JBC Papers in Press. Published on July 31, 2002 as Manuscript M

Similar documents
Association of the Ste20-like Kinase (SLK) with the Microtubule

Beta3 integrin promotes long-lasting activation and polarization of Vascular Endothelial Growth Factor Receptor 2 by immobilized ligand

Cdc42 Activation Assay Kit

Gα 13 Activation Assay Kit

B. ADM: C. D. Apoptosis: 1.68% 2.99% 1.31% Figure.S1,Li et al. number. invaded cells. HuH7 BxPC-3 DLD-1.

CD93 and dystroglycan cooperation in human endothelial cell adhesion and migration

Rabbit (monoclonal) Anti-FAK [py 397 ] Phosphospecific Antibody, Unconjugated

Supplementary information

Lab Module 7: Cell Adhesion

Supplementary Material

Gα i Activation Assay Kit

T H E J O U R N A L O F C E L L B I O L O G Y

SUPPLEMENTARY INFORMATION

SUPPLEMENTAL MATERIAL. Supplemental Methods:

Supplementary Figure 1 Pfn1, but not other Pfn isoforms are expressed in

SUPPLEMENTARY INFORMATION FIGURE 1 - 1

Rab5 Activation Assay Kit

Arf6 Activation Assay Kit

Journal of Cell Science Supplementary Material

Supplemental Data Supplementary Figure Legends and Scheme Figure S1.

invasion and motility

Supporting Information

SUPPLEMENTARY INFORMATION

RheB Activation Assay Kit

Rho activation kit. Catalog Number: ADI-EKS-465. Table of Contents

Post-translational modification

Supplemental Materials and Methods

Supplementary Materials and Methods

Required role of focal adhesion kinase (FAK) for integrin-stimulated cell migration

MCF10A cells were trypsinized and attached onto fibronectin-coated petri dishes

Focal adhesion kinase suppresses Rho activity to promote focal adhesion turnover

IgG TrueBlot Protocol for Mouse, Rabbit or Goatderived Antibodies - For Research Use Only

*Corresponding author. Tel: ;

System. Dynamic Monitoring of Receptor Tyrosine Kinase Activation in Living Cells. Application Note No. 4 / March

T H E J O U R N A L O F C E L L B I O L O G Y

Protocol for induction of expression and cell lysate production

ApoTrack Cytochrome c Apoptosis ICC Antibody Kit

Rabbit (polyclonal) Anti-Rac1/cdc42 [ps 71 ] Phosphospecific Antibody, Unconjugated

Apoptosis And Anti-tumor Effect Induced By Mtor Inhibitor And Autophagy Inhibitor In Human Osteosarcoma Cells

ab Ran Activation Assay Kit

Flag-Rac Vector V12 V12 N17 C40. Vector C40 pakt (T308) Akt1. Myc-DN-PAK1 (N-SP)

LINGO-1, A TRANSMEMBRANE SIGNALING PROTEIN, INHIBITS OLIGODENDROCYTE DIFFERENTIATION AND MYELINATION THROUGH INTERCELLULAR SELF- INTERACTIONS.

RNA was isolated using NucleoSpin RNA II (Macherey-Nagel, Bethlehem, PA) according to the

Supplementary Table 1. The Q-PCR primer sequence is summarized in the following table.

Androgen Receptor (Phospho-Tyr363) Colorimetric Cell-Based ELISA Kit. Catalog #: OKAG02138

T H E J O U R N A L O F C E L L B I O L O G Y

Figure legends for supplement

This Document Contains:

Supplementary Fig.1 Luton

ApoTrack Cytochrome c Apoptosis ICC Antibody

Supporting Online Material Y. Tang et al., published 1/24/03

Description of supplementary material file

ApoTrack Cytochrome c Apoptosis ICC Antibody Kit: 2 color immunocytochemistry of cytochrome c and mitochondria.

Nature Immunology: doi: /ni.1744

Segments of the obstructed intestinal loops were fixed in 4% paraformaldehyde

Promotion of HDF Cell Attachment and Proliferation

1. QUANTITY OF LYSATE 2. LYSIS BUFFER

Time allowed: 2 hours Answer ALL questions in Section A, ALL PARTS of the question in Section B and ONE question from Section C.

Protocol(Research use only)

Methods Western blot analysis of plg Quantification of plasminogen accumulation by ELISA Immunohistochemical analysis

Anti-HB-EGF (Human) mab

Phos-tag beads as an immunoblotting enhancer for selective detection of phosphoproteins in cell lysates

Supplemental Information

AlphaScreen SureFire Histone H3 (p-ser10) Assay Kits. Manual

THE BASICS OF IMMUNOHISTOCHEMISTRY

Supplementary data. sienigma. F-Enigma F-EnigmaSM. a-p53

CytoGLOW. IKK-α/β. Colorimetric Cell-Based ELISA Kit. Catalog #: CB5358

TECHNICAL BULLETIN. MEK Activity Assay Kit. Product Code CS0490 Storage Temperature 20 C

Supplemental Online Material. The mouse embryonic fibroblast cell line #10 derived from β-arrestin1 -/- -β-arrestin2 -/-

Mechanism of Induction and Suppression of Antiviral Immunity Directed by Virus-Derived Small RNAs in Drosophila

INOS. Colorimetric Cell-Based ELISA Kit. Catalog #: OKAG00807

ab G alpha i Activation Assay Kit

Kinase Reaction and Alkylation Protocol

Supporting Information

Single cell imaging of Bruton's Tyrosine Kinase using an irreversible inhibitor

AlphaScreen SureFire STAT3 Total Assay Kits. Manual

14_integrins_EGFR

Protein A Agarose Immunoprecipitation Kit

FIGURE S1. Representative images to illustrate RAD51 foci induction in FANCD2 wild-type (wt) and

AlphaScreen SureFire EGF Receptor (p-tyr1068) Assay Kits. Manual

Rabbit (polyclonal) Anti-IRS-1 [ps 616 ] Phosphospecific Antibody, Unconjugated

Supplementary Material - Methods

Four different active promoter genes were chosen, ATXN7L2, PSRC1, CELSR2 and

For gel-shift assays, 2 ul ivtt synthesized protein (Promega) was incubated at room temperature

Figure 1: TDP-43 is subject to lysine acetylation within the RNA-binding domain a) QBI-293 cells were transfected with TDP-43 in the presence or

Please read manual carefully before starting experiment

Introduction. Candice K. Klingbeil, Christof R. Hauck, Datsun A. Hsia, K.C. Jones, Shannon R. Reider, and David D. Schlaepfer

Western Blot Tool Box

Culture media, trypsin, penicillin and streptomycin were from Invitrogen (Breda, the Netherlands).

