The FERM-Domain Protein Expanded Regulates Hippo Pathway Activity via Direct Interactions with the...

10
Developmental Cell Article The FERM-Domain Protein Expanded Regulates Hippo Pathway Activity via Direct Interactions with the Transcriptional Activator Yorkie Caroline Badouel, 1,2,5 Laura Gardano, 1,2,3,5 Nancy Amin, 1,2 Ankush Garg, 1,2 Robyn Rosenfeld, 1,2 Thierry Le Bihan, 4 and Helen McNeill 1,2, * 1 Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada 2 Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada 3 Welcome Trust Centre for Cell Biology, Edinburgh EH9 3JR, Scotland, UK 4 Centre for System Biology at Edinburgh (CSBE), University of Edinburgh, The Kings Buildings, Edinburgh EH9 3JR, Scotland, UK 5 These authors contributed equally to this work *Correspondence: [email protected] DOI 10.1016/j.devcel.2009.01.010 SUMMARY The Hippo kinase pathway plays a central role in growth regulation and tumor suppression from flies to man. The Hippo/Mst kinase phosphorylates and activates the NDR family kinase Warts/Lats, which phosphorylates and inhibits the transcriptional acti- vator Yorkie/YAP. Current models place the FERM- domain protein Expanded upstream of Hippo kinase in growth control. To understand how Expanded regulates Hippo pathway activity, we used affinity chromatography and mass spectrometry to identify Expanded-binding proteins. Surprisingly we find that Yorkie is the major Expanded-binding protein in Drosophila S2 cells. Expanded binds Yorkie at endogenous levels via WW-domain-PPxY interac- tions, independently of Yorkie phosphorylation at S168, which is critical for 14-3-3 binding. Expanded relocalizes Yorkie from the nucleus, abrogating its nuclear activity, and it can regulate growth down- stream of warts in vivo. These data lead to a new model whereby Expanded functions downstream of Warts, in concert with 14-3-3 proteins to sequester Yorkie in the cytoplasm, inhibiting growth activity of the Hippo pathway. INTRODUCTION Understanding how cells regulate proliferation and apoptosis is crucial for understanding developmental events, such as organ size control, as well as disease conditions such as cancer, in which the regulation of growth and apoptosis is lost. Studies in recent years, primarily conducted in Drosophila, have implicated the Hippo (Hpo) signaling pathway as a central mechanism that regulates organ size by controlling both cell proliferation and cell death (reviewed in Bandura and Edgar, 2008; Pan, 2007). The Hpo pathway core consists of a kinase cascade in which the Ste20-like kinase Hpo, facilitated by the WW-domain-con- taining adaptor protein Salvador (Sav), phosphorylates and acti- vates the NDR family kinase Warts (Wts) (Pantalacci et al., 2003; Tapon et al., 2002; Udan et al., 2003; Wu et al., 2003). Wts, facil- itated by the Mob family protein Mats (Justice et al., 1995; Lai et al., 2005; Wei et al., 2007), phosphorylates the transcriptional coactivator Yorkie (Yki) (Huang et al., 2005; Oh and Irvine, 2008). This phosphorylation provides a 14-3-3-binding site on Yki, allowing 14-3-3 to shuttle Yki from the nucleus, inhibiting its function (Dong et al., 2007; Oh and Irvine, 2008). Yki binds Scal- loped (Bandura and Edgar, 2008; Goulev et al., 2008; Wu et al., 2008; Zhang et al., 2008b), and possibly other transcription factors, to regulate the expression of Cyclin E, Diap1, Fj, and Bantam, promoting cell proliferation and inhibiting apoptosis (reviewed in Bandura and Edgar, 2008; Edgar, 2006; Hariharan, 2006; Pan, 2007; Reddy and Irvine, 2008; Saucedo and Edgar, 2007; Zeng and Hong, 2008). Hpo pathway activation also leads to increased transcription of upstream components such as mer and ex, providing feedback inhibition of the pathway. Mutation of hpo, sav, or wts, or overexpression of yki, results in massive tissue overgrowth characterized by excessive cell proliferation and diminished apoptosis. The Hpo pathway plays a conserved role in organ size control and tissue homeostasis in mammals. YAP, the mammalian homolog of Yki, is upregulated in some tumors and can transform immortalized mammary epithelial cells in vitro (Overholtzer et al., 2006) and accelerate tumorigenesis (Zender et al., 2006). NF2, the human counterpart of Mer, is a known tumor suppressor gene whose mutations lead to neurofibromatosis (McClatchey and Giovannini, 2005). The mammalian homologs of Hpo (MST1/2), Sav (WW45), and Wts (Lats1/2) constitute a kinase cascade that inactivates YAP function via similar phosphorylation events (Dong et al., 2007; Hao et al., 2008; Praskova et al., 2008; Zhang et al., 2008a; Zhao et al., 2007). Whereas the molecular details of the core Hpo kinase cascade have been well worked out, it is less clear how the activity of this pathway is regulated. Current models propose that two membrane-associated FERM-domain proteins, Expanded (Ex) and Merlin (Mer), function as upstream components of the Hpo pathway (Hamaratoglu et al., 2006; Zeng and Hong, 2008). Hpo pathway activity is controlled in part by the large cadherin Fat, which is proposed to recruit Ex to apical membranes (Ben- nett and Harvey, 2006; Silva et al., 2006; Willecke et al., 2006). Ex Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 411

Transcript of The FERM-Domain Protein Expanded Regulates Hippo Pathway Activity via Direct Interactions with the...

Developmental Cell

Article

The FERM-Domain Protein Expanded RegulatesHippo Pathway Activity via Direct Interactionswith the Transcriptional Activator YorkieCaroline Badouel,1,2,5 Laura Gardano,1,2,3,5 Nancy Amin,1,2 Ankush Garg,1,2 Robyn Rosenfeld,1,2 Thierry Le Bihan,4

and Helen McNeill1,2,*1Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada2Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada3Welcome Trust Centre for Cell Biology, Edinburgh EH9 3JR, Scotland, UK4Centre for System Biology at Edinburgh (CSBE), University of Edinburgh, The Kings Buildings, Edinburgh EH9 3JR, Scotland, UK5These authors contributed equally to this work

*Correspondence: [email protected] 10.1016/j.devcel.2009.01.010

SUMMARY

The Hippo kinase pathway plays a central role ingrowth regulation and tumor suppression from fliesto man. The Hippo/Mst kinase phosphorylates andactivates the NDR family kinase Warts/Lats, whichphosphorylates and inhibits the transcriptional acti-vator Yorkie/YAP. Current models place the FERM-domain protein Expanded upstream of Hippo kinasein growth control. To understand how Expandedregulates Hippo pathway activity, we used affinitychromatography and mass spectrometry to identifyExpanded-binding proteins. Surprisingly we findthat Yorkie is the major Expanded-binding proteinin Drosophila S2 cells. Expanded binds Yorkie atendogenous levels via WW-domain-PPxY interac-tions, independently of Yorkie phosphorylation atS168, which is critical for 14-3-3 binding. Expandedrelocalizes Yorkie from the nucleus, abrogating itsnuclear activity, and it can regulate growth down-stream of warts in vivo. These data lead to a newmodel whereby Expanded functions downstream ofWarts, in concert with 14-3-3 proteins to sequesterYorkie in the cytoplasm, inhibiting growth activity ofthe Hippo pathway.

INTRODUCTION

Understanding how cells regulate proliferation and apoptosis is

crucial for understanding developmental events, such as organ

size control, as well as disease conditions such as cancer, in

which the regulation of growth and apoptosis is lost. Studies in

recent years, primarily conducted in Drosophila, have implicated

the Hippo (Hpo) signaling pathway as a central mechanism that

regulates organ size by controlling both cell proliferation and cell

death (reviewed in Bandura and Edgar, 2008; Pan, 2007).

