Top Banner
2880 Research Article Introduction Prostate cancer is the most commonly diagnosed cancer and is the second leading cause of cancer mortality in US males. This is a complex disease to which many genetic and environmental factors contribute. As with all carcinomas, prostate cancer results from mutations in oncogenes and tumor suppressor genes that cause sequential changes in cellular behavior. Of these changes, those causing a loss of epithelial cellular adhesion, acquisition of an invasive motile phenotype and ability to metastasize to secondary sites are the most devastating, but also the least understood at the molecular level. To gain a better understanding of the molecular details underlying the complex series of events leading to advanced prostate cancer, a potentially rewarding approach is to focus on proteins that function in normal epithelial cells to maintain strong intercellular adhesion and promote normal cell polarity. In this regard, E-cadherin is involved in Ca 2+ -dependent cell–cell adhesion and establishment of epithelial polarity (Yeaman et al., 1999). Loss of E-cadherin function is associated with increased cellular invasiveness and dedifferentiation of many carcinomas (Takeichi, 1993), and aggressive prostate tumors of both rat and human origin exhibit decreased E-cadherin expression (Bussemakers et al., 1992; Ross et al., 1994). In addition, E-cadherin-mediated cell–cell adhesion is important for recruitment and assembly of proteins involved in tethering post-Golgi transport vesicles to sites of membrane growth and remodeling during cell polarization (‘targeting patches’) (Grindstaff et al., 1998; Yeaman et al., 1999). One important component of targeting patches is the Exocyst, a hetero-octameric protein complex first identified in budding yeast but later found to be ubiquitously expressed in eukaryotes (TerBush et al., 1996; Wang and Hsu, 2006). In budding yeast, its localization corresponds to sites of vesicle docking and fusion throughout the cell cycle (Finger and Novick, 1998; TerBush and Novick, 1995). In epithelial cells, Exocyst redistributes from cytosol to plasma membrane sites of cell–cell contact upon initiation of cadherin-mediated adhesion, and serves to ensure efficient delivery of post-Golgi transport vesicles to these sites (Grindstaff et al., 1998; Yeaman et al., 2004). To function in tethering secretory vesicles to sites of exocytosis, Exocyst must contact both cargo-laden transport vesicles and target sites on the plasma membrane (Guo et al., 1999). Studies in yeast have shown that Exocyst holocomplex assembly involves association between plasma membrane-bound subunits that mark sites of exocytosis and vesicle-associated subunits (Boyd et al., 2004; Guo et al., 1999). Similarly, cell fractionation studies indicate that the mammalian Exocyst may be present as distinct hemicomplexes on vesicle and plasma membranes, and that holocomplex assembly mediates the tethering event (Moskalenko et al., 2003). Exocyst localization, assembly and function is regulated by at least four small GTPases representing members of the Rab, Rho, Arf and Ral subfamilies (Guo et al., 1999; Moskalenko et al., 2002; Novick and Guo, 2002; Prigent et al., 2003). Two of these, Arf6 and Ral, are associated with cell invasiveness and metastasis (Hashimoto et al., 2004; Tchevkina et al., 2005). RalA and RalB are closely related GTPases (~82% Changes in cellular behavior that cause epithelial cells to lose adhesiveness, acquire a motile invasive phenotype and metastasize to secondary sites are complex and poorly understood. Molecules that normally function to integrate adhesive spatial information with cytoskeleton dynamics and membrane trafficking probably serve important functions in cellular transformation. One such complex is the Exocyst, which is essential for targeted delivery of membrane and secretory proteins to specific plasma membrane sites to maintain epithelial cell polarity. Upon loss of cadherin-mediated adhesion in Dunning R3327-5A prostate tumor cells, Exocyst localization shifts from lateral membranes to tips of protrusive membrane extensions. Here, it colocalizes and co-purifies with focal complex proteins that regulate membrane trafficking and cytoskeleton dynamics. These sites are the preferred destination of post-Golgi transport vesicles ferrying biosynthetic cargo, such as α 5 -integrin, which mediates adhesion of cells to the substratum, a process essential to cell motility. Interference with Exocyst activity impairs integrin delivery to plasma membrane and inhibits tumor cell motility and matrix invasiveness. Localization of Exocyst and, by extension, targeting of Exocyst- dependent cargo, is dependent on Ral GTPases, which control association between Sec5 and paxillin. Overexpression of Ral- uncoupled Sec5 mutants inhibited Exocyst interaction with paxillin in 5A cells, as did RNAi-mediated reduction of either RalA or RalB. Reduction of neither GTPase significantly altered steady-state levels of assembled Exocyst in these cells, but did change the observed localization of Exocyst proteins. Supplementary material available online at http://jcs.biologists.org/cgi/content/full/121/17/2880/DC1 Key words: Cell motility, Cell polarity, Exocyst, Metastasis, Ral Summary Ral-regulated interaction between Sec5 and paxillin targets Exocyst to focal complexes during cell migration Krystle S. Spiczka and Charles Yeaman* Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA *Author for correspondence (e-mail: [email protected]) Accepted 16 June 2008 Journal of Cell Science 121, 2880-2891 Published by The Company of Biologists 2008 doi:10.1242/jcs.031641 Journal of Cell Science
12

Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

Apr 28, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2880 Research Article

IntroductionProstate cancer is the most commonly diagnosed cancer and is thesecond leading cause of cancer mortality in US males. This is acomplex disease to which many genetic and environmental factorscontribute. As with all carcinomas, prostate cancer results frommutations in oncogenes and tumor suppressor genes that causesequential changes in cellular behavior. Of these changes, thosecausing a loss of epithelial cellular adhesion, acquisition of aninvasive motile phenotype and ability to metastasize to secondarysites are the most devastating, but also the least understood at themolecular level.

To gain a better understanding of the molecular details underlyingthe complex series of events leading to advanced prostate cancer,a potentially rewarding approach is to focus on proteins that functionin normal epithelial cells to maintain strong intercellular adhesionand promote normal cell polarity. In this regard, E-cadherin isinvolved in Ca2+-dependent cell–cell adhesion and establishmentof epithelial polarity (Yeaman et al., 1999). Loss of E-cadherinfunction is associated with increased cellular invasiveness anddedifferentiation of many carcinomas (Takeichi, 1993), andaggressive prostate tumors of both rat and human origin exhibitdecreased E-cadherin expression (Bussemakers et al., 1992; Rosset al., 1994). In addition, E-cadherin-mediated cell–cell adhesionis important for recruitment and assembly of proteins involved intethering post-Golgi transport vesicles to sites of membrane growthand remodeling during cell polarization (‘targeting patches’)(Grindstaff et al., 1998; Yeaman et al., 1999). One important

component of targeting patches is the Exocyst, a hetero-octamericprotein complex first identified in budding yeast but later found tobe ubiquitously expressed in eukaryotes (TerBush et al., 1996; Wangand Hsu, 2006). In budding yeast, its localization corresponds tosites of vesicle docking and fusion throughout the cell cycle (Fingerand Novick, 1998; TerBush and Novick, 1995). In epithelial cells,Exocyst redistributes from cytosol to plasma membrane sites ofcell–cell contact upon initiation of cadherin-mediated adhesion, andserves to ensure efficient delivery of post-Golgi transport vesiclesto these sites (Grindstaff et al., 1998; Yeaman et al., 2004).

To function in tethering secretory vesicles to sites of exocytosis,Exocyst must contact both cargo-laden transport vesicles and targetsites on the plasma membrane (Guo et al., 1999). Studies in yeasthave shown that Exocyst holocomplex assembly involvesassociation between plasma membrane-bound subunits that marksites of exocytosis and vesicle-associated subunits (Boyd et al.,2004; Guo et al., 1999). Similarly, cell fractionation studies indicatethat the mammalian Exocyst may be present as distincthemicomplexes on vesicle and plasma membranes, and thatholocomplex assembly mediates the tethering event (Moskalenkoet al., 2003). Exocyst localization, assembly and function isregulated by at least four small GTPases representing members ofthe Rab, Rho, Arf and Ral subfamilies (Guo et al., 1999;Moskalenko et al., 2002; Novick and Guo, 2002; Prigent et al.,2003). Two of these, Arf6 and Ral, are associated with cellinvasiveness and metastasis (Hashimoto et al., 2004; Tchevkina etal., 2005). RalA and RalB are closely related GTPases (~82%

Changes in cellular behavior that cause epithelial cells to loseadhesiveness, acquire a motile invasive phenotype andmetastasize to secondary sites are complex and poorlyunderstood. Molecules that normally function to integrateadhesive spatial information with cytoskeleton dynamics andmembrane trafficking probably serve important functions incellular transformation. One such complex is the Exocyst, whichis essential for targeted delivery of membrane and secretoryproteins to specific plasma membrane sites to maintain epithelialcell polarity. Upon loss of cadherin-mediated adhesion inDunning R3327-5�A prostate tumor cells, Exocyst localizationshifts from lateral membranes to tips of protrusive membraneextensions. Here, it colocalizes and co-purifies with focalcomplex proteins that regulate membrane trafficking andcytoskeleton dynamics. These sites are the preferred destinationof post-Golgi transport vesicles ferrying biosynthetic cargo, suchas α5-integrin, which mediates adhesion of cells to the

substratum, a process essential to cell motility. Interference withExocyst activity impairs integrin delivery to plasma membraneand inhibits tumor cell motility and matrix invasiveness.Localization of Exocyst and, by extension, targeting of Exocyst-dependent cargo, is dependent on Ral GTPases, which controlassociation between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction withpaxillin in 5�A cells, as did RNAi-mediated reduction of eitherRalA or RalB. Reduction of neither GTPase significantly alteredsteady-state levels of assembled Exocyst in these cells, but didchange the observed localization of Exocyst proteins.

Supplementary material available online athttp://jcs.biologists.org/cgi/content/full/121/17/2880/DC1

Key words: Cell motility, Cell polarity, Exocyst, Metastasis, Ral

Summary

Ral-regulated interaction between Sec5 and paxillintargets Exocyst to focal complexes during cellmigrationKrystle S. Spiczka and Charles Yeaman*Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA*Author for correspondence (e-mail: [email protected])

Accepted 16 June 2008Journal of Cell Science 121, 2880-2891 Published by The Company of Biologists 2008doi:10.1242/jcs.031641

Jour

nal o

f Cel

l Sci

ence

Page 2: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2881Ral targets Exocyst to focal contacts

identical) that interact with two different Exocyst subunits (Sec5and Exo84) in vitro (Jin et al., 2005; Moskalenko et al., 2003).Recent evidence suggests that each GTPase regulates differentExocyst activities in vivo (Chien et al., 2006; Lim et al., 2005; Rosseet al., 2006; Shipitsin and Feig, 2004). Ral GTPases may be activatedby a family of guanine nucleotide exchange factors (RalGEFs), fourof which (RalGDS, RGL1, RGL2 and Rgr) are downstreameffectors of Ras (Hofer et al., 1994; Spaargaren and Bischoff, 1994).In recent years, it has become increasingly clear that RalGEFs, andby extension Ral GTPases, mediate many of the prometastaticfunctions of oncogenic Ras mutants (Bodemann and White, 2008;Camonis and White, 2005; Feig, 2003). Although the Exocyst hasbeen implicated in some of these activities, the process of identifyingthe exact function of this complex in tumorigenesis is far fromcomplete.

