Wiskott-Aldrich syndrome (WAS) is an X-linked recessive disorder characterized by thrombocytopenia, eczema, and a progressive deterioration of immune function. WAS is caused by mutations in an intracellular protein, WASP, that is involved in signal transduction and regulation of actin cytoskeleton rearrangement. Because immune dysfunction in WAS may be due to an accelerated destruction of lymphocytes, we examined the susceptibility to apoptosis of resting primary lymphocytes isolated from WAS patients in the absence of exogenous apoptogenic stimulation. We found that unstimulated WAS lymphocytes underwent spontaneous apoptosis at a greater frequency than unstimulated normal lymphocytes. Coincident with increased apoptotic susceptibility, WAS lymphocytes had markedly attenuated Bcl-2 expression, whereas Bax expression did not differ. A negative correlation between the frequency of spontaneous apoptosis and the level of Bcl-2 expression was demonstrated. These data indicate that accelerated lymphocyte destruction by spontaneous induction of apoptosis may be one pathogenic mechanism by which the progressive immunodeficiency in WAS patients develops.

WISKOTT-ALDRICH SYNDROME (WAS) is a rare X-linked recessive disorder.1,2 Affected males are clinically characterized by eczema, thrombocytopenia with platelets of reduced size, immunodeficiency, and an increased susceptibility to hematopoietic malignancy and autoimmunity (Remold-O’Donnell et al3 and references therein). WAS patients suffer from chronic and recurrent opportunistic and viral infections as a consequence of immunologic abnormalities. A progressive deterioration of T-lymphocyte function in affected children accompanies the development of T lymphopenia by 6 years of age.4Cell-mediated immunity is further compromised by poor macrophage and neutrophil motility and chemotactic responses.4-7 WAS patients commonly have low isohemagglutinin titers and a depressed response to polysaccharide antigens3; but, their impaired humoral immunity has been at least partially attributed to T-lymphocyte dysfunction.8 WAS T lymphocytes have diminished proliferative responses to mitogens, some specific antigens, and allogeneic stimulation.3 Moreover, WAS T lymphocytes fail to proliferate or upregulate interleukin-2 gene expression in response to treatment with immobilized anti-CD3 antibody.9,10Deformation and deregulation of the actin cytoskeleton is likely to be the basis for many of the clinical defects associated with WAS, particularly immune dysfunction.3,10,11 WAS lymphocytes are frequently morphologically abnormal, with irregular and bulbous cellular projections,10,12 a paucity of microvilli,13 and a poorly delineated actin cortex.10 CD3-mediated stimulation of WAS T-lymphoblastoid cell lines results in abnormal actin polymerization, marked by the absence of specific cytoskeleton rearrangements that occur in normal T-lymphoblastoid cell lines.10 Epstein-Barr virus–transformed B-cell lines from WAS patients also have marked abnormalities in actin distribution and polymerization.11 

The WAS gene was mapped to the short arm of the X chromosome at Xp11.2214 and identified in 1994.15 The gene encodes a 66-kD intracellular protein, WASP, that is expressed exclusively in blood cells15,16 throughout hematopoiesis.17 More than 138 unique mutations to WASP have been described,18 which include frameshifts and substitutions that generally nullify expression by causing mRNA instability and protein truncation.11,19-21 There is considerable variation in WAS severity; even members of the same kindred may have markedly different phenotypes,22 possibly indicating polygenic or environmental contributions. WAS severity may be partially determined by the level at which WASP is stably expressed.11,19-21 

The function of WASP is unknown; however, it has been surmised that WASP is involved in the regulation of actin polymerization and actin cytoskeleton organization from observations that WAS lymphocytes, platelets, granulocytes, and monocytes are defective in regulating the cortical actin cytoskeleton (Remold-O’Donnell et al3 and references therein). In support of this presumption, N-WASP, a ubiquitous mammalian homolog of WASP, has been demonstrated to directly induce actin depolymerizaton and actin cytoskeleton reorganization in nerve growth factor-stimulated cells.23 Although WASP apparently lacks catalytic activity, the protein colocalizes with actin and interacts with WIP24 and PSTPIP,25 2 proteins that regulate actin polymerization. The Rho family small guanosine triphosphatases (GTPases), Cdc42 and Rac, which have been shown to specifically control actin cytoskeleton reorganization,26 interact with WASP.27-30 WASP also associates with numerous other proteins, including various tyrosine kinases (Btk, Itk, Tec, Fyn, and c-Src), adaptor proteins (Nck and Grb2), and phospholipase Cγ1, all of which are involved in lymphoid signal transduction.31-36 Therefore, it has been proposed that WASP functions as a molecular scaffold28 that docks and aligns other proteins for more specific interaction.37 Thus, WASP may link components of the cytoskeleton with key signal transduction elements to integrate signaling in response to various intrinsic or extrinsic stimuli to regulate actin cytoskeleton reorganization.

A WASP deficiency or the expression of dysfunctional WASP is probably detrimental to the development and survival of hematopoietic cell lineages.3 A uniformly nonrandom pattern of X-chromosome inactivation in all blood cells from WAS obligate heterozygous carriers and platelet loss in WAS patients suggests that mutations to WASP probably confer a growth and/or survival disadvantage to affected cells.3 Consequently, we reasoned that the development of lymphopenia and other progressive immune defects associated with WAS may be caused by an inherently decreased potential for survival and the accelerated destruction of peripheral lymphocytes.

In this study, we compared the levels of spontaneous apoptosis in unstimulated resting lymphocytes isolated from WAS patients and from normal, healthy controls. We found that WAS lymphocytes underwent spontaneous apoptosis at a greater frequency than did normal lymphocytes. Bcl-2 family members regulate the onset of apoptosis, acting as either agonists or inhibitors of programmed cell death.38 Because the deregulation of apoptosis in WAS lymphocytes might be caused by aberrant expression of one or more of the Bcl-2 family members, we determined that the level of Bcl-2 expression in WAS lymphocytes was attenuated. We suggest that the accelerated destruction of lymphocytes by spontaneous apoptosis could account for the progressive deterioration of immune function in WAS patients.

Patient samples.

Samples from 5 male patients previously diagnosed with WAS were used in this study. At the time of analysis, patients no. 1 and 2 were 2.5 and 4 years of age, respectively. Patients no. 3 and 4 are siblings and were 4.5 and 14 years of age, respectively. Patient no. 5 was 14 months of age. Diagnosis of WAS was initially based on family history and presentation of immunodeficiency with recurrent infection, thrombocytopenia, platelets with reduced volume, and eczema. To define the WAS phenotype of the patients, clinical scores were assigned to each according to published recommendations.18,21 Patients no. 1, 2, 3, 4, and 5 were assigned clinical scores of 4, 4, 3, 3, and 3, respectively. The genotypes of 4 of 5 patients have been determined. Patient no. 1 has a point mutation, C290T, resulting in an amino acid substitution of R86C in exon 2. The genotype of patient no. 2 was not determined. Patients no. 3 and 4, the siblings, have a deletion mutation in exon 8 (John Bastian, personal communication, July 1999). Patient no. 5 has a splice site mutation (4-bp deletion in intron 8), resulting in the deletion of exon 8; patient no. 5 does not express WASP. Patients no. 1, 2, and 5 were treated at Childrens Hospital (CHLA) in Los Angeles, CA; each has since received bone marrow transplantations. Patients no. 3 and 4 were treated at Children’s Hospital in San Diego, CA. These studies were performed in accord with protocols approved by the Committee on Clinical Investigations of the Institutional Review Board at CHLA. None of the patients had been splenectomized. Normal controls (8) were obtained from either healthy volunteer adult donors (5) or healthy pediatric patients (3) at CHLA. No significant difference between the normal pediatric and normal adult means was noted (P = .978). Whenever possible, patient and normal blood was collected and processed under paired conditions. For the comparison of normal pediatric and normal adult blood, specimens were collected and processed under paired conditions.

Isolation and in vitro treatment of peripheral blood lymphocytes (PBL).

Peripheral blood was collected from WAS patients or normal, healthy individuals in heparinized Vacutainer tubes (Becton Dickinson, Franklin Lakes, NJ). Within 8 hours, mononuclear cells were isolated from whole blood by Ficoll-Hypaque (Pharmacia Biotech, Piscataway, NJ) density gradient centrifugation. Mononuclear cells were washed in Hanks’ balanced salt solution (HBSS; BioWhittaker, Walkersville, MD) and resuspended in R-10 medium (RPMI-1640 supplemented with 10% heat-inactivated human A+ serum [CHLA Blood Bank, Los Angeles, CA], 2 mmol/L L-glutamine [Gemini BioProducts, Calabasas, CA], 1 × 10−6 mol/L 2-mercaptoethanol [Sigma, St Louis, MO], and 1× penicillin-streptomycin [Gemini BioProducts]). Cells were plated at a density of 5 × 106 cells/mL in 100-mm tissue culture dishes (Corning, Corning, NY) and placed in a 37°C humidified incubator conditioned with 5% CO2 for 1 hour. Nonadherent PBL were collected and resuspended in R-10. PBL were plated at a density of 0.5 × 106 cells/mL in 24-well flat-bottomed dishes (Costar, Cambridge, MA) and maintained in a humidified atmosphere with 5% CO2 at 37°C for up to 10 days. Some experiments were performed after maintaining PBL in R-10 containing 10% heat-inactivated fetal calf serum (Summit Biotechnology, Fort Collins, CO) instead of human serum, but no differences in the apoptotic frequencies of PBL were apparent. PBL were not stimulated in vitro.

