the kinase domain of drosophila tribbles is required for ... · the kinase domain of drosophila...

12
The kinase domain of Drosophila Tribbles is required for turnover of fly C/EBP during cell migration Venessa Masoner a , Rahul Das a , Laramie Pence a , Gaurav Anand a , Holly LaFerriere b,1 , Troy Zars b , Samuel Bouyain a , Leonard L. Dobens a,n a Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA b Division of Biological Sciences, University of Missouri-Columbia, Columbia, MO 65211, USA article info Article history: Received 21 November 2012 Received in revised form 22 December 2012 Accepted 28 December 2012 Available online 7 January 2013 Keywords: Drosophila oogenesis Trib protein family abstract Drosophila Tribbles (Trbl) encodes the founding member of the Trib family of kinase-like proteins that regulate cell migration, proliferation, growth and homeostasis. Trbl was identified in a misexpression screen in the ovary as an antagonist of border cell migration and acts in part by directing turnover of the C/EBP protein encoded by the gene slow border cells (slbo). The ability of mammalian Trib isoforms to promote C/EBP turnover during tissue differentiation indicates that this function is highly conserved. To better understand the role of Trbl in cell migration, we tested specific Trbl antisera, a trbl null allele and Trbl transgenes bearing site-directed mutations. Trbl is expressed at high levels in the nuclei of follicle cell epithelia and is downregulated in delaminating epithelia as expression of Slbo (C/EBP) is upregulated. This complementary pattern of expression during subsequent cell migration is achieved by negative feedback whereby slbo represses Trbl expression and trbl is necessary and sufficient to promote Slbo protein turnover. A series of point mutations that scan the conserved kinase domain of Trbl reveal that the conserved DLK catalytic loop is required for Trbl–Slbo binding and turnover, as well as for interactions between Trbl subunits, suggesting a mechanism of Trbl function. & 2013 Elsevier Inc. All rights reserved. Introduction Cell migration is a tightly regulated process, in which loss of polarity and delamination of epithelial cells is followed by changes in cell adhesion and the formation and extension of cellular processes during a migratory phase. During normal development as well as abnormal tumor progression, the levels of key regulatory proteins that trigger, direct and terminate cell migration are strictly controlled. The Drosophila ovary presents two excellent model tissues to study collective cell migration: (1) the border cells (BC), which delaminate as a small cluster and migrate through the nurse cells to reach the posterior oocyte from stages 9-10B and (2) the centripetal follicle cells (centripetal migrating FC or CMFC) which migrate as a cell sheet between the nurse cells and oocyte from stages 10B-13 (Fig. 1A). A key regulator of migration in both these tissues is the C/EBP protein Slow border cells (Slbo). Levels of Slbo are critical for proper migration, and negative feedback regulates Slbo expression both transcriptionally (Levine et al., 2010) and post-transcriptionally. A search for genes that regulate Slbo protein levels during BC migration identified the gene tribbles (trbl; Rorth et al., 2000), which encodes a member of the Trib protein family, which include mammalian isoforms Trib1, Trib2 and Trib3. Trib family members share three conserved motifs (reviewed in Angyal and Kiss-Toth, 2012; Dobens and Bouyain, 2012; Kiss- Toth, 2011; Yokoyama and Nakamura, 2011). First, members have a central kinase-like domain that retains a conserved DLK cata- lytic loop but lacks other conserved motifs critical for kinase activity. Consequently, it remains unclear whether Tribs are pseudokinases or act to phosphorylate substrates via a novel mechanism. The presence of conserved binding sites for Mitogen- activated protein kinase or extracellular signal-regulated kinase kinase (MEK1) and constitutive photomorphogenesis protein 1 (COP1) in the C-terminal region has led to competing ideas that Tribs act either as docking kinases (Remenyi et al., 2006) or as ‘molecular scaffolds,’ pseudokinases that bind multiple regulatory proteins and balance the activities of several signaling pathways simultaneously (Kiss-Toth et al., 2006). In support of both these ideas, work done in vertebrates has connected Tribs to regulation of the TGFb/BMP, ras/MAPK and insulin/Akt signaling pathways (Hegedus et al., 2006, 2007; Chan et al., 2007; Hua et al., 2011). During mammalian and fly development, Tribs regulate cell proliferation during tissue differentiation (reviewed in Dobens Contents lists available at SciVerse ScienceDirect journal homepage: www.elsevier.com/locate/developmentalbiology Developmental Biology 0012-1606/$ - see front matter & 2013 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ydbio.2012.12.016 n Corresponding author. Fax: þ1 816 523 5995. E-mail address: [email protected] (L.L. Dobens). 1 Current address: Department of Biology, Culver-Stockton College, Canton, MO 63435, USA. Developmental Biology 375 (2013) 33–44

Upload: phungkhue

Post on 02-Apr-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

Developmental Biology 375 (2013) 33–44

Contents lists available at SciVerse ScienceDirect

Developmental Biology

0012-16

http://d

n Corr

E-m1 Cu

MO 634

journal homepage: www.elsevier.com/locate/developmentalbiology

The kinase domain of Drosophila Tribbles is required for turnoverof fly C/EBP during cell migration

Venessa Masoner a, Rahul Das a, Laramie Pence a, Gaurav Anand a, Holly LaFerriere b,1, Troy Zars b,Samuel Bouyain a, Leonard L. Dobens a,n

a Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USAb Division of Biological Sciences, University of Missouri-Columbia, Columbia, MO 65211, USA

a r t i c l e i n f o

Article history:

Received 21 November 2012

Received in revised form

22 December 2012

Accepted 28 December 2012Available online 7 January 2013

Keywords:

Drosophila oogenesis

Trib protein family

06/$ - see front matter & 2013 Elsevier Inc. A

x.doi.org/10.1016/j.ydbio.2012.12.016

esponding author. Fax: þ1 816 523 5995.

ail address: [email protected] (L.L. Dobens).

rrent address: Department of Biology, Culv

35, USA.

a b s t r a c t

Drosophila Tribbles (Trbl) encodes the founding member of the Trib family of kinase-like proteins that

regulate cell migration, proliferation, growth and homeostasis. Trbl was identified in a misexpression

screen in the ovary as an antagonist of border cell migration and acts in part by directing turnover of

the C/EBP protein encoded by the gene slow border cells (slbo). The ability of mammalian Trib isoforms

to promote C/EBP turnover during tissue differentiation indicates that this function is highly conserved.

To better understand the role of Trbl in cell migration, we tested specific Trbl antisera, a trbl null allele

and Trbl transgenes bearing site-directed mutations. Trbl is expressed at high levels in the nuclei of

follicle cell epithelia and is downregulated in delaminating epithelia as expression of Slbo (C/EBP) is

upregulated. This complementary pattern of expression during subsequent cell migration is achieved

by negative feedback whereby slbo represses Trbl expression and trbl is necessary and sufficient to

promote Slbo protein turnover. A series of point mutations that scan the conserved kinase domain of

Trbl reveal that the conserved DLK catalytic loop is required for Trbl–Slbo binding and turnover, as well

as for interactions between Trbl subunits, suggesting a mechanism of Trbl function.

& 2013 Elsevier Inc. All rights reserved.

Introduction

Cell migration is a tightly regulated process, in which loss ofpolarity and delamination of epithelial cells is followed bychanges in cell adhesion and the formation and extension ofcellular processes during a migratory phase. During normaldevelopment as well as abnormal tumor progression, the levelsof key regulatory proteins that trigger, direct and terminate cellmigration are strictly controlled. The Drosophila ovary presentstwo excellent model tissues to study collective cell migration:(1) the border cells (BC), which delaminate as a small cluster andmigrate through the nurse cells to reach the posterior oocyte fromstages 9-10B and (2) the centripetal follicle cells (centripetalmigrating FC or CMFC) which migrate as a cell sheet betweenthe nurse cells and oocyte from stages 10B-13 (Fig. 1A). A keyregulator of migration in both these tissues is the C/EBP proteinSlow border cells (Slbo). Levels of Slbo are critical for propermigration, and negative feedback regulates Slbo expression bothtranscriptionally (Levine et al., 2010) and post-transcriptionally.

ll rights reserved.

er-Stockton College, Canton,

A search for genes that regulate Slbo protein levels during BCmigration identified the gene tribbles (trbl; Rorth et al., 2000),which encodes a member of the Trib protein family, whichinclude mammalian isoforms Trib1, Trib2 and Trib3.