Because polarization of epithelial cells is dependent, in part, onadhesive spatial cues that direct recruitment and assembly ofExocyst to sites of membrane growth, it is of interest to determinethe fate and function of this essential protein complex in cells thathave lost the ability to undergo cadherin-mediated intercellularadhesion. In this study, Exocyst assembly and activity is examinedin metastatic prostate tumor cells. The guiding hypothesis is thatfollowing loss of E-cadherin expression in prostatic tumor cells,Exocyst responds to distinct spatial cues, assumes a novellocalization within protrusive cell extensions, and functions to directthe targeted delivery of membrane components required duringinvasive cell motility.

ResultsLoss of E-cadherin expression drives Exocyst recruitment toprotrusive cell extensions in metastatic prostate tumor cellsExpression, assembly status and localization of each Exocystsubunit was examined in two clonal cell lines derived from theDunning rat prostate tumor model (Luo et al., 1997). R3327-5�Bcells express several cadherins, including E-cadherin, formmonolayers in culture, are poorly invasive in vitro and are non-metastatic in vivo. By contrast, R3327-5�A cells do not expressdetectable levels of cadherins, exhibit a fibroblast-like morphologyand have enhanced invasive and metastatic potential compared with5�B cells. In vitro, 5�A cells are highly motile, migrating byextending long slender cellular processes. During ECM invasion,these cells assemble circular dorsal ruffles (invadopodia) throughwhich matrix metalloproteinases are secreted.

Quantitative immunoblot analysis shows that 5�A and 5�B cellsexpress almost identical levels of each Exocyst subunit (Fig. 1A).Furthermore, quantitative immunoprecipitation with anti-Sec8antibodies recovered equivalent amounts of each Exocyst subunitfrom 5�A and 5�B cells, indicating that the status of Exocystholocomplex assembly is similar in metastatic and non-metastaticprostate tumor cells (Fig. 1A).

Localization of endogenous Sec6 and Sec8 (Fig. 1B), as well asother Exocyst subunits (Sec3, Sec5, Sec10, Sec15, Exo70 andExo84; data not shown), was examined in 5�A and 5�B cells byimmunofluorescence microscopy. This analysis revealed strikingdifferences in subcellular localization of Exocyst complexes in thetwo cell types. In non-invasive 5�B cells, Sec6 and Sec8 wereassociated with E-cadherin-based adherens junctions along lateralplasma membranes between adjacent cells. No labeling with Sec6/8antibodies was observed at free, non-contacting membranes of cells.This distribution is similar to that reported previously for kidneyepithelial cells (Grindstaff et al., 1998; Yeaman et al., 2004). By

contrast, in 5�A cells Sec6 and Sec8 were observed to accumulatein a juxtanuclear region and also within specific sites at the cellperiphery, showing especially high concentrations at distal tips ofprotrusive cell extensions (Fig. 1B, arrows). Tip staining was notobserved in all cells. Consistent with previous findings that Exocyst

Fig. 1. Exocyst expression, assembly and localization in non-metastatic andmetastatic prostate tumor cells. (A) Dunning rat R3327-5�A (‘A’) and R3327-5�B (‘B’) cells were extracted in 1% Triton X-100. Extracts were subjected toimmunoprecipitation with antibodies to Sec8. Presence of Sec3, Sec5, Sec6,Sec8, Sec10, Sec15, Exo70 and Exo84 in equivalent amounts of whole cellextracts (‘total’) and precipitated immune complexes (‘αSec8’) was assessedby SDS-PAGE followed by immunoblotting with specific antibodies. (B) Subconfluent cultures of R3327-5�B, R3327-5�A or R3327-5�A cellsstably expressing human E-cadherin were cultured on type I collagen-coatedcoverslips, and then processed for immunofluorescent staining with antibodiesto Sec6 or Sec8, as described in the Materials and Methods. Samples wereviewed with a Nikon Microphot-FX microscope (63� objective) andepifluorescent digital images were obtained using a Kodak DCS 760 digitalcamera. Arrows indicate accumulations of Exocyst proteins in protrusiveextensions of R3327-5�A cells. (C) Sub-confluent cultures of R3327-5�A cellswere cultured on Matrigel-coated coverslips, and then processed forimmunofluorescent staining with phalloidin (to label f-actin) and antibodies toSec6. Arrows indicate an accumulation of Exocyst within an actin-richinvadopodium. Scale bars: 20 μm.

Jour

nal o

f Cel

l Sci

ence

Page 3: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2882

recruitment to intercellular junctions is dependent on cadherin-mediated cell–cell adhesion (Yeaman et al., 2004), noimmunostaining of Sec6 or Sec8 was ever observed at sites ofcell–cell contact in 5�A cultures, even when cells were seeded athigh density (Fig. 1B). In addition, Exocyst labeling wasoccasionally observed in invadopodia when 5�A cells were seededon Matrigel-coated coverslips (Fig. 1C, arrowhead). Interestingly,when invadopodia localization was evident in a cell, Exocystaccumulation within pseudopod tips appeared to be reduced orabsent.

Exocyst enrichment within pseudopods was quantified using afilter assay. This is based on the finding that motile cells, whenseeded on porous filter supports (3 μm pores), will attempt tomigrate towards a source of chemoattractant placed in the lowerchamber, but are prevented from doing so because their nuclei aretoo large to pass through the pores in the filter (Cho and Klemke,2002). Hence, only protrusive cellular extensions at the leading edgeof cells extend through the filter, and these can be isolated fromthe remainder of the cell by removing cell bodies from the top ofthe filter with a cotton swab. The relative enrichment of eachExocyst subunit within pseudopods was quantified and twodistribution patterns were observed (Fig. 2A). Some subunits (Sec3,Sec5, Sec6, Sec8 and Exo70) were roughly 1.5-2.7-fold moreconcentrated within the protrusions than they were elsewhere in thecell. By contrast, other subunits (Sec10, Sec15 and Exo84) werenot enriched in pseudopods. That these latter subunits were notconcentrated in protrusive cell extensions was supported byimmunofluorescence microscopy (Fig. 2B).

If the Exocyst functions to tether post-Golgi transport vesiclesto sites of membrane fusion at distal tips of pseudopods andinvadopodia, then it would be anticipated that other proteins thatmediate membrane fusion would also accumulate at these sites.Therefore, the localization of two plasma membrane t-SNAREs(syntaxins 3 and 4) and proteins that regulate their activities(Munc18b and Munc18c) were also examined in 5�A cells. Likethe Exocyst, each of these proteins was found to localize to both ajuxtanuclear region and pseudopod tips (Fig. 2B). The appearanceof tip accumulation was not merely a consequence of tips beingthicker than other regions of the cell periphery, because cytosolicgreen fluorescent protein (GFP) did not accumulate at these siteswhen expressed in cells (Fig. 2B). Quantification of pseudopodaccumulation revealed that both t-SNAREs and their cognateMunc18 proteins were ~1.5-fold more concentrated withinprotrusions than they were elsewhere in the cell.

If loss of E-cadherin expression in invasive prostate tumor cellsis responsible for redirecting Exocyst assembly to pseudopods, thenit should be possible to restore lateral plasma membrane Exocystlocalization by re-expressing E-cadherin in these cells. To test thisprediction, clonal populations of 5�A cells stably transfected withhuman E-cadherin cDNA were examined. It has previously beenreported that ectopic E-cadherin expression was sufficient to repressthe elevated invasive and metastatic potential of these cells (Luoet al., 1999). In addition, E-cadherin expression in 5�A cells droverecruitment of Exocyst complexes to intercellular adhesion sitesand promoted development of epithelial morphology, similar to thatobserved in non-metastatic 5�B cells (Fig. 1B). Together, theseresults highlight an important function for E-cadherin in regulatingthe subcellular localization of the Exocyst in prostate tumor cells,and are consistent with the hypothesis that loss of E-cadherin maycontribute to tumorigenesis by facilitating recruitment of Exocystcomplexes into pseudopods and invadopodia.

Journal of Cell Science 121 (17)

Exocyst is associated with paxillin-containing focal complexeswithin protrusive cellular extensions at the leading edge ofmigrating prostate tumor cellsIf metastatic prostate tumor cells organize Exocyst assembly withinpseudopods in order to target the delivery of cargo to these sitesduring cell migration, then the Exocyst should be enriched at sitesof newly forming cell–substratum adhesions at the leading edge ofcells, but not with older cell–substratum contacts, such as focaladhesions at the center or rear of cells. In order to promote directedmotility of prostate tumor cells, cultures of 5�A cells were seededat high density and experimental wounds were introduced by

Fig. 2. Exocyst subunits and SNARE proteins are enriched within protrusivecell extensions. (A) Dunning rat R3327-5�A prostatic tumor cells were seededon 75 mm Transwell filters (3.0 μm pore size) and medium containing 10%FBS was added basolaterally to stimulate pseudopod extension, as described inthe Materials and Methods. An enriched pseudopod fraction was obtained byremoving cell bodies from the top of filters with a cotton swab. Indicatedproteins were identified by immunoblotting with specific antibodies, andprotein levels were quantified using a Molecular Dynamics Typhoonphosphorimager. To determine the fold enrichment of each protein withinpseudopods, values were normalized to protein levels present in an equivalentamount of whole cell extract. (B) Distribution of endogenous Sec6, Sec8,Sec15, Exo84, syntaxin 3, syntaxin 4, Munc18c and exogenous GFP in 5�Acells. Subconfluent cultures of R3327-5�A cells were cultured on type Icollagen, and then processed for immunofluorescent staining with indicatedantibodies, as described in the Materials and Methods. Bound antibodies weredetected with appropriate FITC or Texas Red-conjugated secondaryantibodies, and epifluorescence images were obtained. Arrows and double-headed arrows indicate accumulation of membrane trafficking components atthe tips of pseudopods. Accumulation of Sec15, Exo84 and GFP was neverobserved within pseudopods. Scale bar: 20 μm.

