• Keine Ergebnisse gefunden

Focal adhesion kinase facilitates platelet-derived growth factor-BB-stimulated ERK2 activation required for chemotaxis migration of vascular smooth muscle cells

N/A
N/A
Protected

Academic year: 2022

Aktie "Focal adhesion kinase facilitates platelet-derived growth factor-BB-stimulated ERK2 activation required for chemotaxis migration of vascular smooth muscle cells"

Copied!
8
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Focal Adhesion Kinase Facilitates Platelet-derived Growth Factor-BB-stimulated ERK2 Activation Required for

Chemotaxis Migration of Vascular Smooth Muscle Cells*

Received for publication, June 21, 2000, and in revised form, September 6, 2000 Published, JBC Papers in Press, September 20, 2000, DOI 10.1074/jbc.M005450200

Christof R. Hauck‡, Datsun A. Hsia, and David D. Schlaepfer§

From the Department of Immunology, The Scripps Research Institute, La Jolla, California 92037

The focal adhesion (FAK) non-receptor protein-tyro- sine kinase (PTK) links both extracellular matrix/inte- grin and growth factor stimulation to intracellular sig- nals promoting cell migration. Here we show that both transient and stable overexpression of the FAK C-termi- nal domain termed FRNK (FAK-related non-kinase) in- hibits serum and platelet-derived growth factor (PDGF)- BB-induced vascular smooth muscle cell (SMC) migration in wound healing andin vitroBoyden Cham- ber chemotaxis assays, respectively. Expression of FRNK, but not a point mutant of FRNK (FRNK L1034S), disrupted the formation of a complex containing both FAK and the activated PDGF-receptor and resulted in reduced tyrosine phosphorylation of endogenous FAK at the Tyr-397 binding site for Src family PTKs. As dem- onstrated using FAK-deficient and FAK-reconstituted fibroblasts, FAK positively contributed to PDGF-BB- stimulated ERK2/MAP kinase activity, and in SMCs, ERK2/MAP kinase activity was required for PDGF-BB- stimulated chemotaxis. Stable expression of FRNK but not FRNK L1034S expression in SMCs lowered the ex- tent and duration of stimulated ERK2/MAP kinase acti- vation at low but not at high PDGF-BB concentrations.

Importantly, stable expression of FRNK in SMCs did not affect SMC morphology or proliferation in culture. Be- cause the increased migration of vascular SMCs in re- sponse to extracellular matrix proteins and growth fac- tors contributes to neointima formation, our results show that FAK inhibition by FRNK expression may pro- vide a novel approach to regulate abnormal vascular SMC migrationin vivo.

Several vascular diseases result from neointima formation, a process that is characterized by the accumulation of vascular smooth muscle cells (SMCs)1and extracellular matrix (ECM)

proteins in the intima of blood vessels (1). Neointima formation is triggered upon damage to the endothelial lining by the local release of chemotactic cytokines or growth factors (2) and by increased production of ECM proteins (3). Because combina- tions of these factors can stimulate cell division, it was origi- nally hypothesized that neointimal hyperplasia resulted from enhanced SMC proliferation (4). However, further studies have shown that SMC migration also plays a significant role in neointima formation (5). Growth factors acting as chemotactic agents (6) and ECM molecules acting as haptotactic factors (7) can independently stimulate SMC migration. Therefore, a com- mon signaling component that coordinates both chemotactic and haptotactic cell migration events would be a promising target for intervention strategies.

Investigations of the molecular regulation of cell migration have demonstrated an important role for the focal adhesion kinase (FAK), a protein-tyrosine kinase (PTK) that localizes to cell-substratum contact sites also known as focal adhesions (for a review see Ref. 8). Genetic support for FAK in promoting cell motility comes from studies using FAK-null fibroblasts that exhibit refractory responses to normal motility-promoting stimuli (9). Importantly, these defects are rescued by stable re-expression of FAK in FAK-null cells (10, 11). Work from a number of laboratories using a variety of cell types has pro- vided evidence that FAK promotes cell migration potentially through the activation of multiple downstream targets such as Src family PTKs (10, 12) and phosphatidylinositol 3-kinase (13, 14), by the increased phosphorylation of p130Cas (15, 16) or paxillin adaptor proteins (17), or through the association with other signaling proteins such as Grb7 (18) and SHP-2 (19 –21).

Although no clear consensus has emerged on the molecular mechanism(s) of how FAK promotes cell migration, studies with FAK-null cells have shown that FAK is required for both integrin- and growth factor-stimulated cell motility (10, 12).

FAK activity is regulated by protein-tyrosine phosphatase action (16) and inhibited by the overexpression of the FAK C-terminal domain termed FRNK (FAK-related non-kinase) (22–24). Importantly, the regulated and autonomous expres- sion of FRNK during embryonic development is under the control of alternative intronic promoter region and therefore may function as an endogenous inhibitor of FAKin vivo(22, 25) In this study, we employed the transient and stable overex- pression of hemagglutinin (HA)-tagged FRNK to investigate the role of endogenous FAK in promoting platelet-derived

* This work was supported in part by Grant-in-Aid 9750682N from the American Heart Association and by Grant CA-75240 from the National Cancer Institute (to D. D. S.). This is manuscript number 13287-IMM from the Scripps Research Institute. The costs of publica- tion 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.

‡ Supported by a fellowship from the Deutsche Forschungsgemein- schaft (Ha2856/1-1).

§ To whom correspondence should be addressed: The Scripps Re- search Institute, Dept. of Immunology, IMM-26, 10550 North Torrey Pines Rd., La Jolla, CA 92037. Tel.: 858-784-8207; Fax: 858-784-8227;

E-mail: dschlaep@scripps.edu.

1The abbreviations used are: SMC, smooth muscle cell; ERK, extra- cellular signal-regulated kinase; MAP kinase, mitogen-activated ki- nase; MEK, MAP kinase/ERK kinase; ECM, extracellular matrix; FAK, focal adhesion kinase; FRNK, FAK-related non-kinase; GFP, green fluorescence protein; IP, immunoprecipitation; PDGF, platelet-derived

growth factor; PDGFr, PDGF receptor-␤; PTK, protein-tyrosine kinase;

HA, hemagglutinin; P-ERK, phosphorylated ERK/MAP kinase; FACS, fluorescence-activated cell sorting; mAb, monoclonal antibody; DMEM, Dulbecco’s modified Eagle’s medium; CS, calf serum; PBS, phosphate- buffered saline; SAP, stress-activated protein; BSA, bovine serum al- bumin; MBP, myelin basic protein; EGF, epidermal growth factor;

TRITC, tetramethylrhodamine isothiocyanate.

