Interleukin-3 (IL-3)–induced activation of endogenous Rac-1, Rac-2, and Cdc42. Rac-1 was also activated by colony-stimulating factor-1 (CSF-1), Steel locus factor (SLF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-5 or by cross-linking the B-lymphocyte receptor for antigen (BCR). The activation of Rac-1 induced by cross-linking the BCR or by IL-3 stimulation was blocked only partially by Ly294002, with about 25% to 30% of Rac-1 activation still occurring in the absence of detectable increases in phosphatidyl-inositol-3 kinase (PI-3K) activity. Overexpression of constitutively active mutants of H-Ras, N-Ras, or M-Ras resulted in activation of coexpressed Rac-1 through an Ly29402-resistant, PI-3K–independent mechanism. Overexpression of constitutively active mutants of p21 Ras, or Rac-1, but not of PI-3K, was sufficient for activation of p38 mitogen-activated protein kinase (MAPK) in cells of hemopoietic origin. Inhibition of increases in PI-3K activity by Ly294002 had no effect on the IL-3–induced activation of p38 MAPK. In contrast, Ly294002 partially inhibited the activation of p38 MAPK induced by cross-linking of the BCR, although some p38 MAPK activation occurred in the absence of increases in the activity of Rac-1 or PI-3K. The activation of Rac-1, Rac-2, and Cdc42 by IL-3 and other hemopoietic growth factors is likely to be an important component of their actions in promoting growth, survival, and function.

Small guanosine triphosphatases (GTPases) of the Ras and Rho families cycle between an inactive, guanosine diphosphate (GDP)–bound form, and a GTP-bound form with affinity for a series of effector proteins that control signal transduction cascades regulating a multiplicity of cellular changes. Rac-1, Rac-2, and Rac-3 form a closely related subfamily of the Rho GTPases. Cdc42 is more distantly related and, like Rac-1, is expressed ubiquitously.1-3 In contrast, the expression of Rac-2 is restricted to cells of the lymphohemopoietic system.1 Most clues to the function of the Rac family have come from experiments involving either overexpression of constitutively active or dominant-negative mutants of Rac-1 or microinjection of constitutively active Rac-1.4-6They have implicated Rac-1 in actin-mediated processes including membrane-ruffling and chemotaxis, and in signaling paths involving the p21 activated kinase (PAK) family of serine/threonine kinases, p38 mitogen–activated protein kinase (p38 MAPK), and the c-jun-terminal kinase (JNK).4-6 Similar approaches have implicated Rac in a variety of functions in the immune system including phagocytosis, chemotaxis, production of oxidants by neutrophils, cell-mediated cytotoxicity, and the differentiation of lymphocytes of the T helper-1 subclass.7-14 

An obvious caveat to experiments that involve supraphysiologic levels of constitutively active or dominant-negative mutant proteins is that they may not accurately identify physiologic functions. The interpretation of experiments using dominant-negative mutants of Rac is particularly problematic because there are exchange factors (eg, mSos-1, Ras GRF1 and 2) with 2 catalytic domains that are able to bind to and activate, respectively, members of the Ras and Rac families.4,15-18 Thus, dominant-negative Rac mutants will bind to and sequestrate activators of Rac that also activate members of the Ras pathway, and vice versa. Analysis of hemopoietic cells from gene-targeted mice lacking Rac-2 has provided clear evidence for specific functions of Rac-2 that are not complemented by other Rac family members. Thus, the absence of Rac-2 resulted in defects in chemotaxis, adhesion, and superoxide production in neutrophils,19 and growth, survival, chemotaxis, adhesion, and degranulation in mast cells.20 There were also surprises not predicted by previous experimentation with the methodologies discussed above, including indications that Rac-2 was upstream of Erk MAPK,19 and Akt.20 There were also observations revealing an unexpected interregulation of Rac-2 and Rac-1 and Cdc42. Mast cells from mice lacking Rac-2 expressed much higher levels of Rac-1 (which were not diminished when Rac-2 was reintroduced),20 whereas hemopoietic stem cells lacking Rac-2 exhibited increased activation of Cdc42.21 These observations emphasize the limitations to our current picture of signaling events involving Rac.

There is only one report of a direct biochemical examination of the ability of hemopoietic growth factors to activate endogenous Rac. Surprisingly, this study concluded that, whereas treatment of neutrophils with fMetLeuPhe (fMLP) induced strong activation of Rac, treatment with granulocyte-macrophage colony-stimulating factor (GM-CSF), or tumor necrosis factor-α (TNF-α) did not.22 

Biochemical evidence for the activation of Rac by platelet-derived growth factor (PDGF), fMLP, leukotrienes, and cross-linking of the T-cell receptor has been previously reported.23-26 The general mechanisms of activation are thought to involve the recruitment to the plasma membrane of guanine-nucleotide exchange factors (GEFs), which trigger the exchange of Rac-bound GDP for GTP. With one notable exception,27,28 the GEFs that are active on the Rho family contain pleckstrin homology (PH) domains that bind to or have the potential to bind to membrane-bound lipid products of the action of phosphatidyl-inositol-3 kinase (PI-3K).29,30 Moreover, studies with pharmacologic inhibitors of PI-3K (Ly294002 and wortmannin) have demonstrated that PI-3K is necessary for activation of Rac by many stimuli.23-25,31 However, activation of Rac-2 by treatment of neutrophils with phorbol myristate acetate (PMA) was not blocked by an inhibitor of PI-3K activity, pointing to the existence of Rac GEFs that do not require PI-3K activity for their action on Rac.25 

Overexpression of dominant-active or dominant-inhibitory mutants of Rac have implicated it in the activation of both families of “stress activated kinases” (p38 MAPK and JNK) in fibroblasts,32-35 and of JNK in hemopoietic cells.36,37 Hemopoietic growth factors, whether acting through receptor tyrosine kinases (eg, Steel locus factor [SLF] or colony-stimulating factor 1 [CSF-1]), or receptors of the cytokine receptor superfamily (eg, interleukin-3 [IL-3] or GM-CSF) activate both p38 MAPK and JNK.36,38-40 Activation of p38 MAPK results from dual phosphorylation on threonine and tyrosine residues of a TGY motif in its activation loop by the upstream MAPK kinases (MKKs), MKK3 and MKK6.41-43 

Here we have directly investigated activation of endogenous Rac-1, Rac-2, and Cdc42 using antibodies we have demonstrated to be operationally specific for these molecules. We have tested as stimuli a variety of hemopoietic growth factors active on 2 classes of receptors and ligation of the B-cell receptor for antigen (BCR). Further, we have examined whether activation of one Rac isoform, Rac-1, correlates with activation of p38 MAPK. We observed that endogenous Rac-1 was activated by 5 hemopoietic growth factors acting through 2 different classes of receptor and by cross-linking of the BCR. However, activation of p38 MAPK was not always correlated with the degree of activation of Rac-1. Thus, whereas activation of Rac-1 via the IL-3 receptor was largely PI-3K dependent (70% inhibition by Ly294002), activation of p38 MAPK remained unaffected by inhibition of PI-3K activity. Moreover, the activation of Rac-1 induced by overexpression of the constitutively active mutants of 3 isoforms of Ras, H-Ras, N-Ras, and M-Ras, was completely insensitive to Ly294002. These results suggest that IL-3 can activate Rac-1 and p38 MAPK via mechanisms that do not depend on PI-3K and may involve Ras.

cDNA constructs, antibodies, and reagents

The vectors encoding G12V H-Ras, Q61K N-Ras, V12L Rac-1, and constitutively active PI-3K (p110*) were gifts from Dr A. Ambrosini (DiBiT, H. San Raffaele, Milan, Italy), Dr Rob Kay (The Terry Fox Laboratories, Vancouver, BC, Canada), Dr Frank McCormick (University of California, San Francisco Cancer Research Institute), and Dr Lewis T. Williams (Chiron, Emeryville, CA), respectively. M-Ras was cloned as described previously.44 The bacterial expression vector, pGEX, encoding a glutathione-S-transferase (GST)–PAK fusion protein (amino acids 59-145 of human PAK-1β; GST-PAK) and the mammalian expression vector, pRK5, encoding myc tagged Rac-1 were kind gifts from Dr Alan Hall (MRC Laboratory for Molecular Cell Biology, London, England). The pEGFP-C1 vector was purchased from Clontech (Palo Alto, CA). The Rac-1 mouse monoclonal antibody was purchased from Upstate Biotechnology (Lake Placid, NY). Rabbit polyclonal antibodies specific for Rac-225 and Cdc42 (Figure1A-B and data not shown) were from Santa Cruz Biotechnology (Santa Cruz, CA). Antibodies specific for phosphorylated molecules, including phospho-Erk (mouse monoclonal), phospho-serine 473 of Akt (rabbit polyclonal), and phospho-p38 MAPK (rabbit polyclonal) were from New England Biolabs (Beverly, MA). Polyclonal rabbit antibodies recognizing unphosphorylated Erk and p38 MAPK were from Santa Cruz Biotechnology and Akt was from New England Biolabs. The F(ab′)2 fragments of goat antimouse IgM were purchased from Jackson Immunoresearch Laboratories (West Grove, PA). Recombinant murine cytokines: CSF-1, SLF, and IL-5 were obtained from R & D Systems (Windsor, ON, Canada). Dr Ian Clark-Lewis (The Biomedical Research Centre, Vancouver, BC, Canada) generously provided synthetic murine IL-3 and GM-CSF. Ly294002 was obtained from Calbiochem-Novabiochem (La Jolla, CA) and stock solutions were made at 50 mM in 100% ethanol unless indicated otherwise.

Fig. 1.

