Patients with the immunodeficiency disorder Wiskott-Aldrich syndrome (WAS) have lymphocytes with aberrant microvilli, and their T cells, macrophages, and dendritic cells are impaired in cytoskeletal-dependent processes. WAS is caused by a defective or a missing WAS protein (WASP). Signal mediators interleukin-4 (IL-4) and CD40 are important for actin-dependent morphology changes in B cells. A possible function of WASP and its interacting partners, Cdc42 and Rac1, was investigated for these changes. It was found that active Cdc42 and Rac1 induced filopodia and lamellipodia, respectively, in activated B cells. Evidence is given that IL-4 has a specific role in the regulated cycling of Cdc42 because IL-4 partially and transiently depleted active Cdc42 from detergent extract of activated B cells. WASP-deficient B lymphocytes were impaired in IL-4– and CD40-dependent induction of polarized and spread cells. Microvilli were expressed on WASP-deficient B cells, but they appeared shorter and less dense in cell contacts than in wild-type cells. In conclusion, evidence is provided for the involvement of Cdc42, Rac1, and WASP in the cytoskeletal regulation of B lymphocytes. Aberrations in WASP-deficient B lymphocytes, described here, provide further evidence that WAS is a cytoskeletal disease of hematopoietic cells.

Activation of lymphocytes is often accompanied by changes in cell morphology. Transendothelial migration requires microvilli-dependent adhesion and spreading of lymphocytes.1 The fine regulation of immune activation in lymph nodes and spleen is dependent on migratory and adhesive capacities of B and T cells.2,3 For migration and eventual contact with other immune cells to occur, adhesion receptors are essential and appear at the trailing uropod of a migrating cell. In contrast, chemoattractant receptors are expressed at the leading edge.4 Reorganization of microtubuli and dynamics of the actomyosin filaments are crucial events during migration and adhesion.5 

T-cell–derived signals through interleukin-4 (IL-4) receptors or CD40 on B cells induce cytoskeletal rearrangements, altering B-cell morphology. Thymus-independent stimulus lipopolysaccharide (LPS) from gram-negative bacteria, and even more pronounced IL-4 and agonistic anti-CD40 antibodies (anti-CD40 monoclonal antibody [mAb]), mimicking the CD40 ligand, stimulate B lymphocytes to acquire a motile shape.6-9 It has been shown that anti-CD40 or LPS plus IL-4, but not LPS alone, induces the formation of dendritic protrusions when B cells are cultured on immobilized antibodies directed to B-cell surface determinants.10,11 LPS plus IL-412 or anti-CD4013 induces homotypic B-cell aggregates that are tight and spherical, whereas LPS-induced aggregates are loose and irregularly shaped.14 The induction of spread cells and tight aggregates are correlated with an increase in number and length of villous structures on the B-cell surface.9 

Wiskott-Aldrich syndrome (WAS) is a severe X-linked immunodeficiency disorder caused by mutations in the gene encoding the WAS protein (WASP). Lymphocytes from patients with WAS have aberrant formation of microvilli.15-17 In addition, T cells16,17and B cells18 have been reported to have abnormal cytoarchitecture. WASP-deficient dendritic cells19 and macrophages20,21 have aberrant polarization and are impaired in stimulated migration. So far, WASP expression has only been detected in cells of the hematopoietic lineage,22 whereas its homologue N-WASP is more ubiquitously expressed.23 WASP24 and N-WASP25interact directly with the Arp2/3 complex, which is important for polymerization from barbed ends and branching of actin filaments.26 Similarly, WASP27,28 and N-WASP23 colocalize with polymerized actin. Recently, mice carrying a WASP null allele were described,29,30 and it was found that T-cell function was impaired, whereas mutant B cells proliferated normally to B-cell stimuli.

WASP was identified as a binding partner for the small GTPase Cdc42.27,31 Cdc42 and its relative Rac1 belong to the family of Rho GTPases, known to operate as molecular switches, cycling between an active guanosine triphosphate (GTP)-bound and an inactive guanosine diphosphate (GDP)-bound state. This cycling is tightly regulated by guanosine nucleotide exchange factors (GEFs), which stimulate the exchange of GDP to GTP, and GTPase-activating proteins (GAPs), which increase the intrinsic rate of GTP hydrolysis.32 Rho GTPases are involved in many cellular processes in which the cell cytoskeleton is the final target, such as migration,33,34 cytokinesis,35 and phagocytosis.36,37 Effector proteins of Cdc42 and Rac1 are numerous, and many of them, including WASP and N-WASP, have Cdc42/Rac-interactive binding (CRIB) motifs that specifically recognize the GTP-bound form.32 

In this paper, we have investigated the intracellular pathways induced by LPS, IL-4, and CD40 activation in B lymphocytes. We show that active Cdc42 and Rac1 induced morphology changes in activated B cells. IL-4 partially and transiently depleted active Cdc42 from detergent extract of B cells. Furthermore, we found that WASP-deficient B cells were impaired in many morphologic processes. Interestingly, N-WASP was clearly expressed in WASP-deficient B cells. The data suggest that Cdc42, Rac1, and WASP are important for the induction of morphologic changes leading to B-cell migration and adhesion.

Reagents and antibodies

Recombinant murine IL-4 was either derived from supernatants of the plasmacytoma X63 Ag8-653 transfected with IL-4 cDNA38or purchased from Peprotech EC (London, United Kingdom). Endotoxin levels of the purified IL-4 were less than 0.1 ng/μg (1 EU/μg). Alone, neither X63 IL-4 nor purified IL-4 induced B-cell proliferation, as measured by 3H-thymidine incorporation, excluding an effect of contaminating endotoxins because LPS alone induced high proliferation. The amount of IL-4 inducing a half-maximal DNA synthesis response in concanavalin A–activated T cells was defined as 1 U. Unless otherwise indicated, 2 U/mL IL-4 (2% X63 supernatant or 2 ng/mL purified IL-4) was used. LPS from Escherichia coli O55:B5 was either prepared at the Department of Microbiology and Tumour Biology (Karolinska Institutet) or purchased from Sigma Aldrich (Stockholm, Sweden). Batches of fetal calf serum (FCS; Labora, Stockholm, Sweden) were selected for low endotoxin levels (less than 0.05 EU/mL). These FCS batches, when applied alone, did not induce B-cell proliferation, as measured by 3H-thymidine incorporation, or polarization (Figure 3). Rat anti–mouse CD40 (1C10)39 and CD44 (RK3G9)40 mAbs were prepared as previously described.11 Rabbit antibodies to the GTPase-binding domain (CRIB, amino acids 201-321) of human WASP were prepared as previously described.41 Rabbit anti–N-WASP antibodies were raised against a peptide (amino acids 173-189) of human N-WASP. Rabbit anti-myc and anti-Cdc42 antibodies were purchased from Santa Cruz Biotechnology (Scandinavian Diagnostic Service, Falkenberg, Sweden). Goat anti–rat immunoglobulin G (IgG; H&L) antibodies and biotinylated-F(ab′)2 donkey anti–rabbit IgG (H&L) were from Jackson ImmunoResearch Laboratories (West Grove, PA). Horseradish peroxidase–conjugated swine anti–rabbit IgG antibodies and fluorescein isothiocyanate (FITC)–coupled streptavidin were from Dako A/S (Glostrup, Denmark).

