14
Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination Benjamin Levine, Jennifer F. Hackney, Andrew Bergen, Leonard Dobens III, Angela Truesdale, Leonard Dobens Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA abstract article info Article history: Received for publication 12 August 2009 Revised 26 April 2010 Accepted 27 April 2010 Available online 5 May 2010 Keywords: Drosophila oogenesis Notch signaling Cux family Stage 10 of Drosophila oogenesis can be subdivided into stages 10A and 10B based on a change in the morphology of the centripetal follicle cells (FC) from a columnar to an apically constricted shape. This coordinated cell shape change drives epithelial cell sheet involution between the oocyte and nurse cell complex which patterns the operculum structure of the mature eggshell. We have shown previously that proper centripetal FC migration requires transient expression of the C/EBP encoded by slow border cells (slbo) at 10A, due in part to Notch activation followed by slbo autorepression (Levine et al., 2007). Here we show that decreased slbo expression in the centripetal FC coincides with increased expression of the transcription factor Cut, a Cut/Cux/CDP family member, at 10B. The 10A/10B temporal switch from Slbo to Cut expression is rened by both cross repression between Slbo and Cut, Slbo auto repression and Cut auto activation. High Cut levels are necessary and sufcient to direct polarized, supracellular accumulation of Actin, DE-cadherin and Armadillo associated with apical constriction of the centripetal FC. Separately, Slbo in the border cell rosette and Cut in the pole cells have antagonistic interactions to restrict Fas2 accumulation to the pole cells, which is important for proper border cell migration. The opposing effects of Cut and Slbo in these two tissues reect the opposing interactions between their respective mammalian homologs CAAT Displacement Protein (CDP; now CUX1) and CAAT Enhancer Binding Protein (C/EBP) in tissue culture. © 2010 Elsevier Inc. All rights reserved. Introduction Cell migration is fundamental to the construction of three- dimensional tissues from cellular units. Cells can move either as loosely organized streams, such as during vertebrate neural crest or neuronal precursor migration (reviewed in Sandell and Trainor, 2006), tightly adherent clusters, such as during border cell migration in the Drosophila ovary, or as coherent cell sheets, such as during Drosophila tracheal tube migration or convergent extension during vertebrate gastrulation (reviewed in Locascio and Nieto 2001; Berg, 2008; Caussinus et al., 2008; Friedl and Gilmour, 2009). The complex cytoskeletal rearrangements and changes in cell adhesion that occur as cells switch from stationary to invasive behaviors is coordinated by both intercellular signals and intrinsic cues, notably transcription factors. Examples include the C/EBP homolog encoded by the gene slow border cells (slbo; Montell et al., 1992; Rorth and Montell, 1992), which regulates levels of the homotypic adhesion molecule DE- cadherin necessary for migration of the border cell cluster, and the transcription factor Snail, which directs a switch from E- to N- cadherin expression required for mesoderm invagination (Cano et al., 2000), a molecular mechanism that is conserved with mass cell migration in vertebrates (reviewed in Come et al., 2004). In some cases, the opposing activities of two transcription factors control the timing and extent of tissue morphogenesis, illustrated by Drosophila knirps and spalt that promote the anterior and posterior tissue migratory behaviors, respectively, of the tracheal tube (Chen et al., 1998). Elucidating how transcription factors modulate the genetic circuits underlying the initiation, guidance and cessation of cell migration will be fundamental to understanding the molecular basis of tissue morphogenesis, wound repair and cancer invasion. Drosophila oogenesis is an excellent model to probe the role of cell signaling during migration (reviewed in Horne-Badovinac and Bilder, 2005; Wu et al., 2008). The ovary is a paired structure composed of 1520 ovarioles, each consisting of a series of attached egg chambers. Individual egg chambers are composed of a single oocyte connected to 15 nutritive nurse cells surrounded by a somatic follicle cell (FC) epithelium. Egg chamber production begins in the proximal germarium where a set of mesodermally-derived FC stem cells proliferate and encapsulate the oocyte-nurse cell syncytium in an epithelial monolayer. Like all epithelia, the FC are polarized cells that contact each other through specialized junction complexes connected to the internal cytoskeleton (Knust and Bossinger, 2002; Dow and Humbert, 2007). Adherens junctions (AJ) interconnect polarized FC, and apical and basal junctions connect the FC sheet to the oocyte and outer basement Developmental Biology 344 (2010) 196209 Corresponding author. Fax: +1 816 523 5995. E-mail address: [email protected] (L. Dobens). 0012-1606/$ see front matter © 2010 Elsevier Inc. All rights reserved. doi:10.1016/j.ydbio.2010.04.030 Contents lists available at ScienceDirect Developmental Biology journal homepage: www.elsevier.com/developmentalbiology

Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Embed Size (px)

Citation preview

Page 1: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Developmental Biology 344 (2010) 196–209

Contents lists available at ScienceDirect

Developmental Biology

j ourna l homepage: www.e lsev ie r.com/deve lopmenta lb io logy

Opposing interactions between Drosophila Cut and the C/EBP encoded by SlowBorder Cells direct apical constriction and epithelial invagination

Benjamin Levine, Jennifer F. Hackney, Andrew Bergen, Leonard Dobens III,Angela Truesdale, Leonard Dobens ⁎Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA

⁎ Corresponding author. Fax: +1 816 523 5995.E-mail address: [email protected] (L. Dobens).

0012-1606/$ – see front matter © 2010 Elsevier Inc. Adoi:10.1016/j.ydbio.2010.04.030

a b s t r a c t

a r t i c l e i n f o

Article history:Received for publication 12 August 2009Revised 26 April 2010Accepted 27 April 2010Available online 5 May 2010

Keywords:Drosophila oogenesisNotch signalingCux family

Stage 10 of Drosophila oogenesis can be subdivided into stages 10A and 10B based on a change in themorphology of the centripetal follicle cells (FC) from a columnar to an apically constricted shape. Thiscoordinated cell shape change drives epithelial cell sheet involution between the oocyte and nurse cellcomplex which patterns the operculum structure of the mature eggshell. We have shown previously thatproper centripetal FC migration requires transient expression of the C/EBP encoded by slow border cells(slbo) at 10A, due in part to Notch activation followed by slbo autorepression (Levine et al., 2007). Here weshow that decreased slbo expression in the centripetal FC coincides with increased expression of thetranscription factor Cut, a Cut/Cux/CDP family member, at 10B. The 10A/10B temporal switch from Slbo toCut expression is refined by both cross repression between Slbo and Cut, Slbo auto repression and Cut autoactivation. High Cut levels are necessary and sufficient to direct polarized, supracellular accumulation ofActin, DE-cadherin and Armadillo associated with apical constriction of the centripetal FC. Separately, Slbo inthe border cell rosette and Cut in the pole cells have antagonistic interactions to restrict Fas2 accumulation tothe pole cells, which is important for proper border cell migration. The opposing effects of Cut and Slbo inthese two tissues reflect the opposing interactions between their respective mammalian homologs CAATDisplacement Protein (CDP; now CUX1) and CAAT Enhancer Binding Protein (C/EBP) in tissue culture.

ll rights reserved.

© 2010 Elsevier Inc. All rights reserved.

Introduction

Cell migration is fundamental to the construction of three-dimensional tissues from cellular units. Cells can move either asloosely organized streams, such as during vertebrate neural crest orneuronal precursor migration (reviewed in Sandell and Trainor,2006), tightly adherent clusters, such as during border cell migrationin the Drosophila ovary, or as coherent cell sheets, such as duringDrosophila tracheal tube migration or convergent extension duringvertebrate gastrulation (reviewed in Locascio and Nieto 2001; Berg,2008; Caussinus et al., 2008; Friedl and Gilmour, 2009). The complexcytoskeletal rearrangements and changes in cell adhesion that occuras cells switch from stationary to invasive behaviors is coordinated byboth intercellular signals and intrinsic cues, notably transcriptionfactors. Examples include the C/EBP homolog encoded by the geneslow border cells (slbo; Montell et al., 1992; Rorth and Montell, 1992),which regulates levels of the homotypic adhesion molecule DE-cadherin necessary for migration of the border cell cluster, and thetranscription factor Snail, which directs a switch from E- to N-cadherin expression required for mesoderm invagination (Cano et al.,

2000), a molecular mechanism that is conserved with mass cellmigration in vertebrates (reviewed in Come et al., 2004). In somecases, the opposing activities of two transcription factors control thetiming and extent of tissue morphogenesis, illustrated by Drosophilaknirps and spalt that promote the anterior and posterior tissuemigratory behaviors, respectively, of the tracheal tube (Chen et al.,1998). Elucidating how transcription factors modulate the geneticcircuits underlying the initiation, guidance and cessation of cellmigration will be fundamental to understanding the molecular basisof tissue morphogenesis, wound repair and cancer invasion.

Drosophila oogenesis is an excellent model to probe the role of cellsignaling during migration (reviewed in Horne-Badovinac and Bilder,2005; Wu et al., 2008). The ovary is a paired structure composed of15–20 ovarioles, each consisting of a series of attached egg chambers.Individual egg chambers are composed of a single oocyte connectedto 15 nutritive nurse cells surrounded by a somatic follicle cell (FC)epithelium. Egg chamber production begins in the proximal germariumwhere a set of mesodermally-derived FC stem cells proliferate andencapsulate the oocyte-nurse cell syncytium in an epithelialmonolayer.Like all epithelia, the FC are polarized cells that contact each otherthrough specialized junction complexes connected to the internalcytoskeleton (Knust and Bossinger, 2002; Dow and Humbert, 2007).Adherens junctions (AJ) interconnect polarized FC, and apical and basaljunctions connect the FC sheet to the oocyte and outer basement

Page 2: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

197B. Levine et al. / Developmental Biology 344 (2010) 196–209

membrane, respectively (reviewed in Tanentzapf et al., 2000; Knust,2002; Szafranski and Goode, 2007). Genetic and biochemicalapproaches show that the AJ, a complex of proteins including asE-Cadherin, Armadillo/β-catenin and α-catenin, is confined to theapicolateral domain by the combined action of (1) the apical Par6/Bazooka/PKC complex and (2) the marginal zone Crb complex,consisting of Crumbs, Stardust and Discs lost. These polarized epithelialjunction complexes stably maintain epithelial sheet tension and tissuerigidity.

