+ All Categories
Home > Documents > The APC tumor suppressor counteracts -catenin activation...

The APC tumor suppressor counteracts -catenin activation...

Date post: 15-Jul-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
16
The APC tumor suppressor counteracts -catenin activation and H3K4 methylation at Wnt target genes Jose Sierra, 1,3 Tomonori Yoshida, 1,3 Claudio A. Joazeiro, 2 and Katherine A. Jones 1,4 1 Regulatory Biology Laboratory, The Salk Insitute for Biological Studies, La Jolla, California 92037, USA; 2 Department of Cancer and Cell Biology, Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, USA The APC tumor suppressor controls the stability and nuclear export of -catenin (-cat), a transcriptional coactivator of LEF-1/TCF HMG proteins in the Wnt/Wg signaling pathway. We show here that -cat and APC have opposing actions at Wnt target genes in vivo. The -cat C-terminal activation domain associates with TRRAP/TIP60 and mixed-lineage-leukemia (MLL1/MLL2) SET1-type chromatin-modifying complexes in vitro, and we show that -cat promotes H3K4 trimethylation at the c-Myc gene in vivo. H3K4 trimethylation in vivo requires prior ubiquitination of H2B, and we find that ubiquitin is necessary for transcription initiation on chromatin but not nonchromatin templates in vitro. Chromatin immunoprecipitation experiments reveal that -cat recruits Pygopus, Bcl-9/Legless, and MLL/SET1-type complexes to the c-Myc enhancer together with the negative Wnt regulators, APC, and TrCP. Interestingly, APC-mediated repression of c-Myc transcription in HT29-APC colorectal cancer cells is initiated by the transient binding of APC, TrCP, and the CtBP corepressor to the c-Myc enhancer, followed by stable binding of the TLE-1 and HDAC1 corepressors. Moreover, nuclear CtBP physically associates with full-length APC, but not with mutant SW480 or HT29 APC proteins. We conclude that, in addition to regulating the stability of -cat, APC facilitates CtBP-mediated repression of Wnt target genes in normal, but not in colorectal cancer cells. [Keywords: Wnt signaling; -catenin; MLL1/MLL2/SET1; H3K4 methylation; APC tumor suppressor; CtBP; TrCP] Received October 18, 2005; revised version accepted January 9, 2006. Wnt/Wg signaling events control body axis formation, cell proliferation, and organogenesis in many organisms (Cadigan 2002; Bienz and Clevers 2003; Gregorieff and Clevers 2005) and are important for self-renewal of in- testinal epithelial and hematopoietic stem cells (Radtke and Clevers 2005; Reya and Clevers 2005). In the canoni- cal Wnt/Wg signaling pathway, -catenin (-cat) func- tions as a dedicated transcriptional coactivator of LEF-1/ TCF HMG proteins (Cadigan 2002; Bienz and Clevers 2003). In the absence of Wnt signaling, newly synthe- sized -cat is targeted to a cytoplasmic “destruction complex” that contains Axin, protein phosphatase 2A, and the adenomatous polyposis coli (APC) tumor sup- pressor (Xing et al. 2003, 2004; Ha et al. 2004), where it undergoes sequential phosphorylation by casein kinase I (CKI) and glycogen synthase kinase-3 (GSK-3). Subsequent phosphorylation of APC by these protein ki- nases induces high-affinity binding to -cat and disas- sembly of the Axin complex (Xing et al. 2004). The phos- phorylated -cat is then ubiquitinated by TrCP and de- stroyed by proteasome-mediated proteolysis. Wnt ligands initiate a signaling cascade through Frizzled and LDL cell-surface receptors and the Dishevelled protein to ultimately inactivate GSK-3 and break the destruc- tion cycle. The unphosphorylated -cat then enters the nucleus, binds LEF-1/TCF proteins, and activates Wnt target genes (Bienz and Clevers 2003; Gregorieff and Clevers 2005). Misregulation of the Wnt signaling pathway is a hall- mark of many aggressive human cancers, including co- lon carcinomas and melanomas (Moon et al. 2004; Reya and Clevers 2005). The vast majority of colorectal adeno- mas and carcinomas contain sporadic or inherited trun- cations of the APC tumor suppressor (Kinzler and Vogel- stein 1996) or oncogenic stabilizing mutations in -cat, resulting in the chronic induction of c-Myc and other Wnt target genes. The most frequent cancer-causing mu- tations of APC are truncations that remove the C-termi- nal half of the protein, including binding sites for Axin and nuclear export signals, which render the mutant APC proteins unable to target -cat to the destruction complex and unable to function as a tumor suppressor (for reviews, see Bienz 2002; Henderson and Fagotto 3 These authors contributed equally to this work. 4 Corresponding author. E-MAIL [email protected]; FAX (858) 535-8194. Article and publication are at http://www.genesdev.org/cgi/doi/10.1101/ gad.1385806. 586 GENES & DEVELOPMENT 20:586–600 © 2006 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/06; www.genesdev.org Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.org Downloaded from
Transcript
Page 1: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

The APC tumor suppressor counteracts�-catenin activation and H3K4methylation at Wnt target genesJose Sierra,1,3 Tomonori Yoshida,1,3 Claudio A. Joazeiro,2 and Katherine A. Jones1,4

1Regulatory Biology Laboratory, The Salk Insitute for Biological Studies, La Jolla, California 92037, USA; 2Department ofCancer and Cell Biology, Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, USA

The APC tumor suppressor controls the stability and nuclear export of �-catenin (�-cat), a transcriptionalcoactivator of LEF-1/TCF HMG proteins in the Wnt/Wg signaling pathway. We show here that �-cat and APChave opposing actions at Wnt target genes in vivo. The �-cat C-terminal activation domain associates withTRRAP/TIP60 and mixed-lineage-leukemia (MLL1/MLL2) SET1-type chromatin-modifying complexes in vitro,and we show that �-cat promotes H3K4 trimethylation at the c-Myc gene in vivo. H3K4 trimethylation invivo requires prior ubiquitination of H2B, and we find that ubiquitin is necessary for transcription initiationon chromatin but not nonchromatin templates in vitro. Chromatin immunoprecipitation experiments revealthat �-cat recruits Pygopus, Bcl-9/Legless, and MLL/SET1-type complexes to the c-Myc enhancer togetherwith the negative Wnt regulators, APC, and �TrCP. Interestingly, APC-mediated repression of c-Myctranscription in HT29-APC colorectal cancer cells is initiated by the transient binding of APC, �TrCP, andthe CtBP corepressor to the c-Myc enhancer, followed by stable binding of the TLE-1 and HDAC1corepressors. Moreover, nuclear CtBP physically associates with full-length APC, but not with mutant SW480or HT29 APC proteins. We conclude that, in addition to regulating the stability of �-cat, APC facilitatesCtBP-mediated repression of Wnt target genes in normal, but not in colorectal cancer cells.

[Keywords: Wnt signaling; �-catenin; MLL1/MLL2/SET1; H3K4 methylation; APC tumor suppressor; CtBP;�TrCP]

Received October 18, 2005; revised version accepted January 9, 2006.

Wnt/Wg signaling events control body axis formation,cell proliferation, and organogenesis in many organisms(Cadigan 2002; Bienz and Clevers 2003; Gregorieff andClevers 2005) and are important for self-renewal of in-testinal epithelial and hematopoietic stem cells (Radtkeand Clevers 2005; Reya and Clevers 2005). In the canoni-cal Wnt/Wg signaling pathway, �-catenin (�-cat) func-tions as a dedicated transcriptional coactivator of LEF-1/TCF HMG proteins (Cadigan 2002; Bienz and Clevers2003). In the absence of Wnt signaling, newly synthe-sized �-cat is targeted to a cytoplasmic “destructioncomplex” that contains Axin, protein phosphatase 2A,and the adenomatous polyposis coli (APC) tumor sup-pressor (Xing et al. 2003, 2004; Ha et al. 2004), where itundergoes sequential phosphorylation by casein kinaseI� (CKI�) and glycogen synthase kinase-3� (GSK-3�).Subsequent phosphorylation of APC by these protein ki-nases induces high-affinity binding to �-cat and disas-sembly of the Axin complex (Xing et al. 2004). The phos-

phorylated �-cat is then ubiquitinated by �TrCP and de-stroyed by proteasome-mediated proteolysis. Wntligands initiate a signaling cascade through Frizzled andLDL cell-surface receptors and the Dishevelled proteinto ultimately inactivate GSK-3� and break the destruc-tion cycle. The unphosphorylated �-cat then enters thenucleus, binds LEF-1/TCF proteins, and activates Wnttarget genes (Bienz and Clevers 2003; Gregorieff andClevers 2005).

Misregulation of the Wnt signaling pathway is a hall-mark of many aggressive human cancers, including co-lon carcinomas and melanomas (Moon et al. 2004; Reyaand Clevers 2005). The vast majority of colorectal adeno-mas and carcinomas contain sporadic or inherited trun-cations of the APC tumor suppressor (Kinzler and Vogel-stein 1996) or oncogenic stabilizing mutations in �-cat,resulting in the chronic induction of c-Myc and otherWnt target genes. The most frequent cancer-causing mu-tations of APC are truncations that remove the C-termi-nal half of the protein, including binding sites for Axinand nuclear export signals, which render the mutantAPC proteins unable to target �-cat to the destructioncomplex and unable to function as a tumor suppressor(for reviews, see Bienz 2002; Henderson and Fagotto

3These authors contributed equally to this work.4Corresponding author.E-MAIL [email protected]; FAX (858) 535-8194.Article and publication are at http://www.genesdev.org/cgi/doi/10.1101/gad.1385806.

586 GENES & DEVELOPMENT 20:586–600 © 2006 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/06; www.genesdev.org

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 2: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

2002). Because Class I mutant APC proteins accumulatein the nucleus, they may also disrupt Wnt signaling byother means. For example, nuclear APC has been sug-gested to bind and sequester excess �-cat in the nucleus,preventing it from associating with the DNA-boundLEF-1/TCF proteins and dampening transcription in sig-naling cells (Neufeld et al. 2000a,b; Rosin-Arbesfeld et al.2000, 2003). Wild-type and mutant APC proteins werealso recently discovered to bind to the nuclear transcrip-tional corepressor, CtBP, potentially to redirect �-cat awayfrom Wnt target genes in vivo (Hamada and Bienz 2004).

Emerging biochemical and genetic studies have elabo-rated important aspects of the mechanism of Wnt tran-scriptional regulation. The central armadillo (ARM) re-peats of �-cat interact with the Bcl-9/Legless coactivator,which serves to connect �-cat to the Pygopus (Pygo) PhDfinger protein (for review, see Bienz and Clevers 2003;Gregorieff and Clevers 2005). Bcl-9/Lgs and Pygo havealso been shown to be required for �-cat to accumulatewithin the nucleus (Townsley et al. 2004). Although it isnot clear whether Bcl-9/Lgs and Pygo can interact withDNA-bound �-cat:LEF-1 complexes, genetic studiesstrongly implicate a role for these coactivators in tran-scription (Hoffmans and Basler 2004; Hoffmans et al.2005). Other studies have shown that �-cat is trans-ported from the nucleus by APC, which contains a cen-tral nuclear export sequence that is critical for its func-tion (Rosin-Arbesfeld et al. 2003). APC and LEF-1 com-pete for overlapping binding sites within the �-catarmadillo (ARM) repeats, suggesting that �-cat mustswitch between different complexes containing APC,Bcl-9/Lgs:Pygo, and LEF-1 before and after transcription.These steps are not well defined biochemically, and vari-ous models have been proposed to explain how �-catexchanges between APC and LEF-1 in the nucleus (forreview, see Tolwinski and Wieschaus 2004).