Animals

A pair of WASP-deficient animals, kindly provided by Dr Fredrick Alt (Boston, MA),29 were bred and maintained in the animal facility of the Department of Cell and Molecular Biology (Karolinska Institutet, Stockholm). Because WAS is an X-linked trait, males are hemizygous for the WASP allele (denoted as WASPthroughout the paper). WASP−/− females × WASP+ CBA male (purchased from Charles River, Uppsala, Sweden) breeding was established, followed by WASP+/−× WASP (F1) breeding to provide litters with WASP mutants and WASP+ sibling controls (F2). The genotype of the mice was controlled by polymerase chain reaction on tail-prepared DNA. CBA/J × C57BL/6 (F1) mice (referred to as CB throughout the paper) were purchased from Charles River. Males were used at 4 to 9 weeks.

Cell culture

Small resting B cells were prepared from splenic cell suspensions after the removal of T cells and Percoll gradient centrifugation as previously described.14,42 Cells were cultured at 0.5 × 106 cells/mL in RPMI 1640 supplemented with 2 mM L-glutamine, 1 mM sodium pyruvate, 50 U/mL penicillin, 50 μg/mL streptomycin, 5 μM 2-mercaptoethanol (all from Gibco BRL, Life Technology, Paisley, United Kingdom) and 5% to 10% of batch-selected FCS at 37°C in a humidified atmosphere containing 5% CO2. Cell lines secreting monoclonal antibodies were cultured in the same way but with 1% to 5% batch-selected FCS.

Migration assay

To exclude contaminants of chemoattractants, both the X63-derived IL-4 and the purified IL-4 (Peprotech) were tested in a migration assay using a 2-chamber system (polycarbonate filter plates, 5-μm pore size; Transwell, Costar, New York, NY). B cells (100 000) were loaded to the upper wells, and no stimulus (5% FCS in RPMI), X63 IL-4, purified IL-4, or SDF-1 (50 ng/mL; R&D Systems, Oxon, United Kingdom) was added onto the lower wells. B cells were allowed to migrate for 2 hours, and then cells in the lower wells were collected and enumerated in triplicate. FCS induced low migration (2400 ± 670 cells, background). X63 IL-4 and purified IL-4 induced a slightly higher number of migrating cells (3300 ± 370 and 3600 ± 350 cells, respectively). Compared to background levels, these changes were nonsignificant (P > .05) using a t test. SDF-1, a potent B-cell chemoattractant, induced the migration of 8300 ± 800 cells (P < .001 compared to background). Figures indicate the mean of triplicate results ± 1 SD. Thus, the X63 supernatant and purified IL-4 seem to lack classical chemokines inducing migration with fast kinetics.

Transfections and stainings

cDNAs of myc-tagged Cdc42L61, Cdc42N17, wild-type Cdc42, RacL61, and RacN17 in the pRK5 vector (Pharmingen, Stockholm, Sweden) was used. CB B lymphocytes were stimulated with LPS for 46 to 50 hours. Fourteen micrograms DNA was electroporated (960 μF, 350 V, and 200 Ω) into activated B cells (10 × 106), then diluted in medium containing LPS or LPS plus IL-4. Cells were transferred to anti–CD44-coated coverslip and cultured for 3 to 4 hours. They were thereafter fixed in either acetone–methanol (1:1) for 20 minutes at −20°C or 4% phosphate-buffered paraformaldehyde for 10 minutes at room temperature, followed by 0.1% Triton X-100 for 1 minute. Cells were first blocked in 10% mouse serum in phosphate-buffered saline (PBS), stained with rabbit anti-myc antibodies, biotinylated antirabbit antibodies, and streptavidin-FITC. All solutions contained 10% mouse serum in PBS.

Antibody coating of plastic culture vessels or coverslips was performed as follows: goat anti–rat IgG was added and incubated for 1 hour at 37°C, followed by addition of rat anti–mouse CD44 (RK3G9) antibodies, incubated overnight at room temperature. Coverslips were washed in balanced salt solution before cells were added.

Glutathione-S-transferase pull-down assay

Residues 201-321 (CRIB) of human WASP were cloned into the pGEX-2T (Pharmacia Biotech, Uppsala, Sweden). Production and purification of recombinant GST-CRIB was described elsewhere.43 Precipitation of Cdc42-GTP was performed as previously described.43 Briefly, CB B lymphocytes (25 or 50 × 106 cells per sample) were stimulated with LPS for 48 hours, and IL-4 or FCS-containing RPMI was added in the last half hour of incubation. Cells were centrifuged at 4°C and lysed for 10 minutes on ice in RIPA buffer (100 μL/106 cells) containing 2 μg/mL aprotinin and leupeptin, 1 mM phenylmethylsulfonyl fluoride, and 5 mM MgCl2. Cdc42-GTP was precipitated from lysates with glutathione–Sepharose beads (Pharmacia Biotech), preloaded with GST-CRIB, for 30 minutes at 4°C. The beads were washed, resuspended in reduced sample buffer (50 mM Tris, 50 mM dithiothreitol, 0.1% bromphenol blue, 10% glycerol, and 5% sodium dodecyl sulfate [SDS]), boiled, and loaded on a 15% polyacrylamide gel. Separated proteins were transferred to a nitrocellulose membrane by Western blotting.

Cell polarization assays

B lymphocytes, isolated from spleens of WASP or WASP+ mice, were cultured in 24-well tissue culture plates in the presence of LPS, IL-4 (derived from the X63 plasmacytoma or purified from Peprotech), anti-CD40 mAb, or no stimulus for 20 to 24 hours. Cells were fixed by adding an equal volume of 2.5% phosphate-buffered glutaraldehyde and were washed twice in PBS before they were counted using an inverted light microscope. The percentage of polarized cells—cells that were not spherical but were tapered and had uropods—was determined.

Cell spreading assay

Cell spreading assays were performed in antibody-coated 96-well plates as previously described.11 Briefly, cells were stimulated with LPS, LPS plus IL-4 (derived from the X63 plasmacytoma or purified from Peprotech), or anti-CD40 mAb and were transferred to anti–CD44-coated plates for 17 to 19 additional hours of culture. Cells were thereafter fixed in 2.5% phosphate-buffered glutaraldehyde, and the percentage of spread cells—defined as having at least one dendritic process of at least one cell diameter—was determined using an inverted light microscope.

Aggregation assays

B lymphocytes, isolated from spleens of WASP or WASP+ mice, were cultured for 46 to 50 hours. The morphology of B-cell aggregates, from cultures stimulated with LPS, LPS plus IL-4 (derived from the X63 plasmacytoma or purified from Peprotech), anti-CD40 mAb or anti-CD40 plus IL-4, was photographed directly using a camera connected to an inverted microscope. Percentage cell aggregation was measured by adding 100 μL trypan blue to 200 μL/well cultures. Cells were then resuspended by gentle pipetting up and down 6 times in a blunted disposable pipette and were transferred to a hemocytometer. Aggregated cells (2 or more cells in association) and single cells were enumerated under an inverted light microscope. The percentage of aggregated cells was then calculated.