3 P p25. p43 p41 28 FADD. cflips. PE-Cy5 [Fluorescence intensity]

For Research Use Only. Not for use in diagnostic procedures.

SensoLyte Anti-alpha-Synuclein Quantitative ELISA Kit (Human/Mouse/Rat) *Colorimetric*

RhoC Activation Assay Kit

J. Cell Sci. 128: doi: /jcs : Supplementary Material. Supplemental Figures. Journal of Cell Science Supplementary Material

Fig. S1 TGF RI inhibitor SB effectively blocks phosphorylation of Smad2 induced by TGF. FET cells were treated with TGF in the presence of

Supplementary Information (Ha, et. al) Supplementary Figures Supplementary Fig. S1

Recombinant adenoviruses. A TRB3-expressing adenovirus was generated through

EGFR (Phospho-Ser695)

The following antibodies were used in this study: NuMA - rabbit-anti NuMA (gift from

VEGFR2 (Phospho-Tyr1175)

Transcription:

JBC Papers in Press. Published on July 31, 2002 as Manuscript M205899200 Association of the Ste20-like Kinase SLK with the Microtubule: Role in Rac1-Mediated Regulation of Actin Dynamics during Cell Adhesion and Spreading Simona Wagner 1, Trevor A. Flood 1, Paul O Reilly 1, Karri Hume 2 and Luc A. Sabourin 1,2 1 Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada 2 Ottawa Health Research Institute, Neuroscience Program, Ottawa, ON, Canada Running Title: Regulation of cell adhesion and spreading by SLK (Total 39,694 characters) Key Words: SLK, fibronectin, actin, microtubule, adhesion Corresponding author: Luc A. Sabourin Neuroscience Research Institute 451 Smyth Rd, Ottawa 1 Copyright 2002 by The American Society for Biochemistry and Molecular Biology, Inc.

Ontario, Canada K1H 8M5 lsab@uottawa.ca Summary Cytoskeletal remodeling events are tightly regulated by signal transduction systems that impinge on adhesion components and modulators of cellular architecture. We have previously shown that the Ste20-like kinase SLK can induce apoptosis through the induction of actin disassembly and cellular retraction (Sabourin et al., (2000) Mol Cel Biol 20(2): 684-696). Using immunofluorescence studies, we report that SLK is redistributed with adhesion components at large podosome-like adhesion sites in fibronectin-stimulated fibroblasts. However, in vitro kinase assays demonstrate that its activity is not modulated following FN stimulation. Double immunofluorescence studies in exponentially growing or spreading fibroblasts shows that SLK is associated with the microtubule network and can be co-precipitated with α-tubulin. Furthermore, the stimulation of adhesion site formation by microtubule disrupting agents induces the relocalization of SLK with unpolymerized α-tubulin to large vinculin-containing adhesion complexes. Using microinjection studies we show that ectopic expression of activated SLK induces the disassembly of actin stress fibers, a process which can be inhibited by dominant negative Rac1. Significantly, endogenous SLK can be colocalized with Rac1 in spreading cells on FN. Finally, overexpresion of SLK by adenoviral infection inhibits cell spreading on fibronectin. These results suggest that SLK is part of a microtubule-associated complex that is targeted to adhesion sites and implicated in the regulation of cytoskeletal dynamics. 2

INTRODUCTION Cells reside in a protein network, the extracellular matrix (ECM), which they secrete and mold into the intercellular space. The ECM exerts profound control over the cells. The effects of the matrix are primarily mediated by integrins, a family of cell surface receptors that attach cells to the matrix and mediate mechanical and chemical signals from it. These signals regulate the activities of cytoplasmic kinases, growth factor receptors, ion channels and control the organization of the intracellular actin cytoskeleton (1). Integrin receptor binding to ECM proteins generates intracellular signals through tyrosine phosphorylation events that are important for cell growth, survival, and migration (2). In various cell types, integrin clustering triggers tyrosine phosphorylation of signaling proteins through the activation of a large number of non-receptor protein tyrosine kinases (PTK) such as FAK and the Src-family of kinases. Increased tyrosine phosphorylation of intracellular proteins is one of the earliest responses stimulated by integrin receptor activation when cells contact matrix protein such as fibronectin (FN). Upon replating of cells on fibronectin, FAK becomes activated either through conformational changes or as a result of aggregation mediated by integrin clustering. FAK autophosphorylation at Tyr-397 under these conditions promotes the transient association of Src-family PTKs with FAK and the formation of a signaling complex. In addition, FAK associates with several different signaling proteins such as p130 Cas, Shc, Grb2, PI3- kinase, and paxillin. This enables FAK to function within a network of integrin stimulated signaling pathways leading to the activation of targets such as the ERK and JNK/mitogen activated protein kinase pathways (1-5). For many cellular processes, microtubules and the actin cytoskeleton dynamics must be coordinated. Recent studies suggest that there may be signal transduction systems that integrate the responses of the two systems (6-15). Previous data have suggested the existence 3