In addition to APC and LEF-1, the �-cat ARM repeatsinteract with the DNA helicases TIP49a/Pontin52 andTIP49b/TIP48/Reptin52 (Bauer et al. 1998, 2000), whichare subunits in the TRRAP/TIP60 histone acetyltrans-ferase (HAT) and mammalian INO80 and SWRCAP/SWR1 chromatin-remodeling complexes (Cai et al. 2003,2005; Jin et al. 2005). Both TIP60 and TIP49 directlyregulate Wnt target genes in vivo (Bauer et al. 2000; Fenget al. 2003; Kim et al. 2005b). Other �-cat-interactingproteins required for Wnt signaling include Brg1-con-taining chromatin remodeling complexes (Barker et al.2001) and CBP/p300 (Hecht et al. 2000; Miyagishi et al.2000). The region C-terminal to the �-cat ARM repeatscontains a very strong activation domain; however, thefactors that bind this domain are unknown. In vivo, theLEF-1/TCF proteins bind constitutively to Wnt targetgenes and repress transcription in the absence of Wntsignaling in conjunction with Gro/TLE-1, CtBP,HDAC1, and other corepressors (for review, see Coureyand Jia 2001; Chinnadurai 2002).

Here, we report that the �-cat C-terminal activationdomain and adjacent ARM 11/12 repeats (CTARM) se-lectively associate with nuclear chromatin remodelingsubunits of TRRAP/TIP60 histone acetyltransferase

(HAT), ISW1, and mixed-lineage-leukemia (MLL1/MLL2) SET1-type histone methyltransferase (HMT)complexes, and that �-cat promotes H3K4 trimethyl-ation (H3K4Me3) at the c-Myc gene in vivo. SET1 pro-teins mediate H3K4Me3 in a manner that depends uponprior ubiquitination of H2B by mammalian BRE1:Rad6ubiquitin ligase complexes (Kim et al. 2005a; Zhu et al.2005). The mammalian SET1-related HMT proteins (in-cluding hSET1, ALL-1/MLL1, MLL2, ALR-1, and HALR)reside in complexes with other conserved SET1 subunits(including Ash2, menin, HCF, WRD5, and RbBP5), andare frequent direct targets of transcriptional activators(Nakamura et al. 2002; Yokoyama et al. 2004; Dou et al.2005; Guenther et al. 2005; Milne et al. 2005; Schneideret al. 2005; Wysocka et al. 2005b). We show here thatH3K4Me3 levels increase upon induction of c-Myc tran-scription by �-cat, and decline when transcription is shutoff in cells expressing the APC tumor suppressor. Inter-estingly, repression of c-Myc transcription is accompa-nied by transient recruitment of APC, �TrCP, and CtBP.Moreover, nuclear CtBP coimmunoprecipitates withfull-length APC, but not with Class I or Class II mutantAPC proteins. We conclude that �-cat regulates H3K4methylation and is counteracted directly by APC at Wnttarget genes in vivo.

Results

The �-cat activation domain associates with nuclearTRRAP/TIP60, ISW1, and MLL1/MLL2 SET1-typecomplexes

We showed previously that �-cat:LEF-1 transcription onchromatin in vitro can be selectively inhibited, or“squelched,” by a fragment of �-cat that spans the Cterminus through ARM repeats 11 and 12 (GST-CTARM) (Tutter et al. 2001). The inhibition was specificbecause the �-cat CTARM fragment did not block thetranscriptional activity of the Notch enhancer complexin vitro or disrupt the cooperative binding of �-cateninand LEF-1 to chromatin. Shorter fragments of �-cat con-taining either the C-terminal domain (GST-CT) or ARMrepeats 11 and 12 alone (GST-ARM11/12) had no effecton �-cat activity in vitro. To characterize the proteinsthat bind this region of �-cat, the GST-CTARM proteinwas coupled to glutathione-S-Sepharose beads and incu-bated with nuclear extracts from SW480 colorectal can-cer (CRC) cells in GST-pulldown experiments. Thenuclear proteins that remained bound to the GST-CTARM beads after stringent washing were visualizedby SDS-PAGE and silver staining (Fig. 1A). Multiplenuclear proteins were found to bind GST-CTARM (Fig.1A, lane 4, asterisks), but not GST (Fig. 1A, lane 2), orGST-CT (Fig. 1A, lane 3) beads. These factors were notpresent in the purified GST-CTARM preparation (Fig.1A, lane 7), indicating that they derive from the SW480nuclear extract (Fig. 1A, lane 7).

To identify the �-cat-interacting proteins, the proteinsmarked by asterisks in lane 4 of Figure 1A were excisedin sections from the gel, eluted by trypsin proteolysis,

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 587

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 3: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

and analyzed by MALDI-TOF mass-spectrometry. Themajor CTARM-interacting proteins identified in theseexperiments were subunits from three different chroma-tin remodeling complexes. These include the transcrip-tion/transformation domain-associated protein TRRAP,the SNF2-related helicase p400, and the bacterial RuvB-related single-stranded DNA-dependent AAA+ ATPasesTIP49a/Pontin52 and TIP49b/TIP48/Reptin, all of whichare subunits of TRRAP/TIP60 HAT complexes (Cai et al.2005). Interestingly, TIP49a was previously shown to in-teract directly with �-cat ARM repeats 2–5 (Bauer et al.1998; Bauer et al. 2000), and both TIP49a and TIP60 havebeen shown to be important coactivators for �-cat invivo (Feng et al. 2003; Kim et al. 2005b).

The CTARM pulldown fraction also included subunitsfrom two other chromatin complexes, specifically the

Imitation Switch nucleosome-remodeling ATPase,ISW1, which is found in several distinct remodelingcomplexes, and the SET1-type complex protein, ALL-1/MLL1. Immunoblots confirmed the identification ofTRRAP, MLL1, and ISW1 in the CTARM pulldown frac-tions from HeLa (Fig. 1B, lanes 1–3) and SW480 (Fig. 1B,lanes 4–7) nuclear extracts, and further revealed the pres-ence of the MLL2 HMT (Fig. 1B, lanes 2,5) as well as thecommon SET1 subunits, menin (MEN-1 tumor suppres-sor), the Rb-interacting protein, RbBP5, and Ash2. Addi-tional pulldown experiments indicated that GST-�-cat-FL (full-length �-catenin) also associates with nuclearMLL1 and MLL2 proteins (Fig. 1B, lanes 8–11). As ex-pected, none of the GST-CTARM-interacting factorsbound to either GST-CT or GST-beads. Moreover, theGST-CTARM protein did not interact with other nuclear

Figure 1. Identification of proteins that bind to the�-cat CTARM domain in GST-pulldown experimentscarried out using SW480CRC nuclear extracts. (A) SDS-PAGE analysis of the proteins bound to GST-CTARM(lane 4), GST-CT (lane 3), and GST (lane 2) after incu-bation with SW480 nuclear extract, or of GST-CTARM(lane 7), GST-CT (lane 6), or GST (lane 5) proteins in theabsence of extract; proteins were visualized by silverstain. The GI protein accession numbers of theCTARM-interacting proteins identified by MALDI-TOF are listed at the right. (B, left) Immunoblot analy-sis of HeLa nuclear proteins bound to either GST-CTARM (lane 2) or GST-CT (control; lane 3) beads. Forcomparison, 2% of each input extract is shown in lane1. (Middle) Immunoblot comparison of SW480 nuclearproteins bound to GST-CTARM (lane 5), GST (control,lane 6), or GST-VP16 activation domain (lane 7). Theinput SW480 nuclear extract is shown in lane 4. (Right)Analysis of MLL1, MLL2, and Ash2L in SW40 nuclearextract (input, lane 8) or bound to GST-�-cat-FL (lane 9),GST-CTARM (lane 10) or GST (lane 11) beads. (C)Analysis of HMT (lanes 1–3) or HAT (lanes 4–6) activi-ties present in the GST-CTARM (lanes 1,4), GST (lanes3,6), or GST-VP16 (lanes 2,5) pulldown fractions. Theinput core histones are shown in lane 7. (Bottom) Thecore histones in each reaction were visualized by Coo-massie stain. (D) Analysis of exogenous �-cat:LEF-1 ac-tivation of the endogenous c-Myc gene in HeLa cellstransfected with siRNAs against �-cat or MLL2. Theinset shows an immunoblot of MLL2 and �-cat proteinlevels in control cells (lane 1), or cells expressing thesiRNA targeted to each protein (lane 2).

Sierra et al.

588 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 4: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

proteins required for H2B ubiquitination, such as Paf1 orRad6, nor did it bind the 19S proteasomal subunit Rpt6/SUG1, or the BAF57 or Brm chromatin remodeling sub-units (Fig. 1B; data not shown). Although it has beenreported that the �-cat CTARM domain interacts withthe p300/CBP histone acetyltransferase (Hecht et al.2000; Miyagishi et al. 2000), this association must berelatively weak, because it was not detected in our ex-periments (Fig. 1B, lanes 4–7).

To evaluate the relative strengths of these interac-tions, immunoblots were also carried out with parallelfractions obtained using the GST-VP16 activation do-main, which is known to bind MLL1 and SAGA/GCN5complexes. As shown in Figure 1B, the �-cat CTARMprotein binds MLL1/MLL2/SET1 complex subunitsmore strongly than does the VP16 activation domain,whereas the SAGA GCN5 subunit binds more avidly tothe VP16 activation domain (cf. lanes 5 and 7). TheTRRAP protein associates very tightly with both activa-tion domains, whereas the ISW1 subunit preferentiallybinds the CTARM protein. Because the TRRAP, TIP49,and ISW1 proteins are each present in multiple remod-eling complexes, more detailed studies will be needed tofurther characterize these CTARM-interacting com-plexes, and in particular, to identify which subunit(s)interact directly with the �-cat activation domain.

To assess whether the CTARM protein associateswith a functional HMT complex, we analyzed the abilityof the GST-CTARM and GST-VP16 pulldown fractionsto methylate and acetylate purified core histones invitro. As shown in Figure 1C, the CTARM pulldownfraction contained high levels of a histone H3-specificmethyltransferase activity (lane 1), but relatively weakHAT activity (lane 4), whereas the GST-VP16 fractiondisplayed weak HMT activity (lane 2), but strong HATactivity (lane 5), consistent with the enrichment ofGCN5 in this fraction. To assess whether the SET1-typecomplexes are required for �-cat activity in vivo, levelsof MLL2 in HeLa cells were reduced by a targeted smallinterfering RNA (siRNA), and the effect on c-MycmRNA was examined in the presence of transfected�-cat and LEF-1 expression vectors. As shown in Figure1D, endogenous c-Myc mRNA levels declined modestlyin the presence of the MLL2-specific siRNA, and moresubstantially with an siRNA directed against �-cat. Inall, these data suggest that the �-cat activation domainassociates with active histone H3 methylation com-plexes, and that MLL2 might contribute to �-cat-medi-ated induction of c-Myc transcription in vivo.