Genetic analysis reveals that junction complexes are required notonly for themaintenance of the FC epithelial organization during earlyoogenesis, but also for cell migrations and rearrangements necessaryto pattern the eggshell at later stages (Tepass, 2002; Carthew, 2005;Tepass and Harris, 2007; Niessen and Gottardi, 2008). At stage 9,Notch signaling in the anterior FC directs AJ remodeling duringflattening of the nurse cell FC, a process coordinatedwith formation ofthe columnar main body FC, which pattern the main body features ofthe eggshell (Grammont, 2007). From stages 8–10, the anteriorborder cells (BC) delaminate from the FC epithelium, and migrate as acluster (consisting of two polar cells surrounded by 10–12 rosettecells) through the nurse cell complex to arrive at the border betweenthe nurse cell/oocyte syncytium (Montell, 2003). BC migration isassociated with reorganization of the AJ components Armadillo andCadherin to generate protrusive cell shape changes (Geisbrecht andMontell, 2002). At stage 14, coordinated cell shape changes in thedorsal anterior follicle cells drive the formation of the appendage tubestructures (Berg, 2008).

At stage 10B, the centripetal migrating FC invaginate as an intactepithelium to cover the anterior of the oocyte (Figs. 5A,B). Toaccomplish this, the centripetal FC coordinately constrict apically atstage 10B and acquire adhesivity for the germ line. A set of leadingedge FC derived from the nurse cell FC guides the cell sheet to migratebetween the oocyte and nurse cells. These morphogenetic changescorrelate with increased accumulation of non-muscle myosin and DE-cadherin in the centripetal FC, and mutations in either gene disruptcentripetal migration (Edwards and Kiehart, 1996; Niewiadomska etal., 1999). We have shown previously that Notch activates thetransient expression of the C/EBP homolog encoded by slow bordercells (slbo) in the centripetal FC at stage 10A prior to their involution(Levine et al., 2007). Here we show that as Slbo levels decrease in thecentripetal FC at stage 10B, levels of the Cut gene increase and thathigh levels of Cut at stage 10B are necessary and sufficient for theapical constriction of these cells during centripetal migration. Ourdata indicate that slbo has opposing effects on cut expression topattern both centripetal FC migration and migration of the bordercells. Cut encodes a conserved member of the Cux/CAAT Displace-ment Protein family of proteins with diverse functions in metazoantissue. In mammalian tissue culture, Cux opposes the activity of theSlbo homolog CAAT/Enhancer binding protein (C/EBP) to mediateNotch regulation of proliferation and differentiation (Nepveu, 2001;Sansregret and Nepveu, 2008). Thus the interaction between Cut andSlbo is likely conserved throughout the metazoan lineage.

Materials and methods

Drosophila strains

We used the following stocks: (1) Slbo2.6GAL4;UAS-GFP (geno-type {GAL4-slbo.2.6}1206 P{UAS-GFP.S65T}T2/CyO; Rorth, 1996); (2)cutc145 P{neoFRT}19A /FM2y c ty (Blochlinger et al., 1993; Jackson andBlochlinger, 1997); a gift of Steven Jackson and Celeste Berg,University of Washington; (3) cutDB7 P{neoFRT}19A /FM2y c ty; (4)slbo-lacZ (genotype slbo01310 cn1/CyO; Montell et al., 1992); (5) bun-lacZ (genotype bun2687/CyO; Treisman et al., 1995); (6) w; P{neoFRT}42D slbo8ex2/CyO (a gift of Pernille Rorth, Temasek Life SciencesLaboratory; Rorth, 1994); (7) hsFLP1; bun6903 P{neoFRT}40A /CyO,

(Treisman et al., 1995); (8) UAS-Slbo (Montell et al., 1992); (9) UAS-Bun1/CyO (Treisman et al., 1995); (10) UAS-Cut (Blochlinger et al.,1990); (11) UAS-Nintra (Rebay et al., 1993); (9) Flp-out GAL4, UAS-GFP (genotype y1 w*; P{w+mC=AyGAL4}25 P{w[+mC]=UAS-GFP.S65T}T2/CyO; Ito et al., 1997; Pignoni and Zipursky, 1997); (12) y+,snhsFLP1 w1118; P{neoFRT}42D P{Ubi-GFP(S65T)nls}2R /CyO (gift ofPernille Rorth); (13) P{neoFRT}19A, P{tubP-GAL80}LL1, P{hsFLP}1, w*;P{UAS-mCD8::GFP.L}LL5; actinGAL4; UAS-GFP (a gift of Ting Xie,Stowers Institute); (14) y1 w*; P{Ubi-GFP}33 P{Ubi-GFP}38 P{neoFRT}40A/CyO; (Xu and Rubin, 1993); (15) N55e11 P{neoFRT}18A /FM7a (agift from Kenneth Irvine, Rutgers University; Grammont and Irvine,2001); (16) y1 w; fng13 P{neoFRT}80B /TM6B, Tb1 (Grammont andIrvine, 2001); and (17) DlRevF10 SerRX82 P{neoFRT}82B /TM6B, Tb1

(Heitzler et al., 1996).

Generation of mitotic clones and FLP-out clones

FLP-out clones indicated in the text as Flp-out GAL4 were createdusing the stock y1 w*; P{w+mC=AyGAL4}25 P{w+mC=UAS-GFP.S65T}T2/CyO, y+ (Ito et al., 1997; Pignoni and Zipursky, 1997) infemales of the indicated phenotype. Clones were induced with 30–60 min heat shock at 37 °C 2–4 days prior to dissection. Mitotic cloneswere made by a 60 min heat shock at 37 °C four days prior todissection.

Histochemistry and immunochemistry

Ovaries were dissected and fixed in 4% paraformaldehyde in PBSfor 20 min and then washed 3 times for 5 min in NP-40 wash (50 mMTris–HCl pH 7.4, 150 mM NaCl, 0.5% NP-40, 1 mg/ml BSA). Ovarieswere then blocked for 2 h in 20% BSA in NP-40 wash (Sun and Deng,2005). After blocking, ovaries were incubated in primary antibodyovernight at 4 °C, washed 3 times for 20 min at room temperature andthen incubated in secondary antibody for 2 h at 25 °C. Ovaries werethen washed three times for 20 min and mounted on slides in 50%glycerol in PBS. The following antibodies were used: rabbit anti-β-galactosidase (1:50,000 following extensive preadsorption to ovariantissue; Cappell, West Chester, PA); anti-SLBO (1:2500; Szafranski andGoode, 2004); anti-DECAD (1:100, DCAD2; Oda et al., 1994); mouseanti-Discs large (4F3; Parnas et al., 2001) and anti-Armadillo (1:100,N27A1; Riggleman et al., 1990; Parnas et al., 2001), mouse anti-Fas2(1:100, 1D4; Hummel et al., 2000), mouse anti-Cut (1:100; 2B10,Blochlinger et al., 1990), mouse anti-Eyes Absent (1:100, eya10H6;Bonini et al., 1993). 1:10,000 phalloidin (Sigma, P-1951) and1:100,000 Sytox green (Invitrogen, S7020) were added for 5 min.Confocal images were collected on a Zeiss LSM 410 and analyzed onLSM Image Examiner software. Figures were prepared using Photo-shop CS.

Results

Ordered expression of slbo at 10A and cut at 10B in the centripetal FCresults from cross repression

Previously we identified the C/EBP homolog slow border cells(slbo) in amisexpression screen for targets of the bunched genewhoseexpression is limited to the centripetal FC (Levine et al., 2007). Fromthis screen we identified also the Cux/CDP homolog encoded by thecut gene (Garcia-Bellido, 1975; Johnson and Judd, 1979). As shownpreviously using the monoclonal antisera 2B10, Cut expressionincreases in the FC at stage 2 and Cut levels remain high until stage6 (Fig. 1A; Blochlinger et al., 1993). At these early stages, Cut inhibitsdegradation of CycE; Notch signaling represses Cut expression at stage7, leading to a switch in DNA replication to the endocycle (Sun andDeng, 2005).

Page 3: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 1. slbo expression in the centripetal FC and border cells is complementary to Cut. For all egg chambers anterior is to the left and dorsal is up. (A–A"). Cut expression (red, A")decreases in all FC at stage 7. At stage 9, onset of slbo-lacZ expression from enhancer trap slbo01310 in the migratory border cell is detected (green, arrow, A') from stages 9–14.Genotype: slbo01310/CyO. (B). At stage 10A, slbo01310 expression can be detected in the border cells (BC), centripetal FC (CMFC) and nurse cell FC (NCFC). Cut expression is retained inthe pole cells (PC) at this stage (B"). Two polar cells (PC) retain Cut expression and are visible in yellow (see inset in B", arrowhead). Genotype: slbo01310/CyO. (C). At stage 10B,slbo01310 expression (green) is now absent from the centripetal FC but is retained in the leading edge FC (LE, arrowhead) and in cells at the posterior pole. Onset of late Cut expressionoccurs in a complementary pattern (red): at high levels in the centripetal FC and at lower levels in the main body FC and at higher levels in the posterior FC. Genotype: slbo01310/CyO.(D). At 10B, Cut is expressed at high levels in the centripetal FC (red) where Slbo2.6GAL4;UAS-GFP Slbo levels are now reduced (green, bar). Genotype in D–G: slbo2.6GAL4,UAS-GFP.(E). At 10A, Eyes absent (Eya) is expressed (red) in the nurse cell FC and also marks the leading edge FC (LE); slbo is expressed the centripetal FC (bar) and leading edge FC. (F). At10B, slbo expression (green) decreases in the centripetal FC (bar) but is retained in the leading edge FC (LE). (G). At stage 11, slbo expression is undetectable in the centripetal FC andremains expressed in the leading edge FC (green) along with Eya (red).

198 B. Levine et al. / Developmental Biology 344 (2010) 196–209

Following this switch to the endocycle, Cut expression remainslow inmost FC from stages 7–10A though high Cut levels persist in thepole cells, a subset of the border cells, throughout their migration(Figs. 1A" arrow and B" inset). At stage 10B Cut levels increase in themain body FC, with significantly higher levels in the centripetal FC(Figs. 1C–C" and Blochlinger et al., 1993), marking these cells as theyinvolute between the nurse cell/oocyte complex to cover the anteriorof the oocyte and pattern the operculum.

To better define Cut expression in the centripetal FC, we comparedCut to the expression of the slbo reporter gene (slbo01310; hereafterslbo-lacZ), which is expressed in this tissue at stage 10A (Figs. 1A–C,and 1D; Montell et al., 1992; Rorth, 1996). When slbo-lacZ levels arehigh at stage 10A, Cut levels are low (nuclear lacZ shown in Figs. 1B–B"). Subsequently at stage 10B, slbo-lacZ levels decrease (Levine et al.,2007) concomitant with an increase in Cut levels, again highest in thecentripetal FC and with lower Cut levels in the main body FC (Fig. 1C).As shown in Figs. 1D–G, by stage 11 slbo-lacZ expression persists onlyin the FC at the leading edge of centripetal migration, a set of cells thatexpress Cut as well and are marked effectively by Eyes Absentexpression (Eya, Figs. 1E–G; Bonini et al., 1993). In the border cellcluster, an overlapping pattern of slbo-lacZ and Cut can be seen aswell, where Slbo is expressed in both the inner polar cells and theouter border cell rosette (latter revealed by slbo-lacZ, Fig. 1B, inset). Insum, expression of Cut and Slbo from stages 10–14 marks distinctgroups of migrating cells that comprise the centripetal FC and bordercells, respectively.

