Stromal cell–derived factor (SDF)-1α and its receptor, CXCR4, play an important role in cell migration, embryonic development, and human immunodeficiency virus infection. However, the cellular signaling pathways that mediate these processes are not fully elucidated. We and others have shown that the binding of SDF-1α to CXCR4 activates phosphatidylinositol-3 kinase (PI-3 kinase), p44/42 mitogen-associated protein kinase, and the transcription factor nuclear factor–κB, and it also enhances the tyrosine phosphorylation and association of proteins involved in the formation of focal adhesions. In this study, we examined the role of phosphatases in CXCR4-mediated signaling pathways. We observed significant inhibition of SDF-1α–induced migration by phosphatase inhibitors in CXCR4-transfected pre-B lymphoma L1.2 cells, Jurkat T cells, and peripheral blood lymphocytes. Further studies revealed that SDF-1α stimulation induced robust tyrosine phosphorylation in the SH2-containing phosphatase SHP2. SHP2 associated with the CXCR4 receptor and the signaling molecules SHIP, cbl, and fyn. Overexpression of wild-type SHP2 increased SDF-1α–induced chemotaxis. Enhanced activation of fyn and lyn kinases and the tyrosine phosphorylation of cbl were also observed. In addition, SDF-1α stimulation enhanced the association of cbl with PI-3 kinase, Crk-L, and 14-3-3β proteins. Our results suggest that CXCR4-mediated signaling is regulated by SHP2 and cbl, which collectively participate in the formation of a multimeric signaling complex.

Chemokines and their receptors play important roles in immune and inflammatory responses through the regulation of cell migration and growth. In addition, chemokines participate in the pathogenesis of several diseases.1-5 Chemokine receptors act as coreceptors for the human immunodeficiency virus (HIV), and the expression of both chemokines and chemokine receptor homologues by Kaposi's sarcoma herpes virus (KSHV/HHV-8) has been implicated in the development of Kaposi's sarcoma.6 7 

The α-chemokine, stromal cell–derived factor (SDF), is a widely expressed ligand of the CXCR4 receptor.8-11 It is a potent chemotactic factor for mature leukocytes such as monocytes and T lymphocytes as well as for CD34+progenitors.12-14 CXCR4 is a 7-transmembrane G protein–coupled receptor that is expressed by a variety of cells, including peripheral blood lymphocytes (PBLs), monocytes, thymocytes, pre-B cells, dendritic cells, and endothelial cells.15-17CXCR4 serves as a coreceptor for T-cell tropic HIV-1 strains and plays an important role in HIV pathogenesis.1,17,18 In addition, CXCR4 and SDF-1α are critical for embryonic development. Targeted disruption of either protein leads to severe defects in mice embryos that are lethal.19-22 Although CXCR4 and/or SDF-1α play important roles in both physiologic and pathologic processes, the cellular signaling pathways that mediate these effects are not fully elucidated.

Previously, we and others have shown that the association of SDF-1α with CXCR4 activates multiple signaling pathways.23-25 The tyrosine phosphorylation and association of proteins such as RAFTK, Crk, and paxillin, which are involved in the formation of focal adhesions, were enhanced upon SDF-1α stimulation.23,24Phosphatidylinositol-3 kinase (PI-3 kinase), which is an essential component of signaling pathways that induce chemotaxis, was also activated.23,26 In addition, selective activation of the downstream signaling molecule, p44/42 mitogen-activated protein kinase (MAPK), but not p38 MAPK or Jun N-terminal kinase, was observed as well as the activation of the transcription factor, nuclear factor (NF)-κB.23 

In the present study, we have characterized the role of the tyrosine phosphatase SHP2 and adaptor molecule cbl in CXCR4-mediated signaling pathways. Protein tyrosine phosphatases (PTPs) play an important role in the regulation of signals generated by various stimuli.27-30 However, the role of individual PTPs in signaling is not uniform. SHP1, a PTP expressed predominantly in hematopoietic cells, and SHIP (SH2-containing inositol phosphatase) act as negative regulators of signaling.29-31 In contrast, the ubiquitously expressed PTP, SHP2, appears to play a positive role in growth factor–induced signaling pathways.32-35 In addition to their tyrosine phosphatase activity, SHP1 and SHP2 can function as adaptor molecules. Through their SH2 domains, SHP1 and SHP2 can bind to several proteins and then transduce signals.36-39 

Both SHP1 and SHP2 play important roles in immune regulation and development.40-42 However, their roles in chemokine-mediated signaling are not fully understood. SHP1 was recently shown to mediate SDF-1α–induced chemotaxis. Alterations in chemotactic response to SDF-1α were observed in hematopoietic cells derived from mice lacking SHP1 (moth-eaten or viable).43 

Cbl is a 120-kd protein present in lymphocytes that can function as an adaptor molecule in tyrosine phosphorylation–dependent signaling. It is rapidly phosphorylated by Src-like kinases upon stimulation of cell surface receptors and binds to several other signaling molecules, including ZAP-70, Syk, PI-3 kinase, Crk-L, and Vav.44-48Cbl also contains a ubiquitin-associated domain. It binds to and stimulates the ubiquitin-mediated degradation of active platelet-derived growth factor, epidermal growth factor, and colony-stimulating factor-1 receptors.49-51 In this way, cbl can act as a negative regulator of receptor and nonreceptor tyrosine kinases.

In this study, we demonstrate that SDF-1α treatment of CXCR4-positive cells resulted in the tyrosine phosphorylation of SHP2 and cbl and the activation of fyn kinase. Furthermore, we show that SHP2 is constitutively associated with the CXCR4 receptor and is recruited into the protein tyrosine kinase machinery by forming a complex with SHIP, cbl, and fyn. Cbl was also shown to form an activation-induced multimeric complex with PI-3 kinase, 14-3-3β, and Crk-L. Moreover, pretreatment of cells with the phosphatase inhibitors phenylarsine oxide (PAO) and sodium orthovanadate reduced the SDF-1α–induced migration of T and B cells. These results suggest that SHP2 and cbl are key mediators of SDF-1α–induced functional responses.

Reagents and materials

Antibodies to SHP2, cbl, fyn, lyn, 14-3-3β, and Crk-L were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). Monoclonal antiphosphotyrosine antibody (4G10) was a generous gift from Dr Brian Druker (Oregon Health Sciences University, Portland, OR). Antibody to PI-3 kinase was from Upstate Biotechnology (Lake Placid, NY). The GST-SH2 domain of SHP2 was obtained from Santa Cruz Biotechnology. Electrophoresis reagents and the nitrocellulose membrane were obtained from Bio-Rad Laboratories (Hercules, CA). The phosphatase inhibitors PAO and sodium orthovanadate and the protease inhibitors leupeptin and α 1-antitrypsin and all other reagents were acquired from Sigma Chemical Co (St Louis, MO).

