PTEN/MMAC1 is a major new tumor suppressor gene that encodes a dual-specificity phosphatase with sequence similarity to the cytoskeletal protein tensin. Recently, we reported that PTEN dephosphorylates focal adhesion kinase (FAK) and inhibits cell migration, spreading, and focal adhesion formation. Here, the effects of PTEN on cell invasion, migration, and growth as well as the involvement of FAK and p130 Crk-associated substrate (p130Cas) were investigated in U87MG glioblastoma cells missing PTEN. Cell invasion, migration, and growth were down-regulated by expression of phosphatase-active forms of PTEN but not by PTEN with an inactive phosphatase domain; these effects were correlated with decreased tyrosine phosphorylation levels of FAK and p130Cas. Overexpression of FAK concomitant with PTEN resulted in increased total tyrosine phosphorylation levels of FAK and p130Cas and effectively antagonized the effects of PTEN on cell invasion and migration and partially on cell growth. Overexpression of p130Cas increased total tyrosine phosphorylation levels of p130Cas without affecting those of FAK; however, although p130Cas could reverse PTEN inhibition of cell invasion and migration, it did not rescue cell growth in U87MG cells. In contrast to FAK, p130Cas could not be shown to interact with PTEN in cells, and it was not dephosphorylated directly by PTEN in vitro. These results suggest important roles of PTEN in the phenotype of tumor progression, and that the effects of PTEN on cell invasion, migration, and growth are mediated by distinct downstream pathways that diverge at the level of FAK.

PTEN3 (also called MMAC1, mutated in multiple advanced cancers 1) is a major new tumor suppressor gene recently identified on human chromosome 10q23. It was identified as a candidate tumor suppressor gene based on the presence of inactivating mutations in human brain, breast, and prostate cancers (1, 2). PTEN is also frequently deleted or mutated in a wide range of human tumors and tumor cell lines such as glioblastoma, melanoma, and lymphoid, lung, and endometrial cancers (3, 4, 5, 6, 7, 8, 9, 10, 11). Furthermore, germ-line PTEN mutations have been found in patients with juvenile polyposis coli (12), Cowden disease, a multiple hamartoma syndrome with a high risk of breast and thyroid cancer (13), and the related hamartomatous polyposis syndrome, Bannayan-Zonana syndrome (14), suggesting that inactivation of PTEN plays important roles in tumorigenicity.

Recently, suppression of tumorigenicity and cell growth by expression of PTEN cDNA in glioma cells with mutated PTEN alleles was reported (15, 16). PTEN encodes the catalytic signature motif of protein tyrosine phosphatases and functions as a dual-specificity phosphatase in vitro(3, 17, 18). Although mutations of PTEN are seen throughout the gene in various cancers, the phosphatase domain seems to be critical for tumor suppression, because most missense mutations are observed to cluster around the phosphatase domain, and most remaining mutations are predicted to truncate the protein because of nonsense or frameshift mutations (1, 2, 10). In fact, growth suppression in glioma cells with mutated PTEN alleles by PTEN cDNA expression was shown to require a functional phosphatase catalytic domain (15). Furthermore, mutation or a reported decreased expression of PTEN is associated with advanced glioma, melanoma, and prostate cancer, implicating losses of PTEN mutation in tumor progression (6, 19, 20).

PTEN also has sequence similarity to the cytoskeletal protein tensin (1, 2, 3), which binds to actin filaments at focal adhesions. We have reported recently that PTEN inhibits cell migration, spreading, and focal adhesions (21). PTEN interacts with FAK and reduces its tyrosine phosphorylation, as well as that of a potential downstream effector, p130Cas. FAK is thought to be a key molecule implicated in integrin signaling pathways. The activation of integrins by cell binding to extracellular matrix leads to increases in FAK tyrosine phosphorylation and enhances kinase activity (22, 23, 24, 25, 26, 27). Activation of FAK also leads to its association with several kinases and signal transduction molecules including Grb2, Src, and Shc, followed by activation of extracellular signal-regulated kinase 2/MAP kinase (28, 29, 30, 31). In addition to its potential involvement in integrin stimulation of MAP kinase, FAK promotes cell migration through the activation of p130Cas(32). FAK activation may lead to increases in tyrosine phosphorylation of p130Cas directly (33, 34) or via c-Src binding to FAK (35). In fact, this pathway may be essential for cell migration in several cell lines, and the FAK/p130Cas complex appears to target downstream pathways other than MAP kinases in mediating FAK-promoted cell migration (32, 36).

In the present study, we investigated the ability of PTEN to suppress glioblastoma cell invasion and migration and whether mutations in its phosphatase domain could ablate these abilities. To explore PTEN signaling pathways, we also tested whether overexpression of FAK and its downstream effector p130Cas could antagonize the effects of PTEN. We have found that FAK overexpression can rescue cell invasion, migration, and even to some extent growth, whereas p130Cas can rescue only cell invasion and migration. These findings suggest that PTEN suppresses cell invasion, migration, and growth, possibly through FAK dephosphorylation, and that p130Cas is a downstream mediator of FAK involved in mediating cell invasion and migration, but not in cell growth, showing distinct downstream pathways that diverge at the level of FAK.

Expression Plasmids.

GFP expression plasmids based on pGZ21δxZ containing no insert, full-length wild-type PTEN, or HA-tagged FAK were constructed as described (21). The point mutants D92A (Asp92→Ala), C124A (Cys124→Ala), or G129E (Gly129→Glu) were introduced into PTEN by site-directed mutagenesis as described (21). pSSRα-Cas-FLAG was constructed by inserting the epitope tag FLAG (Eastman Kodak Co., New Haven, CT) at the 3′ end of the p130Cas coding sequence in the expression vector pSSRα-Cas (37) that was kindly provided by Hisamaru Hirai (University of Tokyo, Tokyo, Japan). The puromycin resistance plasmid pHA262pur (38) was kindly provided by Hein te Riele (Netherlands Cancer Institute, Amsterdam, the Netherlands).

Antibodies.

The monoclonal antibody 2A7 (Upstate Biotechnology, Lake Placid, NY) directed against FAK was used for immunoprecipitation, and a second monoclonal antibody against FAK (Transduction Laboratories, Lexington, KY) was used for immunoblotting. Monoclonal antibodies for p130Cas and phosphotyrosine (RC20) were obtained from Transduction Laboratories. Monoclonal antibody against FLAG (M2) was from Eastman Kodak, against HA (12CA5) was from Boehringer Mannheim (Indianapolis, IN), and against GFP was from Clontech (Palo Alto, CA). Rabbit polyclonal antibody MT478 against PTEN was raised by immunization with His6-tagged recombinant PTEN expressed in Escherichia coli as described (21). IgG was purified from antiserum using GammaBind G Sepharose (Amersham Pharmacia Biotech, Piscataway, NJ).

Cell Culture and Transfections.

The PTEN-mutated glioblastoma cell line U87MG was obtained from American Type Culture Collection (Manassas, VA). Human foreskin fibroblasts were kindly provided by Susan Yamada (National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD) and used as a positive control of cells expressing endogenous PTEN. Cells were maintained in DMEM supplemented with 10% fetal bovine serum, 100 units/ml penicillin, and 100 μg/ml streptomycin and cultured in 10% CO2 at 37°C. Calf serum (10%) was substituted for culturing NIH 3T3 cells. Transfections were performed by electroporation (39). Briefly, pGZ21δxZ (20 μg; alternatively, 10 μg in the case of cotransfection with either 10 μg HA-FAK or 10 μg FLAG-Cas) containing either no insert or PTEN was transfected into 1.5 × 106 U87MG cells by electroporation together with 10 μg of pHA262pur. Cells were subcultured at a 1:3 dilution 24 h after transfection and were maintained for 3 days in 1 μg/ml puromycin-containing medium. For cell invasion and migration assays, cells were serum starved by incubating in DMEM containing 0.5% serum during the last 24 h of puromycin selection to exclude any effects of growth suppression by PTEN on cell invasion and migration assays. For coimmunoprecipitation experiments, NIH 3T3 cells were transfected with either HA-FAK or FLAG-Cas (3 μg each) plus GFP-tagged wild-type PTEN or trapping mutant D92A (30 μg each) and used for immunoprecipitations 24 h after transfection. Cells were analyzed for expression of various constructs by Western blotting for PTEN, FAK, and p130Cas as described below; GFP-expressing cells were also quantified by scoring at least 100 cells per transfection using a fluorescence microscope.