THEJOURNAL OFBIOLOGICALCHEMISTRY Vol. 275, No. 52, Issue of December 29, pp. 41092–41099, 2000

© 2000 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org

41092

First publ. in: Journal of Biological Chemistry 275 (2000), 52, pp. 41092–41099

Konstanzer Online-Publikations-System (KOPS) URL: http://www.ub.uni-konstanz.de/kops/volltexte/2007/4022/

URN: http://nbn-resolving.de/urn:nbn:de:bsz:352-opus-40228

(2)

growth factor (PDGF)-BB-stimulated vascular SMC migration.

Although FRNK expression did not affect PDGF receptor-␤ (PDGFr) activation or serum-stimulated cell proliferation, FRNK interfered with the PDGF-BB-stimulated recruitment of endogenous FAK to an activated PDGFr complex, decreased FAK phosphorylation at the Tyr-397 site, and reduced the extent and duration of PDGF-BB-stimulated ERK2/MAP ki- nase activation. Because pharmacological inhibition of ERK activity prevented efficient PDGF-BB-stimulated SMC cell mi- gration, our results point to an important role for FAK in relaying cell motility promoting signals from the PDGFr to MAP kinases.

EXPERIMENTAL PROCEDURES

Reagents, Plasmids, and Antibodies—Recombinant human PDGF- BB, the ERK kinase inhibitor PD98059, and the p38 kinase inhibitor SB203580 were purchased from Calbiochem (La Jolla, CA). Rat tail collagen was from Roche Molecular Biochemicals (Indianapolis, IN).

Eukaryotic expression plasmids pCDNA3.1 (Invitrogen, La Jolla, CA) encoding HA-tagged murine FRNK or FRNK L1034S (10) and vectors pEGFP and pEGFP-FRNK have been described (26). Affinity-purified antibodies to the FAK C-terminal domain (#5992) were used as de- scribed (27), to FAK N-terminal residues (A17) and to ERK2 (C14), were from Santa Cruz Biotechnologies (Santa Cruz, CA), and antibodies to phosphorylated ERK/MAP kinases (P-ERK) were from New England BioLabs (Beverly, MA). Phosphorylation and site-specific affinity puri- fied antibodies to FAK pTyr-397 were from BIOSOURCE International (Hopkinton, MA) and polyclonal rabbit antiserum to the PDGFr-␤was a generous gift from Jill Meisenhelder (Salk Institute). Monoclonal antibody (mAb) to phosphotyrosine (clone 4G10) was purchased from Upstate Biotechnology (Lake Placid, NY), mAb to vinculin (clone hVIN-1) was from Sigma, mAb (clone 16B12) to the HA-epitope was from Covance Research (Berkeley, CA), and mAb to ERK2 (clone B3B9) was used as described previously (27).

Cells and Transfection—Rat aortic SMCs (ATCC, CRL-2018) were grown in DMEM, 10% calf serum (CS) containing sodium pyruvate, penicillin, and streptomycin (SMC medium). Primary mouse fibroblasts derived from FAK-null mice (FAK⫺/⫺cells) (9) and FAK-reconstituted FAK/cells (clone DA2) were cultured as described previously (10).

SMC in 10-cm dishes (migration assay) or on collagen-coated glass coverslips in 24-well plates (wound healing assay) were transfected with the indicated amounts of DNA using Effectene (Qiagen, Valencia, CA) 36 – 48 h prior to experiments. Cell transfection efficiency (⬃15%) was determined by FACS analysis on green fluorescence protein (GFP)- transfected cells. For selection of stable cell lines, hygromycin-resistant populations of cells were single-cell-sorted by FACS and expanded, and clones were analyzed by blotting for expression of HA-tagged FRNK.

Control cells were transfected with the pCDNA3.1 vector and selected for growth in hygromycin (150␮g/ml). PDGF-BB stimulation at the indicated concentrations was performed with serum-starved (0.5% CS for 18 h) cells, and lysates were made at the indicated time after PDGF-BB addition.

Wound Healing Assay—SMCs plated onto collagen-coated (10␮g/ml) glass coverslips were transfected with GFP or GFP-FRNK (0.5 ␮g), serum-starved (18 h in 0.5% CS), wounded with a 200-␮l pipette tip, washed with PBS, and incubated in SMC medium. Cells were fixed 18 or 36 h after wounding with 3.7% paraformaldehyde for 15 min, washed with PBS, and permeabilized for 10 min with 0.2% saponin in PBS containing 5% normal goat serum (blocking solution). Samples were incubated for 45 min in blocking solution containing TRITC-phalloidin (Molecular Probes), washed with PBS, embedded in Vectashield (Vec- tor), viewed with an Olympus BX60 epifluorescence microscope, and documented using Ektachrome 400 film. Pictures were taken sepa- rately for GFP and TRITC fluorescence, and the images were merged using the program Adobe Photoshop. For assays with stably-transfected cells, migration of wounded cells was evaluated after 0 and 20 h with an inverted Nikon phase-contrast microscope and photographed with Kodak TMAX-400 film.

Chemotaxis Migration Assay—MilliCell modified Boyden chamber (Millipore, Bedford, MA) migration assays were performed as described previously (12). Briefly, serum-starved cells were detached, resus- pended in migration medium (DMEM containing 0.5% BSA), and counted. Both sides of MilliCell chambers were precoated with rat tail collagen (5␮g/ml in PBS) overnight at 4 °C, washed with PBS, and air-dried. Chambers containing serum-starved cells (1⫻105cells/0.3 ml) were placed in 24-well dishes containing DMEM with 0.5% BSA

with or without PDGF-BB at the indicated concentrations. Transiently transfected migratory cells on the membrane underside were identified by GFP fluorescence, and the migration of stable cell lines was visual- ized by Crystal Violet staining (0.1% Crystal Violet, 0.1Mborate, pH 9.0, 2% EtOH) and cell counting (cells/field using a 40⫻ objective).

Background cell migration in the absence of PDGF-BB (0.5% BSA only) was less than 5% of stimulated values.

Cell Growth Assay and Immunofluorescence Staining—1⫻103cells/

well were seeded in 96-well plates and incubated at 37 °C in complete SMC medium. After 24, 48, or 72 h, cells were fixed, stained with Crystal Violet, and air-dried. Cell-associated dye was eluted with 10%

acetic acid, and absorbance values were determined at 600 nm. Immu- nofluorescence staining of cells attached to fibronectin-coated (10␮g/

ml) glass coverslips for 2 h was performed as described previously (10).

Cell Lysis, Immunoprecipitation, and Immunoblotting—Lysates were made in modified lysis buffer containing 1% Triton X-100, 1% sodium deoxycholate, and 0.1% SDS as described (28). Antibodies were incubated for 4 h at 4 °C and collected on protein A (Repligen, Cambridge, MA) or protein G-plus (Calbiochem) agarose beads. Precipitated protein com- plexes were washed twice in Triton-only lysis buffer followed by washing in HNTG buffer (50 mMHepes, pH 7.4, 150 mMNaCl, 0.1% Triton X-100, 10% glycerol). For immunoblotting, proteins were transferred to polyvi- nylidene fluoride membranes (Millipore). Blots were incubated with ei- ther 1␮g/ml monoclonal or a 1:1000 dilution of polyclonal antibodies for 2 h at room temperature. Bound primary antibody was visualized by enhanced chemiluminescent detection, and subsequent reprobing of membranes was performed as described previously (28).