Trib family members share three conserved motifs (reviewedin Angyal and Kiss-Toth, 2012; Dobens and Bouyain, 2012; Kiss-Toth, 2011; Yokoyama and Nakamura, 2011). First, members havea central kinase-like domain that retains a conserved DLK cata-lytic loop but lacks other conserved motifs critical for kinaseactivity. Consequently, it remains unclear whether Tribs arepseudokinases or act to phosphorylate substrates via a novelmechanism. The presence of conserved binding sites for Mitogen-activated protein kinase or extracellular signal-regulated kinasekinase (MEK1) and constitutive photomorphogenesis protein 1(COP1) in the C-terminal region has led to competing ideas thatTribs act either as docking kinases (Remenyi et al., 2006) or as‘molecular scaffolds,’ pseudokinases that bind multiple regulatoryproteins and balance the activities of several signaling pathwayssimultaneously (Kiss-Toth et al., 2006). In support of both theseideas, work done in vertebrates has connected Tribs to regulationof the TGFb/BMP, ras/MAPK and insulin/Akt signaling pathways(Hegedus et al., 2006, 2007; Chan et al., 2007; Hua et al., 2011).

During mammalian and fly development, Tribs regulate cellproliferation during tissue differentiation (reviewed in Dobens

Fig. 1. Trbl accumulates in the nuclei of non-migratory FC groups (A). Outline of migratory cell types during oogenesis. At stage 9, posterior pole FC (PP) delaminate to

form the border cell cluster (BC) which migrate through the nurse cells (NC) to reach the oocyte (O) at stage 10, when the centripetal FC (CMFC) migrate between the NC/O

boundary. (B). Trbl-GAL4 enhancer trap (trblP{GawB}NP4027) expresses UAS-GFP (green) in the main body FC with low levels in the centripetal FC (empty arrow) that express

Slbo (red). (C). At stage 10B, Trbl accumulates at high levels in the nuclei of the main body FC. Lower levels of Trbl occur in the border cells (black arrow) at stage 10B. Inset

contrasts high Slbo protein levels in the border cells (white arrow, right) with low levels of Trbl (left, empty arrow). (D). Low levels of Trbl (red) accumulate in the

centripetal FC that express Slbo (green). DAPI reveals the location of all nuclei in the cell sheet. (E) At stage 10, Trbl levels are high and nuclear. (F) At stage 12, Trbl levels

are low and non-nuclear.

V. Masoner et al. / Developmental Biology 375 (2013) 33–4434

and Bouyain, 2012). In flies, Trbl blocks (1) embryonic cell divisionduring mesodermal migration (Grosshans and Wieschaus, 2000;Seher and Leptin, 2000), (2) the step-wise cell division connected topatterning of the bristle primordia in the peripheral neurons(Abdelilah-Seyfried et al., 2000; Norga et al., 2003; Fichelson andGho, 2004) and (3) stem cell proliferation during germ linedifferentiation (Mata et al., 2000; Schulz et al., 2004). MammalianTrib isoforms regulate differentiation linked to cell division duringhematopoesis (Lin et al., 2007; Eder et al., 2008; Sathyanarayanaet al., 2008), myogenesis (Kato and Du, 2007; Sung et al., 2007),

lymphogenesis (Selim et al., 2007) and adipogenesis (Naiki et al.,2007). The connections observed between Tribs and diverse cellsignaling pathways regulating cell growth, proliferation and differ-entiation likely underlie the involvement of Tribs in cancer anddisease (Kiss-Toth, 2011; Yokoyama and Nakamura, 2011; Angyaland Kiss-Toth, 2012; Prudente et al., 2012).

One common mechanistic feature shared by all Tribs is the abilityto bind key regulatory proteins and either block their activity or directtheir turnover by the proteosome. In the mesoderm and germ line ofthe fly, Trbl degrades String/cdc25 phosphatase to regulate the entry

V. Masoner et al. / Developmental Biology 375 (2013) 33–44 35

into S phase (Grosshans and Wieschaus, 2000; Seher and Leptin,2000). In mouse osteoblasts, Trib3 binds and degrades SMURF1, an E3ubiquitin ligase specific for the BMP mediator Smad, effectivelystabilizing Smads (Chan et al., 2007). During adipogenesis, Trib3binds and degrades acetyl coenzyme A carboxylase (ACC), the rate-limiting enzyme in fatty acid synthesis (Qi et al., 2006). As well, Trib3(and Trib2 less effectively) binds to Akt to inhibit phosphorylation ofFoxo1 to permit adipocyte development to proceed (Naiki et al.,2007). During BC migration, Trbl binds to Slbo to direct its degrada-tion by the proteosome, and this interaction is conserved in mam-mals, where Trib1 and Trib2 accelerate degradation of C/EBPa andC/EBPb, respectively, during normal tissue differentiation and intumors (Keeshan et al., 2006, 2008; Naiki et al., 2007; Yamamotoet al., 2007).

Here we show, using a cell migration model in the fly ovary, thatSlbo and Trbl have a complex negative feedback whereby Slborepresses Trbl expression and Trbl – via its conserved DLK motif –directs Slbo turnover. Mechanistically, we show that Trbl binds toitself and more strongly to Slbo, and both interactions require theDLK motif. Our results shed light on the conserved mechanism ofTrbl function, and assign a role for Trbl interactions in a negativefeedback loop regulating C/EBP-regulated cell migration.

Materials and methods

Drosophila strains

Stocks used were (1) P{wþ(mC)¼AyGAL4}25 P{wþ(mC)¼UAS-

GFP.S65T}T2/CyO (Pignoni and Zipursky, 1997), (2) slbo01310/CyO

(Montell et al., 1992), (3) P}1206 P} T2/CyO (Rorth et al., 2000), (4)P{XP}trbld07751, (5) P{XP}trbld03251 (4 and 5 are from the Harvard StockCenter; Thibault et al., 2004), (6) hsFLP1 (Xu and Rubin, 1993), (7)UAS-Trbl, (8) trblEP1119, (9) trblEP3519, (10) hsSlbo (7–9 are gifts fromPernille Rorth (Rorth et al., 2000)), (12) w1118; {Ubi-GFP(S65T)nls}3L

P{neoFRT}80B/TM3 (Xu and Rubin, 1993), (12) trblP{GawB}NP4027, (13)CG33969EP20583, (14) fbl1, (15) Df(3L)rdgC-co2, (16) y(1) w(1);P{wþ(mW.hs)¼en2.4-GAL4}e22c/SM5 (10–15 are from the Blooming-ton Stock Center), (17) UAS-Trbl IR22114 and (18) UAS-Trbl IR22113 (17and 18 are from the Vienna Stock Center).

Generation of clones and immunochemistry

Flp-out clones misexpressing UAS-transgenes were created asdescribed previously (Levine et al., 2010). The production of mitoticclones was performed as before (Dobens et al., 2000).

Immunohistochemistry procedures have been described (Levineet al., 2010) and the following primary antisera were used: (1) poly-clonal antisera were raised against the peptide CZDKHEYEDIGVEPL-DYTR in chickens and affinity purified with the peptide (Aves Lab,Tigard, OR) and was used at 1:1000 (in PBSþ1% BSAþ0.5% Triton-X100 solution after preadsorption to ovarian tissue); (2) rabbit anti-b-galactosidase (1:50,000 following preadsorption to ovarian tissue;Cappell, West Chester, PA); (3) rabbit anti-Slbo at 1:2000 (Szafranskiand Goode, 2004); (4) monoclonal mouse and rabbit GFP (Invitro-gen) were used at 1:200 and 1:1000, respectively; and (5) mousemonoclonal anti-Flag (M2, Sigma) was used at 1:200. Secondaryantibodies used were goat anti-rabbit, mouse and chicken Alexa-Fluors488, 594 and 633 (1:200; Invitrogen). Confocal images werecollected on an Olympus Fluoview 300 confocal microscope orNikon 90i microscope with Optigrid paddle and Metamorph Imageacquisition software. DIC images were collected on a Nikon TE-2000with attached Colorview camera and Analysis image acquisitionsoftware. Figures were prepared using Photoshop CS.

Fluorescence intensity (measured in arbitrary units) wasobtained by collecting confocal sections through the BC cluster

taken under identical settings and the section where slbo expres-sion is maximum was selected using ‘plot Z-axis profile’ function ofthe ImageJ software (http://rsbweb.nih.gov/ij/). The correspondingTrbl fluorescence intensity in this section was measured by the‘measure’ function of ImageJ and the background intensity (whichis the average of the maximum intensity of three separate emptyspots within that specific egg chamber) subtracted. Finally, the Trblintensities obtained from multiple egg chambers of a givengenotype were averaged and used for plotting. The graphs andstatistical analysis were done using GraphPad Prism software.

