CD95 (Fas)-induced apoptosis plays a critical role in the elimination of activated lymphocytes and induction of peripheral tolerance. Defects in CD95/CD95L (Fas-Ligand)-apoptotic pathway have been recognized in autoimmune lymphoproliferative diseases (ALPS) and lpr or gld mice and attributed to CD95 and CD95L gene mutations, respectively. Large granular lymphocyte (LGL) leukemia is a chronic disease characterized by a proliferation of antigen-activated cytotoxic T lymphocytes. Autoimmune features such as hypergammaglobulinemia, rheumatoid factor, and circulating immune complexes are common features in LGL leukemia and ALPS. Therefore, we hypothesize that expansion of leukemic LGL may be secondary to a defective CD95 apoptotic pathway. In this study, we investigated expression of CD95 and CD95L in 11 patients with CD3+ LGL leukemia and explored the apoptotic response to agonistic CD95 monoclonal antibody (MoAb). We found that leukemic LGL from each patient expressed constitutively high levels of CD95/CD95L, similar to those seen in normal activated T cells. However, cells from 9 of these 11 patients were totally resistant to anti-CD95–induced apoptosis. Similarly, cells were resistant to anti-CD3-MoAb–triggered cell death. Lack of anti-CD95–induced apoptosis was not due to mutations in the CD95 antigen. Leukemic LGL were not intrinsically resistant to CD95-dependent death, because LGL from all but 1 patient underwent apoptosis after phytohemagglutinin/interleukin-2 activation. The patient whose leukemic LGL were intrinsically resistant to CD95 had an aggressive form of LGL leukemia that was resistant to combination chemotherapy. These findings that leukemic LGL are resistant to CD95-dependent apoptosis despite expressing high levels of CD95 are similar to observations made in CD95L transgenic mice. These data suggest that LGL leukemia may be a useful model of dysregulated apoptosis causing human malignancy and autoimmune disease.

LARGE GRANULAR lymphocyte (LGL) leukemia can be classified into CD3+ (T cell) and CD3 (natural killer [NK] cell) type depending on the cell lineage of the leukemic cells.1 Autoimmune manifestations are a prominent and characteristic feature of T-LGL leukemia. Serologic abnormalities are frequent, including autoantibodies such as rheumatoid factor and antinuclear antibody, as well as high levels of circulating immune complexes and polyclonal hypergammaglobulinemia.1-5 Autoimmune disease, particularly rheumatoid arthritis, also occurs frequently in LGL leukemia. Increased numbers of LGL could be explained either by stimulation of proliferation or by inhibition of apoptosis. Circulating leukemic LGL are in the Go/G1 phase of the cell cycle6; therefore, we hypothesize that extended cell survival may be secondary to defects in apoptosis.

Leukemic LGL show many characteristics of antigen-activated T cells.6 The physiological deletion of antigen-activated T cells occurs through apoptosis, mediated through CD95 antigen (Fas).7,8 CD95 is a transmembrane protein, belonging to the tumor necrosis receptor family.9 Ligation of CD95 by CD95 Ligand (FasL) or by anti-CD95 monoclonal antibody (MoAb) induces apoptosis of target cells bearing CD95.10-14CD95/CD95L-triggered apoptosis is involved in control of the immune response, induction of peripheral tolerance, and killing of viral-infected or malignant cells.8,15,16 

A defect in CD95-dependent apoptosis is the underlying pathogenetic mechanism in animal models of lymphoproliferative disorders associated with autoimmune manifestations. Lpr/Lpr mice have mutations in CD95, whereas gld/gld mice have mutations in CD95L.17,18 Both animal models are characterized by hypergammaglobulinemia, rheumatoid factor, and circulating immune complexes, features similar to those observed in LGL leukemia.19,20 Dysregulation of CD95/CD95L is also seen in CD95L transgenic mice.21 In this model, high levels of CD95L result in the selection of a novel population of activated T cells that express high levels of CD95, but that are resistant to CD95-mediated apoptosis.

In this study, we examined expression of CD95 and CD95L by leukemic LGL and evaluated whether leukemic LGL were susceptible to CD95 and T-cell receptor (TCR)-induced apoptosis. We found that leukemic LGL expressed high levels of CD95 and CD95L, similar to levels seen on normal activated T cells. Despite high CD95 expression, leukemic LGL were resistant to CD95 or TCR-triggered cell death.

Patients.

All patients met clinical criteria of T-LGL leukemia, with LGL counts ranging from 600 to 27,000/μL (normal, 223 ± 99/μL) and evidence of clonal TCR gene rearrangement.1 Clinical and laboratory features of these patients are shown in Table 1. Nine patients had chronic disease not requiring treatment. Patient no. 5 was receiving methotrexate for neutropenia and rheumatoid arthritis; on therapy, the neutrophil count increased into the normal range. Patient no. 10 had an aggressive form of LGL leukemia, refractory to methotrexate. He presented with massive, painful hepatomegaly with increased circulating LGL (16 × 109/L) and thrombocytopenia (20 to 30 × 109 platelets/L). This patient did not have γδ T-cell lymphoma, because the leukemic cells were αβ+ and γδ as determined by MoAb staining. Four cycles of CHOP produced no response.

Table 1.