of a factor associated with the microtubules which can be released and affect assembly of the actin meshwork (16). Interestingly, microtubule breakdown stimulates stress fiber formation in fibroblasts (17), requires active Rho and involves elevated myosin light chain phosphorylation (14,18,19), suggesting a coordinate regulation of the actin cytoskeleton and the microtubule network. This is accompanied by assembly of adhesion plaques and tyrosine phosphorylation of the adhesion components FAK and paxillin (10). Tension exerted by microtubules on the actin cytoskeleton has been hypothesized to counteract stress fiber formation (10). Recently, it has been shown that there is direct physical interaction between the microtubules and adhesion sites, suggesting that relaxation signals are delivered, via the microtubules, to focal contacts in order to modulate the development of adhesions in a region-specific manner (11,15,20,21). We and others, have previously identified a novel Ste-20 related kinase (22-25), termed SLK, involved in cytoskeletal reorganization and apoptosis (24,25). Overexpression of SLK in various cell lines was shown to induce the rapid disassembly of actin stress fibers and cell death (25). Relatively high levels of SLK protein and activity have been observed in all cell types analysed, suggesting a functional role for SLK in physiological processes other than apoptosis. Because of its ability to induce cytoskeletal remodeling when overexpressed in a variety of cultured cell lines, we have analysed the levels, distribution and activity of SLK protein during cell spreading on fibronectin (FN), a process that requires extensive cellular remodeling (2,26). Using indirect immunofluorescence microscopy and biochemical techniques, we have determined that SLK is redistributed to large adhesion complexes during cell spreading on FN, and that it is associated with the microtubule network. Using microinjection studies we show that ectopic expression of activated SLK induces the disassembly of actin stress fibers, a process which can be inhibited by dominant negative Rac1. Finally, overexpresion of SLK by adenoviral infection inhibits cell spreading on FN. These results suggest that SLK may be delivered to 4

adhesion sites via the microtubule network where it regulates actin dynamics. 5

Experimental Procedures Cell lines and culture. The mouse fibroblast cell lines MEF-3T3 (MEF Tet-Off, C3018, Clontech) and Swiss 3T3 mouse embryonic fibroblast (ATCC cat no.4925) were used in all experiments. MEF-3T3 and Swiss 3T3 were maintained at 37 o C/5% CO2 in Dulbecco s modified Eagle s medium (DMEM, Bio-Whitaker) supplemented with 10% fetal bovine serum (FBS), 2mM L-glutamine, 50 ¼g/ml penicilin, and 50 ¼g/ml streptomycin. For fibronectin (FN) replating assays, sub-confluent MEF-3T3 cultures were serum starved in 0.25% FBS-DMEM for 24 hours and harvested by trypsin-edta treatment as previously described (27). The trypsin was inactivated using soybean trypsin inhibitor (0.5 mg/ml) and the cells were collected by centrifugation and resuspended in 0.1% BSA-DMEM medium. After 1 hour at 37 C in suspension, the cells were plated onto fibronectin-coated plates or coverslips. The culture dishes (or coverslips) were precoated with fibronectin (10 µg/ml, Sigma-Aldrich) or poly-l-lysine (100 µg/ml, Sigma- Aldrich) in PBS overnight at 4 C, rinsed with PBS and warmed to 37 C for one hour prior to replating. For cytoskeletal disruption experiments, MEF 3T3 cells were plated on FN-coated (10 µg/ml) plates and then serum-starved overnight on 0.25% FBS-DMEM. The plates were then treated with colchicine (10 µg/ml) or nocodazole (10 µm) for 30 minutes prior to lysis. Analysis of SLK protein expression and activity were performed by western blot and in vitro kinase assay as described below. To disrupt the microtubule network prior to FN-stimulation, colchicine was added to the cells in suspension in the final 30 min of incubation. For immunofluorescence studies, the coveslips were fixed in 4% paraformaldehyde for 10 minutes at room temperature, washed in PBS, and blocked with 0.1% BSA and 200 µg/ml ChromaPure Goat IgG (Jackson Immunoresearch Laboratories). SLK was detected using an 6

anti-slk rabbit polyclonal antibody (24) in conjunction with fluorescein isothiocyanate (FITC-) or tetramethyl rhodamine isothiocyanate (TRITC)-labeled secondary antibodies. Vinculin protein was detected with a mouse anti-vinculin monoclonal (clone VIN11-5, Sigma-Aldrich) in conjunction with FITC-labeled secondary antibodies. Tubulin protein was detected using an anti-α-tubulin monoclonal antibody (clone DM1, Sigma-Aldrich) and TRITC-labeled secondary antibodies. The anti-rac1 monoclonal antibody was obtained from Transduction Laboratories. Actin was detected by incubation with TRITC-conjugated phalloidin. Samples were visualized with a Zeiss Axioskop100 epifluorescence microscope equipped with appropriate filters and photographed with a digital camera (Sony Corporation HBO50) using the Northern Eclipse software package. Western blot and in vitro kinase assays. At various times after replating, the attached cells were rinsed in PBS and protein extracts were made in modified RIPA buffer containing 50 mm Tris-HCl (ph 7.4), 150mM NaCl, 1mM EDTA, 1% Triton X-100, 0.5 % sodium deoxycholate, and 0.1% sodium dodecyl sulfate (SDS), 1% NP40 and protease inhibitors (Sigma-Aldrich inhibitor cocktail). For SLK expression analysis, 20 ¼g of total cell lysate was subjected to Western blot with anti-slk rabbit polyclonal antibodies. Reactive proteins were detected by enhanced chemiluminescence using a goat anti-rabbit horseradish peroxidase (HRP)- labeled secondary antibody, and visualized using a chemiluminescence reagent (Perkin Elmer Life Science Inc.). For in vitro kinase assays, 200 ¼g of total cell lysate was immunoprecipitated using 2 ¼g anti-slk antibodies and 20 µl of ProteinA sepharose (Amersham Pharmacia) for 2 hours at 4 C. Immunoprecipitates were washed three times with NETN (50 mm Tris-HCl [ph 7.5], 150mM NaCl, 1mM EDTA, 0.1% Nonidet P-40) and once with kinase buffer (20mM Tris-HCl 7