Ubiquitin is required for �-cat trans-activationof chromatin pBRE templates in vitro

These findings raised the question of whether the H3K4methylation or H2B ubiquitination steps might be im-portant for �-cat activity in vitro. The in vitro chroma-tin-based transcription assays measure RNA initiationby primer extension, and thus do not require RNA poly-merase II (RNAPII) Ser-2 phosphorylation or elongationfactors. Although H3K4Me3 has been strongly linked to

transcription elongation, MLL1/Set1 is present at mostactive RNAPII promoters in mammalian cells (Guentheret al. 2005), and thus could be required generally forRNA initiation or promoter clearance. Indeed, recentstudies indicate that MLL1 complexes strongly stimu-late transcription initiation on chromatin templates invitro (Dou et al. 2005).

These observations lead us to ask whether the endog-enous ubiquitin present in the Drosophila embryo chro-matin-assembly extract might be necessary for �-cat:LEF-1 transcription in vitro. To address this question,chromatin transcription reactions were incubated with aprotein containing the Vsp9p CUE domain, which bindstightly to monoubiquitin (Lima 2003). Interestingly, theGST-CUE protein strongly inhibited �-cat:LEF-1 activa-tion of pBRE chromatin templates in vitro (Fig. 2A, lane3). Mutant GST-CUE proteins that are unable to bindubiquitin failed to block transcription (F420A [Fig. 2A,lane 4] or P421A [Fig. 2A, lane 5]), and GST alone had noeffect (Fig. 2A, lane 6). The GST-CUE protein did notblock the cooperative binding of �-cat and LEF-1 to chro-matin (Fig. 2A, cf. lanes 9–12 and 8), nor did it interferewith nucleosome assembly (data not shown). Impor-tantly, GST-CUE inhibition could be overcome com-pletely by adding exogenous ubiquitin, either as His-tagged ubiquitin, GST-tagged ubiquitin, or a chain-ter-minating form of ubiquitin (Fig. 2B, cf. lanes 3 and 8; datanot shown). The chromatin footprint experiments re-vealed a slight but reproducible change in DNase I hy-persensitivity at one of the �-cat:LEF-1-binding sites (Fig.2A), which might result from modification of the pBREtemplate. Ubiquitination of �-cat itself is unlikely, be-cause the �N �-cat protein used in these experimentslacks the N-terminal residues for phosphorylation andubiquitination.

Interestingly, the GST-CUE protein also blocked tran-scription of pNRE chromatin templates by a recombi-nant Notch enhancer complex (containing CBF1, theNotch intracellular domain, and Mastermind) (Fig. 2C,lanes 4–7), indicating that the requirement for ubiquitinmay be general. Both Notch and �-cat-regulated tran-scription were also blocked by ubiquitin-aldehyde (Fig.2C, lane 9; data not shown), which inhibits deubiquiti-nating enzymes. In contrast, the Vsp9p CUE domain didnot block transcription from �-globin (�-glo) promoteron a nonchromatin (DNA) template (Fig. 2A–C), nor didit interfere with basal- or Tat-activated HIV-1 transcrip-tion elongation on naked DNA in vitro (Fig. 2D). Thesedata suggest that ubiquitination, like acetylation, maybe generally required for enhancer- and chromatin-de-pendent initiation of transcription in vitro, and that fur-ther studies in this system should be useful in definingthe enzymes and mechanism involved.

�-cat and other Wnt pathway-specific regulators cycleon and off the c-Myc enhancer in LiCl-treated cellsin vivo

We next used chromatin immunoprecipitation (ChIP)experiments to assess whether �-cat regulates H3K4 tri-

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 589

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 5: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

methylation at Wnt target genes in vivo. C2C12 (mousemyoblast) cells were synchronized and �-cat was in-duced by treatment with lithium, a GSK-3� inhibitor,under conditions shown previously to induce �-catenin(Baek et al. 2003). As shown in Figure 3A, c-Myc RNAlevels increased rapidly upon the addition of LiCl (lanes1–6), accompanied by an increase in nuclear �-cat pro-tein (lanes 7–12). Although the C2C12 cells were incu-bated continuously with lithium, �-cat and associatedcoactivators appeared transiently at either 30–45 min(Fig. 3B; lanes 9,10), or 30–60 min (Fig. 3C; lanes 3–5),and then were replaced by the TLE1 corepressor. In con-trast, LEF-1 bound constitutively to the c-Myc enhancer(Fig. 3B, lanes 7–12). The ChIP conditions used here werespecific, because LEF-1 was not detected at a distal up-stream region of the c-Myc gene (Fig. 3B, lanes 1–6), norwas c-Myc enhancer DNA recovered in immunoprecipi-tates with control IgG serum (Fig. 3B, lanes 7–12).

�-Catenin was recruited to the c-Myc enhancer to-gether with Pygopus and Bcl-9/Lgs (Fig. 3B, lanes 9,10),which are Wnt pathway-specific coregulators that appearto be required for both transcription and nuclear trans-

port of �-cat. MLL2 and the SET1-type subunits Ash2and menin were also present with �-cat (Fig. 3C, lanes3–5), and H3K4Me3 increased strongly at the c-Mycgene. Because the GSK-3� kinase, which is the directtarget of LiCl, has also been detected in the nucleus, weasked whether it might also be present at the c-Myc en-hancer. As shown in Figure 3B, GSK-3� was present atthe enhancer with kinetics similar to those of the TLE-1corepressor (Fig. 3B, lanes 7,11,12), and opposite of thatof �-cat. The rapid disappearance of the GSK-3� kinaseupon treatment of cells with lithium raises the possibil-ity that it functions as part of the corepressor complex atinactive Wnt target genes. More detailed kinetic ChIPanalyses indicate that this cyclic pattern of alternatingcoactivator and corepressor complexes repeats for mul-tiple cycles in cells that are persistently exposed to LiCl(S. Wang and K.A. Jones, unpubl.). Although the en-hancer factors cycle on and off of the enhancer, levels ofRNAPII and the CDK9 transcription elongation factorsteadily accumulate at the gene (Fig. 1B), in parallel withthe rise in steady-state c-Myc mRNA levels.

Given that GSK3� was present at the c-Myc enhancer,

Figure 2. Ubiquitin is required for �-cat:LEF-1transcription of chromatin templates in vitro.(A) The CUE domain competes for transcrip-tion on chromatin in vitro. Primer-extensionanalysis of RNA from a chromatin-assembledLEF-1 reporter gene (pBRE) in the presence(lanes 2–6) or absence (lane 1) of 120 nM re-combinant �N �-cat protein. RecombinantLEF-1 (�AD; 120 nM) was present in all reac-tions, and the �-globin (�-glo) gene was addedas a control for nonchromatin (DNA) tran-scription. Where indicated, reactions con-tained 15 µg (2.5 µM) each of GST-CUE (lane3), GST-CUE-F420A (lane 4), GST-CUE-P421A (lane 5), or GST alone (lane 6). Lanes7–12 show DNase I footprint analyses of thebinding of �-cat:LEF-1 in reactions 1–6, re-spectively. (B) Ubiquitin rescues the inhibi-tory effect of GST-CUE on �-cat-regulatedtranscription in vitro. (Top) Primer extensionanalysis of pBRE and �-glo (control) RNA inreactions containing (lanes 2–10) or lacking(lane 1) recombinant �-cat and LEF-1. Whereindicated, reactions contained low (2.5 µM,lanes 3,7,8) or high (10 µM, lane 4) levels ofGST-CUE, or low (2.5 µM, lanes 5,9,10) orhigh (10 µM, lane 6) GST control, in either theabsence of exogenous ubiquitin (lanes 1–6) orin the presence of either 1 µM (lanes 7,9) or 10µM (lanes 8,10) of His-tagged ubiquitin. (C)The GST-CUE domain blocks Notch en-hancer activation of chromatin templates invitro. Where indicated, reactions contained (lanes 2–10) or lacked (lane 1) the recombinant Notch intracellular domain (NICD) andCBF1 DNA-binding protein. Reactions 3–10 also contained the Notch coactivator Mastermind (MAM). GST-CUE levels were 170 nM(lane 4), 830 nM (lane 5), 1.7 µM (lane 6), and 3.3 µM (lane 7), and the GST control was added at 5.3 µM (lane 8). Ubiquitin aldehyde(1.8 nM) and MG132 (4 nM) in DMSO were present in lane 9, and the DMSO alone was tested as a control in lane 10. RNA synthesizedfrom the CBF1-dependent promoter was analyzed by primer extension; �-glo was added as a control for nonchromatin transcription.(D) The CUE domain does not block HIV-1 Tat-regulated transcription initiation or elongation on the HIV-1 promoter DNA in vitro.HIV-1 RNA was analyzed by the run-off elongation assay in the presence of GST-Tat (lanes 2,4–6), GST-CUE (lanes 3–6), or GST alone(lanes 1,3).

Sierra et al.

590 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 6: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

Figu

re3.

Ch

IPan

alys

isof

�-c

atin

duct

ion

ofm

uri

ne

c-M

yctr

ansc

ript

ion

inli

thiu

m-t

reat

edC

2C12

cell

s.(A

,le

ft,

lan

es1–

6)R

T–P

CR

anal

ysis

ofc-

Myc

RN

Ain

C2C

12ce

lls

trea

ted

wit

h10

mM

LiC

lfo

rth

eti

mes

indi

cate

dab

ove

each

lan

e;�

-act

inm

RN

Ale

vels

wer

em

onit

ored

asa

con

trol

.(R

igh

t,la

nes

7–12

)Th

ele

vels

of�

-cat

,Pyg

o,an

dL

EF-

1in

the

lith

ium

-tre

ated

C2C

12ce

lls

wer

em

onit

ored

byim

mu

nob

lot.

(B)C

hIP

anal

ysis

ofth

ec-

Myc

enh

ance

rat

diff

eren

tti

mes

foll

owin

gin

duct

ion

of�

-cat

byex

posu

reof

cell

sto

10m

ML

iCl.

Th

epr

imer

su

sed

tom

onit

orth

eu

pstr

eam

LE

F-1

site

sin

the

mou

sec-

Myc

gen

een

han

cer

are

indi

cate

din

the

sch

emat

icdi

agra

mre

lati

veto

the

P1

RN

Ast

art

site

(+1)

.(L

anes

1–6)

Adi

stal

(10

Kb)

ups

trea

mre

gion

was

mon

itor

edas

aco

ntr

ol.

Lan

es7–

12sh

owth

etr

ansc

ript

ion

fact

orbi

ndi

ng

toth

eW

nt-

resp

onsi

vec-

Myc

enh

ance

r(r

ef).

(C)

Ch

IPan

alys

isof

ML

L2

and

SET

1-ty

peco

mpl

exsu

bun

its

atth

ec-

Myc

enh

ance

rfo

llow

ing

expo

sure

to10

mM

LiC

lfo

rth

eti

mes

indi

cate

dab

ove

each

lan

e.(D

)Qu

anti

tati

veP

CR

anal

ysis

ofso

me

ofth

eC

hIP

sam

ples

show

nin

Ban

dC

.In

each

grap

h,

the

kin

etic

sof

�-c

atlo

cali

zati

onto

the

c-M

ycen

han

cer

(sh

own

ingr

een

)is

com

pare

dw

ith

the

spec

ific

tran

scri

ptio

nfa

ctor

indi

cate

dat

the

bot

tom

ofea

chpa

nel

.