Construction of stable CXCR4 transfectants

CXCR4 complementary DNA, tagged at the amino-terminus with a Flag epitope (Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys), was subcloned into the pcDNAIII expression vector and then stably transfected into cells from the murine pre-B lymphoma cell line, L1.2, as described.52-54 Geneticin (G418)–containing medium was used to select for transfectants. Cell-surface expression of CXCR4 on the L1.2 transfectants was confirmed by flow cytometry.

Isolation of PBLs

PBLs were generated from peripheral blood mononuclear cells as described.55 56 Briefly, peripheral blood mononuclear cells were separated by Ficoll-Hypaque gradient centrifugation. After 2 rounds of adherence to plastic in RPMI 1640 with 10% fetal calf serum, 2 mM glutamine, 50 μg/mL penicillin, and 50 μg/mL streptomycin, the cells were grown in medium containing 5 μg/mL phytohemagglutinin for 3 days. The PBLs were then washed and grown in RPMI 1640 containing 15% fetal calf serum and 5% interleukin-2 (Advanced Biotechnologies, Columbia, MD). After 3 weeks, the activated T cells were found to be 50% to 60% positive for CXCR4 by flow cytometry and were then used for further studies.

Cell culture

The CXCR4 L1.2 cells were grown in RPMI 1640 with 10% fetal bovine serum, 2 mM glutamine, 1 mM sodium pyruvate, 50 μg/mL penicillin, 50 μg/mL streptomycin, 55 μM 2-mercaptoethanol, and 0.8 mg/mL G418 (Gibco BRL, Grand Island, NY). Jurkat T cells were grown in RPMI 1640 with 10% fetal calf serum, 2 mM glutamine, 50 μg/mL penicillin, and 50 μg/mL streptomycin.

Flow cytometry

Cells were washed twice with phosphate-buffered saline (PBS), resuspended in 500 μL of wash buffer (PBS containing 5% fetal calf serum) and 12G5 anti-CXCR4 antibody, and then incubated at 4°C for 30 minutes. The cells were next washed 3 times with wash buffer, resuspended in 500 μL of wash buffer and fluorescein isothiocyanate–coupled secondary antibody, and then incubated at 4°C for 30 minutes. Thereafter, the cells were washed 3 times, resuspended in 500 μL PBS, and analyzed by flow cytometry.

Stimulation of cells

Cells were washed twice with RPMI 1640 and serum-starved for 2 hours at 37°C in 1 × Hank's buffered salt solution at a concentration of 10 × 106 cells/mL. The cells were then treated with 100 ng/mL SDF-1α at 37°C. After different time periods, cells were lysed in modified radioimmunoprecipitation assay (RIPA) buffer (50 mM Tris-HCl, pH 7.4; 1% Nonidet P-40; 150 mM NaCl; 1 mM phenylmethylsulfonyl fluoride; 10 μg/mL each of aprotinin, leupeptin, and pepstatin; and 10 mM each of sodium vanadate, sodium fluoride, and sodium pyrophosphate). Total cell lysates were clarified by centrifugation at 10 000g for 10 minutes. Protein concentrations were determined by protein assay (Bio-Rad Laboratories).

Immunoprecipitation and immunoblotting

Equal amounts of protein from each sample were clarified by incubation with protein A–Sepharose CL-4B (Amersham Pharmacia Biotech, Piscataway, NJ) for 1 hour at 4°C. After the removal of protein A–Sepharose by brief centrifugation, the samples were incubated with a primary antibody for 2 hours or overnight at 4°C. The antibody-antigen complexes were immunoprecipitated by incubation with 50 μL protein A–Sepharose for 3 hours or overnight at 4°C. Nonspecific bound proteins were removed by washing the protein A–Sepharose beads 3 times with modified RIPA buffer and one time with PBS. Bound immunocomplexes were solubilized in 30 μL 2 × Laemmli buffer, separated on 8% or 12% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), and transferred to nitrocellulose membranes. The membranes were blocked in 5% nonfat milk protein for 1 hour and probed with primary antibody for 2 hours at room temperature or overnight at 4°C. Immunoreactive bands were visualized using horseradish peroxidase–conjugated secondary antibody and the enhanced chemiluminescent system (Amersham Pharmacia Biotech). The immunoprecipitation and Western blotting data shown in “Results” are representative of findings from 3 independent experiments.

Glutathione-S-transferase–fusion protein binding studies

SH-PTP2 glutathione-S-transferase (GST)–fusion proteins were purchased from Santa Cruz Biotechnology. Fifty micrograms of GST-fusion protein were added to 1 mg cell lysate and incubated for 1 hour at 4°C. GST protein (Santa Cruz Biotechnology) was used as a control. The complexes were then preabsorbed with 50 μL Glutathione-Sepharose 4B beads (Amersham Pharmacia Biotech) and incubated for 3 hours or overnight at 4°C. The beads were centrifuged and washed 3 times with modified RIPA buffer and once with 1 × PBS. Bound proteins were eluted by boiling in Laemmli sample buffer and separated on 8% SDS-PAGE.

Migration assays

Cells were resuspended at 6.6 × 106/mL in RPMI 1640 medium containing 2.5% fetal calf serum. Twenty-four–well plates containing 5 μm porosity inserts (CoStar Corp, Kennebunk, ME) were used for the assays. A total of 25 ng/mL SDF-1α in 600 μL of medium was added to the bottom wells, and 150 μL of cells, untreated or treated with the phosphatase inhibitor PAO or sodium orthovanadate, was placed in the inserts. Controls were treated with appropriate solvents and SDF-1α under similar conditions. After 3 hours, cells that migrated to the bottom wells were collected and counted on a hemacytometer.

Fyn, lyn, and p44/42 MAPK assays

Cell lysates were immunoprecipitated with fyn, lyn, or Erk1 and Erk2 antibodies. For fyn and lyn kinase assays, the immune complexes were washed twice with RIPA buffer and twice with kinase buffer (20 mM HEPES, 50 mM NaCl, 10 μM Na3VO4, 5 mM MgCl2, and 5 mM MnCl2). The complexes were then incubated in kinase buffer containing 0.18 MBq (5 μCi) γ-32P-adenosine triphosphate (ATP) for 20 minutes at 30°C. The reaction was terminated by adding 4 × Laemmli sample buffer and boiling the samples for 5 minutes. Proteins were then separated on 8% SDS-PAGE and viewed by autoradiography. For p44/42 MAPK assays, the immune complexes were washed twice with RIPA buffer and twice with kinase buffer (50 mM HEPES, pH 7.4; 10 mM MgCl2; and 20 μM ATP). The complexes were then incubated in kinase buffer containing 7 μg of myelin basic protein (Upstate Biotechnology) and 0.18 MBq (5 μCi) of γ-32P-ATP for 30 minutes. Proteins were separated on 15% SDS-PAGE and viewed by autoradiography.

