Department of Pathology and Laboratory Medicine, University of Pennsylvania, 6 Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
Correspondence to: M. L. Tykocinski
![]() |
Abstract |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Keywords: apoptosis, cell-cell interactions, human, T lymphocytes
![]() |
Introduction |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The Fas (CD95) receptor on T cell surfaces participates in activation-induced cell death (3). Fas ligand (FasL; CD95L), which binds to Fas, is a member of the tumor necrosis factor superfamily and is expressed by a variety of immune cells, including monocytes, NK cells, and activated B and T cells (46). FasL is a trimeric, type II membrane-associated protein (7,8) that is released as soluble FasL (sFasL) trimers from membranes by metalloproteinase cleavage (911).
Several observations have prompted us to devise FasL protein derivatives that can associate with cell membranes. Membrane-associated FasL induces apoptosis more effectively than does sFasL (1216), possibly stemming from the higher functional valency expected for membrane-associated ligands, as compared to their soluble forms. Indeed, experiments with different sFasL proteins support the notion that FasL's functional valency dictates its potency. For example, whereas naturally processed, trimeric sFasL induces apoptosis relatively poorly, a longer recombinant sFasL variant incorporating the entire FasL ectodomain forms aggregates of trimers which display higher apoptotic activity (12,17). In fact, for non-aggregating recombinant sFasL trimers, antibody cross-linking is essential for the lysis of Fas-sensitive cells (13).
Additional potential benefits for membrane-associated FasL, as opposed to sFasL, relate to toxicity. When administered systemically, aggregated sFasL (18,19), as well as Fas-specific antibody (20), are toxic in vivo, causing rapid-onset liver damage. Hence, while in vivo loss of FasFasL manifests as lymphoproliferation and autoimmune disease syndromes in both mice and humans, exaggerated sFasL function is implicated in tissue destruction (21). Membrane association represents a reasonable strategy for bypassing the toxic effects of sFasL. This is supported by the finding that transfected FasL on APC surfaces is associated with less systemic toxicity, while maintaining desired FasL-mediated function (22,23).
CD28 is a co-stimulator receptor on T cells that is triggered by B7 surface proteins (B7-1/CD80 and B7-2/CD86) of APC. A second receptor for B7 proteins, cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), is a type I, disulfide-linked homodimeric glycoprotein that emerges on activated T cells and functions as an inhibitory receptor (2426). CTLA-4Ig, a soluble fusion protein in which the CTLA-4 ectodomain is linked to the Fc domain of IgG1, competitively blocks B7:CD28 interaction (27,28), and in so doing, suppresses T cell immunopathogenesis in animal models of alloimmune (2933) and autoimmune (34,35) diseases. Given the soluble CTLA-4's well-documented immunoregulatory activity, it represents an attractive APC `homing element' to be appended to sFasL derivatives.
Previously, we have demonstrated that glycosyl-phosphatidylinositol (GPI)-modified derivatives of proteins are amenable to exogenous re-incorporation into membranes and can be used for the engineering, or painting, of APC surfaces (2,3638). However, since GPI anchors are appended to the C-termini of proteins, type II membrane proteins such as FasL, with their functional structures positioned near their C-termini, cannot be made as functional GPI derivatives. In this study, we report an alternative class of membrane-associating fusion proteins that is suited for type II membrane proteins and combines APC binding potential along with the additional capacity to send trans signals to interacting T cells. The model protein described here is CTLA-4FasL (CD152CD95L), which both associates with, and thereby masks, B7 co-stimulators on APC membranes, and simultaneously delivers FasL inhibitory signals in trans to T cells. The rationale for linking B7 blockade and FasL inhibition is made more compelling by findings that CTLA-4Ig increases T cell susceptibility to Fas-dependent apoptosis and B7 counteracts FasL's apoptosis-inducing activity (3942). The present data establish that CTLA-4FasL's T cell inhibitory function is substantially augmented when this fusion protein is anchored on APC surfaces. Moreover, CTLA-4FasL's inhibitory activity exceeds that of the extensively studied CTLA-4Ig fusion protein, which has well-documented therapeutic potential.