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 591

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 7: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

we decided to test for the presence of other negative Wntregulators, in particular the APC tumor suppressor and�-cat ubiquitin ligase, �TrCP. Interestingly, both factorswere detected at the c-Myc enhancer; however, they didnot colocalize with TLE1, but rather appeared togetherwith �-cat (Fig. 3C, lanes 3–5). Although proteins in-volved in degradation of other short-lived activators havebeen localized to enhancer elements of target genes (Mu-ratani and Tansey 2003), it was unexpected to find APCwith �-cat at the c-Myc gene, because the two proteinsinteract in a mutually exclusive manner in vitro. Con-sequently, it has been assumed that �-cat must transferbetween APC-containing nuclear import/export com-plexes and DNA-bound LEF-1/TCF enhancer complexes.The ChIP data suggest an alternate scenario in whichAPC, functioning as a nuclear chaperone, accompanies�-cat both on and off the DNA. A more quantitativeassessment of the exchange kinetics for some of thesetranscription factors was provided by real-time PCRanalysis of chromatin complexes (Fig. 3D), which sup-ports the conclusion that APC associates with the c-Mycenhancer with kinetics similar to those observed for�-cat, Pygo, and Bcl-9/Lgs.

Repression of c-Myc transcription in HT29-APC cellsis accompanied by binding of APC, �TrCP, and CtBPto the enhancer

These observations suggested to us that destructioncomplex subunits like APC and �TrCP might participatein transcription, for example, to disassemble the Wntenhancer complex or mediate the exchange of coactiva-tors and corepressor complexes between transcriptioncycles. This possibility was difficult to assess in thelithium-treated C2C12 cells, which continuously induce�-cat, but we reasoned that it might be useful to examineby ChIP the shut-off of c-Myc transcription upon expres-sion of APC in colon cancer cells. For these experiments,we used an engineered HT29-APC colorectal cancer cellline (Morin et al. 1996), which contains a truncated ClassII mutant APC that is unable to degrade the endogenous�-cat, as well as an integrated full-length APC transgeneunder the control of a zinc-inducible metallothionein(MT) promoter. The induction of wild-type APC by zincin these cells promotes the degradation of �-cat and re-presses Wnt target gene expression. As a control, ChIPexperiments were carried out in HT29-�-Gal cells,which contain a �-Galactosidase transgene expressedfrom the identical zinc-inducible MT promoter (Morin etal. 1996).

As shown in Figure 4A, exposure of HT29-APC cells tozinc caused a rapid induction of APC RNA (lanes 1–7),whereas steady-state c-Myc RNA levels noticably de-clined between 2 and 3 h later (lanes 8–15). In contrast,�-actin mRNA levels were unaffected by zinc in eithercell line, and zinc had no effect on c-Myc mRNA levelsin the control HT29-�-Gal cells (Fig. 4A, lanes 16–22).Immunoblotting of full-length APC protein was rela-tively insensitive, but showed a gradual accumulation ofthe protein at 8–12 h after induction with zinc, whereas

steady-state levels of both �-cat and c-Myc protein sig-nificantly declined between 4 and 8 h (Fig. 4B, lanes 1–5).No changes were observed in the level of the Sp1 tran-scription factor in these extracts, which was monitoredas a control. ChIP analysis revealed that LEF-1, �-cat,Pygo, Bcl-9/Lgs, and the MLL2/SET1-type complex sub-units were present at the active c-Myc enhancer in bothcell lines in the absence of zinc (Fig. 4C, lane 1 for HT29-�-Gal cells, and lane 8 for the HT29-APC cells). As ex-pected, RNAPII, CDK9, acetylated H4K8, and trimeth-ylated H3K4 were also present at high levels at the activec-Myc gene (Fig. 4C, lanes 1–7). Interestingly, the endog-enous Class II mutant HT29 APC protein was also re-cruited to the active c-Myc enhancer in vivo. However,unlike the situation in LiCl-treated C2C12 cells, �-catand the associated coactivators did not cycle on and offof the enhancer, but rather remained stably bound, likeLEF-1, to the c-Myc gene.

Although exposure to zinc had no effect on any ofthese transcription factor interactions in the controlHT29-�-Gal cells (Fig. 4C, lanes 1–7), induction of thefull-length APC protein caused a rapid dissociation ofthe Wnt enhancer proteins (�-cat, Bcl-9/Lgs, and Pygo)and associated coactivators (MLL2, menin, RbBP5, Ash2)from the c-Myc gene in HT29-APC cells (Fig. 4C, lanes8–14). RNAPII and H3K4Me3 levels declined graduallyin these experiments, whereas LEF-1 levels remained un-changed. Most interestingly, the full-length APC proteintransiently appeared at the c-Myc enhancer during theloss of the �-cat and coactivator proteins (Fig. 3C,D), inconjunction with �TrCP, CtBP, and YY1 proteins. Atlater times, only the TLE-1 corepressor and HDAC1 co-repressors were detected with LEF-1 at the repressedgene (Fig. 4C, lanes 8–14). These data suggest that theAPC-mediated shut-off of c-Myc transcription proceedsin two steps, initiated by the transient binding of full-length APC and the CtBP corepressor to the enhancer,and followed by the stable binding of TLE-1 and HDAC1.

To examine the exchange of transcription coactivatorand corepressor complexes in more detail, the associa-tion of LEF-1, �-cat, Pygo, Bcl-9/Lgs, APC, and CtBPwith the c-Myc enhancer in HT29-APC and HT29-�-Galcells was analyzed by RT–PCR (Fig. 4D). These resultsindicate that �-cat, Pygo, and Bcl-9/Lgs exit the promoterwith similar kinetics, whereas Ash2 and H3K4Me3 lev-els decline more slowly. Both APC-FL and CtBP weredetected at the c-Myc enhancer in HT29-APC cells attimes overlapping the loss of �-cat and coactivator sub-units. To further assess whether the truncated mutantAPC resides in a stable multiprotein complex with �-catin HT29 cells, chromatin complexes from the HT29-�-Gal cell line were isolated with �-cat antibody and sub-jected to reChIP with antisera to APC and LEF-1. Asshown in Figure 4E, both APC and LEF-1 could be im-munoprecipitated from the initial �-cat-containing chro-matin complex (lanes 8,9), indicating that the three fac-tors are closely associated in vivo. Most importantly, wenote that the exchange of coactivator and corepressorcomplexes at the c-Myc enhancer significantly preceedsthe decline in �-cat protein levels that result from APC-

Sierra et al.

592 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 8: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

Figu

re4.

An

alys

isof

AP

C-i

ndu

ced

shu

t-of

fof

hu

man

c-M

ycge

ne

tran

scri

ptio

nin

the

HT

29-A

PC

CR

Cce

llli

ne.

(A,t

op,l

anes

1–7)

RT

–PC

Ran

alys

isof

the

indu

ctio

nof

AP

C-F

Lm

RN

Ain

HT

29-A

PC

cell

saf

ter

expo

sure

toZ

nC

l 2fo

rdi

ffer

ent

tim

es(i

nm

inu

tes)

,as

indi

cate

dab

ove

each

lan

e.R

T–P

CR

anal

ysis

of�

-act

inm

RN

Ais

show

nb

elow

asa

con

trol

.(M

idd

le,l

anes

8–15

)RT

–PC

Ran

alys

isof

the

decl

ine

inc-

Myc

mR

NA

leve

lsin

the

Zn

Cl 2

-tre

ated

HT

29-A

PC

cell

s,w

ith

tim

esof

expo

sure

toZ

nC

l 2(i

nh

ours

)in

dica

ted

abov

eea

chla

ne.

RT

–PC

Ran

alys

isof

�-a

ctin

mR

NA

issh

own

asa

con

trol

.(B

otto

m,l

anes

16–2

2)R

T–P

CR

anal

ysis

ofc-

Myc

,AP

C-F

L,a

nd

�-a

ctin

mR

NA

leve

lsin

the

con

trol

HT

29-�

-Gal

cell

s,w

hic

hw

ere

expo

sed

toZ

nC

l 2fo

rth

eti

mes

indi

cate

dab

ove

each

lan

e.(B

,la

nes

1–5)

Imm

un

oblo

tan

alys

isof

stea

dy-s

tate

leve

lsof

AP

C-F

L,

�-c

at,

and

c-M

ycpr

otei

ns

inex

trac

tsfr

omH

T29

-AP

Cce

lls

trea

ted

wit

hZ

nC

l 2fo

rth

edi

ffer

ent

tim

es(i

nh

ours

)in

dica

ted

abov

eea

chla

ne.

Lev

els

ofth

eSp

1tr

ansc

ript

ion

fact

orar

ein

clu

ded

asa

con

trol

.(C

)C

hIP

anal

ysis

oftr

ansc

ript

ion

fact

ors

bou

nd

toth

ec-

Myc

gen

ein

HT

29-�

-Gal

cell

s(l

anes

1–7)

orin

HT

29-A

PC

cell

s(l

anes

8–14

)by

expo

sure

toZ

nC

l 2fo

rth

edi

ffer

ent

amou

nts

ofti

me

(in

min

ute

s)in

dica

ted

abov

eea

chla

ne.

(D)C

hIP

anal

ysis

byR

T–P

CR

for

som

eof

the

sam

efa

ctor

san

alyz

edin

C.E

ach

pan

elan

alyz

estr

ansc

ript

ion

fact

oroc

cupa

ncy

atth

ec-

Myc

enh

ance

rin

HT

29-A

PC

cell

s(b

lue)

vs.

the

con

trol

HT

29-�

-Gal

cell

s(r

ed).

FL-A

PC

was

dete

cted

wit

han

tise

rasp

ecif

icto

aC

-ter

min

aldo

mai

n(A

PC

-C),

wh

erea

sth

em

uta

nt

AP

Cpr

otei

nw

asde

tect

edu

sin

gan

anti

sera

spec

ific

toth

eN

term

inu

sof

the

prot

ein

(AP

C-N

).(E

)R

e-C

hIP

anal

ysis

tote

stw

het

her

the

mu

tan

tH

T29

AP

Cpr

otei

nan

den

doge

nou

s�

-cat

enin

co-o

ccu

pyth

ec-

Myc

enh

ance

rin

HT

29C

RC

cell

s.C

hIP

inH

T29

-�-G

alce

lls

(wit

hou

tzi

nc)

was

firs

tca

rrie

dou

tu

sin

gth

e�

-�-c

atan

tibo

dy,

and

the

imm

un

ocom

plex

was

dilu

ted

in10

mM

dith

ioth

reit

ol.

Ali

quot

sw

ere

then

imm

un

opre

cipi

tate

dw

ith

�-A

PC

(Nte

rmin

us-

spec

ific

)or

�-L

EF-

1an

tise

ra,

and

the

prec

ipit

ated

chro

mat

inw

asam

plif

ied

byP

CR

usi

ng

prim

ers

spec

ific

for

the

c-M

ycen

han

cer.

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 593

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 9: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

mediated proteolytic degradation, strongly suggestingthat APC acts directly and immediately to facilitate therepression of Wnt target genes.