The Hpo pathway core consists of a kinase cascade in which

the Ste20-like kinase Hpo, facilitated by the WW-domain-con-

taining adaptor protein Salvador (Sav), phosphorylates and acti-

Develo

vates the NDR family kinase Warts (Wts) (Pantalacci et al., 2003;

Tapon et al., 2002; Udan et al., 2003; Wu et al., 2003). Wts, facil-

itated by the Mob family protein Mats (Justice et al., 1995; Lai

et al., 2005; Wei et al., 2007), phosphorylates the transcriptional

coactivator Yorkie (Yki) (Huang et al., 2005; Oh and Irvine, 2008).

This phosphorylation provides a 14-3-3-binding site on Yki,

allowing 14-3-3 to shuttle Yki from the nucleus, inhibiting its

function (Dong et al., 2007; Oh and Irvine, 2008). Yki binds Scal-

loped (Bandura and Edgar, 2008; Goulev et al., 2008; Wu et al.,

2008; Zhang et al., 2008b), and possibly other transcription

factors, to regulate the expression of Cyclin E, Diap1, Fj, and

Bantam, promoting cell proliferation and inhibiting apoptosis

(reviewed in Bandura and Edgar, 2008; Edgar, 2006; Hariharan,

2006; Pan, 2007; Reddy and Irvine, 2008; Saucedo and Edgar,

2007; Zeng and Hong, 2008). Hpo pathway activation also leads

to increased transcription of upstream components such as mer

and ex, providing feedback inhibition of the pathway. Mutation of

hpo, sav, or wts, or overexpression of yki, results in massive

tissue overgrowth characterized by excessive cell proliferation

and diminished apoptosis.

The Hpo pathway plays a conserved role in organ size control

and tissue homeostasis in mammals. YAP, the mammalian

homolog of Yki, is upregulated in some tumors and can transform

immortalized mammary epithelial cells in vitro (Overholtzer et al.,

2006) and accelerate tumorigenesis (Zenderet al., 2006). NF2, the

human counterpart of Mer, is a known tumor suppressor gene

whose mutations lead to neurofibromatosis (McClatchey and

Giovannini, 2005). The mammalian homologs of Hpo (MST1/2),

Sav (WW45), and Wts (Lats1/2) constitute a kinase cascade

that inactivates YAP function via similar phosphorylation events

(Dong et al., 2007; Hao et al., 2008; Praskova et al., 2008; Zhang

et al., 2008a; Zhao et al., 2007).

Whereas the molecular details of the core Hpo kinase cascade

have been well worked out, it is less clear how the activity of this

pathway is regulated. Current models propose that two

membrane-associated FERM-domain proteins, Expanded (Ex)

and Merlin (Mer), function as upstream components of the Hpo

pathway (Hamaratoglu et al., 2006; Zeng and Hong, 2008).

Hpo pathway activity is controlled in part by the large cadherin

Fat, which is proposed to recruit Ex to apical membranes (Ben-

nett and Harvey, 2006; Silva et al., 2006; Willecke et al., 2006). Ex

pmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 411

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Figure 1. Discrete Domains Regulate Ex Apical and Junctional

Localization

(A) Schematic of the Ex protein. Ex consists of an N-terminal FERM domain,

a linker region, and a C-terminal proline-rich domain.

(B–F) High magnification of lateral views of imaginal eye discs expressing

different GFP- or HA-tagged Ex constructs (green, [B]–[F]) and stained for actin

(Rhodamine-phalloidine, red, [B0]–[E0]). (B) Full-length Ex localizes apically and

is enriched to apical junctions. (C) The Ex FERM domain primarily localizes

412 Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevie

consists of an N-terminal FERM domain, a linker region, and

a C-terminal proline-rich domain (Figure 1A). Expression of the

C-terminal half of Ex in the Drosophila wing can suppress growth

(Boedigheimer et al., 1997), and mutations that truncate the C-

terminal domain are functionally null and lethal (Pellock et al.,

2007). How the C-terminal proline-rich region of Ex inhibits

growth remains unknown. In addition, it is unclear how the Hpo

signal is terminated, as 14-3-3 proteins, which can function to

shuttle Yki out of the nucleus, have the potential to shuttle Yki

back in, in the absence of a cytoplasmic anchor.

To address the biochemical basis of Ex regulation of growth

and apoptosis, we purified Ex-binding proteins from Drosophila

cells by using affinity chromatography coupled with mass spec-

trometry. Surprisingly, we find that the major Ex-binding protein

is Yki. We show that Ex binds Yki at endogenous levels and regu-

lates Hpo pathway activity downstream of wts in vivo. Ex func-

tions to relocate Yki from the nucleus to the cytoplasm, thus

inhibiting Yki transcriptional activity. We demonstrate that the

Ex-Yki interaction occurs via Yki WW domains and three partially

redundant PPxY motifs in the Ex C-terminal domain. Binding of

Ex to Yki is independent of phosphorylation at S168, suggesting

that Ex can bind Yki in both phospho and dephosphorylated

forms. We propose a model whereby Ex functions in concert

with 14-3-3 proteins to sequester Yki in the cytoplasm, inhibiting

its activity in the regulation of growth and apoptosis.

RESULTS

In Vivo Analysis of Ex Localization ElementsPrevious data have indicated that Ex localizes apically, with a

concentration at cell-cell junctions (Bennett and Harvey, 2006;

Boedigheimer and Laughon, 1993; McCartney et al., 2000; Silva

et al., 2006; Willecke et al., 2006). To determine which portions of

Ex function to mediate its localization, we adopted an in vivo

structure-function analysis. Transgenic Drosophila lines were

established expressing full-length Ex and Ex deletion constructs

tagged with GFP or HA (Figure 1A). Full-length Ex-HA shows

a localization pattern similar to that of endogenous Ex, with

a localization at apical surfaces of epithelial cells, and a concen-

tration at apical junctions (Figure 1B). A construct with only the

N-terminal FERM domain (aa 1–400) localizes most strongly to

the apical membrane domain (Figure 1C). The linker region (aa

400–747) mediates weak membrane localization (Figure 1D),

whereas the C-terminal region (aa 748–end) becomes strongly

localized to apical junctions (Figures 1E and 1F). These data indi-

cate that the N terminus, containing the FERM domain, mediates

apical localization, that there is a weak apical localization element

located in the linker region, and that the C-terminal domain, previ-

ously identified as containing the growth-suppressive portions of

Ex, localizes to apical junctions.

Purification and Mass Spectrometric Identificationof an Ex-Yki ComplexTo understand how Ex regulates Hpo kinase pathway activity,

we sought to identify Ex-binding proteins in Drosophila cells.

apically. (D) The Ex linker domain appears mostly cytoplasmic, with a weak

apical enrichment. (E and F) The Ex C terminus localizes apically, with an

enrichment near apical junctions.

r Inc.

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Figure 2. Identification of Yki as an Ex-Binding Protein

(A) Identification of Ex-binding proteins. Ex was immunoprecipitated, by using anti-Flag antibody, from S2 cells transfected with Ex-Flag or Flag as a control.

Proteins coimmunoprecipitating with Ex-Flag or Flag were silver stained in a polyacrylamide gel. Arrows indicated two bands isolated from cells expressing

Ex-Flag, but not in the control, running at the expected sizes of Ex and Yki.