Detection of apoptosis.

Either of 2 methods, terminal deoxynucleotidyl transferase-mediated dUTP nicked end labeling (TUNEL) or annexin V-labeling, was used to mark apoptotic cells. For TUNEL,39 cells were harvested and washed in 3 mL phosphate-buffered saline (PBS; Sigma). At least 105 cells were stained on ice and in the dark with fluorochrome-conjugated antihuman antibodies recognizing specific cell surface markers to be used in immunophenotyping. Incubations with the antibodies were performed in PBS containing 0.1% human intravenous Ig (Sandoz Pharmaceuticals, East Hanover, NJ) for 15 minutes. All allophycocyanin- and phycoerythrin-conjugated antibodies were used according to the supplier’s recommendations (Becton Dickinson Immunocytometry Systems [BDIS], San Jose, CA). Stained cells were washed in 3 mL PBS and fixed in 0.5 mL 2% paraformaldehyde (Sigma) on ice for 15 minutes. Cells were washed in 3 mL PBS and permeabilized in 0.5 mL PBS containing 0.5% Tween-20 (Sigma) and 0.2% bovine serum albumin (BSA; fraction V; Sigma). Samples were mixed gently and placed on ice for 15 minutes. Cells were washed in 3 mL PBS and resuspended in 50 μL TUNEL reaction mix containing 5 U terminal deoxynucleotidyl transferase (TdT; Promega, Madison, WI), 1× TdT reaction buffer (Promega; containing 100 mmol/L cacodylate buffer [pH 6.8], 1 mmol/L cobalt chloride, and 0.1 mmol/L dithiothreitol), and 10 μmol/L biotin-16-dUTP (Boehringer Mannheim Biochemicals, Indianapolis, IN). Cells were incubated in the reaction mix at 37°C for 30 minutes. Cells were washed in 3 mL PBS and resuspended in 100 μL labeling solution containing 4× SSC, 0.1% Triton X-100 (Sigma), 10% nonfat dry milk (Carnation, Glendale, CA), and 0.1% sodium azide (Sigma). Ten microliters of avidin-fluorescein (BDIS) was mixed with the cells. Samples were kept at room temperature for 30 minutes in the dark. Cells were washed in 3 mL PBS containing 0.1% Triton X-100. Cells were resuspended in 0.5 mL PBS containing 5 μg/mL propidium iodide (PI; Sigma) and 0.1% DNase heat-inactivated RNase A (Sigma). Negative control samples were treated in an identical manner except that TdT was omitted from the TUNEL reaction mix. Labeled cells were analyzed promptly by flow cytometry.

Annexin V-labeling was used to confirm the results of TUNEL.40 Briefly, cells were harvested and washed in 3 mL PBS. Cells were stained with immunophenotyping antibodies as described above. Cells were then washed in 3 mL PBS and resuspended at a concentration of 1 × 106 cells/mL in 1 × annexin V binding buffer (all reagents are supplied with the Apoptosis Detection Kit; R&D Systems, Minneapolis, MN). One hundred microliters (105) of cells was transferred to another tube. Ten microliters of fluorescein-conjugated annexin V (10 μg/mL) and 10 μL PI (50 μg/mL) were mixed with the cells. Samples were kept at room temperature for 15 minutes in the dark. Four hundred microliters of 1× binding buffer was used to dilute the labeled cells and analysis was performed by flow cytometry within 1 hour. Controls were prepared according to the recommendations of the supplier.

Determination of relative Bcl-2 expression levels.

The relative level of Bcl-2 expression in PBL was determined by intracellular staining using a hamster antihuman monoclonal antibody, an assay similar to that used by von Freeden-Jeffry et al.41 Briefly, at least 105 cells were harvested and stained with immunophenotyping antibodies, as described above. Cells were washed in 3 mL PBS and fixed in 0.5 mL 2% paraformaldehyde for 15 minutes on ice. Cells were washed in 3 mL PBS and permeabilized in 200 μL PBS containing 0.3% saponin (Sigma) and 0.2% BSA. All subsequent steps were performed in this buffer. Samples were placed on ice for 15 minutes. Two micrograms of anti–Bcl-2 antibody (clone 6C8; Pharmingen, San Diego, CA) was mixed with the cells. The negative control for measuring background fluorescence was prepared in a similar manner, except that 2 μg hamster IgG isotype control polyclonal antibody (Pharmingen) was added to the cells. Samples were placed on ice for 20 minutes. Cells were washed in 3 mL PBS and resuspended in 100 μL of the buffer. One microgram of fluorescein-conjugated mouse antihamster IgG antibody (clone G70-204; Pharmingen) was mixed with the cells. Samples were placed on ice in the dark for 20 minutes. Cells were washed in 3 mL PBS and resuspended in 0.5 mL 2% paraformaldehyde. Samples were stored at 4°C until analysis was performed by flow cytometry.

Flow cytometry and data analysis.

Analysis of TUNEL samples was performed using a FACSVantage flow cytometer (BDIS) equipped with an argon laser tuned to 488 nm and a helium-neon (HeNe) laser tuned to 633 nm. A 610 nm short-pass splitter was used to divert PI fluorescence to FL-3. After electronic compensation, FL-3 fluorescence was measured using both linear and logarithmic amplification; FL-1 (fluorescein), FL-2 (phycoerythrin), and FL-4 (allophycocyanin) fluorescences were measured using logarithmic amplification. The flow rate was not permitted to exceed 200 events per second. The data from at least 5 × 104events were collected for analysis of freshly isolated cells. For all other experiments, the data from at least 104 events were collected. The data were analyzed using CELLQuest software (BDIS). For analysis of TUNEL data, fragmented cells and debris were electronically excluded. Another region was set to electronically eliminate multiplet events from the analysis. A negative control sample (no TdT added) was used to establish TUNEL-negative and -positive regions, such that at least 99% of the events were in the lower TUNEL-negative region. Background events were then subtracted from the TUNEL-positive events.

All other flow cytometry was performed using a FACSCalibur instrument (BDIS) fitted with both argon and HeNe lasers. The data from at least 104 events were collected. Analysis of fluorescein-annexin V-labeled cells was performed according to the recommendations provided with the Apoptosis Detection Kit (R&D Systems). For analysis of annexin V binding data, fragmented cells and debris were electronically excluded. For analysis of Bcl-2 expression, a region was set to include the enriched lymphocyte population, excluding all other cells and debris. The same region was used for analysis of both normal and WAS lymphocytes so that differences in cell size and granularity, and possible differences in mitochondrial copy number on a per cell basis, were not reflected in the values reported. Background fluorescence was measured using a negative isotype control. The mean fluorescence intensity (MFI) reported for a given sample is the difference between the sample value and the background value.

Statistical analysis of results.

Number Crunching Statistical Systems software (Dr Gerry L. Hintze, Kayszille, UT) was used for the statistical analysis of data. Where applicable, an unbalanced repeated measures analysis of variance (ANOVA) was used to determine the significance of differences between normal and WAS sample values. This method eliminates bias possibly associated with analysis of unbalanced repeated measures. The TUNEL data were also analyzed as a set of randomly chosen duplicate repeated measures to test the validity of the first method of analysis. This analysis showed that the first method was indeed valid, confirming the statistical significance of the data. A probability level (P) of ≤.05 is termed significant. An analysis of correlation between Bcl-2 expression and level of apoptosis was performed using logarithmic transformation. A correlation coefficient (r2) was determined using the nonparametric Spearman method. Increases in the frequency of apoptosis over time were determined by linear regression.

WAS PBL undergo spontaneous apoptosis in vitro at a greater rate than normal PBL.

The levels of spontaneous apoptosis in unstimulated, resting PBL were determined in this study. To compare the frequency of apoptosis in WAS PBL and normal PBL, we determined the fraction of cells undergoing apoptosis by 2 quantitative methods. TUNEL39 and annexin V-labeling40 measure oligonucleosomal DNA degradation and externalization of phosphatidylserine, respectively, 2 hallmarks of apoptosis.42,43 Flow cytometry was used to quantify marked cells undergoing apoptosis immediately after PBL isolation or after a period of in vitro incubation. Importantly, PBL were not treated with cytokines or any other mitogenic or antigenic stimuli and neither was an extrinsic stimulus used to induce apoptosis.