Immunoprecipitation and Western Blotting.

After puromycin selection of cells expressing the various constructs, U87MG cells (5 × 105) were allowed to spread for 2 h on 10-cm Petri dishes coated with 10 μg/ml fibronectin. The cells were washed with ice-cold PBS and solubilized in RIPA lysis buffer for 20 min on ice (40). For coimmunoprecipitation experiments, NIH 3T3 cells were solubilized in modified CSK buffer (100 mm NaCl, 0.5% Triton X-100, 300 mm sucrose, 3 mm MgCl2, and 10 mm 1,4-piperazinediethanesulfonic acid, pH 6.8) containing 2 mm phenylmethylsulfonyl fluoride, 50 mm sodium fluoride, 1 mm sodium orthovanadate, and protease inhibitor mixture (Boehringer Mannheim). The homogenates were clarified by centrifugation at 20,000 × g for 15 min at 4°C, protein concentrations were determined using a bicinchoninic acid protein assay kit (Pierce, Rockford, IL), and samples were adjusted to equal protein concentration and volume. Immunoprecipitation was performed as described (40), except using anti-FAK (5 μg/ml), anti-p130Cas (4 μg/ml), anti-HA (4 μg/ml), or anti-FLAG (5 μg/ml) antibodies. Immunoprecipitates (or total cell lysates for anti-PTEN blotting) were resolved in 4–12% SDS-polyacrylamide gels (Novex, San Diego, CA) and electrophoretically transferred to nitrocellulose membrane (Novex) for 1.5 h at 150 mA. The filters were incubated with blocking buffer (5% nonfat dry milk; alternatively, 5% BSA for anti-phosphotyrosine antibody, in T-TBS: 150 mm NaCl, 50 mm Tris-HCl, and 0.1% Tween 20, pH 7.4) for 1 h and then incubated for 1 h with either anti-PTEN (2 μg/ml) or horseradish peroxidase-conjugated RC20 (1:2500) antibodies. Blots were visualized by the enhanced chemiluminescence (ECL) reaction (Amersham Life Science, Arlington Heights, IL). Membranes were then stripped with 2% SDS, 100 mm β-mercaptoethanol in 62.5 mm Tris-HCl (pH 6.8) for 30 min at 70°C, reprobed for FAK (1:1000), p130Cas (1:1000), or GFP (1:1000), and visualized again by ECL.

Cell Invasion and Migration.

Cell invasion assays were carried out using modified Boyden chambers consisting of Transwell (Corning Costar Corp., Cambridge, MA) membrane filter inserts in 24-well tissue culture plates. The Transwell filters were 6.5-mm diameter, 8-μm pore size, 10-μm thickness polycarbonate membranes. The upper surfaces of the Transwell membranes were coated with 1 mg/ml Matrigel matrix (Collaborative Biomedical Products, Bedford, MA; Becton Dickinson Labware, Franklin Lakes, NJ) overnight at 4°C, rinsed once with invasion buffer (DMEM containing 1% BSA and 0.5% serum), and then placed into 24-well tissue culture plates containing 600 μl of invasion buffer/well. After selection and serum starvation of transfected cells as described above, U87MG cells were harvested using 0.53 mm EDTA and resuspended in invasion buffer. Cells (2 × 105) in 100 μl of invasion buffer were added to each Transwell chamber and allowed to invade toward the underside of the membrane for 16 h. Noninvading cells were removed by wiping the upper side of the membrane, and the invaded cells were fixed and stained using Diff-Quik (Baxter Healthcare Corp., McGraw Park, IL). The number of invaded cells per membrane was counted under a light microscope at ×200.

Cell migration was measured using an in vitro wound healing assay as described (21). Briefly, in vitro “scratch” wounds were created by scraping confluent cell monolayers in fibronectin-coated (10 μg/ml) Petri dishes with a sterile pipette tip. After 24 h incubation in DMEM with 0.5% serum, migration was quantified by counting cell numbers migrating from the wound edge. Each determination represents the average of four individual experiments, and error bars represent SE. Ps were calculated by Student’s t test.

In-Blot Phosphatase Assay.

PTEN dephosphorylation of FAK and p130Cas was examined by an in-blot phosphatase assay as described (21). Briefly, histidine-tagged PTEN (His6-PTEN) was generated by inserting full-length PTEN cDNA into the pQE30 vector (Qiagen, Valencia, CA). The protein was purified using Ni-NTA beads (Qiagen) under denaturing conditions and then renatured by sequential dilution and concentration in renaturation buffer[PBS (pH 7.0) containing 2 mm MgCl2, 0.5 mm phenylmethanesulfonyl fluoride, 0.005% Tween 20, 10 mm DTT, and protease inhibitor mixture]. Purity (>90%) was confirmed by SDS-PAGE and Coomassie blue staining, and preparations were used within 1 h. U87MG cells that had spread on fibronectin for 2 h were lysed in RIPA buffer, and FAK and p130Cas were immunoprecipitated using either anti-FAK or anti-p130Cas antibodies, respectively. Equal amounts of immunoprecipitated proteins were subjected to 4–12% SDS-PAGE and electrotransferred to nitrocellulose. Blots were incubated with 20 μg/ml recombinant His6-PTEN in 50 mm HEPES buffer (pH 7.0) containing 10 mm MgCl2, 10 mm DTT at 30°C for 1 h. Phosphoproteins were detected with anti-phosphotyrosine antibody.

Cell Growth.

After puromycin selection for transfected cells, cells were harvested by trypsin-EDTA. Transfected U87MG cells (5 × 104 cells) were cultured in six-well plates with 10% serum in DMEM. Cell number was counted after 24 h incubation by harvesting the cells using trypsin-EDTA and counting in a hemocytometer. [3H]Thymidine incorporation was measured using 1 μCi/ml [3H]thymidine (Amersham Pharmacia Biotech) for 1 h after 24 h incubation. The cells were then washed three times with ice-cold PBS and detached from the plates with trypsin-EDTA. The radioactivity of samples was determined with a liquid scintillation counter. In situ detection of cells undergoing apoptosis was done by fluorescent-conjugated annexin V staining (Clontech) as described (41) or by TUNEL assay (Boehringer Mannheim), which was detected by anti-fluorescein antibody conjugated with horseradish peroxidase according to the manufacturer’s protocol.

PTEN Inhibits Cell Invasion, and Its Activity Is Phosphatase Dependent.

U87MG cells (which have a PTEN frameshift mutation at codon 54) were cotransfected with GFP-PTEN (wild-type), GFP-PTEN phosphatase-inactivating mutant (C124A), the control mutant GFP-PTEN (G129E), or GFP without insert, together with a plasmid encoding a puromycin resistance gene to select cells coexpressing the expression plasmids. After selection with puromycin for 3 days, cells were analyzed for PTEN expression by immunoblotting. As shown in Fig. 1, a Mr ∼82,000 band corresponding to GFP-PTEN was recognized by anti-PTEN antibody MT478 in wild-type or mutant PTEN-transfected cells. This antibody also recognized both recombinant full-length PTEN and endogenous PTEN in human foreskin fibroblasts (Fig. 1). PTEN protein migrated at Mr ∼55,000, in agreement with previous reports (3, 16). In contrast, no endogenous PTEN protein could be detected in U87MG cells transfected with GFP; the PTEN gene is mutated in this cell. We also determined the percentage of cells expressing GFP-PTEN by scoring for GFP-positive cells using a fluorescence microscope. The percentage of cells that were GFP positive after cotransfection with GFP, GFP-PTEN (wild type), GFP-PTEN (C124A), or GFP-PTEN (G129E) were 95 ± 2%, 91 ± 4%, 89 ± 4%, and 95 ± 1%, respectively (mean ± SE of four independent experiments).