(B) Yki coimmunoprecipitates with Ex-Flag. S2 cells were transfected with Ex-Flag and V5-Wts as indicated on top, and a-Flag immunoprecipitates were probed

with a-Flag (Ex), a-Yki, a-Hpo, and a-V5 (Wts). One-tenth of the cell lysates used for immunoprecipitation were probed with the same antibodies as control. Note

that Yki, but not Hpo or Wts, coimmunoprecipitates with Ex.

(C) Ex and Yki interact at endogenous levels. Yki immunoprecipitates from untransfected S2 cells were subjected to western blot with a-Ex. Note that endogenous

Ex coimmunoprecipitates with Yki, and not with the control IgG.

(D) Junctional localization in the eye disc. High magnification of third-instar eye imaginal discs stained for Actin (red), Armadillo (green), and DAPI (blue) to illustrate

the morphology of the eye imaginal disc when viewed from the side. Apical is facing up (marked by an arrow), and junctional clusters are highlighted by Armadillo

(marked by an asterisk).

(E) Yki and Ex colocalize at apical junctions. Yki (red), Ex (blue), and the junctional marker Armadillo (green). As in (D), except that the discs were permeablized

before fixation and stained with Yki and Ex. The pictures represent a lateral view of an eye imaginal disc; the apical membrane is on top. The single channels are

represented in black and white for Ex and Yki. Note that Ex and Yki partially colocalize apically.

Drosophila S2 cells were transfected with Flag-tagged Ex. Ex

and its interacting proteins were immunoprecipated by using

Flag antibody. S2 cells transfected with the Flag empty vector

were used as a control. Elution from Flag-agarose beads (see

Experimental Procedures) was conducted in phosphoric acid.

A nano-LC/MS/MS analysis was performed on the tryptic digest

of the proteins associated with the immunoprecipitate in a gel-

free approach.

Table S1 reports the proteins found uniquely in the Ex-trans-

fected sample and not in controls. Surprisingly, one of the

most abundant coimmunoprecipitating proteins, detected with

the highest spectral counts, is Yki, detected with a high number

of peptides and a good level of sequence coverage in all exper-

iments. Consistent with our mass spectrometry results, silver-

stain analysis of the eluates revealed the presence of two major

bands specific to the Ex-Flag immunoprecipitates: A band at

200 kDa corresponds to Ex-Flag, and a 50 kDa band corre-

sponds to Yki molecular size (Figure 2A). We confirmed by immu-

noblot the presence of endogenous Yki immunoprecipitating

with Ex-Flag in S2 cells, whereas other components of the Hpo

Develo

pathway, such as Hpo and Wts, were not present in the immuno-

precipitates (Figure 2B and data not shown).

To determine if the interaction between Ex and Yki can occur

at physiological levels, we tested whether endogenous levels of

Ex and Yki coimmunoprecipitate in Drosophila S2 cells. Indeed,

western blot analysis of Yki immunoprecipitates demonstrated

that Ex also coimmunoprecipitates with Yki from untransfected

S2 cells, showing that these proteins can interact at endogenous

levels (Figure 2C).

To determine if there is a population of Yki that colocalizes with

Ex in vivo, we examined Yki and Ex localization in the eye imag-

inal disc, by using antibodies against the endogenous proteins.

Ex localizes strongly to apical junctions in Drosophila imaginal

discs (Figure 1). Strikingly, a population of Yki is found near the

apical cortex, colocalizing with Ex (Figures 2D and 2E and data

not shown). Treatment of the discs with Triton X-100 before fixa-

tion confirmed that a population of Yki is membrane associated

and colocalized with Ex at apical junctions (Figure 2E). Taken

together, these results strongly suggest that Ex and Yki interact

in vivo.

pmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 413

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Figure 3. The PPxY Motifs of Ex Bind Yki WW Domains

(A) Yki directly binds Ex. Bacterially expressed GST-Yki bound to glutathione Sepharose beads was incubated with the purified His-tagged Ex C terminus (His-Ex-

CT). Proteins associating resin-bound Yki were immunoblotted and probed with a-His to detect Ex. One-tenth of the Ex proteins used for the pull-down was

probed with a-His as a control (input). Note that the C-terminal region of Ex interacts with Yki, and not with a GST-tagged fragment of the intracellular part of

Fat used as a negative control (GST-Ctl).

(B) Yki structure. Yki contains two WW domains and is phosphorylated by Wts on S168. Yki-N and Yki-C are the deletion constructs used for the pull-down assay.

(C) Ex binds the Yki C terminus. GST-Yki mutants, bound to glutathione Sepharose beads, were incubated with the purified His-tagged Ex C terminus (His-Ex-

CT). Note that Ex is pulled down by the Yki C terminus (lane 4), but not by the N terminus (lane 3).

(D) Mutations in the WW domains of Yki disrupt interaction with Ex. Same pull-down experiments as in (C), with different Yki mutants. Mutation of the Wts phos-

phorylation site of Yki, S168A, does not disrupt the interaction with Ex (lane 4). Yki WW domains were disrupted by substituting two critical amino acids with

alanine. Disruption of both WW domains, DWW1 and DWW2, inhibits binding with Ex (lane 5), whereas disruption of only one of them (DWW1 and DWW2)

decreases but does not abrogate the interaction (lane 6 and 7).

(E) Mutations in the PPxY motifs of Ex disrupt interaction with Yki. Mutation of the three PPxY motifs were introduced in the Ex C-terminal construct by mutating

the critical tyrosine of each motif into an alanine (Ex3Y). This mutant and the wild-type Ex-CT were incubated with GST-Yki, and the pulled-down samples were

probed with a-His. Note that Ex3Y does not associate with GST-Yki (lane 8).

(F) S2 cells were transfected with Ex-Flag or Ex3Y-Flag, and a-Yki immunoprecipitates were probed with a-Flag (Ex) and a-Yki. One-tenth of the cell lysates used

for the immunoprecipitation was probed with the same antibodies. Contrary to the wild-type Ex, Ex3Y does not coimmunoprecipitate with Yki.

414 Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc.

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Ex Binds Directly to YkiTo determine if Ex directly interacts with Yki, we used in vitro pull-

down assays with proteins purified from bacteria. Pull-down

experiments revealed that Ex can directly bind Yki (Figure 3A).

To determine which portion of Ex binds Yki, we generated a series

of Ex deletion constructs tagged with a histidine epitope tag.

These studies demonstrated that the C-terminal part of Ex (His-

Ex-CT) is able to bind GST-Yki in vitro, whereas the N-terminal

part of Ex does not (Figure 3A; see Figure S1B available online).

Moreover, we confirmed by immunoprecipitation in S2 cells

that a Flag-tagged C-terminal Ex, but not DCT-terminal Ex, binds

Yki (Figure S1A). Thus, the Yki-binding region of Ex maps to the

C-terminal domain, which has been previously shown to be

essential for growth regulation in vivo.

The PPxY Motifs of Ex Bind Yki WW DomainsYki contains two WW domains in its C-terminal region, which

bind Wts (Huang et al., 2005), allowing Wts to phosphorylate

Yki on Serine 168. This phosphorylation generates a 14-3-3-

binding site, enabling 14-3-3 to shuttle Yki out of the nucleus

(Dong et al., 2007; Zhao et al., 2007). To determine if Wts phos-

phorylation of Yki is required for the binding between Ex and Yki,

we generated a nonphosphorylatable GST-Yki in which Serine

168 was replaced by an alanine (GST-Yki S168A). This mutant

binds His-Ex-CT as efficiently as wild-type in vitro, demon-

strating that Yki phosphorylation on S168 is dispensable for Ex

binding (Figure 3D, lane 4).