Electron microscopy

B lymphocytes, isolated from WASP and WASP+ mice, were stimulated with LPS or anti-CD40 mAb plus IL-4 for 48 hours. Aggregates were gently pelleted to preserve cell–cell adhesion and microvilli and then were fixed in glutaraldehyde. Specimen preparation was described previously.11 For examination, a Jeol 100CX electron microscope (Tokyo, Japan) was used at 80 kV.

Western blot analysis

B lymphocytes, isolated from WASP and WASP+ mice, were stimulated with LPS or LPS plus IL-4 for 46 to 50 hours. Cells were harvested, lysed in buffer (50 mM Tris, 150 mM NaCl, 1% Triton X-100, 1 mM phenylmethylsulfonyl fluoride, and 2 μg/mL aprotinin and leupeptin) at 4°C for 15 minutes. Equal protein concentrations of lysates were boiled in reduced sample buffer (described above) containing 2% SDS and were thereafter loaded on a 10% polyacrylamide gel. Separated proteins were transferred to a polyvinylidene difluoride membrane by Western blotting. Membranes were blocked in Tris-buffered saline, 0.05% Tween 20, and 5% fat-free dry milk (Blotto A) for 1 hour at 4°C and then incubated with primary antibodies in Blotto A overnight at 4°C. Horseradish peroxidase–conjugated secondary antibody in Blotto A was added and incubated for 1.5 hours at 4°C. Between steps, membranes were extensively washed in Tris-buffered saline with 0.05% Tween 20. Blots were developed with enhanced chemiluminescence (Pierce, Rockford, IL). Membranes were stained with Ponceau Red to confirm equal loading.

Cdc42 and Rac1 induce filopodia and lamellipodia in activated B lymphocytes

We wanted to investigate a potential role of Cdc42 and Rac1 in the cytoskeletal rearrangements of B lymphocytes. These GTPases induced morphologic changes in fibroblasts after microinjection.44,45 Notably, active Cdc42 induced filopodia, a structure that seems similar to the protrusions found on spread B cells.10,11 We therefore considered Cdc42 and Rac1 as having a potential role in IL-4–induced spreading. To elucidate the role of Cdc42 and Rac1 in B lymphocytes, we used the fact that the Rho GTPases can be converted to constitutively active (CA) and dominant negative (DN) forms using single amino acid substitutions. CA mutants are locked in the GTP-bound form, thereby preventing intrinsic and GAP-induced GTP hydrolysis.46,47 DN mutants are locked in the GDP-bound form, thus forming a nonfunctional complex with cellular GEFs and competing with endogenous proteins for binding to GEFs.48 CA, DN, or wild-type (WT) myc-tagged constructs of Cdc42 or Rac1 were transiently electroporated into LPS-activated B lymphocytes. Cells were then cultured in the presence of LPS or LPS plus IL-4 on anti-CD44 antibody layers (B cells, in contrast to fibroblasts, can only spread in the presence of antibody layers). Cells were fixed, permeabilized, and stained with anti-myc antibodies. CA Cdc42 induced long filopodia in cells stimulated with LPS or LPS plus IL-4, whereas DN Cdc42 failed to induce these structures (Figure 1A). WT Cdc42 induced short filopodia structures or lamellipodia (Figure 1A). CA Rac1 mainly induced lamellipodia in cells stimulated with LPS or LPS plus IL-4 and, rarely, short filopodia (Figure 1B). DN Rac1 did not induce these membrane structures (Figure 1B). Morphologic changes induced by CA and WT Cdc42 or Rac1 in LPS-stimulated cells were indistinguishable from those in LPS plus IL-4 stimulated cells. Thus, in this system, the LPS stimulus alone is sufficient to induce membrane alterations. Induction of morphologic changes was dependent on coating with anti-CD44 antibodies because, in the absence of antibody layers, no morphologic changes were detected and cells were spherical (not shown).

Fig. 1.

Cdc42 and Rac1 induce filopodia and lamellipodia in activated B lymphocytes.

Constitutively active (CA), dominant negative (DN), or wild-type (WT)myc-tagged constructs of Cdc42 (A) or Rac1 (B) were transiently electroporated into B lymphocytes, preactivated with LPS for 46 to 50 hours. Cells were diluted in medium containing LPS or LPS plus IL-4 and cultured on anti-CD44–coated glass coverslips for an additional 3 to 4 hours, fixed, permeabilized, and visualized with anti-myc antibodies. Cells were photographed using a camera connected to a fluorescence microscope with a × 100 objective. (A) CA Cdc42 induced filopodia, long microspikes of polymerized actin, whereas DN Cdc42 failed to induce this structure and transfected cells were spherical. WT Cdc42 induced either short filopodialike structures or lamellipodia (not shown). (B) CA Rac1 induced lamellipodia, flattened sheets of polymerized actin, whereas DN Rac1-expressing cells were spherical. Similar observations were made using LPS or LPS plus IL-4–activated B cells. These cells are representative of the morphologic phenotypes detected in more than 5 experiments, including screening of at least 300 cells in each experiment.

Fig. 1.

Cdc42 and Rac1 induce filopodia and lamellipodia in activated B lymphocytes.

Constitutively active (CA), dominant negative (DN), or wild-type (WT)myc-tagged constructs of Cdc42 (A) or Rac1 (B) were transiently electroporated into B lymphocytes, preactivated with LPS for 46 to 50 hours. Cells were diluted in medium containing LPS or LPS plus IL-4 and cultured on anti-CD44–coated glass coverslips for an additional 3 to 4 hours, fixed, permeabilized, and visualized with anti-myc antibodies. Cells were photographed using a camera connected to a fluorescence microscope with a × 100 objective. (A) CA Cdc42 induced filopodia, long microspikes of polymerized actin, whereas DN Cdc42 failed to induce this structure and transfected cells were spherical. WT Cdc42 induced either short filopodialike structures or lamellipodia (not shown). (B) CA Rac1 induced lamellipodia, flattened sheets of polymerized actin, whereas DN Rac1-expressing cells were spherical. Similar observations were made using LPS or LPS plus IL-4–activated B cells. These cells are representative of the morphologic phenotypes detected in more than 5 experiments, including screening of at least 300 cells in each experiment.

Close modal

Short pulses of IL-4 partially and transiently deplete active Cdc42 from the B-cell cytosol

Many proteins, including specific GAPs and GEFs, regulate the cycling of Cdc42. IL-4 may have a potential role in this process, thereby inducing cytoskeletal changes in activated B cells. We have used a GST-CRIB (from WASP) fusion protein to precipitate the cytosolic fraction of active Cdc42 from detergent lysates of LPS-stimulated B lymphocytes given short pulses of IL-4. In each experiment, extract from cells stimulated with LPS alone was used as a standard for the detected level of Cdc42. A clear decrease in the level of cytosolic Cdc42-GTP was detected after IL-4 stimulation for 5 minutes (Figure2, lane 4) compared to the LPS standard (lane 1). No reduction was detected after IL-4 stimulation for 30 minutes, 10 minutes (lanes 2 and 3, respectively), or 1 minute (not shown). By adding FCS-containing medium alone, we controlled for the reduction being caused by IL-4 stimulation (not shown). Similar results were obtained in 4 experiments, suggesting that IL-4 stimulation of B cells leads to a transient depletion of active Cdc42 from the cytosol. In some other experiments, however, no difference between the groups was obtained. It is unlikely that these data were obtained by chance or the amount of active Cdc42 in the different experimental groups would have been totally variable, which was not the case. Instead, we believe this reflects some of the difficulties when working with rapidly cycling proteins.49 

Fig. 2.