To examine the effects of PTEN on cell invasion, U87MG cells were transfected with GFP plasmids either without insert or with wild-type PTEN or PTEN mutants and were selected by puromycin as described above. Because PTEN suppresses growth in cell lines missing the PTEN gene (Ref. 15; Fig. 6), cells were serum starved to suppress growth, thereby ruling out contributions of PTEN growth suppression on cell invasion assays. As shown in Fig. 2 A, cell invasion through membranes coated with Matrigel was decreased by 45% (55.6 ± 7.5% of control) in cells expressing wild-type PTEN and by 42% (58.0 ± 7.0% of control) in cells expressing the phosphatase-active mutant of PTEN (G129E), whereas no significant change was observed after transfection with the phosphatase-inactivated mutant of PTEN (C124A), indicating that the phosphatase activity of PTEN is required for inhibition of cell invasion. There were no significant differences in total cell numbers of each transfectant, which were determined by counting invading plus noninvading cells on Matrigel after 16 h incubation (data not shown). These results indicate that the suppression of cell invasion by PTEN was not due to PTEN effects on cell growth. The marked inhibition of cell invasion after PTEN expression was not accompanied by any change in cell viability (∼95% viability by trypan blue assay with or without PTEN expression).

We also examined whether tyrosine phosphorylation levels of FAK correlated with the effects of PTEN on cell invasion. Immunoprecipitation of FAK showed decreases in tyrosine phosphorylation of FAK by phosphatase-active forms of PTEN (wild type and G129E) but not by a phosphatase-inactive form of PTEN (C124A; Fig. 2,B), consistent with our previous experiments that FAK is dephosphorylated both in vivo and in vitro(21). The amount of FAK protein in each sample assessed by immunoblotting was similar (Fig. 2 B). Thus, PTEN inhibition of cell invasion is associated with FAK dephosphorylation.

FAK Overexpression Rescues PTEN Inhibition of Cell Invasion.

The correlation of FAK dephosphorylation with inhibition of cell invasion suggests that the inhibition of invasion may be a downstream event of FAK dephosphorylation by PTEN. Next, we tested whether FAK overexpression could attenuate the effects of PTEN on cell invasion. As shown in Fig. 3,A, overexpression of FAK resulted in an increase in total FAK protein levels and abrogated PTEN-induced down-regulation of FAK phosphorylation. We then examined the phosphorylation levels of p130Cas. Tyrosine phosphorylation of p130Cas was decreased in PTEN-expressing cells; however, it was increased after overexpression of FAK cotransfected with PTEN (Fig. 3,A), suggesting that p130Cas is located downstream of FAK. Although FAK overexpression alone had minimal to only very slightly positive effects on cell invasion (115.6 ± 2.3% of control), coexpression of FAK with PTEN effectively rescued the ability of cells to invade (99.5 ± 4.0% of control) (Fig. 3 B), suggesting that FAK is an important mediator of PTEN-induced inhibition of cell invasion.

p130Cas Overexpression Also Rescues PTEN Inhibition of Cell Invasion.

The adapter protein p130Cas is a putative downstream effector of FAK, and it is characteristically tyrosine phosphorylated after increases in FAK and c-Src kinase activity in response to attachment to the extracellular matrix (33, 34, 35). We therefore investigated whether p130Cas is also implicated in PTEN-induced inhibition of cell invasion as a downstream event of FAK dephosphorylation. Again, tyrosine phosphorylation levels of p130Cas were decreased in PTEN-expressing cells (Fig. 4,A). Both total protein and tyrosine phosphorylation levels of p130Cas were increased after p130Cas overexpression. The phosphorylation levels of FAK in p130Cas-overexpressing cells were not affected, suggesting that p130Cas phosphorylation was downstream of FAK. Although p130Cas overexpression alone had minimal effects on cell invasion (109.8 ± 4.9% of control), coexpression of p130Cas with PTEN effectively rescued the ability of cells to invade (increasing from 53.5 ± 6.5% to 96.5 ± 3.1% of control; Fig. 4 B). These results are consistent with a role for p130Cas in PTEN-induced inhibition of cell invasion as a downstream effector of FAK.

Direct Effects of PTEN on FAK but not on p130Cas.

Because our data suggested that p130Cas was located downstream of FAK, we next tested whether PTEN could directly dephosphorylate tyrosine-phosphorylated p130Casin vitro using an in-blot phosphatase assay, with FAK as a positive control (21). Tyrosine-phosphorylated FAK and p130Cas were immunoprecipitated from U87MG cells, electroblotted, and incubated with or without His6-PTEN (Fig. 5,A). Recombinant PTEN reduced tyrosine phosphorylation of FAK by 78% (22 ± 15% of control, three experiments). No dephosphorylation occurred when blots were incubated with control E. coli lysate (data not shown) or without PTEN (Fig. 5,A). In contrast, PTEN could not dephosphorylate p130Cas (Fig. 5 A).

We also tested for physical interactions of PTEN with p130Cas in living cells using a “trapping” mutant of PTEN (21, 42). Overexpression of a PTEN mutant with inactivated phosphatase activity (potentially able to bind and protect a substrate) resulted in elevated phosphorylation of HA-FAK in NIH 3T3 cells that had endogenous PTEN (Fig. 5,B), consistent with our previous report (21). Moreover, immunoprecipitated HA-FAK retained bound GFP-PTEN, indicating formation of a FAK-PTEN complex (Fig. 5,B). In control plasmid GFP (−) transfected cells, no association of GFP with HA-FAK could be seen (data not shown). In contrast, phosphorylation levels of p130Cas were not increased, and no association of GFP-PTEN with FLAG-Cas was observed when cells were transfected with the PTEN trapping mutant (Fig. 5 C). Thus, PTEN could not interact with p130Cas both in vitro and in vivo under conditions where FAK was readily dephosphorylated, indicating that p130Cas is not a physiological substrate for PTEN. Our results suggest that PTEN indirectly inhibits phosphorylation of p130Cas, e.g., through its effects on FAK.

PTEN Inhibition of Cell Migration Is Phosphatase Dependent, and Both FAK and p130Cas Overexpression Rescue PTEN Inhibition of Cell Migration.

We reported recently that PTEN overexpression reduces cell migration, whereas the expression of antisense PTEN enhances cell migration in stable transfectant lines of NIH 3T3 cells (21). In this study, we examined the effects of phosphatase mutants of PTEN and of FAK or p130Cas overexpression on PTEN suppression of cell migration. Phosphatase-active forms of PTEN significantly inhibited cell migration as measured by in vitro wound healing assays (Fig. 6,A). In contrast, the expression of the phosphatase-inactive form of PTEN had no significant effect on cell migration (Fig. 6,A). Next, either FAK or p130Cas was coexpressed with PTEN to test the involvement of FAK and p130Cas in PTEN-induced inhibition of cell migration. Both FAK and p130Cas overexpression individually could enhance U87MG cell migration (Fig. 6,B) as reported previously in other cell types (32, 36, 43, 44), in clear contrast to their minimal effects on cell invasion (Figs. 3 and 4). Furthermore, in PTEN-expressing cells, both FAK and p130Cas overexpression effectively reversed the reduction of cell migration by PTEN (Fig. 6 B), suggesting that both FAK and p130Cas are intermediates in PTEN-induced inhibition of cell migration; their effects on PTEN action were similar to the results of the cell invasion assay. Similar effects of FAK and p130Cas on PTEN-induced reduction of cell migration were observed in DBTRG-05 MG glioblastoma cells (data not shown).

FAK, but not p130Cas, Reverses Cell Growth Inhibition by PTEN.

Because both FAK and p130Cas could rescue the inhibition of cell migration and invasion by PTEN, we examined for effects of FAK and p130Cas overexpression on cell growth. Cells were cultured for 24 h after puromycin selection for transfected cells. Cell growth in cells transfected with phosphatase-active forms of PTEN was suppressed by >50% (wild type: reduced to 45 ± 6% of control; G129E: 48 ± 7% of control; Fig. 7,A). In contrast, no reduction was seen when cells were transfected with C124A, a phosphatase-inactive form of PTEN (Fig. 7,A), compatible with a previous report (15). Next, cells were cotransfected with either FAK or p130Cas. Neither FAK nor p130Cas alone had any significant effect on the number of cells after transfection (Fig. 7,B). In PTEN and FAK coexpressing cells, the cell number was significantly, but partially, increased compared with cells expressing PTEN alone (increasing from 44 ± 6% to 83 ± 4% of control, P < 0.005; Fig. 7,B). The changes in cell number were also confirmed by [3H]thymidine incorporation (wild-type PTEN transfection reduced incorporation to 65 ± 5% of control; increased to 84 ± 6% by coexpression of FAK). On the other hand, p130Cas overexpression did not result in significant changes in cell numbers of PTEN expressing cells (Fig. 7 B), suggesting that p130Cas is not involved in PTEN-induced regulation of cell growth.