Specificity of Rac antibodies and activation of Rac-1 by β-common family cytokines.

Baf/3 cells were electroporated with 15 μg constitutively active Rac-1 (Rac-1 myc) or Rac-2 (Rac-2 myc). At 16 hours after electroporation, cells were lysed and both overexpressed and endogenous Rac GTPases were precipitated using recombinant GST-PAK bound to GT beads. Antibodies specific for Rac-1 or Rac-2 were used for immunoblotting serial 2-fold dilutions (1, ½, ¼) of lysates with (A) overexpressed, myc-tagged Rac-1 or Rac-2 as indicated or (B) endogenous and overexpressed Rac-1 or Rac-2 isoforms. (C) MC/9 cells were incubated for 5 minutes with PBS (con), GM-CSF (10 μg/mL), IL-3 (5 μg/mL), or IL-5 (50 ng/mL) and lysed. Immunoblots of Rac-1 GTP bound by beads and in WCLs were performed using a Rac-1–specific monoclonal antibody (Rac-1 GTP). One half of the total GTP-bound Rac-1 precipitated was run in immunoblots. The WCLs corresponded to 1.3%, 1.2%, and 1.2% of the total Rac available for precipitation for GM-CSF, IL-3, and IL-5, respectively. Numbers below immunoblots of Rac-1 GTP (and of other GTP-bound GTPases in succeeding figures) represent the ratios of the optical density of the immunoblotted band of GST-PAK precipitated Rac-1 from stimulated cells to the optical density of the immunoblotted band of GST-PAK precipitated Rac-1 from control, unstimulated cells.

Fig. 1.

Specificity of Rac antibodies and activation of Rac-1 by β-common family cytokines.

Baf/3 cells were electroporated with 15 μg constitutively active Rac-1 (Rac-1 myc) or Rac-2 (Rac-2 myc). At 16 hours after electroporation, cells were lysed and both overexpressed and endogenous Rac GTPases were precipitated using recombinant GST-PAK bound to GT beads. Antibodies specific for Rac-1 or Rac-2 were used for immunoblotting serial 2-fold dilutions (1, ½, ¼) of lysates with (A) overexpressed, myc-tagged Rac-1 or Rac-2 as indicated or (B) endogenous and overexpressed Rac-1 or Rac-2 isoforms. (C) MC/9 cells were incubated for 5 minutes with PBS (con), GM-CSF (10 μg/mL), IL-3 (5 μg/mL), or IL-5 (50 ng/mL) and lysed. Immunoblots of Rac-1 GTP bound by beads and in WCLs were performed using a Rac-1–specific monoclonal antibody (Rac-1 GTP). One half of the total GTP-bound Rac-1 precipitated was run in immunoblots. The WCLs corresponded to 1.3%, 1.2%, and 1.2% of the total Rac available for precipitation for GM-CSF, IL-3, and IL-5, respectively. Numbers below immunoblots of Rac-1 GTP (and of other GTP-bound GTPases in succeeding figures) represent the ratios of the optical density of the immunoblotted band of GST-PAK precipitated Rac-1 from stimulated cells to the optical density of the immunoblotted band of GST-PAK precipitated Rac-1 from control, unstimulated cells.

Close modal

Cell culture

Cells were grown at 37°C in humidified incubators gassed with 5% CO2 using RPMI 1640 medium (Stem Cell Technologies, Vancouver, BC, Canada), supplemented with 10% (vol/vol) fetal calf serum (FCS; Cansera, Rexdale, ON, Canada), 50 μM β-mercaptoethanol, 0.2 mM l-glutamine, and 1 mM sodium pyruvate. The IL-3–dependent cell lines, Baf/3, WEHI 274.3, R6/X, and MC/9, were cultured in 4% WEHI 3B-conditioned medium as a source of IL-3. Primary bone marrow–derived mast cells (BMMCs) were generated from 4- to 8-week-old (C57BL/6 x DBA/2) F1 hybrid mice (BDF1) or Balb/c mice by culturing bone marrow cells in 4% WEHI 3B-conditioned medium for 3 to 4 weeks to generate mature BMMCs as described previously.45 Primary B-cell blasts were generated from the spleens of 4- to 8-week-old BDF1 hybrid mice. A single-cell suspension of splenocytes was obtained and red cells lysed using red cell removal buffer (0.017 M NH4Cl, 0.14 M Tris [tris(hydroxymethyl)aminomethane], pH 7.2). After depletion of adherent cells by incubation for 2 hours on tissue culture–treated plastic dishes in 10% FCS and RPMI, splenocytes were resuspended at 2 × 106 cells/mL with 15 μg/mL lipopolysaccharide (LPS) and cultured for 72 hours to generate B-cell blasts.

Cell stimulation

Prior to stimulation, factor-dependent cell lines or BMMCs were cultured overnight in one tenth the concentration of growth factor in which they were normally grown. Cells were then washed 2 times in serum-free media (SFM) containing 20 mM HEPES (N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid; pH 7.2). MC/9 or WEHI 274.3 were resuspended at 107 cells/mL and BMMCs at 1.5 × 107 cells/mL in SFM for 1.5 hours at 37°C. Cells were stimulated in 1 mL SFM by addition of saturating concentrations of recombinant or synthetic cytokines: IL-5 (50 ng/mL), SLF (50 ng/mL), CSF-1 (200 ng/mL), IL-3 (5 μg/mL), and GM-CSF (10 μg/mL). After incubation with LPS for 72 hours, cultures of splenocytes were washed 2 times in SFM, resuspended in SFM at 1.5 × 107 cells/mL, and incubated for 3 hours at 37°C in SFM. B lymphocytes (1.5 × 107 cells in 1 mL) were stimulated by addition of 40 μg F(ab′)2 fragments of goat antimouse IgM. Ly294002 or solvent was added to the cells during the final 15 minutes of serum starvation (prior to stimulation) where indicated.

Electroporation

Baf/3, MC/9, or R6/X cells (2.0 × 107) were incubated in 500 μL SFM with 10 μg/mL diethylaminoethyl (DEAE) dextran for 20 minutes at 37°C. The suspension was transferred to a 0.4-cm electroporation cuvette and electroporated at 300 V and 975 μF using a Gene Pulser II from Bio-Rad (Hercules, CA) yielding a time constant of 20 to 25 milliseconds. After electroporation, cells were incubated at 37°C in RPMI, 10% FCS, and 4% WEHI 3B-conditioned medium for 10 to 16 hours at 106 cells/mL. Cells were then washed twice in SFM and incubated at 1.0 × 107 cells/mL in SFM for 2 hours. Where indicated, cells were treated with Ly294002 for 30 minutes during the last half-hour of serum starvation.

Assay for activation of Rac-1, Rac-2, and Cdc42

Recombinant GST-PAK was prepared by growing Escherichia coli (DH5α) to an optical density (600 nm) of 0.6 at 37°C and inducing fusion protein production for 2 hours at 25°C with 0.1 mM isopropyl β-d-thiogalactoside (IPTG). Cells were pelleted and sonicated in phosphate-buffered saline (PBS) containing 1% Triton X-100 and protease inhibitors (protease inhibitor cocktail tablets; Boehringer Mannheim, Mannheim, Germany). The soluble fraction of bacterial sonicate was applied to glutathione (GT) Sepharose 4B beads (Amersham Pharmacia Biotech, Baie d'Urfe, QC, Canada) for 30 minutes at 4°C, which were washed twice in PBS containing 0.1% Tween-20 (PBST) and 1 mM dithiothreitol (DTT) and twice in PBST without DTT. To assess Rac/Cdc42 activation, equal numbers of cells were lysed in a buffer containing 50 mM Tris, pH 7.5, 10% glycerol, 1% Nonidet P-40, 30 mM MgCl2, 150 mM NaCl, 1 mM sodium molybdate, 200 mM sodium orthovanadate, 1 mM sodium fluoride, 50 mM β-glycerol phosphate, 1 μg/mL microcystin, 10 μg/mL aprotinin, 10 μg/mL soy bean trypsin inhibitor, 2 μg/mL leupeptin, 0.7 μg/mL pepstatin, and 40 μg/mL phenylmethylsulfonyl fluoride (PMSF). Total protein in whole cell lysates (WCLs) was quantified using a BCA protein assay kit (Pierce, Rockford, IL). Aliquots of WCLs were added to 20 μL GT beads with bound GST-PAK and were rotated for 30 minutes at 4°C. The supernatant was removed from the beads and retained for immunoblotting. Beads were then washed 3 times with 1 mL of the same buffer used to lyse the cells. Sodium dodecyl sulfate (SDS) sample buffer was applied to aliquots of whole cell lysates or bead pellets and heated to 80°C for 10 minutes. Known aliquots of SDS eluates of beads or WCLs were subject to SDS-polyacrylamide gel electrophoresis (SDS-PAGE; 12% acrylamide) and immunoblotting. Blots were probed with a Rac-1–specific mouse monoclonal antibody or rabbit polyclonal antibodies specific for Cdc42 or Rac-2 to quantify levels of the respective activated GTPases. The ratio of GTP-bound GTPase in stimulated cells versus unstimulated controls was estimated from measurements of arbitrary units of optical density of bands corresponding to the GTPase in immunoblots of the respective samples. The ratio of the optical density of the band corresponding to the GTPase in immunoblots of eluates from the GST-PAK–coated beads, and in an immunoblot of a known aliquot of the WCLs, provided an estimate of the absolute amount of GTP-bound GTPase as a fraction of the total cellular GTPase. The amount of total Rac/Cdc42 in the sample of WCLs, which was subjected to SDS-PAGE and immunoblotting, corresponded to 1% to 4% of that applied to the GST-PAK–coated beads as indicated. The degree of activation of GTPases was also expressed as the ratio of the optical density of the immunoblotted band of activated GTPase precipitated from stimulated cells to the optical density of the immunoblotted band of activated GTPase precipitated from unstimulated cells. Goat antibodies specific for rabbit or mouse immunoglobulins and conjugated to horseradish peroxidase (Dako, Glostrup, Denmark) were used as secondary antibodies. Immunoblots were developed using enhanced chemiluminescence (Amersham Pharmacia Biotech).