Clinical and Laboratory Features of LGL Leukemia Patients

Patient No. Age/Sex Splenomegaly*RA WBCANC ALC HCTPLT
1  29/F  Yes  No 30,800  310  30,000  26  365,000  
2  68/M  No No  3,800  1,600  1,630  34  158,000  
3  79/M No  No  2,700  100  2,450  31  145,000  
66/M  Yes  No  15,400  8,800  5,100  31 368,000  
5  58/M  Yes  Yes  17,400  3,800 13,050  42  221,000  
6  75/M  No  No  15,700 1,700  13,700  27  386,000  
7  67/M  No  No 12,200  2,400  9,300  42  206,000  
8  68/M  No No  4,500  720  3,375  24  95,000  
9  60/M No  Yes  3,200  1,700  1,000  36  173,000  
10 56/M  Yes  No  15,800  1,100  11,500  30 96,000  
11  63/F  No  No  6,800  270  6,100 37  189,000 
Patient No. Age/Sex Splenomegaly*RA WBCANC ALC HCTPLT
1  29/F  Yes  No 30,800  310  30,000  26  365,000  
2  68/M  No No  3,800  1,600  1,630  34  158,000  
3  79/M No  No  2,700  100  2,450  31  145,000  
66/M  Yes  No  15,400  8,800  5,100  31 368,000  
5  58/M  Yes  Yes  17,400  3,800 13,050  42  221,000  
6  75/M  No  No  15,700 1,700  13,700  27  386,000  
7  67/M  No  No 12,200  2,400  9,300  42  206,000  
8  68/M  No No  4,500  720  3,375  24  95,000  
9  60/M No  Yes  3,200  1,700  1,000  36  173,000  
10 56/M  Yes  No  15,800  1,100  11,500  30 96,000  
11  63/F  No  No  6,800  270  6,100 37  189,000 

All cell counts are per microliter and represent blood values at the time of laboratory studies.

Abbreviations: RA, rheumatoid arthritis; WBC, white blood cell counts; ANC, absolute neutrophil count; ALC, absolute lymphocyte count; HCT, hematocrit (%); PLT, platelet count.

*

Patients no. 1 and 10 had prior splenectomies for symptomatic splenomegaly; patient no. 4 had splenectomy for ITP.

Patients no. 4, 6, and 8 were transfusion-dependent.

CD95 and CD95L expression.

Fresh peripheral blood mononuclear cells (PBMC) were obtained from the 11 LGL leukemia patients and from the buffy coat of healthy donors. PBMC were isolated by Ficoll-Hypaque (Pharmacia Biotech, Uppsala, Sweden) gradient centrifugation and then analyzed by flow cytometry (FacScan; Becton Dickinson, Mountain View, CA).

Analysis of CD95 surface expression was performed using phycoerythrin (PE)-conjugated UB2 MoAb (Kamiya Biomedical Corp, Tukwila, WA). To measure CD95 expression on LGL leukemic cells, the cells were incubated with UB2 MoAb and anti-CD57-fluorescein isothiocyanate (FITC) MoAb (Immunotech, Marseille, France). Negative isotype control MoAbs were IgG1-PE (Immunotech) and IgM-FITC (Dako, Copenhagen, Denmark). PBMC from healthy donors and leukemic LGL were cultured in RPMI medium (Fischer Scientific, Pittsburgh, PA) supplemented with 10% fetal calf serum and activated for 2 days with 1 μg/mL phytohemagglutinin (PHA; Sigma, St Louis, MO) and for 10 additional days with recombinant interleukin-2 (IL-2; 100 U/mL; Chiron, Emeryville, CA). CD95 expression was determined before and after activation. For determination of CD95 expression on normal CD57+ T cells or NK cells, three-color staining was performed using MoAbs: CD95-PE UB2, CD57-FITC, and CD3-Cy5 (Immunotech). CD3+/CD57+ and CD3/CD57+ cells were gated for determination of CD95 expression.

Detection of CD95L expression required intracellular staining as previously described.22,23 PBMC were washed twice with phosphate-buffered saline (PBS) and fixed for 10 minutes at 4°C with 0.5% paraformaldehyde-PBS. After centrifugation, PBMC were resuspended in 0.1% Triton X-100–PBS for 3 minutes. PBMC were then washed and incubated with 2 μg anti-CD95L C-20 MoAb (Santa Cruz Biotechnology, Santa Cruz, CA) for 30 minutes at 4°C and then rinsed three times in PBS containing 1% bovine serum albumin and 0.1% sodium azide. Nonspecific binding sites were blocked for 30 minutes with 20% normal swine serum. The cells were washed again and incubated with an FITC-conjugated swine antirabbit Ig MoAb (Dako) for 30 minutes at 4°C. Normal rabbit Ig (Dako) diluted to the same protein concentration as the primary antibody was used as a negative control. After washing, the cells were analyzed using flow cytometry.24,25 

Apoptosis assay.

For the apoptosis assays, unactivated or activated PBMC were transferred to a 96-well culture plate at a concentration of 5 × 105/mL. The cells were then incubated with 1 μg/mL of anti-CD95 MoAb (CH 11; Kamiya Biomedical Corp) or 10 μg/mL of anti-CD3 MoAb (BC3; kindly provided by C. Anasetti, FHCRC, Seattle, WA) for 24 or 48 hours. The same conditions were used to induce cell death induced by 50 μmol/L of C2-ceramide (Sigma). Determination of apoptosis was performed by staining with 7-amino-actinomycin D (7-AAD; Calbiochem, San Diego, CA) and propidium iodide (PI; Molecular Probes, Inc, Eugene, OR), as described.24,25 For 7-AAD staining, cells were incubated with 7-AAD at a concentration of 20 μg/mL for 30 minutes at 4°C in the dark. The cells were then resuspended in PBS and analyzed using flow cytometry. For PI staining, cells were incubated with 50 μg/mL of PI for 30 minutes at room temperature, after fixation overnight with 70% ethanol. To ensure that apoptotic signal was related specifically to anti-CD95, the cells were preincubated with 500 ng/mL CD95 blocking ZB4 MoAb (Kamiya Biomedical Corp) for 60 minutes before treatment with the apoptosis-inducing MoAb CH11. T-cell leukemia CEM cell line was used as a positive control. CD95- or TCR-specific apoptosis was determined as follows: (% of apoptotic cells in the assay well − % of apoptotic cells in the control well)/(100 − % of apoptotic cells in the control well) × 100.