Design and construction of Trbl mutants

Construction of FLAGTRBLWT

The complete ORF of trbl was amplified from cDNA using theoligonucleotides ATGGATTACAAGGATGACGACGATAAGATGGATA-ACAGTAGCGGTC and TCAGCCCATGTCCACATCCGTATCGGGTTC togenerate a N-terminal FLAG fusion (FLAG sequence in bold),cloned into pSTBlue-1 (AccepTor Vector kit, Novagen) and con-firmed by DNA sequencing. An EcoRI fragment containing the full-length FLAGTRBL was then cloned into pUASTattB and againconfirmed by DNA sequencing.

Construction of UAS-FLAGTRBLD/NLK, UAS-FLAGTRBLFLCR/A, and

UAS-FLAGTRBLSLE/G

Mutated versions of Trbl were generated from pSTBlue-1þFLAGTRBL using the QuikChange II XL Site-Directed MutagenesisKit (Stratagene) and oligonucleotides were designed as describedtherein (FLAGTRBLD/NLK: CGGGATTATCCTCAGGAACCTCAAGCTCA-AGCG and CGCTTGAGCTTGAGGTTCCTGAGGATAATCCCG; UAS-FLAGTRBLFLCR/A: GGGAGCAGTTCCTCTGCGCTATTGTAAACGAACCG-TTG and CAAGCGTTCGTTTACAATAGCGCAGAGGAACTGCTCCC; UAS-FLAGTRBLSLE/G: GCAGTATGAATCACTGGGAGGCTCAATGATCCTCG andCGAGGATCATTGAGCCTCCCAGTGATTCATACTGC). Site-directed muta-tions (shown in bold) were confirmed by DNA sequencing.FLAGTRBLSLE/G was constructed slightly differently by adding flankingattB1 and attB2 sites (using the oligonucleotides GGGGACAAGTTTG-TACAAAAAAGCAGGCTTCATGGATTACAAGGATGACGACGATAAG andGGGGACCACTTTGTACAAGAAAGCTGGGTCTCAGCCCATGTCCA-CATCCGTATC; attB1 and attB2 sequences in bold), using recombina-tion to insert the fragment into the GATEWAY donor vector pDONR-21 and then into the destination vector pUASgattB (Bischof andBasler, 2008).

Embryo injections were performed as a fee-for-service(Genetic Services, Inc., Cambridge, MA) and transgenic linesestablished were confirmed for the presence of the WT or mutanttransgene by sequencing of PCR product.

Yeast two-Hybrid interaction screen

The ProQuest Two-Hybrid System (Invitrogen) was used toperform yeast two-hybrid interaction tests. Plasmid DNA trans-formations of Saccharomyces cerevisiae strain MaV203 were per-formed as outlined in the kit manual and grown on SC medialacking both tryptophan and leucine to confirm the presence ofboth the bait and prey plasmids. The cells were then transferredto a series of plates to test for interactions: SC-Leu-Trp-His,SC-Leu-Trp-Hisþ3AT (at various concentrations), and SC-Leu-Trp-HisþXGAL. To confirm presence of protein expression, yeastextracts were prepared with Laemmli buffer and detected viaWestern blot using Tribbles and Slbo antibodies (data not shown).A quantitative b-galactosidase assay was performed on transfor-mants grown in SC-Leu-Trp broth using CPRG as a substrate.

Fig. 2. slbo and trbl have opposing effects in migratory FC groups (A). Prior to delamination at stage 8, Trbl (red) accumulates at high levels in the border cells nuclei (arrow,

counterstained with Fas2 in magenta). (B) At stage 9, Trbl levels remain high as border cell migration begins. (C) By stage 10, only low levels of Trbl occur in the border cells at the

nurse cell/oocyte boundary (arrow). (D) In the slbo mutant slbo01310, Trbl levels remain high at stage 10 in the border cells (anterior arrow) and in the centripetal migrating FC

(CMFC). (E) Trbl expression in WT egg chambers, measured as fluorescence intensity (see section ‘‘Materials and methods’’) showing the decrease in Trbl expression as BC migration

progresses from stages 8-10B. Error bars represent mean7standard deviation. The difference between groups is statistically significant (P value for one way ANOVA testo0.0001,

Student’s T-test between each groupo0.05). (F) Trbl expression, measured as fluorescence intensity, in WT (slbo01310/CyO genotype) egg chambers is lower when compared to

mutant (slbo01310/slbo01310) egg chambers both at stage 9 and stage 10A/B. Error bars represent mean7standard deviation. The difference between groups is statistically significant

(P value for one way ANOVA testo0.0001, Student’s T-test value between each groupo0.5, except WT stage 10A and 10B, where the value is 40.05). (G) Misexpression of UAS-Trbl

using the Flp-out actin driver can be detected in FC clones located in the centripetal FC (arrow) by GFP expression (green, arrow center panel) and have no effect on levels of slbo-lacZ

(red). Genotype: hsFLP1; slbo01310/ P{wþ(mC)¼AyGAL4}25 P{wþ(mC)¼UAS-GFP.S65T}T2; UAS-Trbl. Failure to repress slbo-lacZ was observed in 10/10 clones located in the

centripetal FC.

V. Masoner et al. / Developmental Biology 375 (2013) 33–4436

V. Masoner et al. / Developmental Biology 375 (2013) 33–44 37

To construct bait and prey plasmids FLAGTRBL, FLAGTRBLD/NLK,and SLBO cDNAs were amplified using PCR to add flanking attB1and attB2 sites. Forward and reverse primers for FLAGTRBL andFLAGTRBLD/NLK were, respectively: 50–GGGGACAAGTTTGTACAAAAAAGCAGGCTTCATGGATTACAAGGATGACGACGATAAG–30, 50–GGGGACCACTTTGTACAAGAAAGCTGGGTCTCAGCCCATGTCCACATCCGTATC–30; and for SLBO, respectively, 50–GGGGACAAGTTTGTACAAAAAAGCAGGCTTCATGCTGAACATGGAGTCGCCGCAG–30, 50–GGG-GACCACTTTGTACAAGAAAGCTGGGTCCTACAGCGAGTGTTCGTTGGTG-TTG–30 (attB1 and attB2 sequences in bold). The PCR product wascloned into the donor vector pDONR-21 and then into eitherpDEST32 (bait vector) or pDEST22 (prey vector). All constructs wereconfirmed by DNA sequencing.

Construction of pDEST32 UAS-FLAGTRBLCOP-1

pDEST32 UAS-FLAGTRBLCOP-1 mutant was generated from pre-viously constructed pDEST32 UAS-FLAGTRBL using the QuikChangeKit and the following oligonucleotides: FLAGTRBLCOP-1: 50–GATG-GATGATGACGAGGCAGGACTCTGTCCCTTGG–30 and 50–CCAAGGGA-CAGAGTCCTGCCTCGTCATCATCCATC–30 (mutated nucleotides inbold). Site-directed mutations were confirmed by DNA sequencing.

Fig. 3. Construction of a trbl null allele: (A) map of the trbl gene region (after McQuilto

(genes at top and respective transcripts below, with introns indicated as thin lines and

inverted triangles (CG33969EP20583, trblEP119, trblEP3519). Below these, the insertion site

transposon contains the mini-white gene (represented by red half triangle) and the

chromosome 3L) and includes both mini-white genes (conceptually, a fusion of the two

reduction in nuclear staining of Trbl using the Trbl antisera (red and black and white,

localization (right panel and R. Das, unpublished data). White arrows indicate the locatio

nuclear Trbl (left panel); adjacent mutant cells (filled arrows) show (1) low levels of Trb

of Dlg (yellow, right panel). (C) Scheme for examining Slbo turnover in trbl mutant ce

P(Ubi-GFP(S65T)nls)3L P(neoFRT)80B/ trblD13 P(neoFRT)80B. After FLP induction, trblD13

induction of hs-Slbo, high levels of nuclear Slbo can be detected throughout the FC epith

cells (black) can be compared to WT cells (green) in panel 4. (D) In stage 10 egg cha

induction. This was at 3 h after heat shock recovery in 28/32 clones detected in the m

Results

Trbl is a nuclear protein expressed in non-migratory follicle cells

whose expression is repressed by Slbo (C/EBP)