To map the regions in Yki responsible for interaction with Ex,

we performed pull-down assays with purified GST-Yki fusion

proteins containing different deletions constructs. We observed

that the N-terminal region of Yki (aa 1–295) does not bind Ex

(Figure 3C, lane 3). In contrast, the C-terminal region of Yki (aa

206–418), which includes both WW domains, binds strongly to

Ex (Figure 3C, lane 4).

To determine if one or both of Yki’s WW domains are respon-

sible for binding to Ex, we mutated two critical residues of the

WW domains, disrupting one (DWW1 [W292A/P295A] or DWW2

[W361A/P364A]) or both (DWW1 and DWW2 [W292A/P295A/

W361A/P364A]) domains. Disruption of both WW domains in

Yki abrogates its binding to Ex, whereas both single WW domain

disruptions induce only a decrease in Ex binding (Figure 3D,

lanes 5–7). These results suggest that both WW domains of Yki

are needed for efficient binding to Ex.

The WW domains of the human ortholog of Yki, the Yes-Asso-

ciated protein, YAP, belongs to a subgroup of WW domains that

are known to specifically bind PPxY motifs (Macias et al., 2002).

Interestingly, three of these motifs can be found in the C-terminal

region of Ex (aa 786–789, 844–847, and 1203–1206: see

Figure S2). To test the role of the PPxY motifs in Yki binding,

the critical tyrosine residue of each motif was mutated into an

alanine (Ex3Y). Using GST pull-down assays, we found that

Ex3Y is no longer able to bind Yki, suggesting that these PPxY

motifs are critical for interaction with Yki WW domains

(Figure 3E, lane 8). Moreover, we find that a Flag-tagged version

Develo

of Ex3Y expressed in S2 cells is not coimmunoprecipitated with

endogenous Yki, whereas the wild-type version of Ex is effi-

ciently precipitated with Yki (Figure 3F).

Our structure-function analysis revealed that Ex, like Wts,

binds to the WW domains of Yki, suggesting that the interaction

of Ex and Wts with Yki may be mutually exclusive. To directly test

whether Ex and Wts function competitively in binding Yki, we

examined if the Yki-Wts coimmunoprecipitation is affected by

expressing increasing amounts of Ex. When an increasing

amount of Ex is expressed in cells, the coimmunoprecipitation

of Ex with Yki increases, whereas the amount of Wts coimmuno-

precipitated with Yki decreases (Figure 3G). Taken together with

our observation that Wts was not present in Ex immunoprecipi-

tates (Figure 2B), these data suggest that Ex and Wts may

compete for binding to Yki. Interestingly, increasing levels of

Ex markedly compete Wts-Yki binding (Figure 3G), whereas

increasing Wts is less effective in inhibiting Ex-Yki binding

(Figure 2B), suggesting that the Ex-Yki interaction is stronger

than the Yki-Wts interaction.

Ex Inhibits Yki Transcriptional Activity in S2 CellsTo test the consequences of the Yki-Ex interaction on Yki

activity, we used a well-established transcriptional assay (Huang

et al., 2005). This assay relies on a fusion protein of the Gal4-

DNA-binding domain with Yki that can activate a UAS-luciferase

reporter in S2 cells. The luciferase activity, which reflects Yki

cotranscriptional activity, is reduced by the expression of Ex or

other members of the Hpo pathway (Hamaratoglu et al., 2006;

Huang et al., 2005) (Figure 4). We found that the C-terminal

domain of Ex, responsible for Yki binding, is necessary and suffi-

cient to strongly decrease Yki activity in this assay (Figures 4B

and 4C), suggesting that the N-terminal part of Ex may be

responsible for the regulation of Ex activity (Figure 4B). To iden-

tify the Yki-inhibiting domain in the C-terminal part of Ex, we first

subdivided it into two parts, CT1 and CT2, and tested their

effects on Yki transcriptional activity (Figure 4A). This assay

revealed that both CT1 and CT2 are able to decrease Yki activity;

however, they are less effective than the entire C terminus

(Figure 4C). Addition of CT1 and CT2 led to increased transcrip-

tional inhibition. The PPxY motifs involved in Yki binding are

found both in CT1 and CT2 (Figure 4A). To test the role of these

motifs in the inhibition of Yki activity, we used the Yki-binding

mutant Ex3Y. Ex3Y is significantly less potent than wild-type Ex

in repressing Yki activity, supporting a role for these Yki-binding

motifs in Yki inhibition (Figure 4D).

The observation that the C-terminal domain of Ex, with its PPxY

motifs, directly binds Yki (Figure 3A), and inhibits its cotranscrip-

tional activity in S2 cells (Figures 4B and 4C), suggests that Ex

might be able to inhibit Yki directly, in an Hpo/Wts-independent

fashion.

To test this hypothesis, we first asked whether the suppression

of Yki activity by Ex required Hpo. We observed that knockdown

of Hpo by RNAi fails to rescue the decrease in Yki activity induced

by Ex, despite a dramatic decrease in Hpo levels after RNAi

(G) Ex-Yki and Wts-Yki interactions are mutually exclusive. S2 cells were transfected with V5-Wts, HA-Yki, and an increasing amount of Ex-Flag plasmids, and

a-HA immunoprecipitates were probed with a-V5 (Wts), a-HA (Yki), and a-Flag (Ex). One-tenth of the cell lysates used for the immunoprecipitation was probed

with the same antibodies. Note that increasing amounts of Ex decrease the amount of Wts coimmunoprecipitating with Yki.

pmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 415

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

treatment (Figure 4E). The same treatment is able to inhibit the

effects of Hpo on Yki activity (Figure 4E). Thus, the effects of Ex

on Yki activity, at least in S2 cells, appears to be Hpo indepen-

dent.

Ex Expression Relocalizes Yki to the CytoplasmicCompartmentWe next observed the consequences of Ex expression on Yki

localization by expressing tagged versions of Ex and Yki in S2

cells. Yki expressed in S2 cells can be observed both in the

nucleus and the cytoplasm (Figure 4F). In contrast, Ex expressed

in S2 cells is predominantly cytoplasmic, with a component that

is enriched at the plasma membrane (Figure 4G). Coexpression

of Ex with Yki results in a dramatic relocalization of Yki to the

cytoplasmic compartment (Figures 4G). We confirmed that this

occurs with the DBD-Gal4-Yki fusion protein used in the tran-

scription assay as well (Figure S3), and in both cases quantitation

reveals that this alteration in localization is statistically significant

Figure 4. Ex Inhibits Yki Transcriptional Activity in S2 Cells

(A) Representation of Ex structure, with an indication of the different deletion

constructs.

(B–D) S2 cells were transfected with UAS-luciferase plasmid along with plas-

mids expressing the Gal4 DNA-binding domain fused to yki and plasmids

expressing hpo or different ex mutants as indicated on the bottom. Graphs

represent luciferase activity normalized with an internal control (Renilla lucif-

erase); error bars represent standard deviation. Note that the Ex C terminus

is necessary and sufficient to decrease Yki transcriptional activity, and that

Ex3Y is less potent than the wild-type at inhibiting Yki transcriptional activity.

(E) S2 cells were transfected as in (B)–(D) and treated with a specific hpo

dsRNA. In addition to the luciferase assay, a western blot was performed

and probed with a-Hpo. Note that Hpo quantity is largely decreased after

dsRNA treatment, and that Yki inhibition by Ex is Hpo independent.

(F and G) S2 cells transfected with (F) Yki-HA or (G) Flag-Ex and Yki-HA were

subjected to immunofluorescence with a-Flag (Ex, red) and a-Yki (green) anti-

bodies and DAPI (nucleus, blue). Note that coexpression of Ex leads to a reduc-

tion in Yki nuclear staining.

