A recent study identified a clonal expansion of CD3CD4+cells secreting Th2-type cytokines in 4 patients with chronic hypereosinophilia. Because interferon α (IFN-α) is used in the therapy of the idiopathic hypereosinophilic syndrome, the effects of this cytokine on the survival of clonal Th2 cells isolated from the blood of 2 patients were determined. First, these cells displayed a high rate of spontaneous apoptosis on culture in cytokine-free medium and were also sensitive to Fas-mediated apoptosis induced by soluble Fas ligand. Addition of IFN-α or interleukin-2 (IL-2) to culture medium resulted in significant protection against spontaneous but not Fas-induced apoptosis. Although spontaneous apoptosis of the clonal Th2 cells was clearly associated with down-regulation of both bcl-2 and bcl-xL levels, IFN-α had no significant effect on the expression of these antiapoptotic proteins, whereas addition of IL-2 resulted in higher levels of bcl-2. On the other hand, IFN-α decreased the numbers of cells with disrupted mitochondrial transmembrane potential both during spontaneous apoptosis and after exposure to protoporphyrin IX. Thus, IFN-α might promote the survival of clonal Th2 cells, an effect that could be relevant to the therapeutic approach for patients with chronic hypereosinophilia caused by clonal expansion of Th2-type cells.

Interferon α (IFN-α) has been used successfully to treat the idiopathic hypereosinophilic syndrome (HES).1-6 The beneficial effects of IFN-α in this disease have been attributed to the direct actions of this immunomodulatory molecule on eosinophils because it inhibits eosinophilopoiesis,7 several effector functions of eosinophils,8,9 and their recruitment into tissues.10 Moreover, it has been suggested that IFN-α might directly promote eosinophil apoptosis.11 Most clinical studies evaluating the benefits of IFN-α in the setting of the idiopathic HES have been conducted on patients presenting clinical and biologic parameters consistent with the myeloproliferative variant of this disease.1-6,12,13 

We and others recently demonstrated that a subgroup of patients with idiopathic HES actually present an underlying T-lymphocyte disorder, characterized by the presence in peripheral blood of clonally expanded interleukin-5 (IL-5)–producing T cells with an aberrant surface phenotype.14-20 In our own series of 4 such patients, the clonal T cells presented a CD3CD4+phenotype.14-17 Absent or low membrane expression of the T-cell receptor (TCR)/CD3 complex on helper T cells has been observed in several pathologic settings including (retro-)viral infections,21-23 chronic antigenic stimulation,24 and cancer.25,26 Whatever the cause of their lack of CD3/TCR expression, the pathogenic role of the clonal CD3CD4+ cells in the induction of hypereosinophilia in our patients is highly likely. Indeed, on polyclonal activation or after contact with dendritic cells, these clonal cells secrete high levels of IL-5, together with IL-4 and IL-13, indicating their Th2 nature.14-17 

We have previously demonstrated that IFN-α is a potent inhibitor of IL-5 synthesis by differentiated human Th2 clones, including cells obtained from 2 patients in our series, providing rationale for treatment of such patients with this molecule.15 However, the premalignant nature of clonal Th2 cells found in patients with chronic hypereosinophilia has been suggested by the subsequent development of T-cell lymphoma.14,17,20 We were therefore interested in investigating the influence of IFN-α on the survival of clonal Th2 cells associated with chronic hypereosinophilia. We first observed that these clonal T cells are highly prone to undergo apoptosis both spontaneously in culture with cytokine-free medium and on Fas engagement. We then determined the effects of IFN-α on spontaneous and Fas-mediated apoptosis. The observation that IFN-α protects Th2 cells against spontaneous apoptosis led us to determine the effects of this cytokine on bcl-2 and bcl-xL expression as well as on mitochondrial changes implicated in the apoptotic program, including disruption of the mitochondrial transmembrane potential (Δψm) and the production of reactive oxygen species (ROS) in those cells.27-29 

Patients

As previously described,15-17 patient 1 was a 20-year-old woman presenting with severe pruritus, eczema, and tenosynovitis of the right ankle. At presentation, the circulating leukocyte count was 16 900/μL, including 8923 eosinophils and 4630 lymphocytes. CD4+ T cells represented 87% of total lymphocytes and were composed of 88% CD3CD4+cells (3545 CD3CD4+ cells/μL) and 12% CD3+CD4+ cells. Serum IgE and IgM levels were 340 U/mL (normal, < 20) and 310 mg/dL (normal, 40-250), respectively. Treatment with IFN-α was followed by regression of both clinical manifestations and decrease of eosinophil blood count, although the clonal CD3CD4+ population remained stable. However, recurrence of hypereosinophilia was observed less than a year after initiation of IFN-α. Subsequent evolution has been characterized by expansion of the circulating CD3CD4+ population (reaching 4800/μL) and appearance of chromosomal abnormalities in these cells. The patient has now developed peripheral T-cell lymphoma characterized by bilateral masses in the parotid regions and a submandibular mass composed of CD3CD4+ cells.

Patient 216,17 was a 21-year-old woman also presenting with severe pruritus and eczema, as well as cyclic angioedema. The circulating leukocyte count was 14 800/μL, including 9102 eosinophils and 3419 lymphocytes. CD4+ T cells represented 87% of total lymphocytes and were composed of 83% CD3CD4+ cells (2469 CD3CD4+ cells/μL) and 17% CD3+CD4+ cells. Serum IgE and IgM levels reached 15 640 U/mL and 1253 mg/dL, respectively. The clinical and biologic findings were consistent with the diagnosis of Gleich syndrome. Two-year follow-up has been characterized by good response to glucocorticoid treatment, as evidenced by normalization of eosinophil levels. Although the CD3CD4+ cell population had decreased significantly (662/μL) over this period, tapering of steroid dosage was quickly followed by recurrence of hypereosinophilia and clinical manifestations. In both patients, bone marrow aspiration showed abundant eosinophil precursors and absence of blastic cells. They were free of treatment at the time of blood sampling for the present study.

Reagents

Human recombinant IFN-α (rIFN-α) 2b with a specific activity of 3.106 IU/mL was obtained from Schering-Plough (Innishannon, Ireland). Human recombinant IL-2 (rIL-2) with a specific activity of 3.104 IU/mL was kindly provided by K. Willard-Gallo (Université Libre de Bruxelles, Rhode Saint-Genèse, Belgium). The following monoclonal antibodies (mAb) were purchased from Becton Dickinson (Mountain View, CA): fluorescein isothiocyanate (FITC)–labeled anti-CD45RA; peridinin chlorophyll protein (PerCP)–labeled anti-CD4; phycoerythrin (PE)–labeled anti-Fas (CD95), anti-DR, and anti-CD45RO; purified anti-CD3; and anti-CD45RA. The neutralizing anti-Fas ligand (clone NOK-1) mAb was purchased from Pharmingen (San Diego, CA). The soluble human recombinant APO-1/Fas ligand (sFasL) and associated enhancer (antibody reacting with sFasL) were purchased from Alexis Corporation (Laüfelfingen, Switzerland). Mouse monoclonal anti-bcl-xL (IgG1, H-5) antibody was obtained from Santa Cruz Biotechnology (Santa Cruz, CA) and mouse IgG1 isotype control from R & D Systems Europe (Abingdon, United Kingdom). FITC-labeled rabbit antimouse Ig used as second layers, FITC-conjugated anti-bcl-2 mAb (IgG1k, clone 124), and IgG1 control mAb for intracellular staining of bcl-2 were obtained from Dako (Glostrup, Denmark).