To examine the endogenous expression of Trbl in the eggchamber, we took two approaches. First, we evaluated availabletrbl enhancer trap reporter genes and found one (trblP{GawB}NP4027)that expressed in the FC with high levels in the main body FC andat low levels in the centripetal FC (CMFC) and border cells (BC),two cell types that express high levels of Slbo protein (Fig. 1B). Toexamine directly Trbl protein levels in this tissue, we raisedantisera to Trbl protein (see section ‘‘Materials and methods’’)and used it to detect a protein that migrates at 65 kDa (supple-mental Fig. S1B) with widespread nuclear expression in thesomatic follicle cells and germ line nurse cells from stages 2 to10 of oogenesis (Fig. 1C–F). Counterstaining for a slbo-lacZ

reporter gene (slbo01310) revealed that, like expression oftrblP{GawB}NP4027, Trbl protein levels are low in both the bordercells and in the leading edge CMFC (Fig. 1C and D). At later stages

n and Pierre, 2012) showing the location of Trbl (blue) and adjacent gene CG33969

coding regions as orange boxes). Insertion sites of alleles used are shown as black,

s of FRT-bearing transposons P{XP}trbld07751 and P{XP}trbld03251 are shown; each

hybrid element (trblD13) deletes the intervening region (20888086-20394680 of

red half triangles). (B) Mutant clones of trblD13 (absence of GFP, center panel) show

left panel). As well, these clones have uncharacterized defects in cell size and Dlg

n of two wild type cells positive for GFP (center panel) that express strong levels of

l (left panel), (2) nuclear size defects (DAPI, right panel) and (3) reduced expression

lls. Panel 1 shows the FC epithelium in a female of the genotype hsFLP1; hs-Slbo;

clones can be detected by the absence of GFP, in panel 2. After 30 min heat shock

elium (red) in panel 3. Three hours after recovery, Slbo turnover in trblD13 mutant

mbers, trblD13 mutant cells (no GFP) show higher levels of Slbo, 3 h after hs-Slbo

ain body FC.

V. Masoner et al. / Developmental Biology 375 (2013) 33–4438

the subcellular accumulation of Trbl in the main body FC isdynamic: we observe strong nuclear accumulation at stage 10(Fig. 1E) that decreases to low levels in the cytoplasm by stage 12(Fig. 1F).

Closer examination revealed that Trbl expression is high at thebeginning of BC delamination (respectively stage 7, Fig. 2A andstage 8, Fig. 2B) but subsequently Trbl levels decrease throughoutBC migration so that levels are low when the border cells arrive atthe nurse cell/oocyte boundary (stage 10B, Fig. 2C and E). Based onthe complementary expression of Trbl and Slbo during BC andCMFC migration, we tested the interaction between these genes inthese two tissues in several ways. First we examined the effect ofslbo mutations and Slbo misexpression on Trbl levels, measuredwith Trbl antisera. In the slbo mutant background (slbo01310/slbo01310), Trbl expression increased in the border cells and cen-tripetal FC (Fig. 2D) both at stages 9 and 10B (Fig. 2F), indicatingthat slbo is required for Trbl repression throughout BC migration. Incontrast, Flp-out misexpression of Slbo has no effect on Trblexpression in the main body FC (supplemental Fig. S2A and B). Ithas been shown that mouse Trbl acts as a transcriptional cofactor torepress transcription (e.g. Ohoka et al., 2005; Takahashi et al., 2008),so we sought to test if Trbl misexpression could repress slbo-lacZ, anin vivo reporter of slbo transcription. As shown in Fig. 2G, Flp-outactin-GAL4 misexpression of Trbl in the centripetal FC was insuffi-cient to reduce slbo reporter gene expression. Complementarily,examination of trbl null mutant clones revealed no change in slbo-

lacZ reporter gene expression (data not shown). Thus Slbo isnecessary but not sufficient to repress Trbl expression in the eggchamber while Trbl is neither necessary nor sufficient to repressslbo transcription.

Table 1Complementation analysis of trbl deletion allele.

% expected

trblD13 trblD3 trblEP1119 trblEP3519

trblD13 0% (n¼4300)

trblD3 0%

(n¼4300)

trblEP1119 49% (n¼264) 41% (n¼111)

trblEP3519 23% (n¼130) 22.5%

(n¼148)

Df(3L)rdgC-co2 10% (n¼156) 16% (n¼107)

CG33969EP20583 113% (n¼96) 108% (n¼134) 96.2% (n¼117) 137% (n¼54)

Table 2Genetic interactions between trbl and slbo during BC migration.

% migration

No

Slbo2.6GAL4;UAS-Trbl IR22114 5%

Slbo2.6GAL4;UAS-Trbl IR22113 0

trblEP1119/trblEP1119 0

trblEP1119/trblD13 0

trblEP3519/trblD13 Reduced ovaries

slbo10301/slbo10310; trblEP1119/þ 58.3%

slbo10301/slbo10310; trblEP1119/trblEP1119 17.7

trbl is necessary to direct Slbo turnover

In flies and mouse cell lines, Trib family members are necessaryto direct turnover of Slbo protein (Rorth et al., 2000; Keeshan et al.,2010). Because existing trbl alleles are not protein null and mayretain Trbl activity, we sought to create a deletion in trbl codingsequence. To accomplish this, we used a regimen of hsFLP inductionto sponsor FRT-mediated recombination in males transheterozy-gous for two FRT-bearing P-elements, P{XP}trbld07751 andP{XP}trbld03251 inserted at sites flanking the trbl gene and markedby a single mini-white marker (Fig. 3A). Recombinants that deletedthe trbl gene could be detected fortuitously by the presence of twomini-white markers in the hybrid transposon, and two resulting trbl

deletions (designated trblD13 and trblD3) were confirmed by PCR toremove sequences (from proximal to distal, respectively) 498,698base pairs (bp) downstream from the 30 end of the gene to 28 bpdownstream from the start site of transcription (data not shown).

By several criteria trblD13 and trblD3 are bonafide trbl deletionalleles. First, both trblD13 and trblD3 resulted in reduced number ofadults in combination with weaker trbl alleles trbEP119 and trblEP3519

(order of allelic strength inferred from Table 1 was trblD13¼

trblD34trblEP35194trblEP1119). Second, the trblD13 allele recombinedonto an FRT80 chromosome showed reduced antisera staining inmain body FC clones (Fig. 3B) similar to reduced antisera stainingseen in clones of trblEP1119 (supplemental Fig. S1A). And third, escapertrblD13/trblEP1119 and trblD13/Df(3L)rdgC animals have defects in nursecell number (data not shown) similar to those reported for trbl

mutations (data not shown and Mata et al., 2000). Because trblD13

and trblD3 complement mutations in the proximal genes CG33969

(Table 1) and fbl (data not shown), we conclude that the deletion ofsequences downstream of the designated 30 end of trbl in these allelesdoes not affect adjacent genes.

We used the trblD13FRT80 chromosome to test the requirementfor trbl in Slbo turnover in the main body FC, where Trbl levels arehigh. Main body FC clones of trblD13 had no effect on endogenousSlbo levels (data not shown), so we examined the effect on Slboturnover in a scheme outlined in Fig. 3C. We produced FLP-FRTtrblD13 mutant clones in females bearing a hs-Slbo inducibletransgene (genotype hsFLP; hs-Slbo; UbiGFPFRT80/trblD13 FRT80)and 3 h following recovery from a 30 min heat shock to induceubiquitous Slbo, we used Slbo antisera to compare Slbo turnoverin WT cells to turnover in trblD13 mutant clones (Szafranski andGoode, 2004). As shown in Fig. 3D, Slbo levels are noticeably morestable in main body FC that are mutant for trblD13, indicating thattrbl is necessary for Slbo turnover in this tissue.

Partial Full

42% 53% (n¼38)

2 98 (n¼54)

1.7 98.3 (n¼291)

0 100% (n¼145)

37.4% 4.2% (n¼350)

61.5 20.8 (n¼96)

V. Masoner et al. / Developmental Biology 375 (2013) 33–44 39

In the border cells, Slbo and Trbl have opposing effects on eachother’s expressions, so we examined their genetic interactionsduring the progress of BC migration (Table 2). While strong alleliccombinations (trblEP3519/trblD13) led to rudimentary, undevelopedovaries that lacked late stage egg chambers (Table 2 and data notshown), RNAi knockdown of Trbl activity specifically at late stagesin the border cells (Slbo2.6GAL4; UAS-Trbl IR22114; Table 2) partiallyblocked BC migration. Weaker mutant allelic combinations(trblEP1119/trblEP1119) had no effect on BC migration but did effec-tively suppress defects seen in slbo01310 females (Montell et al.,1992), resulting in an increased frequency of partial and completeBC migration (Table 2). We conclude that opposing activities of trbl

and slbo contribute to the progress of BC migration, which isconsistent with previous work (Rorth et al., 2000).