416 Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevie

(Figure S4). This relocalization of Yki away from the nucleus

provides a mechanistic explanation for the ability of Ex to repress

Yki transcriptional activity. Consistent with this finding in S2

cells, reducing the levels of Ex in the posterior compartment of

wing discs, by using UAS-ExRNAi and Engrailed-Gal4, leads to

increased levels of nuclear Yki (Oh and Irvine, 2008) (Figure S5).

Ex Regulates Growth In Vivo Downstream of wts

Taken together, these data lead to a model in which Ex can func-

tion in parallel to or downstream of Wts to regulate Yki transcrip-

tional activity. To determine if this is true in vivo, we examined the

effects of expressing Ex in the eye imaginal disc in a wts mutant

background. Mutations in hpo or wts lead to an overgrowth

phenotype characterized by overexpression of the Yki transcrip-

tional targets Cyclin E and DIAP1 (Hansen et al., 2003; Huang

et al., 2005; Pantalacci et al., 2003; Tapon et al., 2002; Udan

et al., 2003). Using the MARCM system, we analyzed the effect

of expression of Ex in wts mutant clones in developing

Drosophila eye discs. As previously reported, induction of wts

mutant clones results in a dramatic overgrowth phenotype,

with the mutant cells covering almost the entire eye disc. The

overgrowth phenotype is clearly visible, as the wts mutant eye

disc appeared deformed, with a folded excessive epithelium in

third-instar larval eye imaginal discs (Figure 5A). If Ex regulates

Yki only through the activation of Wts, its overexpression in

wts mutant clones should have no effect. On the contrary, we

observed that expression of Ex largely rescues the overgrowth

phenotype induced by wts mutation (Figure 5B). In fact, this

suppression is so dramatic that late third-instar eye discs are

not drastically deformed but retain a more normal shape (Fig-

ure 5B). These data indicate that Ex can function downstream

of wts to regulate growth in the eye imaginal disc.

We next examined the ability of Ex to regulate the expression of

a known transcriptional target of Yki, Cyclin E. Cyclin E is normally

expressed in a narrow stripe of cells posterior to the morphoge-

netic furrow (arrowhead: Figure 5C). Overexpression of Cyclin E

is clearly visible in wts tissue at early third instar as a dramatic

broadening of the expression domain of Cyclin E (white brackets:

Figure 5D). Strikingly, expression of Ex largely rescues Cyclin E

overexpression in wts mutant tissue (Figure 5E). The overgrowth

of the imaginal discs induced by wts clones leads to a lethality at

the pupal stage in all animals (Figure 5F). Expression of Ex in wts

mutant clones leads to significant rescue of the pupal lethality

induced by loss of wts (Figure 5F). Therefore, Ex can function

downstream of wts, both in the regulation of the Hpo pathway

target Cyclin E, and in blocking the lethality induced by the unre-

strained overgrowth of wts mutant cells.

We next tested if expression of the C-terminal region of Ex,

responsible for Yki binding, was also able to rescue the wts over-

growth phenotype, and we found that the C-terminal domain of

Ex is even more potent than full-length Ex in blocking the over-

growth of wts clones, rescuing both Cyclin E overexpression

and lethality (Figure 5F and data not shown). As a control, we

tested the expression of another portion of Ex, the loop domain

(also known as the linker domain; between the FERM domain

and the C terminus) (Figure S2). Expression of the loop is not

able to rescue the wts mutant phenotype, and it showed no

detectable rescue of either growth or Cyclin E expression

(Figure 5F and data not shown).

r Inc.

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

These results strongly suggest that Ex is able to regulate

growth by inhibiting Yki directly, independent of Hpo/Wts kinase,

both in vitro and in vivo. Taken together, these data suggest

a model in which the binding of Ex to Yki leads to reduced levels

of Yki in the nucleus, thereby terminating growth promotion and

apoptosis inhibition by the Hpo pathway (Figure S6).

DISCUSSION

The Hpo pathway has recently emerged as a central mechanism

that restricts organ size in Drosophila and controls tissue growth

and cancer susceptibility in mammals. Here, we provide several

Figure 5. Ex Regulates In Vivo Growth Downstream of wts(A and B) Third-instar eye imaginal discs containing (A) wtsx1 clones or (B) wtsx1

clones overexpressing Ex-GFP obtained by using the MARCM technique. �/�clones were marked by the presence of GFP. Note that the overgrowth and the

‘‘undulating’’ shape phenotypeofwtsx1 clonesare rescuedbyExoverexpression.

(C) Wild-type eye imaginal disc stained with a-Cyclin E. A thin band of cells

along the morphogenic furrow (MF) stain positively for Cyclin E (the arrowhead

marks the location of the MF).

(D–E00) Third-instar eye imaginal discs containing (D) wtsx1 clones or (E) wtsx1

clones overexpressing Ex-GFP. �/� clones were marked by the presence of

GFP (D and E), and discs were subjected to immunostaining with a-Cyclin E

(D0 and E0). The range of Cyclin E expression is greatly increased in the wtsx1

clone (compare the size of the bracket within the clone versus that outside of

the clone [D0]) and is rescued by the overexpression of Ex. Note some nonau-

tonomous proliferation is induced by wts clones in the presence or absence

of Ex. (D00) and (E00) represent an overlay of GFP and Cyclin E expression.

(F) Table summarizing the recovery of adults flies with the indicated genotypes.

A total of 25% of flies with the correct genotypes are expected in each case.

Note that Ex and Ex-CT rescue the lethality induced by wtsx1 clones.

Develo

lines of evidence demonstrating that Ex functions to regulate the

Hpo pathway by direct binding of Yki, antagonizing Yki function

by inhibiting its nuclear localization.

Current models propose that ex and mer function together to

restrict tissue growth upstream of hpo (Edgar, 2006; Hamarato-

glu et al., 2006; Hariharan, 2006; McCartney et al., 2000; Pan,

2007; Saucedo and Edgar, 2007). Mer and Ex colocalize with

cortical actin in the apical region of the cell (Boedigheimer and

Laughon, 1993; Boedigheimer et al., 1997; McCartney et al.,

2000). Both genetic and physical interactions have been

observed between Mer and Ex: loss of one copy of mer domi-

nantly enhanced wing overgrowth in ex mutants. In addition,

fragments of Mer and Ex protein can interact physically with

each other in far-western experiments or when overexpressed

in cultured cells (McCartney et al., 2000). Clones doubly mutant

for mer and ex have more dramatic overgrowth than either single

mutant (Hamaratoglu et al., 2006; Maitra et al., 2006), and mer,ex

double mutants phenocopy hpo mutants.

However, despite the widespread acceptance that ex and mer

function upstream of Hpo (Edgar, 2006; Hamaratoglu et al.,

2006; Pan, 2007; Saucedo and Edgar, 2007; Zeng and Hong,

2008), some data are difficult to reconcile with ex acting strictly

upstream of hpo in activation of the pathway. Genetic analysis

indicates that ex is downstream of dachs, which has been shown

to be directly upstream of wts in growth control (Cho et al., 2006).

Biochemical analysis of the effects of overexpressing Mer and

Ex also suggested that ex may not act simply upstream of hpo.

For example, overexpression of Mer in S2 cells leads to a shift

in Wts mobility, whereas overexpression of Ex does not alter

Wts mobility (Hamaratoglu et al., 2006). In vivo analysis has

also suggested that mer and ex may have different roles in

controlling growth and apoptosis (Blaumueller and Mlodzik,

2000; Pellock et al., 2007).