![]() |
Methods |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
A ß2-microglobulin (ß2m)FasL expression construct was produced in several steps. First, the human ß2m cDNA sequence was PCR-amplified from pß2m/REP12ß. The primers used for this amplification (5'-TTGGGGTACCATGTCTCGCTCCGTGG-3' and 5'-AAAGGATCCAAGCTTTCCATGTCTCGATCCCACTT-3') were designed to replace the stop codon of human ß2m with a HindIII restriction site. Following DNA purification, the 360 bp ß2m PCR product was digested with KpnI and HindIII, and subcloned into the respective sites of pCEP9ß, yielding pß2mX/CEP9ß. Human FasL sequence was mobilized from pXFasL/CEP9ß using HindIII and BamHI, and this 480 bp DNA fragment was subcloned into the respective sites of pß2mX/CEP9ß to generate pß2mFasL/CEP9ß. Partially overlapping synthetic oligonucleotides (5'-AGCTTAGGT-GGTGGTTCTGGTGGTGGTTCTGACTACAAGGACGACGA-3' and 5'-AGCTACCTCCTCCAGATCCTCCTCCCTTGTCATCGTCGTCCTTGTAGT-3'), encoding the Flag epitope tag flanked by a linker sequence, (GGGS)2DYKDDDDK(GGGS)2, were subcloned into the HindIII site of pß2mFasL/CEP9ß, yielding pß2mFlagFasL/CEP9ß. All constructs were confirmed by DNA sequencing.
Transfectant cell lines were generated by introducing the various EBV episomal expression constructs (within the pREP and pCEP vector backbones) into 293 cells, using Lipofectin reagent (Life Technologies, Bethesda, MD). Selection of stable transfectants was performed in D10 growth medium (vide infra) supplemented with 0.4 mg/ml G418 (Life Technologies). Multiple individual colonies were picked and screened for recombinant protein expression.
Cell culture
Human cell lines obtained from the ATCC (Rockville, MD) included Jurkat T cells, 293 embryonic kidney cells, and Raji and Daudi EBV-transformed B cell lines. The EBV-transformed B cell line, JY, was provided by Dr F. Chisari (Scripps Institute). All cells were maintained in humidified incubators providing 5% CO2. RPMI 1640 and high glucose DMEM media, antibiotics, glutamine, and FBS were obtained from Biowhittaker (Bethesda, MD). Jurkat cells were cultured in RPMI 1640 supplemented with 100 µg/ml penicillin, 100 U/ml streptomycin, 2 mM glutamine and 10% heat-inactivated FBS (together designated as R10 medium). The 293 cells were cultured in DMEM supplemented with 100 µg/ml penicillin, 100 U/ml streptomycin, 2 mM glutamine and 10% heat-inactivated FBS (together designated as D10 medium). Conditioned media were prepared by culturing cells for 56 days in Cellgro-FREE supplemented with penicillin/streptomycin and L-glutamine (Mediatech, Herndon, VA). Cellular debris was removed from cell culture supernatants by centrifugation (6 min at 400 g) followed by filtration through 0.2 µm sterile syringe filters. Clarified supernatants were stored at 4°C for 2 months without significant loss of activity. A commercially available solid-phase assay (MBL, Cambridge, MA) was used to quantitate sFasL in samples.
Human peripheral blood mononuclear cells (PBMC) were isolated from healthy volunteers by Ficoll-Hypaque (Pharmacia Biotech, Uppsala, Sweden). Proliferation assays with human PBMC were performed in 96-well U-bottom tissue culture plates. The assay medium consisted of R10 supplemented with 25 mM HEPES pH 7.4 (R10-H). Assays were performed in triplicate. Purified CTLA-4FasL (vide infra), CTLA-4Ig (43) or commercial sFasL (Alexis Biochemicals, San Diego, CA) were diluted in R10-H, and 50 µl of each diluted protein sample, along with 2x105 PBMC in 100 µl, was added per well. Cultures were then pre-incubated at 37°C for 3060 min, prior to the addition of graded amounts of mitogenic anti-CD3 mAb (OKT3) in 50 µl R10-H. At 22 h and every 24 h thereafter, culture wells were pulsed with 25 µl of R10-H containing 0.5 µCi of [methyl-3H]thymidine (ICN, Chicago, IL) for 18 h and then harvested onto glass fiber filters for scintillation counting.
