuing signals (Siegel et al., 2000). We have recently generated a chimeric SA-FasL protein that forms tetramers and higher structures and has potent apoptotic activity on Fas+ cells in soluble form. Immunomodulation with biotinylated donor cells decorated with the chimeric molecule resulted in effective elimination of alloreactive T cells, leading to the survival of transplanted islet and cardiac allografts without any sign of general toxicity or chemotactic function on neutrophils (Askenasy et al., 2005, Yolcu et al., 2002, Askenasy et al., 2003). In this study, we tested whether chimeric SA-FasL can be utilized to modulate autoreactive responses by altering the ability of transferred cells to mediate IDDM. Ex vivo treatment of diabetogenic splenocytes with SA-FasL resulted in significant apoptosis of T cells as well as slower progression and lower incidence of disease in an adoptive transfer model. The observed effect was associated with increased percentage and absolute number of CD4+CD25+ T regulatory cells. The implication of these findings for the use of chimeric FasL to alter autoreactive T cell populations from IDDM patients under extracorporeal conditions as a novel therapeutic intervention is discussed.

Section snippets

Mice and glucose monitoring

Eight weeks old female NOD/Lt mice and NOD/Lt-Rag1null breeder pairs were purchased from The Jackson Laboratory (Bar Harbor, ME) and maintained under specific pathogen-free conditions according to standards and guidelines stipulated by the University of Louisville Institutional Animal Care and Use Committee. NOD/Lt-Rag1null mice for adoptive transfer experiments were used from the breeding colony at 4–6 weeks. NOD/Lt-Rag1null mice lack functional T and B cells and do not spontaneously develop

Ex vivo SA-FasL treatment induces apoptosis in NOD T cells

T cells upregulate Fas and become sensitive to Fas/FasL-mediated apoptosis upon repeated antigenic stimulation (Krammer, 2000, Van Parijs and Abbas, 1998, Ju et al., 1995). Inasmuch as T cells are critical to the induction of diabetes, we reasoned that NOD mice with active disease will have a pool of autoantigen activated T cells that may be sensitive to Fas-mediated apoptosis. This notion was tested by treating splenocytes from diabetic NOD mice with equimolar amounts of SA (control protein)

Discussion

We herein demonstrate that a novel form of chimeric FasL with potent apoptotic activity induced apoptosis in CD4+ T cells without a significant effect on CD8+ T and CD4+CD25+ Treg cell populations from diabetic NOD mice. Ex vivo SA-FasL-treated cells caused delayed onset and decreased incidence of diabetes in an adoptive transfer model. The observed immunomodulatory effect of SA-FasL may operate through inducing apoptosis in pathogenic T cells while sparing CD4+CD25+ Treg cells. A limited

Acknowledgements

This research was supported by grants from NIH DK61333, AI47864, AI57903, Juvenile Diabetes Research Foundation (1-2001-328), American Diabetes Association (1-05-JF-56), and the Commonwealth of Kentucky Research Challenge Trust Fund. Additionally, we would like to thank Heather Stowers, Lisa Colbert, Janice Ditslear, and Victoria Kyle for their technical support and maintenance of our animal colony and Sarah Parnell for her time and technical expertise in executing the real-time PCR analyses.

References (53)

  • X.X. Zheng et al.

    Favorably tipping the balance between cytopathic and regulatory T cells to create transplantation tolerance

    Immunity

    (2003)
  • N. Askenasy et al.

    Display of Fas ligand protein on cardiac vasculature as a novel means of regulating allograft rejection

    Circulation

    (2003)
  • A. Banz et al.

    Modulation of fas-dependent apoptosis: a dynamic process controlling both the persistence and death of CD4 regulatory T cells and effector T cells

    J. Immunol.

    (2002)
  • A. Bendelac et al.

    Syngeneic transfer of autoimmune diabetes from diabetic NOD to healthy neonates

    J. Exp. Med.

    (1987)
  • M.A. Curotto de Lafaille et al.

    CD25-T cells generate CD25 + Foxp3+ regulatory T cells by peripheral expansion

    J. Immunol.

    (2004)
  • V.R. Dharnidharka et al.

    Fas stimulation results in selective islet infiltrate apoptosis in situ and reversal of diabetes

    Ann. N. Y. Acad. Sci.

    (2002)
  • D.T. Felson et al.

    The Prosorba column for treatment of refractory rheumatoid arthritis: a randomized, double-blind, sham-controlled trial

    Arthritis Rheum.

    (1999)
  • T.A. Ferguson et al.

    Apoptosis, tolerance, and regulatory T cells—old wine, new wineskins

    Immunol. Rev.

    (2003)
  • B. Fritzsching et al.

    In contrast to effector T cells, CD4 + CD25 + FoxP3+ regulatory T cells are highly susceptible to CD95 ligand- but not to TCR-mediated cell death

    J. Immunol.

    (2005)
  • H.J. Garchon et al.