ERK2 in Vitro Kinase Reactions—ERK-2 was immunoprecipitated with polyclonal anti-ERK2 antibodies (Santa Cruz Biotechnologies), the IPs were washed in Triton-only lysis buffer, followed by HNTG buffer, and kinase buffer (20 mMHepes, pH 7.4, 10% glycerol, 10 mMMgCl2, 10 mMMnCl2, 150 mMNaCl) before they were incubated for 15 min at 32 °C in 30␮l of kinase buffer containing 2.5␮g of myelin basic protein (MBP), 20␮MATP, and 10␮Ci/nmol [␥-32P]ATP (3000 Ci/mmol). Reac- tions were stopped with 2⫻ SDS-polyacrylamide gel electrophoresis sample buffer, resolved by SDS-polyacrylamide gel electrophoresis, stained with Coomassie Blue, visualized by autoradiography, and the radioactivity incorporated into MBP was determined by Cerenkov counting of the excised MBP bands.

Statistical Analyses—Ordinary one-way analysis of variance was used to determine the overall significance within data groups. If a significant result was obtained by analysis of variance, the Tukey- Kramer multiple comparisonsttest was used to determine significance between individual groups.

RESULTS

Transient FRNK Expression Inhibits Wound Healing Re- sponse and PDGF-BB-stimulated Migration of Rat Aortic SMC—To determine whether FAK promotes vascular SMC cell migration, rat aortic SMCs were transfected with an expres- sion vector encoding green fluorescence protein (GFP) or a GFP fusion protein of the FAK-specific inhibitor, FRNK (GFP- FRNK) and analyzed in anin vitrowound healing assay (Fig.

1). When confluent monolayers of transfected cells were ana- lyzed for their ability to repopulate the wounded area, non- transfected and GFP-expressing cells migrated into the open space within the first 18 h (Fig. 1A) and closed the wound within 36 h (Fig. 1C). In contrast, GFP-FRNK-expressing cells remained at the initial wound margin and did not migrate into the open space after 18 h (Fig. 1B). After 36 h, GFP-FRNK- expressing cells were not found in the previously wounded area, whereas the non-transfected cells had migrated into and closed this space (Fig. 1D).

Because matrix proteins remaining in the wound area and growth factors present in the serum-containing medium pro- duce combined motility-promoting signals in wound healing assays, the GFP- or GFP-FRNK-transfected cells were also analyzed for migratory responses in modified Boyden chamber assays where cells migrate through membrane pores in re- sponse to defined chemotactic stimuli. PDGF is a potent che- moattractant for vascular SMCs in culture and is believed to play a major role in neointima formation in atherosclerosis and in restenosis in vivo(2). GFP-transfected SMCs migrated to-

FAK Regulates PDGF-stimulated Smooth Muscle Cell Motility 41093

(3)

ward a PDGF-BB gradient with a maximum response at 5 ng/ml, whereas GFP-FRNK-expressing cells did not readily migrate in response to the PDGF-BB stimulation (Fig. 2A).

This inhibitory effect of FRNK was dose-dependent, as increas- ing amounts of transfected HA-tagged FRNK led to a strong inhibition of PDGF-BB-stimulated (5 ng/ml) SMC cell motility (Fig. 2B). These results show that transient FRNK expression in SMCs disrupts serum and PDGF-BB-stimulated motility- promoting events.

Stable Expression of FRNK Does Not Affect SMC Viability or Morphology—Because FAK has been shown to transduce ex- tracellular matrix-stimulated survival signals (for a review see Ref. 8), FRNK-mediated interference with endogenous FAK function could potentially result in decreased SMC cell viability and thereby affect cell motility responses. To determine the effects of FRNK overexpression on SMCs, stable cell lines were generated expressing either HA-FRNK or HA-FRNK L1034S, a point mutant of FRNK that does not bind paxillin (29). As opposed to FRNK, FRNK L1034S expression in fibroblasts does not localize to focal contacts (10, 29) and does not interfere with FAK-mediated cell motility (10, 12).

Several SMC clones that expressed either FRNK (FRNK-1 and -2) or FRNK L1034S (FRNK L1034S-1 and -2) were iso- lated and expanded (Fig. 3A). As determined by immunoblot- ting whole cell lysates with antibodies to the HA-tag, FRNK and FRNK L1034S were equivalently expressed in the FRNK-1, FRNK-2, and FRNK L10234S-1 clones (Fig. 3A). Rep- robing these whole cell lysates with antibodies to FAK showed that neither FRNK nor FRNK L1034S expression affected en- dogenous FAK expression. However, sequential reprobing this blot with antibodies to the PDGFr-␤showed that its expression

was slightly decreased in both FRNK- and FRNK L1034S- expressing cells compared with the empty vector-transfected control (pCDNA) SMCs (Fig. 3A). Additional comparisons showed that stable FRNK expression was less than the endog- enous level of FAK found in the SMCs (data not shown). Con- trary to previous reports of FRNK expression-promoting cell apoptosis (30, 31), stable expression of FRNK in SMCs did not detectably affect cell viability. Because FRNK expression has also been shown to inhibit cell cycle progression (32, 33), se- rum-stimulated cell proliferation comparisons were made be- tween FRNK, FRNK L1034S, and pCDNA control SMCs (Fig.

3B). After 48 h, no significant differences in cell growth were observed between the different cell clones. After 72 h, a signif- icant difference in the total pCDNA control SMC cell number was observed compared with both FRNK and FRNK L1034S clones (Fig. 3B). No significant differences in serum-stimulated cell proliferation were observed between FRNK- and FRNK L1034S-expressing SMCs after 72 h.