Representative TUNEL data shown in Fig 1show that a larger fraction of cells isolated from a WAS patient were undergoing spontaneous apoptosis compared with cells from a normal control at the time of analysis. Another prominent characteristic of apoptotic cells detectable by flow cytometry is cytoplasmic shrinkage, resulting in decreased forward light scatter and increased side light scatter, caused by increases in cellular density during volume reduction.44 A direct comparison of the light scattering properties of normal PBL with those isolated from a WAS patient showed that a relatively larger subpopulation of WAS PBL had apoptotic characteristics (Fig 1A). The results of an analysis of freshly isolated PBL performed immediately after isolation are shown in Fig 1C. The mean levels of apoptotic DNA fragmentation in freshly isolated PBL from the group of 5 WAS patients and the group of 8 normal individuals (5 adults and 3 children) differed significantly (P = .0123). WAS PBL underwent spontaneous apoptosis at a 10-fold greater frequency immediately after isolation. Considerable variation in the levels of spontaneous apoptosis in freshly isolated PBL from different WAS patients was evident, possibly reflecting genotypic differences. However, differences between the siblings are also noted and suggest that polygenic factors may determine the susceptibility to apoptosis in WAS PBL. Therefore, these data suggest that WAS PBL, as compared with normal PBL, may have a lower threshold of susceptibility to apoptogenic signals.

Fig. 1.

Analysis of apoptotic lymphocytes isolated from WAS patients. The levels of apoptosis were measured using TUNEL immediately after isolation or after incubation in vitro. PBL were not stimulated. (A) WAS lymphocytes had increased side scatter and decreased forward scatter, characteristics of apoptosis, after 4 days of incubation in vitro relative to normal lymphocytes. (B) Representative data acquired after TUNEL indicate that a higher fraction of WAS lymphocytes were undergoing apoptosis relative to normal, healthy donor lymphocytes after 4 days of incubation in vitro. The fraction of TUNEL-positive cells is indicated in the upper right corner. (C) WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Levels of apoptosis were measured immediately after isolation of PBL from WAS patients or from normal individuals using the TUNEL assay. N, the mean value for a group of 8 different normal controls (3 children and 5 adults). W, the mean value for a group of 5 different WAS patients. 1 through 5, the mean values for each individual WAS patient. Patient no. 5 was analyzed twice in 2 independent experiments (with 6 and 3 replicates); all other patients were analyzed once in single experiments (with 6 replicates). An unbalanced repeated measures analysis of variance (ANOVA) showed that the values for patients no. 2, 3, 4, and 5 differed significantly from the mean normal value (P ≤ .0001). The mean value for the group of 5 WAS patients (W) also differed significantly from the mean normal (P = .0123). n, the number of different samples; *, statistical significance. (D) WAS lymphocytes were more susceptible to apoptosis than normal lymphocytes after in vitro incubations of 24, 48, and 96 hours. Considerable variability in the apoptotic susceptibility of PBL from different WAS patients was apparent. Mean levels of apoptosis are reported in Table 1. Patient no. 1 was analyzed once in a single experiment, patient no. 2 was analyzed 3 times in 3 independent experiments, and patients no. 3, 4, and 5 were analyzed twice in 2 independent experiments. Results from 8 different normal controls (3 children and 5 adults) are shown. The number of repeated measures made in each experiment was varied, ranging from 2 to 6 (generally 4), depending on the number of PBL isolated from the blood samples. An unbalanced repeated measures ANOVA showed that differences between the groups, WAS and normal, are significant at 24, 48, and 96 hours (P = .000181, .00283, and .000190, respectively). *Statistical significance.

Fig. 1.

Analysis of apoptotic lymphocytes isolated from WAS patients. The levels of apoptosis were measured using TUNEL immediately after isolation or after incubation in vitro. PBL were not stimulated. (A) WAS lymphocytes had increased side scatter and decreased forward scatter, characteristics of apoptosis, after 4 days of incubation in vitro relative to normal lymphocytes. (B) Representative data acquired after TUNEL indicate that a higher fraction of WAS lymphocytes were undergoing apoptosis relative to normal, healthy donor lymphocytes after 4 days of incubation in vitro. The fraction of TUNEL-positive cells is indicated in the upper right corner. (C) WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Levels of apoptosis were measured immediately after isolation of PBL from WAS patients or from normal individuals using the TUNEL assay. N, the mean value for a group of 8 different normal controls (3 children and 5 adults). W, the mean value for a group of 5 different WAS patients. 1 through 5, the mean values for each individual WAS patient. Patient no. 5 was analyzed twice in 2 independent experiments (with 6 and 3 replicates); all other patients were analyzed once in single experiments (with 6 replicates). An unbalanced repeated measures analysis of variance (ANOVA) showed that the values for patients no. 2, 3, 4, and 5 differed significantly from the mean normal value (P ≤ .0001). The mean value for the group of 5 WAS patients (W) also differed significantly from the mean normal (P = .0123). n, the number of different samples; *, statistical significance. (D) WAS lymphocytes were more susceptible to apoptosis than normal lymphocytes after in vitro incubations of 24, 48, and 96 hours. Considerable variability in the apoptotic susceptibility of PBL from different WAS patients was apparent. Mean levels of apoptosis are reported in Table 1. Patient no. 1 was analyzed once in a single experiment, patient no. 2 was analyzed 3 times in 3 independent experiments, and patients no. 3, 4, and 5 were analyzed twice in 2 independent experiments. Results from 8 different normal controls (3 children and 5 adults) are shown. The number of repeated measures made in each experiment was varied, ranging from 2 to 6 (generally 4), depending on the number of PBL isolated from the blood samples. An unbalanced repeated measures ANOVA showed that differences between the groups, WAS and normal, are significant at 24, 48, and 96 hours (P = .000181, .00283, and .000190, respectively). *Statistical significance.

Close modal

We examined the effects of maintaining isolated PBL in vitro for varying periods in medium containing 10% human A+ serum, which minimizes lymphocyte activation. The levels of spontaneous apoptosis in WAS PBL (5 patients) and normal PBL (5 adults and 3 children) differed significantly after incubations of 24, 48, and 96 hours (Fig 1D). We observed a steady increase in the fraction of WAS PBL undergoing spontaneous apoptosis over time. In contrast, the frequency of spontaneous apoptosis in normal PBL leveled off after 48 hours in vitro. Over the entire duration of the incubation, the difference between the increases in WAS and normal PBL tendency to undergo apoptosis was statistically significant (P < .0001). We observed that most of the cells undergoing apoptosis were resting in either G0 or G1 of the cell cycle (data not shown).

Annexin V-labeling confirmed and extended the results obtained using TUNEL to mark cells undergoing spontaneous apoptosis. Apoptotic cells are distinguished by flow cytometry from live and dead cells by fluorescein-conjugated annexin V binding and exclusion of the vital dye, PI, respectively (Fig 2A). As shown in Fig 2B, over the entire duration of incubation, WAS PBL from the 4 patients (no. 1 through 4) analyzed underwent spontaneous apoptosis at a significantly greater frequency than normal PBL (P = .025).

Fig. 2.

Annexin V staining confirmed that WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Representative data indicate that a higher percentage of WAS lymphocytes had externalized phosphatidylserine and bound annexin V compared with normal lymphocytes. Furthermore, a higher percentage of lymphocytes from WAS patients were inviable, no longer excluding PI, relative to lymphocytes from normal individuals. Apoptotic cells are represented by events in the lower right-hand quadrant, with viable and dead cells depicted by events in the lower left-hand and upper right-hand quadrants, respectively. The fraction of cells undergoing apoptosis and the fraction of dead cells are indicated at the right of each plot. (B) Both methods of analysis, annexin V staining and TUNEL, indicated that WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Samples from 4 different WAS patients (no. 1 through 4) and 6 different normal controls were analyzed. Statistical analysis by an unbalanced repeated measures ANOVA showed that the differences between WAS and normal levels of apoptosis as measured using annexin V were significant after in vitro incubations of 48 and 96 hours, as indicated by the asterisks (P = .0013 and .0181, respectively). TUNEL data (shown in Fig 1) is duplicated for easy comparison.

Fig. 2.

Annexin V staining confirmed that WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Representative data indicate that a higher percentage of WAS lymphocytes had externalized phosphatidylserine and bound annexin V compared with normal lymphocytes. Furthermore, a higher percentage of lymphocytes from WAS patients were inviable, no longer excluding PI, relative to lymphocytes from normal individuals. Apoptotic cells are represented by events in the lower right-hand quadrant, with viable and dead cells depicted by events in the lower left-hand and upper right-hand quadrants, respectively. The fraction of cells undergoing apoptosis and the fraction of dead cells are indicated at the right of each plot. (B) Both methods of analysis, annexin V staining and TUNEL, indicated that WAS lymphocytes underwent apoptosis at a greater frequency than normal lymphocytes. Samples from 4 different WAS patients (no. 1 through 4) and 6 different normal controls were analyzed. Statistical analysis by an unbalanced repeated measures ANOVA showed that the differences between WAS and normal levels of apoptosis as measured using annexin V were significant after in vitro incubations of 48 and 96 hours, as indicated by the asterisks (P = .0013 and .0181, respectively). TUNEL data (shown in Fig 1) is duplicated for easy comparison.