Assay for activation of p38 MAPK, Akt, or Erk

Blots of WCLs (50-100 μg total protein) were probed with phospho-specific antibodies to assess levels of activated p38 MAPK, Akt, or Erk as described above. To confirm equivalency of loading, membranes were stripped of bound antibodies by exposure to a solution containing 62.5 mM Tris, 0.2% SDS, and 100 mM β-mercaptoethanol for 30 minutes at 55°C, followed by washing with PBST and reprobing with antibodies specific for the unphosphorylated forms of p38 MAPK, Erk, or Akt.

Activation of endogenous Rac-1, Rac-2, and Cdc42 by multiple hemopoietic growth factors

The activation of Rac-1, Rac-2, and Cdc42 was measured by specifically coprecipitating GTP-bound forms of these GTPases using a recombinant fusion protein (GST-PAK) of GST and amino acid residues 59-145 of PAK-1β, including the Cdc42/Rac-interactive binding domain.46,47 Because the GST-PAK coprecipitates a variety of activated Rho family GTPases, it was critical to use antibodies specific for the desired GTPase. To assess the specificity of the antibodies we have used to identify Rac-1 and Rac-2, we expressed myc-tagged constitutively active mutants of these proteins in Baf/3 cells. We coprecipitated the exogenous GTPases with GST-PAK and split the precipitates into aliquots that were subjected to SDS-PAGE and immunoblotting with either a monoclonal antibody specific for Rac-1 or polyclonal antibodies specific for Rac-2. As shown in Figure 1A, the Rac-1–specific monoclonal antibody specifically recognized constitutively active Rac-1 while detecting absolutely none of the constitutively active Rac-2 present. The converse was observed with immunoblotting with polyclonal antibodies specific for Rac-2 (Figure1A), in agreement with previous assessments of the specificity of these antibodies.25 Moreover, as shown in Figure 1B, the Rac-1–specific monoclonal antibody was able to specifically detect overexpressed myc-tagged Rac-1, but not myc-tagged Rac-2 while recognizing endogenous Rac-1. The converse was observed when fractions of the same coprecipitation were immunoblotted with the Rac-2–specific polyclonal antibodies. These results indicated that the antibodies used to detect Rac-1 and Rac-2 were indeed operationally specific for their respective targets and had the necessary specificity and sensitivity for use in assessing activation of specific endogenous isoforms that occur naturally as a mixture in murine hemopoietic cells. The polyclonal antibodies specific for Cdc42 have been used previously23,24,25 and we confirmed that they recognized recombinant Cdc42, but did not cross-react with Rac-1 or p21 Ras (data not shown).

The mouse mast cell-like cell line, MC/9, was chosen as a convenient model for study of the activation of endogenous Rac-1 because it responds to multiple hemopoietic growth factors. These include IL-3, GM-CSF, and IL-5, all of which act through receptors of the hemopoietin receptor family, as well as SLF, which acts through a receptor-tyrosine kinase.

In contrast to published data indicating that GM-CSF failed to activate Rac in human neutrophils,22 we observed rapid activation of Rac-1 by GM-CSF (Figure 1C). This activation was maximal at 1 minute and was maintained for at least 10 minutes. Stimulation of MC/9 cells with IL-3 also induced activation of Rac-1, although the kinetics were slower with maximal activation occurring after 5 minutes and being maintained for at least 10 minutes (Figure 1C). Similar results were observed with IL-5 (Figure 1C). The levels of Rac-1 GTP reached after stimulation by these 3 growth factors was estimated to range from 0.8% to 1.4% (0.8% for GM-CSF, 1.2% for IL-3, and 1.4% for IL-5) as a percentage of the total Rac-1 present in WCL.

We also used cultures of BMMCs to confirm that IL-3 activated endogenous Rac-1 in primary cells. IL-3 treatment of BMMCs resulted in greater increases in the levels of activated Rac-1 over those seen in resting cells (4.3-fold), than those seen in MC/9 (2.4-fold; Figure2A compared with Figure 1C). The estimated percentage of total cellular Rac-1 activated in IL-3–treated BMMCs was also greater than that seen in IL-3–treated MC/9 cells (3.6% in BMMCs compared with 1.2% in MC/9).

Fig. 2.

IL-3 activates multiple Rho family GTPases in BMMCs.

BMMCs generated from BDF1 mice were incubated for 5 minutes with PBS (con) or IL-3 (5 μg/mL). Cells were lysed and GST-PAK was used to precipitate endogenous, GTP-bound Rac-1, Rac-2 or Cdc42. Immunoblots were performed with (A) a Rac-1 specific monoclonal antibody, (B) polyclonal antibodies specific for Rac-2, or (C) polyclonal antibodies specific for Cdc42. Immunoblots of Rac/Cdc42 present in WCLs were performed with the same antibodies as used to visualize Rac/Cdc42-GTP precipitated with GST-PAK (except for the case of Rac-2 where, for technical reasons, the Rac-1 antibody was used to probe the immunoblot of the WCL). One half of total GTP-bound Rac-1 precipitated was run in the Rac-1 immunoblot; the WCL corresponded to 3% of total Rac-1 available for precipitation. The total amount of GTP-bound Cdc42 precipitated was run in the Cdc42-specific immunoblot; the WCL corresponded to 4% of the total Cdc42 available for precipitation. Shown below immunoblots GTP-bound GTPases in this and succeeding figures are numbers corresponding to the ratios of the optical densities of the bands from stimulated versus control cells.

Fig. 2.

IL-3 activates multiple Rho family GTPases in BMMCs.

BMMCs generated from BDF1 mice were incubated for 5 minutes with PBS (con) or IL-3 (5 μg/mL). Cells were lysed and GST-PAK was used to precipitate endogenous, GTP-bound Rac-1, Rac-2 or Cdc42. Immunoblots were performed with (A) a Rac-1 specific monoclonal antibody, (B) polyclonal antibodies specific for Rac-2, or (C) polyclonal antibodies specific for Cdc42. Immunoblots of Rac/Cdc42 present in WCLs were performed with the same antibodies as used to visualize Rac/Cdc42-GTP precipitated with GST-PAK (except for the case of Rac-2 where, for technical reasons, the Rac-1 antibody was used to probe the immunoblot of the WCL). One half of total GTP-bound Rac-1 precipitated was run in the Rac-1 immunoblot; the WCL corresponded to 3% of total Rac-1 available for precipitation. The total amount of GTP-bound Cdc42 precipitated was run in the Cdc42-specific immunoblot; the WCL corresponded to 4% of the total Cdc42 available for precipitation. Shown below immunoblots GTP-bound GTPases in this and succeeding figures are numbers corresponding to the ratios of the optical densities of the bands from stimulated versus control cells.

Close modal

Because Rac-2 is specifically expressed in hemopoietic cells, it was interesting to examine activation of this Rac isoform.1,48Endogenous Rac-2, as detected by antibodies specific for Rac-2 (Figure1),25 was clearly activated by IL-3 in BMMCs (Figure 2B), although limitations of the Rac-2 antibodies prevented accurate quantitation of the percentage of the total cellular Rac-2 that was activated. IL-3 stimulation of BMMCs also induced increased levels of GTP-bound Cdc42 (activation of approximately 1% of total cellular Cdc42; Figure 2C). We observed that CSF-1 and SLF, 2 other hemopoietic growth factors, acting through a different class of receptor (receptor tyrosine kinases), also activated Rac-1. SLF stimulation of MC/9 cells induced maximal increases in levels of Rac-1 GTP by 1 minute, but levels had dropped considerably by 5 minutes (Figure3A). SLF also induced activation of endogenous Rac-1 in primary BMMCs (Figure 3B). Stimulation of the myelomonocytic cell line, WEHI 274.3, with CSF-1, led to rapid activation of Rac-1, with levels reaching a maximum at 1 minute, and dropping significantly by 5 minutes (Figure 3C). The percentages of Rac-1 GTP (as a fraction of the total Rac-1 pool present) in cells activated by SLF (4% in BMMCs and 2.4% in MC/9) or by CSF-1 (5%) were again relatively small.

Fig. 3.

Activation of Rac-1 stimulated by hemopoietic growth factors acting through receptor tyrosine kinases.

(A) MC/9 cells were stimulated with SLF (50 ng/mL), (B) BMMCs from BDF1 mice were stimulated with SLF (50 ng/mL), or (C) WEHI 274.3 cells were stimulated with CSF-1 (200 ng/mL) for the indicated number of minutes or with PBS for 5 minutes (con). Following stimulation, lysates were assayed for GTP-bound/activated endogenous Rac-1 (Rac-1 GTP) as specified earlier. Immunoblots were performed using a Rac-1–specific monoclonal antibody. One half of the total GTP-bound Rac-1 precipitated was run in immunoblots for cells described in panels A, B, and C. The WCLs corresponded to 1.9%, 2.6%, and 2.1% of the total Rac-1 available for precipitation from cells in panels A, B, and C, respectively.

Fig. 3.

Activation of Rac-1 stimulated by hemopoietic growth factors acting through receptor tyrosine kinases.