Jour

nal o

f Cel

l Sci

ence

Page 4: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2883Ral targets Exocyst to focal contacts

scratching the culture. Behavior of cells at the wound margin wasthen observed.

Paxillin is a molecular scaffold that organizes signaling proteinsresponsible for remodeling the plasma membrane and the actincytoskeleton to orchestrate protrusive activity required for cellmotility (Turner et al., 2001). In migrating fibroblasts, paxillin isassociated with newly forming focal complexes at the leading edge,as well as with stable focal adhesions at the center and rear of cells.Immunofluorescent labeling studies were performed to determinewhether the Exocyst colocalizes with components of paxillin-basedsignaling complexes. These included paxillin itself (Fig. 3A), theArf6 GTPase-activating protein GIT1, the SH2/SH3 adaptor proteinNck1/2 and the Rac guanine nucleotide exchange factor β-PIX (Fig.3B). That each of these proteins was expressed by 5�A cells andthat antibodies to each were specific was confirmed by western blotanalysis (Fig. 3C).

Paxillin, GIT1, Nck1/2 and β-PIX were all enriched atpseudopods tips, similar to the Exocyst (Fig. 3). Each of theseproteins was also present at juxtanuclear sites. Close inspection ofcells by confocal microscopy revealed that Sec6 and paxillin werecolocalized at distal tips of pseudopods at the leading edge ofmigrating cells at the wound margin (Fig. 3A, lower panels,arrows), but that behind these sites, more proximal to cell bodies,the two proteins had distinct distributions (Fig. 3A, lower panels,arrowheads and asterisks). Importantly, paxillin was also enrichedwithin adhesion sites elsewhere in the cell, but Sec6 was not enrichedat these sites. Paxillin-associated signaling proteins were, like Sec6,enriched within pseudopods tips but did not appear to accumulatewithin other paxillin-positive structures (Fig. 3B). Therefore, theseresults indicate that Exocyst is enriched at sites containing newlyforming cell–substrate focal complexes at the leading edge ofmigrating cells, but is absent from older contact sites elsewhere incells.

Paxillin, β-PIX, GIT1 and Nck1/2 co-fractionate with Sec8 indensity gradients of post-nuclear supernatant obtained frommechanically homogenized 5�A prostate tumor cells (Fig. 4A).Analysis of gradient fractions by western blotting with specificantibodies reveals a peak of Sec8 in fractions corresponding to adensity (δ) of ~1.15-1.17 g/ml (Fig. 4A, peak ‘A’). Thus, Sec8 isassociated with membranes that have a higher density than the bulkof plasma membrane, as defined by the presence of Na+-K+ ATPase(Fig. 4A, NaK-ATPase, peak δ ~1.10-1.13 g/ml). A fraction ofpaxillin was recovered in the Sec8-containing peak, although asecond peak of paxillin was recovered in higher density fractionsthat did not contain a peak of Sec8 (Fig. 4A, peak ‘B’). Importantly,β-PIX and GIT1 were primarily recovered within the Sec8-containing fractions, and not within the higher density paxillin-containing fractions. The SH2/SH3 adaptor Nck1/2 was found toco-fractionate with both paxillin-containing peaks, but a slightlylarger (~47 kDa) protein that was detected by anti-Nck1/2 antibodieswas present exclusively in fractions that contained Sec8. This bandappears to represent a phosphorylated form of Nck1/2, becausetreatment of fractions with alkaline phosphatase caused it todisappear (data not shown). These results indicate that Sec8 co-fractionates in density gradients with membranes containing paxillinand associated proteins involved in regulating vesicle traffickingand cytoskeleton dynamics.

Results of density gradient fractionation suggest that fractionscontaining the peak of Sec8 (Fig. 4A, peak ‘A’) may containpseudopod tip complexes, whereas fractions containing the peak ofpaxillin (Fig. 4A, peak ‘B’) may contain older focal adhesion

Fig. 3. Exocyst colocalizes with focal complexes at pseudopod tips ofmigrating prostate tumor cells. (A) Distribution of endogenous Sec6 andpaxillin in R3327-5�A prostatic tumor cells. Cells were seeded on fibronectin-coated glass coverslips for 18 hours, then were fixed with 2%paraformaldehyde, permeabilized with 1% Triton X-100, incubated withmouse anti-Sec6 (mAb 9H5) and rabbit anti-paxillin antibodies, then stainedwith FITC-conjugated goat anti-mouse and Texas Red-conjugated donkeyanti-rabbit IgG. Epifluorescence images were obtained as described in Fig. 1.Arrows indicate tips of protrusive pseudopods, within which Sec6 and paxillinappear to colocalize. In lower panels, arrowheads indicate structures withinpseudopods that stained with anti-paxillin antibodies, but not anti-Sec6antibodies, and asterisks indicate structures that stained with anti-Sec6antibodies but not anti-paxillin antibodies. (B) Distribution of endogenousSec6, Git1, Nck1/2 and β-PIX in 5�A cells. Cells were cultured, fixed andpermeabilized as described in the Materials and Methods. Sec6 distributionwas compared with that of Git1, β-PIX and Nck1/2. Sec6/Git1 and Sec6/β-PIX images were collected by epifluorescence microscopy. Sec6/Nck1/2images were obtained with a Zeiss confocal laser-scanning microscope (63�objective) using a krypton/argon laser with 488 nm (FITC) and 568 nm (TexasRed) laser lines. (C) Specificity of antibodies. 5�A cells were lysed and ~1 μgof protein was loaded per lane onto a 10% SDS-PAGE gel. The proteins weretransferred to Immobilon PVDF membranes and incubated with rabbitpolyclonal antibodies to paxillin, GIT1 or Nck1/2 or mouse mAb to β-PIX.Blots were then probed with HRP-conjugated secondary antibodies anddeveloped for ECL detection.

Jour

nal o

f Cel

l Sci

ence

Page 5: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2884

complexes that do not contain Exocyst proteins. To confirm theprediction that Exocyst associates preferentially with focalcomplexes at the leading edge of migrating cells, paxillin-containingprotein complexes were immunoprecipitated from cell bodies orpseudopods isolated from 5�A cells induced to undergo frustratedchemotaxis, as described for Fig. 2. This analysis confirmed thatExocyst complexes were associated only with paxillin-containingcomplexes isolated from pseudopods, and not with more abundantpaxillin complexes associated with other cellular structures (Fig.4B).

To determine whether the Exocyst directly binds paxillin, and toidentify which subunit is responsible for this interaction, eachindividual Exocyst subunit was expressed together with paxillin incoupled in vitro transcription/translation reactions. This analysisrevealed a direct interaction between Sec5 and paxillin (Fig. 4C).None of the other Exocyst subunits associated with paxillin in thisassay (data not shown). This interaction was independent of the N-terminal Ral-binding domain of Sec5, indicating that paxillin bindsa different part of Sec5 from that involved in binding Ral GTPases(Fig. 4C).

Biosynthetic cargo is delivered to Exocyst-containingpseudopodsMorphological transport assays were performed to determinewhether exocytic cargo is preferentially targeted to Exocyst-enriched sites in pseudopods. For these studies, cells weretransfected with plasmids encoding GFP-tagged forms of either α5-

Journal of Cell Science 121 (17)

integrin (α5-GFP) or a temperature-sensitive mutant of the vesicularstomatitis virus (VSV) G protein (tsG-GFP). The latter protein isa convenient marker of exocytosis because its transport through thesecretory pathway can be manipulated by shifting the temperatureat which cells are cultured. Following transfection, cells wereincubated at 39.5°C for 4 hours to accumulate tsG-GFP in theendoplasmic reticulum, then shifted to 19°C for 2 hours to allowthe protein to accumulate within the trans-Golgi network (TGN).Finally, cells were shifted to 32°C for various lengths of time topermit protein trafficking from the TGN to the plasma membrane.In cells fixed after incubation at 32°C for 30 minutes, much of theGFP-tagged cargo remained in perinuclear compartments (Fig. 5A,arrowheads). In addition, post-Golgi transport intermediatescontaining tsG-GFP or α5-GFP were observed in the cytoplasm atsome distance from the perinuclear vicinity, and these appeared tobe enriched in pseudopods compared with other parts of the cell(Fig. 5A, arrows).

Upon docking and fusion with the plasma membrane, it is nolonger possible to observe accumulation of GFP-labeled transportvesicles in the cytoplasm. It could be argued, therefore, that post-Golgi transport intermediates appear only to be targeted topseudopods because they have farther to travel than do intermediatesthat dock and fuse within the cell body. Therefore, terminal dockingand fusion events were impaired by application of the non-permeable fixative tannic acid to cells in order to resolve more-definitively whether exocytosis was polarized towards pseudopods.Tannic acid crosslinks proteins at the plasma membrane, thereby

Fig. 4. Exocyst is associated with paxillin-containingcomplexes within pseudopods of migrating prostatetumor cells. (A) Fractionation of R3327-5�A cells iniodixanol gradients. 5�A cells were homogenized in aball-bearing cell-cracker. Post-nuclear supernatant wasfractionated by isopycnic centrifugation through five-step iodixanol gradients, as described in the Materialsand Methods. Fractions (0.5 ml) were collected anddensities determined with a refractometer. Presence ofSec8, paxillin, NaK-ATPase α subunit, β-PIX, GIT1and Nck1/2 in gradient fractions was assayed by SDS-PAGE followed by immunoblotting with specificantibodies. Protein levels were quantified using aMolecular Dynamics Phosphorimager. Fractionscorresponding to peak levels of NaK-ATPase, Sec8and paxillin are labeled ‘plasma membrane’, ‘A’ and‘B’, respectively. (B) Co-immunoprecipitation of Sec8with paxillin from isolated 5�A pseudopods. 5�A cellswere cultured on 75 mm Transwell filters (3 μmpores), and induced to extend pseudopods, asdescribed for Fig. 2. Whole cells (wc), isolatedpseudopods (p) or cell bodies (cb) were isolated inCSK buffer after rubbing either the top or bottom offilters with a cotton swab, as appropriate. Extracts(total) and precipitated immune complexes (αPaxillinIP), normalized to total protein content, were assessedby SDS-PAGE followed by immunoblotting withspecific antibodies to paxillin or Sec8. Sec8 co-precipitates with paxillin immune complexes, but onlyfrom pseudopods and not cell bodies. (C) Sec5 bindspaxillin in vitro. Plasmids encoding paxillin and/ormyc-Sec5 or Sec5 lacking its Ral-binding domain(myc-Sec5ΔRBD) were used to prime coupledtranscription and translation reactions in the presenceof [35S]methionine/cysteine. Aliquots of translationproducts were assessed directly (total) or afterimmunoprecipitation with anti-myc antibodies (α-mycip). Bands representing paxillin that co-immunoprecipitated with Myc-tagged Sec5 areindicated (asterisks).