Isolation of CD3CD4+ T cells from hypereosinophilic patients

Blood leukocytes were obtained from both patients by cytapheresis, after informed consent was obtained. Peripheral blood mononuclear cells (PBMC) were isolated by density gradient centrifugation on Lymphoprep (Nycomed, Oslo, Norway). After 3 washings with Hanks balanced salt solution (HBSS) (Gibco, Life Technologies, Paisley, Scotland), cells were resuspended in cold-conservation medium composed of 50% RPMI 1640, 10% dimethylsulfoxide (DMSO) (Sigma Chemical Co, St Louis, MO), and 40% heat-inactivated fetal calf serum (FCS) (BioWhittaker, Europe, Verviers, Belgium). Cells were quickly frozen in a −80°C freezer and then stored in liquid nitrogen until use. CD3CD4+ T cells from both patients were prepared from CD4+ T cells (see below) by selective depletion of CD3+ T cells through incubation with anti-CD3 mAb for 30 minutes at 4°C, washing, and then incubation with immunomagnetic particles coated with sheep antimouse IgG (Dynabeads, Dynal, Oslo, Norway). The resulting cell preparations contained more than 90% viable CD3CD4+ T cells as determined by flow cytometry.

Isolation of CD3+CD4+CD45RO+ T cells from healthy volunteers

The PBMC were isolated from buffy coats of healthy individuals by density gradient centrifugation on Lymphoprep. Cells were resuspended in cold-conservation medium, quickly frozen, and then stored in liquid nitrogen until use. After 3 washings with HBSS, PBMC were resuspended in complete medium (RPMI 1640, BioWhitakker, supplemented with 10% FCS and 40 μg/mL gentamicin). CD4+lymphocytes were purified using the MACS CD4+ T cell isolation kit (Miltenyi Biotec Gmbh, Bergisch Gladbach, Germany). The CD4+ T-cell suspensions were further incubated with an anti-CD45RA mAb for 30 minutes at 4°C, washed, and incubated with immunomagnetic particles coated with sheep antimouse IgG (Dynabeads, Dynal). Coated cells were removed with a magnet, leaving purified CD3+CD4+CD45RO+ cells in suspension. The resulting cell preparations routinely contained more than 90% viable CD3+CD4+CD45RO+T cells.

Lymphocyte phenotyping

The expression of HLA-DR, CD45RO, and Fas (CD95) on CD4+ T cells was determined by flow cytometry. For that purpose, PBMC from patients and from healthy blood donors were washed in phosphate-buffered saline (PBS) and incubated for 30 minutes on ice with appropriate mAbs: FITC-, PE-, or PerCP-conjugated mAbs against CD4, CD3, CD45RO, HLA-DR, and CD95 antigens. After washing, 10 000 cells were acquired using a FACScan flow cytometer (Becton Dickinson) and analyzed with the Cellquest software (Becton Dickinson).

Cell culture and determination of apoptosis

Normal CD3+CD4+CD45RO+ and clonal CD3CD4+ T cells were seeded at 2.105/well per 200 μL at 37°C and 5% CO2-humidified atmosphere in flat-bottomed 96-well plates (Nunc, Gibco) in complete medium in the presence or absence of IL-2 (100 U/mL) or IFN-α (104-1 IU/mL). For induction of Fas-mediated apoptosis, CD4+ T cells were incubated in the presence of sFasL at concentrations ranging from 50 to 1.5 ng/mL and associated enhancer (1 μg/mL). After 24 to 96 hours of incubation, CD4+ T cells were collected and examined for apoptosis. In most experiments, recovered viable lymphocytes were detected by flow cytometry according to their light forward and side scatter properties (FSC/SSC). Indeed, apoptotic lymphocytes were smaller in size than resting viable lymphocytes but were distinct from debris by an increase in 90° scatter. After culture, numbers of viable CD3+CD4+CD45RO+ or CD3CD4+ T lymphocytes (FSChigh/SSClow) recovered were determined and expressed as a percentage of the original input viable cells. Based on these measurements, results were expressed as percentage of apoptotic cells. Measurement of apoptosis by the light scatter method was verified by 2 additional methods for apoptosis detection, namely annexin V binding and terminal deoxynucleotidyl transferase (TdT)–mediated deoxyuridine triphosphate (dUTP) nick end-labeling (TUNEL). FITC–annexin V binding detects phosphatidyl serine on the outer surface of cells undergoing apoptosis and was performed using a commercially available kit (Bender MedSystems, Vienna, Austria). TUNEL was performed using a kit according to the manufacturer's protocol (Boehringer Mannheim, Mannheim, Germany). We confirmed that FSClow/SSChigh cells were clearly enriched in apoptotic cells (98% annexin V positive [An+] and 97% TUNEL positive), whereas FSChigh/SSClow cells contained few apoptotic cells (9% An+ and 7% TUNEL positive). Double-labeling experiments with FITC–annexin V and propidium iodide (PI) revealed that most of the An+ cells were PI+ after 24 hours of culture (not shown).

Expression ofbcl-2 and bcl-xL

Levels of bcl-2 and bcl-xL expression were determined by flow cytometry. Intracellular staining for bcl-2 was performed as follows. Cells were washed in PBS supplemented with 10% FCS and 0.1% azide (PBS-FCS-NaN3), fixed in FACS lysing solution (Becton Dickinson) for 10 minutes at room temperature, washed again in PBS, and then incubated for 15 minutes at room temperature in permeabilizing solution (Becton Dickinson) in PBS-FCS-NaN3. Finally, cells were intracellularly stained with FITC-conjugated anti–bcl-2 mAb, FITC-isotype control mAb, nonlabeled anti–bcl-xL mAb, and isotype control for 30 minutes at room temperature. Bcl-xL–labeled cells were washed in PBS-FCS-NaN3 buffer and further incubated in presence of a FITC-labeled rabbit antimouse Ig for 30 minutes at room temperature. After washing, cells were immediately applied to a FACScan flow cytometer and analyzed with the Cellquest software (Becton Dickinson). Apoptotic cells were excluded on basis of FSC/SSC parameters. The expression of bcl-2 and bcl-xL was reported in specific molecule equivalents of soluble fluorochrome (MESF) using QuickCal beads (Flow Cytometry Standards Corp, San Juan, Puerto Rico).