Transient transfection

Wild-type SHP2 (SHP2 WT) construct and control vector (kindly provided by Dr Benjamin G. Neel, Beth Israel Deaconess Medical Center) were transiently transfected into the JMC.T5 Jurkat cell line (kindly provided by Dr Hamid Band, Harvard Medical School) by the electroporation method as described.55 Briefly, 10 μg of plasmid carrying the control vector or SHP2 WT34 35 was added to the cell suspension (20 × 106 cells/mL) in a Gene Pulser cuvette and then incubated on ice for 10 minutes. Electric pulse was given by using the Gene Pulser II (Bio-Rad) set at 250 V, 950 μF. The cells were transferred to RPMI 1640 medium containing 10% fetal bovine serum and then grown for 72 hours. The expression of SHP2 was examined by immunoprecipitation as described above.

Phosphatase inhibitors reduce SDF-1α– induced migration of CXCR4 L1.2, Jurkat T cells, and PBLs

SDF-1α has been shown to act as a potent chemoattractant for various cell types.8,9,12 We and others have recently shown that SDF-1α–induced chemotaxis is regulated by PI-3 kinase.23,26 Because several signaling components are required for mediating chemotaxis, we analyzed the effect of the phosphatase inhibitors PAO and sodium orthovanadate on SDF-1α–induced migration of CXCR4 transfectants, Jurkat T cells, and PBLs. PAO and sodium orthovanadate inhibit the catalytic activity of PTPs, which modulate growth factor–mediated chemotaxis.57As shown in Figure 1A,B, PAO led to a dose-dependent decrease in SDF-1α–induced chemotaxis in CXCR4 L1.2 and Jurkat cells as compared with cells treated with solvent controls. Similarly, sodium orthovanadate treatment reduced the migration of CXCR4 L1.2, Jurkat T cells, and PBLs in a dose-dependent manner (Figure2A-C). At the highest concentration (1.0 μM), PAO reduced the migration of the CXCR4 transfectants to less than 5% (P ≤ .001) and the Jurkat cells to less than 10% (P ≤ .002) (Figure 1A,B). At 100 μM of sodium orthovanadate, the migration of the CXCR4 transfectants was reduced to 58% (P ≤ .034), the Jurkat T cells to 42% (P ≤ .002), and the PBLs to 39% (P ≤ .009) (Figure 2A-C). Under similar conditions, the inhibitors had no effect on the viability of the cells (data not shown). In addition, the phosphatase inhibitors had no effect on SDF-1α–induced p44/42 MAPK activation at 1 μM (PAO) and 100 μM (sodium orthovanadate), indicating that the observed inhibition of migration was not due to toxicity (data not shown).

Fig. 1.

Effect of PAO on SDF-1α–induced chemotaxis of CXCR4 L1.2 and Jurkat cells.

CXCR4 L1.2 (A) or Jurkat (B) cells were pretreated with 0, 0.1, 0.3, or 1.0 μM concentration of PAO for 45 minutes. Migration toward 25 ng/mL SDF-1α was measured after 3 hours. Chemotaxis of cells in the absence of inhibitor is considered 100% migration. *P < .05; **P < .005.

Fig. 1.

Effect of PAO on SDF-1α–induced chemotaxis of CXCR4 L1.2 and Jurkat cells.

CXCR4 L1.2 (A) or Jurkat (B) cells were pretreated with 0, 0.1, 0.3, or 1.0 μM concentration of PAO for 45 minutes. Migration toward 25 ng/mL SDF-1α was measured after 3 hours. Chemotaxis of cells in the absence of inhibitor is considered 100% migration. *P < .05; **P < .005.

Close modal
Fig. 2.

Effect of sodium orthovanadate on SDF-1α–induced chemotaxis of CXCR4 L1.2 cells, Jurkat cells, and PBLs.

CXCR4 L1.2 (A), Jurkat (B), or PBL (C) cells were untreated (0) or pretreated with different concentrations of sodium orthovanadate for 45 minutes. Migration toward 25 ng/mL SDF-1α was measured after 3 hours. Chemotaxis of cells in the absence of inhibitor is considered 100% migration. *P < .05; **P < .005.

Fig. 2.

Effect of sodium orthovanadate on SDF-1α–induced chemotaxis of CXCR4 L1.2 cells, Jurkat cells, and PBLs.

CXCR4 L1.2 (A), Jurkat (B), or PBL (C) cells were untreated (0) or pretreated with different concentrations of sodium orthovanadate for 45 minutes. Migration toward 25 ng/mL SDF-1α was measured after 3 hours. Chemotaxis of cells in the absence of inhibitor is considered 100% migration. *P < .05; **P < .005.

Close modal

SHP2 is tyrosine-phosphorylated upon SDF-1α treatment of CXCR4 L1.2 and Jurkat T cells

Recently, the involvement of the tyrosine phosphatase, SHP1, in SDF-1α–induced chemotaxis has been shown in cells derived from SHP1-deficient moth-eaten mice.43 SHP2 has also been shown to regulate migration induced by various growth factors and cytokines.57 58 To characterize the role of SHP2 in CXCR4-mediated signaling pathways, CXCR4 transfectants and Jurkat T cells were treated with SDF-1α, and cell lysates were then analyzed for SHP2 tyrosine phosphorylation. Figure3A-B (top panels) shows that stimulation with SDF-1α resulted in a marked increase in the tyrosine phosphorylation of SHP2. This phosphorylation of SHP2 was rapid, and maximum phosphorylation was obtained after 5 to 10 minutes. Equal amounts of SHP2 protein were present in each lane (Figure 3A-B, bottom panels).

Fig. 3.

Tyrosine phosphorylation of SHP2 upon SDF-1α stimulation.

CXCR4 L1.2 (A) or Jurkat (B) cells were either unstimulated (0) or stimulated with 100 ng/mL SDF-1α for varying time periods. Cells were then lysed and immunoprecipitated with anti-SHP2 antibody. The immune complexes were run on SDS-PAGE and immunoblotted with antiphosphotyrosine antibody (top panels), followed by anti-SHP2 antibody (bottom panels). C represents the antibody control, and TCL is 50 μL of the total cell lysate. IP indicates immunoprecipitation; WB, Western blot.

Fig. 3.

Tyrosine phosphorylation of SHP2 upon SDF-1α stimulation.