(A) MC/9 cells were stimulated with SLF (50 ng/mL), (B) BMMCs from BDF1 mice were stimulated with SLF (50 ng/mL), or (C) WEHI 274.3 cells were stimulated with CSF-1 (200 ng/mL) for the indicated number of minutes or with PBS for 5 minutes (con). Following stimulation, lysates were assayed for GTP-bound/activated endogenous Rac-1 (Rac-1 GTP) as specified earlier. Immunoblots were performed using a Rac-1–specific monoclonal antibody. One half of the total GTP-bound Rac-1 precipitated was run in immunoblots for cells described in panels A, B, and C. The WCLs corresponded to 1.9%, 2.6%, and 2.1% of the total Rac-1 available for precipitation from cells in panels A, B, and C, respectively.

Close modal

Activation of endogenous Rac-1 by cross-linking of the BCR

To investigate whether Rac-1 was activated by ligation of the endogenous receptor for antigen on B lymphocytes (BCRs), we used primary cultures of B lymphoblasts that had been generated by treatment of resting splenocytes with LPS. We cross-linked the BCR without engaging the Fc receptors by using the F(ab′)2 fragment of antibodies to IgM. This resulted in rapid activation of Rac-1, with maximal stimulation occurring at 1 minute (5.3-fold over background; Figure 4). This level of activated Rac-1 was maintained for 10 minutes. Once again, the absolute amount of Rac-1 activated by BCR-mediated stimulation represented only a small fraction of the total cellular Rac-1 (about 1%).

Fig. 4.

Activation of Rac-1 induced by cross-linking of the BCR.

Splenocytes from BDF1 mice were stimulated with LPS (15 μg/mL) for 72 hours to generate B lymphoblasts. B lymphoblasts were stimulated with F(ab′)2 fragments of antimouse IgM (40 μg/mL) for the indicated length of time or treated with PBS for 5 minutes (con). Following stimulation, lysates were assayed for GTP-bound endogenous Rac-1 (Rac-1 GTP) as specified earlier. Immunoblots were performed using a Rac-1 monoclonal antibody. One half of the total GTP-bound Rac-1 precipitated was run and the WCL corresponded to 1.4% of the total Rac-1 available for precipitation.

Fig. 4.

Activation of Rac-1 induced by cross-linking of the BCR.

Splenocytes from BDF1 mice were stimulated with LPS (15 μg/mL) for 72 hours to generate B lymphoblasts. B lymphoblasts were stimulated with F(ab′)2 fragments of antimouse IgM (40 μg/mL) for the indicated length of time or treated with PBS for 5 minutes (con). Following stimulation, lysates were assayed for GTP-bound endogenous Rac-1 (Rac-1 GTP) as specified earlier. Immunoblots were performed using a Rac-1 monoclonal antibody. One half of the total GTP-bound Rac-1 precipitated was run and the WCL corresponded to 1.4% of the total Rac-1 available for precipitation.

Close modal

Different requirements for PI-3K activity for activation of Rac-1 induced by IL-3 or cross-linking of the BCR

All known Rho family GEFs, which contain the Dbl homology (DH) catalytic domain, also contain a PH domain that is likely to bind phosphoinositides generated by PI-3K.29,49 Thus, if activation of Rac-1 by hemopoietic growth factors or ligation of the BCR involves these GEFs, it is likely to depend on PI-3K. To test this hypothesis, we investigated the effect of a pharmacologic inhibitor of PI-3K, Ly294002. The activation of Rac-1 induced by cross-linking the BCR was reduced by approximately 75% with concentrations of Ly294002 (3.1 μM) that reduced Akt phosphorylation to background levels (Figure 5B). This suggested that the remaining 25% of Rac-1 activation did not require increases in levels of PI-3K activity. With higher concentrations of Ly294002, levels of Akt phosphorylation were reduced to well below the background level in unstimulated cells. We observed that when background PI-3K activity was so reduced, Rac-1 activation was also reduced to near background levels (93% inhibition at 25 μM Ly294002; Figure 5B). This suggests that low background levels of overall PI-3K activity may be permissive for Rac-1 activation.

Fig. 5.

Effects of the PI-3K inhibitor, Ly294002, on activation of Rac-1 and p38 MAPK induced by IL-3 or cross-linking of the BCR.

(A) BMMCs from Balb/c mice or (B) LPS-stimulated B lymphoblasts from BDF1 mice were pretreated with Ly294002 or with ethanol (solvent), and subsequently stimulated for 5 minutes with PBS (con), IL-3 (1 μg/mL), or F(ab′)2 fragments of antimouse IgM (40 μg/mL). Activated, endogenous Rac-1 (Rac-1 GTP) was precipitated by GST-PAK bound to GT beads. Immunoblots were performed using a Rac-1–specific monoclonal antibody. Activation levels of endogenous Akt, Erk, and p38 MAPK were measured from WCLs using antibodies capable of specifically recognizing the phosphorylated form of these molecules (p-Akt, p-Erk, p-p38 MAPK). Equivalency of loading was confirmed by reprobing phosphoblots with antibodies capable of recognizing both phosphorylated and nonphosphorylated proteins (Akt, Erk, p38 MAPK).

Fig. 5.

Effects of the PI-3K inhibitor, Ly294002, on activation of Rac-1 and p38 MAPK induced by IL-3 or cross-linking of the BCR.

(A) BMMCs from Balb/c mice or (B) LPS-stimulated B lymphoblasts from BDF1 mice were pretreated with Ly294002 or with ethanol (solvent), and subsequently stimulated for 5 minutes with PBS (con), IL-3 (1 μg/mL), or F(ab′)2 fragments of antimouse IgM (40 μg/mL). Activated, endogenous Rac-1 (Rac-1 GTP) was precipitated by GST-PAK bound to GT beads. Immunoblots were performed using a Rac-1–specific monoclonal antibody. Activation levels of endogenous Akt, Erk, and p38 MAPK were measured from WCLs using antibodies capable of specifically recognizing the phosphorylated form of these molecules (p-Akt, p-Erk, p-p38 MAPK). Equivalency of loading was confirmed by reprobing phosphoblots with antibodies capable of recognizing both phosphorylated and nonphosphorylated proteins (Akt, Erk, p38 MAPK).

Close modal

Similar results were obtained with IL-3 stimulation of BMMCs, where activation of Rac-1 was only partially inhibited (by 70%) by Ly294002 even at high concentrations (50 μM; Figure 5A). Immunoblotting of cell lysates with an antibody specific for phosphorylated Akt confirmed that the IL-3–induced increase in PI-3K activity had been completely inhibited. These results suggested the possible existence of a pool of Rac-1 that was activated by IL-3 or ligation of the BCRs through mechanism(s) that did not require stimulus-mediated increases in PI-3K activity. However, because levels of Akt phosphorylation were never reduced below background to undetectable levels, we cannot exclude the possibility that a low level of PI-3K activity was permissive for IL-3–induced Rac-1 activation.

There were also differences in the sensitivity to inhibition by Ly294002 of activation of the p38 MAPK and Erk induced by IL-3 or by cross-linking of the BCR. Thus, Ly294002 had no inhibitory effect on the IL-3–induced activation of p38 MAPK or Erk (Figure 5A). The absence of any effect of Ly294002 on activation of these MAP family kinases was surprising given evidence implicating the Rac pathway in their activation,33-35 and our finding that Ly294002 inhibited IL-3–induced activation of Rac by 70%. These results suggested that, in the context of signaling through endogenous IL-3 receptors, part of the activation of Rac and all of the activation of p38 MAPK and Erk, was independent of IL-3–induced increases in PI-3K activity. However, in the case of ligation of the BCR, the presence of Ly294002 resulted in partial inhibition of the activation of both p38 MAPK and Erk 1/2 (Figure 5B). Thus, activation of Erk and p38 MAPK downstream of the BCR involved both PI-3K–dependent and PI-3K–independent pathways.

Ras-mediated activation of Rac-1 is PI-3K independent

Our observation that IL-3–induced activation of Rac-1 was not completely inhibited by concentrations of Ly294002 that completely blocked increases in PI-3K–mediated activation of Akt suggested the existence of alternative mechanisms of activating Rac-1 that did not require increased PI-3K activity. There is evidence that Ras proteins are upstream activators of Rac,4,18 and hemopoietic growth factors are known to activate Ras family members.50,51 To test the hypothesis that members of the Ras family could activate Rac-1 in hemopoietic cells, and to investigate whether this occurred through a mechanism involving increased PI-3K activity, we coexpressed wild-type Rac-1 and a constitutively active mutant of 1 of 2 p21 Ras isoforms, H-Ras and N-Ras, in the IL-3–dependent cell line, Baf/3. A plasmid encoding GFP, pEGFP-C1, was coexpressed in all samples to monitor cell viability and electroporation efficiency. Overexpression of either a constitutively active mutant of p21 H-Ras (G12V H-Ras) or N-Ras (Q61K N-Ras) induced activation of coexpressed Rac-1 (Figure6A). To investigate whether Ras-induced activation of Rac-1 was dependent on Ras-mediated increases in PI-3K activity, we examined the effects of treating cells with Ly294002. To determine the concentration of Ly294002 needed to give maximal inhibition of Akt phosphorylation in Baf/3 cells exhibiting high levels of PI-3K activity, we treated Baf/3 cells transfected with either constitutively active H-Ras (G12 H-Ras) or PI-3K (p110*) with titrated doses of Ly294002 (Figure 6C). We observed that 25 μM Ly294002 resulted in the reduction of Akt phosphorylation to background levels in cells overexpressing either the G12 H-Ras or the constitutively active PI-3K. Moreover, there was no further decrease in Akt phosphorylation in cells treated with 50 μM Ly294002. These experiments showed that, despite the fact that Ly294002 (25 μM) reduced the phosphorylation of Akt induced by expression of G12 H-Ras or Q61 N-Ras to background levels, it had no effect on the activation of exogenous Rac-1 (Figure 6A). Thus, expression of constitutively active p21 H or N-Ras activated coexpressed Rac-1 through a mechanism that did not depend on overall increases in PI-3K activity. Because we have shown that the method used to assay activation of “p21 Ras” by hemopoietic growth factors in published studies does not discriminate between isoforms of p21 Ras or the “nonclassical” member of the Ras family, M-Ras,44 it was important to determine whether M-Ras was upstream of Rac-1 as well. This was particularly relevant, because we have shown (A. Schallhorn and J.W.S., unpublished observations, September 2000) that hemopoietic growth factors such as IL-3 were strong activators of M-Ras but not of p21 H-Ras. Expression of a constitutively active mutant of M-Ras (Q71L M-Ras) in Baf/3 cells resulted in activation of coexpressed Rac-1 (Figure 6B). Moreover, treatment of cells expressing constitutively active M-Ras with Ly294002 (25 μM) had no effect on activation of coexpressed Rac-1, although the phosphorylation of Akt induced by expression of Q71L M-Ras was reduced to background levels (Figure 6B). Therefore, we have concluded that both p21 Ras isoforms (H-Ras and N-Ras) and M-Ras are capable of activating exogenous Rac-1 through mechanisms that do not depend on overall increases in PI-3K activity.