Close modal
Both WAS B and T lymphocytes are more susceptible to apoptosis than normal B and T lymphocytes.

WAS patients have defects in both humoral and cell-mediated immunity; thus, we sought to determine if both T and B lymphocytes have increased susceptibility to spontaneous apoptosis. We measured the frequencies of spontaneous apoptosis in resting T and B lymphocytes isolated from WAS patients (no. 1 through 4) and normal controls after incubation in vitro. Figure 3A shows that unstimulated WAS CD3+ T lymphocytes undergo spontaneous apoptosis at a greater rate than do unstimulated normal CD3+ T lymphocytes. In vitro incubation augmented the level of spontaneous apoptosis in unstimulated T lymphocytes. Over time, the frequency of spontaneous apoptosis in WAS T lymphocytes increased more rapidly than occurred normally (P = .024). Unstimulated WAS CD19+ B lymphocytes also underwent spontaneous apoptosis at an increasing frequency during in vitro incubation (Fig 3B). The mean level of spontaneous apoptosis in WAS B lymphocytes was higher than in normal B lymphocytes at each time point. This trend was consistent, although statistical significance could not be demonstrated due to the presence of very low numbers of circulating B lymphocytes in our WAS patients. In vitro incubation also augmented spontaneous apoptosis in unstimulated WAS B lymphocytes by 2-fold over normal.

Fig. 3.

Both T and B lymphocytes from WAS patients are more susceptible to apoptosis than T and B lymphocytes from normal individuals. Analysis of apoptosis was performed using TUNEL after staining with immunophenotyping cell surface markers. Four different WAS patients (no. 1 through 4) and 4 different normal controls were analyzed. (A) CD3+ T lymphocytes from WAS patients underwent apoptosis at a greater frequency than did normal lymphocytes. Statistical significance was shown after 96 hours of incubation in vitro (P = .0239) by an unbalanced repeated measures ANOVA. (B) WAS B lymphocytes (CD19+) are more susceptible to apoptosis than were normal B lymphocytes. The mean levels of apoptosis were clearly different, although, because of small cell numbers, the standard errors were too high to show statistical significance. The scales used in (A) and (B) are identical.

Fig. 3.

Both T and B lymphocytes from WAS patients are more susceptible to apoptosis than T and B lymphocytes from normal individuals. Analysis of apoptosis was performed using TUNEL after staining with immunophenotyping cell surface markers. Four different WAS patients (no. 1 through 4) and 4 different normal controls were analyzed. (A) CD3+ T lymphocytes from WAS patients underwent apoptosis at a greater frequency than did normal lymphocytes. Statistical significance was shown after 96 hours of incubation in vitro (P = .0239) by an unbalanced repeated measures ANOVA. (B) WAS B lymphocytes (CD19+) are more susceptible to apoptosis than were normal B lymphocytes. The mean levels of apoptosis were clearly different, although, because of small cell numbers, the standard errors were too high to show statistical significance. The scales used in (A) and (B) are identical.

Close modal
Bcl-2 expression is attenuated in WAS PBL.

Because Bcl-2 family members have been shown to regulate apoptosis in hematopoietic cells, the accelerated spontaneous apoptosis in WAS PBL could result from deregulated expression of regulatory factors, such as Bcl-2,45 that repress apoptosis. We examined the relative levels of Bcl-2 expression in WAS and normal PBL by measuring the MFI of cells stained indirectly with anti–Bcl-2 antibody in situ. Measurements were made either immediately after isolation of PBL (Table 1) or after in vitro incubation (Table 2). WAS PBL from 5 different patients had a significantly reduced level of Bcl-2 compared with normal PBL from 8 different individuals (Fig 4). The relative levels of Bcl-2 expressed in isolated PBL did not change significantly over time during in vitro incubation. Wide ranges in Bcl-2 expression are evident, both in PBL isolated from WAS patients and from normal controls. Analysis showed that PBL from patients no. 2 and 5 had Bcl-2 levels that differed significantly from normal (P = .00477 and .0254, respectively) at the initial time point. There was no significant difference between the levels of Bax46 expression in WAS and normal PBL (data not shown). Therefore, we have demonstrated that Bcl-2, an inhibitor of apoptosis, but not Bax, an activator of apoptosis, is aberrantly expressed in WAS PBL relative to normal PBL.

Table 1.

Comparing the Frequency of Apoptosis and the Level of Bcl-2 Expression in Freshly Isolated WAS Lymphocytes From Different Patients

Mean Apoptotic Fraction of Lymphocytes*Mean Bcl-2 Expression in Lymphocytes
Mean normal 0.00144 ± 0.00120  68.0 ± 19.7  
Patient no. 1 0.00184 ± 0.000372  51.12 ± 3.63  
Patient no. 2 0.0112 ± 0.000844  6.51 ± 1.20  
Patient no. 3 0.00421 ± 0.000886  34.51 ± 2.16  
Patient no. 4 0.0121 ± 0.00247  38.31 ± 0.834  
Patient no. 5 0.0327 ± 0.0107  12.11 ± 2.73 
Mean Apoptotic Fraction of Lymphocytes*Mean Bcl-2 Expression in Lymphocytes
Mean normal 0.00144 ± 0.00120  68.0 ± 19.7  
Patient no. 1 0.00184 ± 0.000372  51.12 ± 3.63  
Patient no. 2 0.0112 ± 0.000844  6.51 ± 1.20  
Patient no. 3 0.00421 ± 0.000886  34.51 ± 2.16  
Patient no. 4 0.0121 ± 0.00247  38.31 ± 0.834  
Patient no. 5 0.0327 ± 0.0107  12.11 ± 2.73 

Mean normal represents the mean value for a group of 8 different normal controls (5 adults and 3 children). Patient no. 1 was analyzed once, patient no. 2 was analyzed 3 times, and all others were analyzed twice in independent experiments with unbalanced numbers of replicates.

*

Mean apoptotic fraction of lymphocytes was measured immediately after isolation using flow cytometry to quantify cells marked using TUNEL.

Mean Bcl-2 expression in lymphocytes was determined by measuring the MFI of cells stained with fluorescein-conjugated secondary antibody directed against bound anti–Bcl-2 monoclonal antibody.

Table 2.

Comparing the Frequency of Apoptosis and the Level of Bcl-2 Expression in WAS and Normal Lymphocyte Populations

Incubation Time After IsolationMean Apoptotic Fraction of Lymphocytes*Mean Bcl-2 Expression in Lymphocytes
WAS Normal WAS Normal
None  0.014 ± 0.013 0.0014 ± 0.0012  25.3 ± 17.9  54.9 ± 28.3 
24 h  0.17 ± 0.078  0.048 ± 0.018 34.5 ± 15.2  59.7 ± 19.8  
48 h  0.27 ± 0.18 0.10 ± 0.035  33.8 ± 10.1  62.0 ± 25.4  
96 h 0.37 ± 0.15  0.12 ± 0.044  ND ND 
Incubation Time After IsolationMean Apoptotic Fraction of Lymphocytes*Mean Bcl-2 Expression in Lymphocytes
WAS Normal WAS Normal
None  0.014 ± 0.013 0.0014 ± 0.0012  25.3 ± 17.9  54.9 ± 28.3 
24 h  0.17 ± 0.078  0.048 ± 0.018 34.5 ± 15.2  59.7 ± 19.8  
48 h  0.27 ± 0.18 0.10 ± 0.035  33.8 ± 10.1  62.0 ± 25.4  
96 h 0.37 ± 0.15  0.12 ± 0.044  ND ND 

WAS indicates the mean value for a group of 5 WAS patients; normal indicates the mean value for a group of 8 normal controls (5 adults and 3 children). Patient no. 1 was analyzed once, patient no. 2 was analyzed 3 times, and all others were analyzed twice in independent experiments with unbalanced numbers of replicates.

Abbreviation: ND, not determined.

*

Mean apoptotic fraction of PBL was measured using flow cytometry to quantify cells marked by TUNEL.

Mean Bcl-2 expression in lymphocytes was determined by measuring the MFI of cells stained with fluorescein-conjugated secondary antibody directed against bound antihuman Bcl-2 monoclonal antibody.

Fig. 4.

Bcl-2 expression in WAS lymphocytes is attenuated as compared with Bcl-2 expression in normal lymphocytes. MFI of the Bcl-2 signal was measured either immediately after isolation or after in vitro incubation for 24 or 48 hours. Samples from 5 different WAS patients and 7 different normal controls were analyzed. An unbalanced repeated measures ANOVA showed that the differences in Bcl-2 expression levels between the 2 groups, normal and WAS, at each time point (0, 24, and 48 hours) are significant (P = .0233, .00985, and .0192, respectively).

Fig. 4.