Slbo protein accumulation (detected by antisera to Slbo, Figs. 2A,B)parallels transient slbo-lacZ expression in the centripetal FC so thatSlbo protein levels are high at stage 10A (Fig. 2A') and subsequentlyundetectable by 10B (Fig. 2B') at the onset of Cut expression (2B); Slboprotein expression in leading edge FC could not be detected by this

ratherweak antisera. The strict exclusivity of Slbo andCut in these cellssuggests that these genes may repress each other's expression. To testthis, we examined cut or slbo expression in genetic backgrounds thatmanipulate levels of the reciprocal gene. In FLP-out FC clonesexpressing Cut prior to 10B (Fig. 2C), we observed cell autonomousreduction of slbo-lacZ levels in both in the centripetal FC (inset fromFig. 2C shown in Figs. 2E–E") and border cells (inset fromFig. 2C shownin Figs. 2D–D"), the effect of which we will describe in more detail,below. Cut misexpression also reduced Slbo protein levels in bothtissues as well (Supplemental Fig. 3A,B). One concern was thatmisexpression of Cut simply generally reduces expression of all genesin the centripetal FC, so we examined the effect of Cut misexpressionon the expression of the centripetal FC marker genes A359-lacZ(Fig. 2G, Dobens et al., 2000) and pP[ryVM32E(-112/-39)-lacZ] (Fig. 2I,Cavaliere et al., 1997). As shown in Figs. 2G–J, Slbo2.6GAL4 drivingmisexpression of UAS-Cut in the centripetal FC lead to either increasedlevels of pP[ryVM32E(-112/-39)-lacZ] or an increased number of cellsexpressing both markers, in sharp contrast to slbo-lacZ whichdecreased under these conditions (not shown), indicating that Cuteffects are gene-dependent. Complementary to Cut effects on slboexpression, misexpression of Slbo after 10A using the FLP-out driverresulted in reduced levels of Cut, both in the centripetal FC and in themain body FC where low levels of Cut occur (Figs. 2F–F"). FLP-outclones expressing Slbo recovered prior to stage 6 were sufficient torepress early Cut expression (Supplemental Fig. 1), indicating that atall stages of oogenesismisexpression of Slbo effectively can repress Cutexpression.

To determine whether cut was necessary for slbo repression in thecentripetal FC, we examined the effects of cut null allele clones on slboexpression. Consistent with the role of cut in promoting cellproliferation before stage 6, cutc145 null clones were small at late

Page 4: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 2. Cut and Slbo are sufficient to repress each other's expression. (A,B). Accumulation of Slbo (red) and Cut (green) proteins detected by specific antisera. Slbo levels are high inthe centripetal FC at 10A (A') when Cut levels are low (A). Complementarily, at 10B Slbo levels are low (B') while Cut levels are high (B) in the centripetal FC and low in the mainbody FC. (C). slbo expression (slbo01310, red) at stage 10A is repressed by FLP-out misexpression of Cut (clones detected by GFP expression, green). Insets in D and E are boxed.Genotype in C–E: Actin5CNCD2NGAL4; slbo01310/UAS-Cut;UAS-GFP. Phenotype observed in 8/8 clones recovered in the CMFC and 6/9 clones recovered in the BC. (D). Inset from Cshows close-up of a FLP-out Cut-expressing clone in the border cells (marked by GFP, green, D') showing cell autonomous reduction of slbo-lacZ levels (arrowhead, red). (E). Insetfrom C of close-up of FLP-out Cut-expressing clones located in the centripetal FC (Marked by GFP, green, E') showing cell autonomous reduction of slbo-lacZ (arrowhead, E").(F). FLP-out Slbo-expressing clones (marked by GFP, green, F') located in the centripetal FC (arrowheads) and main body FC (one clone indicated by arrow) result in cellautonomous repression of Cut (red, F") in these cells at 10B. Genotype: Actin5CNCD2NGAL4;UAS-Slbo;UAS-GFP. Phenotype observed in 24/24 clones. (G). Wild type expression ofcentripetal FC marker gene A359 at 10B (surface view). Genotype: slbo2.6GAL4; A359-lacZ. (H). Increased expression of A359 occurs in centripetal FC expressing Cut under control ofthe Slbo2.6GAL4 promoter. Genotype: slbo2.6GAL4/UAS-Cut; A359-lacZ. (I). Wild type expression of centripetal FC marker gene pP[ryVM32E(-112/-39)-lacZ] at 10B (cross-sectionview). Genotype: slbo2.6GAL4; pP[ryVM32E(-112/-39)-lacZ]. (J). Increased expression of pP[ryVM32E(-112/-39)-lacZ] occurs in centripetal FC expressing Cut under control of theSlbo2.6GAL4 promoter. Genotype: slbo2.6GAL4/UAS-Cut; pP[ryVM32E(-112/-39)-lacZ].

199B. Levine et al. / Developmental Biology 344 (2010) 196–209

stages, but these clones could be unambiguously detected using apositive marking system (Lee and Luo, 2001). cutc145 clones examinedbefore stage 10B had no effect on slbo-lacZ expression (data notshown), consistentwith low levels of Cut seen at these stages (2A). cutmutant clones (positively marked in Figs. 3A–C) recovered in thecentripetal FC at the onset of Cut expression at late stage 10A showedhigher levels of slbo-lacZ (in cutc145 clones, 3A) or Slbo protein (incutDB7 clones, 3B) and in the posterior polar FC where Cut levels arelow, cutc145 clones led to higher levels of slbo-lacZ (Fig. 3C). Incontrast, cut mutant clones recovered in the main body columnar FCat 10B showed no slbo-lacZ or Slbo misexpression (not shown). Theseresults suggest that cut is necessary and sufficient during lateoogenesis to repress slbo expression in a subset of anterior andposterior follicle cells, presumably cells in which slbo can be activated.

While Cut is necessary and sufficient to repress Slbo, and Slbo issufficient to repress Cut, slbo null clones recovered in the anterior FCresulted in no Cutmisexpression prior to 10B (data not shown) and noincreased Cut expression after (Figs. 3D–D"). In weak slbo mutantallelic combinations (slbo01310/slboex2; low Slbo protein levels areseen in this genotype as shown in Fig. 7B) we noted that Cut and slbo-lacZ expression patterns do overlap (cf. WT in Figs. 3E–E" to 3F–F"),but this is due likely to misexpression of slbo-lacZ at 10B resulting

from a loss of Slbo autorepression, as described previously (Levine etal., 2007). In sum, these data indicate that Slbo is sufficient to repressCut, but reduced slbo activity is not sufficient to trigger precocious Cutexpression before 10B.

High Cut levels in the centripetal FC is regulated by autoactivation

Previously, we have shown that Notch signaling tightly regulatesSlbo expression in the centripetal FC: at 10A, Notch blocks Slboautorepression so that high levels of Slbo expression can be detected(Fig. 2A); at 10B, decreased Notch signaling permits Slbo autorepres-sion with reduced levels of Slbo as a consequence (Levine et al., 2007).Because Notch represses Cut at early stages of oogenesis (Sun andDeng, 2005), we tested whether Notch represses Cut at later stages byexamining Cut levels in clones mutant for Notch (N55e11; Figs. 4A–E)or in clones expressing an activated version of the Notch receptor(Nintra; Fig. 4F).

Consistent with previous reports (Sun and Deng, 2005), N55e11 nullclones resulted in increased Cut expression at stages 9 and 10A (Figs.4A,B). At early 10B, N55e11 clones showed slightly reduced Cutaccumulation compared to 10A clones stained in parallel (cf. Figs.4B to 4C). This suggests that ectopic Cut expression in these 10B Notch

Page 5: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 3. cut is necessary to repress slbo expression. (A). At 10B, slbo expression (slbo01310, red, A") increases in cutmutant clones recovered in the centripetal FC that are positivelymarked byGFP (green, arrowhead, A'). Genotype for A–C: FRT19A, tubP-GAL80 hsFLP1 w−/ FRT19A cutc145; UAS-mCD8::GFP.L/slbo01310; UAS-SrcGFP. Phenotype observed in 5/6 egg chambers. (B). At10B, Slboprotein levels detectedwith specific antisera (red) increase in cutmutant clones recovered in the centripetal FC that are positivelymarkedbyGFP (green, arrowhead). Phenotypeobserved in 3/3 clones. (C). At 10B, slbo expression (slbo01310, red, B") increases in cutmutant clones located in the posterior FC that are positively marked by GFP (green, B'). Phenotypeobserved in2/2 clones. (D).At10B,Cut expression in thecentripetal FC isnot affectedby slbomutant clone located there (markedbyabsence ofGFP, outlined).Note that gainwas increasedto visualize the effects on low levels of Cut expression in these cells. Genotype: hsFLP1; FRT42slbory8ex2d/ FRT42UbiGFP. Phenotypeobserved in 5/5 clones. (E). At 10B, confocal cross-sectionshows slbo expression (green) is limited to leading edge FC (arrowhead) andCut expression (red) in the centripetal FC. Genotype: slbo01310/+. (F). At 10B, a confocal cross-section of a slbomutant egg chamber reveals overlapping expression of slbo-lacZ and Cut in the centripetal FC. Genotype: slbo01310/slboex2.

200 B. Levine et al. / Developmental Biology 344 (2010) 196–209

mutant clones may reflect perdurance of Cut from earlier stages.Consistent with this, at late 10B no increase in Cut expression abovenormal levels can be detected in Notch mutant clones, located eitherin the centripetal FC (Figs. 4D,E) or in the main body FC (Fig. 4E). Aswell, the onset of Cut expression at 10B appears normal in Nts ovariesshifted to the restricted temperature (data not shown; Xu et al., 1992;Dobens et al., 2005) and in clones of fringe or double mutant Delta andSerrate clones (data not shown). Misexpression of the intracellularportion of the Notch receptor, which activates Notch signaling, hasbeen shown to repress Cut expression prior to stage 7 (Sun and Deng,2005). We used the Slbo2.6GAL4 driver to misexpress Nintra in the FCspecifically at 10B and observed no repression of late Cut expressionin the centripetal FC (Fig. 4F). Interestingly, scattered groups of GFPpositive cells misexpressing Nintra that were located in themain bodyFC did repress effectively the low levels of Cut found there (Fig. 4F,arrow) indicating that Cut expression in the main body FC remainssensitive to ectopic Notch signaling, in while Cut regulation in thecentripetal FC is not.