    Genetic analysis of immune dysfunction in non-obese diabetic (NOD) mice: mapping of a susceptibility locus close to the Bcl-2 gene correlates with increased resistance of NOD T cells to apoptosis induction

    Eur. J. Immunol.

    (1994)
  • M.A. Gavin et al.

    Homeostasis and anergy of CD4 + CD25+ suppressor T cells in vivo

    Nat. Immunol.

    (2002)
  • E.A. Green et al.

    CD4 + CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-β- TGF-β receptor interactions in type 1 diabetes

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • S.T. Ju et al.

    Fas(CD95)/FasL interactions required for programmed cell death after T-cell activation

    Nature

    (1995)
  • N. Kayagaki et al.

    Polymorphism of murine Fas ligand that affects the biological activity

    Proc. Natl. Acad. Sci. U.S.A.

    (1997)
  • S. Kim et al.

    Soluble Fas ligand-susceptible “memory” cells in mice but not in human: Potential role of soluble Fas ligand in deletion of auto-reactive cells

    Autoimmunity

    (2002)
  • S. Kim et al.

    Inhibition of autoimmune diabetes by Fas ligand: the paradox is solved

    J. Immunol.

    (2000)
  • Cited by (18)

    • Localized immune tolerance from FasL-functionalized PLG scaffolds

      2019, Biomaterials
      Citation Excerpt :

      Two lines of evidences from our own studies support this notion. First, we have demonstrated that Treg cells in NOD are preferentially more resistant to SA-FasL as compared with Teff cells [25]. Second, in collaboration with Askenasy and group, we have shown that sorted Treg cells modified to display SA-FasL on their surface have improved regulatory function and following adoptive transfer into prediabetic NOD prevented the development of diabetes [65,66].

    • Enhanced killing activity of regulatory T cells ameliorates inflammation and autoimmunity

      2013, Autoimmunity Reviews
      Citation Excerpt :

      We focused on three disease models: non-obese diabetic mice (NOD) as a genetic model of human type 1 diabetes (T1D) [33,34], toxic colitis as a model of chronic inflammatory bowel disease (IBD) [35], and graft versus host disease (GvHD) as a model of inflammation caused by adoptive transfer of T cells [36]. These models were chosen because T1D and IBD are characterized by relative resistance of pathogenic cells to negative regulation, but are sensitive to AICD by Fas cross-linking [37,38]. These models have been effectively treated by elimination of effector cells using a fusion protein targeting the IL-2 receptor to internalize active caspase-3 (IL2-cas) [39–41].

    • Immunomodulation with SA-FasL protein as an effective means of preventing islet allograft rejection in chemically diabetic NOD mice

      2013, Transplantation Proceedings
      Citation Excerpt :

      Complete lack of allogeneic islet rejection during the course of the 250-day observation period provides important evidence for the efficacy of our immunomodulatory regimen for the control of not only allo, but also autoreactive immunity. This prediction is consistent with our previous observations that SA-FasL protein preferentially induces apoptosis in autoreactive T-effector cells in NOD while sparing CD4+CD25+FoxP3+ T-regulatory (Treg) cells.11 Although we do not know the mechanistic basis of long-term allogeneic islet survival in the present study, it is tempting to speculate that active immune regulation, plausibly by Treg cells, may play a role.

    • Depletion of Naïve Lymphocytes with Fas Ligand Ex Vivo Prevents Graft-versus-Host Disease without Impairing T Cell Support of Engraftment or Graft-versus-Tumor Activity

      2013, Biology of Blood and Marrow Transplantation
      Citation Excerpt :

      Presentation of antigens in conjunction with apoptotic signals eliminates activated cells [46] and has been used successfully for antigen-specific abrogation of immune responses in vivo [38,47-50]. This simulation of physiologic AICD has also been implemented for ex vivo elimination of activated cells from donors with a pre-existing autoimmune disease such as diabetes [51] and selective depletion of host-sensitized donor T cells [8,9,11,12]. Clinical implementation of this method of allosensitization and depletion ex vivo is cumbersome and involves mixed lymphocyte culture of the graft with cells that present the host alloantigen.

    • Killer Treg restore immune homeostasis and suppress autoimmune diabetes in prediabetic NOD mice

      2011, Journal of Autoimmunity
      Citation Excerpt :

      Fourth, FasL can be employed for immunomodulation in conjunction with other strategies aiming to sensitize, expand and enhance the suppressive activity of Treg to arrest autoimmunity [16,26–28]. FasL is a physiological constituent of apoptotic signals transduced by human and murine adaptive Treg cells [13] and has the significant advantage of selective and antigen-specific elimination of autoreactive and alloreactive effector cells [18,19,23,32,53]. Finally, the most prominent feature of CD25+-FasL immunomodulation is to create a substantial disease-free window of opportunity during which additional approaches to definitive arrest of autoimmunity might be implemented prior to institution of therapies to replenish the damaged tissue [54].

    View all citing articles on Scopus
    View full text