Bcl-2 expression in WAS lymphocytes is attenuated as compared with Bcl-2 expression in normal lymphocytes. MFI of the Bcl-2 signal was measured either immediately after isolation or after in vitro incubation for 24 or 48 hours. Samples from 5 different WAS patients and 7 different normal controls were analyzed. An unbalanced repeated measures ANOVA showed that the differences in Bcl-2 expression levels between the 2 groups, normal and WAS, at each time point (0, 24, and 48 hours) are significant (P = .0233, .00985, and .0192, respectively).

Close modal
Correlation between the level of Bcl-2 expression and susceptibility to induction of spontaneous apoptosis in PBL.

The relationship between Bcl-2 expression and susceptibility to induction of spontaneous apoptosis was studied in freshly isolated PBL from 5 different WAS patients and from 4 normal, healthy individuals. Relative Bcl-2 expression was measured immediately after isolation without an extended in vitro incubation. At the same time and using the same sample, we measured the level of apoptosis in the PBL population after TUNEL. As illustrated in Fig 5, there was an inverse relationship between Bcl-2 expression and the susceptibility to induction of spontaneous apoptosis (r2 = .744). Unstimulated normal PBL that expressed relatively high levels of Bcl-2 were less susceptible to induction of spontaneous apoptosis; the frequency of apoptosis in normal populations was reduced when compared with the frequency of apoptosis in WAS populations. In contrast, unstimulated WAS PBL that expressed relatively low levels of Bcl-2 tended to be more susceptible to induction of spontaneous apoptosis than were normal PBL.

Fig. 5.

Correlation between the relative level of Bcl-2 expression and the susceptibility to spontaneous apoptosis in PBL isolated from WAS patients and normal controls. The mean value for replicate measures (2 to 3) of the MFI of the Bcl-2 signal was plotted on the y-axis. The mean fraction of PBL undergoing spontaneous apoptosis was determined using TUNEL and was plotted on the x-axis. Both measurements were made immediately after isolation of PBL using the same sample. Samples from 5 different WAS patients were analyzed; patient no. 5 was analyzed twice in 2 independent experiments. The data were fitted logarithmically. There is an inverse correlation between Bcl-2 expression and the level of susceptibility to apoptosis (r2 = .744).

Fig. 5.

Correlation between the relative level of Bcl-2 expression and the susceptibility to spontaneous apoptosis in PBL isolated from WAS patients and normal controls. The mean value for replicate measures (2 to 3) of the MFI of the Bcl-2 signal was plotted on the y-axis. The mean fraction of PBL undergoing spontaneous apoptosis was determined using TUNEL and was plotted on the x-axis. Both measurements were made immediately after isolation of PBL using the same sample. Samples from 5 different WAS patients were analyzed; patient no. 5 was analyzed twice in 2 independent experiments. The data were fitted logarithmically. There is an inverse correlation between Bcl-2 expression and the level of susceptibility to apoptosis (r2 = .744).

Close modal

We have shown that, in the absence of extrinsic apoptotic stimuli, resting WAS PBL undergo spontaneous apoptosis at a significantly greater frequency than resting normal PBL. We also have found that the expression of Bcl-2 is attenuated in WAS PBL. Because Bcl-2 inhibits the onset of apoptosis, insufficient levels of Bcl-2 expression could diminish the survival potential and shorten the life span of lymphocytes. We suggest then that peripheral T and B lymphocytes in WAS patients undergo accelerated spontaneous apoptosis as a consequence of a partial Bcl-2 deficiency. The accelerated destruction of peripheral T lymphocytes by spontaneous apoptosis could account for the progressive deterioration of T-lymphocyte function and lymphopenia in WAS patients. Moreover, a poor humoral response to certain specific antigens by WAS patients may not be strictly a consequence of T-lymphocyte abnormalities, as has been previously suggested.3,8 The accelerated destruction of memory B lymphocytes with diminished survival potential may preclude secondary immune responses.47 Therefore, chronic and recurrent infection in WAS patients may result from a failure to develop immunologic memory caused by accelerated spontaneous apoptosis of peripheral T and B lymphocytes.

Marked by DNA degradation, phosphatidylserine externalization, and cytoplasmic shrinkage, the abnormally high incidence of apoptosis in WAS lymphocytes suggests that inherent abnormalities of the actin cytoskeleton and cell surface glycoproteins3 may engender their self-destruction by programmed cell death (PCD). We found that the frequency of WAS PBL apoptosis was augmented to a greater extent than normal by in vitro incubation. This suggests that PCD was not triggered before isolation or in a manner dependent on the removal of lymphocytes from the periphery, where perhaps an extrinsic factor enabled survival. Moreover, because an external stimulus was not required to induce apoptosis in WAS PBL, we suggest that an internal mechanism induces unstimulated, resting WAS PBL to undergo spontaneous apoptosis at an accelerated frequency relative to normal PBL. The necessary impetus to undergo spontaneous apoptosis might be provided by the inherent abnormalities in WAS cells; these defects may conceivably trigger intrinsic apoptogenic signaling by surveillance proteins that monitor cellular damage.

Previous studies of WAS patients have demonstrated profound lymphopenia in peripheral lymphoid organs, corroborating our results and suggesting that our observations are clinically relevant. For example, the spleen and lymph nodes of WAS patients were largely devoid of T lymphocytes.48,49 Moreover, another case study of a WAS patient reported that the germinal centers and follicles of the lymph node and spleen were poorly developed or absent.49Hypocellularity and abnormal tissue architecture of these secondary lymphoid tissues in WAS patients is consistent with the our observation that WAS lymphocytes undergo accelerated spontaneous apoptosis and support the possible contention that WAS PBL have an inherently diminished potential for survival in vivo.

Studies of other affected blood cell lineages in WAS patients have also provided data that are consistent with our observations. It has been proposed that thrombocytopenia in WAS patients develops as a consequence of the accelerated destruction of platelets.3Studies of WAS heterozygotes have suggested that affected hematopoietic stem cells and their progeny that carry an active mutant X-chromosome are stringently selected against during development,3conceivably due to a loss of a capacity to either proliferate or survive. Affected blood cells in WAS heterozygotes may either fail to renew or to expand in a manner competitive with their normal, unaffected counterparts. Our study provides a unifying hypothesis that may explain the mechanism of the accelerated destruction of platelets and lymphocytes in WAS patients and the strict exclusion of affected blood cells in WAS carriers. We propose that mutations to WASP cause increased spontaneous apoptosis in all affected blood cells in both WAS patients and WAS carriers.

Mutations to WASP may obstruct hematopoiesis, but clearly the development of mature lymphocytes and platelets in WAS patients is not strictly precluded. Instead, it seems that WASP mutations are a detriment to the survival of mature blood cells in the periphery. In this study, we found abnormally low levels of Bcl-2 in resting, unstimulated WAS PBL. Because other studies have demonstrated the importance of Bcl-2 in regulating the survival of mature lymphocytes,50-52 our results suggest that the increased lability of WAS lymphocytes may be due to insufficient Bcl-2 expression. Interestingly, we note that the development and maintenance of lymphocytes in WAS patients is mirrored by lymphopoiesis in Bcl-2–deficient mice. Postnatally, it appears that lymphopoiesis in Bcl-2–deficient mice is normal; however, older mice eventually develop lymphopenia, as the thymus and spleen of these mice undergo massive apoptotic involution.53 Furthermore, whereas Bcl-2–deficient hematopoietic progenitor cells (HPC) in mouse chimeras differentiate into phenotypically mature lymphocytes, their progeny have markedly shortened life spans relative to the progeny of normal HPC.54 It seems, therefore, that murine Bcl-2 is dispensable for lymphocyte maturation but is required for the maintenance of viability afterward. We now describe a similar phenomena that occurs in WAS patients. Consistent with this observation, WASP-deficient mice, despite having apparently normal lymphopoiesis, have markedly decreased numbers of mature lymphocytes in the periphery relative to wild-type mice.55 

Considerable variation in the levels of susceptibility to spontaneous apoptosis was found in PBL from different WAS patients. Despite genotypic identity and presumably similar environments, PBL isolated from the siblings, patients no. 3 and 4, had strikingly different levels of susceptibility to spontaneous apoptosis. PBL from patient no. 4 were about 3 times more sensitive to intrinsic apoptotic stimuli than were PBL from patient no. 3. Although both patients had grade 3 WAS, patient no. 3 is several years younger than patient no. 4. Because immunodeficiency associated with WAS is progressive, it is intriguing to speculate that such phenotypic differences may arise with the increasing age of the patient. Of course, there is the possibility that multiple genetic factors determine the clinical severity of WAS. Certainly, it is not uncommon to find that different members of the same kindred have different manifestations of the disease.18 

We have observed considerable variation in Bcl-2 expression in freshly isolated PBL from different WAS patients (Table 1). The level of spontaneous apoptosis appears to be inversely related to the level of Bcl-2 expression. For example, PBL from WAS patient no. 1, which expressed relatively higher levels of Bcl-2 than PBL from the other patients, were less susceptible to the induction of spontaneous apoptosis. Because reduced Bcl-2 expression in WAS PBL could account for the increased susceptibility to intrinsic apoptotic stimuli, we speculate that WASP may be involved in signaling pathways that regulate Bcl-2 expression. Many mutations entirely nullify WASP expression, causing a clinically severe form of the disease.11,19-21Other mutations cause only trace amounts of WASP to be stably expressed and manifest a milder form of the disease. We suggest that WAS severity could be determined by the variable susceptibility of different patient’s lymphocytes to spontaneous apoptosis, which, in turn, could be determined by variable levels of Bcl-2 expression. If Bcl-2 expression is regulated by WASP-dependent signaling pathways, as we have proposed, then insufficient WASP activity could cause a reduction in the expression of Bcl-2 in WAS lymphocytes and thereby increase their sensitivity to apoptogenic signals. It will be of interest to examine the possible relationship between WASP insufficiency and the expression level of Bcl-2 using a larger number of WAS patients in future studies.