Using biochemical purification and mass-spectrometic anal-

ysis, we identified Yki as a major Ex-binding protein in Drosophila

S2 cells. We demonstrate that the binding of Yki to Ex is direct

and is mediated by a WW domain-PPxY interaction. We further

show that this interaction is independent of Wts-dependent

phosphorylation at S168, a site previously shown to be essential

for strong interactions of Yki with 14-3-3 proteins. Consistent

with our biochemical analysis, we show that ex can act down-

stream of wts in the regulation of growth in eye imaginal discs

and can repress the pupal lethality caused by excessive growth

of wts clones. In addition, we find that loss of hpo does not alter

the ability of ex to regulate yki activity, as indicated by transcrip-

tional assays in S2 cells, and that expression of Ex is sufficient to

relocalize Yki to the cytoplasm. These data lead to a model in

which Ex functions to repress Yki activity at least in part by

keeping Yki out of the nucleus. Intriguingly, the Yki homolog,

YAP, was first identified as a protein that binds Src family kinases

at the cell membrane (Sudol, 1994). Subsequent studies have

focused on the role of Yki in the nucleus. Interestingly, our immu-

nohistochemical analysis reveals that a portion of Yki colocalizes

with Ex at the cell membrane in Drosophila imaginal discs.

Once Yki is phosphorylated by Wts, it can bind 14-3-3 proteins

and can be transported out of the nucleus. However, since 14-3-

3-bound Yki can also shuttle back into the nucleus, Ex binding

Yki provides an anchor that can effectively dampen Yki activity.

14-3-3 shuttling activity results in an equilibrium of distribution of

pmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 417

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Yki between the nucleus and the cytoplasm. This equilibrium is

biased in favor of Yki in the cytoplasm in the presence of Ex

acting as an anchor. The presence of a tether of Yki in the cyto-

plasm was already suggested (Oh and Irvine, 2008) based on the

distribution of YkiS168A between the cytoplasm and the

nucleus, instead of predominantly in the nucleus.

The strong nuclear localization of Yki is only seen in Drosophila

tissues in cases of pathological stimulation of growth, such as in

wts loss-of-function clones, which lead to massive overgrowth.

The lack of detectable Yki nuclear localization during normal

growth regulation suggests that Yki is an exceedingly potent

growth regulator, and points to why there are many layers of

regulation of Yki localization. The need for Ex to dampen Yki

signaling in the nucleus is reflected by the increase of Cyclin E

and Diap1 transcription in ex mutant clones.

We speculate that the regulation of the Hpo pathway by

combined loss of Ex and Mer is so potent because one acts as

the brake and the other controls the accelerator. Ex restricts

Yki to the cytoplasm, thus blocking activity downstream,

whereas Mer activates Hpo activity, thereby restricting Yki via

inhibitory phosphorylation. Thus, loss of Ex on its own does

not have a dramatic effect on cell proliferation and apoptosis,

since the activity of the kinase cascade is regulated via Mer.

Conversely, as long as Ex is present, excessive pathway activity

induced by loss of Mer can be effectively modulated by the

dampening activity of Ex.

Our data strongly suggest that Ex regulates Yki activity down-

stream of wts, by directly binding Yki and inhibiting Yki nuclear

localization and transcriptional activity. We cannot, however,

exclude the possibility that Ex also has additional upstream roles

in regulating Hpo activity. Interestingly, as previously shown

(Hamaratoglu et al., 2006), overexpressed Ex does not induce

apoptosis in a wts mutant background, although it can block

growth (Figure S7), suggesting that ex is upstream of wts in

apoptosis control, yet downstream of wts in growth control.

Genetic dissection of this pathway is complicated by the well-

documented feedback loops in the Hpo pathway: for example,

Yki regulates the expression of both mer and ex. In addition,

genetic evidence suggests that ex and mer function together

to regulate endocytosis and growth factor signaling (Maitra

et al., 2006). Further biochemical dissection of Hpo pathway

activity will be required to fully elucidate the diverse ways in

which growth and apoptosis are controlled in response to

various developmental and environmental signals.

The binding of Ex to Yki is likely to be 14-3-3 independent, as

mutation of S168, the predominant Wts phosphorylation site,

impairs 14-3-3 binding, yet does not affect the ability of Ex to

bind Yki. Thus, Ex binding to Yki could provide a pool of Yki

that is nonphosphorylated and poised for release by upstream

growth regulators. The binding of 14-3-3 to Yki can protect Yki

from dephosphorylation. This provides a problem for the cell,

since 14-3-3 must dissociate from Yki to allow it to become

dephosphorylated, thus releasing a potent activator of prolifera-

tion and inhibitor of cell death, allowing it to re-enter the nucleus.

The ability of Ex to bind both phosphophorylated and dephos-

phorylated Yki provides a mechanism by which to anchor

dephosphorylated Yki in the cytoplasm.

An appealing model is that the binding of Ex to Yki may be

modulated, once in the cytoplasm, as an additional control point

418 Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevie

for the Hpo pathway. FERM-domain proteins frequently form

inhibitory intramolecular associations, blocking the activity of

the protein until the repression is relieved. Modifications of the

Ex FERM domain or linker region could thus alter the ability of

Ex to bind Yki in vivo. Ex localization at apical junctions is at least

partially dependent upon the atypical cadherin Fat, which can

regulate Hpo pathway activity. The recruitment of Ex complexes

(directly or indirectly) to Fat may modify the ability of Ex to

interact with Yki.

Our in vivo analysis indicates that the N-terminal FERM

domain of Ex contains apical localization elements, whereas

the C-terminal region contains junctional localization elements.

Thus, each of these localization elements might be regulated

independently and might impact on the ability of Ex to sequester

Yki. Identification of which protein(s) Ex binds at junctions may

illuminate the mechanisms by which Ex responds to external

inputs to regulate Yki activity.

All of the components of the Hpo pathway are well conserved

in mammals and have been shown to have conserved functions

in regulating growth. Loss of Hpo and Wts orthologs and overex-

pression of the Yki ortholog, YAP, have been implicated in

a variety of human cancers. FERM6, the human ortholog of Ex,

also regulates Hpo pathway activity (Zhao et al., 2007) in

mammals. Future studies will determine if FERM6 directly binds

YAP, and if disrupting YAP-FERM6 interactions is a tumor-

promoting event.

EXPERIMENTAL PROCEDURES

Molecular Biology

Full-length Ex, the FERM domain (aa 1–400), the linker region (aa 400–747),

and the C-terminal region (aa 748–end) were cloned into pAFW and pAWF

for expression of Flag-tagged proteins in S2 cells, and into pPGW and

pPHW for expression of GFP- and HA-tagged proteins in flies. N-terminal

Flag-tagged CT1 (aa 748–1136) and CT2 (aa 1129–end) were cloned into

pAc5.1. The Ex C terminus and N terminus (aa 1–747) were cloned in

pEXP1-DEST for expression in bacteria. Mutagenesis of the PPxY domains

of Ex was conducted by PCR; tyrosines 789, 847, and 1206 were changed

to alanines. Yki was clone into pGEX4T1 to express a GST-tagged protein in

bacteria. Mutagenesis of the WW domains of Yki was conducted by PCR,

and a critical tryptophan and proline were mutated into alanine, for one

(DWW1 [W292A/P295A] or DWW2 [W361A/P364A]) or both (DWW1 and

DWW2 [W292A/P295A/W361A/P364A]) domains.