Cell sorting.

In experiments examining apoptosis of activated LGL, flow cytometry was used to isolate the leukemic CD57+ cells. Cells (12 to 15 × 106) were washed in PBS, resuspended in 300 μL of PBS, and incubated for 30 minutes at 4°C with CD57 FITC MoAb. CD57+ cells were then sorted on a FACStar (Becton Dickinson) cell sorter and directed to apoptosis assay as described above. The purity of enriched CD57+ cells was 93% to 96%.

Western blotting.

Cells were lysed in a buffer composed of 1% Nonidet P-40, 10 mmol/L Tris (pH 7.4), 0.1 mmol/L phenylmethylsulfonyl fluoride, EDTA, 10 mmol/L iodometacine, 1 μg/mL leupeptin, 1 μg/mL apoprotin, 0.4 mmol/L Na orthovanadate, and antipain for 45 minutes at 4°C. Equal amounts of protein were loaded on sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and transferred to nitrocellulose membrane. After blocking with PBS buffer containing 5% milk and 0.1% tween, the membranes were probed with N-20 anti-CD95L (Santa Cruz Biotechnology) for 2 hours at room temperature followed by horseradish peroxidase (HRP)-conjugated secondary antibody. Membranes were washed and developed using a chemoluminescent detection system (ECL; Amersham Life Science, Arlington Heights, IL).

Analysis of CD95 gene.

Sufficient material was available for mutational analyses of the CD95 coding sequence in 5 patients. These analyses were performed by reverse transcription-polymerase chain reaction (RT-PCR) single-stranded conformation polymorphism (SSCP) followed by cDNA sequencing. Total RNA was extracted from PBMC of LGL leukemia patients or normal subjects and transcribed to cDNA. The open reading frame of the CD95 cDNA was amplified with three overlapping sets of primers and analyzed by SSCP under previously established conditions.26,27 The cytoplasmic region, which encompasses the signal transducing death domain, was further examined for mutations by denaturing gradient gel electrophoresis (DGGE). The cDNAs showing mobility shifts were extracted from the gel and reamplified using the same primer set. PCR products were subcloned into the TA cloning vector pCR2.1 (Invitrogen, La Jolla, CA), and multiple clones were selected for bidirectional sequencing (ALF, Piscataway, NJ).

CD95/CD95L expression.

PBMC from all patients expressed constitutively high levels of surface CD95 and CD95L protein (Table 2). The mean percentage of CD95+ LGL cells was 88% ± 7%, similar to that of activated T cells (94% ± 3%) and much higher than that in normal PBMC, in which CD95 is expressed at a relatively low level (35% ± 11%). Almost 80% of leukemic CD57+ cells coexpressed CD95 (Fig 1), whereas in normal PBMC, only a minority of CD57+ cells expressed CD95 (32% ± 12%). Because normal CD57+ cells may be either CD3 or CD3+, we performed three-color analysis of normal PBMC to further delineate CD95 expression. We found that CD95 was expressed on 32% ± 10% of normal CD3+, CD57+ cells (n = 10). Therefore, there was a much higher frequency of CD95 expression on leukemic LGL compared with their normal CD3+, CD57+ counterparts. It is of interest that the percentage of normal CD57+ cells expressing CD95 after activation was similar to that seen constitutively in leukemic LGL. Figure 2 shows the results of flow cytometry detection of CD95L. As previously described, CD95L was expressed on normal PBMC only after activation. In contrast, we found constitutive expression of CD95L on PBMC from all LGL leukemia patients. The level of expression of CD95L on leukemic LGL appeared higher than that observed on activated normal T cells. The mean fluorescence intensity of CD95L was 3.9 (range, 1.7 to 7) in leukemic LGL, as compared with 1.5 (range, 1.43 to 1.6) in activated PBMC. Western blot analysis of whole cell lysates confirmed the elevated levels of CD95L in leukemic LGL compared with normal activated PBMC (not shown).

Table 2.

Expression of CD95 and CD95L on Leukemic LGL

Samples Phenotype (%)
CD95CD57 CD57/CD95*CD95L
PBMC  35 ± 11 6 ± 2  32 ± 12  2 ± 1  
PBMC post-PHA/IL-2 94 ± 3   9 ± 2  90 ± 3   42 ± 5  
CD3+ LGL leukemia (patient no.)  
 1  96 44  80  90  
 2  78  30  62  43  
 3  85 74  85  ND  
 4  95  65  84  44 
 5  95  52  93  90  
 6  78  33  77  79 
 7  84  76  84  35  
 8 96  15  72 80  
 9  91  41  82  50  
 10 87  12 64  89  
 11  82  38  63  88  
Mean ± SD 88 ± 7   44 ± 22  77 ± 10 69 ± 23 
Samples Phenotype (%)
CD95CD57 CD57/CD95*CD95L
PBMC  35 ± 11 6 ± 2  32 ± 12  2 ± 1  
PBMC post-PHA/IL-2 94 ± 3   9 ± 2  90 ± 3   42 ± 5  
CD3+ LGL leukemia (patient no.)  
 1  96 44  80  90  
 2  78  30  62  43  
 3  85 74  85  ND  
 4  95  65  84  44 
 5  95  52  93  90  
 6  78  33  77  79 
 7  84  76  84  35  
 8 96  15  72 80  
 9  91  41  82  50  
 10 87  12 64  89  
 11  82  38  63  88  
Mean ± SD 88 ± 7   44 ± 22  77 ± 10 69 ± 23 

Abbreviations: PBMC, peripheral blood mononuclear cells from normal controls, n = 6; ND, not determined.