Because cut activates its own expression in some embryonic celltypes (Blochlinger et al., 1991), we examined whether cut regulatesits own expression in the FC. Cut protein levels were significantlyreduced in positively marked mainbody FC clones of the protein nullcutDB7 allele (Fig. 4G), indicating this allele is a protein null and hencenot useful to document effects on Cut protein levels using the Cut2B10 antisera. In contrast, clones of the genetic null allele cut145

recovered in the mainbody FC resulted in normal levels of Cut proteinaccumulation as detected by 2B10 antisera (Fig. 4H), indicating thatthis null allele is not a protein null andwould be useful to examine Cutautoregulation. In support of this, when we examined cut145 mutantclones recovered in the centripetal FC, we observed a significantdecrease in levels of Cut protein accumulation (Fig. 4I), suggesting

that Cut is required to activate its own expression in this tissue. Tofurther test this, we used hsGAL4 to transiently express UAS-Cut inthis tissue. Following a 30 min heat shock induction of hsGAL4, UAS-Cut females (Brand and Perrimon, 1993), Cut levels increased from 3to 6 h after recovery (data not shown). In contrast, a similar transientinduction of hsGAL4,UAS-Slbo led to peak Slbo levels at 2 H andundetectable Slbo by 4 h (data not shown). By 24 h, very high levels ofCut could be detected throughout the FC layer in degrading eggchambers (Fig. 4J). Similar results were obtained following transienthsCut misexpression (data not shown). We conclude that Cut issufficient to activate its own expression in all FC sub-types, butautoactivation has significance only for highest Cut levels in thecentripetal FC.

cut and slbo have opposing effects on follicle cell polarity and apicalconstriction

High levels of Cut in the centripetal FC coincide with a their apicalconstriction at stage 10B (Figs. 5A,B) and is attendant with increasedaccumulation of several apical junction (AJ) components includingDE-cadherin (Fig. 5B), β-catenin and F-actin (Tepass et al., 2000; Yagiand Takeichi, 2000; Gumbiner, 2005). Mutations in DE-cadherin andnon-muscle myosin (NMM) have been shown to result in defectivecentripetal migration, indicating cell shape changes associated withthis migration require cell–cell cohesion and coordinated cytoskeletalactivity (Edwards and Kiehart, 1996; Tepass et al., 1996; Niewia-domska et al., 1999). To examine the significance of high Cut levels forFC shapes, we produced FLP-out Actin5C-GAL4 clones misexpressingUAS-Cut in the FC layer and noted the effect on both cell shapes andaccumulation of AJ components. As can be seen in Fig. 5C, Cut-expressing clones detected by Cut antisera showed levels of Cut

Page 6: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 4. Cut expression in the centripetal FC is Notch independent but cut dependent. (A–E). The effect of Notch mutant clones on Cut accumulation is documented for stages 9 (A) tolate 10B (E). In all, arrows point to clones and arrowheads indicate the posterior edge of the centripetal FC. Genotype for all: FRT18 N55e11/FRT18Ubi-GFP; SbhsFLP. (A,B). At stage 9 (A)and 10 (B), Notch mutant clones show ectopic Cut accumulation (cross-section shown in A, surface view in B). Phenotype observed in 12/12 egg chambers. (C). Faint Cut expressioncan be seen initiating in the centripetal FC (between two arrowhead) at early stage 10B, and main body Notch mutant clones express slightly lower levels of Cut compared to 10A(arrow, egg chambers in A–E are stained in parallel). Phenotype observed in 3/3 egg chambers. (D and E). At late 10B, anterior Notch mutant clones do not lead to increased levels ofCut when compared to adjacent cells, either in the centripetal FC (D cross-section and E surface view) or in themain body FC. No effect on Cut levels observed in 8/8 clones recoveredat this stage. (F). At late 10B, ectopic expression of Nintra has no effect on high Cut levels in the centripetal FC (arrowhead) but leads to repression of main body Cut expression(arrow). Note that gain was increased to visualize the effects on low levels of Cut expression in these cells. Genotype: slbo2.6GAL4/UAS-Nintra. Phenotype observed in 10/10 eggchambers. (G). At 10B, cutDB7 mutant clones recovered in the centripetal FC positively marked by GFP (green, arrow) lead to reduced detection of the Cut epitope, indicating that thisis a protein null allele. Twin spot with 2x levels of Cut epitope is outlined in white. Genotype: FRT19A, tubP-GAL80 hsFLP1 w−/ FRT19A cutDB7; UAS-mCD8::GFP.L; UAS-SrcGFP. (H). cutmutant clone positively marked by GFP (green) located in the main body FC (not shown) and posterior FC has no effect on Cut expression (red, arrow), indicating that Cut has noeffect on its own low levels of expression in these cells and that cutc145 is not a protein null allele. Genotype in I and J: FRT19A, tubP-GAL80 hsFLP1 w−/ FRT19A cutc145; UAS-mCD8::GFP.L; UAS-SrcGFP. Phenotype observed in 8/8 egg chambers. (I). At 10B, cut mutant clones recovered in the centripetal FC positively marked by GFP (green) lead to reduced Cutexpression, indicating that Cut activates its own expression in these cells. Phenotype observed in 3/3 centripetal FC clones. (J). 24 h following 30 min heat shock induction of UAS-Cut, high levels of Cut persist throughout the FC.

201B. Levine et al. / Developmental Biology 344 (2010) 196–209

comparable to the high Cut expression in the centripetal FC. Whenmarked by UAS-lacZ and detected by histological staining, Cut-expressing clones showed two conspicuous phenotypes (Figs. 5D,E):in the columnar FC, clones were thickened compared to adjacent cells(Fig. 5D, arrow); and in the squamous FC, clones failed to flattenproperly (cf. three closely arranged nuclei in Fig. 5D with WTarrangement in Fig. 5E; also see Supplemental Fig. 3C).

These phenotypes led us to examine the effect of Cut expression onepithelial polarity. As can be seen in Fig. 5F, FLP-out Cut-expressingclones in the main body at 10B showed increased DE-cadherinaccumulation to levels comparable to centripetal FC (data not shown).Confocal cross-sections (Figs. 5F–I, panels from left to right,respectively, show sections of the FC epithelia from basal to apicalsides) revealed that cells in Cut-expressing clones constrict apically

Page 7: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 5. High levels of Cut alter FC cell shapes and polarity. (A). In a WT egg chamber at stages 9 and 10A, DE-Cadherin (red) is apically localized in the FC (arrow, top) as anterior FCflatten and posterior FC rearrange to surround the oocyte (O). These changes in the FC epithelium occur in a manner coordinated with border cell migration (border cells, BC). DNA isvisualized with Sytox-green here and in B. (B). At 10B, the centripetal FC (CMFC, arrow) accumulate high levels of DE-cadherin and involute between oocyte (O) and nurse cells.(C). FLP-out expression of UAS-Cut leads to ectopic small clones of Cut-expressing cells in stage 10B egg chambers (arrow) with ectopic Cut levels (in green) similar to the high Cutlevels that occur in the centripetal FC (arrowhead) at this stage. Egg chamber counterstained with DE-cadherin (red). Genotype in C, F–K: Actin5CNCD2NGAL4;UAS-Cut;UAS-GFP.(D). FLP-out expression of UAS-Cut marked by UAS-lacZ and visualized by histological staining reveals two significant phenotypes. First, anterior nurse cell FC expressing UAS-lacZand UAS-Cut fail to flatten so that three nuclei (arrows) are closely apposed in this tissue (compare to clones expressing UAS-lacZ alone in E). Phenotype observed in 18/18 eggchambers. Second, clones expressing UAS-Cut thicken compared to WT FC (arrow). Phenotype observed in 7/10 egg chambers. Genotype: Actin5CNCD2NGAL4;UAS-Cut;UAS-lacZ.(E). FLP-out misexpression of UAS-lacZ alone marks nuclei in the nurse cell FC revealing the normal spreading of cells in this tissue. (F–I). FLP-out actin5CNGAL4 expression of UAS-Cut is marked by GFP (green) and counterstained (in red) for either DE-cadherin (F), Armadillo (G), Actin (phalloidin, H), and Discs large (I). From left to right in each row areconfocal slices moving from a basal view at the left to an apical view of the epithelial sheet (indicated in F). In F, G and H, AJs components are concentrated at the apical face of the FCsheet in cells expressing high levels of Cut. In contrast the basolateral junction protein Dlg shows no apical accumulation in Cut-expressing clones (I). (J). View of (F), stacked androtated to show that cells in Cut-expressing clone of five cells (outlined) lose wild type hexagonal shape (cells numbered 0–6) and form contacts with the five neighbors in the clone.(K). View of (I) rotated and showing that cells in a larger Cut-expressing clone of seven cells (outlined) take on a rounded shape to minimize contacts with surrounding WT cells.

202 B. Levine et al. / Developmental Biology 344 (2010) 196–209

and accumulate DE-cadherin (Fig. 5F), Armadillo (Fig. 5G) and F-actin(Fig. 5H) strongest at the apical vertices where cells in the clone makecontact. As can be seen in Fig. 5F, right-most panel, the concentrationof apically polarized DE-cadherin was greatest at areas of contactbetween the GFP positive, Cut-expressing cells, implying that theseclones have a greater affinity for one another compared to the flanking

WT cells. Cut-expressing cells can appear to pile up on adjacent cellsin cross-section (Fig. 6B), but the epithelium remains intact. Thiscollective change in cell shape and affinity in Cut-expressing cloneslead to the loss of the normal FC hexagonal arrangements (numbered0–6 in Fig. 5J) with the formation of aberrant FC contacts in small Cut-expressing clones of either five neighboring cells (numbered 1*–5*

Page 8: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 6. Cut and Slbo have opposing effects on FC cell shapes and polarity. (A–F). Cross-section of FC epithelium reveals effects of Cut and Slbo on cell polarity. Apical is to the top in all.(A). DE-Cadherin (red) is apically localized in the FC (arrowhead) at stage 10. DNA is stained with Sytox-green. (B). FLP-out clones expressing high levels of Cut (GFP, green)accumulate high levels of apical DE-cadherin (arrowhead, red, cross-section shown). Genotype in B and C: Actin5CNCD2NGAL4;UAS-Cut;UAS-GFP. Phenotype observed in 12/12 eggchambers. (C). FLP-out clones expressing high levels of Cut (GFP, green) accumulate high levels of apical β-catenin (arrowhead, red, cross-section shown). Phenotype observed in8/8 egg chambers. (D). Positively marked cutmutant clones (green) exhibit diffuse basolateral accumulation of DE-cadherin (arrowhead, red, cross-section shown). Genotype in D, Eand H: FRT19A, tubP-GAL80 hsFLP1 w−/ FRT19A cutc145; UAS-mCD8::GFP.L; UAS-SrcGFP. Phenotype observed in 3/3 egg chambers. (E). Positively marked of cutmutant clones (green)exhibit ectopic basolateral accumulation of β-catenin (arrowhead, red, cross-section shown). Phenotype observed in 5/5 egg chambers. (F). FLP-out clones expressing high levels ofSlbo (GFP, green) exhibit ectopic basolateral accumulation of DE-cadherin (arrowhead, red, cross-section shown). Genotype: y1 w*; P(w+mC=AyGAL4)25 P(w+mC=UAS-GFP.S65T)T2;UAS-Slbo;UAS-GFP. Phenotype observed in 6/6 egg chambers. (G). Anterior slbo mutant clones (marked by absence of GFP) show increased accumulation DE-cadherin andconvergent phenotype (arrow, red, surface-section shown). Genotype: hsFLP1; FRT42slbory8ex2d/ FRT42 UbiGFP. Phenotype observed in 3/5 anterior clones. (H). Anterior cutmutantclone (positively marked by GFP) show decreased and disorganized accumulation DE-cadherin (arrow, red, surface-section shown). Phenotype observed in 5/6 anterior clones.