[ph 7.5], 15mM MgCl2, 10 mm NaF, 10 mm ²-glycerophosphate, 1mM orthovanadate). Reactions (20 ¼l in kinase buffer) were initiated by the addition of 5 µci of γ[ 32 P]ATP. After a 30 minute incubation at 30 C, reactions were terminated by the addition of 4 x SDS sample buffer and 20 ¼l aliquots were fractionated by 8% SDS-PAGE. Gels were transferred to PVDF membranes, exposed to X-ray film and probed for SLK to evaluate the efficiency of the immunoprecipitation. For the detection of focal adhesion kinase (FAK), 50 ¼g of cell lysate was subjected to Western blot with anti-fak rabbit polyclonal IgG antibody (Upstate Biotechnology) or with anti-fak [py397] phosphospecific rabbit polyclonal antibody (Biosource International). To evaluate the level of paxillin tyrosine phosphorylation, 500 ¼g of total cell lysate in modified RIPA was immunoprecipitated using 2 ¼l of anti-paxillin monoclonal antibody (clone 349, Transduction Laboratories), washed in NETN and transferred to PVDF membranes (NEN Life Science Products). The membranes were probed with an anti-phosphotyrosine antibody (RC20:HRPO; Transduction Laboratories) and subjected to chemiluminescence detection (Perkin Elmer Life Science Inc). In co-immunoprecipitation experiments, equal amounts of total cell lysate (200 µg in modified RIPA or Triton-X buffer (50 mm Tris ph7.4, 150 mm NaCl, 1mM EDTA, 1% Triton-X 100) were immunoprecipitated with anti-slk, washed in NETN, resolved on 8% SDS-PAGE gels, transferred to PVDF membranes (NEN Life Science Products) and probed for the indicated proteins. Microinjection and adenovirus infection For microinjection studies, plasmid DNA in water (100ng/µl) was injected into the nuclei of cells plated on FN-coated coverslips using a semi-automated Eppendorf System equipped with a 8

5171 Micromanipulator on a Zeiss Axiovert S100 microscope. The SLK expression plasmids consisted of HA (hemagglutinin)-tagged SLK C-terminal truncation which results in SLK activation (HA-SLK1-373). Plasmid HA-SLK1-373 K63R is the kinase inactive version of HA- SLK1-373 (25). For co-injections, FLAG-tagged RacN17 and HA-SLK1-373 were mixed (each at 100ng/ml) and injected in MEF-3T3 cells. The injected cells were incubated for 3h (or 8h in co-injections) at 37 C, fixed in 4 % paraformaldehyde and processed for indirect immunofluorescence staining. Coverslips were double stained for HA, using anti-ha rabbit polyclonal antibody (sc-805, Santa Cruz Biotechnology, Inc.) and anti-vinculin or actin as described above. To quantitatively measure the phenotypic changes induced by SLK ectopic expression, cells that stained positive for the HA epitope and displayed an absence of cytoplasmic stress fibers were scored. At least 50 injected cells were counted in three separate experiments and the average is shown. In spreading assays, MEF-3T3 cells were infected with adenoviruses expressing either of the two HA-tagged truncations of SLK. The cells were serum-starved in 0.25% FBS-DMEM overnight and infected for 90 minutes at a MOI of 100 in serum-free medium and allowed to express the tagged proteins for 5h. The cells were then collected by centrifugation, held in suspension for 1h and plated onto fibronectin coated coverslips and allowed to spread for 10-20 minutes prior to fixation and immunofluorescence staining. To quantitatively assess the effect of SLK overexpression during cell spreading, HApositive cells that displayed a rounded, non-spread morphology (HA-SLK1-373) or increased density of stress fibers (HA-SLK1-373 K63R ) were enumerated. At least 100 HA-positive cells were counted in three separate experiments. 9

RESULTS We have previously demonstrated that overexpression of the Ste20-like kinase SLK induces actin stress fiber disassembly and cell death (24,25). Because of its ability to induce cytoskeletal remodeling when overexpressed in a variety of cultured cell lines, we analysed the levels, distribution and activity of SLK protein during cell spreading on fibronectin (FN), a process that requires extensive cellular remodeling (2,26). To gain insight into the role of SLK during normal cell growth, we first investigated the cellular distribution of endogenous SLK in exponentially growing MEF-3T3 cells. Supporting the observation that SLK can induce stress fiber disassembly, endogenous SLK distribution was found to correlate with an absence of actin stress fibers (see Figure 1), suggesting a role for SLK in the regulation of actin dynamics. Interestingly, the levels of SLK protein were also found to be variable, suggesting that its expression may be regulated throughout the cell cycle. The replating of suspended cells on FN-coated substrates has been shown to trigger the recruitment of adhesion complexes and subsequent actin polymerization from adhesion sites (2,28). To investigate the function of SLK in cytoskeletal remodeling events, MEF-3T3 cells were immunostained for SLK during adhesion and spreading shortly after FN stimulation. When MEF cells held in suspension were replated onto FN-coated substrates, SLK was found to be redistributed to the cell periphery and to colocalize with the adhesion protein vinculin (Figure 1E and F) and FAK (not shown). Surprisingly, SLK was observed to colocalize with vinculin at large structures resembling lamellipodia that formed abundantly 20 minutes postreplating (Figure 1E and F). A poor association was observed at discrete adhesion complexes that formed in the initial 10 minutes of spreading (Figure 1C and D), suggesting that SLK may be associated with adhesion complex disassembly or turnover. To investigate potential changes 10