Cytofluorometric analysis of Δψm and reactive oxygen species generation

To evaluate Δψm, clonal Th2 cells (106/mL) from patient 1 were incubated in complete medium with 2.5 nmol/L 3,3′-dihexyloxacarbocyanine (DiOC6; Molecular Probes, Eugene, OR) for 15 minutes at 37°C followed by analysis on a FACScan flow cytometer, according to a published protocol.30 The fluorescence of DiOC6 (green), a potential-sensitive cationic lipophilic dye that incorporates into the mitochondrial matrix, correlates with the Δψm and is oxidation independent. Therefore, DiOC6low cells correspond to cells with disrupted Δψm. Superoxide generation was also measured by flow cytometry using 0.25 μmol/L hydroethidine (HE; Molecular Probes), a dye that is oxidized into fluorescent ethidium (red) by superoxide anion.30HEhigh cells therefore correspond to cells with increased production of superoxide anions. In some experiments, the clonal Th2 cells were cultured in the presence of 30 μmol/L protoporphyrin IX (PPIX; Sigma), alone or in combination with IFN-α (104 IU/mL).

Clonal CD3CD4+ Th2 cells associated with chronic hypereosinophilia present an activated memory phenotype

We described elsewhere the Th2-like cytokine profile of the monoclonal CD3CD4+ T lymphocytes present in the circulation of the 2 patients with chronic hypereosinophilia.15-17 As shown in Figure1, by flow cytometry, their phenotype was further characterized by the expression of the CD45RO isoform and the activation antigen HLA-DR and by the absence of the CD7 and CD27 markers16,17 (not shown). Moreover, the clonal T cells expressed high levels of CD95 as compared with CD3+CD4+CD45RO+ T cells obtained from healthy individuals. The phenotype of the clonal Th2 cells is therefore consistent with that of activated memory T cells.

Fig. 1.

The clonal Th2 cells present an activated memory phenotype.

PBMC from 2 hypereosinophilic patients (P1 and P2) and from healthy blood donors were stained with FITC-, PE-, or PerCP-conjugated mAbs against CD4, CD3, CD45RO, HLA-DR, and CD95 antigens. Surface phenotype was determined by flow cytometry after gating on CD3+CD4+ or CD45RO+ lymphocytes from healthy subjects and clonal CD3CD4+ Th2 cells from both patients. The figure shows histograms from 1 experiment, representative of 3. The background staining using isotype-matched control Abs is represented by the broken line.

Fig. 1.

The clonal Th2 cells present an activated memory phenotype.

PBMC from 2 hypereosinophilic patients (P1 and P2) and from healthy blood donors were stained with FITC-, PE-, or PerCP-conjugated mAbs against CD4, CD3, CD45RO, HLA-DR, and CD95 antigens. Surface phenotype was determined by flow cytometry after gating on CD3+CD4+ or CD45RO+ lymphocytes from healthy subjects and clonal CD3CD4+ Th2 cells from both patients. The figure shows histograms from 1 experiment, representative of 3. The background staining using isotype-matched control Abs is represented by the broken line.

Close modal

Interferon α protects clonal Th2 cells against spontaneous but not Fas-mediated apoptosis

In a first series of experiments, we compared the rate of spontaneous apoptosis of clonal CD3CD4+ cells from patients with that of CD3+CD4+CD45RO+ T cells from healthy individuals during a culture period of 1 to 4 days in the absence of any growth factor. The percentage of apoptotic cells was then quantified using flow cytometry analysis according FSC/SSC parameters, measuring DNA fragmentation using TUNEL assay and phosphatidylserine externalization using FITC–annexin V binding. The data presented in Figure 2 are a graphic representation of the flow cytometry data for FSC/SSC parameters. We found at all time points a significantly increased proportion of apoptotic cells within the clonal Th2 cells as compared with control CD3+CD4+CD45RO+ T cells. Similar results were obtained using either FITC–annexin V binding or TUNEL assay, except that in every case more apoptotic cells were detected as defined by FSC/SSC or annexin V binding than as defined by the TUNEL assay. This difference was probably due to the fact that DNA degradation is a relatively late event in apoptosis.

Fig. 2.

The clonal Th2 cells are highly prone to undergo spontaneous apoptosis.

Purified CD3+CD4+CD45RO+ cells from healthy individuals (♦) and CD3CD4+ cells from hypereosinophilic patient 1 (●) and patient 2 (▪) were cultured in the absence of exogenous cytokine. Cells were recovered after 24, 48, 72, and 96 hours of culture. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of percent apoptotic cells of 5 experiments). *P < .005 as compared with control CD3+CD4+CD45RO+ cells from healthy individuals (ANOVA test).

Fig. 2.

The clonal Th2 cells are highly prone to undergo spontaneous apoptosis.

Purified CD3+CD4+CD45RO+ cells from healthy individuals (♦) and CD3CD4+ cells from hypereosinophilic patient 1 (●) and patient 2 (▪) were cultured in the absence of exogenous cytokine. Cells were recovered after 24, 48, 72, and 96 hours of culture. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of percent apoptotic cells of 5 experiments). *P < .005 as compared with control CD3+CD4+CD45RO+ cells from healthy individuals (ANOVA test).

Close modal

The increased expression of the CD95 antigen on the clonal Th2 cells prompted us to examine whether the propensity of these cells to undergo apoptosis could be enhanced through ligation of Fas. As shown Figure3, most of the clonal Th2 cells (> 90%) underwent apoptosis after 24 hours of incubation with 50 ng/mL sFasL, whereas less than 40% of apoptotic cells were found within control CD3+CD4+CD45RO+ T cells under the same experimental condition. The apoptosis induction by Fas engagement led us to determine whether this pathway was involved in the spontaneous apoptosis of those cells as they express mRNA encoding FasL (not shown). As shown in Table 1, addition of a blocking anti-FasL mAb did not affect the spontaneous apoptosis of the clonal Th2 cells, whereas it inhibited sFasL-induced apoptosis. These data establish that the spontaneous apoptosis of clonal Th2 cells is independent of the Fas/FasL pathway.

Fig. 3.

The clonal Th2 cells are sensitive to Fas-mediated apoptosis.

Purified CD3+CD4+CD45RO+ cells from healthy individuals (♦) and CD3CD4+ cells from hypereosinophilic patient 1 (▴) and patient 2 (▪) were cultured in the presence of graded concentrations of sFasL. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of percent apoptotic cells of 3 experiments. *P < .05 as compared with CD3+CD4+CD45RO+ cells from healthy individuals (ANOVA test).

Fig. 3.

The clonal Th2 cells are sensitive to Fas-mediated apoptosis.

Purified CD3+CD4+CD45RO+ cells from healthy individuals (♦) and CD3CD4+ cells from hypereosinophilic patient 1 (▴) and patient 2 (▪) were cultured in the presence of graded concentrations of sFasL. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of percent apoptotic cells of 3 experiments. *P < .05 as compared with CD3+CD4+CD45RO+ cells from healthy individuals (ANOVA test).

Close modal
Table 1.