Immunoaffinity purification
Immunoaffinity beads specific for human FasL were prepared according to a standard protocol. First, 0.1 ml of Protein Aagarose beads (Life Technologies, Bethesda, MD) was sequentially loaded with 100 µg of rabbit anti-mouse Ig and after washing, with 50 µg of the mouse anti-human FasL mAb NOK-1 (PharMingen, La Jolla, CA). After washing, antibodies were cross-linked to beads in 0.2 M sodium borate, pH 9.0, with 5 mg/ml of dimethyl pimelimidate (Pierce, Rockford, IL). Chemical cross-linking reactions were quenched with 0.2 M ethanolamine, pH 8.0, for 2 h, and the beads were washed with PBS containing 0.05% sodium azide and then stored at 4°C.
Immunoaffinity purification of CTLA-4FasL was performed in batches. An aliquot of 0.1 ml of the FasL-specific beads was added to cell culture supernatants containing CTLA-4FasL and rocked overnight at 4°C. Beads were collected by centrifugation and washed with 0.2 M sodium phosphate, pH 6.8. CTLA-4FasL was batch-eluted from the beads with 0.1 M citrate, pH 2.5, and transferred into tubes containing 1 M phosphate, pH 8.0, for neutralization. Purified CTLA-4FasL was quantitated by the commercial sFasL solid-phase assay cited above.
Gel filtration chromatography and Western blotting
For analyses of molecular size and stoichiometry, clarified cell culture supernatants containing CTLA-4FasL were concentrated 15- to 30-fold with Centricon-10 ultrafiltration devices (Amicon, Beverly, MA). The retentate was applied to a pre-calibrated Superdex-200 (Pharmacia Biotech) column in 1xPBS, and 0.5 ml fractions were collected and analyzed using a sFasL ELISA. Fractions containing active CTLA-4FasL were concentrated 50-fold and then co-incubated with the chemical cross-linker dithiobis(succinimidyl proprionate) (DSP; Pierce) at room temperature for 30 min, prior to quenching with 0.1 M Tris base/0.1M glycine. Samples for SDSPAGE analysis were prepared in Laemmli sample buffer, with or without 2-mercaptoethanol (2-ME), and were boiled prior to loading onto 10% SDSPAGE gels. Following electrophoresis, gels were blotted onto Immobilon P membranes (Millipore, Beverly, MA), blocked with 5% milk/PBS and probed with anti-human FasL antiserum (anti-C20; Santa Cruz Biologicals, Santa Cruz, CA). After extensive washing, blots were incubated with horseradish peroxidase-conjugated anti-rabbit antiserum (BioRad, Richmond, CA) and developed with enhanced chemiluminescent substrate (NEN, Boston, MA) prior to exposure to X-ray film.
Flow cytometry
Cells were pre-incubated at 37°C for 3045 min with various dilutions of cell culture supernatants containing either ß2mFasL or CTLA-4FasL. These cells were then pelleted, washed twice with FACS buffer (1xPBS/0.5% BSA/0.02% sodium azide) and incubated on ice for 3045 min with 10 µg/ml of primary anti-human FasL mAb (NOK-1; PharMingen) or anti-human CTLA-4 mAb (BNI3; PharMingen), with mouse IgG1 (Dako, Carpenteria, CA) serving as a negative control. Cells were then incubated on ice for 3045 min with FITC-conjugated goat F(ab')2 anti-mouse IgG (Boerhinger Mannheim, Indianapolis, IN). Following washes, fluorescence-labeled cells were run on a FACScan and propidium iodide-excluding populations were gated for analysis with the CellQuest software package (Becton Dickinson, Mountain View, CA).