Short pulses of IL-4 partially and transiently deplete active Cdc42 from the B-cell cytosol.

Detergent extracts of LPS-activated B cells were stimulated with IL-4 for 5 to 30 minutes. Cdc42-GTP was removed from extracts using GST-CRIB (from WASP) coupled to glutathione–Sepharose beads. Bound proteins were identified with Western blotting using anti-Cdc42 antibodies. Experimental groups were as follows: no addition of IL-4 (lane 1), addition of IL-4 for 30 (lane 2), 10 (lane 3), or 5 (lane 4) minutes. LPS extracts preloaded with GTPγS are shown in lane 5. All lanes were on the same membrane, but the exposure time was shorter for lane 5 because the signal was strong. Data are representative of 4 experiments.

Fig. 2.

Short pulses of IL-4 partially and transiently deplete active Cdc42 from the B-cell cytosol.

Detergent extracts of LPS-activated B cells were stimulated with IL-4 for 5 to 30 minutes. Cdc42-GTP was removed from extracts using GST-CRIB (from WASP) coupled to glutathione–Sepharose beads. Bound proteins were identified with Western blotting using anti-Cdc42 antibodies. Experimental groups were as follows: no addition of IL-4 (lane 1), addition of IL-4 for 30 (lane 2), 10 (lane 3), or 5 (lane 4) minutes. LPS extracts preloaded with GTPγS are shown in lane 5. All lanes were on the same membrane, but the exposure time was shorter for lane 5 because the signal was strong. Data are representative of 4 experiments.

Close modal

GTPγS, a nonhydrolyzable GTP analogue, arrests Cdc42 in its active state. B-cell lysates from LPS-stimulated cells, preloaded with GTPγS, showed a strong band of Cdc42-GTP after precipitation (Figure2, lane 5). This indicates that Cdc42-GDP is the predominant form in the cytoplasm under the experimental conditions used.

Induction of B-cell polarization is impaired in the absence of WASP

We have investigated whether the defects in polarization and migration of WAS dendritic cells and macrophages also apply to B cells from WASP-deficient mice. Lymphocytes undergoing locomotion characteristically have a polarized or prolonged cell body. Thus, assessment of cells with a motile cell shape can be used to determine locomotion capacity of lymphocytes.6,7,50 In WASP+ B cells (Figure 3, open bars), IL-4, anti-CD40 mAb, and LPS all induced a higher frequency of motile-shaped cells than noninduced cells, the 2 former stimuli giving higher responses than the latter. In WASP B cells (Figure3, black bars), LPS stimuli induced a frequency of motile-shaped cells similar to that in WASP+ cells. However, IL-4 and anti-CD40 induction of motile-shaped WASP B cells was impaired in comparison to WASP+ cells. This suggests that WASP has an important role for IL-4– and CD40-induced motility but that LPS-induced signaling can bypass the requirement for WASP or use completely different pathways.

Fig. 3.

Induction of B-cell polarization is impaired in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured with indicated stimuli for 24 hours, after which they were fixed. Polarized cells were defined as having a tapered cell body and uropods, whereas nonpolarized cells were spherical. Polarized cell frequency was determined. Results are presented as mean values of triplicate experiments, error bars represent 1 SD, and brackets indicate P < .005 using a 2-tailed t test. At least 300 cells were counted in each group, and the data are representative of at least 4 experiments.

Fig. 3.

Induction of B-cell polarization is impaired in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured with indicated stimuli for 24 hours, after which they were fixed. Polarized cells were defined as having a tapered cell body and uropods, whereas nonpolarized cells were spherical. Polarized cell frequency was determined. Results are presented as mean values of triplicate experiments, error bars represent 1 SD, and brackets indicate P < .005 using a 2-tailed t test. At least 300 cells were counted in each group, and the data are representative of at least 4 experiments.

Close modal

Impaired spreading in the absence of WASP

We also investigated the capacity of B-cell spreading in the absence of WASP. LPS plus IL-4 or anti-CD40 mAb induced a high number of spread WASP+ B cells (Figure4, open bars). However, WASP B cells were impaired in spreading in response to the corresponding stimuli (Figure 4, black bars). Only occasional spread cells were seen in LPS cultures. Still, in the absence of WASP, spreading was not completely abolished because the percentage of WASP spread cells was higher in the presence of suboptimal concentrations of IL-4 (Figure 4) or anti-CD40 (not shown) than with LPS alone. Spreading did not occur in the absence of immobilized antibodies or on surfaces coated with irrelevant antibodies (not shown).

Fig. 4.

Impaired spreading in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured with indicated stimuli for 46 to 50 hours and were thereafter transferred to anti-CD44–coated plates. Cells were incubated for 17 to 19 more hours and fixed. The frequency of spread cells, having at least one protrusion longer than one cell diameter, was determined. Mean values of 5 samples (LPS and LPS plus IL-4) or triplicate experiments (anti-CD40 mAb, 30 μg/mL) in each group are presented. Error bars represent 1 SD, and at least 300 cells were counted in each group. Brackets indicate P < .001 using a 2-tailed t test. This observation is representative of 3 similar experiments.

Fig. 4.

Impaired spreading in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured with indicated stimuli for 46 to 50 hours and were thereafter transferred to anti-CD44–coated plates. Cells were incubated for 17 to 19 more hours and fixed. The frequency of spread cells, having at least one protrusion longer than one cell diameter, was determined. Mean values of 5 samples (LPS and LPS plus IL-4) or triplicate experiments (anti-CD40 mAb, 30 μg/mL) in each group are presented. Error bars represent 1 SD, and at least 300 cells were counted in each group. Brackets indicate P < .001 using a 2-tailed t test. This observation is representative of 3 similar experiments.

Close modal

Aggregation is mildly impaired in the absence of WASP

IL-4 and CD40 are known to induce B-cell homotypic adhesion, resulting in the formation of tight, spherical aggregates.12-14 We investigated whether WASP was important for this process. LPS-stimulated WASP+ B cells formed loose, irregularly shaped aggregates, and these were indistinguishable from those of WASP B cells (Figure5A). LPS plus IL-4 induced tight, spherical aggregates to a similar extent in WASP+ and WASP B cells. However, anti-CD40 mAb- or anti-CD40 plus IL-4–induced WASP aggregates were smaller, though still spherical, and less dense than WASP+ aggregates. Another method to estimate cellular aggregation is to gently resuspend aggregate cultures and then enumerate single and aggregated cells. Again, using this method, no differences in percentage of aggregated cells were detected in LPS or LPS plus IL-4 cultures (Figure 5B). In anti-CD40 or anti-CD40 plus IL-4 cultures, a mild defect was detected in WASP B cells (black bars) compared to WASP+ B cells (open bars). Enumeration and morphologic characterization of aggregates suggest that WASP can be a component in the induction of homotypic B-cell adhesion, resulting in the formation of tight aggregates. A mild defect in aggregation can be a secondary cause of impaired polarization of WASP B cells because motility most likely is required for the formation of big aggregates.