In fibroblasts, FRNK strongly localizes to focal contact sites, whereas FRNK L1034S exhibits a perinuclear distribution FIG. 1. Transient FRNK expression inhibits SMC in vitro

wound healing migration.Monolayers of SMCs transiently trans- fected with GFP (AandC) or GFP-FRNK (BandD) were wounded by a pipette tip. At 18 h (AandB) and 36 h (CandD) after wounding, samples were fixed, actin was stained with TRITC-phalloidin (red) and cells expressing GFP or GFP-FRNK (green) were visualized.Scale bar

represents 20␮m. FIG. 2.Transient FRNK expression inhibits PDGF-BB-stimu-

lated SMC migration. A, SMCs were transiently transfected with expression vectors (5 ␮g each) for GFP (closed bars) or GFP-FRNK (open bars) and used in Boyden chamber migration assays with the indicated concentrations of PDGF-BB as the stimulus. After 5 h, GFP- containing cells on the underside of the membrane were counted. Data represent the mean⫾S.D. of three independent experiments.B, SMCs were transiently co-transfected with the indicated amounts of pCDNA3.1 HA-FRNK and 3 ␮g of GFP-encoding plasmid. Samples were normalized to 7␮g of total DNA with the addition of control vector (pCDNA3.1). SMC chemotaxis in modified Boyden chambers was stim- ulated with 5 ng/ml PDGF-BB. After 5 h, GFP-containing cells on the underside of the membrane were counted.Barsshow the percentage of migration compared with control cells (pCDNA3.1- and GFP-trans- fected) and represent mean⫾S.D. of two independent experiments.

(4)

(10). Indirect immunofluorescence detection of FRNK in the SMC clone FRNK-1 showed that HA-tag staining was concen- trated at the tips of cell protrusions, which correspond also to the vinculin-positive sites of focal contact formation (Fig. 3C).

Importantly, no specific HA-tag staining was detected in the pCDNA control SMCs that overlapped with vinculin staining of focal contact sites (Fig. 3C). Similar to results with fibroblasts, FRNK L1034S expression in SMC clone L1034S-1 was present in a perinuclear distribution and did not significantly co-local- ize with focal contact sites (Fig. 3C). Importantly, no significant differences were observed in either cell morphology, vinculin staining at focal contact sites, or in the general actin cytoskel- etal organization in FRNK-expressing SMC cells compared with the pCDNA and FRNK L1034S-expressing SMCs (Fig.

3C). These results show that stable expression of FRNK in SMCs was associated with focal contact sites but did not neg- atively affect cell viability or morphology.

FRNK Expression Inhibits Specific PDGF-BB-stimulated Ty- rosine Phosphorylation Events—To determine whether the sta- ble expression of FRNK in SMCs was able to disrupt motility- promoting signals as did transient FRNK overexpression, anti- P.Tyr blotting analyses were performed on either serum- starved or PDGF-BB-stimulated (10 ng/ml, 5 min) pCDNA control, FRNK-, or FRNK L1034S-expressing SMC clones (Fig.

4A). Compared with normal non-transfected SMCs (data not shown), the pCDNA control SMCs exhibited an identical pat- tern of tyrosine phosphorylation events under starved and PDGF-BB-stimulated conditions (Fig. 4A,lanes 1and 2). In- terestingly, in both the FRNK and FRNK L1034S clones, the tyrosine phosphorylation of unknown 116- to 130-kDa pro- tein(s) were increased under both serum-starved and PDGF- BB-stimulated conditions compared with pCDNA SMCs (Fig.

4A,lanes 3–10). Although no significant difference in PDGFr-␤ tyrosine phosphorylation upon PDGF-BB addition to the FRNK or FRNK L1034S clones was observed, the stimulated tyrosine phosphorylation of⬃150- and⬃42-kDa proteins was noticeably reduced in the FRNK-expressing compared with the pCDNA and FRNK L1034S-expressing SMCs (Fig. 4A,lanes 4 and 6). These results show that stable FRNK but not FRNK L1034S expression in SMCs selectively disrupts the cascade of PDGF-BB-stimulated tyrosine phosphorylation to distinct downstream targets.

FAK Positively Contributes to PDGF-BB-stimulated ERK Ac- tivation at Low but Not at High Growth Factor Concentra- tions—Previous studies have demonstrated that FAK relays integrin-initiated signals to the activation of the ERK/MAP kinase cascade (34). To determine whether the⬃42-kDa pro- tein showing reduced phosphorylation after PDGF-BB stimu- lation in the FRNK-expressing SMC clones represents ERK2/

MAP kinase, the blot shown in Fig. 4A was reprobed with antibodies recognizing only the active and dually phosphoryl- ated form of ERK kinases (P-ERK). As shown in Fig. 4B, activation of ERK2 in response to PDGF-BB stimulation was markedly reduced in both FRNK-expressing clones when com- pared with pCDNA control cells or FRNK L1034S SMC clones.

Importantly, equal amounts of ERK2 were present in all sam- ples (Fig. 4B,lower panel).

Because previous studies have shown that FAK tyrosine phosphorylation is increased after low but not high concentra- tions of PDGF-BB addition to fibroblasts (35), the pCDNA, FRNK-1, and FRNK L1034S-1 SMC clones were stimulated with increasing concentrations of PDGF-BB for 10 min and whole cell lysates were analyzed for ERK activity by phospho- ERK blotting (Fig. 5A). Compared with the pCDNA and FRNK L1034S-1 clones, the maximal inhibitory effect of FRNK ex- pression was observed at low 10 ng/ml motility-promoting con- FIG. 3.Stable FRNK expression does not affect SMC viability

or morphology.A, SMCs were transfected with HA-FRNK, HA-FRNK L1034S, or with the empty pCDNA3.1 vector and selected for growth in hygromycin. Clones expressing FRNK or FRNK L1034S were identified by HA-tag blotting of whole cell lysates (upper panel). Blots were re- probed with polyclonal antibodies to PDGFr-␤(middle panel) and to FAK (lower panel).B, serum-stimulated proliferation rates of SMC clones expressing FRNK or FRNK L1034S or transfected with the pCDNA control vector. Cells were seeded in 96-well plates (1 ⫻103 cells/well), fixed after the indicated time, and stained with Crystal Violet, and dye absorbance was measured at 600 nm. Values (mean from eight determinations) represent the -fold increase in absorbance compared with cultures after 24 h. After 72 h (asterisks), the number of pCDNA SMCs was significantly different (p ⬍ 0.001) from both the FRNK and FRNK L1034S SMC clones. The experiment was repeated three times with similar results.C, FRNK and FRNK L1034S distri- bution in SMC clones was visualized by immunofluorescence staining with HA-tag antibodies. Control pCDNA SMCs did not show specific HA-tag staining. Vinculin staining highlights focal adhesion structures, and TRITC-phalloidin was used to visualize the actin cytoskeleton (actin).Arrowheadsindicate focal contact sites.Scale barsrepresent 10

M.

FAK Regulates PDGF-stimulated Smooth Muscle Cell Motility 41095

(5)

centrations of PDGF-BB, whereas at higher mitogenic 50 ng/ml concentrations of PDGF-BB, similar levels of ERK activation were observed in lysates from the FRNK-1 SMC clone com- pared with the pCDNA control cells (Fig. 5A). Unexpectedly, the level of PDGF-BB-stimulated ERK activity in the FRNK L1034S-1 clone at 10 ng/ml was higher than the stimulated ERK level in pCDNA control SMCs (Fig. 5A) even though PDGFr-␤expression levels are lower in the FRNK L1034S-1 clone than in the pCDNA control SMCs (Fig. 3A). Although the mechanism(s) for increased PDGF-BB stimulated ERK activity in the FRNK L1034S-1 clones is(are) not known, the observed increased tyrosine phosphorylation of ⬃116- to 130-kDa pro- teins in these cells (Fig. 4A) may be indicative of cellular compensatory events.