CXCR4 L1.2 (A) or Jurkat (B) cells were either unstimulated (0) or stimulated with 100 ng/mL SDF-1α for varying time periods. Cells were then lysed and immunoprecipitated with anti-SHP2 antibody. The immune complexes were run on SDS-PAGE and immunoblotted with antiphosphotyrosine antibody (top panels), followed by anti-SHP2 antibody (bottom panels). C represents the antibody control, and TCL is 50 μL of the total cell lysate. IP indicates immunoprecipitation; WB, Western blot.

Close modal

SHP2 associates with the CXCR4 receptor

SHP1 was recently shown to associate with CXCR4 upon SDF-1α stimulation.25 In the present study, we determined the association of SHP2 with CXCR4 upon SDF-1α stimulation of Jurkat cells. As shown in Figure 4, SHP2 constitutively associated with CXCR4, and this association was enhanced upon SDF-1α stimulation.

Fig. 4.

CXCR4 associates with SHP2 upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated for varying time periods with 100 ng/mL SDF-1α were immunoprecipitated with CXCR4. The immune complexes were subjected to SDS-PAGE and immunoblotted with anti-SHP2 antibody. C represents the antibody control, and TCL is 50 μL of the total cell lysate.

Fig. 4.

CXCR4 associates with SHP2 upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated for varying time periods with 100 ng/mL SDF-1α were immunoprecipitated with CXCR4. The immune complexes were subjected to SDS-PAGE and immunoblotted with anti-SHP2 antibody. C represents the antibody control, and TCL is 50 μL of the total cell lysate.

Close modal

SHP2 associates with SHIP, fyn, and cbl upon SDF-1α stimulation of Jurkat T cells

SHP2 has been shown to associate with several signaling components upon growth factor or cytokine stimulation.36-39 Here, we examined its association with other signaling molecules upon SDF-1α stimulation. As shown in Figure 5A-F, SHP2 associated with the phosphatase SHIP, the adaptor molecule cbl, and fyn kinase upon SDF-1α stimulation of Jurkat T cells. Association with SHIP was rapid, reached a maximum level at 5 to 10 minutes, and was reduced thereafter (Figure 5A). This association was further confirmed by the reverse immunoprecipitation of blotting SHP2 immunoprecipitates with SHIP (Figure 5B). Association of SHP2 with cbl was rapid and sustained for up to 20 minutes of stimulation with SDF-1α (Figure 5C). The cbl association was also reconfirmed by blotting SHP2 immunoprecipitates with cbl (Figure 5D). Furthermore, cbl was shown to associate with the amino-terminal SH2 domain of SHP2 when using GST-fusion protein for the immunoprecipitation (Figure 5E). Fyn constitutively associated with SHP2, was slightly enhanced after 2 to 5 minutes of stimulation with SDF-1α, and was reduced thereafter (Figure 5F).

Fig. 5.

SHP2 associates with SHIP, fyn, and cbl upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods were immunoprecipitated with either anti-SHIP (A), anti-SHP2 (B), anti-cbl (C), anti-SHP2 (D), the GST SH2-domain of SHP2 (E), or anti-Fyn (F). The immune complexes were then run on SDS-PAGE and immunoblotted with anti-SHP2 antibody (A,C,F) or anti-SHIP (B) or anti-cbl antibody (D,E). C represents the antibody control, and TCL is 50 μL of total cell lysate.

Fig. 5.

SHP2 associates with SHIP, fyn, and cbl upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods were immunoprecipitated with either anti-SHIP (A), anti-SHP2 (B), anti-cbl (C), anti-SHP2 (D), the GST SH2-domain of SHP2 (E), or anti-Fyn (F). The immune complexes were then run on SDS-PAGE and immunoblotted with anti-SHP2 antibody (A,C,F) or anti-SHIP (B) or anti-cbl antibody (D,E). C represents the antibody control, and TCL is 50 μL of total cell lysate.

Close modal

SHP2 overexpression enhances SDF-1α–induced migration

To address the functional role of SHP2 in SDF-1α–induced chemotaxis, we transiently transfected Jurkat cells with control vector or SHP2 WT. As shown in Figure 6A, a higher amount of SHP2 was expressed in SHP2 WT–transfected cells (lane 3) than in untransfected or control vector–transfected cells (lanes 1 and 2, respectively). Chemotaxis studies carried out with these transfectants revealed that overexpression of SHP2 WT protein enhanced SDF-1α–induced chemotaxis by 50% to 60% as compared with cells overexpressing the control vector (Figure 6B). Similar results were obtained in 3 different transfection experiments.

Fig. 6.

Overexpression of SHP2 in transfected JMC.T5 Jurkat cells enhances SDF-1α–induced migration.

(A) JMC.T5 Jurkat cells were untransfected or transfected with either control vector or SHP2 WT and then lysed with lysis buffer. Lysates (500 μg protein) from untransfected (lane 1), control vector–transfected (lane 2), or SHP2 WT–transfected (lane 3) cells were immunoprecipitated with SHP2 antibody. The immune complexes were then resolved on SDS-PAGE and blotted with SHP2 antibody. (B) The cells transfected with the control vector or SHP2 WT were subjected to migration assay with SDF-1α (50 ng/mL). The cells migrating to the bottom chamber were counted. *P < .005.

Fig. 6.

Overexpression of SHP2 in transfected JMC.T5 Jurkat cells enhances SDF-1α–induced migration.

(A) JMC.T5 Jurkat cells were untransfected or transfected with either control vector or SHP2 WT and then lysed with lysis buffer. Lysates (500 μg protein) from untransfected (lane 1), control vector–transfected (lane 2), or SHP2 WT–transfected (lane 3) cells were immunoprecipitated with SHP2 antibody. The immune complexes were then resolved on SDS-PAGE and blotted with SHP2 antibody. (B) The cells transfected with the control vector or SHP2 WT were subjected to migration assay with SDF-1α (50 ng/mL). The cells migrating to the bottom chamber were counted. *P < .005.

Close modal

Fyn and lyn kinases are activated upon SDF-1α stimulation of Jurkat T cells

Because fyn associated with SHP2, we examined fyn and lyn kinase activation in SDF-1α–stimulated cells. We found that both fyn and lyn kinases were activated after 2 minutes of SDF-1α treatment, although the activation of fyn was greater than that of lyn (Figure7A-B). The activation of both fyn and lyn kinases was reduced after 5 to 10 minutes of stimulation.

Fig. 7.

Fyn and Lyn are activated upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods were immunoprecipitated with anti-fyn (A) or anti-lyn (B) antibody. The immune complexes were subjected to in vitro kinase reactions. C represents the control antibody.