in SLK kinase activity during cell spreading on FN, extracts from replated cells were prepared and subjected to Western blot analysis and in vitro kinase assays. Following replating and Western blot analysis, total SLK levels remained unchanged during cell spreading (Figure 2A). Interestingly, following in vitro kinase assay and normalization to total immunoprecipitated SLK (not shown), SLK activity remained unchanged following replating onto FN-coated plates (Figure 2A). Similarly, no changes were observed during cell adhesion (20 minutes) to poly-llysine-coated substrates. Over the same time period, phosphorylation of FAK at tyrosine 397 (28) and increased paxillin tyrosine phosphorylation (2,28) did occur, indicating that the adhesion components were activated (Figure 2B). These results suggest that SLK distribution is modulated by adhesion without any detectable changes in its kinase activity. One possibility is that specific redistribution of SLK leads to localized changes in substrate phosphorylation and downstream signaling events. SLK is associated with the microtubules and adhesion components during cell spreading. The observation that SLK colocalizes with adhesion components during late spreading events raises the possibility that it is implicated in adhesion complex disassembly or turnover. Recently, adhesion site contact by the microtubule network has been shown to promote their dissociation (11,15). In addition, disruption of microtubules has been shown to induce the assembly of focal adhesions and to stimulate the formation of actin stress fibers (10). Therefore, we investigated the distribution of SLK and α-tubulin in double immunostaining experiments. As shown in figure 3, a very strong colocalization was observed between the microtubule network and endogenous SLK protein in exponentially growing MEF cells (>95% of exponentially growing cells). In some areas at the cell periphery SLK staining presented as a filament-like pattern (see Figure 3C and D) that colocalized with the microtubule network, suggesting that 11

SLK is a microtubule-associated kinase. Interestingly, this association of SLK with the microtubules appeared to be preferentially non-centrosomal. Similarly, non-centrosomal microtubule nucleation has been observed at focal contacts in 3T3 cells during recovery from nocodazole or taxol treatment (15). To test whether SLK forms part of a complex with the microtubule, whole cell extracts obtained under different lysis conditions were subjected to immunoprecipitations using anti-slk followed by Western blot analysis. Our results show that under all the conditions tested, α-tubulin could be co-precipitated with SLK (Figure 3E). However, in GST pulldown assays using GST-tubulin, SLK did not interact directly with α- tubulin (not shown), suggesting that SLK is indirectly associated with the microtubule and that they are part of a higher order protein complex. Treatment of serum starved fibroblasts with microtubule disrupting agents has been previously shown to stimulate the formation of large adhesion complexes containing tyrosine phosphorylated FAK and paxillin at the cell periphery (10,14). To test whether SLK can redistribute to newly formed adhesion sites following microtubule depolimerization, serum starved MEF cells grown on FN were treated with nocodazole for 30 minutes and subjected to double immunostaining for SLK and vinculin or α-tubulin. Induction of focal adhesion assembly by nocodazole resulted in the redistribution of SLK to the cell periphery with vinculin and α- tubulin, suggesting that SLK can be targeted to newly formed adhesion sites (Figure 4D and E). Interestingly, this can be achieved in the absence of an intact microtubule network (Figure 4F and G). Microtubule disrupting agents have been shown to impair cell spreading and focal adhesion turnover (11). Therefore, we next examined whether SLK was also associated with the microtubule network during cell adhesion and spreading on FN. Replating of suspended cells on FN-coated substrates resulted in SLK localization to the cell periphery with polymerized 12

microtubules (Figure 5A and B). Interestingly, in the presence of colchicine, SLK was also colocalized with α-tubulin at the cell periphery even though the vast majority of the cells fail to spread (Figure 5C and D). Similarly, SLK was co-localized with large vinculin containing structures in colchicine treated cells (Figure 5E and F). Similar large adhesions were also observed to form following taxol freezing of microtubules (11). Overall, these results suggest that SLK is associated with the microtubule network but can also be targeted to adhesion sites independently of polymerized microtubules. Overexpression of SLK promotes cellular retraction and inhibits cell spreading To gain insight into the role of SLK during cell adhesion and spreading, MEF cells grown on FN-coated substrates were microinjected with epitope tagged SLK expression vectors encoding the activated truncation SLK1-373 (or the kinase inactive mutant SLK1-373 K63R ) and analysed for the presence of actin stress fibers and focal adhesions. Cells overexpressing activated SLK1-373 displayed a retracted morphology (rounded appearance and loosely adherent), a marked reduction in actin stress fibers and extensive actin reorganization at the periphery (Figure 6A and B). Quantitative analysis showed that more than 85% (86.6 ± 7%) of cells expressing SLK1-373 displayed that phenotype. In contrast, no change in stress fiber density was observed in cells injected with the kinase inactive SLK1-373 K63R (97 ± 2% retained stress fibers; Figure 6C and D). When SLK1-373-overexpressing cells were surveyed for focal adhesions using anti-vinculin antibodies, peripheral adhesion sites were smaller and fibrillar adhesions were greatly reduced or absent, consistent with the disassembly of actin stress fibers (Figure 6E and H). Overexpression of SLK1-373 K63R had no effect on the density, size and distribution of vinculin-containing adhesion sites in all injected cells (Figure 6G 13

and H). No obvious differences were observed in the organization of the microtubule network of microinjected cells (data not shown). These results suggest a functional role for SLK in the process of actin stress fiber disassembly rather than a direct destabilization of adhesion structures. The small GTPase Rac1 has been previously shown to induce stress fiber disassembly as well as lamellipodia formation (29). Interestingly, Rac1 also binds directly to the microtubule in its GTP-bound form (30) and becomes activated by microtubule growth in fibroblasts (21). Therefore, we tested the possible involvement of Rac1 in SLK-mediated stress fiber disassembly. As shown in figure 6I, 6J and 6K, co-injection of both HA-SLK1-373 and a dominant negative form of Rac1 (RacN17) resulted in a marked inhibition of stress fiber disassembly (86% compared to 32% in the presence of RacN17) and subsequent cellular retraction for up to 8 hours post-injection. Over the same period, cells expressing HA-SLK1-373 alone had completely retracted and displayed extensive blebbing, a hallmark of apoptotic cells. Furthermore, as for vinculin, in 60% (60±5%) of the cells SLK was found to colocalize with Rac1 following replating of MEF-3T3 cells onto FN (Figure 7). Together, these results suggest a functional interaction between these two proteins and that Rac1 may be a mediator of SLKinduced actin remodeling. Whether SLK directly regulates Rac1 or an upstream guanine exchange pathway remains to be elucidated. To test the effect of SLK overexpression on cell spreading, MEF cells were infected with adenoviral constructs encoding SLK1-373 or SLK1-373 K63R and then replated on FN-coated substrates for 10-20 minutes. Because overexpression of SLK1-373 induces a rapid apoptotic response (24,25), the cells were only allowed to express the HA-tagged constructs for 6 hours prior to plating. As shown in figure 8, cells expressing the kinase inactive protein SLK1-373 K63R assembled vinculin-containing adhesion sites but displayed an increased stress fiber 14