Fig. 6.

Overexpression of activated mutants of H-Ras, N-Ras, or M-Ras results in activation of Rac-1 via a mechanism, which is not inhibited by Ly294002.

Baf/3 cells were coelectroporated with 3 μg pEGFP-C1 empty vector, 3 μg wild-type, myc-tagged Rac-1, and 15 μg of (A and C) control plasmid (vector), a constitutively active mutant of PI-3K (p110*), or constitutively active mutants of one of 2 p21 Ras isoforms (G12 H-Ras and Q61 N-Ras), (B) control plasmid (vector), wild-type M-Ras (WT M-Ras), or a constitutively active mutant of M-Ras (Q71 M-Ras). At 16 hours after electroporation, cells were resuspended in serum-free medium for 2 hours. Ly294002 (25 μM dissolved in DMSO) was present for these 2 hours for N-RAS (A). Ly294002 (25 μM dissolved in ethanol) was present for the last 30 minutes for H-Ras (A), M-Ras (B), and H-Ras/p110* (C). After this, cells were lysed. Activated Rac-1 myc (Rac-1 myc GTP) was precipitated from lysates using GST-PAK bound to GT beads and detected by immunoblotting with a Rac-1 specific monoclonal antibody. The activation status of Akt and Erk was also determined from the same lysates using phospho-specific antibodies (p-Akt, p-Erk). Equivalency of loading was confirmed as described earlier (Akt, Erk).

Fig. 6.

Overexpression of activated mutants of H-Ras, N-Ras, or M-Ras results in activation of Rac-1 via a mechanism, which is not inhibited by Ly294002.

Baf/3 cells were coelectroporated with 3 μg pEGFP-C1 empty vector, 3 μg wild-type, myc-tagged Rac-1, and 15 μg of (A and C) control plasmid (vector), a constitutively active mutant of PI-3K (p110*), or constitutively active mutants of one of 2 p21 Ras isoforms (G12 H-Ras and Q61 N-Ras), (B) control plasmid (vector), wild-type M-Ras (WT M-Ras), or a constitutively active mutant of M-Ras (Q71 M-Ras). At 16 hours after electroporation, cells were resuspended in serum-free medium for 2 hours. Ly294002 (25 μM dissolved in DMSO) was present for these 2 hours for N-RAS (A). Ly294002 (25 μM dissolved in ethanol) was present for the last 30 minutes for H-Ras (A), M-Ras (B), and H-Ras/p110* (C). After this, cells were lysed. Activated Rac-1 myc (Rac-1 myc GTP) was precipitated from lysates using GST-PAK bound to GT beads and detected by immunoblotting with a Rac-1 specific monoclonal antibody. The activation status of Akt and Erk was also determined from the same lysates using phospho-specific antibodies (p-Akt, p-Erk). Equivalency of loading was confirmed as described earlier (Akt, Erk).

Close modal

To determine whether increased PI-3K activity alone was sufficient for activation of Rac in cells of hemopoietic origin, we performed parallel experiments in which groups of cells were cotransfected with Rac-1 and a constitutively active mutant of the p110 catalytic subunit of PI-3K. These experiments demonstrated that overexpression of activated PI-3K in hemopoietic cells was sufficient for activation of coexpressed Rac-1 (Figure 6A).

Activation of Ras or Rac-1, but not PI-3K, is sufficient for activation of p38 MAPK in hemopoietic cells

To assess whether activation of Ras, Rac-1, or PI-3K was sufficient for activation of p38 MAPK in hemopoietic cells, constitutively active mutants were transiently overexpressed in 2 IL-3–dependent cell lines, Baf/3 and R6/X. In both cell lines, overexpression of G12V H-Ras, Q61K N-Ras, or V12L Rac-1 was sufficient for the induction of increased p38 MAPK phosphorylation (Figure7A-B and data not shown). However, overexpression of constitutively active PI-3K did not result in increased phosphorylation of p38 MAPK in either cell type (Figure 7A-B). Microscopic observation of levels of GFP expression in transfected cells ruled out the possibility that overexpression of the p110* construct was toxic (data not shown). Moreover, immunoblotting for phospho-Akt demonstrated not only that overexpression of p110* resulted in increased PI-3K activity, but that the levels induced were considerably higher than those observed in cells that were overexpressing G12V H-Ras and in which activation of p38 MAPK was induced. To investigate the role of PI-3K in the induction of p38 MAPK activation downstream of Ras, cells expressing Q61K N-Ras were treated with Ly294002. Treatment with Ly294002 did not inhibit activation of p38 MAPK induced by overexpression of activated Q61K N-Ras, instead inducing a reproducible increase in phosphorylation of p38 MAPK (data not shown).

Fig. 7.

Overexpression of activated H-Ras or Rac-1, but not p110 PI-3K, is sufficient for activation of p38 MAPK in hemopoietic cells.

(A) Baf/3 cells or (B) R6/X cells were electroporated with 5 μg pEGFP-C1 empty vector, and 30 μg (unless otherwise specified) control plasmid (vector), constitutively active p21 Ras (G12 H-Ras), constitutively active Rac-1 (V12 Rac-1), or constitutively active PI-3K (p110*). At 16 hours after electroporation, activation of p38 MAPK or PI-3K was measured using phospho-p38 MAPK (p-p38 MAPK) or phospho-Akt (p-Akt) specific antibodies, respectively. Equivalency of loading was confirmed using antibodies capable of recognizing both the phosphorylated and nonphosphorylated form of p38 MAPK (p38 MAPK), or Akt (Akt). The relative expression levels of constitutively active mutants of Ras and Rac-1 were estimated using a monoclonal antibody specific for an epitope tag from human c-myc (myc) common to both.

Fig. 7.

Overexpression of activated H-Ras or Rac-1, but not p110 PI-3K, is sufficient for activation of p38 MAPK in hemopoietic cells.

(A) Baf/3 cells or (B) R6/X cells were electroporated with 5 μg pEGFP-C1 empty vector, and 30 μg (unless otherwise specified) control plasmid (vector), constitutively active p21 Ras (G12 H-Ras), constitutively active Rac-1 (V12 Rac-1), or constitutively active PI-3K (p110*). At 16 hours after electroporation, activation of p38 MAPK or PI-3K was measured using phospho-p38 MAPK (p-p38 MAPK) or phospho-Akt (p-Akt) specific antibodies, respectively. Equivalency of loading was confirmed using antibodies capable of recognizing both the phosphorylated and nonphosphorylated form of p38 MAPK (p38 MAPK), or Akt (Akt). The relative expression levels of constitutively active mutants of Ras and Rac-1 were estimated using a monoclonal antibody specific for an epitope tag from human c-myc (myc) common to both.

Close modal

We show here using both primary cells and cell lines that endogenous Rac-1 is activated by stimulation of receptors of the hemopoietin family (the receptors for IL-3, GM-CSF, or IL-5), of the tyrosine kinase family (receptors for SLF and CSF-1), or of the receptor for antigen on BCRs. IL-3 induced activation of both Rac-2 and the related GTPase, Cdc42 (Figure 2). Although stimulation of endogenous receptors induced significant increases in levels of GTP-bound Rac-1 or Cdc42 relative to those in unstimulated cells (2.5- to 12-fold factors of induction; Figures 1-4), the absolute amount of activated Rac-1 or Cdc42 remained small (ranging from approximately 1% to 5% depending on the stimuli). These results are in broad agreement with reported levels of GTP-bound Rac-2 in neutrophils stimulated with fMLP (2.5% of total cellular Rac-2), or in porcine aortic endothelial cells stimulated with PDGF (4% of total cellular Rac-1).23,24 These results should be taken as minimal estimates of Rac activation, as in vivo levels of Rac-1 or 2 GTP will rapidly decrease due to the high intrinsic GTPase activity of Rac GTPases. Moreover, some activated Rac may be associated with a detergent insoluble fraction, for example, the cytoskeleton, meaning it will be unavailable for detection by this assay.