*

Percentage of CD57+ cells expressing CD95 is indicated.

Sixty-one percent of the cells were CD16+.

Thirty-eight percent of the cells were CD56+.

Fig. 1.

Representative flow cytometry results showing coexpression of CD95 on CD57+ leukemic LGL from patient no. 9. Ninety-one percent and 41% of the cells express CD95 (graph on the left) and CD57 (graph on the center), respectively. In dual fluorescence, 82% of the CD57+ cells coexpress CD95 (graph on the right).

Fig. 1.

Representative flow cytometry results showing coexpression of CD95 on CD57+ leukemic LGL from patient no. 9. Ninety-one percent and 41% of the cells express CD95 (graph on the left) and CD57 (graph on the center), respectively. In dual fluorescence, 82% of the CD57+ cells coexpress CD95 (graph on the right).

Close modal
Fig. 2.

Determination of CD95L expression on normal PBMC (before and after PHA/IL-2 activation) and in 2 cases of freshly isolated LGL leukemia (cases no. 4 and 11). The cells were fixed and permeabilized and then stained with anti-CD95L MoAb (C-20) followed by secondary antirabbit-FITC MoAb. The shaded area represents the level of fluorescence obtained with an isotype control MoAb, the unshaded area represents the level with CD95L MoAb.

Fig. 2.

Determination of CD95L expression on normal PBMC (before and after PHA/IL-2 activation) and in 2 cases of freshly isolated LGL leukemia (cases no. 4 and 11). The cells were fixed and permeabilized and then stained with anti-CD95L MoAb (C-20) followed by secondary antirabbit-FITC MoAb. The shaded area represents the level of fluorescence obtained with an isotype control MoAb, the unshaded area represents the level with CD95L MoAb.

Close modal
Results of apoptosis assay (Table3 ).

Freshly isolated PBMC from 10 of 11 patients showed no apoptosis after 24 hours of exposure to anti-CD95 MoAb (Fig3). After prolonged incubation with anti-CD95 MoAb for 48 hours, cells still remained resistant, except for patient no. 3. In this patient, 31% of the cells were apoptotic. Similar results were seen after 48 hours of anti-CD3 MoAb activation. Absence of apoptosis was observed in 9 of 10 patients, with only patient no. 3 showing slight susceptibility with 28% apoptotic cells (not shown). Ceramide-induced cell death was detected in all 8 cases studied and was comparable to normal PBMC (47% ± 13% v 42% ± 11%, respectively; not shown).

Table 3.

Results of CD95-Dependent Apoptosis in LGL Leukemia

CD3+ LGL Leukemia (patient no.)% Specific Cell Death After Anti-CD95 MoAb Exposure
Freshly Isolated Post-PHA/IL-2
1  34  50  
<5  33  
3  <5  56  
4  <5  43  
<5  53  
6  <5  53  
7  <5  57  
<5  30  
9  <5  45  
10  <5  <5  
11 <5  76 
CD3+ LGL Leukemia (patient no.)% Specific Cell Death After Anti-CD95 MoAb Exposure
Freshly Isolated Post-PHA/IL-2
1  34  50  
<5  33  
3  <5  56  
4  <5  43  
<5  53  
6  <5  53  
7  <5  57  
<5  30  
9  <5  45  
10  <5  <5  
11 <5  76 

Percentage of specific cell death was calculated as described in the Patients and Methods. Anti-CD95 MoAb was added for 24 hours. As positive controls for apoptosis assays, CEM cell line and normal activated T cells were used. For CEM, apoptosis was 60% ± 5% (n = 5). For normal activated T cells, 50% ± 15% cell death was observed (n = 5).

Fig. 3.

Freshly isolated LGL are resistant to CD95 and anti-CD3–mediated apoptosis. CEM-sensitive cell line was used as a positive control for CD95-induced apoptosis (top panel). The cells were incubated with media alone (histograms on the left) or with anti-CD95 (CH11, 1 μg/mL) for 24 and 48 hours, as shown. The cells were then stained with 7-AAD and analyzed by flow cytometry. The LGL leukemic cells are analyzed in the bottom panel. The percentage of apoptotic cells is indicated in each histogram. Although leukemic LGL were resistant to both anti-CD95– and anti-CD3–induced apoptosis (BC3, 10 μg/mL), they remained susceptible to ceramide.

Fig. 3.

Freshly isolated LGL are resistant to CD95 and anti-CD3–mediated apoptosis. CEM-sensitive cell line was used as a positive control for CD95-induced apoptosis (top panel). The cells were incubated with media alone (histograms on the left) or with anti-CD95 (CH11, 1 μg/mL) for 24 and 48 hours, as shown. The cells were then stained with 7-AAD and analyzed by flow cytometry. The LGL leukemic cells are analyzed in the bottom panel. The percentage of apoptotic cells is indicated in each histogram. Although leukemic LGL were resistant to both anti-CD95– and anti-CD3–induced apoptosis (BC3, 10 μg/mL), they remained susceptible to ceramide.