density and appeared larger than uninjected cells, with numerous lamellipodia (Figure 8 A, B, E and F). This phenotype was observed in 95±5% of HA-positive cells. In contrast, cells expressing activated SLK1-373 at relatively low levels (about 8% of HA-positive cells), displayed very small adhesion sites throughout the cell (Figure 8C and D). Cells (92±6% of HApositive cells) expressing relatively higher levels of SLK1-373 failed to spread and to assemble actin stress fibers on FN-coated substrates (Figure 8G and H). The failure to polymerize actin in SLK1-373 overexpressing cells may lead to adhesion site instability and an inhibition of contractile mechanisms leading to impaired cell spreading. These results are consistent with a role for SLK during late spreading events such as adhesion turnover, actin remodeling and membrane protrusion. DISCUSSION The processes regulating cellular adhesion and migration involve complex signaling mechanisms ultimately resulting in cytoskeletal remodeling, changes in cell shape and cell movement (2,6,26,31). We have recently identified a novel Ste20-like kinase activated during apoptosis and mediating actin reorganization (24,25). To investigate the role of SLK in cytoskeletal remodeling during normal cell growth, we analysed the expression, activity and cellular distribution of endogenous SLK protein following FN stimulation and cell spreading. Our results show that SLK colocalizes with the adhesion protein vinculin at podosomelike structures, which formed predominantly during the later phase of cell spreading (20 minutes) on FN in MEF-3T3 cells. These large vinculin-containing structures are suggestive of loosely assembled adhesion sites, resembling lamellipodia, suggesting that SLK may be involved in regulating cell spreading through destabilization of adhesion contacts. Supporting this, no colocalization with vinculin was observed early (10 minutes) during spreading when focal adhesions are assembling and adhesion components become tyrosine phosphorylated. In 15

addition, overexpression of activated SLK in exponentially growing cells or cells that were replated onto FN-coated substrates induced their retraction from the substrate and inhibited cell spreading, respectively. This appeared to be mediated by a loss of actin stress fibers as well as fibrillar adhesions, presumably located along the fibers. Furthermore SLK-induced actin fiber disassembly could be inhibited by dominant negative Rac (RacN17) suggesting a role for Rac1 downstream of SLK-mediated cytoskeletal remodeling events. These results support a role for SLK in actin dynamics during cell spreading, which may contribute to adhesion site destabilization and turnover (2,4,6,7,26). Recently, the microtubule network has been clearly implicated in adhesion site destabilization and turnover, a process that requires kinesin motors (10,11,13,15,32). Interestingly, Ballestrem and co-workers have demonstrated that microtubules are required for cell migration, tail retraction, and modulation of cell adhesion (32). Similarly, it has been proposed that regional contractility is modulated by the interplay of microtubule-linked events with focal adhesions (12). Interestingly, our results show that SLK is associated with a microtubule-containing protein complex in MEF-3T3 cells. In addition, we find that SLK colocalizes with vinculin-containing structures and the microtubule network during cell spreading. Combined with previous observations, our results suggest that one of the adhesion destabilizing signals delivered by the microtubule may be SLK. One possibility is that the rapid disassembly of actin stress fibers mediated by SLK may represent a regulatory mechanism for the control of microtubule-mediated suppression of contractility during remodeling events. Alternatively, the depolymerization of actin fibers by SLK may allow more efficient targeting of adhesion sites by the microtubule for further delivery of destabilizing signals (15). Direct interaction between SLK and α-tubulin was not observed by in vitro binding assays, suggesting an indirect interaction. One possibility is that post-translational modification of one 16

or both proteins is required for their interaction, a signal that could be transduced downstream of adhesion complex formation. Alternatively, an adapter protein is required to recruit SLK to the microtubule. Whether these interactions are tightly regulated through post-translational modifications downstream of adhesion complex formation remains to be investigated. The Rho family of small GTPases has been implicated as a mediator of cytoskeletal remodeling events such as the formation of stress fibers, lamellipodia and ruffling (29,33-35). The small GTPase Rac1 has been shown to be activated following microtubule growth and to promote lamellipodial protrusion (21). Specifically, GTP-Rac1 can directly bind β-tubulin (30), suggesting that it plays a role in cytoskeletal remodeling events regulated by microtubule growth such as adhesion disassembly. In addition, RhoA and mdia have been demonstrated to stabilize the microtubule network by capping (14,18,19), presumably to stabilize adhesion sites and the actin network (36). Our results show that SLK and Rac1 localization overlap extensively during cell spreading on FN at membrane protrusions resembling lamellipodia, cellular structures that can be induced by activated Rac1, suggesting that Rac1 may be a downstream effector of SLK. Furthermore, dominant negative Rac1 inhibits the cytoskeletal remodeling events induced by activated SLK, suggesting a functional interaction between these proteins. Recently, overexpression of β-tubulin1 in MDCK cells has been demonstrated to inhibit cell spreading and adhesion (8). In addition, delayed formation of adhesion sites were reported in these cells shortly after replating (8). Interestingly, microtubule-disrupting agents induce the formation adhesion sites and actin stress fibers, supporting the notion that the growing microtubule carries actin and adhesion destabilizing signals. Therefore, we propose a model whereby specific adhesion components, through direct interaction with tubulin, can target the microtubule network to adhesion sites (see Figure 9). Targeting of SLK to adhesion sites by the microtubules can lead to actin network destabilization, a process required for further cytoskeletal 17

remodeling and cell movement. Overall, we have established a novel molecular link between the adhesion destabilizing activity of the microtubule network and actin-based cytoskeletal remodeling events. Further studies will be required to identify upstream and downstream signals required for SLKdependent remodeling events implicating Rac1 and the microtubule network. 18