Cytotoxicity assays
Cytotoxicity assays were performed in triplicate as described (44), with a few modifications. Jurkat cells in exponential growth phase were labeled at 37°C for 45 h with 3 µCi/ml of [3H]thymidine (ICN, Costa Mesa, CA) in R10 medium, pelleted and washed twice. Cells were resuspended in R10 at 2x105/ml, and 0.1 ml of the cell suspension was added to each well. Clarified supernatants containing CTLA-4FasL or ß2mFasL were diluted into R10 and 0.1 ml of each diluted sample was added to individual wells of round-bottom 96-well tissue culture plates. Anti-Flag mAb (Sigma) was added as cross-linker at 0.5 µg/ml to indicated samples. Pre-incubation of cells with defined quantities of CTLA-4FasL or ß2mFasL was performed on ice for 1 h, followed by centrifugation (6 min at 400 g) and two washes with R10. In each experiment, a medium-only control was used to determine the spontaneous loss of radiolabeled DNA. For cellcell killing assays, 104 pre-treated effector cells were added per well. Cultures were incubated for 1720 h at 37°C and radiolabeled DNA was then harvested onto glass filters for scintillation counting. In blocking experiments, Jurkat cells and supernatants to be blocked were pre-incubated with anti-Fas mAb (ZB4; Coulter Immunotech, Westbrook, ME) and anti-FasL mAb (NOK-1; PharMingen) respectively. Percent Fas-dependent specific lysis was calculated according to the equation 100x(S E)/S, where S and E represent retained DNA in the absence of added protein and experimentally retained DNA in the presence of added protein respectively. Maximal killing ranged from 40 to 70%, depending on the assay.
Annexin V binding assays
Daudi cells (106) were pre-treated with 300 ng/ml of CTLA-4FasL or ß2mFasL on ice for 1 h, pelleted and washed. Pre-treated Daudi cells (5x105) were plated beneath semipermeable 3 µm membranes (Becton Dickinson) in 24-well tissue culture plates. Jurkat cells (106) were added either above or below the membrane, as indicated. After an 18 h incubation period, cells were pre-stained with CD3FITC (Dako) and then tested for externalization of phosphatidylserine (used as a marker for early apoptosis), as detected by binding of Annexin Vphycoerythrin (PharMingen), according to the manufacturer's protocol. Propidium iodide-excluding populations of CD3+ cells were gated for analysis, and Annexin Vbright cells were counted and plotted as percent of total CD3+ cells.
![]() |
Results |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Both FasL and CTLA-4 proteins form multimers in their native forms, existing as non-covalently-associated homotrimers and disulfide-linked homodimers respectively. The multimerization status of the CTLA-4FasL fusion protein was first evaluated via immunoblotting experiments. Conditioned media from two CTLA-4FasL transfectant clones were processed for SDSPAGE and immunoblotting with FasL-specific antiserum, in the presence or absence of reducing agent. In the absence of reducing agent, two prominent ~7090 kDa molecular species, along with a weaker one at ~45 kDa, were detected (Fig. 1A). In contrast, in the presence of reducing agent, the larger bands disappeared, leaving only the ~45 kDa band. As expected, no significant FasL immunoreactivity was detected in conditioned media from a negative control, i.e. vector-only 293 cell transfectants. This finding was consistent with the presence of inter-chain disulfide bridges between ~45 kDa CTLA-4FasL monomers.
|
To confirm these gel-based CTLA-4FasL multimerization findings, Superdex-200 gel filtration chromatography was performed. Peak column fractions containing FasL immunoreactivity, as determined by ELISA, eluted, as expected, at ~140150 kDa. Chemical cross-linking of peak fractions followed by anti-FasL immunoblotting also revealed the ~7090 and ~140150 kDa CTLA-4FasL complexes (data not shown). Hence, the chemical cross-linking and gel filtration analyses, taken together, suggest that CTLA-4FasL complexes have a trimeric molecular stoichiometry, with the presence of intermolecular disulfide bridges between at least two of the component chains.