Close modal

After activation with PHA and IL-2, PBMC from 10 of 11 patients showed susceptibility to anti-CD95, with a mean percentage of apoptotic cells of 50% (range, 30% to 76%). Likewise, exposure to anti-CD3 MoAb induced an apoptotic response in 10 of 11 patients, with a mean percentage of apoptotic cells of 45% (range, 29% to 63%). The apoptotic effect of anti-CD95 MoAb was specifically inhibited by ZB4 MoAb (Fig 4). To ensure that apoptosis was occurring in LGL leukemic cells rather than in normal cells expanded after 10 days of IL-2 activation, CD57+ cells from 3 patients were sorted after activation and exposed to anti-CD95 and anti-CD3 MoAb. Purity after cell sorting was greater than 93% CD57+ cells in each case. Figure 5 shows that 76% of the CD57+ cells underwent apoptosis after anti-CD95 MoAb in patient no. 4. Similar results were seen after activation with anti-CD3 MoAb (not shown). The same data were obtained with the CD57+-sorted cells from patients no. 5 and 7.

Fig. 4.

Protection of CD95-induced apoptosis by CD95 blocking MoAb ZB 4. The activated PBMC of patient no. 11 were preincubated with ZB 4 (500 ng/mL for 60 minutes) and then exposed to anti-CD95 for 48 hours. The cells were incubated with 50 μg/mL of PI for 30 minutes at room temperature, after fixation overnight with 70% ethanol, and the DNA content was analyzed using flow cytometry. Histograms on the left, center, and right represent the results after incubation with serum alone, anti-CD95, and ZB4 + anti-CD95, respectively. The percentage of apoptotic cells is indicated on the left side of each panel.

Fig. 4.

Protection of CD95-induced apoptosis by CD95 blocking MoAb ZB 4. The activated PBMC of patient no. 11 were preincubated with ZB 4 (500 ng/mL for 60 minutes) and then exposed to anti-CD95 for 48 hours. The cells were incubated with 50 μg/mL of PI for 30 minutes at room temperature, after fixation overnight with 70% ethanol, and the DNA content was analyzed using flow cytometry. Histograms on the left, center, and right represent the results after incubation with serum alone, anti-CD95, and ZB4 + anti-CD95, respectively. The percentage of apoptotic cells is indicated on the left side of each panel.

Close modal
Fig. 5.

Flow cytometry results showing that leukemic LGL are susceptible to anti-CD95–induced apoptosis after activation. The PBMC of patient no. 4 were cultured initially with PHA (1 μg/mL) for 2 days and then with IL-2 (100 U/mL) for 10 more days. The cells were then stained with CD57+ and sorted on FacStar. The purified CD57+ cells (94%) were then incubated with anti-CD95 MoAb (CH11, 1 μg/mL) for 48 hours and stained with 7-AAD before analysis using flow cytometry. The graph on the left represents the control (media alone); the graph on the right represents the cells incubated with CH11. The percentage of apoptotic cells is shown on the upper-right quadrant.

Fig. 5.

Flow cytometry results showing that leukemic LGL are susceptible to anti-CD95–induced apoptosis after activation. The PBMC of patient no. 4 were cultured initially with PHA (1 μg/mL) for 2 days and then with IL-2 (100 U/mL) for 10 more days. The cells were then stained with CD57+ and sorted on FacStar. The purified CD57+ cells (94%) were then incubated with anti-CD95 MoAb (CH11, 1 μg/mL) for 48 hours and stained with 7-AAD before analysis using flow cytometry. The graph on the left represents the control (media alone); the graph on the right represents the cells incubated with CH11. The percentage of apoptotic cells is shown on the upper-right quadrant.

Close modal
Analysis of CD95 gene mutation.

Function ablating mutations in the CD95 gene have been shown to be causative in some autoimmune diseases. Using RT-PCR SSCP and DGGE analysis, followed by cDNA sequencing of PCR products, we examined the CD95 coding sequence to determine if mutations might account for the failure of LGL leukemia cells to undergo apoptosis when exposed to anti-CD95 MoAb. No mutations were detected in the CD95 coding sequence of the LGL leukemia patients studied. Four of the five patients examined expressed a previously documented polymorphism at bp 836, which does not alter the amino acid sequence.

CD3+ LGL leukemia is a chronic clonal T-cell lymphoproliferative disorder recognized as a distinct entity using clinical, immunological, and molecular parameters.1,2,28The clonal expansion of leukemic LGL may require a multistep pathogenesis. Leukemic LGL have many characteristics of antigen-activated cytotoxic T lymphocytes (CTL): (1) They express a T-cell cytotoxic phenotype and can be activated via the CD3/CD16 pathway.6,29 (2) They constitutively express perforin and CD95L.30,31 (3) At least in some cases, they use a restricted Vβ repertoire, reinforcing the hypothesis of antigenic selection.32 Increased numbers of LGL could be explained either by a stimulation of proliferation or inhibition of programmed cell death. CD95/CD95L interactions play a major role in the induction of cell death after T-cell activation.8-11 CD95 is weakly expressed on the surface of resting T cells and is upregulated after antigen activation.9,14 The accumulation of peripheral T cells might result from a defect in removing antigen-activated T cells. Therefore, expansion of LGL leukemic cells could be due to a defect in the CD95 apoptotic pathway.

We studied the CD95/CD95L apoptotic pathway in 11 cases of T-LGL leukemia. We found a constitutively high surface expression of CD95 and CD95L in all 11 cases at levels similar to, if not greater than, those seen in normal activated T cells. LGL specifically expressed CD95, because almost 80% of CD57+ LGL coexpressed CD95. Despite this high level of CD95 expression and evidence of a constitutively activated T-cell phenotype, freshly isolated cells from 9 of 11 patients were completely resistant to anti-CD95–induced apoptosis. In the 2 remaining patients (patients no. 1 and 3), CD95-induced apoptosis was lower than levels seen in control cells, especially in patient no. 3, whose cells displayed a slight apoptotic response only after 2 days of anti-CD95 MoAb exposure. A similar pattern of resistance to TCR-triggered cell death was also observed. Normal T cells become sensitive to apoptosis after activation, when expression of high level of CD95 and CD95L is observed. Our observations that leukemic LGL is resistant to apoptosis despite expressing high levels of both CD95 and CD95L suggest that this apoptotic pathway is dysregulated in LGL leukemia.