The function of WASP is not currently known; however, it has been suggested that, because WASP lacks catalytic activity, the protein may act as a molecular scaffold28 to coordinate the interactions of other signaling proteins that control the dynamic organization of the actin cytoskeleton. In support of this proposal, WASP interacts with Cdc42 and Rac,27-30 small GTPases of the Rho family that regulate the formation of filopodia and lamellipodia, respectively.26 Emerging now is the concept that, along with the actin cytoskeleton, the Rho family GTPases have the ability to coordinately regulate other cellular activities, such as cell cycle progression, activation of mitogen-activated protein (MAP) kinase signaling cascades, and cell survival.26,56 Several studies have demonstrated that Rho family GTPase activity is essential, not only to actin cytoskeleton dynamics, but also to cell survival.57-60 We suggest then that WASP might function as a scaffold for Cdc42- or Rac-dependent effector complexes that coordinately regulate actin cytoskeleton reorganization and cell survival in lymphocytes. This WASP-dependent complex may facilitate the activation of phosphoinositide 3-kinase (PI 3-kinase) by Cdc42.61 PI 3-kinase has been shown to be involved in both suppression of apoptosis62,63 and actin cytoskeleton dynamics.64,65 Alternatively, Btk, a protein kinase that regulates the expression of Bcl-XL in B lymphocytes,66 might become activated by a Cdc42- or Rac-dependent effector complex in a manner that requires its interaction with WASP.34,67,68 Perhaps Itk, another Tec kinase family member expressed in T lymphocytes and shown to interact with WASP,33 will be found to have a role analogous to Btk.69 

If this is true, then the lack of WASP in WAS blood cells could result in irregular signaling through Cdc42 or Rac. A WASP deficiency may result in the random activation of other discrete pools of Rho family GTPase, possibly by the mechanism discussed by Reif and Cantrell.56 The nonspecific activation of different pools of Cdc42/Rac-effector complexes could result in sustained signaling through alternative pathways, such as the c-Jun N-terminal kinase or stress-activated protein kinase (JNK/SAPK) MAP kinase cascades,70,71 which may lead to the induction of apoptosis.72,73 In support of this idea, overexpression of activated Rho family GTPases markedly increases apoptosis.74-76 Consequently, in addition to the effect of decreased Bcl-2 expression by WAS lymphocytes, erratic signaling through Cdc42 or Rac, in the absence of sufficient WASP activity, could possibly promote the onset of apoptosis.

In conclusion, this study implicates a pathogenic mechanism in WAS that involves the accelerated apoptotic depletion of peripheral lymphocytes and that may thereby cause immunodeficiency in WAS patients. We suggest that the progressive immune dysfunction associated with WAS may develop as the result of an inability of the thymus to generate new T lymphocytes at a rate that is compensatory with their depletion in the periphery. The deregulation of apoptosis has now been associated with the pathogenesis of a variety of diseases.77,78 Moreover, in those diseases associated with increased apoptosis, it is apparent that the deficient expression of antiapoptotic regulatory factors, such as Bcl-2 and Bcl-XL, may be responsible for the decreased survival of affected cells. For example, in patients with X-linked agammaglobulinemia (XLA), in which B-lymphocyte development is arrested by mutations to the tyrosine kinase Btk,79 manifest immunodeficiency might develop, in part, due to the attrition of abnormally fragile pre-B lymphocytes. This was suggested by studies ofxid mice, an animal model for XLA, also carrying mutations in Btk,80 that demonstrated that the poor survival of peripheral B lymphocytes may be due to abnormal regulation of both Bcl-2 and Bcl-XL.50,66 In human immunodeficiency virus infection, CD4+ T lymphocytes undergo accelerated destruction,81-83 possibly due to diminished expression of Bcl-2,84,85 and immunodeficiency may then progress from insufficient compensatory lymphopoiesis.86 Thus, together with the results of this study, it would appear that deregulation of Bcl-2 family members may be a common mechanism involved in the pathogenesis of various immunodeficiencies. An analysis of larger numbers of WAS patients, in an effort to correlate susceptibility to spontaneous apoptosis and disease severity, will be important in understanding the clinical course of WAS. Future studies will also address the possible mechanisms by which WASP-dependent signaling pathways may regulate the survival of peripheral lymphocytes.

The authors express our gratitude to Dr Leo Mascarenhas and Dr John Bastian (Children’s Hospital, San Diego, CA) for their assistance in procuring blood samples from WAS patients, and Dr Hans Ochs for providing the genotypic information. We appreciate the efforts of Earl Leonard, our biostatistician. We thank Dr Donald Durden, Dr Jane Fountain, and Brile Chung for the critical reading of this manuscript. We are grateful to Flavia Thiemann and Matthieu DeClerck for their technical assistance. We gratefully acknowledge the CHLA Research Institute, and the Achievement Rewards for College Scientists Foundation (Los Angeles Chapter) for the support of S.L.R., the recipient of the John H. Richardson & Margaret Kersten Ponty Endowed Fellowship through the Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA.

Supported in part by Grants No. AI40581, HL54729, and HL54850 (Specialized Center of Research in Stem Cell Biology) from the National Institutes of Health. S.L.R. is a fellow of the Achievement Rewards for College Scientists Foundation and Childrens Hospital Los Angeles Research Institute.