Apoptotic activity of CTLA-4FasL versus ß2mFasL
Functional differences between recombinant membrane-binding FasL and sFasL proteins have been observed (12,13). Apoptosis of Fas-sensitive target cells is readily detected after exposing these cells to 0.1 ng/ml of membrane-binding FasL on membrane microvesicles (13); in contrast, certain sFasL trimers have been shown to antagonize Fas receptor triggering (12). The relative capacities of CTLA-4FasL versus ß2mFasL to activate (or antagonize) apoptosis in the presence (or absence) of a `membrane-anchoring' cell was determined. To this end, the transfectant supernatants were tested for their respective abilities to induce apoptosis of Fas-sensitive Jurkat T cells, as measured by a radiometric DNA fragmentation assay. [3H]Thymidine-labeled Jurkat cells were co-incubated with comparable amounts, as quantitated by a soluble sFasL ELISA, of CTLA-4FasL or ß2mFasL. CTLA-4FasL demonstrated dose-dependent killing of Jurkat cells from 1100 ng/ml (Fig. 2A). Importantly, the addition of Daudi cells (a Burkitt's lymphoma B cell line), which constitutively express moderate levels of B7-1 and high levels of B7-2, increased CTLA-4FasL cytotoxic activity by ~60-fold and lowered the limit of detection of apoptosis to 0.1 ng/ml of CTLA-4FasL. Two other EBV-transformed B cell lines, JY and Raji, both with B7 expression profiles similar to Daudi cells, also potentiated CTLA-4FasL-mediated cytotoxicity (data not shown).
|
Dependence of CTLA-4FasL trans effector activity upon intercellular contact
CTLA-4Ig binds to cells expressing B7-1 and/or B7-2 (46). To test whether CTLA-4FasL similarly binds to B7+ cell surfaces, we incubated B7-1/B7-2+ Daudi cells with increasing concentrations CTLA-4FasL-containing cell supernatants and detected surface FasL expression by indirect immunofluorescence and flow cytometry. A CTLA-4FasL dose-dependent increase in the detection of cell surface-associated FasL epitopes was observed, with a plateau at ~100 ng/ml (Fig. 3A, upper panel). In parallel, we demonstrated CTLA-4FasL also binds to Fas+ (B7) Jurkat cells, with a dose-dependent increase in CTLA-4 epitopes upon adding 30300 ng/ml of CTLA-4FasL (Fig. 3A
, lower panel). Hence, CTLA-4FasL exhibits the expected binding potentials for cells expressing either B7 or Fas at their surfaces.
|
Next, the cell-contact dependence of the apoptotic effector function of CTLA-4FasL-coated Daudi cells was tested in a dual-chamber experiment. CTLA-4FasL-coated Daudi cells were plated below semipermeable membranes in culture wells, and Jurkat target cells were then added to the same wells both above and below the semipermeable membranes. Following a 20 h co-incubation period, Jurkat cell apoptosis was monitored by flow cytometric detection of fluorochrome-conjugated Annexin V binding to apoptotic cell surfaces. Significant Jurkat cell apoptosis was observed only in the lower compartment where CTLA-4FasL-coated Daudi effector cells and the Jurkat target cells were in direct contact on the same side of the semipermeable membrane (Fig. 3C, lower panel); the Jurkat cells present in isolation above the membrane did not demonstrate Annexin V positivity (Fig. 3C
, upper panel). As expected, no Jurkat cell apoptosis was observed when either untreated or ß2mFasL-treated Daudi cells were used as effectors. This experiment clearly shows that once bound to B7+ cells, CTLA-4FasL function is cellcell contact dependent. Moreover, these dual-chamber experiments indicate that CTLA-4FasL-coated cells do not shed sufficient CTLA-4FasL protein, either through direct protein loss or microvesiculation, to induce apoptosis at a distance in Jurkat cell targets. Furthermore, the experiment rules out the possibility that the Daudi cells, once coated with CTLA-4FasL, release some type of other soluble inhibitory factor that could act on its own.