Jour

nal o

f Cel

l Sci

ence

Page 6: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2885Ral targets Exocyst to focal contacts

‘freezing’ the vesicle fusion machinery(Polishchuk et al., 2004). However,cytoplasmic events, including post-Golgivesicle budding and transport to the cellperiphery proceed unimpeded in thepresence of tannic acid for at least 1 hour.Under these conditions, a clearaccumulation of both tsG-GFP and α5-GFP-containing transport intermediateswas observed within pseudopods (Fig.5B). After prolonged incubation at 32°C,vesicles began to accumulate within thecell body, indicating either thatpseudopods had exceeded their capacityfor vesicles or that transport machinerywas failing owing to extended exposureto tannic acid. Nevertheless, these resultsindicate that exocytosis is polarizedtowards Exocyst-enriched tips ofpseudopods in 5�A prostate tumor cells.

Exocyst assembly within pseudopodsis required for exocytosis and invasivecell motilityTo determine whether the Exocyst isrequired for biosynthetic proteintrafficking in metastatic prostate tumorcells, the efficiency of delivery of newlysynthesized α5-integrin to the surface of5�A cells that had normal or reducedlevels of Sec5 or Sec6 was compared.Cells were transfected with siRNAsspecific for Sec5 or Sec6 or a controlsiRNA. Twenty-four hours later, cellswere transfected with plasmid encodingα5-GFP, then returned to the incubator for another 24 hours. Onthe day of the experiment (48 hours after siRNA transfection), oneset of cultures was lysed and analyzed for expression of Sec5, Sec6or a loading control (β-PIX) by western blotting (Fig. 6A), andanother set was used to quantify α5-GFP trafficking (Fig. 6B).Triplicate wells containing either control or Exocyst knock-downcells were subjected to pulse-chase and surface biotinylationanalysis to quantify the amount of newly synthesized α5-GFP thatarrived at the surface during a 1 hour delivery period. This revealedthat the efficiency of delivery of newly synthesized α5-GFP to theplasma membrane of 5�A cells was significantly reduced followingRNAi-mediated reduction of Sec5 or Sec6 expression. The amountof α5-integrin that was synthesized in control and experimentalcultures was similar (data not shown), but the fraction that wasinserted into the plasma membrane of cells with reduced Sec5 orSec6 levels was reduced to ~18-22 % of the levels observed inmock-transfected cells or in cells transfected with control siRNA.Therefore, Exocyst function is required for trafficking of newlysynthesized α5-integrin from the TGN to the plasma membrane ofprostate tumor cells.

Because Exocyst complexes are enriched within pseudopods andinvadopodia, and are required for exocytosis of α5-GFP-ladentransport vesicles, it is reasonable to expect that Exocyst assemblywithin pseudopods and invadopodia is required for invasive cellmotility. Wound healing experiments were performed initially, inorder to determine whether Exocyst assembly is required for

directed motility of 5�A prostate tumor cells. Following RNAi-mediated reduction of either Sec5 or Sec6, cells were significantlyimpaired in their ability to close wounds relative to control cells(Fig. 7A). Twenty-four hours after wounding monolayers, controlmonolayers had completely healed, whereas wounded monolayersof cells with reduced Sec5 or Sec6 levels had closed wounds toonly ~35-40% of the controls. Scattered individual cells wereobserved within the wounds in these cultures, but coordinatedmigration of the population was impaired following Sec5 or Sec6knockdown.

A hallmark of metastatic tumor cells is their ability to degradebasal laminae and subjacent connective tissue, enter the bloodstreamand migrate to distant sites in the body. To determine whetherExocyst function is required for the invasive motility of 5�A prostatetumor cells, the ability of cells to migrate across Matrigel-coatedfilters was assayed (Fig. 7B). Non-metastatic 5�B prostate tumorcells were not able to degrade Matrigel and so remained on theupper surface of filters during invasion assays. By contrast, asignificant number of metastatic 5�A cells were able to degradeMatrigel and migrate across filters during a 24-hour incubation (Fig.7B). In 5�A cells transfected with Sec5 or Sec6 siRNAs, but notwith control siRNA, invasiveness was impaired and cells remainedin the upper chamber, similar to non-metastatic 5�B cells (Fig. 7B).Co-transfection of cells with Sec5 siRNA and a plasmid encodingmyc-tagged Sec5 harboring a silent mutation to render the mRNAresistant to RNAi-mediated silencing restored Sec5 expression (Fig.

Fig. 5. Polarized trafficking of biosynthetic cargo to pseudopods in prostate tumor cells. R3327-5�A cellswere transfected with plasmids encoding GFP-VSVG (tsG-GFP) or GFP α5-integrin (α5-GFP) andmorphological transport assays were performed as described in the Materials and Methods. Followingaccumulation of cargo proteins in the TGN, cultures were shifted to 32°C either in the absence (A) orpresence (B) of 0.5% tannic acid for various lengths of time to facilitate the trafficking of accumulated GFP-fusion proteins from the TGN to the plasma membrane. After 30 minutes at 32°C (A) or indicated times (B),cells were fixed with 2% paraformaldehyde, permeabilized and stained with anti-Sec8 mAb, which wasdetected using a Texas Red-conjugated secondary antibody. Epifluorescence images were obtained asdescribed in Fig. 1. Arrowheads indicate TGN and arrows indicate examples of post-TGN transportintermediates. More post-TGN transport vesicles appear to be delivered to pseudopods than to other parts ofthe cell, even when the TGN is located on the opposite side of the nucleus from the pseudopod. In tannic acidpre-fixed samples (B), pseudopods accumulate transport vesicles, but with prolonged incubation, vesicles alsobegin to accumulate within the cell body.

Jour

nal o

f Cel

l Sci

ence

Page 7: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2886

7C) and rescued ECM invasiveness to levels comparable withcontrol 5�A cells (Fig. 7B).

Ral GTPases regulate Exocyst association with paxillin-containing focal complexes and polarized growth of protrusivecellular extensionsRal GTPases directly bind Sec5 and Exo84 and somehow regulateExocyst function, either by controlling holocomplex assembly orlocal accumulation at sites of membrane remodeling (Camonis andWhite, 2005). Because Sec5 is required for polarized traffickingand invasive motility of metastatic prostate tumor cells, and in vitrobinding studies suggest that Sec5 mediates Exocyst association withpaxillin, we sought to determine whether Ral binding to Sec5 isrequired for invasive motility, and whether this involves modulatingeither Exocyst assembly or association with paxillin. Similar tofindings in other cell types (Takaya et al., 2004), active Ral GTPaseswere found to be enriched within protrusive cellular extensions atthe leading edge of motile prostate tumor cells and, to a lesser extent,in a perinuclear compartment (Fig. 8A). Thus, Ral GTPases arepositioned to regulate Exocyst activities at two sites in these cells.

Ral binding to Sec5 is required for invasive motility of aggressiveprostate tumor cells. Expression of a Sec5 variant harboring a point

Journal of Cell Science 121 (17)

mutation that disrupts binding to Ral (Sec5T11A) (Fukai et al., 2003)failed to rescue matrix invasiveness following siRNA-mediatedreduction of endogenous Sec5 (Fig. 7B,C). Neither Sec5T11A norSec5R27E, a second Ral-uncoupled variant, bound paxillin whenexpressed in 5�A cells, although both proteins were incorporatedinto Exocyst complexes that could be recovered byimmunoprecipitation with anti-Sec8 antibodies (Fig. 8B). It isunlikely that either mutation disrupts paxillin binding directly, asthe Ral-binding domain of Sec5 was dispensable for paxillinbinding in vitro (Fig. 4C). However, incorporation of Ral-uncoupledectopic Sec5 subunits into endogenous Exocyst complexes blockedthe association between paxillin and Exocyst in 5�A prostate tumor

Fig. 7. Ral-coupled Exocyst activity is required for invasive motility ofprostate tumor cells. (A) Wound healing assay. Confluent monolayers ofR3327-5�A cells, transfected with siRNAs specific for Sec5, Sec6 or a controlnon-targeting siRNA, were experimentally wounded by scratching, andanalysis was performed as described in the Materials and Methods. (B) Matrigel invasion assay. Non-metastatic R3327-5�B cells or metastaticR3327-5�A cells transfected with indicated siRNAs and rescue constructs wereseeded on Matrigel-coated Transwell filters. Invasion assays were performedas described in the Materials and Methods. (C) Sec5 expression analysis. 5�Aprostate tumor cells transfected with indicated siRNAs and/or rescueconstructs, were extracted and analyzed by SDS-PAGE and immunoblottingwith antibodies to Sec5 (endogenous and ectopic proteins detected) or c-myc(ectopic Sec5 detected).

Fig. 6. Exocyst is required for exocytosis of newly synthesized α5-integrin inprostate tumor cells. (A) RNAi-mediated reduction of Sec5 and Sec6expression. R3327-5�A were transfected with either nothing (‘mock’) or withsiRNAs targeting Sec5, Sec6 or a control non-targeting siRNA (‘control’), asdescribed in the Materials and Methods. Sixty hours post-transfection, cellswere lysed and lysates were analyzed by SDS-PAGE and immunoblotting forSec5, Sec6 and β-PIX. Protein levels were quantified using a MolecularDynamics Typhoon phosphorimager. (B) Metabolic pulse-chase and surfacebiotinylation analysis of α5 integrin trafficking in prostate tumor cells.Delivery of newly synthesized α5-GFP to the surface of R3327-5�A cells wasassessed as described in the Materials and Methods. Experiments wereperformed twice, each time with triplicate wells of cells. Relative surfacedelivery was defined as the mean signal obtained from three replicatebiotinylated α5-GFP bands, normalized to the mean of the total α5-GFPrecovered in the initial immunoprecipitates.

Jour

nal o

f Cel

l Sci

ence

Page 8: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2887Ral targets Exocyst to focal contacts

cells, as shown by the failure to co-immunoprecipitate paxillin withSec8 in cells expressing either Sec5T11A or Sec5R27E (Fig. 8B).