Because ERK activity in the FRNK-1 SMC clone was maxi- mally inhibited at 10 ng/ml PDGF-BB (Fig. 5A), experiments were also performed to evaluate whether FRNK expression affected the duration of PDGF-BB-stimulated ERK activation (Fig. 5B). Serum-starved pCDNA, FRNK-1, and FRNK L1034S-1 SMC clones were stimulated with 10 ng/ml PDGF- BB, and at time points between 5 and 120 min, whole cell lysates were analyzed for ERK activity by phospho-ERK blot- ting (Fig. 5B). Compared with both the pCDNA and FRNK L1034S-1 SMC clones, where ERK activity persisted up to 120

min, PDGF-BB-stimulated ERK activity was decreased in the early 5- to 10-min time points and not detectably activated after 60 min in the FRNK-1 SMC clone (Fig. 5B). These com- bined results show that stable FRNK expression in SMCs re- FIG. 4.Stable FRNK expression disrupts specific PDGF-BB-

stimulated tyrosine phosphorylation and signaling events. A, whole cell lysates from serum-starved or PDGF-BB-stimulated (10 ng/

ml, 5 min) SMCs were analyzed by anti-P.Tyr immunoblotting. Proteins showing decreased PDGF-stimulated tyrosine phosphorylation in FRNK-expressing SMCs are indicated (asterisks).B, the lower portion of the membrane shown inAwas reprobed with monoclonal anti-active phospho-ERK (P-ERK) antibodies (upper panel) and with monoclonal anti-ERK2 antibodies (lower panel).

FIG. 5.At low PDGF-BB concentrations, FAK contributes to the magnitude and duration of ERK/MAP kinase activation.A, whole cell lysates from SMCs stimulated with the indicated concentra- tions PDGF-BB (5 min) were analyzed with monoclonal anti-active phospho-ERK (P-ERK) antibodies (upper panel) and then reprobed with monoclonal anti-ERK2 antibodies (lower panel).B, lysates from SMC stimulated for the indicated times with PDGF-BB (10 ng/ml) were analyzed as inA.C, ERK2 activation is impaired in cells that lack FAK.

Whole cell lysates from serum-starved or PDGF-BB-stimulated (10 ng/ml, 5 min) FAK/and FAK re-expressing (DA2) fibroblasts were analyzed by ERK2 blotting. The slower migrating ERK2 band repre- sents phosphorylated and activated ERK2. ERK2 IPs from starved or PDGF-stimulated FAK/, and DA2 cells were assayed for in vitro kinase activity using myelin basic protein (MBP) as a substrate. Radio- activity incorporated into MBP was determined by Cerenkov counting, and values represent the means⫾S.D. of three determinations.Aster- isksindicate ERK2 activity significantly higher in starved FAK/than in DA2 cells (p⬍0.05).Double asterisks indicate ERK2 activity sig- nificantly lower in PDGF-BB-stimulated FAK/ than in DA2 cells (p⬍0.01).

(6)

duces the extent and duration of PDGF-BB-stimulated ERK activity.

To address the issue as to whether FRNK-mediated inhibi- tion of PDGF-BB-stimulated ERK2 activity represents an in- hibition of FAK function, analyses were performed in FAK-null fibroblasts (FAK⫺/⫺) and FAK-reconstituted fibroblasts (clone DA2) to determine whether the presence or absence of FAK affected PDGF-BB stimulated ERK2 activation (Fig. 5C). The DA2 cells were generated from an FAK⫺/⫺ cell clone, and previous studies have shown that the PDGFr is expressed equivalently in both cell lines (12). Compared with DA2 cells, FAK/cells displayed a higher basal level of ERK2 activity under serum-starved conditions (Fig. 5C,lanes 1and3) and a lower level of induced ERK2 activity after PDGF-BB stimula- tion (Fig. 5C,lanes 2and4). Although the FAK-related PTK Pyk2 is highly expressed in the FAK/ cells (27) and may contribute to the high basal ERK activity in the absence of FAK, the DA2 cell results show that FAK positively contributes to PDGF-BB-stimulated ERK2 activation.

PDGF-BB-stimulated ERK Activation Is Required for SMC Motility—To establish the connections between FRNK expres- sion, inhibition of PDGF-BB-stimulated ERK2 activity, and cell migration, SMCs were pretreated for 45 min with increasing concentrations of the MEK/ERK kinase inhibitor PD98059 and subsequently stimulated with PDGF-BB (Fig. 6A). Although 10 –25␮Mof PD98059 did not interfere with PDGF-BB-stimu- lated tyrosine phosphorylation of the PDGFr or FAK, PD98059 inhibited PDGF-BB-stimulated ERK activation in a concentra- tion-dependent manner (Fig. 6A). Additionally, SMCs were either pretreated with PD98059 or a specific inhibitor for p38

kinase (SB203580) and employed in modified Boyden chamber motility assays (Fig. 6B). Although treatment of SMCs with the p38 inhibitor had no effect on PDGF-BB-stimulated cell migra- tion (data not shown), PD98059 reduced PDGF-BB-stimulated SMC chemotaxis in a similar dose-dependent range as ob- served for ERK2 inhibition (Fig. 6B). Taken together, these findings support the conclusion that FRNK may disrupt stim- ulated SMC migration in part by interfering with the FAK-de- pendent aspect of PDGF-BB-mediated ERK2 activation.

Stable FRNK Expression Inhibits Wound Healing Response and PDGF-stimulated Motility of Rat Aortic SMCs—To inves- tigate the effect of stable FRNK overexpression on SMC migra- tion,in vitro wound healing assays were performed (Fig. 7).

Control pCDNA, FRNK-1, or the FRNK L1034S-1 SMC clones were grown to the same density, and then the cell monolayer was wounded with a pipette tip (Fig. 7,A,C, andE). After 20 h in the presence of serum, pCDNA SMCs (Fig. 7, B) and the FRNK L1034S-1 SMCs (Fig. 7F) exhibited cell reorientation responses along the wounded edge margin and had migrated into the wounded area. However, after 20 h, the FRNK-1 SMCs (Fig. 7D) exhibited only limited cell reorientation responses along the wounded edge margin and did not efficiently repop- ulate the open space. Similar results were obtained with wound healing assays using the FRNK-2 and FRNK L1034S-2 SMC clones (data not shown).