Cell Culture and Immunoprecipitation

Drosophila S2 cells, cultured in S2 insect media (Sigma) containing 10% fetal

bovine serum (HyClone) and antibiotics, were transiently transfected by using

Effectine (QIAGEN) and were collected 48 hr or 72 hr after transfection. Western

blots were performed according to standard protocols. Antibodies used were

mouse a-Flag (1:10000, Sigma), rabbit a-Yki (1:1000, D. Pan), rat a-Yki

(1:3000), rabbit a-Hpo (1:6000), mouse a-V5 (1:1000, Invitrogen), rabbit a-Ex

(1:1000, A.S. Laughon), and mouse a-His (1:6000, Sigma). a-Yki antibody

was raised in rat against native GST-tagged protein, and a-Hpo was raised in

rabbit against the peptide CTTAKNNDDQKPRNR. Cells were lysed in buffer

(150 mM Tris [pH 7.5], 1 mM EDTA, 0.5% NP40, 0.2 mM PMSF, 1 mM EGTA,

and protease cocktail inhibitor [Roche]),and proteins were immunoprecipi-

tated. Rat a-Yki was added to the cell lysate for 2 hr at 4�C and was precipitated

by adding protein glutathione Sepharose (GE Healthcare) for 1 hr. Ex-Flag

immunoprecipitation was performed by using a-Flag M2 agarose (Sigma),

and HA-Yki was performed by using rat a-HA affinity matrix (Roche), incubated

for 1 hr at 4�C. SDS sample buffer was added after three washes of the beads

with the lysis buffer. Ex coimmunoprecipitating proteins were analyzed by LC-

MS-MS, as described in Supplemental Experimental Procedures.

r Inc.

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Luciferase reporter gene assays were performed by transfecting 10 ng Yki-

Gal4 plasmid with 2 ng UAS-luc and 1 ng renilla plasmids, with or without Ex or

Hpo plasmids in 96-well plates (5 wells per condition). Luciferase assays were

performed by using the Dual Luciferase Reporter Assay System (Promega).

For hpo knockdown, dsRNAs were synthesized with the MEGAscript RNAi

kit (Ambion), from PCR products with the primers described by Willecke

et al. (2006). Cells were incubated with 2 mg dsRNA per well in 50 ml

Schneider’s medium in 96-well plates. After 2 hr, 50 ml Schneider’s medium

containing 20% FBS was added to each well.

GST Pull-Down

His-tagged ex and GST-yki plasmids were transformed and amplified in BL21

bacteria, and protein production was induced by IPTG. His-tagged Ex proteins

were purified by using nickel beads (QIAGEN) and were eluted with a 10 mM

imidazole buffer. GST-Yki proteins were purified from bacteria by using gluta-

thione Sepharose beads (Amersham, Biosciences). His-tagged Ex proteins

were incubated with GST-Yki bound in glutathione beads in pull-down buffer

(20 mM Tris, 1 mM EDTA, 1% Triton X-100, 1 mM b-meraptoethanol) for

45 min at room temperature or for 2 hr at 4�C. Beads were then washed three

times in pull-down buffer before adding the sample SDS buffer.

Drosophila Genetics and Immunohistochemistry

Drosophila stocks used in this study were y,w1118 (wild-type) and wtsx1 (Xu

et al., 1995). We used the MARCM technique (Lee and Luo, 2001) to examine

wtsx1 clones alone or clones expressing ExFL or ExCTerm by using (1) eyFLP,

UAS-mCD8::GFP; tubGal80 FRT 82b, tubGal4/UAS-ExFL, wtsx1 FRT 82b; (2)

eyFLP, UAS-mCD8::GFP; tubGal80 FRT 82b, tubGal4/UAS-ExFL, wtsx1 FRT

82b; and (3) eyFLP, UAS-mCD8::GFP; tubGal80 FRT 82b, tubGal4/UAS-

ExCT, wtsx1 FRT 82b, respectively. Imaginal discs were fixed and stained

by following standard procedures. The primary antibodies used were mouse

a-Arm (1:400, DSHB), rabbit a-Ex (A.S. Laughon), rat a-Yki, rabbit a-Yki

(D. Pan), rabbit a-Casp3ACT (Cell Signaling Technology), and rat a-Cyclin E.

Actin was stained with rhodamine phalloidin (Invitrogen-Molecular Probes).

SUPPLEMENTAL DATA

Supplemental Data include seven figures, one table, Supplemental Experi-

mental Procedures, and Supplemental References and can be found with

this article online at http://www.cell.com/developmental-cell/supplemental/

S1534-5807(09)00038-0.

ACKNOWLEDGMENTS

We thank Frank Sicheri, Anne-Claude Ginras, Craig Smibert, and Nic Tapon for

advice and critical comments on this manuscript. We thank Ken Irvine, Nic

Tapon, Georg Halder, Duojia Pan, the Developmental Studies Hybridoma

Bank, and Alan Laughton for antibodies and flies. Funding for this work was

supported by a Cancer Research Society grant to H.M., by the ‘‘Fondation

pour la Recherche Medicale’’ to C.B., and Biotechnology and Biological

Sciences Research Council funding to T.B.

Received: September 11, 2008

Revised: December 11, 2008

Accepted: January 23, 2009

Published: March 16, 2009

REFERENCES

Bandura, J.L., and Edgar, B.A. (2008). Yorkie and Scalloped: partners in

growth activation. Dev. Cell 14, 315–316.

Bennett, F.C., and Harvey, K.F. (2006). Fat cadherin modulates organ size in

Drosophila via the Salvador/Warts/Hippo signaling pathway. Curr. Biol. 16,

2101–2110.

Blaumueller, C.M., and Mlodzik, M. (2000). The Drosophila tumor suppressor

expanded regulates growth, apoptosis, and patterning during development.

Mech. Dev. 92, 251–262.

Develo

Boedigheimer, M., and Laughon, A. (1993). Expanded: a gene involved in the

control of cell proliferation in imaginal discs. Development 118, 1291–1301.

Boedigheimer, M.J., Nguyen, K.P., and Bryant, P.J. (1997). Expanded func-

tions in the apical cell domain to regulate the growth rate of imaginal discs.

Dev. Genet. 20, 103–110.

Cho, E., Feng, Y., Rauskolb, C., Maitra, S., Fehon, R., and Irvine, K.D. (2006).

Delineation of a Fat tumor suppressor pathway. Nat. Genet. 38, 1142–1150.

Dong, J., Feldmann, G., Huang, J., Wu, S., Zhang, N., Comerford, S.A.,

Gayyed, M.F., Anders, R.A., Maitra, A., and Pan, D. (2007). Elucidation of

a universal size-control mechanism in Drosophila and mammals. Cell 130,

1120–1133.

Edgar, B.A. (2006). From cell structure to transcription: Hippo forges a new

path. Cell 124, 267–273.

Goulev, Y., Fauny, J.D., Gonzalez-Marti, B., Flagiello, D., Silber, J., and Zider,

A. (2008). SCALLOPED interacts with YORKIE, the nuclear effector of the

hippo tumor-suppressor pathway in Drosophila. Curr. Biol. 18, 435–441.

Hamaratoglu, F., Willecke, M., Kango-Singh, M., Nolo, R., Hyun, E., Tao, C.,

Jafar-Nejad, H., and Halder, G. (2006). The tumour-suppressor genes NF2/

Merlin and Expanded act through Hippo signalling to regulate cell proliferation

and apoptosis. Nat. Cell Biol. 8, 27–36.

Hansen, J., Floss, T., Van Sloun, P., Fuchtbauer, E.M., Vauti, F., Arnold, H.H.,

Schnutgen, F., Wurst, W., von Melchner, H., and Ruiz, P. (2003). A large-scale,

gene-driven mutagenesis approach for the functional analysis of the mouse

genome. Proc. Natl. Acad. Sci. USA 100, 9918–9922.