FAK is also known to protect cells from apoptosis (45, 46). We next examined whether the marked suppression of cell growth after PTEN expression was related to apoptosis. U87MG cells were cotransfected with pHA262pur either without or with GFP or GFP-PTEN (wild type). After puromycin selection for transfectants, cells undergoing apoptosis were detected by annexin V staining or TUNEL assays. The percentages of cells that were annexin V positive were very low without any treatment: 2.1 ± 0.5% (nontransfected cells), 1.8 ± 0.7% (GFP), and 2.2 ± 0.7% (GFP-PTEN), respectively (mean ± SE of three independent experiments), consistent with the results of trypan blue assays. In contrast, positive cells were increased to 74 ± 3% (nontransfected cells), 80 ± 6% (GFP), and 78 ± 7% (GFP-PTEN), respectively, after 24-h UV irradiation (200 mJ/cm2). The TUNEL assay also showed similar percentages of positive cells; there were no significant differences between nontransfected, GFP, and GFP-PTEN-transfected cells. Thus, apoptosis was not involved in the PTEN-mediated growth suppression observed in U87MG cells under standard cell culture conditions.

The PTEN gene is deleted or mutated in ∼45% of endometrial cancers, ∼30% of glioblastomas, and at lower frequencies in a wide range of other human tumors. Recent reports that reexpression of PTEN in human glioma cell lines with mutated PTEN alleles suppresses cell growth and tumorigenicity further established PTEN as a tumor suppressor (15, 16). However, studies characterizing the molecular mechanisms by which PTEN functions as a tumor suppressor have lagged considerably. In this study, we have: (a) established that PTEN can inhibit cell invasion in a phosphatase-dependent manner; (b) confirmed that PTEN can suppress cell growth and migration and established that the latter was also dependent on PTEN phosphatase; (c) demonstrated that overexpression of FAK can restore both FAK and p130Cas tyrosine phosphorylation, and that it can antagonize or reverse the inhibitory actions of PTEN on cell invasion, migration, and even partially on growth suppression; (d) established that p130Cas overexpression restores its own phosphorylation but not that of FAK and can antagonize effects of PTEN on cell invasion and migration, but not on cell growth; and (e) confirmed that PTEN can directly interact with FAK and reduce its tyrosine phosphorylation and established that, in contrast, PTEN does not interact with p130Cas and cannot dephosphorylate p130Cas directly. These effects are summarized in Fig. 8, and they suggest a working mechanistic scheme in which PTEN down-modulates FAK phosphorylation and suppresses all three biological activities. In contrast, p130Cas acts downstream of FAK, because its overexpression affects neither FAK phosphorylation nor growth, and it could not be shown to interact with PTEN either in cells or in vitro.

Our studies provide, to our knowledge, the first evidence that PTEN can restrain the capacity of tumor cells to invade through a basement membrane matrix. Its inhibitory activity required an intact phosphatase domain. PTEN inhibition of tumor cell invasion could be reversed by intracellular overexpression of FAK at levels that restored its initially PTEN-suppressed tyrosine phosphorylation. We speculate that overexpressing FAK may enhance its phosphorylation by increasing the total amounts of FAK available for phosphorylation and out-competing the phosphatase activity of PTEN. It was reported recently that PTEN dephosphorylates phosphatidylinositol 3,4,5-trisphosphate (47, 48) and decreases Shc tyrosine phosphorylation levels (49). It is therefore also possible that overexpression of FAK might play a dominant-negative role in PTEN growth suppression by substrate competition. In addition, we reported previously that cell migration was inhibited in NIH 3T3 cells overexpressing PTEN (21). In this report, we established that an intact PTEN phosphatase domain was needed for this activity, and moreover that FAK overexpression could also reverse PTEN inhibition of migration on a planar surface in two glioma cell lines. Although our similar findings in the invasion assay must be evaluated with caution because they are based on results from a single glioma cell line, rescue of tumor cells by FAK transfection and phosphorylation from PTEN-induced inhibition of tumor cell migration and invasion suggest that PTEN suppresses these activities through dephosphorylating FAK.

General proposed functions of FAK include roles in focal adhesion assembly, cell adhesion, and cell migration. Overexpression of dominant-negative FAK causes a transient reduction in cell spreading (50), consistent with our previous report that PTEN-induced FAK dephosphorylation leads to suppression of cell spreading and impaired focal adhesion and cytoskeletal formation (21). FAK is also an important regulator of integrin-mediated cell migration events. FAK expression is enhanced in rapidly migrating cells during wound healing (51, 52), and FAK overexpression stimulates cell migration (44). Cells in which FAK is inhibited by a tyrosine kinase inhibitor (52), cells from FAK-deficient (gene knockout) mice (43), and cells overexpressing the FAK COOH-terminal domain (53) all exhibit decreased cell migration. Our findings suggest a function for PTEN in down-modulating these FAK activities by dephosphorylation.

Recently, p130Cas has been reported to be a mediator of FAK-promoted cell migration (32, 36). We have shown that p130Cas is also dephosphorylated as well as FAK by PTEN expression (21). We established in this report that either FAK or p130Cas overexpression could induce rephosphorylation of p130Cas and could effectively rescue PTEN-induced inhibition of both cell migration and invasion. p130Cas is a signal transduction protein that binds directly to FAK and is phosphorylated upon cell adhesion to extracellular matrix proteins in a FAK- and Src-dependent manner (33, 34, 35, 54). The proline-rich region of FAK spanning amino acids 711–717 has been demonstrated to be a binding site for the SH3 domain of p130Cas(55). We have been unable to detect direct dephosphorylation of p130Cas by PTEN using an in-blot phosphatase assay under conditions where FAK is readily dephosphorylated, nor could we detect physical association of PTEN with p130Cas. These findings collectively suggest that p130Cas is a downstream effector in a signal transduction pathway leading to cell migration and invasion after PTEN dephosphorylation of FAK (Fig. 8). The findings that both p130Cas and FAK were rephosphorylated after overexpression of FAK, but that p130Cas overexpression resulted in only its own phosphorylation without affecting the phosphorylation level of FAK also indicates that rescue by p130Cas is a downstream event of FAK. Thus, the signaling pathways of PTEN modulating cell invasion, migration, and growth appear to be mediated by distinct downstream pathways that diverge at the level of FAK (Fig. 8). The evidence that both FAK and p130Cas play important roles not only in cell migration but also in cell invasion suggests that FAK/p130Cas activation may contribute to the invasiveness of tumors. Thus, the elevated FAK observed in transformed cells (56, 57, 58) may form complexes with p130Cas, leading to invasion events involved in the dissemination of tumors.

In this study, manipulating the amounts of phosphorylated FAK could also antagonize the biological effects of PTEN on cell growth. Although overexpression of FAK rescued the action of PTEN on cell invasion, migration, and even partially on growth suppression, p130Cas did not affect growth, suggesting that the cell growth-suppressive effect of PTEN is mediated by FAK and downstream effectors other than p130Cas. Activation and autophosphorylation of FAK in response to integrin binding to extracellular matrix leads to its binding to a number of intracellular signaling molecules besides p130Cas including Src, Grb2, and phosphatidylinositol 3-kinase (28, 29, 59). FAK/Src association can lead to activation of the MAP kinase pathway through Grb2 binding to FAK (28, 30), although other mechanisms of integrin-mediated MAP kinase activation also exist (60, 61). Cell adhesion also promotes the association of FAK with phosphatidylinositol 3-kinase (59), which has been implicated in integrin-mediated cell survival through the proto-oncogene Akt and MAP kinase activation (62, 63). The requirement for phosphatase activity in PTEN-mediated growth suppression (Ref. 15 and this study) strongly suggests that the dephosphorylation of a substrate(s) of PTEN such as FAK is critical for growth. Recently, however, phosphatidylinositol 3,4,5-trisphosphate, which is produced by phosphatidylinositol 3-kinase and can activate Akt (64), has also been identified as a substrate of PTEN besides FAK (47, 48). PTEN also decreases phosphorylation levels of Shc in response to growth factor receptor activation and suppresses MAP kinase activity (49). Taken together with our results that PTEN can directly dephosphorylate FAK, but also that FAK rephosphorylation could rescue only about one-half of the PTEN-induced suppression of cell growth, these findings collectively suggest that other pathway(s) independent of FAK, such as PTEN dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate or Shc, may be also involved in this signaling. Therefore, PTEN may regulate cell growth by dephosphorylating plural targets (Fig. 8).