Trbl kinase domain is required for border cell migration

BC migration is blocked by Trbl misexpression (Rorth et al.,2000), so we used this sensitive migration phenotype to conduct astructure–function analysis of Trbl. We used site-directed muta-genesis to make three mutations in the Trbl kinase domain(Fig. 4A): (1) FLCR/A was designed to disrupt a divergent sequencelocated in a region in conventional kinases (VAIK) predicted tobind ATP; (2) D/NLK was designed to disrupt the conserved DLKmotif in the catalytic domain of the kinase region; and (3) SLE/Gwas designed to disrupt a conserved SLE motif predicted tocoordinate Mg2þ . Each of these mutations was made in a Flag-tagged Trbl transgene under UAS control, inserted into an attPvector (Bischof and Basler, 2008) and injected to produce trans-genic animals with insertions recombined into the second chro-mosome landing site (attP40; 25C7 on 2L) or third chromosome

Fig. 4. Trbl kinase domain is required to block BC migration: (A) map of the Trbl pro

degenerate VAIK motif, a conserved catalytic loop (red) and a putative Mg2þ binding d

site-specific mutations produced (red) in three respective motifs are show. (B–E) Pane

Slbo2.6GAL4,UAS-GFP reveals that WT border cell migration shown at stages 9 and 10 pr

between nurse cells and oocyte by stage 10 (white arrow). (C–E) Slbo2.6GAL44 misexp

BC migration, similar to WT Trbl (not shown). In contrast, misexpression of D/NLK (D) f

compartment (G) leads to cross vein defects (box) and increase in cell size (inset), whe

increase in cell size can be seen by misexpression of the D/NLK mutant in the posterio

landing site (attP2; 68A4 on 3L). In this way, we could comparemisexpression of mutant and WT transgenes while minimizingposition effects.

We tested the effect of misexpression of each transgene in theborder cells using the Slbo2.6GAL4 driver (Fig. 4B), and the resultsare summarized in Fig. 4 and Table 3. Following misexpression ofFlag-tagged UAS-Trbl inserted at either attP2 on the third chro-mosome or attP40 on the second, we observed a significant blockin BC migration (Table 3) that was comparable to the block causedby misexpression of previously described transgenes UAS-Trbl orEP3519, which were tested in parallel (Rorth et al., 2000). For eachtransgene, the block observed was more significant at 30 1C,consistent with the temperature sensitivity of GAL4 activity.

As shown in Fig. 4B–E and summarized in Table 3, misexpres-sion of UAS-FlagTrbl transgenes bearing the FLCR/A (Fig. 4C) orSLE/G mutation (Fig. 4E) resulted in a strong block to BC migra-tion, similar to WT UAS-FlagTrbl (Table 3). Thus these two kinaseregion mutations do not affect Trbl activity. In contrast, misex-pression of the D/NLK mutation either at attP40 (Fig. 4D) or attP2(data not shown) failed to block BC migration compared to WTUAS-FlagTrbl inserted at the respective landing site (Table 3).

While these data indicate that the D/NLK mutation disruptsTrbl activity, we were concerned that D/NLK transgenes mightlack activity due instead to either reduced expression or produc-tion of a mutant protein that does not fold properly. Weconfirmed that D/NLK is expressed in the border cells, as bothTrbl antisera (Fig. 6B) and Flag antisera (data not shown) detectedSlbo2.6GAL4 misexpression. While we cannot rule out the possi-bility of D/NLK misfolding, we note that D/NLK misexpressed inthe posterior wing blade under control of the engrailed-GAL4

driver (en2.4-GAL4) caused an increase in cell size similar to

tein coding region shows the conserved central kinase region (blue) containing a

omain. The Drosophila and mouse sequence is aligned below this and each of three

ls show the effect of misexpressing GFP alone and each mutant Trbl protein. (B)

ogresses from the anterior of the egg chamber at stage 9 to a position at the border

ression of the mutants TrblFLCR/A (C) or TrblSLE/G (E) resulted in a strong block to

ailed to block BC migration. (F–H) Misexpression of WT Trbl in the posterior wing

n compared to the engrailedGAL4 driver alone (F). A similar cross vein defect and

r wing (H), suggests that it retains activity in this assay.

Table 3Misexpression assay of UAS-Trbl, UAS-Trbl mutant transgenes and UAS-Trbl RNAi.

% migration

No Partial Full

Slbo2.6GAL4 0% 0% 100% (n¼110)

Slbo2.6GAL4;UAS-FLAGTrbl (III) 57 28 15 (n¼157)

Slbo2.6GAL4;UAS-FLAGTrblD/NLK (III) 0 4 96 (n¼140)

Slbo2.6GAL4/UAS-FLAGTrbl (II) 24 41 35 (n¼100)

Slbo2.6GAL4/UAS-FLAGTrblD/NLK (II) 0 3 97 (n¼59)

Slbo2.6GAL4/UAS-FLAGTrblFLCR/A (II) 58 12 31 (n¼121)

Slbo2.6GAL4/UAS-FLAGTrblSLE/G (II) 70 16 14 (n¼120)

Slbo2.6GAL4;UAS-Trbl (Rorth) 28 30 42 (n¼71)

Slbo2.6GAL4;UAS-EP3519 78 12 10 (n¼51)

Fig. 5. Trbl kinase domain is required for Slbo binding in vitro (Y2H): (A) Yeast

strains harboring bait/prey combinations are spread on sectors of drop-out YPD

plates indicated. (B–E) Increasing amounts of 3-amino-1,2,4-triazol (3-AT) reveal

the following order in strength of interactions: Slbo–Trbl4Trbl–Trbl4 Slbo–D/

NLK¼Trbl–D/NLK.

V. Masoner et al. / Developmental Biology 375 (2013) 33–4440

misexpression of WT Trbl (data not shown and Mata et al., 2000).D/NLK also retains WT activity when misexpressed in the germline (LLD, unpublished data). Thus the D/NLK mutation defines a

specific function for the Trbl catalytic domain to block BCmigration but not to block cell division in the wing.

Trbl kinase domain is required for Slbo binding and turnover

Previously, Trbl has been shown to bind Slbo in a pull downassay in S2 cells (Rorth et al., 2000), and we sought to test thestrength of this interaction using a sensitive yeast two-hybrid assay.As shown in Fig. 5, Trbl in a bait vector effectively interacts with Slboin a prey vector to grow on histidine drop out plates in the presenceof increasing amounts of 3-amino-1,2,4-triazol (3-AT). The reverseinteraction, with Slbo in the bait and Trbl in the prey vector,occurred with similar strength up to 75 mM 3-AT (data not shown).Surprisingly, Trbl in the bait vector is also able to interact with Trblin the prey vector (Fig. 5), but not as strongly as with Slbo in theprey vector (25 mM 3-AT as compared to 100 mM 3-AT, respec-tively). A second b-galactosidase reporter gene assay confirmed thatthe relative strength of Trbl–Slbo interaction is 1400-fold greaterthat the Trbl–Trbl interaction (Table 4).

Because the D/NLK mutation abrogates Trbl function in theborder cells, we used site-directed mutagenesis to make the sameD/NLK mutation in the central catalytic domain of the Trbl baitvector (Fig. 4). When tested on histidine drop out plates in a two-hybrid assay, the D/NLK mutation exhibited significantly weakerinteractions with both Slbo prey and Trbl prey (Fig. 5). In thequantitative b-gal reporter gene assay, the D/NLK mutant resultedin a 16.5-fold reduction in Trbl binding while the very strong Slbointeraction was reduced to undetectable levels. We designed asecond mutation to disrupt the C-terminal COP1 domain of theTrbl bait (Fig. 4), a motif deemed critical for interaction with theproteosome. The COP1 mutant in the Trbl prey resulted in noreduction in binding to either prey Slbo or prey Trbl in both theplate growth and quantitative b-gal reporter gene assays(Table 4). Thus Trbl–Trbl and Trbl–Slbo interactions require anintact catalytic domain but not a COP1 binding site.

We compared the ability of WT Trbl and the D/NLK mutant todirect Slbo turnover in vivo in two ways. First, we misexpressed bothtransgenes in the border cells using the Slbo2.6GAL4 driver. As shownin Fig. 6A, WT Trbl misexpression led to reduced Slbo protein levels inthe border cells, whereas slbo-lacZ reporter gene expression remainedhigh, confirming previous observations that the transcription of slbo isunchanged in this tissue (Rorth et al., 2000). In contrast, Slbo proteinlevels were unaffected by misexpression of D/NLK (Fig. 6B), consistent

Table 4

Bait/prey YPAD -L-

W

-L-W-

H

-L-W-Hþ10 mM

3AT

-L-W-Hþ25 mM

3AT

-L-W-Hþ50 mM

3AT

-L-W-Hþ75 mM

3AT

-L-W-Hþ100 mM

3AT

-L-W-

HþXGALb-gal

assay

Trbl/Trbl þþ þþ þþ þþ þ þ – – – 0.066

DLK/Trbl þþ þþ þþ þ – – – – – 0.004

Trbl/Slbo þþ þþ þþ þþ þþ þþ þþ þ þ 95.019

DLK/Slbo þþ þþ þþ þ – – – – – 0

COP1/Slbo þþ þþ þþ þþ þþ þþ þ þ þ 102.236

Strong control þþ þþ þþ þþ þþ þþ þþ þ þ 14.490

Weak control þþ þþ þþ þþ þþ þþ þ þ – 0.391

Negative

control

þþ þþ þþ þ – – – – – 0.014

MAV203 þþ – – – – – – – – 0

V. Masoner et al. / Developmental Biology 375 (2013) 33–44 41

with the failure of this mutation to bind Slbo (Fig. 5) and block BCmigration (Fig. 4C).