203B. Levine et al. / Developmental Biology 344 (2010) 196–209

and outlined in Fig. 5J) or four neighbors (not shown). In larger clones,the areas of contact between Cut-expressing cells and wild type cellsresulted in a round clone shape (Fig. 5K), a cell arrangement thatlikely reflects differences in DE-cadherin levels and affinity (Carthew,2005).

The striking effect of Cut misexpression on the apical face of the FCepithelium led us to examine the basolateral FC face of Cut clones.Basolateral junction marker Discs Large (Dlg) levels were increasedoverall in Cut misexpressing clones, but no change in either basal orapical accumulation of Dlg was evident (Fig. 5I). These data show thatmisexpression of Cut in the main body columnar FC is sufficient toremodel apical but not basal junctions, resulting in supracellularreorganization of the cytoskeleton that resemble the centripetallymigrating FC at stage 10B (Fig. 5B).

The ability of high Cut levels to generate ectopic centripetal FCphenotypes, including increased accumulation of AJs components(Figs. 6B,C) and apically constricted cell shapes (Fig. 5), led us toexamine the effect of cut mutations on late stages of oogenesis. Weak

cut mutations that affect wing margin patterning resulted in fertilefemales (data not shown), though close inspection of eggs laid by ct6

wing notch females revealed small, irregular eggshell imprints,consistent with the effects of this allele on cell size, and short dorsalappendages with a turned paddle, consistent with a possible effect onepithelial cell morphogenesis (Supplemental Fig. 2). In contrast to Cutmisexpression clones in which DE-cadherin and Arm are apicallylocalized (cross-section in Figs. 6B,C, respectively), cutc145 null clonesresulted in diffuse accumulation of these proteins throughout the cell,both in clones recovered in the mainbody FC (Figs. 6D,E) and in clonesrecovered in the centripetal FC coincident with centripetal migration(Fig. 6H). Together with data from Fig. 4, these results indicate that cutis required to maintain AJ polarization and that high levels of Cut inthe centripetal FC drive apical constriction associated with centripetalFC migration.

Given the antagonistic effects of cut and slbo in the FC, weexamined more closely the effect of both on cell polarity in late stageegg chambers. Consistent with an opposing interaction between Cut

Page 9: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

204 B. Levine et al. / Developmental Biology 344 (2010) 196–209

and Slbo, UAS-Slbo FLP-out clones resulted in elevated levels of DE-Cadherin accumulating at basolateral positions (Fig. 6F), a phenotypesimilar to the expression seen in cutc145mutant clones (cf. Figs. 6D and6F). FLP-out clones that co-expressed Slbo and Cut showed increasedDE-Cadherin levels but the impact of Slbo co-expression on apicalconstriction phenotypes were difficult to interpret (data not shown).slboex2a null clones recovered in the main body FC prior to stage 10Bdid not have any effect on DE-cadherin levels nor did late slboex2a

clones located posterior to the centripetal FC, where slbo levels arelow (data not shown). In contrast, late stage slboex2a mutant clonesthat contact the centripetal FC resulted in increased DE-cadherinlevels and convergence of cells to the clone's center (Fig. 6G), aphenotype superficially similar to that seen in FLP-out Cut-expressingclones. Larger anterior slboex2amutant clones led to a breakdown in FCepithelial integrity and egg chamber collapse and hence were difficultto document (data not shown). These data suggest that reduction ofSlbo activity in anterior cells is necessary for apical constriction of cellsin the sheet.

slbo and cut have opposing effects on Fas2 expression to regulate bordercell migration

The opposing effects of Cut and Slbo on cell shapes and AJaccumulation during centripetal migration led us to examine theirinteractions during border cell migration. Cut protein accumulates inthe pair of polar cells (Fig. 7A', arrowhead), while Slbo protein isexpressed in both the polar cells and border cell rosette (Fig. 7A",arrow). The weak slbo mutant slbo01310/slbo01310 resulted in reducedSlbo protein accumulation throughout the border cells (Fig. 7B") andectopic, nuclear Cut accumulation in the rosette cells (Fig. 7B', arrow),indicating that slbo represses Cut in these cells. We have shownpreviously that slbo represses its own expression measured by slbo-lacZ in the centripetal FC (Levine et al., 2007) and here we observedthat in the slbo transheterozygous mutant slbo01310/ slboex2, (1)misexpression of Cut occurred in the rosette cells (Fig. 7D'; weakmisexpression in this genotype is marked by arrow) and (2) increasedslbo-lacZ (Fig. 7D") expression occurred throughout the border cellcluster, including the polar cells that do not strongly express the slbo-lacZ reporter in the WT (cf. Fig. 7D" to WT in 7C" stained in parallel).Consistent with our observations in the centripetal FC, misexpressionof Cut in the border cell rosette using either the FLP-out (Fig. 2C) orSlbo2.6Gal4 drivers (Rorth, 1996) led to a reduction in Slbo levels(Figs. 7E,E') and a block in border cell migration (Fig. 7F). These dataindicate that patterning of cut and slbo expression in the border cellcluster and centripetal FC relies on both cross repression and slboautorepression (Fig. 2C and Levine et al., 2007).

In the course of these experiments we noted that Cut expression inwild type follicle cells parallels closely the reported expression ofFasciclin2 (Fas2; Hummel et al., 2000), a transmembrane cell

Fig. 7. Cut and Slbo have opposing effects on gene expression in the border cells. (A–A"). Cutstage 9 (arrowhead) while Slbo (red) is expressed in both the polar cells and rosette cells (ar(red) is undetectable and Cut protein (green) is misexpressed in the rosette cells (arrow). Thstained in parallel to (A). Genotype: slbo01310/ slbo01310. (C–C"). Wild type Slbo reporter genpolar cells (seen in C"), a pattern that parallels Slbo protein accumulation (in A") and isGenotype: slbo01310/+. (D–D"). In slbo mutant egg chambers, ectopic expression of Cut (redgenotype. As well, all cells of the border cell cluster including the rosette and polar cells (acompared to control (C). Note that (C) and (D) are stained in parallel. Genotype: slbo01310/slbexpression in the rosette. Genotype in E, F and H: slbo2.6GAL4,UAS-GFP/UAS-Cut. (F,F'). Stageborder cell rosette is sufficient to block centripetal migration. (G,G'). WT expression of Fas2between the two polar cells and at the posterior side of the polar cell pair. Slbo2.6GAL4 drivinGFP. (H,H'). Cut misexpression in FC rosette leads to Fas2 (red) misexpression in the rosette ccharacteristic positioning of the polar cells were also noted (not shown). (I,I'). Stage 6 egg chamreduction in Fas2 levels (red). Genotype in I and K: hsFLP UAS-SrcGFP TubGAL80 FRT19A/cuchamber showing ectopic Fas2 (red) occurs in FLP-out clones expressing Cut (GFP, green). Enseen in this image (arrowhead). Genotype in J and L: hsFLP1; y1 w*; P(w+mC=AyGAL4)25 P(Stage 10B egg chamber with cutc145 mutant clone located in the centripetal FC (positively mobserved in 4/4 egg chambers. (L,L'). Stage 11 egg chamber with ectopic Fas2 (red) in FLP-o

adhesion molecule of the immunoglobulin superfamily that binds tothe scaffolding protein Discs Large (Dlg; Szafranski and Goode, 2004).Prior to stage 7, Cut and Fas2, as detected by monoclonal antisera(1D4; Hummel et al., 2000), are co-expressed in all FC. At stage 7,accumulation of both proteins decreases in all cells except the polarcells (cf. Figs. 7A' and G'). In these cells, Fas2 accumulates in apolarized manner (1) at the membrane where the two polar cellscontact and (2) apically at the leading edge of border cell migration(Szafranski and Goode, 2004). Szafranski and Goode (2004) showedthat ectopic Fas2 expression in the border cell rosette disrupts bordercell migration and concluded that Fas2 restricted to the polar cells isrequired to control the timing of border cell cluster delamination andthe speed and direction of border cell migration.

Given both the similarities of both the expression patterns of Fas2and cut in the pole cells and misexpression phenotypes on border cellmigration, we tested if cut regulates Fas2 in this tissue. cut mutantclones in the outer rosette cells had no effect on border cell migration(not shown) consistent with low levels of Cut in these cells. Clonescould not be recovered in the polar cells during their migration so weexamined Fas2 levels following Slbo2.6GAL4 misexpression of Cut inthe rosette border cells. Misexpression of Cut led to increased Fas2 inthe rosette cells (Figs. 7H,H') resulting in reduced Fas2 at the interfaceof the polar FC and mis-accumulation of Fas2 at rosette cell/polar cellinterfaces (Fig. 7H') in clusters that failed to migrate, a phenotypeidentical to the effect of Fas2 misexpression in the rosette cells(Szafranski and Goode, 2004).

In the main body FC, Fas2 and Cut have overlapping patterns ofexpression at several stages, indicating that Cut may regulate Fas2 inthese cells. Prior to stage 6, positively marked cutmutant clones in theFC resulted in cell autonomous reduction of Fas2 levels and loss ofbasolateral Fas2 accumulation (Figs. 7I,I'). After stage 6 when Cutlevels are low, FLP-out Cut-expressing clones recovered in themainbody FC resulted in high levels of ectopic Fas2 (Fig. 7J,J'). Atstage 10, Fas2 expression in the dorsal centripetal FC appears tooverlap with Cut expression and consistent with activation by Cut atthis stage, cut mutant clones led to reduced Fas2 in the dorsalcentripetal FC (Figs. 7K,K') and FLP-out clones expressing Cut in themain body FC led to ectopic Fas2 expression (Figs. 7L,L'). We concludethat cut is necessary and sufficient for restricted Fas2 expression in thecentripetal FC and border cell cluster and Fas2 regulation by Cut isimportant for cell migration of both cell types.