Relative activity of purified CTLA-4FasL versus CTLA-4Ig against peripheral T cell responders
The immunomodulatory activity of CTLA-4FasL was compared to that of CTLA-4Ig and sFasL, the latter two proteins corresponding to CTLA-4FasL's component parts. For this experiment, peripheral T cells (within PBMC) were used in place of the Jurkat T cell line, and the capacity of the various proteins in their purified forms to inhibit anti-CD3 mAb (OKT3)-induced proliferation of these T cells was assessed. As baseline data, graded doses of OKT3 mAb (from 0.1 to 1.0 ng/ml) yielded the expected increases in T cell proliferation (data not shown). CTLA-4FasL potently inhibited this proliferative response at all time points (Fig. 4A), with complete inhibition observed at 30 ng/ml CTLA-4FasL at an OKT3 concentration of 1 ng/ml (Fig. 4B
). In contrast, the addition of equivalent amounts of CTLA-4Ig or sFasL resulted in significantly less inhibition. At higher concentrations of sFasL, dose-dependent inhibition was detected, reaching ~43% at 1000 ng/ml (Fig. 4C
). Likewise, antibody cross-linked sFasL was inhibitory at higher concentrations (~77% inhibition at 1000 ng/ml). Of note, in control assays, commercial sFasL that was maximally activated by antibody cross-linking demonstrated quantitatively comparable apoptosis-inducing activity to cell-anchored CTLA-4FasL (data not shown).
|
The 50% inhibitory concentration (IC50) for CTLA-4FasL was compared to that of CTLA-4Ig, sFasL, antibody-cross-linked sFasL, and the combination of CTLA-4Ig with sFasL (or antibody-cross-linked FasL). The IC50 versus proliferation for CTLA-4FasL (~45 kDa) was ~6 ng/ml, which on a molar basis translates into ~130 pM. By contrast, the IC50s for CTLA-4Ig (40 kDa) or sFasL (35 kDa) were calculated as ~130 and ~29 nM respectively. Therefore, CTLA-4FasL demonstrates potencies ~1000- and ~220-fold greater than CTLA-4Ig and sFasL respectively. Antibody-cross-linking of sFasL increased inhibitory potency and reduced the IC50 of sFasL to ~5.7 nM (~5-fold).
Combining CTLA-4Ig and sFasL lowered the doses required for inhibition, regardless of the presence of cross-linking antibodies. In calculating IC50s for these combinations, the IC50 of sFasL was determined in the presence of a maximal dose of CTLA-4Ig (10 µg/ml). The inhibition curves take into consideration the baseline offset observed in the presence of 10 µg/ml CTLA-4Ig. IC50s (molar calculations based on the mol. wt of sFasL) for CTLA-4Ig plus sFasL and for CTLA-4Ig plus antibody-cross-linked sFasL were ~26 and ~2.9 nM respectively. Taken together, findings indicate that CTLA-4FasL's inhibitory capacity substantially exceeds that of CTLA-4Ig and sFasL, whether used alone or in combination.
![]() |
Discussion |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The major findings of this study include the following: (i) Like native sFasL, CTLA-4FasL forms functional trimers, with evidence in this case for disulfide bridging between at least two of the component polypeptide chains. This latter feature mirrors the disulfide bridging within native CTLA-4 homodimers. (ii) CTLA-4FasL's capacity to induce apoptosis in Jurkat T cell targets is substantially potentiated by the addition of B7+ cells that serve to anchor the fusion protein to APC membranes. (iii) The apoptosis observed in this system is dependent upon the FasL:Fas molecular axis, as shown by antibody blocking. (iv) CTLA-4FasL binding to both B7+ APC and Fas+ T cells can be readily demonstrated by immunostaining. (v) APC pre-coated with CTLA-4FasL induce apoptosis of Jurkat T cell targets in a cell-contact-dependent fashion. (vi) Immunoaffinity-purified CTLA-4FasL potently inhibits the proliferative response of native T cells (within PBMC pools) triggered via their TCR complexes, with the degree of inhibition substantially surpassing that for CTLA-4Ig or sFasL when used alone or combinatorially.