Fig. 7.

Fyn and Lyn are activated upon SDF-1α stimulation.

Lysates obtained from Jurkat cells unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods were immunoprecipitated with anti-fyn (A) or anti-lyn (B) antibody. The immune complexes were subjected to in vitro kinase reactions. C represents the control antibody.

Close modal

Cbl is phosphorylated upon SDF-1α stimulation of Jurkat T cells and associates with PI-3 kinase, 14-3-3β, and Crk-L

Cbl acts as an adaptor protein in tyrosine-dependent signaling and has been shown to associate with several signaling molecules in response to a variety of stimuli.44-48 Because cbl associated with SHP2, we explored its phosphorylation and association with other signaling molecules in response to SDF-1α treatment. Stimulation with SDF-1α increased the tyrosine phosphorylation of cbl in Jurkat cells (Figure 8A, upper panel). Maximum phosphorylation was obtained after 2 to 5 minutes of stimulation and declined thereafter. Equal amounts of cbl protein were present in each lane (Figure 8A, bottom panel). Furthermore, cbl associated with PI-3 kinase and the adaptor proteins Crk-L and 14-3-3β (Figure 8B-F). Cbl associated constitutively with PI-3 kinase, and the association was only slightly increased after 5 minutes of treatment with SDF-1α (Figure 8B). Similar results were obtained when PI-3 kinase immunoprecipitates were immunoblotted with cbl (Figure8C). The association of cbl with Crk-L was also enhanced upon SDF-1α stimulation (Figure 8D). This association was reconfirmed by immunoblotting Crk-L immunoprecipitates with cbl (Figure 8E). Cbl also associated with 14-3-3β upon SDF-1α stimulation (Figure8F).

Fig. 8.

Cbl is tyrosine-phosphorylated and associates with PI-3 kinase, Crk-L, and 14-3-3β upon SDF-1α stimulation.

Lysates obtained from Jurkat cells were unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods and then lysed. The cell lysates were immunoprecipitated with anti-cbl antibody run on SDS-PAGE and immunoblotted with antiphosphotyrosine antibody (A, top panel), followed by anti-cbl antibody (A, bottom panel). Equivalent amounts of cell lysate (500 μg) were immunoprecipitated with anti-cbl (B,D,F), anti–PI-3 kinase (C), or anti-Crk-L (E) and then run on SDS-PAGE and immunoblotted with anti–PI-3 kinase (B), anti-cbl (C,E), anti–Crk-L (D), or anti–14-3-3β (F). C represents the antibody control, and TCL is 50 μL of total cell lysate.

Fig. 8.

Cbl is tyrosine-phosphorylated and associates with PI-3 kinase, Crk-L, and 14-3-3β upon SDF-1α stimulation.

Lysates obtained from Jurkat cells were unstimulated (0) or stimulated with 100 ng/mL SDF-1α for various time periods and then lysed. The cell lysates were immunoprecipitated with anti-cbl antibody run on SDS-PAGE and immunoblotted with antiphosphotyrosine antibody (A, top panel), followed by anti-cbl antibody (A, bottom panel). Equivalent amounts of cell lysate (500 μg) were immunoprecipitated with anti-cbl (B,D,F), anti–PI-3 kinase (C), or anti-Crk-L (E) and then run on SDS-PAGE and immunoblotted with anti–PI-3 kinase (B), anti-cbl (C,E), anti–Crk-L (D), or anti–14-3-3β (F). C represents the antibody control, and TCL is 50 μL of total cell lysate.

Close modal

The α-chemokine, SDF-1α, and its cognate receptor, CXCR4, play a critical role in embryonic development, neuronal patterning, and HIV pathogenesis.1,17,19-22 The association of SDF-1α with CXCR4 also plays an important role in the immune system through the regulation of myeloid progenitor proliferation and the trafficking of leukocytes.12-14 However, the signaling pathways behind these normal and disease processes are only beginning to be understood. SDF-1α–induced activation of CXCR4 is coupled to the JAK/STAT, PI-3 kinase, p44/p42 MAPK, and NF-κB pathways.23-26 Recently, SDF-1α was shown to induce phosphorylation of the CXCR4 receptor at serine and tyrosine residues and association of the CXCR4 receptor with the tyrosine phosphatase, SHP1.25,59 Furthermore, the altered chemotactic response to SDF-1α observed in hematopoietic cells derived from SHP1-deficient mice indicates that SHP1 mediates SDF-1α–induced chemotaxis.43 

In this study, we have further assessed the role of PTPs in SDF-1α–initiated signaling events. PTPs can regulate various cellular functions by initiating and terminating signals. We observed that the SDF-1α–induced chemotaxis of CXCR4+ cells was significantly reduced by the tyrosine phosphatase inhibitors PAO and sodium orthovanadate. However, PAO and sodium orthovanadate did not affect SDF-1α–induced p44/42 MAPK activation. The strong inhibition of migration by tyrosine phosphatase inhibitors suggests that tyrosine phosphatases are important mediators of SDF-1α–induced chemotaxis. PAO and sodium orthovanadate have been reported to suppress platelet-derived growth factor–induced migration and cell spreading of lung carcinoma cells by inhibiting SHP2 activity.51,60 We found that SDF-1α stimulation of T cells enhanced the tyrosine phosphorylation of SHP2, which has been shown to be tyrosine-phosphorylated in response to stimulation by growth factors and cytokines.32-35 Furthermore, we observed that overexpression of SHP2 WT enhanced SDF-1α–induced chemotaxis. In other studies, SHP2 has been shown to regulate chemotaxis induced by integrins. Its role in cell migration has also been confirmed in SHP2−/− knock-out mice; fibroblasts derived from these mice have slower spreading and motility as compared with cells from WT animals.35 SHP2 has been shown to regulate cell adhesion, spreading, and migration by modulating tyrosine phosphorylation of focal adhesion proteins.35,58,61 We and others have recently shown that SDF-1α induces the tyrosine phosphorylation of components of focal adhesion complexes.23 62 

SHP2 was shown to associate with the CXCR4 receptor and the signaling molecules SHIP and PI-3 kinase. Tyrosine-phosphorylated SHP2 can act as an adaptor molecule and has been shown to bind to several signaling molecules, including growth factor receptors and the adhesion molecule PECAM-1.32-35,38 Recently, another PTP, SHP1, was shown to bind to the CXCR4 receptor.25 SHIP, which is a 145-kd inositol-5-phosphatase that selectively hydrolyzes the 5′ phosphate from both inositol 1,3,4,5 tetraphosphate (IP4) and phosphatidylinositol 3,4,5, triphosphate (PIP3), has previously been shown to bind to SHP2 in various cell types.31,63,64 SHIP seems to play an important role in chemokine-mediated signaling: Hematopoietic cells derived from SHIP−/− knock-out mice showed enhanced chemotactic responses to SDF-1α as compared with the cells from WT mice.65 SHP2 also binds to PI-3 kinase, which recently was shown to regulate SDF-1α–induced chemotaxis.23,26 Furthermore, SHIP may down-regulate PI-3 kinase–initiated signaling events through inhibition of protein kinase B activation by decreasing the levels of PIP3.66 