These results of CD95 resistance are similar to findings observed in animal models of lymphoproliferation and autoimmune disease occurring in lpr and gld mice.19,20 The pathogenesis of CD95 resistance in these murine models is due to mutations in CD95 and CD95L, respectively.17,18 Recently, resistance to anti-CD95–induced apoptosis has been described in a human disease, termed autoimmune lymphoproliferative syndrome (ALPS).33-37The clinical and biological features of ALPS are very similar to those observed in LGL leukemia, including the following: (1) The majority of the uniquely expanded CD4, CD8cells in ALPS are CD57+.33 (2) The patients present with splenomegaly, hypergammaglobulinemia, and autoimmune hemolytic anemia, thrombocytopenia, or neutropenia. CD95 gene mutations primarily involving the death domain have been described in these patients. We previously reported the absence of mutations in the CD95 death domain in seven patients.31 However, some patients with ALPS have had mutations in regions of CD95 other than the death domain.34 For these reasons, we examined the entire CD95 antigen cDNA in the LGL leukemia patients. We found no evidence for CD95 mutation in each of the 5 patients studied. We have also found no evidence for CD95 mutation in 4 additional patients (unpublished observations). Therefore, dysregulated CD95-dependent apoptosis in LGL leukemia does not result from mutations in CD95. Recently, an autoimmune lymphoproliferative disease (ALD) has been described with generalized autoimmune manifestations but without expansion of dual CD4/CD8-negative T cells.38Interestingly, the CD95+ cells are resistant to anti-CD95–induced apoptosis but do not display any CD95 gene mutations. Ceramide is the second messenger produced by hydrolysis of sphingomyelin after a CD95 apoptotic signal.39 In ALD patients, ceramide induced cell death is deficient, suggesting a downstream alteration of the apoptosis pathway.38Ceramide-induced apoptosis was normal in LGL leukemia patients, suggesting that the cause of apoptotic resistance is different in LGL leukemia compared with ALD patients.

The mechanism resulting in resistance to CD95-induced apoptosis in LGL leukemia is not known. From our study we do know that resistance is not due to lack of CD95 surface expression or mutant CD95 protein, as seen in myeloma samples or myeloma cells lines.25,27 We also know that there is not an intrinsic defect in apoptosis in LGL leukemia, because leukemic cells from 10 of 11 patients underwent apoptosis after activation with IL-2. It is conceivable that lack of IL-2 in vivo might explain the apoptotic resistance. IL-2 predisposes peripheral T cells to CD95 and anti-CD3/TCR–induced cell death.14 IL-2–deficient mice (IL-2−/−) develop a fatal disease with lymphadenopathy, splenomegaly, and multiorgan T-cell infiltration.40 Activated T cells from these IL-2−/− mice display a CD95-resistant phenotype, although they express CD95 similarly to cells from their IL-2+/+ littermates. Although LGL leukemic cells constitutively express p75 IL-2 receptor, they do not produce IL-2 gene transcripts or secrete IL-2 even after anti-CD3 MoAb activation.41,42 

Leukemic cells from 1 patient remained resistant to both CD95 and TCR triggered cell death even after activation with IL-2. No abnormalities were observed in CD95 gene, suggesting that the defect in apoptosis is distal to the receptor. This patient had an aggressive clinical course, presenting with pancytopenia and massive hepatomegaly, which was refractory to combination chemotherapy. We recently reported that chronic LGL leukemias express relatively high levels of multidrug resistance gene (MDR1) and that P-glycoprotein was functionally active in CD57+ leukemic LGL.43 It is of interest that leukemic LGL from this patient showed a high level of P-glycoprotein surface expression (not shown). It has been suggested that chemotherapeutic agents may induce apoptosis through the CD95/CD95L pathway.16 Anthracycline-resistant cell lines are also resistant to CD95-induced apoptosis.25 Therefore, elucidation of the mechanism of resistance to CD95-mediated apoptosis in this patient may also help delineate mechanisms involved in drug resistance.

Our data suggest that LGL leukemia can serve as a useful model of dysregulated apoptosis causing human malignancy and autoimmune disease. Our results showing that leukemic LGL express high levels of CD95 yet are resistant to CD95-mediated apoptosis are similar to findings observed in CD95L transgenic mice. In this animal model, expression of CD95L leads to disease manifestations. High levels of soluble CD95L have been found in sera from LGL leukemia patients.44 Growth of hematopoietic colonies in vitro is negatively regulated by activation of the CD95 pathway.45Mature neutrophils undergo apoptotic death through CD95 triggering.46 Taken together, these data suggest that secretion of CD95 ligand may be a mechanism leading to neutropenia in LGL leukemia. Studies investigating this hypothesis are ongoing in our laboratory.

Supported by the Veterans Administration. T.L. is a recipient of “Association pour la Recherche Contre le Cancer” and “Pharmacia” grants. The Flow Cytometry and Molecular Biology Core laboratories at H. Lee Moffitt Cancer Center and Research Institute were used in the course of this work.