Because stable FRNK expression in the SMCs inhibited PDGF-BB-stimulated ERK activity (Figs. 4 and 5) and phar- macological inhibition of the MEK/ERK kinase cascade dis- rupted PDGF-BB-stimulated SMC motility (Fig. 6), the pCDNA, FRNK-, and FRNK L1034S-expressing SMCs were analyzed for PDGF-BB-stimulated (5 ng/ml) motility responses in Boyden chamber assays (Fig. 8A). Similar to the results obtained in the transient transfection experiments (Fig. 2), stable FRNK expression led to a strong reduction (8- to 10-fold) in PDGF-BB-stimulated motility compared with FRNK FIG. 6.ERK activity is required for PDGF-BB-stimulated SMC

chemotaxis migration.A, serum-starved SMC were pretreated with the indicated concentrations of the PD98059 MEK inhibitor for 45 min and then stimulated with PDGF-BB (20 ng/ml, 10 min). Whole cell lysates were analyzed by anti-P.Tyr (upper panel), anti-phospho-ERK (middle panel), and anti-ERK2 blotting (lower panel).B, serum-starved SMCs pretreated with the indicated concentrations of PD98059, were employed in Boyden chamber migration assays with 5 ng/ml PDGF-BB as a stimulus. After 5 h, cells on the lower side of the membrane were fixed, stained, and counted.Barsrepresent means⫾S.D. from three experimental analyses.

FIG. 7. Stable FRNK expression blocks serum-stimulated wound healing response of SMCs.In vitrowound healing assays using control pCDNA (AandB), FRNK- (CandD), and FRNK L1034S- expressing (EandF) SMCs. Phase contrast images represent initial wounding of cell monolayer (0 h;A,C, andE) and migration in the presence of serum for 20 h (B,D, andF).

FAK Regulates PDGF-stimulated Smooth Muscle Cell Motility 41097

(7)

L1034S and pCDNA SMCs (Fig. 8A). These results show that inhibition of FAK function by FRNK but not FRNK L1034S expression results in disrupting both PDGF-BB-stimulated ERK activity and cell motility.

FRNK Blocks the Formation of a Complex between FAK and PDGFr and Promotes FAK Dephosphorylation at Tyr-397—

Although the exact molecular connections of FAK to PDGF-BB- stimulated ERK activation and cell motility remain to be de- fined, FAK has been shown to co-immunoprecipitate with an activated complex containing PDGFr-␤ after PDGF-BB stimulation of fibroblasts (12). To determine whether a FAK

PDGFr-␤ complex is formed in SMCs, serum-starved pCDNA SMCs were stimulated with PDGF-BB, FAK was iso-

lated by immunoprecipitation, and FAK-associated proteins were visualized by anti-P.Tyr blotting (Fig. 8B). Compared with serum-starved cells, PDGF-BB stimulation led to the as- sociation of a⬃190-kDa P.Tyr-containing protein with FAK in SMCs (Fig. 8B). To confirm that the⬃190-kDa tyrosine-phos- phorylated protein associated with FAK upon PDGF-BB stim- ulation was PDGFr-␤, reciprocal co-immunoprecipitation ex- periments with anti-PDGFr antibodies were performed on starved or PDGF-BB-stimulated SMCs (Fig. 8C). Under serum- starved conditions, neither FAK nor other tyrosine-phosphory- lated proteins were detected in association with the PDGFr-␤. However, after PDGF-BB stimulation of SMCs, tyrosine-phos- phorylated FAK was found to co-immunoprecipitate with anti- bodies to PDGFr-␤but not with preimmune serum from PDGF- BB-stimulated SMC lysates (Fig. 8C). These results show that an activated complex containing tyrosine-phosphorylated FAK and PDGFr-␤is formed after PDGF-BB stimulation of SMCs.

To test whether stable FRNK expression in SMCs may in- terfere with the stimulated formation of a FAK- and PDGFr-

␤-containing complex, antibodies specific to the FAK N-termi- nal domain were used to isolate endogenous FAK from either the pCDNA control, FRNK-, or FRNK L1034S-expressing SMCs after PDGF-BB stimulation (Fig. 8D). As detected by anti-P.Tyr blotting, PDGFr-␤ co-immunoprecipitated with FAK from the pCDNA and FRNK L1034S SMCs, but not de- tectably with FAK from FRNK-expressing SMCs (Fig. 8D).

Equal levels of FAK were isolated from the pCDNA, FRNK-, and FRNK L1034S-expressing SMCs. Significantly, FAK iso- lated from the PDGF-BB-stimulated FRNK-1 SMCs exhibited reduced tyrosine phosphorylation levels compared with FAK isolated from pCDNA and FRNK L1034S-1 SMCs (Fig. 8D).

Previous studies in fibroblasts have shown that motility-pro- moting concentrations of PDGF-BB promote FAK phosphoryl- ation at Tyr-397 and the stimulated SH2-mediated binding of Src family PTKs to this site on FAK (12, 36). Using a phospho- specific antibody directed to the FAK Tyr-397 site, stable FRNK expression but not FRNK L1034S resulted in the re- duced phosphorylation of FAK Tyr-397 after PDGF-BB stimu- lation of SMCs (Fig. 8D). Taken together, our results support the conclusion that FRNK expression in SMCs acts in a recep- tor-proximal fashion to uncouple the stimulated linkage be- tween FAK and PDGFr-␤, thereby resulting in the inhibition of FAK Tyr-397 phosphorylation and the reduced propagation of downstream signaling events.

DISCUSSION

Accumulating evidence supports a critical role for FAK in promoting cell migration stimulated by different types of cell surface ligand receptors. In this report, we investigated the involvement of FAK in PDGF-BB-stimulated chemotaxis of vascular SMCs through the transient and stable expression of a FAK-specific inhibitor comprising the FAK C-terminal do- main termed FRNK (37). FRNK encompasses the focal adhe- sion targeting sequences (38) and proline-rich motifs that in- teract with SH3-containing proteins (39, 40). FRNK does not possess kinase activity, nor does it contain the FAK autophos- phorylation/SH2 binding site at Tyr-397. FRNK is expressed as an independent transcript in a tissue- and developmental-spe- cific manner from a cryptic promoter located within an intron of the FAK gene (25). Because transient FRNK expression in fibroblasts promotes the displacement of FAK from focal con- tact sites (22), dephosphorylation of FAK at Tyr-397 (10), and inhibition of FAK-mediated cell motility (10, 17, 23), FRNK is believed to function as a specific inhibitor of FAK function.