IFN-α and IL-2 protect the clonal Th2 cells against spontaneous but not Fas-mediated apoptosis

Agent addedSpontaneousFas-mediated
Patient 1Patient 2Patient 1Patient 2
None 28 ± 1 31 ± 3 89 ± 4 96 ± 2 
Anti-Fas mAb 29 ± 2 28 ± 1 30 ± 3 28 ± 1 
Control mAb 28 ± 1 28 ± 1 90 ± 4 97 ± 1 
IL-2 (100 IU/mL) 11 ± 0.5* 10 ± 1* 78 ± 7 94 ± 1 
IFN-α (104IU/mL) 9 ± 1* 8 ± 0.5* 83 ± 5 89 ± 4 
Agent addedSpontaneousFas-mediated
Patient 1Patient 2Patient 1Patient 2
None 28 ± 1 31 ± 3 89 ± 4 96 ± 2 
Anti-Fas mAb 29 ± 2 28 ± 1 30 ± 3 28 ± 1 
Control mAb 28 ± 1 28 ± 1 90 ± 4 97 ± 1 
IL-2 (100 IU/mL) 11 ± 0.5* 10 ± 1* 78 ± 7 94 ± 1 
IFN-α (104IU/mL) 9 ± 1* 8 ± 0.5* 83 ± 5 89 ± 4 

Purified CD3CD4+ cells from patient 1 and patient 2 were cultured for 24 hours in absence (spontaneous) or presence (Fas-mediated) of sFasL (25 ng/mL). Anti-Fas (1 μg/mL) or control mAb (1 μg/mL), IL-2 (100 IU/mL) or IFN-α (104IU/mL) was added at the beginning of the culture. At the end of the culture, apoptotic lymphocytes were detected by flow cytometry according to their light scatter properties (FSC/SSC). Results are the mean percentages ± SD of 3 experiments.

*

P < .05 as compared with culture without IFN-α or IL-2 (ANOVA test).

We then examined whether the clonal Th2 cells could be rescued from apoptosis by IL-2, which is known to exert antiapoptotic effects on activated T cells. As shown in Figure 4, addition of IL-2 (100 U/mL) to the culture medium rescued more than 80% of the clonal T cells from spontaneous apoptosis. Addition of rIFN-α (104 IU/mL) also exerted a clear-cut antiapoptotic action on clonal Th2 cells from both patients. As shown in Figure5, the protective effect of rIFN-α on spontaneous apoptosis was dependent on the dose, a protective effect being already observed at a concentration of 1 IU/mL. The rescuing effect of IFN-α was also confirmed using FITC–annexin V binding and TUNEL assay (not shown). Contrasting with their inhibitory effects on spontaneous apoptosis, neither rIFN-α nor IL-2 affected Fas-induced apoptosis of the clonal T cells (Table 1). In a previous study, we demonstrated that IL-2 but not IFN-α induced proliferation of the Th2 cells.16 

Fig. 4.

IFN-α and IL-2 protect the clonal Th2 cells against spontaneous apoptosis.

CD3CD4+ cells from hypereosinophilic patient 1 and patient 2 (♦) were cultured in presence of IFN-α (104 IU/mL) (▪) or IL-2 (100 IU/mL) (▴). Cells were recovered after 24, 48, 72, and 96 hours of culture. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of % apoptotic cells of 5 experiments. *P < .005 as compared with culture without IFN-α or IL-2 (ANOVA test).

Fig. 4.

IFN-α and IL-2 protect the clonal Th2 cells against spontaneous apoptosis.

CD3CD4+ cells from hypereosinophilic patient 1 and patient 2 (♦) were cultured in presence of IFN-α (104 IU/mL) (▪) or IL-2 (100 IU/mL) (▴). Cells were recovered after 24, 48, 72, and 96 hours of culture. Apoptotic cells were detected by flow cytometry according to FSC/SSC parameters. Results are the means ± SEM of % apoptotic cells of 5 experiments. *P < .005 as compared with culture without IFN-α or IL-2 (ANOVA test).

Close modal
Fig. 5.

IFN-α prevents spontaneous apoptosis of the clonal Th2 cells in a dose-dependent manner.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured in the presence of graded amounts of IFN-α (1-104 IU/mL) for 24 hours. At the end of the culture, apoptotic lymphocytes were detected by flow cytometry according to their light scatter properties (FSC/SSC). Results are the means ± SEM of 4 experiments. *P < .005, **P < .05 as compared with culture without IFN-α (ANOVA test).

Fig. 5.

IFN-α prevents spontaneous apoptosis of the clonal Th2 cells in a dose-dependent manner.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured in the presence of graded amounts of IFN-α (1-104 IU/mL) for 24 hours. At the end of the culture, apoptotic lymphocytes were detected by flow cytometry according to their light scatter properties (FSC/SSC). Results are the means ± SEM of 4 experiments. *P < .005, **P < .05 as compared with culture without IFN-α (ANOVA test).

Close modal

Interferon α does not prevent bcl-2 and bcl-xLdown-regulation in clonal Th2 cells

To determine whether the susceptibility of the clonal Th2 cells to apoptosis was caused by changes in the constitutive expression of bcl-2 and bcl-xL which are known to exert antiapoptotic effects, purified clonal Th2 cells and normal CD3+CD4+CD45RO+ T cells were stained for these proteins immediately after purification and analyzed by flow cytometry. As shown in Table 2, bcl-2 expression was clearly lower among the clonal Th2 cells, whereas no significant difference was observed as far as bcl-xLexpression is concerned.

Table 2.

Expression of bcl-2 and bcl-xL in clonal Th2 cells

CellsAgent addedBcl-xLBcl-2
024 h024 h
CD4+CD45RO+(controls) — 83 021 ± 6427 71 524 ± 4752 64 986 ± 5483 58 933 ± 4982 
CD3CD4+ (patient 2) — 85 432 ± 5561 — 26 048 ± 6843* — 
CD3CD4+ (patient 1) — 96 953 ± 11704 58 644 ± 10128 29 839 ± 5290* 19 973 ± 2213 
 +IFN-α (104IU/mL) — 62 563 ± 13723 — 19 911 ± 2555 
 +IL-2 (100 IU/mL) — 66 238 ± 22834 — 23 647 ± 3371 
CellsAgent addedBcl-xLBcl-2
024 h024 h
CD4+CD45RO+(controls) — 83 021 ± 6427 71 524 ± 4752 64 986 ± 5483 58 933 ± 4982 
CD3CD4+ (patient 2) — 85 432 ± 5561 — 26 048 ± 6843* — 
CD3CD4+ (patient 1) — 96 953 ± 11704 58 644 ± 10128 29 839 ± 5290* 19 973 ± 2213 
 +IFN-α (104IU/mL) — 62 563 ± 13723 — 19 911 ± 2555 
 +IL-2 (100 IU/mL) — 66 238 ± 22834 — 23 647 ± 3371 

Intracytoplasmic bcl-xL and bcl-2 staining was performed on purified CD3+CD4+CD45RO+ cells from healthy individuals (n = 6) and CD3CD4+ from hypereosinophilic patients (for each patient 6 determinations were performed). For patient 1, bcl-xL and bcl-2 levels were compared with those obtained after 24 hours of incubation in the absence or presence of IFN-α (104 IU/mL) or IL-2 (100 IU/mL). Results shown are the mean ± SD of bcl-xL and bcl-2 staining within live FSChigh/SSClow cells expressed in MESF.

*

P < .05 as compared with CD4+CD45RO+ cells from healthy individuals.

P < .05 as compared with bcl-2 expression obtained in absence of IL-2 (ANOVA test).