Most types of normal cells require attachment to extracellular matrix components to be able to grow. The loss of integrin-mediated cell-matrix contact induces apoptosis, termed anoikis, in many nontransformed cell types (65, 66, 67, 68). In contrast, the ability of malignant cells to proliferate in the absence of adhesion, termed anchorage independence of growth, correlates closely with tumorigenicity. Recent studies have implicated FAK in this cell survival. Inhibition of FAK in fibroblasts and endothelial cells results in growth suppression (53) and apoptosis (46), although FAK may not mediate survival in all cases (69). Conversely, overexpression of activated FAK can rescue MDCK cells from anoikis (45). In one case, activated FAK has even been reported to induce anchorage-independent growth and tumor formation by MDCK cells (45). These results provide evidence that FAK is an important mediator of integrin-mediated survival signals. In fact, several studies have established that levels of FAK expression are often increased in proliferating cells or advanced cancers (56, 57, 58). Interestingly, although PTEN inhibits FAK and suppresses cell growth, we could not detect PTEN-induced apoptosis in this study. PTEN might affect tumor progression by its role in down-regulating cell invasion through basement membranes by changes in focal adhesion or cytoskeletal formation, because mutation or decreased expression of PTEN is associated with tumor progression in certain cancers (6, 19, 20). Moreover, PTEN inactivation has been shown recently to enhance the ability of ES cells to grow in an anchorage-independent manner (70).

In summary, we have shown that PTEN restoration in glioblastoma cells with mutated PTEN alleles results in inhibition of cell invasion, migration, and growth in a process that involves phosphorylation of FAK. Alterations in p130Cas were also implicated in PTEN inhibition of cell invasion and migration but not in its effects on cell growth, suggesting that there are different downstream signaling pathways that diverge at the level of FAK. As documented in this study, such cell surface roles of PTEN appear to distinguish it from other tumor suppressors, such as p53 or Rb, which typically function in the nucleus (71). Recent studies indicate a role for PTEN inactivation in tumor progression, and our findings suggest that potential therapeutic targets of antimetastatic approaches may include FAK and p130Cas.

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

                  
3

The abbreviations used are: PTEN, phosphatase and tensin homologue deleted on chromosome 10; FAK, focal adhesion kinase; p130Cas, p130 Crk-associated substrate; MAP, mitogen-activated protein; GFP, green fluorescent protein; HA, hemagglutinin; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling.

Fig. 1.

Expression of PTEN and its mutants in U87MG cells. Empty GFP vector (control) or vector containing wild-type PTEN (wt) or mutants (C124A and G129E) were transfected into U87MG cells and then selected with puromycin as described in “Materials and Methods.” Equal amounts of total cell lysates (30 μg) were analyzed by anti-PTEN MT478 antibody. As positive controls, recombinant full-length PTEN (10 ng, rPTEN) and total lysate of human foreskin fibroblasts (30 μg, HFF) were also analyzed. Black arrowhead, GFP-PTEN; white arrowhead, recombinant or endogenous PTEN.

Fig. 1.

Expression of PTEN and its mutants in U87MG cells. Empty GFP vector (control) or vector containing wild-type PTEN (wt) or mutants (C124A and G129E) were transfected into U87MG cells and then selected with puromycin as described in “Materials and Methods.” Equal amounts of total cell lysates (30 μg) were analyzed by anti-PTEN MT478 antibody. As positive controls, recombinant full-length PTEN (10 ng, rPTEN) and total lysate of human foreskin fibroblasts (30 μg, HFF) were also analyzed. Black arrowhead, GFP-PTEN; white arrowhead, recombinant or endogenous PTEN.

Close modal
Fig. 2.

Cell invasion assay in wild-type or mutant PTEN-expressing U87MG cells. In A, cell invasion through Matrigel basement membrane extract was analyzed using a modified Boyden chamber as described in “Materials and Methods.” Cells that invaded to the lower surface of the membrane were fixed and stained. Mean cell counts from at least 10 fields and four experiments are shown. Bars, SE. †, P < 0.005 versus control. In B, lysates from each U87MG cell transfectant were analyzed for tyrosine phosphorylation (pTyr) of endogenous FAK. Lysates were immunoprecipitated with FAK (IP:FAK) and immunoblotted with anti-phosphotyrosine (upper panel) or anti-FAK (lower panel).

Fig. 2.

Cell invasion assay in wild-type or mutant PTEN-expressing U87MG cells. In A, cell invasion through Matrigel basement membrane extract was analyzed using a modified Boyden chamber as described in “Materials and Methods.” Cells that invaded to the lower surface of the membrane were fixed and stained. Mean cell counts from at least 10 fields and four experiments are shown. Bars, SE. †, P < 0.005 versus control. In B, lysates from each U87MG cell transfectant were analyzed for tyrosine phosphorylation (pTyr) of endogenous FAK. Lysates were immunoprecipitated with FAK (IP:FAK) and immunoblotted with anti-phosphotyrosine (upper panel) or anti-FAK (lower panel).

Close modal
Fig. 3.

FAK reversal of PTEN-induced reduction of cell invasion. In A, lysates from each U87MG transfectant were immunoprecipitated with either FAK (IP:FAK) or p130Cas (IP:p130Cas) and immunoblotted with anti-phosphotyrosine (pTyr), anti-FAK, or anti-p130Cas. In B, cell invasion through Matrigel was analyzed using a modified Boyden chamber as described in “Materials and Methods.” Cells that invaded through the basement membrane extract to the lower surface of the membrane were fixed, stained, and counted from at least 10 fields and four experiments. Bars, SE. †, P < 0.005 versus control.

Fig. 3.

FAK reversal of PTEN-induced reduction of cell invasion. In A, lysates from each U87MG transfectant were immunoprecipitated with either FAK (IP:FAK) or p130Cas (IP:p130Cas) and immunoblotted with anti-phosphotyrosine (pTyr), anti-FAK, or anti-p130Cas. In B, cell invasion through Matrigel was analyzed using a modified Boyden chamber as described in “Materials and Methods.” Cells that invaded through the basement membrane extract to the lower surface of the membrane were fixed, stained, and counted from at least 10 fields and four experiments. Bars, SE. †, P < 0.005 versus control.

Close modal
Fig. 4.

p130Cas reversal of PTEN-induced reduction of cell invasion. In A, lysates from each U87MG transfectant were immunoprecipitated with either FAK (IP:FAK) or p130Cas (IP:p130Cas) and immunoblotted with anti-phosphotyrosine (pTyr), anti-FAK, or anti-p130Cas. In B, cell invasion through Matrigel was analyzed using a modified Boyden chamber assay as described in “Materials and Methods.” Cells that migrated to the lower side of membrane were counted from at least 10 fields and four experiments. Bars, SE. †, P < 0.005 versus control.

Fig. 4.

p130Cas reversal of PTEN-induced reduction of cell invasion. In A, lysates from each U87MG transfectant were immunoprecipitated with either FAK (IP:FAK) or p130Cas (IP:p130Cas) and immunoblotted with anti-phosphotyrosine (pTyr), anti-FAK, or anti-p130Cas. In B, cell invasion through Matrigel was analyzed using a modified Boyden chamber assay as described in “Materials and Methods.” Cells that migrated to the lower side of membrane were counted from at least 10 fields and four experiments. Bars, SE. †, P < 0.005 versus control.

Close modal
Fig. 5.