To gain further insight into the mechanism by which Ral GTPasesregulate Exocyst-dependent activities in metastatic prostate tumorcells, and to identify which Ral GTPase (RalA or RalB) is requiredfor this regulation, loss-of-function studies were performed. Lentiviralexpression of specific shRNAs efficiently reduced expression of eitherRalA or RalB in 5�A cells (Fig. 8C). Surprisingly, RNAi-mediatedknockdown of either GTPase inhibited association of Exocystcomplexes with paxillin (Fig. 8C). Examination of cells revealed thatreduction of either RalA or RalB impaired the ability of cells to extend

long, slender pseudopods associated with invasive motility (Fig. 9A).Cells with reduced expression of either GTPase were observed togrow as cohesive colonies, and only rarely were cells witnessed todetach from and migrate away from these colonies. These resultsindicate that both RalA and RalB contribute to altered cellularmorphogenesis and motility in prostatic tumor cells, and that targetingof Exocyst complexes to paxillin-containing focal complexes requiresinputs from both GTPases.

RNAi-mediated knockdown of either GTPase did notsignificantly reduce the amount of assembled Exocyst in 5�Aprostate tumor cells, as determined by co-immunoprecipation ofthree subunits (Sec5, Sec15 and Exo84) with Sec8 (supplementarymaterial Fig. S1). However, reduction of Ral expression altered thesubcellular distribution of Exocyst complexes in distinct ways. Incells with reduced expression of RalA, Sec6 and Sec8 wereprimarily present within a perinuclear compartment, whereas in cellswith reduced expression of RalB these proteins accumulated in largecytoplasmic vesicles (Fig. 9B). Therefore, Ral GTPases appear toregulate subcellular localization, rather than assembly of Exocystcomplexes in 5�A prostate tumor cells.

Fig. 8. Ral GTPases are required for Exocyst association with paxillin in cells.(A) Localization of active Ral following wounding of prostatic tumor cells.Cells were transfected with X-Press-tagged Exo84 Ral-binding domain andmonolayers were wounded by scratching. Active Ral GTPase localization wasdetermined by immunofluorescence staining with anti-X-Press antibodies atindicated time points. (B) Co-immunoprecipitation of Exocyst and paxillin isdependent on Ral-binding capability of Sec5. Cells were transfected withplasmids encoding myc-tagged Sec5 [wild type (wt) or Ral-uncoupled mutants(T11A or R27E)]. Cells were extracted in 1% Triton X-100 and extracts weresubjected to immunoprecipitation with antibodies to Sec8 or paxillin. Presenceectopic myc-Sec5 and paxillin in equivalent amounts of whole cell extracts(‘lysate’) and precipitated immune complexes was assessed by SDS-PAGEfollowed by immunoblotting with specific antibodies. (C) RNAi-mediatedreduction of RalA and RalB expression. R3327-5�A were infected withrecombinant lentiviruses coding shRNAs specific for RalA or RalB. Stableclones of cells were selected in puromycin and assessed for RalA and RalBexpression by immunoblotting with specific antibodies. (D) R3327-5�B,R3327-5�A or R3327-5�A cells expressing shRNAs targeting RalA or RalBwere extracted in 1% Triton X-100. Extracts were subjected toimmunoprecipitation with antibody to paxillin. Presence Sec5 and paxillin inequivalent amounts of whole cell extracts (‘lysate’) and precipitated immunecomplexes (‘α-paxillin IP’) was assessed by SDS-PAGE followed byimmunoblotting with specific antibodies.

Fig. 9. Ral GTPases are required for localization of Exocyst to protrusive cellextensions. (A) Morphology of R3327-5�A or R3327-5�A cells expressingshRNAs targeting RalA or RalB. RNAi-mediated reduction of either RalA orRalB suppressed polarized growth of protrusive cell extensions, and cellstended to grow as more tightly compacted colonies than control R3327-5�Aprostate tumor cells. (B) Localization of Sec6 and Sec8 in prostate tumor cellsis altered following reduction of RalA or RalB expression. R3327-5�A cells orR3327-5�A cells expressing shRNAs targeting RalA or RalB were fixed andlabeled with antibodies against Sec6 or Sec8. Exocyst proteins areconcentrated in perinuclear compartments, similar to those observed inparental 5�A cells, when RalA expression is reduced. By contrast, theseExocyst subunits accumulate in large cytoplasmic vesicles in cells when RalBexpression is reduced.

Jour

nal o

f Cel

l Sci

ence

Page 9: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2888

DiscussionMechanisms that regulate assembly and recruitment of the Exocystto sites of active membrane remodeling are complex and poorlyunderstood. Recent work in both budding yeast and mammaliancells has highlighted an essential role for protein-lipid interactionsinvolving Sec3 and Exo70 subunits with phosphatidylinositol 4,5-bisphosphate (He et al., 2007; Liu et al., 2007; Zhang et al., 2008).Although such interactions appear to be essential for overallmembrane binding, they are not sufficient to account for the highlylocalized enrichment of Exocyst complexes often observed inpolarized cells. For example, in renal epithelial cells, the Exocystis concentrated within a relatively narrow region of the lateralplasma membrane, just below the tight junction. Exocystaccumulation at this site requires components of both Ca2+-dependent (E-cadherin) and Ca2+-independent (nectin) cell-adhesionsystems (Yeaman et al., 2004). The importance of E-cadherin-mediated cell–cell adhesion in regulating membrane recruitmentand assembly of Exocyst on lateral plasma membranes is supportedby the current study, because ectopic expression of E-cadherin inmetastatic prostate tumor cells was sufficient to restore assemblyof intercellular junctions and return Exocyst complexes to thesesites. These molecular events are associated with suppression ofinvasive motility in vitro and metastasis in vivo.

Although mechanisms that sort Exocyst complexes into cadherin-based intercellular junctions are incompletely understood, it is likelythat interactions with multidomain scaffolding proteins, such asSAP97 and CASK, which assemble on lateral plasma membranesof epithelial cells, participate in this process. Interactions betweenPDZ domains on these or related proteins and a C-terminal PDZtarget on Sec8 have been shown in neurons (Sans et al., 2003),adipocytes (Inoue et al., 2006) and oligodendrocytes (Anitei et al.,2006), but their significance in sorting Exocyst components inepithelial cells has not been established.

In cells lacking cadherin-mediated adhesion, distinct spatialcues probably mediate recruitment and assembly of Exocystcomplexes at sites of membrane remodeling. In the metastaticprostate cancer cells examined here, Exocyst components wereenriched within protrusive cellular extensions at the leading edgeof motile cells, and were occasionally observed in invadopodia.Paxillin probably represents one important spatial determinant forExocyst localization in these cells. Exocyst componentscolocalized, co-fractionated and co-immunoprecipitated withpaxillin from leading edge complexes. Although molecularspecifics of this interaction not yet known, it is noteworthy thatonly the Sec5 subunit was observed to interact with paxillin invitro. This in vitro interaction did not require Ral GTPases nordid it involve the Ral-binding domain of Sec5. However, Ralbinding to Sec5 was necessary to facilitate Exocyst binding topaxillin in cells. This conclusion is based on findings thatoverexpression of Ral-uncoupled Sec5 mutants inhibited Exocystinteraction with paxillin in 5�A cells, as did RNAi-mediatedreduction of either RalA or RalB. Importantly, neither reductionof RalA nor RalB significantly altered the steady state level ofassembled Exocyst complex in these cells, but the localization ofExocyst proteins was somewhat different when either GTPase wasabsent. In the absence of RalA, Sec6 and Sec8 accumulated in aperinuclear compartment, whereas depletion of RalB resulted inaccumulations of Sec6 and Sec8 in large vesiculated structures inthe cytoplasm. Therefore, Ral GTPases appear to function inspatial regulation of Exocyst function, rather than Exocystassembly per se, during tumor cell motility. Moreover, RalA and

Journal of Cell Science 121 (17)

RalB may regulate different aspects of Exocyst function in theperinuclear compartment and plasma membrane.

A previous study showed that Ral was selectively activated atthe leading edge of motile epithelial cells (Takaya et al., 2004). Wealso observed activation of Ral at the leading edge of motile prostatetumor cells. Therefore, it seems plausible that Ral facilitates Sec5-paxillin interactions specifically at the leading edge of migratingcells and that this contributes to Exocyst enrichment and polarizedexocytosis at these sites. Interestingly, in non-invasive 5�B cells,which express both E-cadherin and paxillin, Exocyst is found to beassociated with lateral membrane cell–cell contacts and is notassociated with paxillin. This implies that a hierarchy of spatial cuesand protein–protein interactions controls the spatial distribution ofExocyst complexes within these cells.

Several reports have highlighted distinct functions for RalA andRalB in tumorigenic transformation (Bodemann and White, 2008;Camonis and White, 2005). There appear to be cell type-specificactivities associated with each GTPase, even within the relativelynarrow biological context of cell migration. Loss-of-functionanalyses have shown that RalB, but not RalA, is limiting formigration of normal rat kidney cells in wound healing assays (Rosseet al., 2006), as well as human bladder and prostate cancer cells intranswell migration assays (Oxford et al., 2005). Interestingly, inboth of these studies, reduction of RalA expression reversed theinhibitory effect of RalB knockdown, suggesting that RalA and RalBmight serve antagonistic functions in cell migration. In contrast tothese reports, other studies have highlighted a role for RalA inchemotactic migration of C2C12 skeletal myoblasts (Suzuki et al.,2000), in human bladder cancer cell lines (Gildea et al., 2002) andin signaling to the Exocyst during neurite branching (Lalli and Hall,2005). The data presented here implicate both RalA and RalB insignaling Exocyst recruitment to paxillin-containing complexesduring polarized growth of protrusive cellular extensions. Thus, ourdata are consistent with previous studies in human pancreaticcarcinoma cells, which showed that both RalA and RalB are requiredfor invasive motility (Lim et al., 2006).

How might Ral activation and Exocyst recruitment to pseudopodtip complexes contribute to cell polarization during invasivemotility? First, the vesicle tethering function of the Exocyst is likelyto be required for delivery of specific components as well as bulkmembrane from the trans-Golgi network (TGN), recyclingendosomes or other internal stores to the growing protrusive cellextension. Results presented here show that Exocyst function isrequired for efficient exocytosis of newly synthesized α5-integrin.This is consistent with results of TIRF microscopy, which revealedthat fusion of vesicles carrying a newly synthesized LDL receptor,which is an Exocyst-dependent cargo molecule (Grindstaff et al.,1998), occurs predominantly at the leading edge of migrating NRKcells (Schmoranzer et al., 2003). In addition, directed motility ofSwiss 3T3 fibroblasts was impaired by expression of dominant-negative protein kinase D mutant (Prigozhina and Waterman-Storer,2004), which has been shown to impair post-TGN membranetrafficking and Exocyst recruitment to the plasma membrane(Liljedahl et al., 2001; Yeaman et al., 2001).