Our conclusion that IL-3 induced activation of Rac-1 and 2 was based on the use of antibodies, which we demonstrated were operationally specific for each Rac isoform (Figure 1A-B). The notion that IL-3 induces activation of Rac-1 is consistent with observations that bone marrow cells from mice that are homozygous null for Rac-2 as a result of gene targeting still retain the capacity to proliferate, survive, and differentiate to BMMCs in response to IL-3.20 The defects that are observed in these Rac-2–deficient cells reflect the differences in the specific functions of Rac-1 and Rac-2.20 Antibodies specific for Rac-3 were not available, so there is also the caveat that some of the Rac-GTP we detected may be Rac-3. A detailed analysis of this question will require the development of specific antibodies capable of differentiating between Rac-1, Rac-2, and Rac-3.

One likely component of the PI-3K–dependent mechanism mediating the activation of Rac-1 downstream of the BCR is the Rac GEF, Vav. Vav contains a PH domain, has enhanced GEF activity in the presence of lipid products of PI-3K,30 and is critical for activation of B cells via the BCR.52 Moreover, Vav becomes tyrosine phosphorylated (an event associated with enhanced GEF activity) in response to IL-3, SLF, or GM-CSF,53 and thus may be responsible for the PI-3K–dependent activation of Rac-1 induced by IL-3. In that the Tec family kinase, Btk, is required for a portion of the activation of Erk induced by ligation of the BCR54 and is activated by products of PI-3K activity,55 it is likely to be involved in the portion of Erk activation that we observed was dependent on PI-3K (Figure 5B). Our observations that overexpression of constitutively active PI-3K resulted in activation of Rac-1, but not of p38 MAPK (Figures 6A and 7), are consistent with the report that overexpression of constitutively active PI-3K was sufficient to induce membrane ruffling, but not activation of transcription factors known to require phosphorylation by p38 MAPK.56 Nevertheless, these observations with overexpressed proteins do not establish that physiologic increases in PI-3K activity are sufficient for activation of Rac-1. Indeed we were unable to observe activation of Rac-1 by IL-4 (B.G. and J.W.S., unpublished observations, April 2001), even though we have previously shown that IL-4 induces increases in PI-3K activity.57 Because IL-4 also fails to activate the Ras/MAPK pathways in lymphohemopoietic cells,38,39,51,58this is compatible with the notion that activation of Rac-1 requires both signals downstream of PI-3K and others downstream of Ras.

It was somewhat unexpected to find that a component of the activation of Rac-1 downstream of the IL-3 receptor and the BCR was not dependent on receptor-induced increases in PI-3K activity (Figure 5A). In this connection, however, it was of interest that expression of constitutively active mutants of H-Ras, N-Ras, or M-Ras resulted in activation of coexpressed Rac-1 through mechanism(s), which did not depend on increases in PI-3K activity (Figure 6). There is indirect evidence consistent with our observation. Thus, dominant active mutants of H-Ras induced membrane ruffling that probably reflected activation of Rac and was not inhibited by Ly294002 or wortmannin.59,60 In Rat-1 fibroblasts, both PI-3K–dependent and –independent pathways led from Ras to the Rac effector PAK.61 Certainly, our results indicate that in hemopoietic cells, activation of Rac-1 downstream of Ras occurs without simultaneous increases in overall PI-3K activity. It should be noted that our results do not exclude that small levels of PI-3K activity corresponding to that in resting, unstimulated cells might still have a permissive role in Rac-1 activation. Indeed this is consistent with our data in B lymphocytes, where high doses of Ly294002 that in these cells (but not mast cells) reduced levels of PI-3K activity to below background levels, blocked Rac-1 activation almost completely. The molecular mechanism for PI-3K–independent activation of Rac-1 is unclear. Only, one candidate, Rac GEF, lacks a PH domain. This protein smgGDS27,28 is structurally unrelated to the other Ras and Rho family GEFs and lacks DH or cdc25 catalytic domains. Nevertheless, smgGDS has GEF activity on a broad range of small GTPases, including Rac, Rap, Ral, and some members of the Ras family such as Ki-Ras 4B,62-64 and M-Ras (C. Korherr, M. Quadroni, and J.W.S., unpublished observations, July 1999). However, there is no evidence that smgGDS is downstream of Ras or is involved in any responses to extracellular signals. Our results raise the possibility that the activation of Rac-1 induced by IL-3, which occurs in the absence of increased PI-3K activity, may be mediated by activation of p21 Ras isoforms, M-Ras, or both. We and others reported that IL-3 and SLF activate Ras isoforms that bound to the monoclonal antibody Y13-259 and were hitherto thought to be p21-Ras alone,50,51 but are now known to include M-Ras.44 However, we have recently shown that IL-3 activates M-Ras and to a lesser extent p21 H-Ras (A. Schallhorn and J.W.S., unpublished observation, September 2001), making both candidate components of IL-3–mediated mechanisms for activation of Rac-1.

The question of whether some or all of the Rac activation induced by hemopoietic growth factors is downstream of Ras is difficult to address experimentally. Transient overexpression of a dominant-negative mutant of p21-Ras, S17N H-Ras, resulted in decreased activation of Rac-1 by IL-3 (B.G., unpublished observations, January 2001). However, dominant-negative Ras mutants such as S17N H-Ras will sequestrate Ras GEF, such as mSos-1 and Ras GRF-1 and -2, that are also direct activators of Rac.15-17 Therefore, we cannot exclude that S17N H-Ras might be exerting its inhibitory effect on activation of Rac-1, not through inhibiting activation of Ras, but by directly sequestrating a Rac GEF or targeting it for destruction.

Our observations on the lack of correlation between activation of Rac-1 and of p38 MAPK in hemopoietic cells contrast with evidence from experiments with other types of cells that suggest that activation of Rac was necessary and sufficient for activation of JNK and p38 MAPK.32-35 Our observations that the activation of Rac-1 resulting from stimulation of the receptors for hemopoietic growth factors or the BCR could be inhibited without corresponding proportional decreases in activation of p38 MAPK (Figure 5) suggested that activation of p38 MAPK was not entirely dependent on activation of Rac-1. Thus, whereas inhibition of PI-3K activity below background levels almost completely abolished the activation of Rac-1 induced by ligation of the BCR, the effect on p38 MAPK activation was much less (Figure 5B). Even greater differences in the effect of Ly294002 on activation of Rac-1 and p38 MAPK occurred in response to IL-3, where Ly294002 reduced activation of Rac-1 by approximately 70%, while having no effect on activation of p38 MAPK (Figure 5A). Similar results were reported in mast cells stimulated with SLF, where PI-3K inhibitors failed to block p38 MAPK activation,65 although not the activation of JNK.65,66 The latter suggests differences in the mechanisms of activation of JNK and p38 MAPK by SLF. This notion is also supported by our observation that overexpression of activated p21 Ras was sufficient to activate p38 MAPK in Baf/3 cells (Figure 7), whereas our published data and that of others show that overexpression of activated Ras was not sufficient to activate JNK.36,37 

Despite the lack of correlation between increases in activity of Rac-1 and p38 MAPK seen when inhibitors of PI-3K were used to reduce levels of activation of endogenous Rac-1, overexpression of constitutively active V12L Rac-1 was sufficient for activation of p38 MAPK (Figure 7). However, in that we and others have shown that the levels of activated Rac-1 induced by physiologic stimuli are relatively low (< 5% of cellular Rac), results of experiments involving overexpression of constitutively active Rac-1 (where cellular levels of activated Rac-1 exceed those observed with physiologic activation of endogenous Rac-1) may not be biologically relevant. Consistent with this notion, overexpression of the Rac GEF, Vav, which probably resulted in levels of activated endogenous Rac-1 that are closer to physiologic levels, failed to induce activation of p38 MAPK in RBL cells.67The notion that physiologically relevant levels of activated Rac-1 are alone, insufficient to activate p38 MAPK, is supported by our observation that overexpression of constitutively active PI-3K in 2 IL-3–dependent cell lines was insufficient for activation of p38 MAPK (Figure 7), although it activated coexpressed, exogenous Rac-1 (Figure6A). These results are consistent with the report that overexpression of constitutively active PI-3K was sufficient to induce membrane ruffling, but not activation of transcription factors known to require phosphorylation by p38 MAPK, including AP-1 and Elk-1.56 

The differential effect of inhibition of PI-3K activity on activation of Rac-1 and p38 MAPK by IL-3 might be explained in several ways. First, p38 MAPK activation might not depend on activation of Rac-1 at all. In that we have shown that IL-3 activates Cdc42 (Figure 2C) and the activation of Cdc42 induced by PDGF occurs independent of PI-3K catalytic activity,68 it is possible that Cdc42 is responsible for the PI-3K–independent activation of p38 MAPK. However, we failed to observe consistent activation of p38 MAPK in cells of hemopoietic origin (Baf/3 or R6X) in which constitutively activated Cdc42 had been overexpressed (data not shown). Alternatively, IL-3 might activate 2 pools of Rac-1 in hemopoietic cells, the minor one of which is activated via a PI-3K–independent mechanism and is responsible for activation of p38 MAPK. Finally, it is possible that the 30% of IL-3–induced Rac-1 activation that was independent of increases in PI-3K activity may synergize with another signal to activate p38 MAPK. Our observations that overexpression of activated Ras (H-Ras, N-Ras, or M-Ras) induced activation of Rac-1 and p38 MAPK (Figures 6 and 7), the latter through mechanisms that could only involve physiologic levels of endogenous Rac-1, raise the possibility that this second pathway may involve Ras. Activation of p38 MAPK induced by granulocyte-colony-stimulating factor (G-CSF) was dependent on activation of Ras.40 Certainly our data show that increased PI-3K activity alone, even at levels greater than those associated with p38 MAPK activation in cells overexpressing constitutively active p21 Ras (Figure 7), was insufficient for activation of p38 MAPK. Finally, activation of p38 MAPK by IL-3 may involve Rac-independent mechanism(s), which as discussed above, appear to be involved in activation of p38 MAPK downstream of cross-linking of the BCR as well.