We found that both transient as well as stable FRNK expres- sion in SMCs potently inhibited PDGF-BB-induced cell migra- tion. Importantly, although FRNK strongly localized to focal FIG. 8.Stable FRNK expression reduces PDGF-stimulated che-

motaxis and FAK䡠PDGFr associations and promotes FAK de- phosphorylation at Tyr-397.A, control pCDNA, FRNK-, or FRNK L1034S-expressing SMC clones were analyzed in Boyden chamber che- motaxis assays with 5 ng/ml PDGF-BB as stimulus. After 5 h, migra- tory cells on the underside of the membrane were fixed, stained, and counted.Barsrepresent means⫾S.D. from two independent experi- ments. B, lysates were made from serum-starved SMCs that were replated onto collagen-coated (10␮g/ml) dishes for 2 h at 37 °C. Re- plated cells were then stimulated with PDGF-BB (20 ng/ml, 10 min).

FAK-associated proteins were visualized by anti-P.Tyr (upper panel), and equal amounts of immunoprecipitated FAK was confirmed by FAK blotting (lower panel).C, lysates were made from either serum-starved or PDGF-BB-stimulated SMCs as described inB, and IPs were made with either preimmune serum (PI) or with anti-PDGFr-␤serum. Se- quential immunoblotting with antibodies to the PDGFr (upper panel), phosphotyrosine (middle panel), or to FAK (lower panel) were used to visualize the formation of a FAK䡠PDGFr complex after PDGF-BB stim- ulation of SMCs.D, N-terminal domain-specific antibodies to FAK were used in IPs with lysates from collagen-replated and PDGF-BB stimu- lated SMCs as inB. FAK-associated proteins were visualized by anti- P.Tyr (upper panel) and sequentially followed by anti-FAK blotting (middle panel). Whole cell lysates (50 ␮g of protein) from collagen- replated and PDGF-BB stimulated SMCs were analyzed with phospho- specific antibodies to the FAK Tyr-397 phosphorylation site (pY397;

lower panel).

(8)

contact sites in SMCs, stable expression of FRNK L1034S, which contains a point mutation disrupting the paxillin bind- ing site, did not localize to focal contact sites and did not block FAK function in PDGF-BB-stimulated SMC migration. Impor- tantly, although FRNK overexpression has been reported to promote cell apoptosis (31) and inhibit growth factor-stimu- lated cell cycle progression (32, 33), stable expression of FRNK within SMCs did not affect cell viability, cell proliferation rates, or alter SMC morphology. Because stable FRNK expres- sion in the SMCs is less than the level of endogenous FAK, it is possible that the relative levels of FRNK and FAK expression within cells may differentially influence cell survival- and cell motility-promoting signaling pathways.

Interestingly, as first observed upon PDGF-BB stimulation of fibroblasts (12), FAK associated with an activated PDGFr-␤ complex upon PDGF-BB stimulation of SMCs. FRNK, but not FRNK L1034S expression, inhibited the stimulated association of FAK with a PDGFr-␤-containing complex. Although the mo- lecular connections linking FAK to the PDGFr-␤remain to be determined, our results showing a strong correlation between PDGFr-␤/FAK association and cell motility in SMCs support previous studies in fibroblasts whereby a stimulated complex between FAK and the epidermal growth factor (EGF) receptor was required for efficient EGF-stimulated cell migration (12).

Together, these results suggest that FAK serves as a receptor- proximal coordinator of cell migration connecting both growth factor receptors and integrins with motility-promoting down- stream signaling events.

In SMCs, we found that FRNK expression inhibited the magnitude and duration of PDGF-BB-stimulated ERK activity at low but not at high growth factor concentrations. We also found that FAK reconstitution of FAK⫺/⫺ cells enhanced PDGF-BB-stimulated ERK activity. Although previous studies with FAK signaling have focused on its role in promoting either integrin-stimulated signaling to the ERK/MAP (8, 32) or JNK/

SAP kinase (26, 33) cascades, our results for the first time demonstrate the importance of FAK in the transduction of signals from a growth factor receptor to ERK2/MAP kinases.

Because the molecular mechanism(s) through which FAK facilitates PDGF-BB-stimulated ERK activation are not known, ongoing investigations are focused on the potential role of FAK in promoting the preferential localization of a motility- promoting signaling complex or the role of FAK in facilitating efficient activation of ERK at synergistic inputs below the level of Ras activation (41). Nevertheless, studies in fibroblasts have shown that the integrity of the SH2 binding site at FAK Tyr- 397 and the recruitment of Src family PTKs into a signaling complex is the first of several important signaling events nec- essary for PDGF-stimulated cell motility (12). Stable FRNK expression in SMCs disrupted the formation of an activated complex containing FAK and the PDGFr-␤, promoted FAK dephosphorylation at Tyr-397, and resulted in reduced PDGF- BB-stimulated ERK activity. Pharmacological inhibition of ERK activation after PDGF stimulation prevented efficient SMC motility, which is consistent with previous findings (42, 43). Because growth factor-stimulated ERK2/MAP kinase acti- vation and subsequent ERK2 connections to increased myosin light chain kinase activity can facilitate cell contractility and cell migration in a variety of cell types (44), decreased PDGF- BB-stimulated ERK activity, due to FAK inhibition, could ex- plain the reduced cell migration observed upon transient and stable FRNK expression in SMCs.

In conclusion, our studies suggest that FAK is a promising target for future therapeutic intervention strategies. We propose that techniques to deliver FRNK to SMCsin vivoor the activation of signaling pathways regulating endogenous FRNK expression

might represent novel approaches to prevent excessive vascular remodeling following endovascular manipulations.

Acknowledgments—We thank Shannon Reider for technical assist- ance and Amanda Moore for administrative support.

REFERENCES 1. Andres, V. (1998)Int. J. Mol. Med.2,81– 89 2. Ross, R. (1993)Nature362,801– 809

3. Matsuno, H., Stassen, J. M., Vermylen, J., and Deckman, H. (1994)Circulation 90,2203–2206

4. Hanke, H., Strohschneider, T., Oberhoff, M., Betz, E., and Karsch, K. R. (1990) Circ. Res.67,651– 659

5. Rutherford, C., Martin, W., Salame, M., Carrier, M., Anggard, E., and Ferns, G. (1997)Atherosclerosis130,45–51

6. Waltenberger, J., Uecker, A., Kroll, J., Frank, H., Mayr, U., Bjorge, J. D., Fujita, D., Gazit, A., Hombach, V., Levitzki, A., and Bohmer, F. D. (1999) Circ. Res.85,12–22

7. Gotwals, P. J., Chi-Rosso, G., Linder, V., Yang, J., Ling, L., Fawell, S. E., and Koteliansky, V. E. (1996)J. Clin. Invest.97,2469 –2477

8. Schlaepfer, D. D., Hauck, C. R., and Sieg, D. J. (1999)Prog. Biophys. Mol. Biol.

71,435– 478

9. Ilic, D., Furuta, Y., Kanazawa, S., Takeda, N., Sobue, K., Nakatsuji, N., Nomura, S., Fujimoto, J., Okada, M., Yamamoto, T., and Aizawa, S. (1995) Nature377,539 –544