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
Wiskott
A
Familiarer, angeborener Morbus Werlhofii?
Monatsschrift fur Kinderheilkunde
6
1937
212
2
Aldrich
RA
Steinberg
AG
Campbell
DC
Pedigree demonstrating a sex-linked recessive condition characterized by draining ears, eczamatoid dermatitis and bloody diarrhea.
Pediatrics
13
1954
133
3
Remold-O’Donnell
E
Rosen
FS
Kenney
DM
Defects in Wiskott-Aldrich syndrome blood cells.
Blood
87
1996
2621
4
Ochs
HD
Slichter
SJ
Harker
LA
von Behrens
WE
Clark
RA
Wedgewood
RJ
The Wiskott-Aldrich syndrome: Studies of lymphocytes, granulocytes, and platelets.
Blood
55
1980
243
5
Altman
LC
Snyderman
R
Blaese
RM
Abnormalities of chemotactic lymphokine synthesis and mononuclear leukocyte chemotaxis in Wiskott-Aldrich syndrome.
J Clin Invest
50
1974
486
6
Zicha
D
Allen
WE
Brickell
PM
Kinnon
C
Dunn
GA
Jones
GE
Thrasher
AJ
Chemotaxis of macrophages is abolished in the Wiskott-Aldrich syndrome.
Br J Haematol
101
1998
659
7
Badolato
R
Sozzani
S
Malcarne
F
Bresciani
S
Fiorini
M
Borsatti
A
Albertini
A
Mantovani
A
Ugazio
AG
Notarangelo
LD
Monocytes from Wiskott-Aldrich patients display reduced chemotaxis and lack of cell polarization in response to monocyte chemoattractant protein-1 and formyl-methionyl-leucyl-phenylalanine.
J Immunol
161
1998
1026
8
Parkman
R
Rappeport
J
Geha
RS
Belli
J
Cassady
R
Levey
R
Nathan
DG
Rosen
FS
Complete correction of the Wiskott-Aldrich syndrome by allogeneic bone-marrow transplantation.
N Engl J Med
298
1978
921
9
Molina
IJ
Sancho
J
Terhorst
C
Rosen
FS
Remold-O’Donnell
E
T cells of patients with the Wiskott-Aldrich syndrome have a restricted defect in proliferative responses.
J Immunol
151
1993
4383
10
Gallego
MD
Santamaria
M
Pena
J
Molina
IJ
Defective actin reorganization and polymerization of Wiskott-Aldrich T cells in response to CD3-mediated stimulation.
Blood
90
1997
3089
11
Facchetti
F
Blanzuoli
L
Vermi
W
Notarangelo
LD
Giliani
S
Fiorini
M
Fasth
A
Stewart
DM
Nelson
DL
Defective actin polymerization in EBV-transformed B-cell lines from patients with the Wiskott-Aldrich syndrome.
J Pathol
185
1998
99
12
Molina
IJ
Kenney
DM
Rosen
FS
Remold-O’Donnell
E
T cell lines characterize events in the pathogenesis of the Wiskott-Aldrich syndrome.
J Exp Med
176
1992
867
13
Kenney
D
Cairns
L
Remold-O’Donnell
E
Peterson
J
Rosen
FS
Parkman
R
Morphological abnormalities in the lymphocytes of patients with the Wiskott-Aldrich syndrome.
Blood
68
1986
1329
14
Kwan
S-P
Hagemann
TL
Blaese
RM
Rosen
FS
A high resolution map of genes, microsatellite markers and new dinucleotide repeats from UBE1 to the GATA locus in the region Xp11.23.
Genomics
29
1995
247
15
Derry
JMJ
Ochs
HD
Francke
U
Isolation of a novel gene mutated in Wiskott-Aldrich syndrome.
Cell
78
1994
635
16
Stewart
DM
Treiber-Held
S
Kurman
CC
Facchetti
F
Notarangelo
LD
Nelson
DL
Studies of the expression of the Wiskott-Aldrich syndrome protein.
J Clin Invest
97
1996
2627
17
Parolini
O
Berardelli
S
Riedl
E
Fernandez
B
Strobl
H
Majdic
O
Knapp
W
Expression of Wiskott-Aldrich syndrome protein (WASP) gene during hematopoietic differentiation.
Blood
90
1997
70
18
Ochs
HD
The Wiskott-Aldrich syndrome.
Semin Hematol
35
1998
332
19
MacCarthy-Morrogh
L
Gaspar
HB
Wang
YC
Katz
R
Thompsom
L
Layton
M
Jones
AM
Kinnon
C
Absence of expression of the Wiskott-Aldrich syndrome protein in peripheral blood cells of Wiskott-Aldrich syndrome patients.
Clin Immunol Immunopathol
88
1998
22
20
Remold-O’Donnell
E
Cooley
J
Shcherbina
A
Hagemann
TL
Kwan
S-P
Kenney
DM
Rosen
FS
Variable expression of WASP in B cell lines of Wiskott-Aldrich syndrome patients.
J Immunol
158
1997
4021
21
Zhu
Q
Watanabe
C
Liu
T
Hollenbaugh
D
Blaese
RM
Kanner
SB
Aruffo
A
Ochs
HD
Wiskott-Aldrich syndrome/X-linked thrombocytopenia: WASP gene mutations, protein expression, and phenotype.
Blood
90
1997
2680
22
Sullivan
KE
Mullen
CA
Blaese
RM
Winkelstein
JA
A multiinstitutional survey of the Wiskott-Aldrich syndrome.
J Pediatr
125
1994
876
23
Miki
H
Sasaki
T
Takai
Y
Takenawa
T
Induction of filopodium formation by a WASP-related actin-depolymerizing protein N-WASP.
Nature
391
1998
93
24
Ramesh
N
Anton
IM
Hartwig
JH
Geha
RS
WIP, a protein associated with Wiskott-Aldrich syndrome protein, induces actin polymerization and redistribution in lymphoid cells.
Proc Natl Acad Sci USA
94
1997
14671
25
Wu
Y
Spenser
SD
Lasky
LA
Tyrosine phosphorylation regulates the SH3-mediated binding of the Wiskott-Aldrich syndrome protein to PSTPIP, a cytoskeletal-associated protein.
J Biol Chem
273
1998
5765
26
Hall
A
Rho GTPases and the actin cytoskeleton.
Science
279
1998
509
27
Aspenstrom
P
Lindberg
U
Hall
A
Two GTPases, Cdc42 and Rac, bind directly to a protein implicated in the immunodeficiency disorder Wiskott-Aldrich syndrome.
Curr Biol
6
1996
70
28
Kolluri
R
Fuchs Tolias
K
Carpenter
CL
Rosen
FS
Kirchhausen
T
Direct interaction of the Wiskott-Aldrich syndrome protein with the GTPase Cdc42.
Proc Natl Acad Sci USA
93
1996
5615
29
Rudolph
MG
Bayer
P
Abo
A
Kuhlmann
J
Vetter
IR
Wittinghofer
A
The Cdc42/Rac interactive binding region motif of the Wiskott-Aldrich syndrome protein (WASP) is necessary by not sufficient for tight binding to Cdc42 and structure formation.
J Biol Chem
273
1998
18067
30
Symons
M
Derry
JMJ
Karlak
B
Jiang
S
Lemanhieu
V
McCormick
F
Francke
U
Abo
A
Wiskott-Aldrich syndrome protein, a novel effector for the GTPase CDC42Hs, is implicated in actin polymerization.
Cell
84
1996
723
31
Rivero-Lezcano
OM
Marcilla
A
Sameshima
JH
Robbins
KC
Wiskott-Aldrich syndrome protein physically associates with Nck through Src homology 3 domains.
Mol Cell Biol
15
1995
5725
32
Banin
S
Truong
O
Katz
DR
Waterfield
MD
Brickell
PM
Gout
I
Wiskott-Aldrich syndrome protein (WASp) is a binding partner for c-Src family protein-tyrosine kinases.
Curr Biol
6
1996
981
33
Bunnell
SC
Henry
PA
Kolluri
R
Kirchhausen
T
Rickles
RJ
Berg
LJ
Identification of Itk/Tsk Src homology 3 domain ligands.
J Biol Chem
271
1996
25646
34
Cory
GOC
MacCarthy-Morrogh
L
Banin
S
Gout
I
Brickell
PM
Levinsky
RJ
Kinnon
C
Lovering
RC
Evidence that the Wiskott-Aldrich syndrome protein may be involved in lymphoid cell signaling pathways.
J Immunol
157
1996
3791
35
Finan
PM
Soames
CJ
Wilson
L
Nelson
DL
Stewart
DM
Truong
O
Hsuan
JJ
Kellie
S
Identification of regions of the Wiskott-Aldrich syndrome protein responsible for association with selected Src homology 3 domains.
J Biol Chem
271
1996
26291
36
She
HY
Rockow
S
Tang
J
Nishimura
R
Skolnik
EY
Chen
M
Margolis
B
Li
W
Wiskott-Aldrich syndrome is associated with the adaptor protein Grb2 and the epidermal growth factor receptor in living cells.
Mol Cell Biol
8
1997
1709
37
Pawson
T
Scott
JD
Signaling through scaffold, anchoring, and adaptor proteins.
Science
278
1997
2075
38
Adams
JM
Cory
S
The Bcl-2 protein family: Arbiters of cell survival.
Science
281
1998
1322
39
Hotz
AH
Gong
J
Traganos
F
Darzynkiewicz
Z
Flow cytometric detection of apoptosis: comparison of the assays of in situ DNA degradation and chromatin changes.
Cytometry
15
1994
237
40
Koopman
G
Reutelingsperger
CPM
Kuijten
GAM
Keehnen
RMJ
Pals
ST
van Oers
MHJ
Annexin V for flow cytometric detection of phosphatidylserin expression on B cells undergoing apoptosis.
Blood
84
1994
1415
41
von Freeden-Jeffry
U
Solvason
N
Howard
M
Murray
R
The earliest T lineage-committed cells depend on IL-7 for Bcl-2 expression and normal cell cycle progression.
Immunity
7
1997
147
42
Wyllie
AH
Morris
RG
Smith
AL
Dunlop
D
Chromatin cleavage in apoptosis: Association with condensed chromatin morphology and dependence on macromolecular synthesis.
J Pathol
142
1984
67
43
Fadok
VA
Voelker
DR
Campbell
PA
Cohen
JJ
Bratton
DL
Henson
PM
Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages.
J Immunol
148
1992
2207
44
Vayssiere
J-L
Petit
PX
Risler
Y
Mignotte
B