The activation of Rac by hemopoietic growth factors is likely to be involved in many of their actions in the regulation of growth, survival, differentiation, and function, all of which are influenced by the absence of Rac-2.19,20 One critical function of the Rac pathway in growth may involve stathmin, first recognized as a protein overexpressed in leukemic blast cells and now known to be a key regulator of microtubule assembly.69 The Rac and Cdc42 pathways activate p65 PAK (PAK-1), which phosphorylates stathmin on serine 16, inactivating it so that it releases tubulin.70We identified stathmin as one of the proteins undergoing serine/threonine phosphorylation after stimulation with IL-3 and showed that one of the sites phosphorylated was serine 16 (M. Quadroni and J.W.S., unpublished observation, June 1997). Our demonstration that IL-3 induces activation of Rac-1, Rac-2, and Cdc42 accounts for the IL-3–induced activation of PAK-171 and phosphorylation of stathmin on serine 16, an essential prelude to cell division. Overexpression of activated Rac promotes survival of hemopoietic cells72 and hemopoietic cells from Rac-2–deficient mice exhibited decreased expression of the antiapoptotic protein, Bcl-XL, and increased expression of the proapoptotic molecule, BAD.20 Hemopoietic growth factors such as GM-CSF, SLF, and CSF-1 are capable of inducing cellular migration and chemotaxis, which in the case of CSF-1 has been shown to be Rac dependent.10,73 Finally, our demonstration that IL-3 and CSF-1 induce activation of Rac provides a molecular basis for observations that these cytokines induce membrane ruffling.73,74 Our findings that hemopoietic growth factors activate Rac-1, Rac-2, and Cdc42 confirms the central role of these GTPases in growth factor–mediated signaling. Further, it raises new questions about the existence of novel mechanisms of activating these GTPases, which do not depend on increases in PI-3K activity and potentially involve novel effectors of the Ras family.

We thank the Canadian Institutes of Health Research (CIHR) for supporting this work.

Prepublished online as Blood First Edition Paper, July 12, 2002; DOI 10.1182/blood-2002-01-0154.

Supported by a grant from the Canadian Institutes of Health Research (CIHR), Canada. B.G. was supported by a National Sciences and Engineering Research Council (NSERC) scholarship and a CIHR scholarship.