10. Sieg, D. J., Hauck, C. R., and Schlaepfer, D. D. (1999) J. Cell Sci.112, 2677–2691

11. Owen, J. D., Ruest, P. J., Fry, D. W., and Hanks, S. K. (1999)Mol. Cell. Biol.

19,4806 – 4818

12. Sieg, D. J., Hauck, C. R., Ilic, D., Klingbeil, C. K., Schaefer, E., Damsky, C. H., and Schlaepfer, D. D. (2000)Nat. Cell Biol.2,249 –256

13. Reiske, H. R., Kao, S. C., Cary, L. A., Guan, J. L., Lai, J. F., and Chen, H. C.

(1999)J. Biol. Chem.274,12361–12366

14. Lai, J.-F., Kao, S.-C., Jiang, S.-T., Tang, M.-J., Chan, P.-C., and Chen, H.-C.

(2000)J. Biol. Chem.275,7474 –7480

15. Cary, L. A., Han, D. C., Polte, T. R., Hanks, S. K., and Guan, J.-L. (1998)J. Cell Biol.140,211–221

16. Gu, J., Tamura, M., Pankov, R., Danen, E. H., Takino, T., Matsumoto, K., and Yamada, K. M. (1999)J. Cell Biol.146,389 – 404

17. Richardson, A., Malik, R. K., Hildebrand, J. D., and Parsons, J. T. (1997)Mol.

Cell. Biol.17,6906 – 6914

18. Han, D. C., and Guan, J. L. (1999)J. Biol. Chem.274,24425–24430 19. Yu, D. H., Qu, C. K., Henegariu, O., Lu, X., and Feng, G. S. (1998)J. Biol.

Chem.273,21125–21131

20. Manes, S., Mira, E., Gomez-Mouton, C., Zhao, Z. J., Lacalle, R. A., and Martinez, A. C. (1999)Mol. Cell. Biol.19,3125–3135

21. Qi, J. H., Ito, N., and Claesson-Welsh, L. (1999) J. Biol. Chem. 274, 14455–14463

22. Richardson, A., and Parsons, J. T. (1996)Nature380,538 –540 23. Gilmore, A. P., and Romer, L. H. (1996)Mol. Biol. Cell7,1209 –1224 24. Zheng, D. Q., Woodard, A. S., Fornaro, M., Tallini, G., and Languino, L. R.

(1999)Cancer Res.59,1655–1664

25. Nolan, K., Lacoste, J., and Parsons, J. T. (1999)Mol. Cell. Biol.19,6120 – 6129 26. Almeida, E. A. C., Ilic, D., Han, Q., Hauck, C. R., Jin, F., Kawakatsu, H.,

Schlaepfer, D. D., and Damsky, C. H. (2000)J. Cell Biol.149,741–754 27. Sieg, D. J., Ilic, D., Jones, K. C., Damsky, C. H., Hunter, T., and Schlaepfer,

D. D. (1998)EMBO J.17,5933–5947

28. Schlaepfer, D. D., Jones, K. C., and Hunter, T. (1998)Mol. Cell. Biol.18, 2571–2585

29. Tachibana, K., Sato, T., D’Avirro, N., and Morimoto, C. (1995)J. Exp. Med.

182,1089 –1100

30. Hungerford, J. E., Compton, M. T., Matter, M. L., Hoffstrom, B. G., and Otey, C. A. (1996)J. Cell Biol.135,1383–1390

31. Xu, L. H., Yang, X., Craven, R. J., and Cance, W. G. (1998)Cell Growth Differ.

9,999 –1005

32. Zhao, J.-H., Reiske, H., and Guan, J.-L. (1998)J. Cell Biol.143,1997–2008 33. Oktay, M., Wary, K. K., Dans, M., Birge, R. B., and Giancotti, F. G. (1999)

J. Cell Biol.145,1461–1469

34. Schlaepfer, D. D., Hanks, S. K., Hunter, T., and van der Geer, P. (1994)Nature 372,786 –791

35. Rankin, S., Hooshmand-Rad, R., Claesson-Welsh, L., and Rozengurt, E. (1996) J. Biol. Chem.271,7829 –7834

36. Salazar, E. P., and Rozengurt, E. (1999)J. Biol. Chem.274,28371–28378 37. Schaller, M. D., Borgman, C. A., and Parsons, J. T. (1993)Mol. Cell. Biol.13,

785–791

38. Hildebrand, J. D., Schaller, M. D., and Parsons, J. T. (1993)J. Cell Biol.123, 993–1005

39. Hildebrand, J. D., Taylor, J. M., and Parsons, J. T. (1996)Mol. Cell. Biol.16, 3169 –3178

40. Polte, T. R., and Hanks, S. K. (1997)J. Biol. Chem.272,5501–5509 41. Renshaw, M. W., Price, L. S., and Schwartz, M. A. (1999)J. Cell Biol.147,

611– 618

42. Graf, K., Xi, X. P., Yang, D., Fleck, E., Hsueh, W. A., and Law, R. E. (1997) Hypertension29(1 Pt 2),334 –339

43. Cospedal, R., Abedi, H., and Zachary, I. (1999)Cardiovasc Res.41,708 –721 44. Klemke, R. L., Cai, S., Giannini, A. L., Gallagher, P. J., de Lanerolle, P., and

Cheresh, D. A. (1997)J. Cell Biol.137,481– 492

FAK Regulates PDGF-stimulated Smooth Muscle Cell Motility 41099

Referenzen

ÄHNLICHE DOKUMENTE

PDGF-BB (5 rig/ml) increased 3H-thymidine incorporation into vascular smooth muscle cells obtained from postmenopausal women (511 rf: 57% above control; Fig. The vehicle,

Recently, it has been shown that satellite cells can express Sema3A in response to hepatocyte growth factor (36). However, the non-endothelial source of Sema3A that we observed

Pyk2 does not strongly localize to focal contact sites in FAK-null cells and targeting the Pyk2 N-terminal and kinase domains to focal adhe- sions by fusion to the FAK C-terminal

Lower levels of both FAK-associated in vitro kinase activity and v-Src co-immunoprecipitation (IP) were detected in lysates from v-Src FRNK compared with both v-Src3T3s and v-Src

Total RNA was isolated from immature and mature MoDCs that had been stimulated with a cocktail of proinflammatory cytokines (TNF-␣, IL-1␤, and IL-6), sCD40L, or polyI:C in the

The studies which have investigated gene expression profiles of circulating leukocytes in response to physical exercise have revealed several candidate genes

Verantwortlich für die Planung und Durchführung auf Konzerten Helfer: Anian Weinberger, Thorsten Ruffer.. Mitgliederverwaltung:

Inhibition of focal adhesion kinase expression or activity disrupts epidermal growth factor- stimulated signaling promoting the migration of invasive human carcinoma cells. Human