Interleukin-2 is known to protect activated T cells against death by increasing bcl-2 and, to a lesser extent, bcl-xLexpression. We therefore decided to compare the effects of IL-2 and IFN-α on expression levels of both bcl proteins within the clonal Th2 cells of patient 1. For this purpose, bcl-2 and bcl-xLprotein expressions were examined by flow cytometry after culture in the absence or presence of the cytokines. Apoptotic and live cells were analyzed separately on the basis of FSC/SSC parameters. As shown in Table 2, bcl-2 and bcl-xL expression by live clonal Th2 cells decreased by about 40% after 24 hours of culture in the absence of exogenous cytokines. The drop in bcl-xL protein expression was even more pronounced within apoptotic cells, resulting in a clear bimodal expression of bcl-xL by the clonal Th2 cells (not shown). On the other hand, normal CD3+CD4+CD45RO+ T cells decreased their bcl-2 and bcl-xL levels only by about 10%. Although addition of IL-2 (100 IU/mL) partially prevented the drop in bcl-2 expression in clonal Th2 cells, the antiapoptotic effect of IFN-α was not associated with any significant change in bcl-2 or bcl-xL down-regulation during the 24-hour culture period (similar data were obtained after 72 hours of culture).

Interferon α prevents Δψm dissipation in clonal Th2 cells

Loss of Δψm, generation of superoxide anions by the uncoupled respiratory chain,27-29 and phosphatidylserine exposure31 at the cell membrane are characteristic features of the apoptotic process. To get further inside the mechanism of action of IFN-α, we monitored these parameters within the clonal Th2 cells.

First, we observed that a fraction of Th2 cells spontaneously become DiOC6low and HEhigh after 24 hours in culture, indicating disrupted Δψm and increased superoxide production, respectively (Figure6). Double-labeling experiments (not shown) revealed that most DiOC6low cells were HEhigh, FSClow/SSChigh, and An+, confirming the association between Δψmdisruption, superoxide generation, and apoptosis. Addition of rIFN-α (104 IU/mL) to the culture medium resulted in a lower number of Th2 cells undergoing Δψm loss and associated HE+/An+ conversion. Indeed, percentages of DiOC6low/HEhigh/An+ cells after 24 hours of culture were 30.5% ± 3.3% and 16.4% ± 0.9% (mean ± SD of 5 determinations) in absence and presence of rIFN-α, respectively (P < .005 by Wilcoxon test). As shown in Figure 6, we also found that an important proportion of the clonal Th2 cells (> 90%) exhibited a drastic Δψmloss 4 hours after Fas cross-linking by sFasL (25 ng/mL). Most of these cells (> 90%) were HE+ and An+. Less than 5% of the clonal Th2 cells were rescued from Fas-mediated Δψm loss and HE+/An+ conversion by rIFN-α added at the beginning of the culture.

Fig. 6.

IFN-α inhibits generation of DiOC6low/HE+ clonal Th2 cells.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured for 24 hours in the absence or presence of sFasL (25 ng/mL). IFN-α (104 IU/mL) was added at the beginning of the culture. At the end of the culture, cells were stained for 15 minutes at 37°C with DiOC6 or HE followed by flow cytometry analysis. The histograms represent DiOC6 (upper) and HE (lower) staining of Th2 cells obtained from patient 1 after 24 hours of culture in the absence (solid lines) or presence (broken lines) of IFN-α.

Fig. 6.

IFN-α inhibits generation of DiOC6low/HE+ clonal Th2 cells.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured for 24 hours in the absence or presence of sFasL (25 ng/mL). IFN-α (104 IU/mL) was added at the beginning of the culture. At the end of the culture, cells were stained for 15 minutes at 37°C with DiOC6 or HE followed by flow cytometry analysis. The histograms represent DiOC6 (upper) and HE (lower) staining of Th2 cells obtained from patient 1 after 24 hours of culture in the absence (solid lines) or presence (broken lines) of IFN-α.

Close modal

To further specify the mechanism of the antiapoptotic effect of IFN-α at the mitochondrial level, we studied the Δψmmodification induced in Th2 cells by PPIX, an agent known to trigger permeability transition (PT) pore opening after binding to benzodiazepine receptors located in the outer mitochondrial membrane.32 After incubation with this agent for 24 hours, we found that more than 60% of the clonal Th2 cells were still alive with a FSChigh/SSClow/Anphenotype. After gating on this population of viable cells, we observed 2 subpopulations of DiOC6low Th2 cells (Figure7), in agreement with a previous study on thymocytes, which demonstrated that the cell population with the lowest DiOC6 fluorescence was the one committed to apoptosis.33 As shown in Figure 7, addition of rIFN-α (104 IU/mL) to PPIX significantly decreased the numbers of cells expressing the lowest DiOC6 fluorescence. Indeed, percentages of those cells after 24 hours of culture were 40 ± 8.9% and 22.6 ± 4.8% (mean ± SEM of 8 determinations) in the absence and presence of rIFN-α, respectively (P < .01 by the Wilcoxon test). Taken together, these data indicate that IFN-α inhibits Δψm decrease induced by PT pore opening in Th2 cells.

Fig. 7.

IFN-α inhibits PPIX-induced Δψm loss in the clonal Th2 cells.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured for 24 hours in presence of PPIX (30 μmol/L). IFN-α (104 IU/mL) was added at the beginning of the culture. At the end of the culture, cells were stained for 15 minutes at 37°C with DiOC6 followed by flow cytometry analysis using gating on live cells. The histograms represent DiOC6 staining of Th2 cells obtained from patient 1 at time 0 (black histogram), after 24 hours of culture in the absence (solid lines) or presence (broken lines) of IFN-α.

Fig. 7.

IFN-α inhibits PPIX-induced Δψm loss in the clonal Th2 cells.

CD3CD4+ cells from hypereosinophilic patient 1 were cultured for 24 hours in presence of PPIX (30 μmol/L). IFN-α (104 IU/mL) was added at the beginning of the culture. At the end of the culture, cells were stained for 15 minutes at 37°C with DiOC6 followed by flow cytometry analysis using gating on live cells. The histograms represent DiOC6 staining of Th2 cells obtained from patient 1 at time 0 (black histogram), after 24 hours of culture in the absence (solid lines) or presence (broken lines) of IFN-α.

Close modal

In this study, we first demonstrated that the clonal Th2-type cells associated with chronic hypereosinophilia displayed a high rate of spontaneous apoptosis on culture in cytokine-free medium. This increased susceptibility to apoptosis was associated with a low level of bcl-2 expression, whereas bcl-xL expression was normal as compared to control cells.