Fig. 5.

Aggregation is mildly impaired in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured for 46 to 50 hours with indicated stimuli. (A) Cells were photographed using a camera connected to an inverted microscope with a × 20 objective. Similar results were obtained in at least 5 experiments. (B) Aggregate cultures were gently resuspended, and single versus aggregated cells were enumerated. Mean values of triplicate experiments are shown. Error bars represent 1 SD, and at least 500 cells were counted in each group. Brackets indicateP < .07 using a 2-tailed t test. This observation is representative of 3 similar experiments.

Fig. 5.

Aggregation is mildly impaired in the absence of WASP.

B cells from WASP (▪) and WASP+ (■) mice were cultured for 46 to 50 hours with indicated stimuli. (A) Cells were photographed using a camera connected to an inverted microscope with a × 20 objective. Similar results were obtained in at least 5 experiments. (B) Aggregate cultures were gently resuspended, and single versus aggregated cells were enumerated. Mean values of triplicate experiments are shown. Error bars represent 1 SD, and at least 500 cells were counted in each group. Brackets indicateP < .07 using a 2-tailed t test. This observation is representative of 3 similar experiments.

Close modal

Activated WASP B lymphocytes express short microvilli on their surfaces

As observed in scanning electron microscopy, lymphocytes from patients with WAS have distorted microvilli on their surfaces.15-17 We used transmission electron microscopy to investigate the expression of microvilli in murine WASP B lymphocytes. LPS-stimulated WASP or WASP+ B cells expressed few and short microvillilike structures (Figure6). WASP B cells stimulated with anti-CD40 mAb and IL-4 expressed more and longer microvilli on their surfaces, but they were shorter than on similarly activated WASP+ B cells. Higher magnification of cell contacts is shown in Figure 7. WASP+ B cells had extensive connections through microvilli in cell contacts (Figure 7A, white arrows). The microvilli connections in cell contacts of WASP B cells was less dense or absent, leading to more frequent close contact through flat surfaces (Figure 7B, white arrows). In addition, WASP B cells had smooth surfaces, whereas WASP+ B cells were more irregularly shaped. In summary, microvilli were expressed on activated WASP B cells, but they appeared shorter and less dense in cell contacts than in wild-type cells.

Fig. 6.

Activated WASP B lymphocytes express short microvilli.

B cells from WASP and WASP+ mice were cultured for 48 hours with indicated stimuli, gently resuspended, then fixed and prepared for transmission electron microscopy. Note the extensive network of microvilli in cell contacts of anti-CD40 plus IL-4–stimulated cells, especially pronounced in WASP+cells. These observations are representative of 2 similar experiments. Size bar represents 1 μm. Original magnification 3800 × (LPS) and 3000 × (anti-CD40 plus IL-4).

Fig. 6.

Activated WASP B lymphocytes express short microvilli.

B cells from WASP and WASP+ mice were cultured for 48 hours with indicated stimuli, gently resuspended, then fixed and prepared for transmission electron microscopy. Note the extensive network of microvilli in cell contacts of anti-CD40 plus IL-4–stimulated cells, especially pronounced in WASP+cells. These observations are representative of 2 similar experiments. Size bar represents 1 μm. Original magnification 3800 × (LPS) and 3000 × (anti-CD40 plus IL-4).

Close modal
Fig. 7.

Microvilli on WASP B lymphocytes are shorter and less pronounced in cell contacts.

Higher magnification of cell contacts from the electron microscopy studies in Figure 6. IL-4– and anti-CD40–stimulated WASP and WASP+ B cells are shown. (A) Note extensive microvilli contacts (white arrows) between WASP+B cells and the irregular cell shapes. (B) Microvilli network between WASP B cells was less extensive or absent, and cells had smooth surfaces (white arrows). Size bar represents 0.4 μm. Original magnification 8000 ×.

Fig. 7.

Microvilli on WASP B lymphocytes are shorter and less pronounced in cell contacts.

Higher magnification of cell contacts from the electron microscopy studies in Figure 6. IL-4– and anti-CD40–stimulated WASP and WASP+ B cells are shown. (A) Note extensive microvilli contacts (white arrows) between WASP+B cells and the irregular cell shapes. (B) Microvilli network between WASP B cells was less extensive or absent, and cells had smooth surfaces (white arrows). Size bar represents 0.4 μm. Original magnification 8000 ×.

Close modal

N-WASP is expressed in activated murine B lymphocytes

We have described partial morphologic defects in B cells lacking expression of WASP. The WASP-deficient mice used here, like some patients with WAS, seem to have a less severe phenotype.18,51 A likely possibility is that another protein can substitute for some of the function of WASP. We investigated the protein levels of N-WASP, a likely substitute candidate, in activated murine B cells. For this purpose, we used specific antibodies directed to either the CRIB domain of WASP or to a peptide of N-WASP. The anti-WASP antibody detected WASP in detergent lysates of WASP+ B cells, whereas WASP was completely absent in WASP cells (Figure8A). On the other hand, N-WASP was clearly detected in both WASP+ and WASP B cells (Figure 8B), in similar concentrations. Thus, N-WASP was expressed in activated B lymphocytes, but it does not appear that in the absence of WASP, the level of N-WASP was increased.

Fig. 8.

N-WASP is expressed in activated murine B lymphocytes.

Detergent lysates of B cells, stimulated with LPS or LPS plus IL-4 for 46 to 50 hours, were separated using SDS-PAGE, blotted, and exposed to antibodies against WASP or N-WASP. (A) Anti-WASP antibodies recognized WASP in WASP+ B cells and in an unknown, higher molecular species (asterisk) in both WASP+ and WASP B cells. (B) Anti–N-WASP antibody recognized a band consisting of 2, perhaps 3, species that most likely represent different phosphorylation states of N-WASP. These data are representative of 3 similar experiments.

Fig. 8.

N-WASP is expressed in activated murine B lymphocytes.

Detergent lysates of B cells, stimulated with LPS or LPS plus IL-4 for 46 to 50 hours, were separated using SDS-PAGE, blotted, and exposed to antibodies against WASP or N-WASP. (A) Anti-WASP antibodies recognized WASP in WASP+ B cells and in an unknown, higher molecular species (asterisk) in both WASP+ and WASP B cells. (B) Anti–N-WASP antibody recognized a band consisting of 2, perhaps 3, species that most likely represent different phosphorylation states of N-WASP. These data are representative of 3 similar experiments.

Close modal

Cdc42, Rac1, and WASP are all implicated as important for the reorganization of the actin cytoskeleton. WASP and N-WASP regulate actin polymerization through direct interaction with the Arp2/3 complex. We investigated morphologic changes in B lymphocytes induced by IL-4 or CD40 activation and found that Cdc42 and Rac1, when overexpressed in activated B cells, induced filopodia and lamellipodia, respectively. WASP-deficient B cells were impaired in polarization, spreading, homotypic aggregation, and microvilli formation in response to IL-4– and CD40-dependent stimuli. Partial morphologic deficiencies can be explained by functional redundancy of a WASP homologous protein. In agreement with this, we found that the WASP homologue, N-WASP, was expressed in B lymphocytes at a level similar to those in WASP knockout and wild-type cells.