Interactions of PTEN with FAK or p130Cas. A, direct tyrosine dephosphorylation of FAK but not p130Cas by PTEN using an in-blot phosphatase assay. FAK and p130Cas were immunoprecipitated (IP:FAK or IP:p130Cas) from U87MG cells. After SDS-PAGE and electroblotting, blots were incubated with or without His6-tagged recombinant PTEN (rPTEN, 20 μg/ml) for 1 h. Tyrosine phosphorylation was visualized with anti-phosphotyrosine antibody (pTyr). B and C, PTEN interaction with FAK but not with p130Cas in intact cells. NIH 3T3 cells transfected with tracer levels of either HA-FAK or FLAG-Cas (3 μg each) plus GFP-tagged wild-type PTEN or trapping mutant D92A (30 μg each) were immunoprecipitated with either anti-HA (IP:HA) or anti-FLAG (IP:FLAG) and immunoblotted for phosphotyrosine, FAK, p130Cas, or GFP; the original lysate was immunoblotted for GFP to confirm a similar amount of GFP.

Fig. 5.

Interactions of PTEN with FAK or p130Cas. A, direct tyrosine dephosphorylation of FAK but not p130Cas by PTEN using an in-blot phosphatase assay. FAK and p130Cas were immunoprecipitated (IP:FAK or IP:p130Cas) from U87MG cells. After SDS-PAGE and electroblotting, blots were incubated with or without His6-tagged recombinant PTEN (rPTEN, 20 μg/ml) for 1 h. Tyrosine phosphorylation was visualized with anti-phosphotyrosine antibody (pTyr). B and C, PTEN interaction with FAK but not with p130Cas in intact cells. NIH 3T3 cells transfected with tracer levels of either HA-FAK or FLAG-Cas (3 μg each) plus GFP-tagged wild-type PTEN or trapping mutant D92A (30 μg each) were immunoprecipitated with either anti-HA (IP:HA) or anti-FLAG (IP:FLAG) and immunoblotted for phosphotyrosine, FAK, p130Cas, or GFP; the original lysate was immunoblotted for GFP to confirm a similar amount of GFP.

Close modal
Fig. 6.

Migration of U87MG cells expressing wild-type or mutant PTEN and modulation by FAK or p130Cas co-expression. In A, in vitro “scratch” wounds were created by scraping confluent cell monolayers in fibronectin-coated dishes. After 24 h, cell numbers migrating from the wound edge were counted. Bars, SE from four independent experiments. †, P < 0.0005 versus control. In B, the effects of FAK or p130Cas co-expression with PTEN on cell migration were analyzed by in vitro wound healing assay as described above. †, P < 0.0005; ‡, P < 0.001 versus control.

Fig. 6.

Migration of U87MG cells expressing wild-type or mutant PTEN and modulation by FAK or p130Cas co-expression. In A, in vitro “scratch” wounds were created by scraping confluent cell monolayers in fibronectin-coated dishes. After 24 h, cell numbers migrating from the wound edge were counted. Bars, SE from four independent experiments. †, P < 0.0005 versus control. In B, the effects of FAK or p130Cas co-expression with PTEN on cell migration were analyzed by in vitro wound healing assay as described above. †, P < 0.0005; ‡, P < 0.001 versus control.

Close modal
Fig. 7.

Growth-suppressive effect of PTEN in U87MG cells and modulation by FAK or p130Cas coexpression. In A, empty vector (control) or vector containing either wild-type or mutant PTEN was transfected in U87MG cells. After selection with puromycin, an equal number of cells were plated and cultured in 10% serum-containing medium for 24 h. Cell numbers were determined in four independent experiments and pooled. Bars, SE. †, P < 0.0005 versus control. In B, the effects of FAK or p130Cas coexpression with PTEN on cell growth were analyzed as described above. Bars, SE. †, P < 0.0005; ‡, P < 0.05; *, P < 0.001 versus Lane 1.

Fig. 7.

Growth-suppressive effect of PTEN in U87MG cells and modulation by FAK or p130Cas coexpression. In A, empty vector (control) or vector containing either wild-type or mutant PTEN was transfected in U87MG cells. After selection with puromycin, an equal number of cells were plated and cultured in 10% serum-containing medium for 24 h. Cell numbers were determined in four independent experiments and pooled. Bars, SE. †, P < 0.0005 versus control. In B, the effects of FAK or p130Cas coexpression with PTEN on cell growth were analyzed as described above. Bars, SE. †, P < 0.0005; ‡, P < 0.05; *, P < 0.001 versus Lane 1.

Close modal
Fig. 8.

Summary and working model of the PTEN signaling network regulating cell migration, invasion, and growth. Extracellular matrix stimulation of cells promotes integrin clustering and signaling accompanied by local accumulation of a number of focal adhesion and signal transduction molecules, including FAK and p130Cas. These multimolecular complexes are involved in focal adhesion formation, cell migration, and cell invasion and potentially in cell growth modulation. PTEN dephosphorylates FAK directly and down-regulates cell migration, invasion, and growth; each of these effects can be antagonized by overexpression of FAK. However, although PTEN expression also results in decreased p130Cas phosphorylation, this protein may be involved in only cell migration and invasion and not in PTEN effects on cell growth. In addition, other pathways involving phosphatidylinositol 3,4,5-trisphosphate, Shc, and MAP kinase can also be modulated by PTEN in an increasingly complex network of signal transduction affecting cell growth and apoptosis (47,48 49). PI3′K, phosphatidylinositol [3′]-kinase; PIP3, phosphatidylinositol 3,4,5-trisphosphate; MAPK, extracellular signal-regulated kinase mitogen-activated protein kinase.

Fig. 8.

Summary and working model of the PTEN signaling network regulating cell migration, invasion, and growth. Extracellular matrix stimulation of cells promotes integrin clustering and signaling accompanied by local accumulation of a number of focal adhesion and signal transduction molecules, including FAK and p130Cas. These multimolecular complexes are involved in focal adhesion formation, cell migration, and cell invasion and potentially in cell growth modulation. PTEN dephosphorylates FAK directly and down-regulates cell migration, invasion, and growth; each of these effects can be antagonized by overexpression of FAK. However, although PTEN expression also results in decreased p130Cas phosphorylation, this protein may be involved in only cell migration and invasion and not in PTEN effects on cell growth. In addition, other pathways involving phosphatidylinositol 3,4,5-trisphosphate, Shc, and MAP kinase can also be modulated by PTEN in an increasingly complex network of signal transduction affecting cell growth and apoptosis (47,48 49). PI3′K, phosphatidylinositol [3′]-kinase; PIP3, phosphatidylinositol 3,4,5-trisphosphate; MAPK, extracellular signal-regulated kinase mitogen-activated protein kinase.