Discussion

cut directs apical constriction of anterior FC

The centripetal FC are patterned at stage 10A and then undergo acoordinated apical cell constriction at 10B leading to a collectiveinvolution of the FC epithelial sheet, which penetrates between the

protein accumulation (green) in wild type egg chambers is restricted to the polar cells atrow). Genotype: slbo01310/+. (B–B"). In a slbomutant background, nuclear Slbo proteine organization of the cluster is aberrant and nuclei are larger in this mutant egg chambere expression (red, slbo01310) occurs in the border cell rosette (arrow) and weakly in thecomplementary to Cut (green, C'), which is limited to the polar cells (arrowhead).

) occurs in the border cell rosette (arrow, red in D') as in (B), but at lower levels in thisrrow and arrowhead, respectively) express higher levels of slbo-lacZ (green, D) whenoex2. (E). Slbo (red) expression in Slbo2.6GAL4; UAS-Cut egg chambers reveals decreased10B egg chamber counterstained for Fas2 reveals that misexpression of UAS-Cut in the(red) shows high levels in the polar cells with the highest concentration at the areas

g GFP expression (green) reveals the location of the rosette. Genotype: slbo2.6GAL4,UAS-ells and accumulation at the anterior and posterior ends of the polar cells. Defects in theberwith a cutc145mutant clone (positivelymarkedwithGFP) showing cell autonomous

tc145 FRT 19A; actinGAL4; UAS-GFP. Phenotype observed in 7/7 clones. (J,J'). Stage 9 eggdogenous Fas2 expression in the inner polar cells of migrating border cell cluster can bew+mC=UAS-GFP.S65T)T2 / UAS-Cut. Phenotype observed in 8/8 egg chambers. (K,K').arked with GFP) showing cell autonomous reduction in Fas2 levels (red). Phenotypeut clones expressing Cut (GFP, green). Phenotype observed in 6/6 egg chambers.

Page 10: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

205B. Levine et al. / Developmental Biology 344 (2010) 196–209

germ line oocyte and nurse cells from stages 11–12. From a geneticscreen for candidate targets of bunched repression that are expressedin the centripetal FC during stage 10, we identified Cut. Classic cutmutant phenotypes include wing margin notches and leg malforma-tions and molecularly these map to a large and complex regulatoryregion that exerts tissue-specific control of cut expression; cut nullmutations are embryonic lethal with defects in neural patterning(Johnson and Judd, 1979; Jack and DeLotto, 1995).

cut encodes a member of the Cux family of proteins foundthroughout themetazoan lineage (Alcalay and Vanden Heuvel, 2009);

these homologs are expressed in diverse tissues and act generally toeither prevent tissue differentiation or promote mitotic cycle entry individing cells or specify cell fate in postmitotic cells (reviewed inNepveu, 2001). Cux familymembers are distinguished by a homeoboxand multiple Cut repeats, which are conserved DNA binding motifsthought to act in a combinatorial manner to allow a single Cut-likeprotein to bind to diverse DNA sequences and regulate distinctpromoters. To add to this complexity, distinct isoforms of human CDPhave specific effects on cell differentiation and cancer (Goulet et al.,2002). Splice isoforms of the Drosophila cut gene have been detected

Page 11: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

206 B. Levine et al. / Developmental Biology 344 (2010) 196–209

recently, but their specific functions and regulation are untested.Here we show that high levels of Cut expression in the centripetal

FC correlate with the apical constriction and coordinated involution ofthese cells during centripetal migration at stage 10B of oogenesis. Thiscollective cell shape change can be recapitulated by misexpression ofhigh levels of Cut in the columnar mainbody FC. Interestingly Cutmisexpressing clones show supracellular cytoskeletal reorganization,remodeling cadherin-based interactions between expressing and non-expressing cells adjacent to the clone. Supracellular reorganization ofcadherin–actin networks is associated with protrusion activities andretraction dynamics that involve many cells (reviewed in Friedl et al.,2004), but the mechanism underlying these shared and coordinatedbehaviors is unclear. cut mutant clones led to reduced DE-cadherinlevels but only subtle effects on apical invagination. Because only smallcut clones could be recovered due to the early requirement for cut incell proliferation, it is possible that these small clones are effectivelycarried by their neighbors during the invagination process.

The correlation between high Cut expression levels in theapically constricted centripetal FC compared to lower Cut levels inthe columnar-shaped main body FC recalls the distinct Cut levels insensory neurons with distinct dendritic branching patterns: highCut in neurons with extensive unbranched terminal protrusions,medium levels in neurons with complex arbors, and low Cut inneurons with simple dendrites (Grueber et al., 2003). Thus Cutlevels may dictate cell shapes in some tissues. It is noteworthythat high Cut levels, either normally in the centripetal FC or in FCmisexpressing Cut, result in no loss of epithelial character, quiteunlike invasive phenotypes associated with mutations in Fas2, dlg,lethal giant larva (lgl), and Neuroglian (Nrg, Goode et al., 1992, 1996;Szafranski and Goode, 2004). The effect of Cut on the apical junctionbut not the basal junction may explain failure of these cells to leavethe epithelium.

Cross repression between cut and slbo refines a genetic switch ingene expression

Cut and Slbo exhibit overlapping expression patterns in theleading edge FC and pole cells but complementary expression in thecentripetal FC and border cell rosette (Figs. 1, 2 and 7). The opposingactivities of their mammalian counterparts suggested to us that cutand slbo might repress each other's expression in the latter cell types,a notion that is supported by our demonstration that (1) Slbo or Cutmisexpression is sufficient to repress the expression of the reciprocalgene in the centripetal FC and border cell rosette and (2) cut mutantclones show increased slbo-lacZ and Slbo protein expression and slbomutants show ectopic Cut in the border cells.

The switch from Slbo at 10A to high levels of Cut at 10B in thecentripetal FC coincides with increased accumulation of key apicaljunction components and our genetic manipulations show that cutand slbo have opposing effects on the accumulation of DE-cadherin inthis cell type. Cadherins are the dominant mediator of collective cellinteractions in cells undergoing diverse motile processes such asgastrulation, primitive streak and neural crest migration, epiblastingression (Edelman et al., 1983; Hatta and Takeichi, 1986; Takeichi,1988; Takeichi et al., 2000) as well as tumor invasiveness (reviewed inPeinado et al., 2004). Increased expression of cadherin isoforms andswitching between cadherin types occurs during cell migration andpositive and negative transcriptional regulation of E- and N-cadherinexpression is exerted in part by a number of zinc-finger transcriptionfactors including Snail, Slug and Twist (reviewed in Peinado et al.,2007). In Drosophila, Twist promotes motile phenotypes by directingE- to N-cadherin switching (Oda et al., 1998; Alexander et al., 2006).While no role for vertebrate Cux-like proteins in cadherin switchinghas been identified previously, the cutmammalian homologue CUTL1mediates TGFβ enhancement of cancer cell motility and invasiveness

by via transcriptional upregulation of N-cadherin (Maeda et al., 2005)and WNT5A (Michl et al., 2005; Michl and Downward, 2006).

Genetic and molecular approaches have characterized the largeupstream regulatory region that controls Cut expression in diversetissues (e.g. Bodmer et al., 1987; Liu and Jack, 1992) and several genesrequired for Cut expression in the embryonic nervous system havebeen identified (reviewed in Bellaiche and Schweisguth, 2001). Notchsignaling has tissue-specific effects on Cut throughout development:during wing patterning, Notch activates Cut at the margin (de Celisand Bray, 1997; Michelli et al., 1997), while in the ovary Cut isrepressed by Notch at stages 7–10A (Sun and Deng, 2005). Our datashow that during late oogenesis, Cut levels in the centripetal FC at 10Bincrease as Notch-activated slbo expression decreases. Thus it wassurprising that Notch mutant clones and Nintra misexpression had noeffect on Cut levels in the centripetal FC, leading us to conclude thatCut activation in these cells occurs in a manner independent of Notch.In the mainbody FC by contrast, Nintra misexpression did effectivelyrepress low Cut levels, indicating that distinct regulatory pathwaysregulate Cut expression in the centripetal FC compared to the mainbody FC. Consistent with this, cutc145 clones in the centripetal FC showsignificant reduction in Cut levels, while mutant clones of cutc145

recovered in the main body FC show no reduction in Cut expression.Evidence that Cut activates its own expression in embryonic cell typescomes from experiments showing that transient, ubiquitous mis-expression of a heat shock inducible Cut transgene during embryo-genesis was sufficient for persistent Cut accumulation in transformedchordotonal cells, but not in other cell types (Blochlinger et al., 1991),and similarly, whenwe expressed heat shock Cut transiently in the FC,we observed a continuous increase in Cut levels so that by 24 h, Cutlevels were very high in all FC subtypes in egg chambers that appeareddegraded. We conclude that if expressed at high enough levels, Cutcan activate its own expression throughout the FC, but Cut autoactivation normally occurs only in the centripetal FC to achieve thehigh Cut levels necessary to direct apical constriction of thesemigrating cells.

A working model summarized in Fig. 8 states that during bordercell migration (8A), Cut is expressed in the polar cells and Slbo isexpressed throughout the border cells, including both the polar cellsand rosette cells and cross repression maintains this pattern ofexpression. In the centripetal FC (8B), transient Slbo expression atstage 10A represses both Cut expression (here) and its ownexpression (Levine et al., 2007). At 10B, Cut represses Slbo andactivates high levels of its own expression in the centripetal FC asthese constrict apically. We showed previously that in the centripetalFC, Notch signaling activates slbo-lacZ expression (Levine et al., 2007)while here we show that ectopic Nintra has no effect on late Cutexpression in this tissue (Fig. 4F). This is difficult to reconcile with theability of Slbo misexpression to effectively repress Cut (Fig. 2F). Basedon work here, we note that slbo-lacZ levels can be high in the bordercells when Slbo protein levels are low (cf. Fig. 7B" to 7D"). Wespeculate that Nintra blocks normal slbo autorepression to increaseslbo-lacZ levels with slight to no affect on Slbo protein levels(unpublished observations) while Flp-out Slbo circumvents auto-repression entirely and effectively represses Cut. Epistasis experi-ments should elucidate further the significance of these proposedinteractions for the Slbo/Cut expression switch.

Opposing activities of cut and slbo pattern border cell migration

Formation of the border cell cluster begins at stage 7 when thecytokine-like signal Unpaired, which is produced by the polar cells,activates the JAK/STAT pathway in neighboring cells recruiting theseto the outer border cell fate. JAK/STAT signaling in turn activates anumber of downstream border cell migration-associated genes,including slbo (Xi et al., 2003; Silver et al., 2005). Stage 7 coincideswith the loss of Cut in all FC except the polar cells while Slbo protein is

Page 12: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

Fig. 8.Model for Cut and Slbo interactions in the follicle cells. (A). During border cell migration, Cut is expressed in the polar cells (green) and Slbo is expressed throughout the bordercells, including both the polar cells and rosette cells (yellow). Cut misexpression in the rosette cells blocks Slbo expression and in slbo mutants Cut is misexpressed in the rosettecells; both manipulations block border cell migration. (B). At 10A, transient Slbo expression (yellow) represses both Cut expression (here) and its own expression (Levine et al.,2007). (C). At 10B, Cut represses Slbo and activates high levels of its own expression (dark green) in the centripetal FC as these constrict apically; Cut is expressed at low levels in themain body FC (light green).