In the early 1990s, Hall and coworkers44,45 performed crucial experiments to explain the function of the Rho GTPases. When active Cdc42 and Rac1 were micro-injected into the Swiss 3T3 fibroblast cell line, specific membrane structures, filopodia, and lamellipodia, respectively, were induced. When we transiently expressed these proteins in activated B cells, we could induce similar phenotypes, indicating that Cdc42 and Rac1 also induce these specific structures in B cells. This activation was dependent on the immobilization of B cells through antibodies to cell membrane molecules. It was clear from our results that LPS alone was sufficient to induce these events, because LPS- and LPS plus IL-4–stimulated cells were indistinguishable in both the morphologic phenotypes and the kinetics of induction (not shown). These results were surprising given that LPS failed to induce spreading in nontransfected B lymphocytes. One reason for this difference may be that the induction of filopodia and lamellipodia is a process separate from the induction of spreading. Another possibility is that these processes may, in part, contain common intracellular pathways but that spreading requires additional factors. However, yet another important difference is that in the transfection experiments, active Cdc42 or Rac1 were in much higher concentrations than their endogenous levels. This may influence downstream effectors and perhaps bypass the tight regulation of signaling by the GTPases. Recently, WASP was shown to have an auto-inhibitory contact between the GTPase binding domain (CRIB) and the C-terminal part, suggesting that WASP activation is also tightly regulated.52 The same mechanism most likely occurs in N-WASP.25 High levels of active Cdc42 or Rac1 can probably shift the GTPase-WASP equilibrium to the favor of GTPase binding to WASP, keeping the latter in an open, active state. Therefore, in this “simplified system,” LPS and LPS plus IL-4 can induce morphologic changes. Interestingly, when active constructs were expressed in WASP B cells, the same phenotypes were induced as in WASP+ B cells (not shown). This suggests that Cdc42 and Rac1 can bypass the requirement for WASP, either through action through WASP-independent pathways or through functional redundancy perhaps by N-WASP. In a model of neutrophil activation, Cdc42 and Rac1 could bypass the requirement for WASP and instead activate actin nucleation through N-WASP.53 

Our results show that IL-4 induces a transient decrease in the level of active cytosolic Cdc42. This observation can be explained by an altered endogenous localization of Cdc42. In the cytoplasm, RhoGDI (guanosine nucleotide dissociation inhibitor) seems to have a major role in sequestering the GDP-bound GTPases, thereby inhibiting the spontaneous exchange of GDP to GTP.32,54 By way of a posttranslationally added geranylgeranyl lipid modification, GTP-bound proteins are able to translocate to membranes.55 For example, phorbol myristate acetate (PMA)-triggered activation of NADPH in human neutrophils correlates with the translocation of Rac2-GTP to the plasma membrane.56 In this system, the translocation is rapid, occurring after a 5-minute stimulation. In a cell-free system, using isolated neutrophil membranes and purified proteins, Rac2 and Cdc42 were rapidly translocated to membranes after GTPγS activation.57 It is tempting to speculate that IL-4 may have a similar role for Cdc42 translocation in B cells. Recent experiments in neutrophils indicate that PMA or the chemoattractant fMet-Leu-Phe (fMLP) induces an increase in the level of active cytosolic Cdc42 and Rac2.58 In a keratinocytic cell line, IL-4 alters the level of active cytosolic Cdc42.59 One explanation for the discrepancy between different results may be found in the experimental procedures. Conditions for cell lysis are important for the solubility of membrane-bound proteins that take part in multimolecular complexes. Together, results of all the experiments mentioned indicate that distinct stimuli transiently and rapidly influence the cytosolic levels of active GTPases.

We also investigated a function for WASP in IL-4– and CD40-induced slow processes, such as the induction of motile and spread cells. WASP B cells were impaired in IL-4– and anti-CD40-induced cell polarization and spreading. These observations suggest that WASP B lymphocytes had intrinsic defects in processes that require a dynamic actin cytoskeleton, such as motility. In contrast, LPS-induced motility can bypass the requirement for WASP or use completely different pathways. Together with defects in stimulated migration and polarization of WAS dendritic cells19 and macrophages,20,21 evidence is accumulating for WASP as important for cell trafficking.60Slow morphologic changes in response to IL-4 and CD40 most likely require gene transcription. IL-4–induced morphology is at least partly mediated by the signal transducer and activator of transcription Stat-6 protein because Stat-6−/− B cells are deficient in the induction of motile and spread cells and also in microvilli formation.9 We are investigating a possible link between Stat-6–activated genes and WASP-dependent signaling pathways.

As judged by scanning electron microscopy, nonactivated human blood lymphocytes have fine villous projections. WAS blood lymphocytes have fewer microvilli, and those expressed are usually abnormally shaped in that they are short and blunted.15-17 In our experiments using transmission electron microscopy, microvilli were clearly induced on anti-CD40 mAb plus IL-4–activated murine WASP B cells. However, the microvilli were shorter than those of activated wild-type B cells. We have previously observed that cell contacts often take place through microvilli and that tight aggregation correlates well with the expression of many long microvilli9 (data not shown). It is interesting that the activated WASP B cells more often contacted each other by way of flat surfaces rather than the wild-type cells. Although a comparison is difficult, it is possible that activated murine WASP B cells have more microvilli than do human lymphocytes from patients with WAS. However, some conditions differ between the present study and published studies. For example, in published studies of human cells, total lymphocytes or T cells were isolated from peripheral blood, whereas we investigated splenic B cells. In addition, in published studies human cells were nonactivated or activated with T-cell stimuli, whereas we studied activated B cells.

In some patients with WAS of less severe phenotype, truncated WASP transcripts and proteins have been detected.18,51 We could confirm that shorter WASP transcripts were completely absent in B cells from WASP-deficient mice (data not shown).29 Thus, partial morphologic deficiencies in murine WASP B cells cannot be explained by truncated WASP. However, we identified similar levels of N-WASP in WASP+ and WASP B cells. Because WASP and N-WASP share 50% homology at the protein level,23 it is tempting to speculate that N-WASP can partially substitute for WASP. Perhaps this is also true for patients with WAS who exhibit a less severe disease phenotype.

Increased surface area through filopodia, lamellipodia, and microvilli is a prerequisite for many cells that make extensive cell-to-cell or cell-to-matrix contacts. For example, in the immunologic synapse,5 a T cell forms filopodia and lamellipodia and clusters membrane molecules on these to sense and contact an antigen-presenting cell. During an immune response, B and T cells encounter the antigen in the spleen and lymph nodes. There, the collaboration between migratory and adhesive capacities is instrumental for B-cell activation to take place.2,3 We suggest that T-cell–derived signals by IL-4 and CD40 can induce morphologic alterations necessary for B-cell migration and interaction with T cells, follicular dendritic cells, and other B cells in B-cell foci and germinal centers (GCs). The importance of CD40–CD40 ligand interactions in GCs was revealed in CD40-deficient mice, which exhibit impaired GC formation.61 Earlier reports, in which electron microscopy was used to examine antigen-specific interaction between B and T cells, revealed extensive membrane contacts between cells.62 Later on, polarized expression of IL-4 in contact areas was shown to enhance conjugate formation.63,64Impaired migration and contact between immune cells from patients with WAS, because of reduced surface plasticity, can most likely explain some of the severity of the disease. In addition, the antigen presentation capacity of WAS B cells may be impaired because antigen presentation on B cells seems to be dependent on CD40–CD40 ligand interaction, whereas other cells can present antigen in a CD40-independent fashion.65 We are investigating how LPS, IL-4, and anti-CD40 activation influences the antigen-presenting capacity of B cells. It will be interesting to include WASP-deficient B cells and to examine whether WASP-dependent morphology changes are important for antigen presentation by B cells.