In a second approach, we misexpressed Trbl or D/NLK in the mainbody FC using a FLP-out driver in females bearing an inducible hsSlbotransgene (genotype: hsFLP; hs-Slbo P{wþ(mC)¼AyGAL4}25 P{w

þ(mc)¼UAS-GFP.S65T}T2/CyO; UAS-FlagTrbl; scheme outlined inFig. 6C). Following 2.5–3 H recovery from heat shock Slbo misexpres-sion, clones misexpressing Trbl in the main body FC were detected byexpression of UAS-GFP and revealed increased Slbo turnover com-pared to adjacent non-GFP, WT cells (Fig. 6D). In contrast, GFP-positive clones misexpressing the D/NLK transgene resulted in a noincrease in Slbo turnover compared to adjacent WT cells (Fig. 6E). FCclones misexpressing either WT Trbl or TrblD/NLK variably exhibitincreased cell size (data not shown), thus we interpret the strongnuclear Slbo accumulation in D/NLK clone shown in (Fig. 6D) as dueto their larger size rather than due to an increase in Slbo stability. Weconclude that the D/NLK catalytic motif mutation that disrupts Slbobinding in a yeast two-hybrid assay also abolishes the ability of Trblto direct Slbo turnover in vivo.

Discussion

Mammalian Trib proteins have diverse functions (reviewed inDobens and Bouyain, 2012), as transcriptional co-activators andrepressors in the nucleus, and as MAP kinase kinase inhibitors andproteosome adapters in the cytoplasm. Tribs bind and direct thedegradation of key regulatory proteins, in particular members ofthe C/EBP family, notably (1) C/EBPa, which is degraded by Trib1to promote the formation of acute myelogenous leukemia (AML)tumors and by Trib2 during myeloid differentiation (Keeshanet al., 2006) and (2) C/EBPb, which is degraded by Trib2 duringdifferentiation of 3T3-L1 preadipocytes (Naiki et al., 2007).As well, C/EBPb levels increase in Trib1 knockout mice (Yamamotoet al., 2007; Keeshan et al., 2008).

For Trib2, it has been demonstrated recently that a pointmutation in the DLK catalytic loop motif disrupts its ability todirect turnover of C/EBPa and promote acute myelogenousleukemia (Keeshan et al., 2010), and here we demonstrate thatthe same DLK/R mutation in fly Trbl compromises its ability todirect Slbo turnover and block BC migration. We also show that anintact DLK catalytic motif is required for Trbl interactions both withSlbo and with Trbl, itself. These data support the notion that theconserved catalytic domain is critical for Trbl function, however itremains unclear if this mutation disrupts ATP binding-dependentprotein folding or whether Tribs are functional kinases, a distinctionwhich will be resolved ultimately by identifying bonafide Tribsubstrates.

The D/NLK Trbl mutant surprisingly retains WT activity whenmisexpressed in the posterior wing compartment, resulting in

larger cells as measured by more widely spaced bristles whencompared to the anterior compartment. We note that a similarmutation in Trbl located just C-terminal to DLK (DLKLK/R) alsoretains WT activity to block cell proliferation when injected intothe blastoderm epithelium (Fig. 7 of Grosshans and Wieschaus,2000). Because WT Trbl is thought to block cell division bydirecting String/cdc25 turnover, our data suggest another domainoutside the Trbl catalytic loop must be required for Stringdegradation.

Several observations are worth noting regarding the trbl

protein null allele and specific antisera that we have analyzed.The trblD13 deletion allele described here (Fig. 3A) is a strongerallele than those used previously (Rorth et al., 2000), but stillyields a few escaper adults in combination with a deficiency ofthe locus (Table 1). Though trbl is not required for viability, wefound that rare escaper animals hatch late and are infertile withvestigial ovaries (data not shown), suggesting trbl has unexploredroles both in larval tissue growth and cell proliferation duringearly oogenesis. Antisera to Trbl reveal dynamic changes insubcellular localization during late oogenesis: Trbl accumulatesin FC nuclei up to stage 11 after which nuclear levels drop andTrbl accumulates at low levels in the cytoplasm (Fig. 1E and F).The notion that Trib localization is regulated and has functionalimportance is supported by observations that (1) GFP-taggedversions of Tribs are localized variously to the cytoplasm, nucleusand even the mitotic spindle (Saka and Smith, 2004), and (2) dur-ing BMP/TGF-b-signaling Tribs bind BMP receptors at the cellcortex, upon signaling are released into the cytoplasm to bind anddegrade SMURFS, and subsequently translocate to the nucleus toserve as SMAD co-activators (Hegedus et al., 2006, 2007; Chanet al., 2007; Hua et al., 2011).

Antisera detect dynamic changes in Trbl levels during BCmigration: Trbl is strongly expressed prior to delamination fromthe anterior epithelium and Trbl levels decrease during poster-iorwards BC migration (Fig. 2A–C). In slbo mutant egg chambers(Fig. 2D and F) Trbl levels are significantly higher compared to WTat all stages examined, indicating that slbo is necessary to repressTrbl. However Slbo misexpression alone is not sufficient torepress Trib expression (supplemental Fig. S2), suggesting thatcofactors mediate Slbo repression of Trbl. Our observation thatslbo represses Trbl stands at odds with previous work showingthat Slbo turnover is reduced in slbo mutant egg chambers (Rorthet al., 2000). While further work must be done to reconcile theseobservations, it is possible that slbo both represses Trbl expres-sion and activates an unidentified Trbl activator, whose existenceis implied by work in embryos (Grosshans and Wieschaus, 2000).In this way, while Trbl levels increase in a slbo mutant, its abilityto direct Slbo turnover could be compromised.

Together these data indicate that Slbo and Trbl are at the core ofa negative feedback loop (Fig. 7) in which Slbo represses Trbl

Fig. 6. Trbl kinase domain is required for Slbo turnover in vivo: (A) misexpression of UAS-FlagTrbl in the border cells using the Slbo2.6GAL4 driver reveals expression of the

transgene using the Trbl antisera (red, arrow) and reduced levels of Slbo protein (green) while in the third panel, slbo-lacZ gene expression remains high (magenta). The

fourth and fifth panels are DAPI staining (blue) and overlay, respectively. (B) Misexpression of UAS-D/NLK in the border cells using the Slbo2.6GAL4 driver also detects

transgene expression using Trbl antisera (red, arrow) and results in normal levels of Slbo protein (green). (C) Scheme for examining Slbo turnover in FC cells misexpressing

Trbl. Panel 1 shows the FC epithelium in a female of the genotype hsFLP1; hs-Slbo/P{wþ(mC)¼AyGAL4}25 P{wþ(mC)¼UAS-GFP.S65T}T2; UAS-Trbl. After FLP induction,

clones misexpressing Trbl can be detected by the expression of GFP, in panel 2. After 30 min heat shock induction of hs-Slbo, high levels of nuclear Slbo can be detected

throughout the FC epithelium (red) in panel 3. Three hours after recovery, Slbo turnover in Trbl-expressing cells (green) can be compared to WT cells (black) in panel 4. (D)

Misexpression of UAS-Trbl in the main body FC (inset) using the Flp-out driver is detected by GFP (green, arrow) and correlates with lower levels of hs-Slbo (red) following

heat shock recovery. Decreased hsSlbo was detected in 20/25 clones in the main body FC. (E) Misexpression of UAS-D/NLK in the main body FC (inset) using the Flp-out

driver is detected by GFP (green, arrow) and correlates with normal to increased levels of hsSlbo (red) following heat shock recovery. Increased hsSlbo was detected in 32/

32 clones in the main body FC.