It is well known that members of the IL-2 family that share the γc as part of their receptors34 can prevent the in vitro death of activated T cells.35,36 IL-2 and IL-4 (not shown) indeed prevented spontaneous apoptosis of the clonal Th2 cells. We observed that IFN-α represents another type of survival factor for the clonal Th2 cells. Autocrine production of IL-2 did not contribute to IFN-α–induced survival of the clonal Th2 cells, because this activity was inhibited neither by anti–IL-2R-α nor anti–IL-2R-γ neutralizing antibodies (data not shown). Several studies indicate that bcl-237 and bcl-xL38 repress cell death of cytokine-deprived activated T cells.35 In this report, when clonal Th2 cells were cultured alone, bcl-2 and bcl-xLprotein expression decreased within live cells, suggesting that down-modulation of these antiapoptotic proteins could be involved in the onset of spontaneous apoptosis. Although addition of IL-2 or IFN-α resulted in an improved survival of the clonal Th2 cells, IFN-α had no significant effect on the expression of these antiapoptotic proteins, whereas addition of IL-2 resulted in higher levels of bcl-2, as expected.35 In contrast to IFN-α, IFN-γ did not influence the death rate of the clonal Th2 cells, although these cells bear receptors for IFN-γ because their expression of class I major histocompatibility complex molecules was up-regulated by IFN-γ (data not shown). Because both IFN-γ and IFN-α activate Stat1,39 whereas only IFN-α protects Th2 cells from apoptosis, it seems evident that Stat1 activation might not be sufficient to obtain the antiapoptotic effect in those cells. However, it is also possible that in the case of IFN-γ, a proapoptotic pathway is simultaneously triggered.

To get further insight into the mechanism of action of IFN-α, we evaluated the possible role of mitochondria in the spontaneous apoptosis of the clonal cells. Indeed, recent studies establish that mitochondria play a critical role in the apoptotic cascade leading to T-cell death.27-29 Early after induction of apoptosis, PT-induced pore opening results in loss of Δψm and the generation of superoxide anions by the uncoupled respiratory chain. The hierarchical relationship between these events appears complex. Along this line, Hildeman and colleagues40 established that the production of superoxide anions associated with a loss of Δψm is a decisive contributor to the in vitro death of activated T cells obtained from mice injected with superantigen. Interestingly, it was reported in another work that type I IFNs can act as a survival factor for these cells.41 Moreover, Hyde and coworkers42 demonstrated that a fibroblast-derived factor, recently recognized as IFN-β,43 enhanced survival of activated human T cells by increasing their intracellular antioxidant glutathione levels.

Others works reported that PT pore opening involved in Δψm loss would constitute a sufficient event able to trigger apoptosis in thymocytes and T cells.44,45 In the present study, the capacity of the PT pore inducer PPIX to trigger Δψm disruption as well as apoptosis in the Th2 clonal cells supported this concept. The inhibitory effect of IFN-α on Δψm changes induced by PPIX strongly suggests that IFN-α promotes the survival of the clonal cells at least in part by preventing mitochondrial changes leading to apoptosis.

Our observations are also consistent with studies demonstrating that type I IFNs display antiapoptotic activity on in vivo activated T cells obtained from human inflamed rheumatoid synovium.43,46Also, IFN-α has been reported to prevent spontaneous apoptosis of chronic lymphocytic leukemia cells47-49 and myeloma cells,50 B-cell receptor-mediated apoptosis in B cells,51 as well as Fas-mediated apoptosis of myeloma cells,52 chronic myelogenous leukemia progenitors,53 and antigen-specific human T-cell clones generated in vitro.54 The fact that IFN-α did not affect Fas-mediated apoptosis in clonal Th2 cells from our patients with hypereosinophilia indicates that the influence of IFN-α on the Fas-mediated death pathway might depend on the cell type considered.

The spontaneous apoptosis of the Th2-like cells contrasts with their persistence in vivo, in association with hypereosinophilia and hyper-IgE.14-17 Furthermore, we observed that the Th2 clonal cells showed no sign of apoptosis immediately after they were purified, suggesting that they were not yet committed to death. This might be related to their in vivo exposure to survival factors such as IL-2, IL-4, or IL-15. As shown in a previous study, IL-2 and IL-4 are produced by the clonal Th2 cells on interaction with dendritic cells.16 It is therefore possible that an autocrine loop promoting Th2-cell survival might be operative in vivo as a consequence of their contact with antigen-presenting cells. In the present study, we demonstrate that a type I IFN, IFN-α, represents another survival factor of Th2 cells that might be operative in vivo in a paracrine fashion. Along this line, recent data indicate that fibroblasts producing IFN-β keep alive activated T cells with low bcl-2 that have reverted to a quiescent memory state.55 

Our findings might be clinically relevant because they suggest that the long-term effects of IFN-α therapy in HES might depend on the subtype of the syndrome considered. Initially, the rationale for use of IFN-α in the management of HES was based on its beneficial effects in patients with chronic myelogenous leukemia56 and the fact that some patients with HES presented several features suggestive of a chronic myeloproliferative disease.57,58 Although it is plausible that some patients with HES actually present a true myeloproliferative disorder, current techniques fail to provide evidence for this in most cases. On rare occasions, clonality of eosinophils has been demonstrated in patients fulfilling the diagnostic criteria of HES, leading to the more appropriate diagnosis of chronic eosinophilic leukemia.59,60 The beneficial effects observed with IFN-α in such patients61,62 may result from its suppressive action both on eosinophil progenitors7 and on differentiated eosinophils.8,9,11 The notion that clonal Th2 cells play a pathogenic role in a subgroup of patients with chronic hypereosinophilia has emerged only recently, and most studies reporting favorable effects of IFN-α in the setting of HES have been conducted on patients with no evidence of an underlying T-cell disorder.1-6,12,13,63-67 The long-term effects of IFN-α in such patients are therefore unclear. Indeed, we observed development of peripheral T-cell lymphoma in 2 of 4 patients in our series, both of whom had received IFN-α for over a year.14,17 In another study of 16 hypereosinophilic patients presenting an aberrant population of IL-5–producing cells, T-cell lymphoma was ultimately diagnosed in 3 patients.20 One of these patients had previously been treated with IFN-α, and therapeutic measures were not specified for the 2 others. In view of the antiapoptotic effects of IFN-α reported in the present paper, we suggest carefully evaluating the long-term outcome of patients receiving isolated IFN-α therapy in the context of the Th2 variant of HES.

The technical assistance of Alain Crusiaux and Martine Ducarme is gratefully acknowledged.