Acknowledgements We thank Dr. Michael Rudnicki for critically reviewing the manuscript. This work was supported by grants from the Canadian Institute of Health Research and the Muscular Dystrophy Association. 19

References 1. Giancotti, F. G., and Ruoslahti, E. (1999) Science 285(5430), 1028-32 2. Schlaepfer, D. D., Hauck, C. R., and Sieg, D. J. (1999) Prog Biophys Mol Biol 71(3-4), 435-78 3. Giancotti, F. G. (1997) Curr Opin Cell Biol 9(5), 691-700 4. Horwitz, A. R., and Parsons, J. T. (1999) Science 286(5442), 1102-3 5. Ilic, D., Damsky, C. H., and Yamamoto, T. (1997) J Cell Sci 110(Pt 4), 401-7 6. Small, J. V., Kaverina, I., Krylyshkina, O., and Rottner, K. (1999) FEBS Lett 452(1-2), 96-9. 7. Small, J. V., Rottner, K., and Kaverina, I. (1999) Curr Opin Cell Biol 11(1), 54-60. 8. Lezama, R., Castillo, A., Luduena, R. F., and Meza, I. (2001) Cell Motil Cytoskeleton 50(3), 147-60. 9. Zhou, X., Li, J., and Kucik, D. F. (2001) J Biol Chem 276(48), 44762-9. 10. Bershadsky, A., Chausovsky, A., Becker, E., Lyubimova, A., and Geiger, B. (1996) Curr Biol 6(10), 1279-89. 11. Kaverina, I., Krylyshkina, O., and Small, J. V. (1999) J Cell Biol 146(5), 1033-44. 12. Kaverina, I., Krylyshkina, O., Gimona, M., Beningo, K., Wang, Y. L., and Small, J. V. (2000) Curr Biol 10(12), 739-42. 13. Krylyshkina, O., Kaverina, I., Kranewitter, W., Steffen, W., Alonso, M. C., Cross, R. A., and Small, J. V. (2002) J Cell Biol 156(2), 349-60. 14. Liu, B. P., Chrzanowska-Wodnicka, M., and Burridge, K. (1998) Cell Adhes Commun 5(4), 249-55. 15. Kaverina, I., Rottner, K., and Small, J. V. (1998) J Cell Biol 142(1), 181-90. 16. Gundersen, G. G., and Cook, T. A. (1999) Curr Opin Cell Biol 11(1), 81-94. 20

17. Danowski, B. A. (1989) J Cell Sci 93(Pt 2), 255-66. 18. Cook, T. A., Nagasaki, T., and Gundersen, G. G. (1998) J Cell Biol 141(1), 175-85. 19. Enomoto, T. (1996) Cell Struct Funct 21(5), 317-26. 20. Waterman-Storer, C. M., and Salmon, E. (1999) Curr Opin Cell Biol 11(1), 61-7. 21. Waterman-Storer, C. M., Worthylake, R. A., Liu, B. P., Burridge, K., and Salmon, E. D. (1999) Nat Cell Biol 1(1), 45-50. 22. Itoh, S., Kameda, Y., Yamada, E., Tsujikawa, K., Mimura, T., and Kohama, Y. (1997) Arch Biochem Biophys 340(2), 201-7 23. Pytowski, B., Hicklin, D. J., Kornhaber, G., Dellaratta, D. V., and Witte, L. (1998) Arch Biochem Biophys 359(2), 310-9 24. Sabourin, L. A., and Rudnicki, M. A. (1999) Oncogene 18, 7566-7575 25. Sabourin, L. A., Tamai, K., Seale, P., Wagner, J., and Rudnicki, M. A. (2000) Mol Cell Biol 20, 684-696 26. Small, J. V., Rottner, K., Kaverina, I., and Anderson, K. I. (1998) Biochim Biophys Acta 1404(3), 271-81. 27. Sieg, D. J., Hauck, C. R., and Schlaepfer, D. D. (1999) J. Cell Science 112, 2677-2691 28. Schaller, M. D., and Parsons, J. T. (1994) Curr Opin Cell Biol 6(5), 705-10 29. Van Aelst, L., and D Souza-Schorey, C. (1997) Genes Dev 11(18), 2295-322 30. Best, A., Ahmed, S., Kozma, R., and Lim, L. (1996) J Biol Chem 271(7), 3756-62 31. Parsons, J. T., Schaller, M. D., Hildebrand, J., Leu, T. H., Richardson, A., and Otey, C. (1994) J Cell Sci Suppl 18, 109-13 32. Ballestrem, C., Wehrle-Haller, B., Hinz, B., and Imhof, B. A. (2000) Mol Biol Cell 11(9), 2999-3012. 33. Oliferenko, S., Kaverina, I., Small, J. V., and Huber, L. A. (2000) J Cell Biol 148(6), 21

1159-64. 34. Ridley, A. J., Allen, W. E., Peppelenbosch, M., and Jones, G. E. (1999) Biochem Soc Symp 65, 111-23 35. Ridley, A. J., Paterson, H. F., Johnston, C. L., Diekmann, D., and Hall, A. (1992) Cell 70(3), 401-10 36. Hollenbeck, P. (2001) Curr Biol 11(20), R820-3. 37. Herreros, L., Rodriguez-Fernandez, J. L., Brown, M. C., Alonso-Lebrero, J. L., Cabanas, C., Sanchez-Madrid, F., Longo, N., Turner, C. E., and Sanchez-Mateos, P. (2000) J Biol Chem 275(34), 26436-40. 22