Hao, Y., Chun, A., Cheung, K., Rashidi, B., and Yang, X. (2008). Tumor

suppressor LATS1 is a negative regulator of oncogene YAP. J. Biol. Chem.

283, 5496–5509.

Hariharan, I.K. (2006). Growth regulation: a beginning for the hippo pathway.

Curr. Biol. 16, R1037–R1039.

Huang, J., Wu, S., Barrera, J., Matthews, K., and Pan, D. (2005). The Hippo

signaling pathway coordinately regulates cell proliferation and apoptosis by

inactivating Yorkie, the Drosophila homolog of YAP. Cell 122, 421–434.

Justice, R.W., Zilian, O., Woods, D.F., Noll, M., and Bryant, P.J. (1995). The

Drosophila tumor suppressor gene warts encodes a homolog of human

myotonic dystrophy kinase and is required for the control of cell shape and

proliferation. Genes Dev. 9, 534–546.

Lai,Z.C.,Wei, X., Shimizu, T., Ramos, E., Rohrbaugh,M.,Nikolaidis, N., Ho, L.L.,

and Li, Y. (2005). Control of cell proliferation and apoptosis by mob as tumor

suppressor, mats. Cell 120, 675–685.

Lee, T., and Luo, L. (2001). Mosaic analysis with a repressible cell marker

(MARCM) for Drosophila neural development. Trends Neurosci. 5, 251–254.

Macias, M.J., Wiesner, S., and Sudol, M. (2002). WW and SH3 domains, two

different scaffolds to recognize proline-rich ligands. FEBS Lett. 513, 30–37.

Maitra, S., Kulikauskas, R.M., Gavilan, H., and Fehon, R.G. (2006). The tumor

suppressors Merlin and Expanded function cooperatively to modulate

receptor endocytosis and signaling. Curr. Biol. 16, 702–709.

McCartney, B.M., Kulikauskas, R.M., LaJeunesse, D.R., and Fehon, R.G.

(2000). The neurofibromatosis-2 homologue, Merlin, and the tumor suppressor

expanded function together in Drosophila to regulate cell proliferation and

differentiation. Development 127, 1315–1324.

McClatchey, A.I., and Giovannini, M. (2005). Membrane organization and

tumorigenesis–the NF2 tumor suppressor, Merlin. Genes Dev. 19, 2265–2277.

Oh, H., and Irvine, K.D. (2008). In vivo regulation of Yorkie phosphorylation and

localization. Development 135, 1081–1088.

Overholtzer,M., Zhang,J.,Smolen, G.A.,Muir, B.,Li,W.,Sgroi,D.C.,Deng, C.X.,

Brugge, J.S., and Haber, D.A. (2006). Transforming properties of YAP, a candi-

date oncogene on the chromosome 11q22 amplicon. Proc. Natl. Acad. Sci. USA

103, 12405–12410.

Pan, D. (2007). Hippo signaling in organ size control. Genes Dev. 21, 886–897.

Pantalacci, S., Tapon, N., and Leopold, P. (2003). The Salvador partner

Hippo promotes apoptosis and cell-cycle exit in Drosophila. Nat. Cell Biol. 5,

921–927.

pmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevier Inc. 419

Developmental Cell

Ex Binds Yki to Inhibit Hpo Growth Signaling

Pellock, B.J., Buff, E., White, K., and Hariharan, I.K. (2007). The Drosophila

tumor suppressors Expanded and Merlin differentially regulate cell cycle

exit, apoptosis, and Wingless signaling. Dev. Biol. 304, 102–115.

Praskova, M., Xia, F., and Avruch, J. (2008). MOBKL1A/MOBKL1B phosphor-

ylation by MST1 and MST2 inhibits cell proliferation. Curr. Biol. 18, 311–321.

Reddy, B.V., and Irvine, K.D. (2008). The Fat and Warts signaling pathways:

new insights into their regulation, mechanism and conservation. Development

135, 2827–2838.

Saucedo, L.J., and Edgar, B.A. (2007). Filling out the Hippo pathway. Nat. Rev.

Mol. Cell Biol. 8, 613–621.

Silva, E., Tsatskis, Y., Gardano, L., Tapon, N., and McNeill, H. (2006). The

tumor-suppressor gene fat controls tissue growth upstream of expanded in

the hippo signaling pathway. Curr. Biol. 16, 2081–2089.

Sudol, M. (1994). Yes-associated protein (YAP65) is a proline-rich phospho-

protein that binds to the SH3 domain of the Yes proto-oncogene product.

Oncogene 9, 2145–2152.

Tapon, N., Harvey, K.F., Bell, D.W., Wahrer, D.C., Schiripo, T.A., Haber, D.A.,

and Hariharan, I.K. (2002). salvador Promotes both cell cycle exit and

apoptosis in Drosophila and is mutated in human cancer cell lines. Cell 110,

467–478.

Udan, R.S., Kango-Singh, M., Nolo, R., Tao, C., and Halder, G. (2003). Hippo

promotes proliferation arrest and apoptosis in the Salvador/Warts pathway.

Nat. Cell Biol. 5, 914–920.

Wei, X., Shimizu, T., and Lai, Z.C. (2007). Mob as tumor suppressor is activated

by Hippo kinase for growth inhibition in Drosophila. EMBO J. 26, 1772–1781.

Willecke, M., Hamaratoglu, F., Kango-Singh, M., Udan, R., Chen, C.L., Tao, C.,

Zhang, X., and Halder, G. (2006). The fat cadherin acts through the hippo

tumor-suppressor pathway to regulate tissue size. Curr. Biol. 16, 2090–2100.

420 Developmental Cell 16, 411–420, March 17, 2009 ª2009 Elsevie

Wu, S., Huang, J., Dong, J., and Pan, D. (2003). hippo encodes a Ste-20 family

protein kinase that restricts cell proliferation and promotes apoptosis in

conjunction with salvador and warts. Cell 114, 445–456.

Wu, S., Liu, Y., Zheng, Y., Dong, J., and Pan, D. (2008). The TEAD/TEF family

protein Scalloped mediates transcriptional output of the Hippo growth-regula-

tory pathway. Dev. Cell 14, 388–398.

Xu, T., Wang, W., Zhang, S., Stewart, R.A., and Yu, W. (1995). Identifying tumor

suppressors in genetic mosaics: the Drosophila lats gene encodes a putative

protein kinase. Development 121, 1053–1063.

Zender, L., Spector, M.S., Xue, W., Flemming, P., Cordon-Cardo, C., Silke, J.,

Fan, S.T., Luk, J.M., Wigler, M., Hannon, G.J., et al. (2006). Identification and

validation of oncogenes in liver cancer using an integrative oncogenomic

approach. Cell 125, 1253–1267.

Zeng, Q., and Hong, W. (2008). The emerging role of the hippo pathway in cell

contact inhibition, organ size control, and cancer development in mammals.

Cancer Cell 13, 188–192.

Zhang, J., Smolen, G.A., and Haber, D.A. (2008a). Negative regulation of YAP

by LATS1 underscores evolutionary conservation of the Drosophila Hippo

pathway. Cancer Res. 68, 2789–2794.

Zhang, L., Ren, F., Zhang, Q., Chen, Y., Wang, B., and Jiang, J. (2008b). The

TEAD/TEF family of transcription factor Scalloped mediates Hippo signaling

in organ size control. Dev. Cell 14, 377–387.

Zhao, B., Wei, X., Li, W., Udan, R.S., Yang, Q., Kim, J., Xie, J., Ikenoue, T., Yu, J.,

Li, L., et al. (2007). Inactivation of YAP oncoprotein by the Hippo pathway is

involved in cell contact inhibition and tissue growth control. Genes Dev. 21,

2747–2761.

r Inc.