We have described impairment in the cytoskeletal regulation of WASP-deficient B cells. This is, to our knowledge, the first time a defect in murine B-cell function is shown. Facchetti et al18 showed that Epstein-Barr virus–transformed B-cell lines from patients with WAS are impaired in F-actin regulation. Together, these reports suggest that impaired B-lymphocyte responses may contribute to the immunodeficiency of WAS.

We thank Dr Johan Thyberg for excellent guidance with the electron microscopy and Karin Blomgren for assistance in specimen preparation. We thank Dr Fredrick Alt for providing the WASP knockout mice. We also thank Dr Edward J Davey for stimulating and helpful discussions.

Supported by grants from the Swedish Natural Science Research Council, the Swedish Cancer Foundation, the Network for Inflammation Research funded by the Swedish Foundation for Strategic Research, the Fifth Framework Programme of the European Commission, the Åke Wiberg Foundation, and the Karolinska Institutet.

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

1
Springer
TA
Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm.
Cell.
76
1994
301
314
2
Han
S
Zheng
B
Takahashi
Y
Kelsoe
G
Distinctive characteristics of germinal center B cells.
Semin Immunol.
9
1997
255
260
3
Tarlinton
D
Germinal centers: getting there is half the fun.
Curr Biol.
8
1998
R753
R756
4
Sánchez–Madrid
F
del Pozo
MA
Leukocyte polarization in cell migration and immune interactions.
EMBO J.
18
1999
501
511
5
Dustin
ML
Cooper
JA
The immunological synapse and the actin cytoskeleton: molecular hardware for T cell signaling.
Nat Immunol.
1
2000
23
29
6
Wilkinson
PC
Islam
LN
Recombinant IL-4 and IFN-γ activate locomotor capacity in human B lymphocytes.
Immunology.
67
1989
237
243
7
Clinchy
B
Elenström
C
Severinson
E
Möller
G
T and B cell collaboration: induction of motility in small, resting B cells by interleukin 4.
Eur J Immunol.
21
1991
1445
1451
8
Komai-Koma
M
Liew
FY
Wilkinson
PC
Interactions between IL-4, anti-CD40, and anti-immunoglobulin as activators of locomotion of human B cells.
J Immunol.
155
1995
1110
1116
9
Davey
EJ
Greicius
G
Thyberg
J
Severinson
E
STAT6 is required for the regulation of IL-4– induced cytoskeletal events in B cells.
Int Immunol.
12
2000
995
1003
10
Santos-Argumedo
L
Kincade
PW
Partida-Sánchez
S
Parkhouse
RME
CD44-stimulated dendrite formation (‘spreading’) in activated B cells.
Immunology.
90
1997
147
153
11
Davey
EJ
Thyberg
J
Conrad
DH
Severinson
E
Regulation of cell morphology in B lymphocytes by IL-4: evidence for induced cytoskeletal changes.
J Immunol.
160
1998
5366
5373
12
Elenström
C
Severinson
E
Interleukin 4 induces cellular adhesion among B lymphocytes.
Growth Factors.
2
1989
73
82
13
Björck
P
Paulie
S
Inhibition of LFA-1–dependent human B-cell aggregation induced by CD40 antibodies and interleukin-4 leads to decreased IgE synthesis.
Immunology.
78
1993
218
225
14
Greicius
G
Tamosiunas
V
Severinson
E
Assessment of the role of leukocyte function-associated antigen-1 in homotypic adhesion of activated B lymphocytes.
Scand J Immunol.
48
1998
642
650
15
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
1332
16
Molina
IJ
Kenney
DM
Rosen
FS
Remold-O′Donnel
E
T cell lines characterize events in the pathogenesis of the Wiskott-Aldrich syndrome.
J Exp Med.
176
1992
867
874
17
Gallego
MD
Santamaria
M
Peña
J
Molina
IJ
Defective actin reorganization and polymerization of Wiskott-Aldrich T cells in response to CD3- mediated stimulation.
Blood.
90
1997
3089
3097
18
Facchetti
F
Blanzuoli
L
Vermi
W
et al
Defective actin polymerization in EBV-transformed B-cell lines from patients with the Wiskott-Aldrich syndrome.
J Pathol.
185
1998
99
107
19
Binks
M
Jones
GE
Brickell
PM
Kinnon
C
Katz
DR
Trasher
AJ
Intrinsic dendritic cell abnormalities in Wiskott-Aldrich syndrome.
Eur J Immunol.
28
1998
3259
3267
20
Badolato
R
Sozzani
S
Malacarne
F
et al
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
1033
21
Zicha
A
Allen
WE
Brickell
PM
et al
Chemotaxis of macrophages is abolished in the Wiskott- Aldrich syndrome.
Br J Haematol.
101
1998
659
665
22
Derry
JMJ
Wiedemann
P
Blair
P
et al
The mouse homolog of the Wiskott-Aldrich syndrome protein (WASP) gene is highly conserved and maps near the Scurfy (sf) mutation on the X chromsome.
Genomics.
29
1995
471
477
23
Miki
H
Miura
K
Takenawa
T
N-WASP, a novel actin-depolymerizing protein, regulates the cortical cytoskeletal rearrangement in a PIP2-dependent manner downstream of tyrosine kinases.
EMBO J.
15
1996
5326
5335
24
Machesky
LM
Insall
RH
Scar1 and the related Wiskott-Aldrich syndrome protein, WASP, regulate the actin cytoskeleton through the Arp2/3 complex.
Curr Biol.
8
1998
1347
1356
25
Rohatgi
R
Ma
L
Miki
H
et al
The interaction between N-WASP and the Arp2/3 complex link Cdc42–dependent signals to actin assembly.
Cell.
97
1999
221
231
26
Machesky
LM
Gould
KL
The Arp2/3 complex: a multifunctional actin organizer.
Curr Opin Cell Biol.
11
1999
117
121
27
Symons
M
Derry
JMJ
Karlak
B
et al
Wiskott-Aldrich syndrome protein, a novel effector for the GTPase Cdc42Hs, is implicated in actin polymerization.
Cell.
84
1996
723
734
28
Linder
S
Nelson
D
Weiss
M
Aepfelbacher
M
Wiskott-Aldrich syndrome protein regulates podosomes in primary human macrophages.
Proc Natl Acad Sci U S A.
96
1999
9648
9653
29
Snapper
SB
Rosen
FS
Mizoguchi
E
et al
Wiskott-Aldrich syndrome protein-deficient mice reveal a role for WASP in T but not B cell activation.
Immunity.
9
1998
81
91
30
Zhang
BJ
Shehabeldin
A
da Cruz
LAG
et al
Antigen receptor-induced activation and cytoskeletal rearrangement are impaired in Wiskott- Aldrich syndrome protein-deficient mice.