Binding of �-cat to CKI�-phosphorylated APC blocksbinding to LEF-1 and inhibits LEF-1:�-cattranscription in vitro

These observations led us to re-examine the question ofwhether �-cat can interact simultaneously with eitherPygo and Bcl-9/Lgs, or APC, when bound to LEF-1 onDNA. We initially assessed whether GST-LEF-1, or dif-ferent domains of APC, could bind the endogenous �-catprotein in pulldown experiments carried with SW480nuclear extracts (Fig. 5A). For these studies, we used hu-man APC proteins GST-APC-A,B,C, which contains allthree 15-amino-acid repeats; or GST-APC-2,3, whichcontains the second and third 20-amino-acid repeat (Xinget al. 2003). The native �-cat in the extract bound withhigh affinity to GST-LEF-1 (Fig. 5A, lane 3), weakly tothe APC-2,3 fragment (Fig. 5A, lane 6), and not at all to

the APC-A,B,C protein (Fig. 5A, lane 4). Phosphorylationof the APC-2,3 by CK1� (P-APC) enhances its affinity forrecombinant �-cat by several hundredfold in vitro (Xinget al. 2004), and strongly increased binding to �-cat inour experiments (Fig. 5A, lane 5). CK1�-phosphorylationof GST-APC-2,3 also strongly enhanced binding to bothfull-length and N-terminal-truncated (�N �-cat) recom-binant �-cat in vitro (Fig. 5B, cf. lanes 4,5 and 6,7).

To evaluate the DNA-binding activity of these com-plexes, EMSA experiments were carried out with recom-binant LEF-1, �-cat, Bcl-9 (HD2 domain), and fragmentsof the APC �-cat-interacting regions in vitro. As shownin Figure 5C, �-cat readily forms a ternary complex withLEF-1 on DNA (lane 4); however, the migration of thiscomplex is unaffected by high levels of GST-APC-2,3 orGST (lanes 7,8,9, respectively). In contrast, the CK1�-phosphorylated GST-APC-2,3 competed for the forma-tion of the �-cat:LEF-1:DNA complex (Fig. 5C, lanes 5,6),indicating that �-cat is unable to bind simultaneously toP-APC-2,3 and LEF-1. The opposite result was obtainedwith the Bcl-9 HD2 domain, which readily bound

Figure 5. Analysis of the interactions be-tween recombinant human �-cat and hu-man LEF-1, human Bcl-9 (HD2 domain;amino acids 288–419), APC-A,B,C, or CKI-phosphorylated or unphosphorylated APC-2,3 proteins in vitro. The (human) APC frag-ments used here are APC-A,B,C, which con-tains all three 15-amino-acid repeats (aminoacids 1100–1189), and APC-2,3, which con-tains the second and third 20-amino-acid re-peat (amino acids 1362–1540; Xing et al.2003). (A) CKI-phosphorylation of APC-2,3enhances binding to �-cat in GST pulldownexperiments carried out using SW480nuclear extracts. SW480 nuclear extract wasincubated with Glutathione-S-Sepharose 4Bbeads coupled to the following GST fusionproteins: GST-LEF-1, GST-APC-A,B,C, CKI-phosphorylated (P-APC) or unphosphory-lated GST-APC-2,3, or GST (control), andexamined for associated nuclear �-cat byWestern blot after stringent washing of thebeads. The �-cat in the SW480 extract (in-put, 1%) is shown in lane 1. The bead-coupled GST fusion proteins are visualizedby Ponceau S stain as a loading control. (B)FL and �N �-catenin were produced by invitro transcription and translation in acoupled reticulocyte lysate (Promega) andtested for binding to unphosphorylated(lanes 6,7) or CK1�-phosphorylated (lanes4,5) GST-APC-2,3. The proteins were visu-alized by Ponceau S stain in the bottompanel. (C) Phosphorylated APC-2,3 blocksbinding of �-cat to LEF-1 in EMSA experiments in vitro. Recombinant LEF-1 (�AD; 1 µM) was present in lanes 2, and 4–10, and �-cat(�N; 1 µM) was present in lanes 3–10. Where indicated, reactions also contained 2 µM (lane 5) or 12 µM (lane 6) of CK1�-phosphory-lated GST-APC-2,3, or 2 µM (lane 7) or 12 µM (lane 8) GST-APC-2,3 (unphosphorylated), or 25 µM GST (lane 9) or GST-HD2 (lane 10).(D) CK1�-phosphorylated APC blocks �-cat:LEF-1 transcription of the chromatin pBRE template in vitro. Reactions contained 100 or600 nM of CK1�-phosphorylated GST-APC-2,3 (lanes 4,5), or GST-APC-2,3 (unphosphorylated; lanes 6,7). The phosphorylated andunphosphorylated GST-APC-2,3 (100 nM) proteins were added after chromatin assembly was completed in the reactions shown inlanes 8 and 9, respectively.

Sierra et al.

594 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 10: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

�-cat:LEF-1 on DNA (Fig. 5C, lane 10), suggesting that�-cat can simultaneously bind LEF-1 and this region ofBcl-9. As expected from these results, the CK1�-phos-phorylated GST-APC-2,3 protein strongly blocked �-cat-regulated transcription in vitro (Fig. 5D, cf. lanes 3 and4,5) and also inhibited the cooperative binding of �-catand LEF-1 to chromatin in DNase I footprint experi-ments (Fig. 5D, bottom, cf. lanes 3 and 5). We concludefrom these experiments that �-cat cannot efficientlybind unphosphorylated APC, and that CK1 phosphory-lation of APC might induce high-affinity binding to �-catand trigger its dissociation from LEF-1.

Wild-type, but not mutant, APC proteins associatewith the CtBP corepressor in extracts

The colocalization of APC with CtBP at the c-Myc en-hancer during the shut-off of c-Myc transcription wasparticularly interesting in light of a recent report thatthe two proteins interact directly, both in vivo and invitro (Hamada and Bienz 2004). However, we notedthat CtBP was not present with the truncated APC atthe active c-Myc in HT29-�-Gal cells, but rather ap-peared only with the full-length APC protein, suggestingthat CtBP does not associate with truncated APC pro-teins on DNA in vivo. Consequently, coimmunoprecipi-tation experiments were carried out to examine the bind-ing of CtBP to native full-length and mutant APC pro-teins in nuclear extracts derived from 293 colorectalcancer cell lines. As shown in Figure 6A, CtBP wasreadily detected in immunoprecipitates of full-length

APC from nuclear extracts of 293 cells (lane 1), but didnot associate with the truncated APC proteins presentin nuclear extracts of HT29 (lane 2) or SW480 (lane 3)CRC cells, despite equivalent levels of CtBP in each ex-tract (lanes 4–6). We conclude that, in nuclear extracts,CtBP preferentially interacts with the full-length APCprotein.

To confirm this observation, we tested whether CtBPassociates with full-length APC protein induced in zinc-treated HT29 cells. As shown in Figure 6B (lane 4), CtBPwas readily detected in immunoprecipitates of APC-FLfrom the HT29-APC cell lines, but did not associate withthe mutant APC protein immunoprecipitated from thecontrol zinc-treated HT29-�-Galactosidase cells (lane 3),despite equal levels of CtBP in the extracts (lanes 1,2).No CtBP was detected with control IgG antiserum (Fig.6B, lanes 5,6). These data confirm the strong interactionbetween APC and CtBP reported previously by Hamadaand Bienz (2004) and extend these findings by showingthat, in nuclear extracts, CtBP does not physically asso-ciate with Class I or Class II mutant APC proteins. Wewere unable to confirm the previous mapping of theCtBP-interacting site to the APC 15-amino-acid repeatdomain, and also failed to detect CtBP in GST-pulldownexperiments with fragments of APC that contain allthree 15-amino-acid repeats (data not shown). The causeof this apparent discrepancy may lie in the stringentbinding conditions of our assays and our use of nuclear,rather than whole-cell, extracts. Taken together, theseexperiments strongly suggest that the full-length APCprotein preferentially interacts with nuclear CtBP. Thesedata suggest an important role for nuclear APC in the

Figure 6. CtBP associates with full-length APC,but not with cancer-associated truncated APC pro-teins. (A) Coimmunoprecipitation of CtBP with full-length or mutant APC proteins was analyzed by im-munoblot using nuclear extracts derived from 293Tcells, SW480, or HT29 CRC cells, as indicated aboveeach lane. (Lanes 1–3) APC was immunoprecipitatedusing an antibody specific to the N-terminal half ofthe protein, and any associated CtBP was detectedby immunoblot. The input CtBP is shown (by im-munoblot) in lanes 4–6, and the input APC proteinsare shown (by immunoblot) in lanes 7–9. (B) Newlyexpressed full-length APC in HT29-APC cells asso-ciates with CtBP. HT29-APC and HT29-�-Gal cellswere treated with 100 µM ZnCl2 to induce full-length APC expression, and association with CtBPwas determined by immunoblot of anti-APC immu-noprecipitates (lanes 1,2) or with control anti-IgGserum (lanes 5,6). The input CtBP protein in eachextract is shown in lanes 3 and 4. A schematic ofwild-type APC and the mutant HT29 APC is shownat the bottom. (S1–S3) Axin-binding sites.

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 595

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 11: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

regulation of Wnt transcription, as outlined schemati-cally in Figure 7 and discussed further below.

Discussion

A role for APC in CtBP-mediated repression of Wnttranscription in vivo.

The data presented here support a model in which theAPC tumor suppressor functions directly to counteract�-cat-mediated transcription at Wnt target genes in vivo.This possibility was first suggested by the finding thatfull-length APC cycles on and off the c-Myc enhancer inconjunction with �-cat and associated coactivators inLiCl-treated C2C12 cells (Fig. 3). In contrast, the en-hancer complex appears to be stable and does not cyclein HT29 CRC cells, which contain a Class II APC mu-tant protein that is unable to degrade �-cat (Fig. 4). Moststrikingly, the binding of the full-length APC protein tothe c-Myc gene in HT29-APC cells correlates with therapid disassembly of the Wnt enhancer complex in vivoand the subsequent decline in steady-state c-Myc mRNAlevels, both of which significantly precede the drop in�-cat protein levels that occurs as a result of proteolytic

degradation in the cytoplasm (Fig. 4). Thus, the effect ofAPC on c-Myc transcription appears to be immediateand direct, and may serve to coordinate the switch be-tween the �-cat coactivator and TLE1 corepressor com-plexes (Fig. 7).

The �-cat enhancer complex includes the Wnt coacti-vators Pygopus and Bcl-9/Lgs, which control the reten-tion of �-cat in the nucleus (Townsley et al. 2004) andmay also function directly in transcription (Hoffmansand Basler 2004; Hoffmans et al. 2005). The observationthat APC can also regulate nuclear transport of �-cat(Rosin-Arbesfeld et al. 2003) raises the possibility thatthese factors may reside within a larger regulatory com-plex that chaperones �-cat in and out of the nucleus andmediates its release from the DNA. Indeed, sequentialChIP (re-ChIP) data indicate that the mutant APC inHT29 colorectal cancer cells exists in a stable complexwith �-cat and LEF-1 at the active c-Myc gene (Fig. 4E).This finding is unexpected because �-cat cannot bindsimultaneously to APC and LEF-1, and thus, if the full-length APC is part of a larger �-cat:LEF enhancer com-plex, it may interact with other subunits. Alternatively,the full-length APC and �-cat may exist in different com-plexes that rapidly exchange at the enhancer. Our cur-rent data indicate that targeting is mediated by the N-terminal half of the APC protein, and that CtBP and�TrCP appear only in conjunction with the full-lengthAPC protein. How APC is recruited to Wnt enhancersremains an open and important question.