207B. Levine et al. / Developmental Biology 344 (2010) 196–209

expressed in both the inner polar and outer rosette cells of the bordercell cluster (Fig. 7A"). The overlapping Slbo/Cut pattern is critical forproper border cell migration because in slbo mutants, ectopic Cutaccumulates in the rosette cells of border cell clusters that fail tomigrate. Complementarily, misexpression of Cut in the outer bordercells leads to a block in border cell migration and Slbo repression. Cutactivates Fas2 in both the pole cells and centripetal FC, the lattersuggesting that polarized Fas2 coordinates centripetal FC invaginationin addition to its role in polarizing the pole cell pair.

Superficially, the border cells and the centripetally migrating FCuse different strategies to accomplish their respective migrations: theborder cell cluster undergoes an epithelial to mesenchymal transitionto delaminate from the FC layer and migrate as a cluster of cells whilethe centripetal FC retain the structure of an epithelial sheetthroughout their involution. Slbo/Cut expression seems distinct aswell: their patterns are complementary in a temporal manner in thesame cells during centripetal migration, but during border cellmigration their expression is complementary in distinct cell typesbut contemporaneous (Fig. 8A,C,D). Similarities in these tissues havebeen noted: the border cell cluster retains a quasi-epithelialorganization throughout its migration; both migrating groups ofcells presumably make similar changes in affinities to enter the germcell environment and proceed following similar cues; and bothmigrating groups are marked by a leading edge cell with cleardifferences in cell morphology and gene expression (Borghese et al.,2006;Wang et al., 2006; Levine et al., 2007). Because diffuse, punctateDE-cadherin accumulation in the rosette cells of the migrating bordercell cluster has been attributed to slbo-directed rapid endosomal-mediated turnover (Niewiadomska et al., 1999; Pacquelet and Rorth,2005), it is possible that transient expression of slbo in the centripetalFC (Levine et al., 2007) at stage 10A initiates rapid endosomal-mediated turnover necessary for the launch of invagination. And asCut regulates Fas2 to organize the border cell cluster, Cut at 10Bdirects polarized accumulation of DE-cadherin and Fas2 to coordinateapical constriction and cell sheet spreading. Thus it is likely that Cut/Slbo interactions during both border cell and centripetal migrationregulate an underlying common molecular mechanism that works inboth multicellular movements.

Acknowledgments

We thank Stephen Jackson, Celeste Berg, Pernille Rorth, GertrudeSchupbach and Kenneth Irvine for fly strains. We thank Jeffrey Price,Erika Geisbrecht, Whitney McBride, lab members and an anonymousreviewer for helpful comments. 2B10, Eya and ID4 antisera wereobtained from the Developmental Studies Hybridoma Bank developedunder the auspices of the NICHD and maintained by The University ofIowa. Slbo antisera were a generous gift from Scott Goode andPrzemyslaw Szafranski. This work was supported by a research grantfrom the National Science Foundation (IOS-0920613) and from theUniversity of Missouri Research Board to LLD.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, inthe online version, at doi:10.1016/j.ydbio.2010.04.030.

References

Alcalay, N.I., Vanden Heuvel, G.B., 2009. Regulation of cell proliferation anddifferentiation in the kidney. Front Biosci. 14, 4978–4991.

Alexander, N.R., Tran, N.L., Rekapally, H., Summers, C.E., Glackin, C., Heimark, R.L., 2006.N-cadherin gene expression in prostate carcinoma is modulated by integrin-dependent nuclear translocation of Twist1. Cancer Res. 66, 3365–3369.

Bellaiche, Y., Schweisguth, F., 2001. Lineage diversity in the Drosophila nervous system.Curr. Opin. Genet. Dev. 11, 418–423.

Berg, C.A., 2008. Tube formation in Drosophila egg chambers. Tissue Eng. Part A 14,1479–1488.

Blochlinger, K., Bodmer, R., Jan, L.Y., Jan, Y.N., 1990. Patterns of expression of cut, aprotein required for external sensory organ development in wild-type and cutmutant Drosophila embryos. Genes Dev. 4, 1322–1331.

Blochlinger, K., Jan, L.Y., Jan, Y.N., 1991. Transformation of sensory organ identity byectopic expression of Cut in Drosophila. Genes Dev. 5, 1124–1135.

Blochlinger, K., Jan, L.Y., Jan, Y.N., 1993. Postembryonic patterns of expression of cut, alocus regulating sensory organ identity in Drosophila. Development 117, 441–450.

Bodmer, R., Barbel, S., Sheperd, S., Jack, J.W., Jan, L.Y., Jan, Y.N., 1987. Transformation ofsensory organs by mutations of the cut locus of D. melanogaster. Cell 51, 293–307.

Bonini, N.M., Leiserson, W.M., Benzer, S., 1993. The eyes absent gene: genetic control ofcell survival and differentiation in the developing Drosophila eye. Cell 72, 379–395.

Borghese, L., Fletcher, G., Mathieu, J., Atzberger, A., Eades, W.C., Cagan, R.L., Rorth, P.,2006. Systematic analysis of the transcriptional switch inducing migration ofborder cells. Dev. Cell 10, 497–508.

Page 13: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

208 B. Levine et al. / Developmental Biology 344 (2010) 196–209

Brand, A.H., Perrimon, N., 1993. Targeted gene expression as a means of altering cellfates and generating dominant phenotypes. Development 118, 401–415.

Cano, A., Perez-Moreno, M.A., Rodrigo, I., Locascio, A., Blanco, M.J., del Barrio, M.G.,Portillo, F., Nieto, M.A., 2000. The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat. Cell Biol. 2,76–83.

Carthew, R.W., 2005. Adhesion proteins and the control of cell shape. Curr. Opin. Genet.Dev. 15, 358–363.

Caussinus, E., Colombelli, J., Affolter, M., 2008. Tip-cell migration controls stalk-cellintercalation during Drosophila tracheal tube elongation. Curr. Biol. 18, 1727–1734.

Cavaliere, V., Spano, S., Andrenacci, D., Cortesi, L., Gargiulo, G., 1997. Regulatoryelements in the promoter of the vitelline membrane gene VM32E of Drosophilamelanogaster direct gene expression in distinct domains of the follicularepithelium. Mol. Gen. Genet. 254, 231–237.

Chen, C.K., Kuhnlein, R.P., Eulenberg, K.G., Vincent, S., Affolter, M., Schuh, R., 1998. Thetranscription factors KNIRPS and KNIRPS RELATED control cell migration andbranch morphogenesis during Drosophila tracheal development. Development125, 4959–4968.

Come, C., Arnoux, V., Bibeau, F., Savagner, P., 2004. Roles of the transcription factorssnail and slug during mammary morphogenesis and breast carcinoma progression.J. Mammary Gland Biol. Neoplasia 9, 183–193.

de Celis, J.F., Bray, S., 1997. Feed-back mechanisms affecting Notch activation at thedorsoventral boundary in the Drosophila wing. Integr. Annu. Index. 124,3241–3251.

Dobens, L., Petersen, J., Treisman, J., Raftery, L., 2000. Drosophila bunched integratesopposing DPP and EGF signals to set the operculum boundary. Development 127,745–754.

Dobens, L., Jaeger, A., Peterson, J.S., Raftery, L.A., 2005. Bunched sets a boundary forNotch signaling to pattern anterior eggshell structures during Drosophilaoogenesis. Dev. Biol. 287, 425–437.

Dow, L.E., Humbert, P.O., 2007. Polarity regulators and the control of epithelialarchitecture, cell migration, and tumorigenesis. Int. Rev. Cytol. 262, 253–302.

Edelman, G.M., Gallin, W.J., Delouvee, A., Cunningham, B.A., Thiery, J.P., 1983. Earlyepochal maps of two different cell adhesion molecules. Proc. Natl. Acad. Sci. U. S. A.80, 4384–4388.

Edwards, K.A., Kiehart, D.P., 1996. Drosophila non-muscle myosin II has multipleessential roles in imaginal disc and egg chamber morphogenesis. Development122, 1499–1511.

Friedl, P., Gilmour, D., 2009. Collective cell migration in morphogenesis, regenerationand cancer. Nat. Rev. Mol. Cell Biol. 10, 445–457.

Friedl, P., Hegerfeldt, Y., Tusch, M., 2004. Collective cell migration inmorphogenesis andcancer. Int. J. Dev. Biol. 48, 441–449.

Garcia-Bellido, A., 1975. Genetic control of wing disc development in Drosophila. CibaFoundation Symposium, pp. 161–182.

Geisbrecht, E.R., Montell, D.J., 2002. Myosin VI is required for E-cadherin-mediatedborder cell migration. Nat. Cell Biol. 4, 616–620.

Goode, S., Wright, D., Mahowald, A.P., 1992. The neurogenic locus brainiac cooperateswith the Drosophila EGF receptor to establish the ovarian follicle and to determineits dorsal-ventral polarity. Development 116, 177–192.

Goode, S., Melnick, M., Chou, T.-B., Perrimon, N., 1996. The neurogenic genes eggheadand brainiac define a novel signaling pathway essential for epithelial morphogensisduring Drosophila oogenesis. Development 122, 3863–3879.

Goulet, B., Watson, P., Poirier, M., Leduy, L., Berube, G., Meterissian, S., Jolicoeur, P.,Nepveu, A., 2002. Characterization of a tissue-specific CDP/Cux isoform, p75,activated in breast tumor cells. Cancer Res. 62, 6625–6633.

Grammont, M., 2007. Adherens junction remodeling by the Notch pathway inDrosophila melanogaster oogenesis. J. Cell Biol. 177, 139–150.

Grammont, M., Irvine, K.D., 2001. fringe and Notch specify polar cell fate duringDrosophila oogenesis. Development 128, 2243–2253.

Grueber, W.B., Jan, L.Y., Jan, Y.N., 2003. Different levels of the homeodomain protein cutregulate distinct dendrite branching patterns of Drosophilamultidendritic neurons.Cell 112, 805–818.

Gumbiner, B.M., 2005. Regulation of cadherin-mediated adhesion in morphogenesis.Nat. Rev. Mol. Cell Biol. 6, 622–634.

Hatta, K., Takeichi, M., 1986. Expression of N-cadherin adhesion molecules associatedwith early morphogenetic events in chick development. Nature 320, 447–449.