J Exp Med.
190
1999
1329
1341
31
Aspenström
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
75
32
Bishop
AL
Hall
A
Rho GTPases and their effector proteins.
Biochem J.
348
2000
241
255
33
Allen
WE
Zicha
D
Ridley
AJ
Jones
GE
A role for Cdc42 in macrophage chemotaxis.
J Cell Biol.
141
1998
1147
1157
34
Nobes
CD
Hall
A
Rho GTPases control polarity, protrusion, and adhesion during cell movement.
J Cell Biol.
144
1999
1235
1244
35
Prokopenko
SN
Saint
R
Bellen
HJ
Untying the Gordian knot of cytokinesis: role of small G proteins and their regulators.
J Cell Biol.
148
2000
843
848
36
Cox
D
Chang
P
Zhang
Q
Reddy
PG
Bokoch
GM
Greenberg
S
Requirements for both Rac1 and Cdc42 in membrane ruffling and phagocytosis in leukocytes.
J Exp Med.
186
1997
1487
1494
37
Caron
E
Hall
A
Identification of two distinct mechanisms of phagocytosis controlled by different Rho GTPases.
Science.
282
1998
1717
1721
38
Karasuyama
H
Melchers
F
Establishment of mouse cell lines which constitutively secrete large quantities of interleukin 2, 3, 4 or 5 using modified cDNA expression vectors.
Eur J Immunol.
18
1988
97
104
39
Heath
AW
Wu
WW
Howard
MC
Monoclonal antibodies to murine CD40 define two distinct functional epitopes.
Eur J Immunol.
24
1994
1828
1834
40
Rao
M
Knox
R
Conrad
DH
Characterization of Pgp-1 antigen on murine B lymphocytes using a new anti–Pgp-1 monoclonal antibody.
Hybridoma.
10
1991
281
284
41
Guinamard
R
Aspenström
P
Fougereau
M
Chavrier
P
Guillemot
J-C
Tyrosine phosphorylation of the Wiskott-Aldrich syndrome protein by Lyn and Btk is regulated by Cdc42.
FEBS Lett.
434
1998
431
436
42
Davey
EJ
Bartlett
WC
Kikutani
H
et al
Homotypic aggregation of murine B lymphocytes is independent of CD23.
Eur J Immunol.
25
1995
1224
1229
43
de Rooij
J
Bos
JL
Minimal Ras-binding domain of Raf1 can be used as an activation-specific probe for Ras.
Oncogene.
14
1997
623
625
44
Ridley
AJ
Paterson
HF
Johnston
CL
Diekmann
D
Hall
A
The small GTP-binding protein rac regulates growth factor-induced membrane ruffling.
Cell.
70
1992
401
410
45
Nobes
CD
Hall
A
Rho, rac and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, filopodia.
Cell.
81
1995
53
62
46
Rittinger
K
Walker
PA
Eccleston
JF
Smerdon
SJ
Gamblin
SJ
Structure at 1.65 Å of Rho A and its GTPase-activating protein in complex with a transition-state analogue.
Nature.
389
1997
758
762
47
Ihara
K
Muraguchi
S
Kato
M
et al
Crystal structure of human RhoA in a dominantly active form complexed with a GTP analogue.
J Biol Chem.
273
1998
9656
9666
48
Feig
LA
Tools of the trade: use of dominant-inhibitory mutants of Ras-family GTPases.
Nat Cell Biol.
1
1999
E25
E27
49
Zhang
B
Wang
Z-X
Zheng
Y
Characterization of the interactions between the small GTPase Cdc42 and its GTPase-activating proteins and putative effectors.
J Cell Biol.
272
1997
21999
22007
50
Newman
I
Wilkinson
PC
Methods for phenotyping polarized and locomotor human lymphocytes.
J Immunol Methods.
147
1992
43
50
51
Lemahieu
V
Gastier
JM
Francke
U
Novel mutations in the Wiskott-Aldrich syndrome protein gene and their effects on transcriptional, translational, and clinical phenotypes.
Hum Mutat.
14
1999
54
66
52
Kim
AS
Kakalis
LT
Abdul-Manan
N
Liu
GA
Rosen
MK
Autoinhibition and activation mechanisms of the Wiskott-Aldrich syndrome protein.
Nature.
404
2000
151
158
53
Glogauer
M
Hartwig
J
Stossel
T
Two pathways through Cdc42 couple the N-formyl receptor to actin nucleation in permeabilized human neutrophils.
J Cell Biol.
150
2000
785
796
54
Hoffman
GR
Nassar
N
Cerione
RA
Structure of the Rho family GTP-binding protein Cdc42 in complex with the multifunctional regulator RhoGDI.
Cell.
100
2000
345
356
55
Cox
AD
Der
CJ
Protein prenylation: more than just glue?
Curr Opin Cell Biol.
4
1992
1008
1016
56
Quinn
MT
Evans
T
Loetterle
LR
Jesaitis
AJ
Bokoch
GM
Translocation of Rac correlates with NADPH oxidase activation.
J Biol Chem.
268
1993
20983
20987
57
Bokoch
GM
Bohl
BP
Chuang
T-H
Guanine nucleotide exchange regulates membrane translocation of Rac/Rho GTP-binding proteins.
J Biol Chem.
269
1994
31674
31679
58
Benard
V
Bohl
BP
Bokoch
GM
Characterization of rac and Cdc42 activation in chemoattractant-stimulated human neutrophils using a novel assay for active GTPases.
J Biol Chem.
274
1999
13198
13204
59
Wery-Zennaro
S
Zugaza
JL
Letourneur
M
Bertologi
J
Pierre
J
IL-4 regulation of IL-6 production involves rac/Cdc42- and p38 MAPK-dependent pathways in keratinocytes.
Oncogene.
19
2000
1596
1604
60
Thrasher
AJ
Jones
GE
Kinnon
C
Brickell
PM
Katz
DR
Is Wiskott-Aldrich syndrome a cell trafficking disorder.
Trends Immunol Today.
19
1998
537
539
61
Kawabe
T
Naka
T
Yoshida
K
et al
The immune responses in CD40-deficient mice: impaired immunoglobulin class switching and germinal center formation.
Immunity.
1
1994
167
178
62
Sanders
VM
Snyder
JM
Uhr
JW
Vitetta
ES
Characterization of the physical interaction between antigen-specific B and T cells.
J Immunol.
137
1986
2395
2404
63
Sanders
VM
Fernandez-Botran
R
Uhr
JW
Vitetta
ES
Interleukin 4 enhances the ability of antigen-specific B cells to form conjugates with T cells.
J Immunol.
139
1987
2349
2354
64
Kupfer
A
Mosmann
TR
Kupfer
H
Polarized expression of cytokines in cell conjugates of helper T cells and splenic B cells.
Proc Natl Acad Sci U S A.
88
1991
775
779
65
Ozaki
ME
Coren
BA
Huynh
TN
Redondo
DJ
Kikutani
H
Webb
SR
CD4+ T cell responses to CD40-deficient APCs: defects in proliferation and negative selection apply only with B cells as APCs.
J Immunol.
163
1999
5250
5256

Author notes

Lisa Westerberg, Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, 171 77 Sockholm, Sweden; e-mail: lisa.westerberg@cmb.ki.se.

Sign in via your Institution