Tyrosine phosphorylation of the adaptor protein cbl was enhanced upon SDF-1α stimulation. Therefore, cbl may play a role in SDF-1α–induced chemotaxis in T cells. Cbl is a prominent component of signaling events downstream of a variety of cell surface receptors, including T cells, B cells, epidermal growth factor, integrins, and cytokines.44-48 It is involved in ubiquitination and the endocytic degradation of various receptors.49-51 

We observed that SDF-1α enhanced the association of cbl with SHP2, PI-3 kinase, Crk-L, and 14-3-3β. The reduction of cbl tyrosine phosphorylation and its association with membrane-localized PI-3 kinase activity have been shown in Src-family kinase mutants, which were defective in their ability to spread on fibronectin-coated surfaces. Furthermore, cbl antisense nucleotides and PI-3 kinase inhibitors also blocked the spreading of WT cells.47 Association of cbl with Crk-L was recently shown to be involved in Ba/F3 cell chemotactic events.67 Cbl also associates with 14-3-3β proteins upon SDF-1α stimulation. In addition, T-cell activation mediated by anti-CD3 has been shown to lead to the enhanced association of cbl with 14-3-3β proteins.68,69 These proteins bind to a wide variety of regulatory proteins and thereby participate in signaling events leading to cell differentiation and proliferation.70-72 

The Src-related tyrosine kinases fyn and lyn were also activated upon SDF-1α stimulation, and fyn associated with SHP2. Fyn and lyn have been shown to associate with cbl and SHP2 and thereby regulate their functions.73-75 Both of these kinases play important roles in various growth factor and T- and B-cell receptor signaling by initiating changes in the phosphorylation of signaling intermediates that modulate further downstream events.76 

Taken together, our studies suggest that the tyrosine phosphatase SHP2 and the adaptor cbl are key components of CXCR4-mediated signaling events induced by SDF-1α. SHP2 associates with CXCR4 and forms a multiprotein signaling complex with cbl, SHIP, and fyn. Cbl also forms an activation-induced complex with PI-3 kinase, Crk-L, and 14-3-3β proteins. The formation of these large multimeric signaling complexes may result in the activation of multiple downstream effectors, which mediate the various functional effects of the CXCR4 receptor.

We are thankful to Dr Jerome E. Groopman for his advice and support for our research project. We also thank Stephanie Brubaker for technical help, Janet Delahanty for editing, Daniel Kelley for preparation of the figures, and Simone Jadusingh for facilitating receipt of the reagents for the experiments.

Supported by National Institutes of Health grant CA76950 (R.K.G.).