The ChIP experiments also suggest that APC-medi-ated inhibition of c-Myc transcription in HT29 cells oc-curs in two steps, initiated by transient binding of APC,�TrCP, CtBP, and YY1 to the enhancer, and followed bystable binding of the TLE-1 and HDAC1 corepressors.The transient recruitment of APC and CtBP, at the timewhen �-cat, Bcl-9, Pygo, and other Wnt enhancer factorsleave the DNA, strongly suggests a role for these factorsin the exchange of Wnt coactivator and corepressor com-plexes. In this respect it is interesting that CtBP wasshown recently to associate with APC, both in vivo andin vitro (Hamada and Bienz 2004). Our results confirm ahigh-affinity interaction between CtBP and the full-length APC protein induced in HT29-APC cells, as wellas with the native (full-length) APC protein in 293 cells(Fig. 6). Consequently, APC may function to recruitCtBP to Wnt enhancers (Fig. 7). Although both CtBP andTLE-1 are well-established corepressors of Wnt targetgenes, the different functions of the two types of core-pressors remain unclear, and the ChIP data suggest thatthey act at distinct steps. Together, these data suggestthat APC counteracts �-cat function in the nucleus, aswell as in the cytoplasm, and may facilitate turnover ofthe enhancer complex at responsive genes by recruiting�TrCP and CtBP.

Cancer-associated mutant APC proteins do notinteract with CtBP

Whereas CtBP interacts strongly with full-length APC, itwas not detected in immunoprecipitates of the mutant

Figure 7. Model for the role of APC in the exchange of coac-tivator and corepressor complexes at Wnt target genes. At theactive gene, �-cat interacts with Bcl-9:Pygo and with MLL1/MLL2 complexes to promote H3K4Me3. The APC tumor sup-pressor controls the switch between transcriptional coactivatorand corepressor complexes, mediated through its ability to in-teract with CtBP. Phosphorylation of APC by GSK-3� and CK1may induce APC to bind �-cat, and dislodge it from LEF-1. �-catmay then be exported from the nucleus as part of an APC com-plex, or, alternatively, it might be ubiquitinated by �TrCP fordegradation at the DNA or elsewhere in the cell.

Sierra et al.

596 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 12: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

APC proteins in SW480 or HT29 CRC cells (Fig. 6). Thesefindings contradict a recent report that CtBP binds to theAPC 15-amino-acid repeat sequences present in both full-length and mutant APC proteins (Hamada and Bienz 2004).We attribute this difference to the stringent binding con-ditions of our assays and to our use of nuclear rather thanwhole-cell extracts. The observation that only the full-length APC protein can interact with CtBP may explainwhy neither CtBP nor �TrCP were present with the mu-tant APC protein at the active c-Myc gene in vivo (Fig. 4).Importantly, the model outlined in Figure 7 strongly sup-ports the earlier conclusion from Hamada and Bienz (2004)that CtBP is not recruited to Wnt enhancers through bind-ing to the LEF-1/TCF proteins, and we speculate insteadthat CtBP and �TrCP are brought to the enhancer by APC.Interestingly, CtBP complexes contain a subunit withamine oxidase activity, LSD1, which is able to reversemono- and di-H3K4 methylation in vitro (Shi et al. 2003;Wysocka et al. 2005a), and therefore can also alter the epi-genetic state of the template. Given these observations, itwill be particularly interesting to assess whether CtBP and�TrCP are also required for the exchange of coactivator andcorepressor complexes at Wnt enhancers in vivo.

A central role for H3K4 trimethylation in�-cat-regulated transcription

To elaborate the mechanism of repression, it is impor-tant to understand how �-cat activates target genes. Thedata presented here indicate that histone H3K4 trimeth-ylation plays a key role in �-cat transactivation. Our bio-chemical studies indicate that the C-terminal activationdomain of �-cat physically associates with chromatinremodeling or modifying subunits from the TRRAP/TIP60, ISW1, and trithorax-related MLL1/MLL2 SET1-type HMT complexes (Fig. 1). SET1-type complexes me-diate trimethylation of histone H3K4, a signature chro-matin modification of highly active genes that has beenstrongly correlated with transcription elongation (for re-view, see Hampsey and Reinberg 2003). Trimethylationof H3K4 requires prior ubiquitination of H2B at K120, andis stimulated in vivo by the Bre1:Rad6 ubiquitin ligase andthe 19S proteasome ATPase subunits, Rpt4 and Rpt6 (Ezh-kova and Tansey 2004). We show that levels of H3K4Me3increase strongly upon induction of c-Myc transcription by�-cat in lithium-treated C2C12 cells (Fig. 3), and graduallydecline when the gene is repressed in HT29-APC cells. Theobservation that �-cat associates with an ISW1 complex isinteresting in light of recent data indicating that yeastIsw1p ATPases appear to remodel chromatin in a mannerthat attenuates RNAPII transcription and facilitates elon-gation in response to H3K4 trimethylation (Morillon et al.2003, 2005; Santos-Rosa et al. 2003). By inference, thesestudies suggest that �-cat regulates early stages of tran-scription elongation at target genes by recruiting chroma-tin complexes that direct H3K4 trimethylation.

Multiple roles for ubiquitin in transcription

The observation that the �-cat activation domain, whichis required for transcription in vitro (Tutter et al. 2001),

associates with H3K4 methylation factors lead us to askwhether ubiquitin is required for �-cat activity in vitro.We find that �-cat transcription of chromatin pBRE tem-plates in vitro is specifically blocked by the monoubiq-uitin-binding Vsp9 CUE domain and can be rescued byexogenous ubiquitin. The conclusion that transcriptioninitiation on chromatin requires ubiquitin supports re-cent reports that purified MLL1/SET1 complex proteinsstrongly enhance transcription initiation on chromatintemplates in vitro (Dou et al. 2005). In contrast, ubiqui-tin was not required for either initiation or elongation oftranscription on naked DNA. Although ubiquitinationof short-lived acidic activators has also been stronglylinked with transcription elongation in vivo (Murataniand Tansey 2003), our experiments were carried out witha truncated �-cat protein lacking the N-terminal regula-tory domain, suggesting that �-cat itself is unlikely to bethe target of ubiquination in these experiments. Thefinding that the transcription by the recombinant Notchenhancer complex also requires ubiquitin further sug-gests that this is a general phenomenon of transcriptionon chromatin, consistent with recent studies showing awidespread requirement for this step in regulatedRNAPII transcription activation (Dou et al. 2005; Guen-ther et al. 2005; Milne et al. 2005; Wysocka et al. 2005b).These findings warrant additional mechanistic studies toestablish whether ubiquitination of H2B enhances tran-scription initiation in vitro, and to better elucidate howubiquitination and methylation steps are coordinatedduring the early stages of transcription.

Because both GSK-3� and �TrCP are present withAPC at the c-Myc enhancer, it is possible that phos-phorylation or ubiquitination of �-cat and phosphoryla-tion of APC may be an important trigger for the disas-sembly of the enhancer complex in vivo. Phosphoryla-tion of the APC 20-amino-acid repeats by CKI� greatlyenhances binding to �-cat and disrupts its interactionwith LEF-1 (Ha et al. 2004), and blocks binding and tran-scription in vitro (Fig. 5). One possibility is that APCmay bind and transport �-cat out of the nucleus for pro-teolytic degradation in the cytoplasm. Alternatively, it ispossible that ubiquitinated �-cat may undergo proteo-lytic degradation at the DNA, as proteolysis was recentlyshown to stimulate the activity of the yeast GCN4 andGAL4 activators in vivo (Lipford et al. 2005). In HT29cells, the mutant APC protein is present at the activegene; however, �TrCP is not recruited and the enhancercomplex does not turn over, which suggests that ubiqui-tination of �-cat may not be required for transcriptionactivation, but instead may serve to promote the ex-change of coactivator and corepressor complexes, and fa-cilitate repression of transcription at Wnt target genes.Collectively, these findings suggest that there may bemultiple ubiquin-dependent steps that regulate in theprocess of activation and turnover at Wnt target genes invivo.

Lastly, the observation that both APC and �-cat enterthe nucleus to regulate Wnt target gene expression fur-ther highlights the need to assess whether the Wnt path-way evolved to directly coordinate changes in cell adhe-

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 597

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 13: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

sion, morphology, polarity, or migration with changes ingene regulation in the nucleus. Thus, �-cat is not only anuclear transcriptional coactivator, but also associateswith membrane-bound cadherins to control cell adhe-sion, and with actin microfilaments, to control cell mor-phology (Nelson and Nusse 2004; Bienz 2005; Harris andPeifer 2005). Similarly, APC controls actin cytoskeletalorganization and cell mobility through its interactionswith microtubules and F-actin (Bienz and Hamada 2004).Given the many emerging connections between actinsignaling and transcription, including the role of histonemethyltransferases in actin polymerization (Nolz et al.2005), these questions have important implications forunderstanding tissue development and tumor metasta-sis.

Materials and methods

Plasmids, recombinant proteins, and antibodies

His-tagged human Pygopus (amino acids 1–160) and His-taggedhuman Bcl-9 (amino acids 1181–1426) cDNAs were PCR-ampli-fied and cloned in-frame with a 6-histidine tag between theBamHI and HindIII restriction sites of the pET-28a(+) (Novagen)expression plasmid. The protocol for His-tagged protein purifi-cation has been described previously (Tutter et al. 2001), and theantigens were used to raise polyclonal antiserum in rabbits (Po-cono Rabbit Farm and Laboratory, Inc.). Commercial antiseraused were �-cat, APC, �TrCP, and GSK-3� (Santa Cruz Biotech-nology); MLL1, MLL2, RbBP5, Menin, Ash2L (Bethyl Laborato-ries), and H4K8Ac and H3K4Me3 (Upstate).

Cells

Mouse C2C12 and human SW480 cell lines were obtained fromthe American Type Culture Collection. C2C12 cells were cul-tured in DMEM medium supplemented with 10% FBS, andSW480 were cultured in Leibovitz L-15 medium supplementedwith 10% FBS. The human HT29-APC and HT-29-�-Galactosi-dase cell lines, which contain integrated APC and �-Gal trans-genes expressed from the MT promoter, were maintained asdescribed previously (Morin et al. 1996) and protein expressionwas induced by exposure of the cells to 100 mM ZnCl2 (Sigma)in the medium for the different times indicated in Figure 4.