Supported by the Télévie program (Belgium). F.R. is a research assistant of the Fonds National de la Recherche Scientifique (Belgium).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Butterfield
JH
Gleich
GJ
Response of six patients with idiopathic hypereosinophilic syndrome to interferon alfa.
J Allergy Clin Immunol.
94
1994
1318
1326
2
Butterfield
JH
Gleich
GJ
Interferon-alpha treatment of six patients with the idiopathic hypereosinophilic syndrome.
Ann Intern Med.
121
1994
648
653
3
Murphy
PT
Fennelly
DF
Stuart
M
O'Donnell
JR
Alfa-interferon in a case of hypereosinophilic syndrome.
Br J Haematol.
75
1990
619
620
4
Zielinski
RM
Lawrence
WD
Interferon-alpha for the hypereosinophilic syndrome.
Ann Intern Med.
113
1990
716
718
5
Bockenstedt
PL
Santinga
JT
Bolling
SF
alpha-Interferon treatment for idiopathic hypereosinophilic syndrome.
Am J Hematol.
45
1994
248
251
6
Fruehauf
S
Fiehn
C
Haas
R
Doehner
H
Hunstein
W
Sustained remission of idiopathic hypereosinophilic syndrome following alpha-interferon therapy.
Acta Haematol.
89
1993
91
93
7
Broxmeyer
HE
Lu
L
Platzer
E
Feit
C
Juliano
L
Rubin
BY
Comparative analysis of the influences of human gamma, alpha and beta interferons on human multipotential (CFU-GEMM), erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) progenitor cells.
J Immunol.
131
1983
1300
1305
8
Aldebert
D
Lamkhioued
B
Desaint
C
et al
Eosinophils express a functional receptor for interferon alpha: inhibitory role of interferon alpha on the release of mediators.
Blood.
87
1996
2354
2360
9
Saito
H
Hayakawa
T
Yui
Y
Shida
T
Effect of influenza virus-sensitized human mononuclear cell supernatant on different functions of human neutrophils and eosinophils: role of interferon.
Arerugi.
35
1986
356
366
10
Nakajima
H
Nakao
A
Watanabe
Y
Yoshida
S
Iwamoto
I
IFN-alpha inhibits antigen-induced eosinophil and CD4+ T cell recruitment into tissue.
J Immunol.
153
1994
1264
1270
11
Morita
M
Lamkhioued
B
Soussi
G
et al
Induction by interferons of human eosinophil apoptosis and regulation by interleukin-3, granulocyte/macrophage-colony stimulating factor and interleukin-5.
Eur Cytokine Netw.
7
1996
725
732
12
Coutant
G
Bletry
O
Prin
L
et al
Treatment of hypereosinophilic syndromes of myeloproliferative expression with the combination of hydroxyurea and interferon alpha. Apropos of 7 cases.
Ann Med Interne (Paris).
144
1993
243
250
13
Kobayashi
M
Katayama
T
Ochiai
S
et al
Interferon-alpha therapy in the myeloproliferative variants of hypereosinophilic syndrome.
Rinsho Ketsueki
34
1993
367
14
Cogan
E
Schandené
L
Crusiaux
A
Cochaux
P
Velu
T
Goldman
M
Brief report: clonal proliferation of type 2 helper T cells in a man with the hypereosinophilic syndrome.
N Engl J Med.
330
1994
535
538
15
Schandené
L
Del Prete
GF
Cogan
E
et al
Recombinant interferon-alpha selectively inhibits the production of interleukin-5 by human CD4+ T cells.
J Clin Invest.
97
1996
309
315
16
Roufosse
F
Schandené
L
Sibille
C
et al
T-cell receptor-independent activation of clonal Th2 cells associated with chronic hypereosinophilia.
Blood.
94
1999
994
1002
17
Roufosse
F
Schandené
L
Sibille
C
et al
Clonal Th2 lymphocytes in patients with the idiopathic hypereosinophilic syndrome.
Br J Haematol.
109
2000
540
548
18
Kitano
K
Ichikawa
N
Mahbub
B
et al
Eosinophilia associated with proliferation of CD(3+)4-(8-) alpha beta+ T cells with chromosome 16 anomalies.
Br J Haematol.
92
1996
315
317
19
Simon
HU
Yousefi
S
Dommann-Scherrer
CC
et al
Expansion of cytokine-producing CD4-CD8- T cells associated with abnormal Fas expression and hypereosinophilia.
J Exp Med.
183
1996
1071
1082
20
Simon
HU
Plötz
SG
Dummer
R
Blaser
K
Abnormal clones of T cells producing interleukin-5 in idiopathic eosinophilia .
N Engl J Med.
341
1999
1112
1120
21
de Waal
M
Yssel
H
Spits
H
et al
Human T cell leukemia virus type I prevents cell surface expression of the T cell receptor through down-regulation of the CD3-gamma, -delta, -epsilon, and -zeta genes.
J Immunol.
145
1990
2297
2303
22
Furukawa
M
Yasukawa
M
Yakushijin
Y
Fujita
S
Distinct effects of human herpesvirus 6 and human herpesvirus 7 on surface molecule expression and function of CD4+ T cells.
J Immunol.
152
1994
5768
5775
23
Willard-Gallo
KE
Van de Keere
F
Kettmann
R
A specific defect in CD3 gamma-chain gene transcription results in loss of T-cell receptor/CD3 expression late after human immunodeficiency virus infection of a CD4+ T-cell line.
Proc Natl Acad Sci U S A.
87
1990
6713
6717
24
Luton
F
Buferne
M
Davoust
J
Schmitt-Verhulst
AM
Boyer
C
Evidence for protein tyrosine kinase involvement in ligand-induced TCR/CD3 internalization and surface redistribution.
J Immunol.
153
1994
63
72
25
Farace
F
Angevin
E
Vanderplancke
J
Escudier
B
Triebel
F
The decreased expression of CD3 zeta chains in cancer patients is not reversed by IL-2 administration.
Int J Cancer.
59
1994
752
755
26
Tartour
E
Latour
S
Mathiot
C
et al
Variable expression of CD3-zeta chain in tumor-infiltrating lymphocytes (TIL) derived from renal-cell carcinoma: relationship with TIL phenotype and function.
Int J Cancer.
63
1995
205
212
27
Green
DR
Reed
JC
Mitochondria and apoptosis.
Science.
281
1998
1309
1312
28
Mignotte
B
Vayssiere
JL
Mitochondria and apoptosis.
Eur J Biochem.
252
1998
1
15
29
Susin
SA
Zamzami
N
Kroemer
G
Mitochondria as regulators of apoptosis: doubt no more.
Biochim Biophys Acta.
1366
1998
151
165
30
Macho
A
Castedo
M
Marchetti
P
et al
Mitochondrial dysfunctions in circulating T lymphocytes from human immunodeficiency virus-1 carriers.
Blood.
86
1995
2481
2487
31
Martin
SJ
Reutelingsperger
CP
McGahon
AJ
et al
Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl.
J Exp Med.
182
1995
1545
1556
32
Pastorino
JG
Simbula
G
Gilfor
E
Hoek
JB
Farber
JL
Protoporphyrin IX, an endogenous ligand of the peripheral benzodiazepine receptor, potentiates induction of the mitochondrial permeability transition and the killing of cultured hepatocytes by rotenone.
J Biol Chem.
269
1994
31041
31046
33
Marchetti
P
Hirsch
T
Zamzami
N
et al
Mitochondrial permeability transition triggers lymphocyte apoptosis.
J Immunol.
157
1996
4830
4836
34
Nakajima
H
Shores
EW
Noguchi