Membrane anchorage potentiates CTLA-4FasL's apoptosis-inducing activity. This was directly demonstrated in experiments where B7+ anchoring cells were either titrated into the killing assay or were pre-coated with the fusion protein prior to the addition of target cells. The membrane anchorage effect likely reflects an augmentation of functional valency. Furthermore, the data and interpretation are consistent with the literature comparing the functional activities of membrane-associated versus sFasL. For example, whereas 0.1 ng of neuro2A cell membrane preparations containing membrane-associated FasL readily kill Jurkat cells (13), 10- to 100-fold more recombinant, sFasL molecular aggregates derived from COS cell transfectants are required to trigger significant Fas-dependent cell death of mouse cell lines (17,47). CTLA-4FasL's activity level contrasts with that of ß2mFasL, which does not detectably bind to APC surfaces and displays no significant cytocidal effects in the absence of cross-linking antibody. The enhancement of ß2mFasL function with epitope tag-specific cross-linking antibody parallels that documented for other recombinant sFasL trimers (13).
As an immunomodulator, CTLA-4FasL combines CD28 antagonism with Fas receptor agonism. This particular combination was motivated by data pointing to an antagonistic relationship between the CD28 and Fas pathways. CD28 signaling inhibits Fas-dependent apoptosis (48), possibly due to up-regulation of Bcl-xL (48,49). Furthermore, CD28 co-stimulator blockade inhibits survival pathway signaling (49) and thereby promotes the Fas apoptosis pathway (39-42). One group has suggested that co-stimulator-induced cytoprotection from Fas-mediated apoptosis stems from FLIP/I-FLICE/FLAME-1/CASH/casper-mediated inhibition of death cascade signaling (50); however, this conclusion was not supported in another study (51). In recent experiments, we have demonstrated B7-1:FasL antagonism by combining B7-1 and FasL protein transfer (A. Chen and M. Tykocinski, unpublished observations). That said, the precise extent to which B7 blockade contributes to CTLA-4FasL's observed activity remains unclear and will require more complicated experimental approaches.
The onset of Fas-mediated, activation-induced cell death in peripheral human T cells, in vitro, occurs ~45 days post-activation (52). Hence, the observation that CTLA-4FasL significantly inhibits T cell proliferation as early as culture days 23 is unexpected. There would appear to be a previously unappreciated kinetic difference between Fas-mediated apoptosis induction versus proliferative inhibition. Since the Fas receptor is up-regulated within 24 h of T cell activation (52,53 and data not shown), it is certainly available to mediate CTLA-4FasL-driven effects at the 23 day time period. There are a variety of downstream intracellular mediators linked to Fas receptor triggering that could potentially explain our observed early effect on T cell proliferation. Additionally, it should be pointed out that a blockade in cell division, regardless of the mechanisms, is likely to impact other T cell functions, such as cytokine secretion. Of note in this regard, secretion of some (e.g. IL-2) but not all (e.g. IFN-) cytokines requires the T cells to undergo multiple rounds of replication (54). Hence, it will be important to determine if CTLA-4FasL differentially regulates the secretion of various cytokines during T cell activation.
FasL is a type II membrane protein, i.e. its functional C-terminus projects outward from the membrane (55). Previously, a variety of heterologous sequences have been appended to type II membrane proteins for purposes such as directing secretion, influencing multimerization and appending epitope tags (17,5659). In the present study, we report a unique type of type II membrane protein derivative, in which the N-terminal intracellular and transmembrane domains of this protein have been replaced with a functional domain that anchors it to cell surfaces. In the case of the CTLA-4FasL fusion protein developed here, the CTLA-4 anchoring domain, derived from a type I membrane protein, confers an added functional feature, i.e. trans signal masking. An elegant aspect of CTLA-4FasL's design is that this chimeric soluble type Itype II protein positions the type II component at the C-terminus, projecting this business end of the molecule away from the membrane following membrane association.