Chromatin immunoprecipitation experiments

Approximately 3 × 107 to 5 × 107 C2C12 cells were plated 48 hprior to formaldehyde cross-linking and syncronized by serumstarvation. Two hours before induction, the medium was re-placed with complete DMEM. The C2C12 cells were then cul-tured in 10 mM LiCl (Sigma) for 0–2 h as indicated for eachexperiment in Figure 2. For ChIP analysis in HT29 cells, full-length APC was induced by the addition of 100 mM ZnCl2 tothe medium. The protocol for the ChIP experiments was de-scribed previously (Weinmann and Farnham 2002). QiaprepSpin Miniprep colums (Qiagen) were used for DNA purification,and immunoprecipitated DNA was analyzed using the HotStartPolymerase kit (Qiagen). PCR primers were designed to ampli-fied regions specific to the human (HT29 experiments) or mouse(C2C12 experiments) c-Myc gene. Under these conditions, thePCR product depended linearly on the amount of genomic DNAadded to the reaction. PCR products were analyzed on 1.8%agarose/TBE gels with ethidium-bromide stain. PCR of the in-

put DNA prior to immunoprecipitation was used as a control.Primers for the human c-Myc promoter monitored the thirdLEF-1 site were as described (Sasaki et al. 2003). The c-Mycprimers were human forward, 5�-GTGAATACACGTTTGCGGGTTAC-3�; human reverse, 5�-AGAGACCCTTGTGAAAAAAACCG-3�; mouse forward, 5�-CTAGAACCAATGCACAGAGC-3�; and mouse reverse, 5�-CTCCCAGGACAAACCCAAGC-3�. The sequence for negative control in mouse c-Myc promoterwas located 10 kb upstream of the TATA box: forward, 5�-ACACACCTTGAATCCCGT-3�; and reverse, 5�-CCCAGCTAGAATGAAGAAG-3�. For the re-ChIP experiments, complexes wereeluted by incubation for 30 min at 37°C in 50 µL 10 mM DTT.After centrifugation, the supernatant was diluted 20 times withDilution Buffer (1.1% Triton X-100, 1.2 mM EDTA, 167 mMNaCl, 16.7 mM Tris-Hcl at pH 8.1) and subjected again to theChIP procedure.

Quantitative PCR experiments were performed in 25-µL re-actions using the 96-well micro titer dish format and biologicalsamples were analyzed in triplicate. SYBR GREEN (Applied Bio-systems) was used as a marker for DNA amplification on an ABIPrism 7900HT apparatus and SDS 2.1 software for the dataanalysis (Applied Biosystems). Line graphs represent the relativevalue of the averaged immunoprecipitation percentage, assign-ing the value 100% to the maximum percentage of immuno-precipitation in each sample. Primers for murine c-Myc gene inC2C12 cells were Mmyc-QPCR-1F, CACACACATACGAAGGCAAAA; Mmyc-QPCR-1R, AAAAGTCGGCCCTGATCAG;and for the human c-Myc gene in HT29 cells: Hmyc-QPCR-1F,CCCAAAAAAAGGCACGGAA; Hmyc-QPCR-2R, TATTGGAAATGCGGTCATGC.

Coimmunoprecipitation and immunoblot analyses

To determine whether APC proteins associate with CtBP bycoimmunoprecipitation, aliquots of 200 µg of preclearednuclear extracts derived from 293, HT29, or SW480 cells werediluted 1:5 in Binding Buffer (20 mM HEPES at pH 7.9, 20%Glycerol, 0.5 mM DTT, 0.5 mM PMSF, 1 µg/mL leupeptin, 1µg/mL aprotinin, 0.1% NP-40), and incubated at 4°C for 90 minin successive steps with 1 µg of anti-APC (Santa Cruz Biotech-nology) and 20 µL of protein-G agarose (Santa Cruz Biotechnol-ogy), respectively. The beads were then washed with 3 × 500 µLWash Buffer (20 mM HEPES at pH 7.9, 75 mM KCl, 2.5 mMMgCl2, 1 mM DTT, 0.1% [v/v] Nonidet P-40, 0.5 mM PMSF, 1µg/mL leupeptin, 1 µg/mL aprotinin) and eluted by boiling inSDS sample buffer and analyzed by SDS-PAGE.

GST-pulldown, HMT/HAT assays, and in vitro transcriptionand binding experiments

GST-pulldown and in vitro transcription experiments were car-ried out using protocols described previously (Tutter et al.2001). For pulldown experiments from SW480CRC extracts, thenuclear extract was dialyzed against HM 0.1M (20 mM HEPESat pH 7.9, 100 mM KCl, 0.2 mM EDTA, 12.5 mM MgCl2, 10%glycerol) before use. An aliquot of 300 µL (500 µg) of nuclearextract was incubated with 700 µL of HM 0.1M, and 40 µLsuspended GST fusion protein-coupled Glutathione Sepharose4B beads (10 µg, final 100 nM), and incubated for 4 h at 4°C withrocking, washed five times in buffer (50 mM HEPES at pH 7.9,150 mM NaCl, 0.2 mM EDTA, 12.5 mM MgCl2, 40 µM ZnSO4,10% Glycerol, and 0.5% Tween20), and analyzed by SDS-PAGEfor silver-stain and immunoblot analysis, or for enzymatic ac-tivity in the HMT and HAT assays. The HMT assay was carriedout in a 50-µL reaction (final volume) in reaction buffer HM0.06M (NaCl). Half of the sample beads were incubated with

Sierra et al.

598 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 14: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

[3H]-S-Adenosyl Methionine (SAM) or [3H]Acetyl-CoA in thepresence of 10 mM ATP, and 2 µg of purified core histones for30 min at 30°C. For in vitro phosphorylation of APC, ∼30 µg ofpurified GST-APC (3 × 20-amino-acid repeat) was incubatedwith 0.5 µL (500 U) of CKI� (New England Biolabs) with orwithout 1 mM ATP in 30 µL reaction in the following finalbuffer concentration: 50 mM Tris-HCl (pH 7.5), 10 mM MgCl2,and 5 mM DTT, and incubated at 30°C for 30 min prior toanalysis by SDS-PAGE. Excised proteins were identified using aBruker Ultraflex TOF/TOF MALDI mass-spectrometer.

For the siRNA knockdown experiments, LEF-1 and �-cat tran-sient expression vectors were transfected into HeLa cellstogether with siRNAs for �-cat (5�-UCAUGCACCUUUGCGUGAGTT-3�; 5�-GCUCAUCAUACUG-GCUAGUTT-3�) orMLL2 (5�-GCUGCUAGAAUCUGCGUUCTT-3�; 5�-AUUCUGCCACGUCUGUGGATT-3�) using Oligofectamine (Invitro-gen). LiCl was added to a final concentration of 15 mM at 2 hprior to harvesting the total RNA, which was 48 h post-trans-fection, and �-cat activity was compared relative to an untrans-fected control.

Acknowledgments

We are grateful to Wolfgang Fischer and Jessica Read of thePeptide Biology Laboratory, The Salk Institute, for mass-spec-trometry identification of CTARM-interacting proteins; to LoniPickle for carrying out the CUE domain inhibition experimenton nonchromatin templates; and to Reiko Landry for expertassistance in the early stages in this project. We also thank BertVogelstein (Johns Hopkins University) for the HT29-APC cellline, Wenqing Xu (University of Washington, Seattle) for theGST-APC expression vectors, and Marianne Bienz (MRC, Cam-bridge, U.K.) and Kurt Basler (University of Zurich, Switzerland)for Pygopus and Bcl-9 cDNAs, respectively, which were used togenerate antisera. We are also grateful to M. Bienz for helpfuldiscussions and suggestions throughout the course of this work.This work was funded by NIH grants to K.A.J. (PO1CA054418and GM067127-03).

References

Baek, S.H., Kioussi, C., Briata, P., Wang, D., Nguyen, H.D.,Ohgi, K.A., Glass, C.K., Wynshaw-Boris, A., Rose, D.W., andRosenfeld, M.G. 2003. Regulated subset of G1 growth-con-trol genes in response to derepression by the Wnt pathway.Proc. Natl. Acad. Sci. 100: 3245–3250.

Barker, N., Hurlstone, A., Musisi, H., Miles, A., Bienz, M., andClevers, H. 2001. The chromatin remodelling factor Brg-1interacts with �-catenin to promote target gene activation.EMBO J. 20: 4935–4943.

Bauer, A., Huber, O., and Kemler, R. 1998. Pontin52, an inter-action partner of �-catenin, binds to the TATA box bindingprotein. Proc. Natl. Acad. Sci. 95: 14787–14792.

Bauer, A., Chauvet, S., Huber, O., Usseglio, F., Rothbacher, U.,Aragnol, D., Kemler, R., and Pradel, J. 2000. Pontin52 andreptin52 function as antagonistic regulators of �-catenin sig-nalling activity. EMBO J. 19: 6121–6130.

Bienz, M. 2002. The subcellular destinations of APC proteins.Nat. Rev. Mol. Cell Biol. 3: 328–338.

———. 2005. �-Catenin: A pivot between cell adhesion and Wntsignalling. Curr. Biol. 15: R64–R67.

Bienz, M. and Clevers, H. 2003. Armadillo/�-catenin signals inthe nucleus—Proof beyond a reasonable doubt? Nat. CellBiol. 5: 179–182.

Bienz, M. and Hamada, F. 2004. Adenomatous polyposis coliproteins and cell adhesion. Curr. Opin. Cell Biol. 16: 528–535.

Cadigan, K.M. 2002. Wnt signaling—20 years and counting.Trends Genet. 18: 340–342.

Cai, Y., Jin, J., Tomonori-Sato, C., Sato, S., Sorokina, I., Parmely,T.J., Conaway, R.C., and Conaway, J.L. 2003. Identificationof new subunits of the multiprotein mammalian TRRAP/Tip60-containing histone acetyltransferase complex. J. Biol.Chem. 278: 42733–42736.

Cai, Y., Jin, J., Florens, L., Swanson, S.K., Kusch, T., Li, B.,Workman, J.L., Washburn, M.P., Conaway, R.C., and Con-away, J.L. 2005. The mammalian YL1 protein is a sharedsubunit of the TRRAP/Tip60 histone acetyltransferase andSRCAP complexes. J. Biol. Chem. 280: 13665–13670.

Chinnadurai, G. 2002. CtBP, an unconventional transcriptionalcorepressor in development and oncogenesis. Mol. Cell 9:213–224.

Courey, A.J. and Jia, S. 2001. Transcriptional repression: Thelong and the short of it. Genes & Dev. 15: 2786–2796.

Dou, Y., Milne, T.A., Tackett, A.J., Smith, E.R., Fukuda, A.,Wysocka, J., Allis, C.D., Chait, B.T., Hess, J.L., and Roeder,R.G. 2005. Physical association and coordinate function ofthe H3 K4 methyltransferase MLL1 and the H4 K16 acetyl-transferase MOF. Cell 121: 873–885.

Ezhkova, E. and Tansey, W.P. 2004. Proteasomal ATPases linkubiquitylation of histone H2B to methylation of histone H3.Mol. Cell 13: 435–442.

Feng, Y., Lee, N., and Fearon, E.R. 2003. TIP49 regulates�-catenin-mediated neoplastic transformation and T-cellfactor target gene induction via effects on chromatin remod-eling. Cancer Res. 63: 8726–8734.

Gregorieff, A. and Clevers, H. 2005. Wnt signaling in the intes-tinal epithelium: From endoderm to cancer. Genes & Dev.19: 877–890.

Guenther, M.G., Jenner, R.G., Chevalier, B., Nakamura, T.,Croce, C.M., Canaani, E., and Young, R.A. 2005. Global andHox-specific roles for the MLL1 methyltransferase. Proc.Natl. Acad. Sci. 102: 8603–8608.

Ha, N.C., Tonozuka, T., Stamos, J.L., Choi, H.J., and Weis, W.I.2004. Mechanism of phosphorylation-dependent binding ofAPC to �-catenin and its role in �-catenin degradation. Mol.Cell 15: 511–521.