V. Masoner et al. / Developmental Biology 375 (2013) 33–4442

expression and conversely Trbl (via its catalytic loop) binds anddegrades Slbo. We used a yeast two-hybrid interaction assay to testthe strength of Trbl interactions in the absence of Trbl-directed Slboturnover and showed (1) that weak interactions occur betweeneither Trbl–Trbl (Fig. 5 and Table 4) or Slbo–Slbo (L.P. and L.D.,unpublished observations), while (2) comparatively stronger Trbl–Slbo interactions occur. These observations adds a key feature to themodel (Fig. 7): the proposition that weak homomeric complexes ofTrbl multimers and Slbo dimers exchange preferentially for strongheteromeric Trbl–Slbo complexes to direct Slbo turnover. Such amodel is consistent with observations that in some instancesprotein kinase dimerization leads to autoinhibited complexes(Parrini et al., 2002) and is supported by our unpublished workshowing that (1) purified Trbl protein expressed in E. coli can form

dimers and tetramers and (2) the strength of Trbl–Trbl interactionimpacts the activity of target pathways (Bouyain, Pence andDobens, unpublished data).

Snail-related transcription factors, which direct E-cadherin-dependent cell migration in a wide range of normal and diseasedtissues, are also regulated by protein turnover (Batlle et al., 2000;Cano et al., 2000; Herranz et al., 2008). Several modificationsaffect Snail stability, including phosphorylation by PAK andGSK3b, dephosphorylation by the small C-terminal domain phos-phatase (SCP), and lysine oxidation is promoted by NFkB. In thelatter case, NFkB prevents Snail phosphorylation by GSK-3 andsubsequent degradation, whereas formation of a ternary complexbetween wild-type p53, the ubiquitin ligase Mdm2, and Snail2promotes degradation (Wang et al., 2009). It is likely that during

Fig. 7. A model for Trbl–Slbo interactions during cell migration. Trbl (green) is expressed in non-migratory epithelial cells and forms multimers (right) in a manner that

requires its catalytic domain (red). Slbo protein is expressed in migrating cells (left) and forms bZIP dimers (orange, zipper; blue, DNA binding domain). Negative feedback

between the two occurs when (1) Slbo binds to the trbl gene and represses its expression (bottom) or (2) Trbl binds Slbo protein via its catalytic domain (red) and directs

Slbo degradation (top) effectively closing the feedback loop. Trbl–Slbo interactions are preferred (top) to maintain low Slbo levels in epithelial cells or to direct oscillating

Slbo levels in migratory cells (see section ‘‘Discussion’’).

V. Masoner et al. / Developmental Biology 375 (2013) 33–44 43

BC migration a similar level of complexity underlies Trbl effectson Slbo stability, and the proposed feedback between these genesresults in oscillating levels of Slbo and corresponding fluctuatingexpression of Slbo target genes, notably DE-cadherin, whoseturnover at the cell membrane has been demonstrated to pro-mote proper BC migration (Mathieu et al., 2007).

In mammals, Trb family members have been identified astumor suppressors or oncogenes, depending on tissue context(Dobens and Bouyain, 2012) and sorting out these conflicting datamay be aided from a simpler Drosophila model. Conservedinteractions between Trbl and C/EBP during cell differentiationin flies and mammals suggest the possibility that interactionsbetween Trbl and the cdc25 phosphatase String observed duringcell division in fly tissue might be conserved in mammals as well.Conversely, mammalian work holds the promise to illuminateand direct further tests of Trbl function in Drosophila, notablydocumented interactions between mouse Trib3 and ATF4, anothermember of the B-Zip class of transcription factors active duringpancreas b-cell differentiation (Liew et al., 2010), and humanTrib3 with Atk kinase during insulin-target cell metabolism(Angyal and Kiss-Toth, 2012). Synergies between these parallellines of investigation will shed light on the diverse roles of Trbfamily members in cell growth, proliferation and differentiation.

Acknowledgments

We thank Pernille Rorth and Gertrude Schupbach for flystrains. We thank Erika Geisbrecht, lab members and studentsin BIO312 (Fall 2012) for helpful comments. Slbo antisera were agenerous gift from Scott Goode and Przemyslaw Szafranski. 2B10and Fas2 antisera were obtained from the Developmental StudiesHybridoma Bank developed under the auspices of the NICHD andmaintained by The University of Iowa. This work was supportedby a research grant from the National Science Foundation (RPG-00251-01-DDC) and support from the University of MissouriResearch Board to LLD. We acknowledge the technical assistanceof Elizabeth Dobens, Denny Swartzlander, Zachary Sebo, andMegan Smith.

Appendix A. Supporting information

Supplementary data associated with this article can be foundin the online version at http://dx.doi.org/10.1016/j.ydbio.2012.12.016.

References

Abdelilah-Seyfried, S., Chan, Y.M., Zeng, C., Justice, N.J., Younger-Shepherd, S.,Sharp, L.E., Barbel, S., Meadows, S.A., Jan, L.Y., Jan, Y.N., 2000. A gain-of-function screen for genes that affect the development of the Drosophila adultexternal sensory organ. Genetics 155, 733–752.

Angyal, A., Kiss-Toth, E., 2012. The tribbles gene family and lipoprotein metabo-lism. Curr. Opin. Lipidol. 23, 122–126.

Batlle, E., Sancho, E., Franci, C., Dominguez, D., Monfar, M., Baulida, J., Garcia DeHerreros, A., 2000. The transcription factor snail is a repressor of E-cadheringene expression in epithelial tumour cells. Nat. Cell Biol. 2, 84–89.

Bischof, J., Basler, K., 2008. Recombinases and their use in gene activation, geneinactivation, and transgenesis. Methods Mol. Biol. 420, 175–195.

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.

Chan, M.C., Nguyen, P.H., Davis, B.N., Ohoka, N., Hayashi, H., Du, K., Lagna, G., Hata,A., 2007. A novel regulatory mechanism of the bone morphogenetic protein(BMP) signaling pathway involving the carboxyl-terminal tail domain of BMPtype II receptor. Mol. Cell Biol. 27, 5776–5789.

Dobens, L.L.J., Bouyain, S., 2012. Developmental roles of tribbles protein familymembers. Dev. Dyn. 241, 1239–1248.

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

Eder, K., Guan, H., Sung, H.Y., Francis, S.E., Crossman, D.C., Kiss-Toth, E., 2008. LDLuptake by monocytes in response to inflammation is MAPK dependent butindependent of tribbles protein expression. Immunol. Lett. 116, 178–183.

Fichelson, P., Gho, M., 2004. Mother-daughter precursor cell fate transformationafter Cdc2 down-regulation in the Drosophila bristle lineage. Dev. Biol. 276,367–377.

Grosshans, J., Wieschaus, E., 2000. A genetic link between morphogenesis and celldivision during formation of the ventral furrow in Drosophila. Cell 101,523–531.

Hegedus, Z., Czibula, A., Kiss-Toth, E., 2006. Tribbles: novel regulators of cellfunction; evolutionary aspects. Cell. Mol. Life Sci. 63, 1632–1641.

Hegedus, Z., Czibula, A., Kiss-Toth, E., 2007. Tribbles: a family of kinase-likeproteins with potent signalling regulatory function. Cell. Signal. 19, 238–250.

Herranz, N., Pasini, D., Diaz, V.M., Franci, C., Gutierrez, A., Dave, N., Escriva, M.,Hernandez-Munoz, I., Di Croce, L., Helin, K., Garcia de Herreros, A., Peiro, S.,

V. Masoner et al. / Developmental Biology 375 (2013) 33–4444

2008. Polycomb complex 2 is required for E-cadherin repression by the Snail1transcription factor. Mol. Cell. Biol. 28, 4772–4781.

Hua, F., Mu, R., Liu, J., Xue, J., Wang, Z., Lin, H., Yang, H., Chen, X., Hu, Z., 2011. TRB3interacts with SMAD3 promoting tumor cell migration and invasion. J. Cell Sci.124, 3235–3246.

Kato, S., Du, K., 2007. TRB3 modulates C2C12 differentiation by interfering withAkt activation. Biochem. Biophys. Res. Commun. 353, 933–938.

Keeshan, K., Bailis, W., Dedhia, P.H., Vega, M.E., Shestova, O., Xu, L., Toscano, K.,Uljon, S.N., Blacklow, S.C., Pear, W.S., 2010. Transformation by Tribbleshomolog 2 (Trib2) requires both the Trib2 kinase domain and COP1 binding.Blood 116, 4948–4957.

Keeshan, K., He, Y., Wouters, B.J., Shestova, O., Xu, L., Sai, H., Rodriguez, C.G.,Maillard, I., Tobias, J.W., Valk, P., Carroll, M., Aster, J.C., Delwel, R., Pear, W.S.,2006. Tribbles homolog 2 inactivates C/EBPalpha and causes acute myelogen-ous leukemia. Cancer Cell 10, 401–411.