It is also likely that other Exocyst-specific functions, distinct froma principle role in vesicle tethering, contribute to its activity inmediating invasive motility of tumor cells. One such function is tocoordinate cytoskeleton remodeling with vesicle trafficking. In oneof the earliest papers describing a connection between Ral and theExocyst, it was suggested that Sec5 mediates Ral-induced filopodiaformation via a mechanism that is distinct from Exocyst-dependent

Jour

nal o

f Cel

l Sci

ence

Page 10: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2889Ral targets Exocyst to focal contacts

membrane trafficking processes (Sugihara et al., 2002).Overexpression of Exo70 has been reported to promote filopodiaformation in NRK cells (Wang et al., 2004), and a similar phenotypehas been seen in R3327-5�A cells (data not shown). In addition,Exo70 was recently shown to interact with the Arp2/3 complex andto regulate its activity within nascent lamellipodia of migrating NRKcells (Zuo et al., 2006). Given that the overall dimensions of a fullyassembled octameric Exocyst holocomplex exceeds 13�30 nm (Hsuet al., 1998), this provides a large interface to accommodatenumerous protein-protein interactions that might serve to integrateExocyst-dependent vesicle tethering with other processes involvedin cellular morphogenesis. Considering the pace with which novelExocyst-interacting partners are being identified, this seems likelyto be the case.

Materials and MethodsReagentsMouse monoclonal antibodies against Sec6 (9H5 and 10C3) and Sec8 (2E9, 2E12,5C3, 8F12 and 10C2) have been described previously (Hsu et al., 1996; Kee et al.,1997). Mouse monoclonal antibodies against Sec5 and Exo84 were generouslyprovided by Dr Richard Scheller (Genentech). Rabbit polyclonal antibodies againstSec3, Sec5, Sec10, Sec15, Exo70 and Exo84 were generated by Covance and havebeen described previously (Yeaman, 2003). Rabbit polyclonal antibodies against theconserved cytoplasmic domain of mouse E-cadherin (E2), α-catenin and the α subunitof the Na+–K+ ATPase (NaKα) have been described previously (Marrs et al., 1993).Rabbit anti-Munc18c was generously provided by Dr David James (Garvan Institute,Australia). Rabbit polyclonal antibodies to RalB (BD Transduction Labs), paxillin,GIT1, Nck1/2 (Santa Cruz Biotechnology) and syntaxin3 (AbCam), and mouse mAbsto β-tubulin (Sigma), β-PIX, Munc18a/b, RalA and syntaxin4 (BD Transduction Labs)were obtained from commercial sources. FITC-goat anti-mouse, Texas Red-donkeyanti-mouse and Texas Red-donkey anti-rabbit IgG were purchased from JacksonImmunoResearch Labs (West Grove, PA). Mouse anti-c-Myc agarose affinity gelwas from Sigma (St Louis, MO).

Plasmid-encoding temperature-sensitive mutant viral glycoprotein VSVG-tsO45fused to GFP (ts-G-GFP) was generously provided by Drs Suzie Scales (Genentech)(Scales et al., 1997). Plasmids encoding α5-integrin or paxillin fused to GFP (α5-GFP, paxillin-GFP) were kindly provided by Dr Alan F. Horwitz (University ofVirginia) (Laukaitis et al., 2001). Myc-tagged rSec5 was expressed from the pCMV-myc vector (Clontech). Ral-uncoupled mutants (rSec5T11A and rSec5R27E) weregenerated by site-directed mutagenesis with the Quick-Change kit (Strategene) andexpressed from the pCMV-myc vector (Clontech). X-Press-tagged rExo84 Ral-bindingdomain was expressed from the pcDNA3.1/HisC vector (Invitrogen). For in vitrotranscription–translation studies, the coding sequence of paxillin was subcloned intothe pcDNA3.1 vector, which contains a T7 RNA polymerase promoter. Myc-taggedrSec5 was expressed from the pGBKT7 plasmid (Clontech). A truncated rSec5construct, deleted of its N-terminal 120 amino acid Ral-binding domain (Myc-rSec5ΔRBD), was expressed from the pcDNA3.1/HisC vector.

Synthetic siRNAs targeting Sec5 or Sec6 and standard negative control non-targeting siRNAs were ordered from Dharmacon. Sec5 and Sec6 targets were designedusing the following sense sequences previously shown to be effective for rat Exocystproteins (Rosse et al., 2006): CGGCAGAAUGGAUGUCUGC (Sec5-1),GGUCGGAAAGACAAGGCAGAU (Sec5-2) and CUGGAGGCAGAGC AU -CAACAC (Sec6). Plasmids coding shRNAs specific for RalA (CGCUG -CAAUUAGAGACAACUA; clone TRCN0000004865) or RalB (GAGUU -UGUAGAAGACUAUGAA; clone TRCN0000072957) in the pLKO.1 vector werepurchased from Open Biosystems (Huntsville, AL).

Cell culture methodsDunning rat prostate tumor cell lines (R3327-5�A and R3327-5�B) were maintainedin high glucose Dulbecco’s modified Eagle’s media (DMEM, Life Technologies)supplemented with 10% fetal bovine serum (FBS), penicillin, streptomycin andkanamycin. Plasmids were transfected using Lipofectamine2000 reagent (LifeTechnologies) according to the manufacturer’s instructions. siRNAs were transfectedat 160 nM using Oligofectamine (Life Technologies) according to the manufacturer’sinstructions. Lentivirus production in 293FT packaging cells followed establishedprotocols (The RNAi Consortium). Stable knockdown of RalA and RalB was achievedby lentiviral infection of R3327-5�A cells and selection in 5 μg/ml puromycin.

Immunofluorescent stainingCells were fixed in 4% paraformaldehyde for 30 minutes, then permeabilized byincubation at 0°C for 10 minutes with 1% Triton X-100 in buffer containing 10 mMPIPES, pH 6.8, 50 mM NaCl, 300 mM sucrose, 3 mM MgCl2 and protease inhibitors(1 mM pefabloc, and 10 μg/ml each of aprotinin, antipain, leupeptin, pepstatin A)(CSK buffer). Antibodies were diluted in blocking buffer [Ringer’s saline (154 mM

NaCl, 1.8 mM Ca2+, 7.2 mM KCl, 10 mM HEPES, pH 7.4)] containing 0.2% BSA,0.5% normal goat serum and 0.5% normal donkey serum) and applied to cells for 2hours at 4°C. After five washes in blocking buffer, FITC and Texas Red-conjugatedsecondary antibodies, diluted 1:200, were applied for 1 hour at 4°C. Coverslips andfilters were washed five times and mounted in VectaShield (Vector Laboratories,Burlingame, CA). Samples were viewed with either a Nikon Microphot-FXmicroscope (63� or 100� objectives) or a Zeiss 510 confocal laser scanningmicroscope (63� objective) using krypton/argon laser with 488 nm (FITC, GFP)and 543 nm (Texas Red) laser lines, as noted in the figure legends. Digital imagesof data collected from the Nikon Microphot-FX microscope were obtained with aKodak DCS 760 digital camera.

Morphological assay for polarized trafficking of α5-GFP and ts-G-GFPR3327-5�A cells were transfected with plasmids encoding either α5-GFP or ts-G-GFP. To accumulate ts-G-GFP in the TGN, cells were suspended with trypsin-EDTA24 hours after transfection, re-plated on collagen-coated coverslips and incubated at40°C for 5 hours to accumulate ts-G-GFP in the endoplasmic reticulum. Cultureswere shifted to 32°C for 7.5 minutes to facilitate protein folding, and subsequentlytransferred to 19°C for 2 hours to accumulate protein in the TGN. To accumulateα5-GFP in the TGN, cells were suspended with trypsin-EDTA 24 hours aftertransfection, re-plated on collagen-coated coverslips and incubated at 37°C for 1 hourthen transferred to 19°C for 4 hours. Trafficking of protein from the TGN to plasmamembrane was initiated by shifting cultures from 19°C to 32°C. In some cases, cultureswere treated with 0.5 % (w/v) tannic acid prior to and during the final 32°C incubationperiod to inhibit vesicle fusion with the plasma membrane (Polishchuk et al., 2004).At indicated time points, cultures were fixed and labeled with antibodies to Sec8followed by Texas Red anti-mouse secondary antibody.

Gel electrophoresis and immunoblottingProtein samples were incubated in SDS-PAGE sample buffer for 10 minutes at 65°Cbefore separation in 7.5%, 10% or 12.5% SDS polyacrylamide gels. Proteins wereelectrophoretically transferred from gels to Immobilon PVDF membrane (Millipore,Bedford, MA). Blots were blocked in Blotto [5% nonfat dry milk, 0.1% sodium azidein 150 mM NaCl, 10 mM TrisHCl, pH 7.5 (TBS)] overnight at 4°C. Primary antibodieswere incubated with blots at room temperature for 1 hour. After five washes, 10minutes each, in TBS containing 0.1% Tween-20, the blots were incubated with 125I-labeled goat anti-mouse or goat anti-rabbit secondary antibody (Amersham) for 1hour at room temperature. Blots were washed as above and exposed to phosphorimagerscreens. The amount of labeled antibody bound to the blots was determined directlyusing a Phosphorimager (Typhoon, Molecular Dynamics, Sunnyvale, CA) andImageQuant software (version 1.2, Molecular Dynamics, Sunnyvale, CA).

Cell fractionation in iodixanol gradientsCells were homogenized in isotonic sucrose buffer [0.25 M sucrose in 20 mM HEPES-KOH, pH 7.2, 90 mM KOAc, 2 mM Mg(OAc)2, and protease inhibitors] by repeatedpassage through a ball bearing homogenizer (Varian Physics, Stanford University).Separation of different membrane compartments was achieved by centrifugation infive-step 10-15-20-25-30% (wt/vol) iodixanol gradients. Briefly, post-nuclearsupernatant was mixed with Opti-Prep [60% (wt/vol) iodixanol, Nycomed, Oslo,Norway] and homogenization buffer to generate a solution containing 30% iodixanol.This was overlaid in centrifuge tubes with equal volumes of 25%, 20%, 15% and10% iodixanol, and samples were centrifuged at 350,000 g for 3 hours at 4°C in aBeckman NVT65 rotor. Fractions (0.5 ml) were collected, refractive indices wereread, and proteins were separated by SDS-PAGE. Proteins were transferred from gelsto Immobilon P membranes for immunoblotting as described above.