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

1
Loughran
TP
Jr
Clonal diseases of large granular lymphocytes.
Blood
82
1993
1
2
Dhodapkar
MV
Li
CY
Lust
JA
Tefferi
A
Phyliky
RL
Clinical spectrum of clonal proliferations of T-large granular lymphocytes: A T-cell clonopathy of undetermined significance?
Blood
84
1994
1620
3
Semenzato
G
Pandolfi
F
Chiesesi
T
De Rossi
G
Pizzolo
G
Zambello
R
Trentin
L
Agostini
C
Dini
E
Vespignani
M
Cafaro
A
Paqualetti
D
Giubellino
MC
Mignole
N
Foa
R
The lymphoproliferative disease of granular lymphocytes. A heterogeneous disorder ranging from indolent to aggressive conditions.
Cancer
60
1987
2971
4
Oshimi
K
Yamada
O
Kanako
T
Nishinarita
S
Iizuka
Y
Urabe
A
Inamori
T
Asano
S
Takahashi
S
Hattori
M
Naohara
T
Ohira
Y
Togawa
A
Masuda
Y
Okubo
Y
Furusawa
S
Sakamoto
S
Omine
M
Mori
M
Tatsumi
E
Mizoguchi
H
Laboratory findings and clinical courses of 33 patients with large granular lymphocyte proliferative disorders.
Leukemia
7
1993
782
5
Gentile
TC
Wener
MH
Starkebaum
G
Loughran
T
Jr
Humoral immune abnormalities in T-cell large granular lymphocyte leukemia.
Leuk Lymphoma
23
1996
365
6
Aprile
JA
Russo
M
Pepe
MS
Loughran
TP
Jr
Activation signals leading to proliferation of normal and leukemic CD3+ large granular lymphocytes.
Blood
78
1991
1282
7
Itoh
N
Yonehara
S
Ishii
A
Yonehara
M
Mizushima
S
Samashima
M
Hase
A
Seto
Y
Nagata
S
The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis.
Cell
66
1991
233
8
Nagata
S
Apoptotis by death factor.
Cell
88
1997
355
9
Nagata
S
Goldstein
O
The Fas death factor.
Science
267
1995
1449
10
Dhein
J
Walczak
H
Baumier
C
Debatin
KM
Krammer
PH
Autocrine T-cell suicide mediated by APO-1(Fas/CD95).
Nature
373
1995
438
11
Alderson
MR
Tough
TW
Davis-Smith
T
Braddy
S
Falk
B
Schooley
KA
Goodwin
RG
Smith
CA
Rambsdell
F
Lynch
DH
Fas ligand mediates activation-induced cell death in human T lymphocytes.
J Exp Med
181
1995
71
12
Glass
A
Walsh
CM
Lynch
DH
Clarck
WR
Regulation of the Fas lytic pathway in cloned CTL.
J Immunol
156
1996
3638
13
Ortaldo
JR
Winkler-Pickett
RT
Nagata
S
Ware
CF
Fas involvement in human NK cell apoptosis: Lack of a requirement for CD16-mediated events.
J Leukoc Biol
61
1997
209
14
Owen-Schaub
LB
Yonehara
S
Crimp
WL
III
Grimm
EA
DNA fragmentation and cell death is selectively triggered in activated human lymphocytes by Fas antigen engagement.
Cell Immunol
140
1992
197
15
Griffith
TS
Brunner
T
Fletcher
SM
Green
DR
Fergusson
TA
Fas ligand-induced apoptosis as a mechanism of immune privilege.
Science
270
1995
1189
16
Friessen
C
Herr
I
Krammer
PH
Debatin
KM
Involvement of the CD95 (APO-1/Fas) receptor/ligand system in drug-induced apoptosis in leukemic cells.
Nat Med
2
1996
574
17
Watanabe-Fukunaga
R
Brannan
CI
Copeland
NG
Jenkins
NA
Nagata
S
Generalized lymphoproliferative disease in mice explained by defects in Fas antigen that mediates apoptosis.
Nature
356
1992
314
18
Takahashi
T
Tanaka
M
Brannan
CI
Jenkins
NA
Copeland
NG
Suda
T
Nagata
S
Generalized lymphoproliferative disease in mice caused by a point mutation in the Fas ligand.
Cell
76
1994
969
19
Steinberg
AD
MRL-lpr/lpr disease: Theories meet Fas.
Semin Immunol
6
1994
55
20
Cohen
PL
Eisenbert
RA
The lpr and gld genes in systemic autoimmunity: Life and death in the Fas lane.
Immunol Today
13
1992
427
21
Cheng
J
Liu
C
Yang
PA
Zhou
T
Mountz
JD
Increased lymphocyte apoptosis in Fas ligand transgenic mice.
J Immunol
159
1997
674
22
Zipp
F
Martin
R
Lichtenfels
R
Roth
W
Dichgans
J
Krammer
PH
Weller
M
Human autoreactive and foreign antigen-specific T cells resist apoptosis induced by soluble recombinant CD95 ligand.
J Immunol
159
1997
2108
23
Truman
JP
Choqueux
C
Tschopp
J
Vedrenne
J
Le Deist
F
Charron
D
Mooney
N
HLA class II-mediated death is induced via Fas/Fas ligand interactions in human splenic B lymphocytes.
Blood
89
1997
1996
24
Nicoletti
I
Migliorati
G
Pagliacci
MC
Grignani
F
Riccardi
C
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry.
J Immunol Methods
139
1991
271
25
Landowski
TH
Gleason-Guzman
MC
Dalton
WS
Selection of drug resistance results in resistance to Fas mediated apoptosis.
Blood
89
1997
1130
26
Fiucci
G
Ruberti
G
Detection of polymorphisms within the Fas cDNA gene sequence by GC-clamp denaturing gradient gel electrophoresis.
Immunogen
39
1995
437
27
Landowski