Commitment to apoptosis is associated with changes in mitochondrial biogenesis and activity in cell lines conditionally immortalized with simian virus 40.
Proc Natl Acad Sci USA
91
1994
11752
45
Hockenbery
D
Nunez
G
Milliman
C
Schreiber
RD
Korsmeyer
SJ
Bcl-2 is an inner mitochondrial membrane protein the blocks programmed cell death.
Nature
348
1990
334
46
Yin
C
Knudson
CM
Korsmeyer
SJ
Van Dyke
T
Bax suppresses tumoirgenesis and stimulates apoptosis in vivo.
Nature
385
1997
637
47
Nunez
G
Hockenberry
D
McDonnell
TJ
Sorensen
CM
Korsmeyer
SJ
Bcl-2 maintains B cell memory.
Nature
353
1991
71
48
Cooper
MD
Chase
HP
Lowman
JT
Krivit
W
Good
RA
Wiskott-Aldrich syndrome: An immunologic deficiency disease involving the afferent limb of immunity.
Am J Med
44
1968
499
49
Snover
DC
Frizzera
G
Specter
BD
Perry
GS
Kersey
JH
Wiskott-Aldrich sydrome: Histopathologic findings in the lymph nodes and spleens of 15 patients.
Hum Pathol
12
1981
821
50
Woodland
RT
Schmidt
MR
Korsmeyer
SJ
Gravel
KA
Regulation of B cell survival in xid mice by the proto-oncogene bcl-2.
J Immunol
156
1996
2143
51
Merino
R
Ding
L
Veis
DJ
Korsmeyer
SJ
Nunez
G
Developmental regulation of the Bcl-2 protein and susceptibility to cell death in B lymphocytes.
EMBO J
13
1994
683
52
Nunez
G
Merino
R
Grillot
D
Gonzalez-Garcia
M
Bcl-2 and Bcl-x: Regulatory switches for lymphoid death and survival.
Immunol Today
15
1994
582
53
Veis
DJ
Sorenson
CM
Shutter
JR
Korsmeyer
SJ
Bcl-2-deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented hair.
Cell
75
1993
229
54
Nakayama
K-I
Nakayama
K
Negishi
I
Kuida
K
Shinkai
Y
Louie
MC
Fields
LE
Lucas
PJ
Stewart
V
Alt
FW
Loh
DY
Disappearance of the lymphoid system in Bcl-2 homozygous mutant chimeric mice.
Science
261
1993
1584
55
Snapper
SB
Rosen
FS
Mizoguchi
E
Cohen
P
Khan
W
Liu
C-H
Hagermann
TL
Kwan
S-P
Ferrini
R
Davidson
L
Bhan
AK
Alt
FW
Wiskott-Aldrich syndrome protein-deficient mice reveal a role for WASP in T but not B cell activation.
Immunity
9
1998
81
56
Reif
K
Cantrell
DA
Networking Rho family GTPases in lymphocytes.
Immunity
8
1998
395
57
Gomez
J
Martinez-A.
C
Giry
M
Garcia
A
Rebollo
A
Rho prevents apoptosis through Bcl-2 expression: Implications for interleukin-2 receptor signal transduction.
Eur J Immunol
27
1997
2793
58
Galandrini
R
Henning
SW
Cantrell
DA
Different functions of the GTPase Rho in prothymocytes and late pre-T cells.
Immunity
7
1997
163
59
Moorman
JP
Bobak
DA
Hahn
CS
Inactivation of the small GTP binding protein Rho induces multinucleate cell formation and apoptosis in murine T lymphoma EL4.
J Immunol
156
1996
4146
60
Henning
SW
Galandrini
R
Hall
A
Cantrell
DA
The GTPase Rho has a critical regulatory role in thymus development.
EMBO J
16
1997
2397
61
Zheng
Y
Bagrodia
S
Cerione
RA
Activation of phosphoinositide 3-kinase activity by Cdc42Hs binding to p85.
J Biol Chem
269
1994
18727
62
Kennedy
SG
Wagner
AJ
Conzen
SD
Jordan
J
Bellacosa
A
Tsichlis
PN
Hay
N
The PI 3-kinase/AKT signaling pathway delivers an anti-apoptotic signal.
Genes Dev
11
1997
701
63
Franke
TF
Kaplan
DR
Cantley
LC
PI3K: Downstream AKTion blocks apoptosis.
Cell
88
1997
435
64
Rodriguez-Viciana
P
Warne
PH
Khwaja
A
Marte
BM
Pappin
D
Das
P
Waterfield
MD
Ridley
A
Downward
J
Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras.
Cell
89
1997
457
65
Gomez
J
Garcia
A
R.-Borlado
L
Bonay
P
Martinez-A.
C
Silva
A
Fresno
M
Carrera
AC
Eicher-Streiber
C
Rebollo
A
IL-2 signaling controls actin organization through Rho-like protein family, phosphatidylinositol 3-kinase, and protein kinase C-ζ.
J Immunol
158
1997
1516
66
Anderson
JS
Teutsch
M
Dong
Z
Wortis
HH
An essential role for tyrosine kinase in the regulation of Bruton’s B-cell apoptosis.
Proc Natl Acad Sci USA
93
1996
10966
67
Kinnon
C
Cory
GO
MacCarthy-Morrogh
L
Banin
S
Gout
I
Lovering
RC
Brickell
PM
The identification of Bruton’s tyrosine kinase and Wiskott-Aldrich syndrome protein associated and signaling pathways.
Biochem Soc Trans
25
1997
648
68
Guinamard
R
Aspenstrom
P
Fougereau
M
Chavrier
P
Guillemot
JC
Tyrosine phosphorylation of the Wiskott-Aldrich syndrome protein by Lyn and Btk is regulated by Cdc42.
FEBS Lett
434
1998
431
69
August
A
Sadra
A
Dupont
B
Hanafusa
H
Src-induced activation of inducible T cell kinase (ITK) requires phosphatidylinositol 3-kinase activity and the Pleckstrin homology domain of inducible T cell kinase.
Proc Natl Acad Sci USA
94
1997
11227
70
Coso
OA
Chiariello
M
Yu
JC
Teramoto
H
Crespo
P
Xu
N
Miki
T
Gutkind
JS
The small GTP-binding proteins Rac1 and Cdc42 regulate the activity of the JNK/SAPK signaling pathway.
Cell
81
1995
1137
71
Lamarche
N
Tapon
N
Stowers
L
Burbelo
PD
Aspenstrom
P
Bridges
T
Chant
J
Hall
A
Rac and Cdc42 induce actin polymerization and G1 cell cycle progression independently of p65PAK and the JNK/SAPK MAP kinase cascade.
Cell
87
1996
519
72
Faris
M
Kokot
N
Latinis
K
Kasibhatla
S
Green
DR
Koretzky
GA
Nel
A
The c-Jun N-terminal kinase cascade plays a role in stress-induced apoptosis in Jurkat cells by up-regulating Fas ligand expression.
J Immunol
160
1998
134
73
Kasibhatla
S
Brunner
T
Genestier
L
Echeverri
F
Mahboubi
A
Green
DR
DNA damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-κB and AP-1.
Mol Cell
1
1998
543
74
Maundrell
K
Antonsson
B
Magnenat
E
Camps
M
Muda
M
Chabert
C
Gillieron
C
Boschert
U
Vial-Knecht
E
Martinou
J-C
Arkinstall
S
Bcl-2 undergoes phosphorylation by c-Jun N-terminal kinase/stress-activated protein kinases in the presence of the constitutively active GTP-binding protein Rac1.
J Biol Chem
272
1997
25238
75
Bazenet
CE
Mota
MA
Rubin
LL
The small GTP-binding protein Cdc42 is required for nerve growth factor withdrawal-induced neuronal death.
Proc Natl Acad Sci USA
95
1998
3984
76
Lores
P
Morin
L
Luna
R
Gacon
G
Enhanced apoptosis in the thymus of transgenic mice expressing constitutively activated forms of human Rac2GTPase.
Oncogene
15
1997
601
77
Hetts
SW
To die or not to die: An overview of apoptosis and its role in disease.
JAMA
279
1998
300
78
Thompson
CB
Apoptosis in the pathogenesis and treatment of disease.
Science
267
1995
1456
79
Conley
ME
Parolini
O
Rohrer
J
Campana
D
X-linked agammaglobulinemia: New approaches to old questions based on the identification of the defective gene.
Immunol Rev
138
1994
5
80
Sher
I
The CBA/n mouse strain; an experimental model illustrating the influence of the X-chromosome on immunity.
Adv Immunol
33
1982
1
81
Meyaard
L
Otto
SA
Jonker
RR
Mijnster
MJ
Keet
RPM
Miedema
F
Programmed death of T cells in HIV-1 infection.
Science
257
1992
217
82
Oyaizu
N
McCloskey
TW
Coronesi
M
Chirmule
N
Kalyanaraman
VS
Pahwa
S
Accelerated apoptosis in peripheral blood mononuclear cells (PBMCs) from human immunodeficiency virus type-1 infected patients and in CD4 cross-linked PBMCs from normal individuals.
Blood
82
1993
3392
83
Gandhi
RT
Chen
BK
Straus
SE
Dale
JK
Lenardo
MJ
Baltimore
D
HIV-1 kills CD4+ T cells by a Fas-independent mechanism.
J Exp Med
187
1998
1113
84
Hashimoto
F
Oyaizu
N
Kalyanaraman
VS
Pahwa
S
Modulation of Bcl-2 protein by CD4 cross-linking: A possible mechanism for lymphocyte apoptosis in human immunodeficiency virus infection and for rescue of apoptosis by interleukin-2.
Blood
90
1997
745
85
Muralidhar
G
Koch
S
Broome
HE
Swain
SL
TCR triggering of anergic CD4 T cells in murine AIDS induces apoptosis rather than cytokine synthesis and proliferation.
J Immunol
157
1996
625
86
Jenkins
M
Hanley
MB
Moreno
MB
Wieder
E
McCune
JM
Human immunodeficiency virus-1 infection interrupts thymopoiesis and multilineage hematopoiesis in vivo.
Blood
91
1998
2672

Author notes

Address reprint requests to Kenneth I. Weinberg, MD, Division of Research Immunology and Bone Marrow Transplantation, Mail Stop #62, Childrens Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027; e-mail: kweinberg@chla.usc.edu.

Sign in via your Institution