Close modal
1
Li J., Yen C., Liaw D., Podsypanina K., Bose S., Wang S. I., Puc J., Miliaresis C., Rodgers L., McCombie R., Bigner S. H., Giovanella B. C., Ittmann M., Tycko B., Hibshoosh H., Wigler M. H., Parsons R. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer.
Science (Washington DC)
,
275
:
1943
-1947,  
1997
.
2
Steck P. A., Pershouse M. A., Jasser S. A., Yung W. K., Lin H., Ligon A. H., Langford L. A., Baumgard M. L., Hattier T., Davis T., Frye C., Hu R., Swedlund B., Teng D. H., Tavtigian S. V. Identification of a candidate tumor suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers.
Nat. Genet.
,
15
:
356
-362,  
1997
.
3
Li D. M., Sun H. TEP1,encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor β.
Cancer Res.
,
57
:
2124
-2129,  
1997
.
4
Cairns P., Okami K., Halachmi S., Halachmi N., Esteller M., Herman J. G., Jen J., Isaacs W. B., Bova G. S., Sidransky D. Frequent inactivation of PTEN/MMAC1 in primary prostate cancer.
Cancer Res.
,
57
:
4997
-5000,  
1997
.
5
Gronbaek K., Zeuthen J., Guldberg P., Ralfkiaer E., Hou-Jensen K. Alterations of the MMAC1/PTEN gene in lymphoid malignancies.
Blood
,
91
:
4388
-4390,  
1998
.
6
Guldberg P., thor Straten P., Birck A., Ahrenkiel V., Kirkin A. F., Zeuthen J. Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event in malignant melanoma.
Cancer Res.
,
57
:
3660
-3663,  
1997
.
7
Rhei E., Kang L., Bogomolniy F., Federici M. G., Borgen P. I., Boyd J. Mutation analysis of the putative tumor suppressor gene PTEN/MMAC1 in primary breast carcinomas.
Cancer Res.
,
57
:
3657
-3659,  
1997
.
8
Risinger J. I., Hayes A. K., Berchuck A., Barrett J. C. PTEN/MMAC1 mutations in endometrial cancers.
Cancer Res.
,
57
:
4736
-4738,  
1997
.
9
Tashiro H., Blazes M. S., Wu R., Cho K. R., Bose S., Wang S. I., Li J., Parsons R., Ellenson L. H. Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies.
Cancer Res.
,
57
:
3935
-3940,  
1997
.
10
Teng D. H-F, Hu R., Lin H., Davis T., Iliev D., Frye C., Swedlund B., Hansen K. L., Vinson V. L., Gumpper K. L., Ellis L., El-Naggar A., Frazier M., Jasser S., Langford L. A., Lee J., Mills G. B., Pershouse M. A., Pollack R. E., Tornos C., Troncoso P., Yung W. K. A., Fujii G., Berson A., Bookstein R., Bolen J. B., Tavtigian S. V., Steck P. A. MMAC1/PTEN mutations in primary tumor specimens and tumor cell lines.
Cancer Res.
,
57
:
5221
-5225,  
1997
.
11
Wang S. I., Puc J., Li J., Bruce J. N., Cairns P., Sidransky D., Parsons R. Somatic mutations of PTEN in glioblastoma multiforme.
Cancer Res.
,
57
:
4183
-4186,  
1997
.
12
Olschwang S., Serova-Sinilnikova O. M., Lenoir G. M., Thomas G. PTEN germ-line mutations in juvenile polyposis coli.
Nat. Genet.
,
18
:
12
-14,  
1998
.
13
Liaw D., Marsh D. J., Li J., Dahia P. L., Wang S. I., Zheng Z., Bose S., Call K. M., Tsou H. C., Peacocke M., Eng C., Parsons R. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome.
Nat. Genet.
,
16
:
64
-67,  
1997
.
14
Marsh D. J., Dahia P. L., Zheng Z., Liaw D., Parsons R., Gorlin R. J., Eng C. Germline mutations in PTEN are present in Bannayan-Zonana syndrome.
Nat. Genet.
,
16
:
333
-334,  
1997
.
15
Furnari F. B., Lin H., Huang H. S., Cavenee W. K. Growth suppression of glioma cells by PTEN requires a functional phosphatase catalytic domain.
Proc. Natl. Acad. Sci. USA
,
94
:
12479
-12484,  
1997
.
16
Cheney I. W., Johnson D. E., Vaillancourt M. T., Avanzini J., Morimoto A., Demers G. W., Wills K. N., Shabram P. W., Bolen J. B., Tavtigian S. V., Bookstein R. Suppression of tumorigenicity of glioblastoma cells by adenovirus-mediated MMAC1/PTEN gene transfer.
Cancer Res.
,
58
:
2331
-2334,  
1998
.
17
Myers M. P., Stolarov J. P., Eng C., Li J., Wang S. I., Wigler M. H., Parsons R., Tonks N. K. P-TEN,the tumor suppressor from human chromosome 10q23, is a dual-specificity phosphatase.
Proc. Natl. Acad. Sci. USA
,
94
:
9052
-9057,  
1997
.
18
Li L., Ernsting B. R., Wishart M. J., Lohse D. L., Dixon J. E. A family of putative tumor suppressors is structurally and functionally conserved in humans and yeast.
J. Biol. Chem.
,
272
:
29403
-29406,  
1997
.
19
Rasheed B. K., Stenzel T. T., McLendon R. E., Parsons R., Friedman A. H., Friedman H. S., Bigner D. D., Bigner S. H. PTEN gene mutations are seen in high-grade but not in low-grade gliomas.
Cancer Res.
,
57
:
4187
-4190,  
1997
.
20
Suzuki H., Freije D., Nusskern D. R., Okami K., Cairns P., Sidransky D., Isaacs W. B., Bova G. S. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues.
Cancer Res.
,
58
:
204
-209,  
1998
.
21
Tamura M., Gu J., Matsumoto K., Aota S., Parsons R., Yamada K. M. Inhibition of cell migration, spreading, and focal adhesions by tumor suppressor PTEN.
Science (Washington DC)
,
280
:
1614
-1617,  
1998
.
22
Hanks S. K., Polte T. R. Signaling through focal adhesion kinase.
Bioessays
,
19
:
137
-145,  
1997
.
23
Schlaepfer D. D., Hunter T. Integrin signaling and tyrosine phosphorylation: just the FAKs?.
Trends Cell Biol.
,
8
:
151
-157,  
1998
.
24
Guan J. L. Focal adhesion kinase in integrin signaling.
Matrix Biol.
,
16
:
195
-200,  
1997
.
25
Clark E. A., Brugge J. S. Integrins and signal transduction pathways: the road taken.
Science (Washington DC)
,
268
:
233
-239,  
1995
.
26
Ilic D., Damsky C., Yamamoto T. Focal adhesion kinase: at the crossroads of signal transduction.
J. Cell Sci.
,
110
:
401
-407,  
1997
.
27
Yamada K. M., Geiger B. Molecular interactions in cell adhesion complexes.
Curr. Opin. Cell Biol.
,
9
:
76
-85,  
1997
.
28
Schlaepfer D. D., Hanks S. K., Hunter T., van der Geer P. Integrin-mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesion kinase.
Nature (Lond.)
,
372
:
786
-791,  
1994
.
29
Schaller M. D., Hildebrand J. D., Shannon J. D., Fox J. W., Vines R. R., Parsons J. T. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src.
Mol. Cell. Biol.
,
14
:
1680
-1688,  
1994
.
30
Schlaepfer D. D., Hunter T. Focal adhesion kinase overexpression enhances ras-dependent integrin signaling to ERK2/mitogen-activated protein kinase through interactions with and activation of c-Src.
J. Biol. Chem.
,
272
:
13189
-13195,  
1997
.
31
Schlaepfer D. D., Jones K. C., Hunter T. Multiple Grb2-mediated integrin-stimulated signaling pathways to ERK2/mitogen-activated protein kinase: summation of both c-Src- and focal adhesion kinase-initiated tyrosine phosphorylation events.
Mol. Cell. Biol.
,
18
:
2571
-2585,  
1998
.
32
Cary L. A., Han D. C., Polte T. R., Hanks S. K., Guan J. L. Identification of p130Cas as a mediator of focal adhesion kinase-promoted cell migration.
J. Cell Biol.
,
140
:
211
-221,  
1998
.
33
Tachibana K., Urano T., Fujita H., Ohashi Y., Kamiguchi K., Iwata S., Hirai H., Morimoto C. Tyrosine phosphorylation of Crk-associated substrates by focal adhesion kinase. A putative mechanism for the integrin-mediated tyrosine phosphorylation of Crk-associated substrates.
J. Biol. Chem.
,
272
:
29083
-29090,  
1997
.
34
Harte M. T., Hildebrand J. D., Burnham M. R., Bouton A. H., Parsons J. T. p130Cas,a substrate associated with v-Src and v-Crk, localizes to focal adhesions and binds to focal adhesion kinase.
J. Biol. Chem.
,
271
:
13649
-13655,  
1996
.
35