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
Premack
BA
Schall
TJ
Chemokine receptors: gateways to inflammation and infection.
Nat Med.
2
1996
1174
1178
2
Rollins
BJ
Chemokines.
Blood.
90
1997
909
928
3
Bokoch
GM
Chemoattractant signaling and leukocyte activation.
Blood.
86
1995
1649
1660
4
Kunkel
SL
Through the looking glass: the diverse in vivo activities of chemokines.
J Clin Invest.
104
1999
1333
1334
5
Luster
AD
Chemokines—chemotactic cytokines that mediate inflammation.
N Engl J Med.
338
1998
436
445
6
Boshoff
C
Weiss
RA
Kaposi's sarcoma-associated herpesvirus.
Adv Cancer Res.
75
1998
57
86
7
Bais
C
Santomasso
B
Coso
O
et al
G-protein-coupled receptor of Kaposi's sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator.
Nature.
391
1998
86
89
8
Hamada
T
Tashiro
K
Tada
H
et al
Isolation and characterization of a novel secretory protein, stromal cell-derived factor-2 (SDF-2) using the signal sequence trap method.
Gene.
176
1996
211
214
9
Bleul
CC
Farzan
M
Choe
H
et al
The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry.
Nature.
382
1996
829
833
10
Feng
Y
Broder
CC
Kennedy
PE
Berger
EA
HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor.
Science.
272
1996
872
877
11
Loetscher
M
Geiser
T
O'Reilly
T
Zwahlen
R
Baggiolini
M
Moser
B
Cloning of a human seven-transmembrane domain receptor, LESTR, that is highly expressed in leukocytes.
J Biol Chem.
269
1994
232
237
12
Aiuti
A
Webb
IJ
Bleul
C
Springer
T
Gutierrez-Ramos
JC
The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood.
J Exp Med.
185
1997
111
120
13
Bleul
CC
Wu
L
Hoxie
JA
Springer
TA
Mackay
CR
The HIV coreceptors CXCR4 and CCR5 are differentially expressed and regulated on human T lymphocytes.
Proc Natl Acad Sci U S A.
94
1997
1925
1930
14
Forster
R
Kremmer
E
Schubel
A
et al
Intracellular and surface expression of the HIV-1 coreceptor CXCR4/fusin on various leukocyte subsets: rapid internalization and recycling upon activation.
J Immunol.
160
1998
1522
1531
15
Gupta
SK
Lysko
PG
Pillarisetti
K
Ohlstein
E
Stadel
JM
Chemokine receptors in human endothelial cells: functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokines.
J Biol Chem.
273
1998
4282
4287
16
Hesselgesser
J
Halks-Miller
M
DelVecchio
V
et al
CD4-independent association between HIV-1 gp120 and CXCR4: functional chemokine receptors are expressed in human neurons.
Curr Biol.
7
1997
112
121
17
Littman
DR
Chemokine receptors: keys to AIDS pathogenesis?
Cell.
93
1998
677
680
18
Berger
EA
Murphy
PM
Farber
JM
Chemokine receptors as HIV-1 coreceptors: roles in viral entry, tropism, and disease.
Annu Rev Immunol.
17
1999
657
700
19
Nagasawa
T
Hirota
S
Tachibana
K
et al
Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1.
Nature.
382
1996
635
638
20
Ma
Q
Jones
D
Borghesani
PR
et al
Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice.
Proc Natl Acad Sci U S A.
95
1998
9448
9453
21
Tachibana
K
Hirota
S
Iizasa
H
et al
The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract.
Nature.
393
1998
591
594
22
Zou
YR
Kottmann
AH
Kuroda
M
Taniuchi
I
Littman
DR
Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development.
Nature.
393
1998
595
599
23
Ganju
RK
Brubaker
SA
Meyer
J
et al
The alpha-chemokine, stromal cell-derived factor-1alpha, binds to the transmembrane G-protein-coupled CXCR-4 receptor and activates multiple signal transduction pathways.
J Biol Chem.
273
1998
23169
23175
24
Davis
CB
Dikic
I
Unutmaz
D
et al
Signal transduction due to HIV-1 envelope interactions with chemokine receptors CXCR4 or CCR5.
J Exp Med.
186
1997
1793
1798
25
Vila-Coro
AJ
Rodriguez-Frade
JM
Martin De Ana
A
Moreno-Ortiz
MC
Martinez-A
C
Mellado
M
The chemokine SDF-1alpha triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway.
FASEB J.
13
1999
1699
1710
26
Sotsios
Y
Whittaker
GC
Westwick
J
Ward
SG
The CXC chemokine stromal cell-derived factor activates a Gi-coupled phosphoinositide 3-kinase in T lymphocytes.
J Immunol.
163
1999
5954
5963
27
Neel
BG
Role of phosphatases in lymphocyte activation.
Curr Opin Immunol.
9
1997
405
420
28
Streuli
M
Protein tyrosine phosphatases in signaling.
Curr Opin Cell Biol.
8
1996
182
188
29
Haque
SJ
Harbor
P
Tabrizi
M
Yi
T
Williams
BR
Protein-tyrosine phosphatase Shp-1 is a negative regulator of IL-4- and IL-13-dependent signal transduction.
J Biol Chem.
273
1998
33893
33896
30
Dong
Q
Siminovitch
KA
Fialkow
L
Fukushima
T
Downey
GP
Negative regulation of myeloid cell proliferation and function by the SH2 domain-containing tyrosine phosphatase-1.
J Immunol.
162
1999
3220
3230
31
Huber
M
Helgason
CD
Damen
JE
et al
The role of SHIP in growth factor induced signalling.
Prog Biophys Mol Biol.
71
1999
423
434
32
Frearson
JA
Alexander
DR
The phosphotyrosine phosphatase SHP-2 participates in a multimeric signaling complex and regulates T cell receptor (TCR) coupling to the Ras/mitogen-activated protein kinase (MAPK) pathway in Jurkat T cells.
J Exp Med.
187
1998
1417
1426
33
Nakamura
K
Cambier
JC
B cell antigen receptor (BCR)-mediated formation of a SHP-2-pp120 complex and its inhibition by Fc gamma RIIB1-BCR coligation.
J Immunol.
161
1998
684
691
34
Gu
H
Griffin
JD
Neel
BG
Characterization of two SHP-2-associated binding proteins and potential substrates in hematopoietic cells.
J Biol Chem.
272
1997
16421
16430
35
Oh
ES
Gu
H
Saxton
TM
et al
Regulation of early events in integrin signaling by protein-tyrosine-phosphatase SHP2.
Mol Cell Biol.
19
1999
3205
3215
36
Bone
H
Dechert
U
Jirik
F
Schrader
JW
Welham
MJ
SHP1 and SHP2 protein-tyrosine phosphatases associate with betac after interleukin-3-induced receptor tyrosine phosphorylation: identification of potential binding sites and substrates.
J Biol Chem.
272
1997
14470
14476
37
Yu
Z
Su
L
Hoglinger
O
Jaramillo
ML
Banville
D
Shen
SH
SHP-1 associates with both platelet-derived growth factor receptor and the p85 subunit of phosphatidylinositol 3-kinase.
J Biol Chem.
273
1998
3687
3694
38
Sagawa
K
Kimura
T
Swieter
M
Siraganian
RP
The protein-tyrosine phosphatase SHP-2 associates with tyrosine-phosphorylated adhesion molecule PECAM-1 (CD31).
J Biol Chem.
272
1997
31086
31091
39
Ong
SH
Lim
YP
Low
BC
Guy
GR
SHP2 associates directly with tyrosine phosphorylated p90 (SNT) protein in FGF-stimulated cells.
Biochem Biophys Res Commun.
238
1997
261
266
40
Shultz
LD
Schweitzer
PA
Rajan
TV
et al
Mutations at the murine motheaten locus are within the hematopoietic cell protein-tyrosine phosphatase (Hcph) gene.
Cell.
73
1993
1445
1454
41
Shultz
LD
Rajan
TV
Greiner
DL
Severe defects in immunity and hematopoiesis caused by SHP-1 protein-tyrosine-phosphatase deficiency.
Trends Biotechnol.
15
1997
302
307
42
Qu
CK
Yu
WM
Azzarelli
B
Cooper
S
Broxmeyer
HE
Feng