The observation that CTLA-4FasL results in apoptosis of B7-1/B7-2 Jurkat cells was unexpected, given that another trimeric sFasL derivative has been reported to function as an antagonist (11). One explanation for this finding revolves around the structure of CTLA-4FasL. Based upon our gel analyses, each CTLA-4FasL trimer likely consists of two disulfide-linked CTLA-4 domains and an unpaired, `free', CTLA-4 domain. Hence, in principle, interactions between the free CTLA-4 domains of two CTLA-4FasL trimers could occur. Our chemical cross-linking data indicate soluble CTLA-4FasL predominates in trimeric complexes, it is unlikely that free CTLA-4 domains dimerize readily in solution. However, the fact that soluble CTLA-4FasL kills Jurkat cells suggests the possible dimerization of unpaired CTLA-4 domain units within cell surface-bound CTLA-4FasL trimers. In principle, this would yield CTLA-4FasL `hexamers' with proapoptotic activity analogous to that of antibody-cross-linked sFasL hexamers. In support of this hypothesis, both CTLA-4FasL and antibody-cross-linked ß2mFasL display similar doseresponse curves for Jurkat cell apoptosis (Fig. 1). This CTLA-4FasL hexamerization hypothesis will require further testing.
Targeting of pathogenic tissues with soluble, Fas-specific agents reveals a significant toxicity issue in vivo. Intraperitoneal or i.v. administration of either Fas-specific antibody (20) or recombinant sFasL (18,19) result in fulminant hepatitis, intrahepatic hemorrhage and rapid death. In principle, such in vivo toxicity might be reduced by directing the Fas triggering agents to the cell surfaces of cells within immune organs. However, it should be noted that Kupffer cells are B7+ and, hence, adjoining sinusoidal hepatocytes could, in principle, be susceptible to apoptosis induction by CTLA-4FasL. However, if this proved to be a problem, therapeutic windows might be exploited, or alternatively, fusion proteins incorporating different co-stimulator receptors could be designed that bypass resident APC. Of note, our preliminary studies have shown no evidence for in vivo lethality or hepatotoxicity of CTLA-4FasL (J. Huang and M. Tykocinski, unpublished observations).
In a mouse transplantation model, APC transduced with an adenovirus vector expressing recombinant FasL effected short-term elimination of alloreactive T cells with little hepatotoxicity. However, the alloreactive T cell depletion achieved by this ex vivo cell engineering method was incomplete, with the eventual repopulation of pathogenic T cells with time. The `in vivo cell engineering' made possible by CTLA-4FasL could potentially yield more effective elimination of pathogenic T cells, while sharing in the lower toxicity of cell-based approaches.
By definition, hetero-bifunctional fusion proteins of the `trans signal converter protein (TSCP)' class serve to block one trans signal and simultaneously confer another. In the case of CTLA-4FasL, APC are being targeted, and the B7-1 co-stimulatory signal is being replaced with a FasL apoptotic signal. However, multiple permutations can be envisioned for TSCP, with alternative designs drawing upon other molecular pairs and cellular effectors and targets. Thus, it should be possible not only to tap into other T cell co-stimulator:receptor combinations, but also to move beyond T cell inhibition altogether, targeting other regulatory endpoints of potential therapeutic benefit.
![]() |
Acknowledgments |
---|
![]() |
Abbreviations |
---|
2-ME 2-mercaptoethanol |
ß2m ß2-microglobulin |
APC antigen-presenting cell |
B7-1 CD80 |
B7-2 CD86 |
DSP dithiobis(succinimidyl proprionate) |
EBV EpsteinBarr virus |
FasL Fas (CD95) ligand |
GPI glycosyl-phosphatidylinositol |
PBMC peripheral blood mononuclear cells |
sFasL soluble FasL |
TSCP trans signal converter protein |
![]() |
Notes |
---|
Received 24 July 2000, accepted 8 January 2001.
![]() |
References |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|