Heitzler, P., Bourois, M., Ruel, L., Carteret, C., Simpson, P., 1996. Genes of the Enhancer ofsplit and acaete–scute complexes are required for a regulatory loop between Notchand Delta during lateral signaling in Drosophila. Development 122, 161–171.

Horne-Badovinac, S., Bilder, D., 2005. Mass transit: epithelial morphogenesis in theDrosophila egg chamber. Dev. Dyn. 232, 559–574.

Hummel, T., Krukkert, K., Roos, J., Davis, G., Klambt, C., 2000. Drosophila Futsch/22C10is a MAP1B-like protein required for dendritic and axonal development. Neuron 26,357–370.

Ito, K., Awano, W., Suzuki, K., Hiromi, Y., Yamamoto, D., 1997. The Drosophilamushroom body is a quadruple structure of clonal units each of which contains avirtually identical set of neurones and glial cells. Development 124, 761–771.

Jack, J., Delotto, Y., 1995. Structure and regulation of a complex locus: the cut gene ofDrosophila. Genetics 139, 1689–1700.

Jackson, S.M., Blochlinger, K., 1997. cut interacts with Notch and Protein kinase A toregulate egg chmber formation and to maintain germline cyst integrity duringDrosophila oogenesis. Development 124, 3663–3672.

Johnson, T.K., Judd, B.H., 1979. Analysis of the Cut Locus of DROSOPHILAMELANOGASTER.Genetics 92, 485–502.

Knust, E., 2002. Regulation of epithelial cell shape and polarity by cell–cell adhesion(Review). Mol. Membr. Biol. 19, 113–120.

Knust, E., Bossinger, O., 2002. Composition and formation of intercellular junctions inepithelial cells. Science 298, 1955–1959.

Lee, T., Luo, L., 2001. Mosaic analysis with a repressible cell marker (MARCM) forDrosophila neural development.[erratum appears in Trends Neurosci 2001 Jul;24(7):385]. Trends Neurosci. 24, 251–254.

Levine, B., Jean-Francois, M., Bernardi, F., Gargiulo, G., Dobens, L., 2007. Notch signalinglinks interactions between the C/EBP homolog slow border cells and the GILZhomolog bunched during cell migration. Dev. Biol. 305, 217–231.

Liu, S., Jack, J., 1992. Regulatory interactions and role in cell type specification of theMalpighian tubules by the cut, Kruppel, and caudal genes of Drosophila. Dev. Biol.150, 133–143.

Locascio, A., Nieto, M.A., 2001. Cell movements during vertebrate development:integrated tissue behaviour versus individual cell migration. Curr. Opin. Genet. Dev.11, 464–469.

Maeda, M., Johnson, K.R., Wheelock, M.J., 2005. Cadherin switching: essential forbehavioral but not morphological changes during an epithelium-to-mesenchymetransition. J. Cell Sci. 118, 873–887.

Michelli, C.A., Rulifson, E.J., Blair, S., 1997. The function and regulation of cut expressionon the wing margin of Drosophil: Notch, wingless and a dominant negative role forDelta and Serrate. Development 124, 1485–1495.

Michl, P., Downward, J., 2006. CUTL1: a key mediator of TGFbeta-induced tumorinvasion. Cell Cycle 5, 132–134.

Michl, P., Ramjaun, A.R., Pardo, O.E., Warne, P.H., Wagner, M., Poulsom, R., D'Arrigo, C.,Ryder, K., Menke, A., Gress, T., Downward, J., 2005. CUTL1 is a target of TGF(beta)signaling that enhances cancer cell motility and invasiveness. Cancer Cell. 7,521–532.

Montell, D.J., 2003. Border-cell migration: the race is on. Nat. Rev. Mol. Cell Biol. 4,13–24.

Montell, D.J., Rorth, P., Spradling, A.C., 1992. slow border cells, a locus required for adevelopmentally regulated cell migration during oogenesis, encodes Drosophila C/EBP.Cell 71, 51–62.

Nepveu, A., 2001. Role of themultifunctional CDP/Cut/Cux homeodomain transcriptionfactor in regulating differentiation, cell growth and development. Gene 270, 1–15.

Niessen, C.M., Gottardi, C.J., 2008. Molecular components of the adherens junction.Biochim. Biophys. Acta 1778, 562–571.

Niewiadomska, P., Godt, D., Tepass, U., 1999. DE-Cadherin is required for intercellularmotility during Drosophila oogenesis. J. Cell Biol. 144, 533–547.

Oda, H., Uemura, T., Harada, Y., Iwai, Y., Takeichi, M., 1994. A Drosophila homolog ofcadherin associated with armadillo and essential for embryonic cell–cell adhesion.Dev. Biol. 165, 716–726.

Oda, H., Tsukita, S., Takeichi, M., 1998. Dynamic behavior of the cadherin-based cell–celladhesion system during Drosophila gastrulation. Dev. Biol. 203, 435–450.

Pacquelet, A., Rorth, P., 2005. Regulatory mechanisms required for DE-cadherinfunction in cell migration and other types of adhesion. J. Cell Biol. 170, 803–812.

Parnas, D., Haghighi, A.P., Fetter, R.D., Kim, S.W., Goodman, C.S., 2001. Regulation ofpostsynaptic structure and protein localization by the Rho-type guanine nucleotideexchange factor dPix. Neuron 32, 415–424.

Peinado, H., Portillo, F., Cano, A., 2004. Transcriptional regulation of cadherins duringdevelopment and carcinogenesis. Int. J. Dev. Biol. 48, 365–375.

Peinado, H., Olmeda, D., Cano, A., 2007. Snail, Zeb and bHLH factors in tumour progression:an alliance against the epithelial phenotype? Nat. Rev. Cancer 7, 415–428.

Pignoni, F., Zipursky, S.L., 1997. Induction of Drosophila eye development byDecapentaplegic. Development 124, 271–278.

Rebay, I., Fehon, R.G., Artavanis-Tsakonas, S., 1993. Specific truncations of DrosophilaNotch define dominant activated and dominant negative forms of the receptor. Cell74, 319–329.

Riggleman, B., Schedl, P., Wieschaus, E., 1990. Spatial expression of the Drosophilasegment polarity gene armadillo is posttranscriptionally regulated bywingless. Cell63, 549–560.

Rorth, P., 1994. Specification of C/EBP function during Drosophila development by thebZIP basic region. Science 266, 1878–1881.

Rorth, P., 1996. A modular misexpression screen in Drosophila detecting tissue-specificphenotypes. Proc. Natl. Acad. Sci. U. S. A. 93, 12418–12422.

Rorth, P., Montell, D.J., 1992. Drosophila C/EBP: a tissue-specific DNA-binding proteinrequired for embryonic development. Genes Dev. 6, 2299–2311.

Sandell, L.L., Trainor, P.A., 2006. Neural crest cell plasticity. size matters. Adv. Exp. Med.Biol. 589, 78–95.

Sansregret, L., Nepveu, A., 2008. The multiple roles of CUX1: insights from mousemodels and cell-based assays. Gene 412, 84–94.

Silver, D.L., Geisbrecht, E.R., Montell, D.J., 2005. Requirement for JAK/STAT signalingthroughout border cell migration in Drosophila. Development 132, 3483–3492.

Sun, J., Deng, W.M., 2005. Notch-dependent downregulation of the homeodomain genecut is required for the mitotic cycle/endocycle switch and cell differentiation inDrosophila follicle cells. Development 132, 4299–4308.

Szafranski, P., Goode, S., 2004. A Fasciclin 2 morphogenetic switch organizes epithelialcell cluster polarity and motility. Development 131, 2023–2036.

Szafranski, P., Goode, S., 2007. Basolateral junctions are sufficient to suppress epithelialinvasion during Drosophila oogenesis. Dev. Dyn. 236, 364–373.

Takeichi, M., 1988. The cadherins: cell–cell adhesion molecules controlling animalmorphogenesis. Development 102, 639–655.

Takeichi, M., Nakagawa, S., Aono, S., Usui, T., Uemura, T., 2000. Patterning of cellassemblies regulated by adhesion receptors of the cadherin superfamily. Philos.Trans. R. Soc. Lond. B Biol. Sci. 355, 885–890.

Tanentzapf, G., Smith, C., Mcglade, J., Tepass, U., 2000. Apical, lateral, and basalpolarization cues contribute to the development of the follicular epithelium duringDrosophila oogenesis. J. Cell Biol. 151, 891–904.

Page 14: Opposing interactions between Drosophila Cut and the C/EBP encoded by Slow Border Cells direct apical constriction and epithelial invagination

209B. Levine et al. / Developmental Biology 344 (2010) 196–209

Tepass, U., 2002. Adherens junctions: new insight into assembly, modulation andfunction. Bioessays 24, 690–695.

Tepass, U., Harris, K.P., 2007. Adherens junctions in Drosophila retinal morphogenesis.Trends Cell Biol. 17, 26–35.

Tepass, U., Gruszynski-Defeo, E., Haag, T.A., Omatyar, L., Török, T., Hartenstein, V., 1996.shotgun encodes Drosophila E-cadherin and is preferentially required during cellrearrangement in the neurectoderm and other morphogenetically active epithelia.Genes Dev. 10, 659–671.

Tepass, U., Truong, K., Godt, D., Ikura, M., Peifer, M., 2000. Cadherins in embryonic andneural morphogenesis. Nat. Rev. Mol. Cell Biol. 1, 91–100.

Treisman, J.E., Lai, Z.-C., Rubin, G.M., 1995. shortsighted acts in the decapentaplegicpathway in Drosophila eye development and has homology to a mouse TGF-b-responsive gene. Development 121, 2835–2845.

Wang, X., BO, J., Bridges, T., Dugan, K.D., Pan, T.C., Chodosh, L.A., Montell, D.J., 2006.Analysis of cell migration using whole-genome expression profiling of migratorycells in the Drosophila ovary. Dev. Cell 10, 483–495.

Wu, X., Tanwar, P.S., Raftery, L.A., 2008. Drosophila follicle cells: morphogenesis in aneggshell. Semin. Cell Dev. Biol. 19, 271–282.

Xi, R., Mcgregor, J.R., Harrison, D.A., 2003. A gradient of JAK pathway activity patternsthe anterior–posterior axis of the follicular epithelium. Dev. Cell 4, 167–177.

Xu, T., Rubin, G.M., 1993. Analysis of genetic mosaics in developing and adult Drosophilatissues. Development 117, 1223–1237.

Xu, T., Caron, L.A., Fehon, R.G., Artavanis-Tsakonis, S., 1992. The involvement of theNotch locus in Drosophila oogenesis. Development 115, 913–922.

Yagi, T., Takeichi, M., 2000. Cadherin superfamily genes: functions, genomicorganization, and neurologic diversity. Genes Dev. 14, 1169–1180.