GS
Biased suppression of hematopoiesis and multiple developmental defects in chimeric mice containing Shp-2 mutant cells.
Mol Cell Biol.
18
1998
6075
6082
43
Kim
CH
Qu
CK
Hangoc
G
et al
Abnormal chemokine-induced responses of immature and mature hematopoietic cells from motheaten mice implicate the protein tyrosine phosphatase SHP-1 in chemokine responses.
J Exp Med.
190
1999
681
690
44
Donovan
JA
Wange
RL
Langdon
WY
Samelson
LE
The protein product of the c-cbl protooncogene is the 120-kDa tyrosine-phosphorylated protein in Jurkat cells activated via the T cell antigen receptor.
J Biol Chem.
269
1994
22921
22924
45
Andoniou
CE
Lill
NL
Thien
CB
et al
The Cbl proto-oncogene product negatively regulates the Src-family tyrosine kinase Fyn by enhancing its degradation.
Mol Cell Biol.
20
2000
851
867
46
Lee
PS
Wang
Y
Dominguez
MG
et al
The Cbl protooncoprotein stimulates CSF-1 receptor multiubiquitination and endocytosis, and attenuates macrophage proliferation.
EMBO J.
18
1999
3616
3628
47
Meng
F
Lowell
CA
A beta 1 integrin signaling pathway involving Src-family kinases, Cbl and PI-3 kinase is required for macrophage spreading and migration.
EMBO J.
17
1998
4391
4403
48
Feshchenko
EA
Langdon
WY
Tsygankov
AY
Fyn, Yes, and Syk phosphorylation sites in c-Cbl map to the same tyrosine residues that become phosphorylated in activated T cells.
J Biol Chem.
273
1998
8323
8331
49
Barinaga
M
A new finger on the protein destruction button.
Science.
286
1999
223
225
50
Yokouchi
M
Kondo
T
Houghton
A
et al
Ligand-induced ubiquitination of the epidermal growth factor receptor involves the interaction of the c-Cbl RING finger and UbcH7.
J Biol Chem.
274
1999
31707
31712
51
Levkowitz
G
Waterman
H
Ettenberg
SA
et al
Ubiquitin ligase activity and tyrosine phosphorylation underlie suppression of growth factor signaling by c-Cbl/Sli-1.
Mol Cell.
4
1999
1029
1040
52
Ponath
PD
Qin
S
Post
TW
et al
Molecular cloning and characterization of a human eotaxin receptor expressed selectively on eosinophils [see comments].
J Exp Med.
183
1996
2437
2448
53
Wu
L
Ruffing
N
Shi
X
et al
Discrete steps in binding and signaling of interleukin-8 with its receptor.
J Biol Chem.
271
1996
31202
31209
54
Wu
L
Gerard
NP
Wyatt
R
et al
CD4-induced interaction of primary HIV-1 gp120 glycoproteins with the chemokine receptor CCR-5.
Nature.
384
1996
179
183
55
Ganju
RK
Dutt
P
Wu
L
et al
Beta-chemokine receptor CCR5 signals via the novel tyrosine kinase RAFTK.
Blood.
91
1998
791
797
56
Alfano
M
Schmidtmayerova
H
Amella
CA
Pushkarsky
T
Bukrinsky
M
The B-oligomer of pertussis toxin deactivates CC chemokine receptor 5 and blocks entry of M-tropic HIV-1 strains.
J Exp Med.
190
1999
597
605
57
Qi
JH
Ito
N
Claesson-Welsh
L
Tyrosine phosphatase SHP-2 is involved in regulation of platelet-derived growth factor-induced migration.
J Biol Chem.
274
1999
14455
14463
58
Manes
S
Mira
E
Gomez-Mouton
C
Zhao
ZJ
Lacalle
RA
Martinez
AC
Concerted activity of tyrosine phosphatase SHP-2 and focal adhesion kinase in regulation of cell motility.
Mol Cell Biol.
19
1999
3125
3135
59
Haribabu
B
Richardson
RM
Fisher
I
et al
Regulation of human chemokine receptors CXCR4. Role of phosphorylation in desensitization and internalization.
J Biol Chem.
272
1997
28726
28731
60
Chintala
SK
Kyritsis
AP
Mohan
PM
et al
Altered actin cytoskeleton and inhibition of matrix metalloproteinase expression by vanadate and phenylarsine oxide, inhibitors of phosphotyrosine phosphatases: modulation of migration and invasion of human malignant glioma cells.
Mol Carcinog.
26
1999
274
285
61
Yu
DH
Qu
CK
Henegariu
O
Lu
X
Feng
GS
Protein-tyrosine phosphatase Shp-2 regulates cell spreading, migration, and focal adhesion.
J Biol Chem.
273
1998
21125
21131
62
Wang
JF
Park
IW
Groopman
JE
Stromal cell-derived factor-1α stimulates tyrosine phosphorylation of multiple focal adhesion proteins and induces migration of hematopoietic progenitor cells: roles of phosphoinositide-3 kinase and protein kinase C.
Blood.
95
2000
2505
2513
63
Liu
L
Damen
JE
Ware
MD
Krystal
G
Interleukin-3 induces the association of the inositol 5-phosphatase SHIP with SHP2.
J Biol Chem.
272
1997
10998
11001
64
Lecoq-Lafon
C
Verdier
F
Fichelson
S
et al
Erythropoietin induces the tyrosine phosphorylation of GAB1 and its association with SHC, SHP2, SHIP, and phosphatidylinositol 3-kinase.
Blood.
93
1999
2578
2585
65
Kim
CH
Hangoc
G
Cooper
S
et al
Altered responsiveness to chemokines due to targeted disruption of SHIP.
J Clin Invest.
104
1999
1751
1759
66
Jacob
A
Cooney
D
Tridandapani
S
Kelley
T
Coggeshall
KM
FcgammaRIIb modulation of surface immunoglobulin-induced Akt activation in murine B cells.
J Biol Chem.
274
1999
13704
13710
67
Uemura
N
Griffin
JD
The adapter protein Crkl links Cbl to C3G after integrin ligation and enhances cell migration.
J Biol Chem.
274
1999
37525
37532
68
Liu
YC
Elly
C
Yoshida
H
Bonnefoy-Berard
N
Altman
A
Activation-modulated association of 14-3-3 proteins with Cbl in T cells.
J Biol Chem.
271
1996
14591
14595
69
Liu
YC
Liu
Y
Elly
C
Yoshida
H
Lipkowitz
S
Altman
A
Serine phosphorylation of Cbl induced by phorbol ester enhances its association with 14-3-3 proteins in T cells via a novel serine-rich 14-3-3-binding motif.
J Biol Chem.
272
1997
9979
9985
70
Garcia-Guzman
M
Dolfi
F
Russello
M
Vuori
K
Cell adhesion regulates the interaction between the docking protein p130(Cas) and the 14-3-3 proteins.
J Biol Chem.
274
1999
5762
5768
71
Luk
SC
Ngai
SM
Tsui
SK
Fung
KP
Lee
CY
Waye
MM
In vivo and in vitro association of 14-3-3 epsilon isoform with calmodulin: implication for signal transduction and cell proliferation.
J Cell Biochem.
73
1999
31
35
72
Tzivion
G
Luo
Z
Avruch
J
A dimeric 14-3-3 protein is an essential cofactor for Raf kinase activity.
Nature.
394
1998
88
92
73
Tang
H
Zhao
ZJ
Huang
XY
Landon
EJ
Inagami
T
Fyn kinase-directed activation of SH2 domain-containing protein-tyrosine phosphatase SHP-2 by Gi protein-coupled receptors in Madin-Darby canine kidney cells.
J Biol Chem.
274
1999
12401
12407
74
Anderson
SM
Burton
EA
Koch
BL
Phosphorylation of Cbl following stimulation with interleukin-3 and its association with Grb2, Fyn, and phosphatidylinositol 3-kinase.
J Biol Chem.
272
1997
739
745
75
Deckert
M
Elly
C
Altman
A
Liu
YC
Coordinated regulation of the tyrosine phosphorylation of Cbl by Fyn and Syk tyrosine kinases.
J Biol Chem.
273
1998
8867
8874
76
Kennedy
JS
Raab
M
Rudd
CE
Signaling scaffolds in immune cells.
Cell Calcium.
26
1999
227
235

Author notes

Ramesh K. Ganju, Division of Experimental Medicine, Harvard Institutes of Medicine-BIDMC, 4 Blackfan Circle, Room 343, Boston, MA 02115; e-mail: rganju@caregroup.harvard.edu.

Sign in via your Institution