Figure Legends Figure 1. SLK localizes to podosome-like structures during cell adhesion and spreading. Exponentially growing MEF cells immunostained for SLK (A) and F-actin (B). Endogenous SLK expression was found to be absent in actin fiber-rich regions (arrows). The asterisk shows a cell bearing stress fibers expressing low levels of SLK. Following replating of MEF cells on FNcoated coverslips (10 min), endogenous SLK was found to be predominantly perinuclear (C) and not associated with newly formed adhesion complexes detected by an anti-vinculin antibody (D). At later time points following replating on FN (20 min), a proportion of SLK (E) was found to be associated with vinculin (F) in diffuse podosome-like adhesion structures (arrows). This colocalization was observed in 60±7% of the cells (n=200) 20 min after replating. The cells were photographed at 400x (A and B) and 1000x (C-F). Figure 2. SLK is not activated during cell adhesion and spreading on fibronectin. (A) Serum starved MEF cells (c) were held in suspension (s), replated on FN-coated dishes (or polylysine; PL) and sampled at timed intervals for SLK expression and activity. The activation of FAK (determined by Y397 phosphorylation) and tyrosine phosphorylation of paxillin were analysed for comparison (B). Normalization to total immunoprecipitated SLK (not shown) shows that SLK is not activated following FN stimulation. In contrast, FAK and paxillin were activated at 10 min following replating. Figure 3. SLK colocalizes with the microtubule network. Exponentially growing Swiss 3T3 cells were double labeled for both SLK (A and C) and tubulin (B and D). Strong colocalization between SLK and microtubules was observed in all cells (arrows). (E) Cells exponentially 23

growing on FN were immunoprecipitated with anti-slk subjected to western blot analysis using an anti-α-tubulin monoclonal. Co-precipitation α-tubulin was more efficient in the triton X-100 and RIPA fraction than in a soluble NP-40 extract. Photomicrographs are shown at 630x (A and B) and 1000x (C and D). Figure 4. SLK is redistributed to adhesion complexes induced by microtubule disruption. Serum starved cells were immunostained for SLK (A), vinculin (B) α-tubulin (C) prior to the addition of nocodazole. Addition of nocodazole to the cultures for 30 min induced the formation of large adhesion complexes (E) and the redistribution of SLK to these sites (D). Similarly, SLK was also localized to these sites (F) along with depolymerized α-tubulin (G). Photomicrographs are shown at 630x. Figure 5. SLK colocalizes with the microtubule network during adhesion and spreading. Cells suspended in serum-free medium were replated onto FN-coated coverslips for 20 min in the absence (A and B) or in the presence (C-E) of nocodazole and immunostained for SLK, a- tubulin and vinculin. SLK (A) was observed to colocalize with the microtubule network (B) during cell spreading on FN. Similarly, in the few cells that spread in the presence of colchicine, SLK (C) was also found to colocalize with depolymerized α-tubulin (D; arrows). As for untreated cells, SLK (E) was also colocalized with vinculin (F) at diffuse adhesion sites in colchicine treated cells (arrows). Photomicrographs are shown at 630x. Figure 6. Microinjection of activated SLK induces actin stress fiber disassembly. MEF cells plated on FN-coated coverslips were microinjected with HA-tagged SLK1-373 and immunostained 3h post-injection. (A and E) Wildtype SLK1-373-injected cells (or the inactive 24

kinase SLK1-373K63R; C and G) were double labeled with anti-ha (A, C, E, G) and TRITCphalloidin (B and F) or anti-vinculin (D and H). Co-injection of dominant negative Rac1 (RacN17) inhibited stress fiber disassembly and cell retraction for up to 8 hours post-injection (I and J). Prolonged expression (8 hours) of SLK1-373 induced retraction and extensive blebbing (I and J; insets). Cells expressing active SLK1-373 displayed altered morphology with actin redistribution to the periphery (arrow in B) and much smaller fibrillar adhesions (double arrow in F). Peripheral adhesions did not appear to be affected (arrow in F). No effect on stress fibers or adhesion sites was observed in SLK1-373K63R expressing cells. Photomicrographs are shown at 630x. (K) Cells that stained positive for the HA epitope and displayed an absence of cytoplasmic stress fibers were scored. At least 50 injected cells were counted in three separate experiments and the average is shown. Stress fiber disassembly induced by expression of SLK1-373 (87±7% of the cells) could be inhibited by co-injection with RacN17 (loss of stress fibers in 32±7% of the injected cells following RacN17 co-injection). Figure 7. Rac1 and SLK co-localization in spreading MEF-3T3 cells. MEF cells held in suspension were replated for 20 min and double labeled for SLK (A) and Rac1 (B). Colocalization was observed at the cell periphery in 60±5% (n=200) of spreading cells (C). Figure 8. Overexpression of activated SLK inhibits cell spreading on FN. MEF monolayers in serum-free medium were infected with Adenoviral vectors carrying HA-tagged SLK1-373 (C and G) (or inactive kinase K63R; A and E), held in suspension and replated on FN-coated coverslips for 20 min. The cells were immunostained using anti-ha and TRITC-phalloidin (F and H) or anti-vinculin antibodies (B and D). Over 90% of SLK1-373 expressing cells (92±6%) failed to spread and did not assemble actin fibers (arrows in G and H). Cells expressing lower 25

levels of SLK1-373 (about 8% of HA-positive cells; see panel C) were able to spread but displayed much smaller adhesion sites. In contrast, 95±5% of cells expressing the kinase inactive SLK1-373K63R displayed an increased density of stress fibers. For both expression plasmids at least 100 HA-positive cells were counted in three separate experiments. Uninfected cells are marked with an asterisk. Photomicrographs are shown at 630x. Figure 9. Proposed model for SLK recruitment and regulation of actin dynamics at adhesion sites. Following integrin binding, adhesion sites assemble through the recruitment of FAK and structural proteins. During adhesion site turnover and disassembly, microtubule (MT) targeting of the adhesion sites, perhaps by direct interaction with paxillin (37), recruits SLK (possibly through interaction with an unknown protein) to the adhesion sites where it regulates actin dynamics. The disassembly of actin stress fibers induced by SLK may be mediated by Rac1 promoting the relaxation and destabilization of adhesion sites, a process that would likely involve additional regulators. 26