Vuori K., Hirai H., Aizawa S., Ruoslahti E. Introduction of p130Cas signaling complex formation upon integrin-mediated cell adhesion: a role for Src family kinases.
Mol. Cell. Biol.
,
16
:
2606
-2613,  
1996
.
36
Klemke R. L., Leng J., Molander R., Brooks P. C., Vuori K., Cheresh D. A. CAS/Crk coupling serves as a “molecular switch” for induction of cell migration.
J. Cell Biol.
,
140
:
961
-972,  
1998
.
37
Nakamoto T., Sakai R., Honda H., Ogawa S., Ueno H., Suzuki T., Aizawa S., Yazaki Y., Hirai H. Requirements for localization of p130Cas to focal adhesions.
Mol. Cell. Biol.
,
17
:
3884
-3897,  
1997
.
38
Lacalle R. A., Pulido D., Vara J., Zalacain M., Jimenez A. Molecular analysis of the pac gene encoding a puromycin N-acetyl transferase from Streptomyces alboniger.
Gene (Amst.)
,
79
:
375
-380,  
1989
.
39
LaFlamme S. E., Thomas L. A., Yamada S. S., Yamada K. M. Single subunit chimeric integrins as mimics and inhibitors of endogenous integrin functions in receptor localization, cell spreading and migration, and matrix assembly.
J. Cell Biol.
,
126
:
1287
-1298,  
1994
.
40
De , Nichilo M. O., Yamada K. M. Integrin αvβ5-dependent serine phosphorylation of paxillin in cultured human macrophages adherent to vitronectin.
J. Biol. Chem.
,
271
:
11016
-11022,  
1996
.
41
Martin S. J., Reutelingsperger C. P., McGahon A. J., Rader J. A., van Schie R. C., LaFace D. M., Green D. R. Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl.
J. Exp. Med.
,
182
:
1545
-1556,  
1995
.
42
Flint A. J., Tiganis T., Barford D., Tonks N. K. Development of “substrate-trapping” mutants to identify physiological substrates of protein tyrosine phosphatases.
Proc. Natl. Acad. Sci. USA
,
94
:
1680
-1685,  
1997
.
43
Ilic D., Furuta Y., Kanazawa S., Takeda N., Sobue K., Nakatsuji N., Nomura S., Fujimoto J., Okada M., Yamamoto T. Reduced cell motility and enhanced focal adhesion contact formation in cells from FAK-deficient mice.
Nature (Lond.)
,
377
:
539
-544,  
1995
.
44
Cary L. A., Chang J. F., Guan J-L. Stimulation of cell migration by overexpression of focal adhesion kinase and its association with Src and Fyn.
J. Cell Sci.
,
109
:
1787
-1794,  
1996
.
45
Frisch S. M., Vuori K., Ruoslahti E., Chan-Hui P. Y. Control of adhesion-dependent cell survival by focal adhesion kinase.
J. Cell Biol.
,
134
:
793
-799,  
1996
.
46
Hungerford J. E., Compton M. T., Matter M. L., Hoffstrom B. G., Otey C. A. Inhibition of pp125FAK in cultured fibroblasts results in apoptosis.
J. Cell Biol.
,
135
:
1383
-1390,  
1996
.
47
Maehama T., Dixon J. E. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate.
J. Biol. Chem.
,
273
:
13375
-13378,  
1998
.
48
Stambolic V., Suzuki A., de la Pompa J. L., Brothers G. M., Mirtsos C., Sasaki T., Ruland J., Penninger J. M., Siderovski D. P., Mak T. W. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN.
Cell
,
95
:
29
-39,  
1998
.
49
Gu J., Tamura M., Yamada K. M. Tumor suppressor PTEN inhibits integrin- and growth factor-mediated MAP kinase signaling pathways.
J. Cell Biol.
,
143
:
1375
-1383,  
1998
.
50
Richardson A., Parsons T. A mechanism for regulation of the adhesion-associated protein tyrosine kinase pp125FAK.
Nature (Lond.)
,
380
:
538
-540,  
1996
.
51
Gates R. E., King L. E., Jr., Hanks S. K., Nanney L. B. Potential role for focal adhesion kinase in migrating and proliferating keratinocytes near epidermal wounds and in culture.
Cell Growth Differ.
,
5
:
891
-899,  
1994
.
52
Romer L. H., McLean N., Turner C. E., Burridge K. Tyrosine kinase activity, cytoskeletal organization, and motility in human vascular endothelial cells.
Mol. Biol. Cell
,
5
:
349
-361,  
1994
.
53
Gilmore A. P., Romer L. H. Inhibition of focal adhesion kinase (FAK) signaling in focal adhesions decreases cell motility and proliferation.
Mol. Biol. Cell
,
7
:
1209
-1224,  
1996
.
54
Nojima Y., Morino N., Mimura T., Hamasaki K., Furuya H., Sakai R., Sato T., Tachibana K., Morimoto C., Yazaki Y., Hirai H. Integrin-mediated cell adhesion promotes tyrosine phosphorylation of p130Cas, a Src homology 3-containing molecule having multiple Src homology 2-binding motifs.
J. Biol. Chem.
,
270
:
15398
-15402,  
1995
.
55
Polte T. R., Hanks S. K. Interaction between focal adhesion kinase and Crk-associated tyrosine kinase substrate p130Cas.
Proc. Natl. Acad. Sci. USA
,
92
:
10678
-10682,  
1995
.
56
Tremblay L., Hauck W., Nguyen L. T., Allard P., Landry F., Chapdelaine A., Chevalier S. Regulation and activation of focal adhesion kinase and paxillin during the adhesion, proliferation, and differentiation of prostatic epithelial cells in vitro and in vivo.
Mol. Endocrinol.
,
10
:
1010
-1020,  
1996
.
57
Weiner T. M., Liu E. T., Craven R. J., Cance W. G. Expression of focal adhesion kinase gene and invasive cancer.
Lancet
,
342
:
1024
-1025,  
1993
.
58
Owens L. V., Xu L., Craven R. J., Dent G. A., Weiner T. M., Kornberg L., Liu E. T., Cance W. G. Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors.
Cancer Res.
,
55
:
2752
-2755,  
1995
.
59
Chen H. C., Guan J. L. Association of focal adhesion kinase with its potential substrate phosphatidylinositol 3-kinase.
Proc. Natl. Acad. Sci. USA
,
91
:
10148
-10152,  
1994
.
60
Wary K. K., Mainiero F., Isakoff S. J., Marcantonio E. E., Giancotti F. G. The adaptor protein Shc couples a class of integrins to the control of cell cycle progression.
Cell
,
87
:
733
-743,  
1996
.
61
Lin T. H., Aplin A. E., Shen Y., Chen Q., Schaller M., Romer L., Aukhil I., Juliano R. L. Integrin-mediated activation of MAP kinase is independent of FAK: evidence for dual integrin signaling pathways in fibroblasts.
J. Cell Biol.
,
136
:
1385
-1395,  
1997
.
62
Khwaja A., Rodriguez-Viciana P., Wennstrom S., Warne P. H., Downward J. Matrix adhesion and Ras transformation both activate a phosphoinositide 3-OH kinase and protein kinase B/Akt cellular survival pathway.
EMBO J.
,
16
:
2783
-2793,  
1997
.
63
King W. G., Mattaliano M. D., Chan T. O., Tsichlis P. N., Brugge J. S. Phosphatidylinositol 3-kinase is required for integrin-stimulated AKT and Raf-1/mitogen-activated protein kinase pathway activation.
Mol. Cell. Biol.
,
17
:
4406
-4418,  
1997
.
64
Downward J. Mechanisms and consequences of activation of protein kinase B/Akt.
Curr. Opin. Cell Biol.
,
10
:
262
-267,  
1998
.
65
Ruoslahti E., Reed J. C. Anchorage dependence, integrins, and apoptosis.
Cell
,
77
:
477
-478,  
1994
.
66
Schwartz M. A. Integrins,oncogenes, and anchorage independence.
J. Cell Biol.
,
139
:
575
-578,  
1997
.
67
Meredith J. E., Schwartz M. A. Integrins,adhesion and apoptosis.
Trends Cell Biol.
,
7
:
146
-150,  
1997
.
68
Frisch S. M., Ruoslahti E. Integrins and anoikis.
Curr. Opin. Cell Biol.
,
9
:
701
-706,  
1997
.
69
Xu L. H., Owens L. V., Sturge G. C., Yang X., Liu E. T., Craven R. J., Cance W. G. Attenuation of the expression of the focal adhesion kinase induces apoptosis in tumor cells.
Cell Growth Differ.
,
7
:
413
-418,  
1996
.
70
Di , Cristofano A., Pesce B., Cordon-Cardo C., Pandolfi P. P. Pten is essential for embryonic development and tumor suppression.
Nat. Genet.
,
19
:
348
-355,  
1998
.
71
Agarwal M. L., Taylor W. R., Chernov M. V., Chernova O. B., Stark G. R. The p53 network.
J. Biol. Chem.
,
273
:
1
-4,  
1998
.