Isolation of pseudopodsPseudopod isolation was performed similar to methods described previously (Choand Klemke, 2002). R3327-5�A cells were incubated for 24 hours in serum-freeDMEM containing 0.5 % (w/v) BSA and antibiotics. Cells were suspended and seededat a density of 1.5�107 cells/filter on Transwell filters (# 3420, 75 mm diameter, 3.0μm pore size). Following attachment at 37°C for 2 hours in serum-free medium,complete medium containing 10% FBS was applied to the basal chamber to stimulatechemotaxis. Cultures were returned to 37°C and incubated for 4 hours to allowpseudopod growth. Cell bodies were manually removed from the tops of filters bygentle scrubbing with a cotton swab, leaving the detached pseudopods adherent tothe bottoms of filters. For analysis of total cellular protein, cell bodies were notremoved. Last, intact cells or isolated pseudopods were solubilized in extraction buffer(CSK with protease inhibitors) and protein content was quantified using a micro BCAassay (Pierce).

ImmunoprecipitationWhole cells, pseudopods or cell bodies were extracted in CSK buffer for 30 minutesat 4°C. Extracts were precleared with 5 μl of nonimmune serum and 50 μlStaphylococcus aureus cells (Pansorbin; Calbiochem Novabiochem, La Jolla, CA)for 1 hour at 4°C. For Sec8 immunoprecipitation, mAbs 2E12, 5C3 and 10C2 werecovalently crosslinked to Protein A Sepharose beads (Pharmacia LKB Nuclear,

Jour

nal o

f Cel

l Sci

ence

Page 11: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2890 Journal of Cell Science 121 (17)

Gaithersburg, MD) with dimethyl pimelimidate (DMP), and 20 μl of immunoadsorbantwas used per immunoprecipitation. Immunoprecipitation of paxillin was performedwith a specific rabbit polyclonal antibody (5 μg per sample) pre-bound to Protein ASepharose beads. Immunoadsorbants were incubated with pre-cleared cell extractsfor 2 hours at 4°C, then washed under stringent conditions and prepared for SDS-PAGE as described previously (Yeaman et al., 2004).

In vitro transcription/translation analysisPlasmids encoding individual myc-tagged Exocyst subunits (in pGBKT7 vectors)were added singly or together with plasmid encoding paxillin (in pcDNA 3.1) to TNTQuick Coupled Transcription/Translation Systems (Promega). Reactions wereincubated in the presence of [35S]methionine/cysteine (Perkin-Elmer), according tomanufacturer’s instructions. Aliquots of total translated product or anti-mycimmunoprecipitated material were analyzed by SDS-PAGE and autoradiography.

Metabolic pulse-chase and surface biotinylation analysisR3327-5�A cells were transfected with siRNAs specific for Sec5 or Sec6 or a controlsiRNA. Twenty-four hours later, cells were transfected with plasmid encoding α5-GFP and returned to the incubator. The following morning, cells were suspendedwith trypsin and split into four identical replicate cultures and returned to the incubatorfor 12-18 hours. On the day of the experiment (~60 hours after siRNA transfection),one set of cultures was lysed and analyzed for expression of Sec5, Sec6 or a loadingcontrol (β-PIX) by western blotting, and the remaining cultures were used to quantifyα5-GFP trafficking. Triplicate wells containing either control or Exocyst knock-downcells were incubated in methionine/cysteine-free culture medium to starve them for30 minutes, then pulse-labeled for 30 minutes in the same medium supplementedwith 1 mCi/ml [35S]methionine/cysteine. Following the labeling period, cultures werewashed three times in chase medium (complete DMEM supplemented with fivefoldexcess methionine and cysteine), and returned to the incubator for 1 hour in the samemedium. At the end of the chase incubation, cultures were placed on ice, washedwith ice-cold Ringer’s saline and labeled with Sulfo-NHS-SS-Biotin (Pierce).Following extensive washes in quenching buffer (TBS containing 50 mM NH4Cland 0.5% BSA), cells were lysed and pre-cleared as described above, and α5-GFPwas immunoprecipitated with anti-GFP antibodies. The biotinylated (e.g. surface-exposed) cohort of α5-GFP was subsequently recovered from total immunoprecipitatesby avidin precipitation. Samples were analyzed by SDS-PAGE and bands werequantified with a phosphorimager following fluorographic enhancement of gels withAmplify solution (Amersham). Relative surface delivery of α5-GFP was defined asthe mean signal obtained from three replicate biotinylated α5-GFP bands, normalizedto the mean of the total α5-GFP recovered in initial immunoprecipitates.

Wound-healing assaysR3327-5�A cells were transfected with siRNAs specific for Sec5, Sec6 or a controlsiRNA using Oligofectamine. Seventy-two hours post-transfection, confluent cellmonolayers were experimentally wounded by scratching with a pipette tip, rinsedwith Ringer’s saline and fed with fresh medium containing 10% serum. Analysis ofwounds was performed by photographing several areas of each wound at multipletime points over a 48 hour period using a Nikon TE300 microscope equipped witha Nikon Coopix 5000 camera. Areas of wound closure were quantified using Image-Pro Plus software (Media Cybernetics, Silver Spring, MD).

Matrigel invasion assay5�B or 5�A cells were transfected without or with Sec5, Sec6 or control siRNAs andcultured for 60 hours. Cells were suspended in serum-free DMEM containing 0.5%(w/v) BSA and antibiotics and seeded at a density of 5�104 cells/filter on Transwellfilters (# 3422, 6.5 mm diameter, 8.0 μm pore size) coated with 20 μg of Matrigel.Following attachment of cells at 37°C for 2 hours in serum-free medium, completemedium containing 10% FBS was applied to the basal chamber to stimulatemigration. Cultures were returned to 37°C and incubated for 24 hours to allow invasionto occur. At the end of the invasion period, cells remaining on the upper surface offilters were manually removed by gentle scrubbing with a cotton swab, and cells thathad invaded through the Matrigel-coated filters were fixed with 100% methanol,stained with DAPI and counted manually. To control for variability in plating efficiencybetween samples, replicate filters were treated identically to those described above,except that cells remaining on the upper surface were not removed at the end of theinvasion period. Cells were fixed with methanol, labeled with DAPI and the meancell density of each culture was calculated by manually counting cells in multiplerandom fields of each filter.

This work was supported by grants from the National Institutes ofHealth (GM067002) and the US Department of Defense (DAMD 17-03-1-0187). Brian Leper, Melinda Schwarz and Hsiang Wen aregratefully acknowledged for their technical assistance in generating andcharacterizing reagents, and performing these studies.

ReferencesAnitei, M., Ifrim, M., Ewart, M. A., Cowan, A. E., Carson, J. H., Bansal, R. and Pfeiffer,

S. E. (2006). A role for Sec8 in oligodendrocyte morphological differentiation. J. CellSci. 119, 807-818.

Bodemann, B. O. and White, M. A. (2008). Ral GTPases and cancer: linchpin supportof the tumorigenic platform. Nat. Rev. Cancer 8, 133-140.

Boyd, C., Hughes, T., Pypaert, M. and Novick, P. (2004). Vesicles carry most exocystsubunits to exocytic sites marked by the remaining two subunits, Sec3p and Exo70p. J.Cell Biol. 167, 889-901.

Bussemakers, M. J., van Moorselaar, R. J., Giroldi, L. A., Ichikawa, T., Isaacs, J. T.,Takeichi, M., Debruyne, F. M. and Schalken, J. A. (1992). Decreased expression ofE-cadherin in the progression of rat prostatic cancer. Cancer Res. 52, 2916-2922.

Camonis, J. H. and White, M. A. (2005). Ral GTPases: corrupting the exocyst in cancercells. Trends Cell Biol. 15, 327-332.

Chien, Y., Kim, S., Bumeister, R., Loo, Y. M., Kwon, S. W., Johnson, C. L., Balakireva,M. G., Romeo, Y., Kopelovich, L., Gale, M., Jr et al. (2006). RalB GTPase-mediatedactivation of the IkappaB family kinase TBK1 couples innate immune signaling to tumorcell survival. Cell 127, 157-170.

Cho, S. Y. and Klemke, R. L. (2002). Purification of pseudopodia from polarized cellsreveals redistribution and activation of Rac through assembly of a CAS/Crk scaffold.J. Cell Biol. 156, 725-736.

Feig, L. A. (2003). Ral-GTPases: approaching their 15 minutes of fame. Trends Cell Biol.13, 419-425.

Finger, F. P. and Novick, P. (1998). Spatial regulation of exocytosis: lessons from yeast.J. Cell Biol. 142, 609-612.

Fukai, S., Matern, H. T., Jagath, J. R., Scheller, R. H. and Brunger, A. T. (2003).Structural basis of the interaction between RalA and Sec5, a subunit of the sec6/8 complex.EMBO J. 22, 3267-3278.

Gildea, J. J., Harding, M. A., Seraj, M. J., Gulding, K. M. and Theodorescu, D. (2002).The role of Ral A in epidermal growth factor receptor-regulated cell motility. CancerRes. 62, 982-985.

Grindstaff, K. K., Yeaman, C., Anandasabapathy, N., Hsu, S. C., Rodriguez-Boulan,E., Scheller, R. H. and Nelson, W. J. (1998). Sec6/8 complex is recruited to cell-cellcontacts and specifies transport vesicle delivery to the basal-lateral membrane in epithelialcells. Cell 93, 731-740.

Guo, W., Roth, D., Walch-Solimena, C. and Novick, P. (1999). The exocyst is an effectorfor Sec4p, targeting secretory vesicles to sites of exocytosis. EMBO J. 18, 1071-1080.

Hashimoto, S., Onodera, Y., Hashimoto, A., Tanaka, M., Hamaguchi, M., Yamada, A.and Sabe, H. (2004). Requirement for Arf6 in breast cancer invasive activities. Proc.Natl. Acad. Sci. USA 101, 6647-6652.

He, B., Xi, F., Zhang, X., Zhang, J. and Guo, W. (2007). Exo70 interacts withphospholipids and mediates the targeting of the exocyst to the plasma membrane. EMBOJ. 26, 4053-4065.

Hofer, F., Fields, S., Schneider, C. and Martin, G. S. (1994). Activated Ras interactswith the Ral guanine nucleotide dissociation stimulator. Proc. Natl. Acad. Sci. USA 91,11089-11093.

Hsu, S. C., Ting, A. E., Hazuka, C. D., Davanger, S., Kenny, J. W., Kee, Y. and Scheller,R. H. (1996). The mammalian brain rsec6/8 complex. Neuron 17, 1209-1219.

Hsu, S. C., Hazuka, C. D., Roth, R., Foletti, D. L., Heuser, J. and Scheller, R. H. (1998).Subunit composition, protein interactions, and structures of the mammalian brain sec6/8complex and septin filaments. Neuron 20, 1111-1122.