Keeshan, K., Shestova, O., Ussin, L., Pear, W.S., 2008. Tribbles homolog 2 (Trib2) andHoxA9 cooperate to accelerate acute myelogenous leukemia. Blood Cells Mol.Dis. 40, 119–121.

Kiss-Toth, E., 2011. Tribbles: ‘puzzling’ regulators of cell signalling. Biochem. Soc.Trans. 39, 684–687.

Kiss-Toth, E., Wyllie, D.H., Holland, K., Marsden, L., Jozsa, V., Oxley, K.M., Polgar, T.,Qwarnstrom, E.E., Dower, S.K., 2006. Functional mapping and identification ofnovel regulators for the Toll/Interleukin-1 signalling network by transcriptionexpression cloning. Cell. Signal. 18, 202–214.

Levine, B., Hackney, J.F., Bergen, A., Dobens, L. r., Truesdale, A., Dobens, L., 2010.Opposing interactions between Drosophila cut and the C/EBP encoded by slowborder cells direct apical constriction and epithelial invagination. Dev. Biol.344, 196–209.

Liew, C.W., Bochenski, J., Kawamori, D., Hu, J., Leech, C.A., Wanic, K., Malecki, M.,Warram, J.H., Qi, L., Krolewski, A.S., Kulkarni, R.N., 2010. The pseudokinasetribbles homolog 3 interacts with ATF4 to negatively regulate insulin exocy-tosis in human and mouse beta cells. J. Clin. Invest. 120, 2876–2888.

Lin, K.R., Lee, S.F., Hung, C.M., Li, C.L., Yang-Yen, H.F., Yen, J.J., 2007. Survival factorwithdrawal-induced apoptosis of TF-1 cells involves a TRB2-Mcl-1 axis-dependent pathway. J. Biol. Chem. 282, 21962–21972.

Mata, J., Curado, S., Ephrussi, A., Rorth, P., 2000. Tribbles coordinates mitosis andmorphogenesis in Drosophila by regulating string/CDC25 proteolysis. Cell 101,511–522.

Mathieu, J., Sung, H.H., Pugieux, C., Soetaert, J., Rorth, P., 2007. A sensitizedPiggyBac-based screen for regulators of border cell migration in Drosophila.Genetics 176, 1579–1590.

McQuilton, P., Pierre, S.E.S., 2012. FlyBase 101—the basics of navigating FlyBase.Nucleic Acids Res..

Montell, D.J., Rorth, P., Spradling, A.C., 1992. Slow border cells, a locus required fora developmentally regulated cell migration during oogenesis, encodes Droso-phila C/EBP. Cell 71, 51–62.

Naiki, T., Saijou, E., Miyaoka, Y., Sekine, K., Miyajima, A., 2007. TRB2, a mouseTribbles ortholog, suppresses adipocyte differentiation by inhibiting AKT andC/EBPbeta. J. Biol. Chem. 282, 24075–24082.

Norga, K.K., Gurganus, M.C., Dilda, C.L., Yamamoto, A., Lyman, R.F., Patel, P.H.,Rubin, G.M., Hoskins, R.A., Mackay, T.F., Bellen, H.J., 2003. Quantitative analysisof bristle number in Drosophila mutants identifies genes involved in neuraldevelopment. Curr. Biol. 13, 1388–1396.

Ohoka, N., Yoshii, S., Hattori, T., Onozaki, K., Hayashi, H., 2005. TRB3, a novel ERstress-inducible gene, is induced via ATF4-CHOP pathway and is involved incell death. EMBO J. 24, 1243–1255.

Parrini, M.C., Lei, M., Harrison, S.C., Mayer, B.J., 2002. Pak1 kinase homodimers areautoinhibited in trans and dissociated upon activation by Cdc42 and Rac1.Mol. Cell 9, 73–83.

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

Prudente, S., Sesti, G., Pandolfi, A., Andreozzi, F., Consoli, A., Trischitta, V., 2012. TheMammalian Tribbles Homolog TRIB3, Glucose Homeostasis, and Cardiovascu-lar Diseases. Endocr. Rev..

Qi, L., Heredia, J.E., Altarejos, J.Y., Screaton, R., Goebel, N., Niessen, S., Macleod, I.X.,Liew, C.W., Kulkarni, R.N., Bain, J., Newgard, C., Nelson, M., Evans, R.M.,Yates, J., Montminy, M., 2006. TRB3 links the E3 ubiquitin ligase COP1 to lipidmetabolism. Science 312, 1763–1766.

Remenyi, A., Good, M.C., Lim, W.A., 2006. Docking interactions in protein kinaseand phosphatase networks. Curr. Opin. Struct. Biol. 16, 676–685.

Rorth, P., Szabo, K., Texido, G., 2000. The level of C/EBP protein is critical for cellmigration during Drosophila oogenesis and is tightly controlled by regulateddegradation. Mol. Cell 6, 23–30.

Saka, Y., Smith, J.C., 2004. A Xenopus tribbles orthologue is required for theprogression of mitosis and for development of the nervous system. Dev. Biol.273, 210–225.

Sathyanarayana, P., Dev, A., Fang, J., Houde, E., Bogacheva, O., Bogachev, O., Menon,M., Browne, S., Pradeep, A., Emerson, C., Wojchowski, D.M., 2008.EPO receptor circuits for primary erythroblast survival. Blood 111, 5390–5399.

Schulz, C., Kiger, A.A., Tazuke, S.I., Yamashita, Y.M., Pantalena-Filho, L.C., Jones, D.L.,Wood, C.G., Fuller, M.T., 2004. A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling on the Drosophila male germ-linestem cell lineage. Genetics 167, 707–723.

Seher, T.C., Leptin, M., 2000. Tribbles, a cell-cycle brake that coordinates prolifera-tion and morphogenesis during Drosophila gastrulation. Curr. Biol. 10,623–629.

Selim, E., Frkanec, J.T., Cunard, R., 2007. Fibrates upregulate TRB3 in lymphocytesindependent of PPAR alpha by augmenting CCAAT/enhancer-binding proteinbeta (C/EBP beta) expression. Mol. Immunol. 44, 1218–1229.

Sung, H.Y., Guan, H., Czibula, A., King, A.R., Eder, K., Heath, E., Suvarna, S.K.,Dower, S.K., Wilson, A.G., Francis, S.E., Crossman, D.C., Kiss-Toth, E., 2007.Human tribbles-1 controls proliferation and chemotaxis of smooth musclecells via MAPK signaling pathways. J. Biol. Chem. 282, 18379–18387.

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

Takahashi, Y., Ohoka, N., Hayashi, H., Sato, R., 2008. TRB3 suppresses adipocytedifferentiation by negatively regulating PPARgamma transcriptional activity.J. Lipid Res. 49, 880–892.

Thibault, S.T., Singer, M.A., Miyazaki, W.Y., Milash, B., Dompe, N.A., Singh, C.M.,Buchholz, R., Demsky, M., Fawcett, R., Francis-Lang, H.L., Ryner, L.,Cheung, L.M., Chong, A., Erickson, C., Fisher, W.W., Greer, K., Hartouni, S.R.,Howie, E., Jakkula, L., Joo, D., Killpack, K., Laufer, A., Mazzotta, J., Smith, R.D.,Stevens, L.M., Stuber, C., Tan, L.R., Ventura, R., Woo, A., Zakrajsek, I., Zhao, L.,Chen, F., Swimmer, C., Kopczynski, C., Duyk, G., Winberg, M.L., Margolis, J.,2004. A complementary transposon tool kit for Drosophila melanogaster usingP and piggyBac.[see comment]. Nat. Genet. 36, 283–287.

Wang, S.P., Wang, W.L., Chang, Y.L., Wu, C.T., Chao, Y.C., Kao, S.H., Yuan, A.,Lin, C.W., Yang, S.C., Chan, W.K., Li, K.C., Hong, T.M., Yang, P.C., 2009. p53controls cancer cell invasion by inducing the MDM2-mediated degradation ofSlug. Nat. Cell Biol. 11, 694–704.

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

Yamamoto, M., Uematsu, S., Okamoto, T., Matsuura, Y., Sato, S., Kumar, H.,Satoh, T., Saitoh, T., Takeda, K., Ishii, K.J., Takeuchi, O., Kawai, T., Akira, S.,2007. Enhanced TLR-mediated NF-IL6 dependent gene expression by Trib1deficiency. J. Exp. Med. 204, 2233–2239.

Yokoyama, T., Nakamura, T., 2011. Tribbles in disease: signaling pathwaysimportant for cellular function and neoplastic transformation. Cancer Sci.102, 1115–1122.