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
Didsbury
J
Weber
RF
Bokoch
GM
Evans
T
Snyderman
R
rac, a novel ras-related family of proteins that are botulinum toxin substrates.
J Biol Chem.
264
1989
16378
16382
2
Johnson
DI
Cdc42: An essential Rho-type GTPase controlling eukaryotic cell polarity.
Microbiol Mol Biol Rev.
63
1999
54
105
3
Haataja
L
Groffen
J
Heisterkamp
N
Characterization of RAC3, a novel member of the Rho family.
J Biol Chem.
272
1997
20384
20388
4
Kjoller
L
Hall
A
Signaling to Rho GTPases.
Exp Cell Res.
253
1999
166
179
5
Mackay
DJ
Hall
A
Rho GTPases.
J Biol Chem.
273
1998
20685
20688
6
Bar-Sagi
D
Hall
A
Ras and Rho GTPases: a family reunion.
Cell.
103
2000
227
238
7
Caron
E
Hall
A
Identification of two distinct mechanisms of phagocytosis controlled by different Rho GTPases.
Science.
282
1998
1717
1721
8
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
9
Massol
P
Montcourrier
P
Guillemot
JC
Chavrier
P
Fc receptor-mediated phagocytosis requires CDC42 and Rac1.
EMBO J.
17
1998
6219
6229
10
Allen
WE
Zicha
D
Ridley
AJ
Jones
GE
A role for Cdc42 in macrophage chemotaxis.
J Cell Biol.
141
1998
1147
1157
11
Knaus
UG
Heyworth
PG
Evans
T
Curnutte
JT
Bokoch
GM
Regulation of phagocyte oxygen radical production by the GTP-binding protein Rac 2.
Science.
254
1991
1512
1515
12
Abo
A
Pick
E
Hall
A
Totty
N
Teahan
CG
Segal
AW
Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1.
Nature.
353
1991
668
670
13
Billadeau
DD
Brumbaugh
KM
Dick
CJ
Schoon
RA
Bustelo
XR
Leibson
PJ
The Vav-Rac1 pathway in cytotoxic lymphocytes regulates the generation of cell-mediated killing.
J Exp Med.
188
1998
549
559
14
Li
B
Yu
H
Zheng
W
et al
Role of the guanosine triphosphatase Rac2 in T helper 1 cell differentiation.
Science.
288
2000
2219
2222
15
Quilliam
LA
Kato
K
Rabun
KM
et al
Identification of residues critical for Ras(17N) growth-inhibitory phenotype and for Ras interaction with guanine nucleotide exchange factors.
Mol Cell Biol.
14
1994
1113
1121
16
Nimnual
AS
Yatsula
BA
Bar-Sagi
D
Coupling of Ras and Rac guanosine triphosphatases through the Ras exchanger Sos.
Science.
279
1998
560
563
17
de Hoog
CL
Fan
WT
Goldstein
MD
Moran
MF
Koch
CA
Calmodulin-independent coordination of Ras and extracellular signal-regulated kinase activation by Ras-GRF2.
Mol Cell Biol.
20
2000
2727
2733
18
Scita
G
Tenca
P
Frittoli
E
et al
Signaling from Ras to Rac and beyond: not just a matter of GEFs.
EMBO J.
19
2000
2393
2398
19
Roberts
AW
Kim
C
Zhen
L
et al
Deficiency of the hematopoietic cell-specific Rho family GTPase Rac2 is characterized by abnormalities in neutrophil function and host defense.
Immunity.
10
1999
183
196
20
Yang
FC
Kapur
R
King
AJ
et al
Rac2 stimulates Akt activation affecting BAD/Bcl-XL expression while mediating survival and actin function in primary mast cells.
Immunity.
12
2000
557
568
21
Yang
FC
Atkinson
SJ
Gu
Y
et al
Rac and Cdc42 GTPases control hematopoietic stem cell shape, adhesion, migration, and mobilization.
Proc Natl Acad Sci U S A.
98
2001
5614
5618
22
Geijsen
N
van Delft
S
Raaijmakers
JA
et al
Regulation of p21rac activation in human neutrophils.
Blood.
94
1999
1121
1130
23
Hawkins
PT
Eguinoa
A
Qiu
RG
et al
PDGF stimulates an increase in GTP-Rac via activation of phosphoinositide 3-kinase.
Curr Biol.
5
1995
393
403
24
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
25
Akasaki
T
Koga
H
Sumimoto
H
Phosphoinositide 3-kinase-dependent and -independent activation of the small GTPase Rac2 in human neutrophils.
J Biol Chem.
274
1999
18055
18059
26
Salojin
KV
Zhang
J
Delovitch
TL
TCR and CD28 are coupled via ZAP-70 to the activation of the Vav/Rac-1-/PAK-1/p38 MAPK signaling pathway.
J Immunol.
163
1999
844
853
27
Chuang
TH
Xu
X
Quilliam
LA
Bokoch
GM
SmgGDS stabilizes nucleotide-bound and -free forms of the Rac1 GTP-binding protein and stimulates GTP/GDP exchange through a substituted enzyme mechanism.
Biochem J.
303
1994
761
767
28
Xu
X
Wang
Y
Barry
DC
Chanock
SJ
Bokoch
GM
Guanine nucleotide binding properties of Rac2 mutant proteins and analysis of the responsiveness to guanine nucleotide dissociation stimulator.
Biochemistry.
36
1997
626
632
29
Quilliam
LA
Khosravi-Far
R
Huff
SY
Der
CJ
Guanine nucleotide exchange factors: activators of the Ras superfamily of proteins.
Bioessays.
17
1995
395
404
30
Han
J
Luby-Phelps
K
Das
B
et al
Role of substrates and products of PI 3-kinase in regulating activation of Rac-related guanosine triphosphatases by Vav.
Science.
279
1998
558
560
31
Kotani
K
Yonezawa
K
Hara
K
et al
Involvement of phosphoinositide 3-kinase in insulin- or IGF-1-induced membrane ruffling.
EMBO J.
13
1994
2313
2321
32
Coso
OA
Chiariello
M
Yu
JC
et al
The small GTP-binding proteins Rac1 and Cdc42 regulate the activity of the JNK/SAPK signaling pathway.
Cell.
81
1995
1137
1146
33
Minden
A
Lin
A
Claret
FX
Abo
A
Karin
M
Selective activation of the JNK signaling cascade and c-Jun transcriptional activity by the small GTPases Rac and Cdc42Hs.
Cell.
81
1995
1147
1157
34
Zhang
S
Han
J
Sells
MA
et al
Rho family GTPases regulate p38 mitogen-activated protein kinase through the downstream mediator Pak1.
J Biol Chem.
270
1995
23934
23936
35
Bagrodia
S
Derijard
B
Davis
RJ
Cerione
RA
Cdc42 and PAK-mediated signaling leads to Jun kinase and p38 mitogen-activated protein kinase activation.
J Biol Chem.
270
1995
27995
27998
36
Rausch
O
Marshall
CJ
Tyrosine 763 of the murine granulocyte colony-stimulating factor receptor mediates Ras-dependent activation of the JNK/SAPK mitogen-activated protein kinase pathway.
Mol Cell Biol.
17
1997
1170
1179
37
Foltz
IN
Gerl
RE
Wieler
JS
Luckach
M
Salmon
RA
Schrader
JW
Human mitogen-activated protein kinase kinase 7 (MKK7) is a highly conserved c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) activated by environmental stresses and physiological stimuli.
J Biol Chem.
273
1998
9344
9351
38
Foltz
IN
Lee
JC
Young
PR
Schrader
JW
Hemopoietic growth factors with the exception of interleukin-4 activate the p38 mitogen-activated protein kinase pathway.
J Biol Chem.
272
1997
3296
3301
39
Foltz
IN
Schrader
JW
Activation of the stress-activated protein kinases by multiple hematopoietic growth factors with the exception of interleukin-4.
Blood.
89
1997
3092
3096
40
Rausch
O
Marshall
CJ
Cooperation of p38 and extracellular signal-regulated kinase mitogen- activated protein kinase pathways during granulocyte colony-stimulating factor-induced hemopoietic cell proliferation.
J Biol Chem.
274
1999
4096
4105
41
Cano
E
Mahadevan
LC
Parallel signal processing among mammalian MAPKs.
Trends Biochem Sci.
20
1995
117
122
42
Doza
YN
Cuenda
A
Thomas
GM
Cohen
P
Nebreda
AR
Activation of the MAP kinase homologue RK requires the phosphorylation of Thr-180 and Tyr-182 and both residues are phosphorylated in chemically stressed KB cells.
FEBS Lett.
364
1995
223
228
43
Enslen
H
Raingeaud
J
Davis
RJ
Selective activation of p38 mitogen-activated protein (MAP) kinase isoforms by the MAP kinase kinases MKK3 and MKK6.
J Biol Chem.
273
1998
1741
1748
44
Ehrhardt
GR
Leslie
KB
Lee
F
Wieler
JS
Schrader
JW
M-Ras, a widely expressed 29-kD homologue of p21 Ras: expression of a constitutively active mutant results in factor-independent growth of an interleukin-3-dependent cell line.
Blood.
94
1999
2433
2444
45
Schrader
JW
Crapper
RM
Autogenous production of a hemopoietic growth factor, persisting-cell-stimulating factor, as a mechanism for transformation of bone marrow-derived cells.
Proc Natl Acad Sci U S A.
80
1983
6892
6896
46
Manser
E
Leung
T
Salihuddin
H
Zhao
ZS
Lim
L
A brain serine/threonine protein kinase activated by Cdc42 and Rac1.
Nature.
367
1994
40
46
47
Morreale
A
Venkatesan
M
Mott
HR
et al
Structure of Cdc42 bound to the GTPase binding domain of PAK.
Nat Struct Biol.
7
2000
384
388
48
Shirsat
NV
Pignolo
RJ
Kreider
BL
Rovera
G
A member of the ras gene superfamily is expressed specifically in T, B and myeloid hemopoietic cells.
Oncogene.
5
1990
769
772
49
Manser
E
Loo
TH
Koh
CG
et al
PAK kinases are directly coupled to the PIX family of nucleotide exchange factors.
Mol Cell.
1
1998
183
192
50
Satoh
T
Nakafuku
M
Miyajima
A
Kaziro
Y
Involvement of ras p21 protein in signal-transduction pathways from interleukin 2, interleukin 3, and granulocyte/macrophage colony-stimulating factor, but not from interleukin 4.
Proc Natl Acad Sci U S A.
88
1991
3314
3318
51
Duronio
V
Welham
MJ
Abraham
S
Dryden
P
Schrader
JW
p21ras activation via hemopoietin receptors and c-kit requires tyrosine kinase activity but not tyrosine phosphorylation of p21ras GTPase- activating protein.
Proc Natl Acad Sci U S A.
89
1992
1587
1591
52
Bachmann
MF
Nitschke
L
Krawczyk
C
et al
The guanine-nucleotide exchange factor Vav is a crucial regulator of B cell receptor activation and B cell responses to nonrepetitive antigens.
J Immunol.
163
1999
137
142
53
Alai
M
Mui
AL
Cutler
RL
Bustelo
XR
Barbacid
M
Krystal
G
Steel factor stimulates the tyrosine phosphorylation of the proto-oncogene product, p95vav, in human hemopoietic cells.
J Biol Chem.
267
1992
18021
18025
54
Jiang
A
Craxton
A
Kurosaki
T
Clark
EA
Different protein tyrosine kinases are required for B cell antigen receptor-mediated activation of extracellular signal-regulated kinase, c-Jun NH2-terminal kinase 1, and p38 mitogen-activated protein kinase.
J Exp Med.
188
1998
1297
1306
55
Scharenberg
AM
El-Hillal
O
Fruman
DA
et al
Phosphatidylinositol-3,4,5-trisphosphate (PtdIns-3,4,5-P3)/Tec kinase-dependent calcium signaling pathway: a target for SHIP-mediated inhibitory signals.
EMBO J.
17
1998
1961
1972
56
Reif
K
Nobes
CD
Thomas
G
Hall
A
Cantrell
DA
Phosphatidylinositol 3-kinase signals activate a selective subset of Rac/Rho-dependent effector pathways.
Curr Biol.
6
1996
1445
1455
57
Gold
MR
Duronio
V
Saxena
SP
Schrader
JW
Aebersold
R
Multiple cytokines activate phosphatidylinositol 3-kinase in hemopoietic cells. Association of the enzyme with various tyrosine- phosphorylated proteins.
J Biol Chem.
269
1994
5403
5412
58
Welham
MJ
Duronio
V
Sanghera
JS
Pelech
SL
Schrader
JW
Multiple hemopoietic growth factors stimulate activation of mitogen-activated protein kinase family members.
J Immunol.
149
1992
1683
1693
59
Nobes
CD
Hawkins
P
Stephens
L
Hall
A
Activation of the small GTP-binding proteins rho and rac by growth factor receptors.
J Cell Sci.
108
1995
225
233
60
Rodriguez-Viciana
P
Warne
PH
Khwaja
A
et al
Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras.
Cell.
89
1997
457
467
61
Tang
Y
Yu
J
Field
J
Signals from the Ras, Rac, and Rho GTPases converge on the Pak protein kinase in Rat-1 fibroblasts.
Mol Cell Biol.
19
1999
1881
1891
62
Kaibuchi
K
Mizuno
T
Fujioka
H
et al
Molecular cloning of the cDNA for stimulatory GDP/GTP exchange protein for smg p21s (ras p21-like small GTP-binding proteins) and characterization of stimulatory GDP/GTP exchange protein.
Mol Cell Biol.
11
1991
2873
2880
63
Hiraoka
K
Kaibuchi
K
Ando
S
et al
Both stimulatory and inhibitory GDP/GTP exchange proteins, smg GDS and rho GDI, are active on multiple small GTP-binding proteins.
Biochem Biophys Res Commun.
182
1992
921
930
64
Strassheim
D
Porter
RA
Phelps
SH
Williams
CL
Unique in vivo associations with SmgGDS and RhoGDI and different guanine nucleotide exchange activities exhibited by RhoA, dominant negative RhoA(Asn-19), and activated RhoA(Val-14).
J Biol Chem.
275
2000
6699
6702
65
Ishizuka
T
Chayama
K
Takeda
K
et al
Mitogen-activated protein kinase activation through Fc epsilon receptor I and stem cell factor receptor is differentially regulated by phosphatidylinositol 3-kinase and calcineurin in mouse bone marrow- derived mast cells.
J Immunol.
162
1999
2087
2094
66
Timokhina
I
Kissel
H
Stella
G
Besmer
P
Kit signaling through PI 3-kinase and Src kinase pathways: an essential role for Rac1 and JNK activation in mast cell proliferation.
EMBO J.
17
1998
6250
6262
67
Song
JS
Haleem-Smith
H
Arudchandran
R
et al
Tyrosine phosphorylation of Vav stimulates IL-6 production in mast cells by a Rac/c-Jun N-terminal kinase-dependent pathway.
J Immunol.
163
1999
802
810
68
Jimenez
C
Portela
RA
Mellado
M
et al
Role of the PI3K regulatory subunit in the control of actin organization and cell migration.
J Cell Biol.
151
2000
249
262
69
Larsson
N
Marklund
U
Gradin
HM
Brattsand
G
Gullberg
M
Control of microtubule dynamics by oncoprotein 18: dissection of the regulatory role of multisite phosphorylation during mitosis.
Mol Cell Biol.
17
1997
5530
5539
70
Daub
H
Gevaert
K
Vandekerckhove
J
Sobel
A
Hall
A
Rac/Cdc42 and p65PAK regulate the microtubule-destabilizing protein stathmin through phosphorylation at serine 16.
J Biol Chem.
276
2001
1677
1680
71
Schurmann
A
Mooney
AF
Sanders
LC
et al
p21-activated kinase 1 phosphorylates the death agonist bad and protects cells from apoptosis.
Mol Cell Biol.
20
2000
453
461
72
Nishida
K
Kaziro
Y
Satoh
T
Anti-apoptotic function of Rac in hematopoietic cells.
Oncogene.
18
1999
407
415
73
Peppelenbosch
M
Boone
E
Jones
GE
et al
Multiple signal transduction pathways regulate TNF-induced actin reorganization in macrophages: inhibition of Cdc42-mediated filopodium formation by TNF.
J Immunol.
162
1999
837
845
74
Romanova
LY
Alexandrov
IA
Blagosklonny
MV
et al
Regulation of actin cytoskeleton in lymphocytes: PKC-delta disrupts IL- 3-induced membrane ruffles downstream of Rac1.
J Cell Physiol.
179
1999
157
169

Author notes

John W. Schrader, The Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada; e-mail:john@brc.ubc.ca.

Sign in via your Institution