Inoue, M., Chiang, S. H., Chang, L., Chen, X. W. and Saltiel, A. R. (2006).Compartmentalization of the exocyst complex in lipid rafts controls Glut4 vesicletethering. Mol. Biol. Cell 17, 2303-2311.

Jin, R., Junutula, J. R., Matern, H. T., Ervin, K. E., Scheller, R. H. and Brunger, A.T. (2005). Exo84 and Sec5 are competitive regulatory Sec6/8 effectors to the RalAGTPase. EMBO J. 24, 2064-2074.

Kee, Y., Yoo, J. S., Hazuka, C. D., Peterson, K. E., Hsu, S. C. and Scheller, R. H.(1997). Subunit structure of the mammalian exocyst complex. Proc. Natl. Acad. Sci.USA 94, 14438-14443.

Lalli, G. and Hall, A. (2005). Ral GTPases regulate neurite branching through GAP-43and the exocyst complex. J. Cell Biol. 171, 857-869.

Laukaitis, C. M., Webb, D. J., Donais, K. and Horwitz, A. F. (2001). Differentialdynamics of alpha 5 integrin, paxillin, and alpha-actinin during formation and disassemblyof adhesions in migrating cells. J. Cell Biol. 153, 1427-1440.

Liljedahl, M., Maeda, Y., Colanzi, A., Ayala, I., Van Lint, J. and Malhotra, V. (2001).Protein kinase D regulates the fission of cell surface destined transport carriers from thetrans-Golgi network. Cell 104, 409-420.

Lim, K. H., Baines, A. T., Fiordalisi, J. J., Shipitsin, M., Feig, L. A., Cox, A. D., Der,C. J. and Counter, C. M. (2005). Activation of RalA is critical for Ras-inducedtumorigenesis of human cells. Cancer Cell 7, 533-545.

Lim, K. H., O’Hayer, K., Adam, S. J., Kendall, S. D., Campbell, P. M., Der, C. J. andCounter, C. M. (2006). Divergent roles for RalA and RalB in malignant growth ofhuman pancreatic carcinoma cells. Curr. Biol. 16, 2385-2394.

Liu, J., Zuo, X., Yue, P. and Guo, W. (2007). Phosphatidylinositol 4,5-bisphosphatemediates the targeting of the exocyst to the plasma membrane for exocytosis inmammalian cells. Mol. Biol. Cell 18, 4483-4492.

Luo, J., Sharma, N., Seftor, E. A., De Larco, J., Heidger, P. M., Hendrix, M. J. andLubaroff, D. M. (1997). Heterogeneous Expression of Invasive and Metastatic Propertiesin a Prostate Tumor Model. Pathol. Oncol. Res. 3, 264-271.

Luo, J., Lubaroff, D. M. and Hendrix, M. J. (1999). Suppression of prostate cancerinvasive potential and matrix metalloproteinase activity by E-cadherin transfection.Cancer Res. 59, 3552-3556.

Jour

nal o

f Cel

l Sci

ence

Page 12: Ral-regulated interaction between Sec5 and paxillin targets ......association between Sec5 and paxillin. Overexpression of Ral-uncoupled Sec5 mutants inhibited Exocyst interaction

2891Ral targets Exocyst to focal contacts

Marrs, J. A., Napolitano, E. W., Murphy-Erdosh, C., Mays, R. W., Reichardt, L. F.and Nelson, W. J. (1993). Distinguishing roles of the membrane-cytoskeleton andcadherin mediated cell-cell adhesion in generating different Na+,K(+)-ATPasedistributions in polarized epithelia. J. Cell Biol. 123, 149-164.

Moskalenko, S., Henry, D. O., Rosse, C., Mirey, G., Camonis, J. H. and White, M. A.(2002). The exocyst is a Ral effector complex. Nat. Cell Biol. 4, 66-72.

Moskalenko, S., Tong, C., Rosse, C., Mirey, G., Formstecher, E., Daviet, L., Camonis,J. and White, M. A. (2003). Ral GTPases regulate exocyst assembly through dual subunitinteractions. J. Biol. Chem. 278, 51743-51748.

Novick, P. and Guo, W. (2002). Ras family therapy: Rab, Rho and Ral talk to the exocyst.Trends Cell Biol. 12, 247-249.

Oxford, G., Owens, C. R., Titus, B. J., Foreman, T. L., Herlevsen, M. C., Smith, S. C.and Theodorescu, D. (2005). RalA and RalB: antagonistic relatives in cancer cellmigration. Cancer Res. 65, 7111-7120.

Polishchuk, R., Di Pentima, A. and Lippincott-Schwartz, J. (2004). Delivery of raft-associated, GPI-anchored proteins to the apical surface of polarized MDCK cells by atranscytotic pathway. Nat. Cell Biol. 6, 297-307.

Prigent, M., Dubois, T., Raposo, G., Derrien, V., Tenza, D., Rosse, C., Camonis, J. andChavrier, P. (2003). ARF6 controls post-endocytic recycling through its downstreamexocyst complex effector. J. Cell Biol. 163, 1111-1121.

Prigozhina, N. L. and Waterman-Storer, C. M. (2004). Protein kinase D-mediatedanterograde membrane trafficking is required for fibroblast motility. Curr. Biol. 14, 88-98.

Ross, J. S., Figge, H. L., Bui, H. X., del Rosario, A. D., Fisher, H. A., Nazeer, T., Jennings,T. A., Ingle, R. and Kim, D. N. (1994). E-cadherin expression in prostatic carcinomabiopsies: correlation with tumor grade, DNA content, pathologic stage, and clinicaloutcome. Mod. Pathol. 7, 835-841.

Rosse, C., Hatzoglou, A., Parrini, M. C., White, M. A., Chavrier, P. and Camonis, J.(2006). RalB mobilizes the exocyst to drive cell migration. Mol. Cell. Biol. 26, 727-734.

Sans, N., Prybylowski, K., Petralia, R. S., Chang, K., Wang, Y. X., Racca, C., Vicini,S. and Wenthold, R. J. (2003). NMDA receptor trafficking through an interactionbetween PDZ proteins and the exocyst complex. Nat. Cell Biol. 5, 520-530.

Scales, S. J., Pepperkok, R. and Kreis, T. E. (1997). Visualization of ER-to-Golgi transportin living cells reveals a sequential mode of action for COPII and COPI. Cell 90, 1137-1148.

Schmoranzer, J., Kreitzer, G. and Simon, S. M. (2003). Migrating fibroblasts performpolarized, microtubule-dependent exocytosis towards the leading edge. J. Cell Sci. 116,4513-4519.

Shipitsin, M. and Feig, L. A. (2004). RalA but not RalB enhances polarized delivery ofmembrane proteins to the basolateral surface of epithelial cells. Mol. Cell. Biol. 24, 5746-5756.

Spaargaren, M. and Bischoff, J. R. (1994). Identification of the guanine nucleotidedissociation stimulator for Ral as a putative effector molecule of R-ras, H-ras, K-ras,and Rap. Proc. Natl. Acad. Sci. USA 91, 12609-12613.

Sugihara, K., Asano, S., Tanaka, K., Iwamatsu, A., Okawa, K. and Ohta, Y. (2002).The exocyst complex binds the small GTPase RalA to mediate filopodia formation. Nat.Cell Biol. 4, 73-78.

Suzuki, J., Yamazaki, Y., Li, G., Kaziro, Y. and Koide, H. (2000). Involvement of Rasand Ral in chemotactic migration of skeletal myoblasts. Mol. Cell. Biol. 20, 4658-4665.

Takaya, A., Ohba, Y., Kurokawa, K. and Matsuda, M. (2004). RalA activation at nascentlamellipodia of epidermal growth factor-stimulated Cos7 cells and migrating Madin-Darby canine kidney cells. Mol. Biol. Cell 15, 2549-2557.

Takeichi, M. (1993). Cadherins in cancer: implications for invasion and metastasis. Curr.Opin. Cell Biol. 5, 806-811.

Tchevkina, E., Agapova, L., Dyakova, N., Martinjuk, A., Komelkov, A. and Tatosyan,A. (2005). The small G-protein RalA stimulates metastasis of transformed cells.Oncogene 24, 329-335.

TerBush, D. R. and Novick, P. (1995). Sec6, Sec8, and Sec15 are components of amultisubunit complex which localizes to small bud tips in Saccharomyces cerevisiae.J. Cell Biol. 130, 299-312.

TerBush, D. R., Maurice, T., Roth, D. and Novick, P. (1996). The Exocyst is a multiproteincomplex required for exocytosis in Saccharomyces cerevisiae. EMBO J. 15, 6483-6494.

Turner, C. E., West, K. A. and Brown, M. C. (2001). Paxillin-ARF GAP signaling andthe cytoskeleton. Curr. Opin. Cell Biol. 13, 593-599.

Wang, S. and Hsu, S. C. (2006). The molecular mechanisms of the mammalian exocystcomplex in exocytosis. Biochem. Soc. Trans. 34, 687-690.

Wang, S., Liu, Y., Adamson, C. L., Valdez, G., Guo, W. and Hsu, S. C. (2004). Themammalian exocyst, a complex required for exocytosis, inhibits tubulin polymerization.J. Biol. Chem. 279, 35958-35966.

Yeaman, C. (2003). Ultracentrifugation-based approaches to study regulation of Sec6/8(exocyst) complex function during development of epithelial cell polarity. Methods 30,198-206.

Yeaman, C., Grindstaff, K. K. and Nelson, W. J. (1999). New perspectives onmechanisms involved in generating epithelial cell polarity. Physiol. Rev. 79, 73-98.

Yeaman, C., Grindstaff, K. K., Wright, J. R. and Nelson, W. J. (2001). Sec6/8 complexeson trans-Golgi network and plasma membrane regulate late stages of exocytosis inmammalian cells. J. Cell Biol. 155, 593-604.

Yeaman, C., Grindstaff, K. K. and Nelson, W. J. (2004). Mechanism of recruiting Sec6/8(exocyst) complex to the apical junctional complex during polarization of epithelial cells.J. Cell Sci. 117, 559-570.

Zhang, X., Orlando, K., He, B., Xi, F., Zhang, J., Zajac, A. and Guo, W. (2008).Membrane association and functional regulation of Sec3 by phospholipids and Cdc42.J. Cell Biol. 180, 145-158.

Zuo, X., Zhang, J., Zhang, Y., Hsu, S. C., Zhou, D. and Guo, W. (2006). Exo70 interactswith the Arp2/3 complex and regulates cell migration. Nat. Cell Biol. 8, 1383-1388.

Jour

nal o

f Cel

l Sci

ence