Hamada, F. and Bienz, M. 2004. The APC tumor suppressorbinds to C-terminal binding protein to divert nuclear�-catenin from TCF. Dev. Cell 7: 677–685.

Hampsey, M. and Reinberg, D. 2003. Tails of intrigue: Phos-phorylation of RNA polymerase II mediates histone meth-ylation. Cell 113: 429–432.

Harris, T.J. and Peifer, M. 2005. Decisions, decisions: �-Cateninchooses between adhesion and transcription. Trends CellBiol. 15: 234–237.

Hecht, A., Vleminckx, K., Stemmler, M.P., van Roy, F., andKemler, R. 2000. The p300/CBP acetyltransferases functionas transcriptional coactivators of �-catenin in vertebrates.EMBO J. 19: 1839–1850.

Henderson, B.R. and Fagotto, F. 2002. The ins and outs of APCand �-catenin nuclear transport. EMBO Rep. 3: 834–839.

Hoffmans, R. and Basler, K. 2004. Identification and in vivo roleof the Armadillo-Legless interaction. Development 131:4393–4400.

Hoffmans, R., Stadeli, R., and Basler, K. 2005. Pygopus and le-gless provide essential transcriptional coactivator functionsto armadillo/�-catenin. Curr. Biol. 15: 1207–1211.

Jin, J., Cai, Y., Yao, T., Gottschalk, A.J., Florens, L., Swanson,S.K., Gutierrez, J.L., Coleman, M.K., Workman, J.L., Mushe-

APC and CtBP at Wnt target genes

GENES & DEVELOPMENT 599

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 15: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

gian, A., et al. 2005. A mammalian chromatin remodelingcomplex with similarities to the yeast INO80 complex. J.Biol. Chem. 280: 41207–41212.

Kim, J., Hake, S.B., and Roeder, R.G. 2005a. The human homo-logue of yeast BRE1 functions as a transcriptional coactiva-tion through direct activator interactions. Mol. Cell 20: 759–770.

Kim, J.H., Kim, B., Cai, L., Choi, H.J., Ohgi, K.A., Tran, C.,Chen, C., Chung, C.H., Huber, O., Rose, D.W., et al. 2005b.Transcriptional regulation of metastasis suppressor gene byTip60 and �-catenin complexes. Nature 434: 921–926.

Kinzler, K.W. and Vogelstein, B. 1996. Lessons from hereditarycolorectal cancer. Cell 87: 159–170.

Lima, C.D. 2003. CUE’d up for monoubiquitin. Cell 113: 554–556.

Lipford, J.R., Smith, G.T., Chi, Y., and Deshaies, R.J. 2005. Aputative stimulatory role for activator turnover in gene ex-pression. Nature 438: 113–116.

Milne, T.A., Dou, Y., Martin, M.E., Brock, H.W., Roeder, R.G.,and Hess, J.L. 2005. MLL associates specifically with a sub-set of transcriptionally active target genes. Proc. Natl. Acad.Sci. 102: 14765–14770.

Miyagishi, M., Fujii, R., Hatta, M., Yoshida, E., Araya, N., Naga-fuchi, A., Ishihara, S., Nakajima, T., and Fukamizu, A. 2000.Regulation of Lef-mediated transcription and p53-dependentpathway by associating �-catenin with CBP/p300. J. Biol.Chem. 275: 35170–35175.

Moon, R.T., Kohn, A.D., De Ferrari, G.V., and Kaykas, A. 2004.WNT and �-catenin signalling: Diseases and therapies. Nat.Rev. Genet. 5: 691–701.

Morillon, A., Karabetsou, N., O’Sullivan, J., Kent, N., Proud-foot, N., and Mellor, J. 2003. Isw1 chromatin remodelingATPase coordinates transcription elongation and termina-tion by RNA polymerase II. Cell 115: 425–435.

Morillon, A., Karabetsou, N., Nair, A., and Mellor, J. 2005. Dy-namic histone methylation on histone H3 defines the regu-latory phase of gene transcription. Mol. Cell 18: 723–734.

Morin, P.J., Vogelstein, B., and Kinzler, K.W. 1996. Apoptosisand APC in colorectal tumorigenesis. Proc. Natl. Acad. Sci.93: 7950–7954.

Muratani, M. and Tansey, W.P. 2003. How the ubiquitin-pro-teasome system controls transcription. Nat. Rev. Mol. CellBiol. 4: 192–201.

Nakamura, T., Mori, T., Tada, S., Krajewski, W., Rozovskaia, T.,Wassell, R., Dubois, G., Mazo, A., Croce, C.M., and Canaani,E. 2002. ALL-1 is a histone methyltransferase that assemblesa supercomplex of proteins involved in transcriptional regu-lation. Mol. Cell 10: 1119–1128.

Nelson, W.J. and Nusse, R. 2004. Convergence of Wnt,�-catenin, and cadherin pathways. Science 303: 1483–1487.

Neufeld, K.L., Nix, D.A., Bogerd, H., Kang, Y., Beckerle, M.C.,Cullen, B.R., and White, R.L. 2000a. Adenomatous polyposiscoli protein contains two nuclear export signals and shuttlesbetween the nucleus and cytoplasm. Proc. Natl. Acad. Sci.97: 12085–12090.

Neufeld, K.L., Zhang, F., Cullen, B.R., and White, R.L. 2000b.APC-mediated downregulation of �-catenin activity in-volves nuclear sequestration and nuclear export. EMBO Rep.1: 519–523.

Nolz, J.C., Gomez, T.S., and Billadeau, D.D. 2005. The Ezh2methyltransferase complex: Actin up in the cytosol. TrendsCell Biol. 15: 514–517.

Radtke, F. and Clevers, H. 2005. Self-renewal and cancer of thegut: Two sides of a coin. Science 307: 1904–1909.

Reya, T. and Clevers, H. 2005. Wnt signalling in stem cells andcancer. Nature 434: 843–850.

Rosin-Arbesfeld, R., Townsley, F., and Bienz, M. 2000. The APCtumour suppressor has a nuclear export function. Nature406: 1009–1012.

Rosin-Arbesfeld, R., Cliffe, A., Brabletz, T., and Bienz, M. 2003.Nuclear export of the APC tumour suppressor controls�-catenin function in transcription. EMBO J. 22: 1101–1113.

Santos-Rosa, H., Schneider, R., Bernstein, B.E., Karabetsou, N.,Morillon, A., Weise, C., Schreiber, S.L., Mellor, J., andKouzarides, T. 2003. Methylation of histone H3 K4 mediatesassociation of the Isw1p ATPase with chromatin. Mol. Cell12: 1325–1332.

Sasaki, T., Suzuki, H., Yagi, K., Furuhashi, M., Yao, R., Susa, S.,Noda, T., Arai, Y., Miyazono, K., and Kato, M. 2003. Lym-phoid enhancer factor 1 makes cells resistant to transform-ing growth factor �-induced repression of c-myc. Cancer Res.63: 801–806.

Schneider, J., Wood, A., Lee, J.S., Schuster, R., Dueker, J., Magu-ire, C., Swanson, S.K., Florens, L., Washburn, M.P., and Shi-latifard, A. 2005. Molecular regulation of histone H3 tri-methylation by COMPASS and the regulation of gene ex-pression. Mol. Cell 19: 849–856.

Shi, Y., Sawada, J., Sui, G., Affar el, B., Whetstine, J.R., Lan, F.,Ogawa, H., Luke, M.P., Nakatani, Y., and Shi, Y. 2003. Co-ordinated histone modifications mediated by a CtBP co-re-pressor complex. Nature 422: 735–738.

Tolwinski, N.S. and Wieschaus, E. 2004. A nuclear escort for�-catenin. Nat. Cell Biol. 6: 579–580.

Townsley, F.M., Cliffe, A., and Bienz, M. 2004. Pygopus andLegless target Armadillo/�-catenin to the nucleus to enableits transcriptional co-activator function. Nat. Cell Biol. 6:626–633.

Tutter, A.V., Fryer, C.J., and Jones, K.A. 2001. Chromatin-spe-cific regulation of LEF-1-�-catenin transcription activationand inhibition in vitro. Genes & Dev. 15: 3342–3354.

Weinmann, A.S. and Farnham, P.J. 2002. Identification of un-known target genes of human transcription factors usingchromatin immunoprecipitation. Methods 26: 37–47.

Wysocka, J., Milne, T.A., and Allis, C.D. 2005a. Taking LSD 1 toa new high. Cell 122: 654–658.

Wysocka, J., Swigut, T., Milne, T.A., Dou, Y., Zhang, X., Bur-lingame, A.L., Roeder, R.G., Brivanlou, A.H., and Allis, C.D.2005b. WDR5 associates with histone H3 methylated at K4and is essential for H3 K4 methylation and vertebrate devel-opment. Cell 121: 859–872.

Xing, Y., Clements, W.K., Kimelman, D., and Xu, W. 2003.Crystal structure of a �-catenin/axin complex suggests amechanism for the �-catenin destruction complex. Genes &Dev. 17: 2753–2764.

Xing, Y., Clements, W.K., Le Trong, I., Hinds, T.R., Stenkamp,R., Kimelman, D., and Xu, W. 2004. Crystal structure of a�-catenin/APC complex reveals a critical role for APC phos-phorylation in APC function. Mol. Cell 15: 523–533.

Yokoyama, A., Wang, Z., Wysocka, J., Sanyal, M., Aufiero, D.J.,Kitabayashi, I., Herr, W., and Cleary, M.L. 2004. Leukemiaproto-oncoprotein MLL forms a SET1-like histone methyl-transferase complex with menin to regulate Hox gene ex-pression. Mol. Cell. Biol. 24: 5639–5649.

Zhu, B., Zheng, Y., Pham, A.-D., Mandal, S., Erdjument-Bro-mage, H., Temptst, P., and Reinberg, D. 2005. Monoubiqui-tination of human histone H2B: The factors involved andtheir roles in HOX gene regulation. Mol. Cell 20: 601–611.

Sierra et al.

600 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from

Page 16: The APC tumor suppressor counteracts -catenin activation ...genesdev.cshlp.org/content/20/5/586.full.pdf · The APC tumor suppressor counteracts-catenin activation and H3K4 methylation

10.1101/gad.1385806Access the most recent version at doi: 20:2006, Genes Dev. 

  Jose Sierra, Tomonori Yoshida, Claudio A. Joazeiro, et al.   methylation at Wnt target genes

-catenin activation and H3K4βThe APC tumor suppressor counteracts

Related Content

  Genes Dev. March , 2006 20: 637-642

Yue Xiong and Yojiro KotakeNo exit strategy? No problem: APC inhibits ²-catenin inside the nucleus

  References

  http://genesdev.cshlp.org/content/20/5/586.full.html#related-urls

Articles cited in: 

http://genesdev.cshlp.org/content/20/5/586.full.html#ref-list-1This article cites 62 articles, 26 of which can be accessed free at:

  License

ServiceEmail Alerting

  click here.right corner of the article or

Receive free email alerts when new articles cite this article - sign up in the box at the top

Cold Spring Harbor Laboratory Press

Cold Spring Harbor Laboratory Press on August 29, 2020 - Published by genesdev.cshlp.orgDownloaded from


Recommended