TH
Qu
N
Buyuksal
I
Painter
JS
Dalton
WS
Mutations in the Fas antigen in patients with multiple myeloma.
Blood
90
1997
4266
28
Scott
CS
Richards
SJ
Classification of large granular lymphocyte (LGL) and NK-associated (Nka) disorders.
Blood Rev
6
1992
220
29
Hoshiro
S
Oshimi
K
Teramura
M
Mizoguchi
H
Activation via the CD3 and CD16 pathway mediates interleukin-2-dependent autocrine proliferation of granular lymphocytes in patients with granular lymphocyte proliferative disorders.
Blood
78
1991
3232
30
Oshimi
K
Shinkai
Y
Okumura
K
Oshimi
Y
Mizoguchi
H
Perforin gene expression in granular lymphocyte proliferative disorders.
Blood
75
1990
704
31
Perzova
R
Loughran
TP
Jr
Constitutive expression of Fas ligand in large granular lymphocytes leukaemia.
Br J Haematol
97
1997
123
32
Zambello
R
Trentin
L
Facco
M
Cerutti
A
Sancetta
R
Milani
A
Raimondi
R
Tassinari
C
Agostini
C
Semenzato
G
Analysis of the T cell receptor in the lymphoproliferative disease of granular lymphocytes: Superantigen activation of clonal CD3+ granular lymphocytes.
Cancer Res
55
1995
6140
33
Le Deist
F
Emile
JF
Rieux-Laucat
F
Benkerou
M
Roberts
I
Brousse
N
Fisher
A
Clinical, immunological, and pathological consequences of Fas-deficient conditions.
Lancet
348
1996
719
34
Fisher
GH
Rosenberg
FJ
Strauss
SE
Dale
JK
Middleton
LA
Lin
AY
Strober
W
Lenardo
MJ
Puck
JM
Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome.
Cell
81
1995
935
35
Drappa
J
Vaishnaw
AK
Sullivan
KE
Chu
JL
Elkon
KB
Fas gene mutations in the Canale-Smith syndrome, an inherited lymphoproliferative disorder associated with autoimmunity.
N Engl J Med
335
1996
1643
36
Bettinardi
A
Brugnoni
D
Quiros-Roldan
E
Malagoli
A
La Grutta
S
Correra
A
Notarangelo
LD
Missense mutations in the Fas gene resulting in autoimmune lymphoproliferative syndrome: A molecular and immunological analysis.
Blood
89
1997
902
37
Sneller
MC
Wang
J
Dale
JK
Strober
W
Middleton
LA
Choi
Y
Fleisher
TA
Lim
MS
Jaffe
ES
Puck
JM
Lenardo
MJ
Straus
SE
Clinical, immunologic, and genetic features of an autoimmune lymphoproliferative syndrome associated with abnormal lymphocytes apoptosis.
Blood
89
1997
1341
38
Dianzani
U
Bragardo
M
DiFranco
D
Alliaudi
C
Scagni
P
Buonfiglio
D
Redoglia
V
Bonissoni
S
Correra
A
Dianzani
I
Ramenghi
U
Deficiency of the Fas apoptosis pathway without Fas gene mutations in pediatric patients with autoimmunity/lymphoproliferation.
Blood
89
1997
2871
39
Cifone
MG
De Maria
R
Roncaioli
P
Rippo
MR
Azuma
M
Lannier
LL
Santoni
A
Testi
R
Apoptotic signaling through CD95 (Fas/Apo-1) activates an acidic sphingomyelinase.
J Exp Med
180
1994
1547
40
Kneitz
B
Herrmann
T
Yonehara
S
Schimpl
A
Normal clonal expansion but impaired Fas-mediated cell death and anergy induction in interleukin-2-deficient mice.
Eur J Immunol
25
1995
2572
41
Zambello
R
Trentin
L
Pizzolo
G
Bullian
P
Masciarelli
M
Feruglio
C
Agostini
C
Raimondi
R
Chisesi
T
Semenzato
G
Cell membrane expression and functional role of the p75 subunit of interleukin-2 receptor in lymphoproliferative disease of granular lymphocytes.
Blood
76
1990
2080
42
Loughran
TP
Jr
Aprile
JA
Ruscetti
FW
Anti-CD3 monoclonal antibody-mediated cytotoxicity occurs through an interleukin-2 independent pathway in CD3+ large granular lymphocytes.
Blood
75
1990
935
43
Lamy
T
Drenou
B
Fardel
O
Amiot
L
Grulois
I
Le Prise
PY
Loughran
TP
Jr
Fauchet
R
Multidrug resistance analysis in lymphoproliferative disease of large granular lymphocytes.
Br J Haematol
100
1998
509
44
Tanaka
M
Suda
T
Haze
K
Nakamura
N
Sato
K
Kimura
F
Motoyoshi
K
Mizuki
M
Tagawa
S
Ohga
S
Hatake
K
Drummond
AH
Nagata
S
Fas ligand in human serum.
Nat Med
2
1996
317
45
Maciejewski
J
Selleri
C
Anderson
S
Young
NS
Fas antigen expression on CD34+ human marrow cells is induced by interferon gamma and tumor necrosis factor alpha and potentiates cytokine-mediated hematopoietic suppression in vitro.
Blood
85
1995
3183
46
Liles
WC
Kiener
PA
Ledbetter
JA
Aruffo
A
Klebanoff
SJ
Differential expression of Fas (CD95) and Fas Ligand on normal human phagocytes: Implications for the regulation of apoptosis in neutrophils.
J Exp Med
184
1996
429

Author notes

Address reprint requests to Thomas P. Loughran, Jr, MD, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612; e-mail: Loughrat@moffitt.usf.edu.

Sign in via your Institution