M
Leonard
WJ
The common cytokine receptor gamma chain plays an essential role in regulating lymphoid homeostasis.
J Exp Med.
185
1997
189
195
35
Akbar
AN
Borthwick
NJ
Wickremasinghe
RG
et al
Interleukin-2 receptor common gamma-chain signaling cytokines regulate activated T cell apoptosis in response to growth factor withdrawal: selective induction of anti-apoptotic (bcl-2, bcl-xL) but not pro-apoptotic (bax, bcl-xS) gene expression.
Eur J Immunol.
26
1996
294
299
36
Vella
AT
Dow
S
Potter
TA
Kappler
J
Marrack
P
Cytokine-induced survival of activated T cells in vitro and in vivo.
Proc Natl Acad Sci U S A.
95
1998
3810
3815
37
Korsmeyer
SJ
Chromosomal translocations in lymphoid malignancies reveal novel proto-oncogenes.
Annu Rev Immunol.
10
1992
785
807
38
Boise
LH
Gonzalez-Garcia
M
Postema
CE
et al
bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death.
Cell.
74
1993
597
608
39
Liu
KD
Gaffen
SL
Goldsmith
MA
JAK/STAT signaling by cytokine receptors.
Curr Opin Immunol.
10
1998
271
278
40
Hildeman
DA
Mitchell
T
Teague
TK
et al
Reactive oxygen species regulate activation-induced T cell apoptosis.
Immunity.
10
1999
735
744
41
Marrack
P
Kappler
J
Mitchell
T
Type I interferons keep activated T cells alive.
J Exp Med.
189
1999
521
530
42
Hyde
H
Borthwick
NJ
Janossy
G
Salmon
M
Akbar
AN
Upregulation of intracellular glutathione by fibroblast-derived factor(s): enhanced survival of activated T cells in the presence of low Bcl-2.
Blood.
89
1997
2453
2460
43
Pilling
D
Akbar
AN
Girdlestone
J
et al
Interferon-beta mediates stromal cell rescue of T cells from apoptosis.
Eur J Immunol.
29
1999
1041
1050
44
Marchetti
P
Castedo
M
Susin
SA
et al
Mitochondrial permeability transition is a central coordinating event of apoptosis.
J Exp Med.
184
1996
1155
1160
45
Zamzami
N
Marchetti
P
Castedo
M
et al
Sequential reduction of mitochondrial transmembrane potential and generation of reactive oxygen species in early programmed cell death.
J Exp Med.
182
1995
367
377
46
Salmon
M
Scheel-Toellner
D
Huissoon
AP
et al
Inhibition of T cell apoptosis in the rheumatoid synovium.
J Clin Invest.
99
1997
439
446
47
Chaouchi
N
Wallon
C
Taieb
J
et al
Interferon-alpha-mediated prevention of in vitro apoptosis of chronic lymphocytic leukemia B cells: role of bcl-2 and c-myc.
Clin Immunol Immunopathol.
73
1994
197
204
48
Jewell
AP
Worman
CP
Lydyard
PM
Yong
KL
Giles
FJ
Goldstone
AH
Interferon-alpha up-regulates bcl-2 expression and protects B-CLL cells from apoptosis in vitro and in vivo.
Br J Haematol.
88
1994
268
274
49
Panayiotidis
P
Ganeshaguru
K
Jabbar
SA
Hoffbrand
AV
Alpha-interferon (alpha-IFN) protects B-chronic lymphocytic leukaemia cells from apoptotic cell death in vitro.
Br J Haematol.
86
1994
169
173
50
Ferlin-Bezombes
M
Jourdan
M
Liautard
J
Brochier
J
Rossi
JF
Klein
B
IFN-alpha is a survival factor for human myeloma cells and reduces dexamethasone-induced apoptosis.
J Immunol.
161
1998
2692
2699
51
Su
L
David
M
Inhibition of B cell receptor-mediated apoptosis by IFN.
J Immunol.
162
1999
6317
6321
52
Egle
A
Villunger
A
Kos
M
et al
Modulation of Apo-1/Fas (CD95)-induced programmed cell death in myeloma cells by interferon-alpha 2.
Eur J Immunol.
26
1996
3119
3126
53
Selleri
C
Sato
T
Del Vecchio
L
et al
Involvement of Fas-mediated apoptosis in the inhibitory effects of interferon-alpha in chronic myelogenous leukemia.
Blood.
89
1997
957
964
54
Scheel-Toellner
D
Pilling
D
Akbar
AN
et al
Inhibition of T cell apoptosis by IFN-beta rapidly reverses nuclear translocation of protein kinase C-delta.
Eur J Immunol.
29
1999
2603
2612
55
Gombert
W
Borthwick
NJ
Wallace
DL
et al
Fibroblasts prevent apoptosis of IL-2-deprived T cells without inducing proliferation: a selective effect on Bcl-XL expression.
Immunology.
89
1996
397
404
56
Talpaz
M
Kantarjian
HM
McCredie
K
Trujillo
JM
Keating
MJ
Gutterman
JU
Hematologic remission and cytogenetic improvement induced by recombinant human interferon alpha A in chronic myelogenous leukemia.
N Engl J Med.
314
1986
1065
1069
57
Bain
BJ
Eosinophilic leukaemias and the idiopathic hypereosinophilic syndrome.
Br J Haematol.
95
1996
2
9
58
Weller
PF
Bubley
GJ
The idiopathic hypereosinophilic syndrome.
Blood.
83
1994
2759
2779
59
Luppi
M
Marasca
R
Morselli
M
Barozzi
P
Torelli
G
Clonal nature of hypereosinophilic syndrome [letter].
Blood.
84
1994
349
350
60
Weide
R
Rieder
H
Mehraein
Y
et al
Chronic eosinophilic leukaemia (CEL): a distinct myeloproliferative disease.
Br J Haematol.
96
1997
117
123
61
Luciano
L
Catalano
L
Sarrantonio
C
Guerriero
A
Califano
C
Rotoli
B
AlphaIFN-induced hematologic and cytogenetic remission in chronic eosinophilic leukemia with t(1;5).
Haematologica.
84
1999
651
653
62
Yamada
O
Kitahara
K
Imamura
K
Ozasa
H
Okada
M
Mizoguchi
H
Clinical and cytogenetic remission induced by interferon-alpha in a patient with chronic eosinophilic leukemia associated with a unique t(3;9;5) translocation.
Am J Hematol.
58
1998
137
141
63
Quiquandon
I
Claisse
JF
Capiod
JC
Delobel
J
Prin
L
alpha-Interferon and hypereosinophilic syndrome with trisomy 8: karyotypic remission [letter].
Blood.
85
1995
2284
2285
64
Schoffski
P
Ganser
A
Pascheberg
U
Busche
G
Gaede
B
Hertenstein
B
Complete haematological and cytogenetic response to interferon alpha-2a of a myeloproliferative disorder with eosinophilia associated with a unique t(4;7) aberration.
Ann Hematol.
79
2000
95
98
65
Malbrain
ML
Van den Bergh
H
Zachée
P
Further evidence for the clonal nature of the idiopathic hypereosinophilic syndrome: complete haematological and cytogenetic remission induced by interferon-alpha in a case with a unique chromosomal abnormality.
Br J Haematol.
92
1996
176
183
66
da Silva
MA
Heerema
N
Schwenk
GRJ
Hoffman
R
Evidence for the clonal nature of hypereosinophilic syndrome.
Cancer Genet Cytogenet.
32
1988
109
115
67
Ceretelli
S
Capochiani
E
Petrini
M
Interferon-alpha in the idiopathic hypereosinophilic syndrome: consideration of five cases.
Ann Hematol.
77
1998
161
164

Author notes

Michel Goldman, Department of Immunology, Hôpital Erasme 808, route de Lennik, B-1070 Brussels, Belgium; e-mail: mgoldman@ulb.ac.be.

Sign in via your Institution