Europe PMC
Updates for full text publications in Europe PMC are currently unavailable due to planned maintenance on 3-4 June 2024. For further information contact Europe PMC Helpdesk.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Wnt/β-catenin-dependent activation of lymphoid enhancer factor 1 (Lef-1) plays an important role in numerous developmental processes. In this context, transcription of the Lef-1 gene is increased by Wnt-mediated TCF4/β-catenin activation on the Lef-1 promoter through mechanisms that remain poorly defined. In mouse airway submucosal gland progenitor cells, Wnt3A transiently induces Lef-1 gene expression, and this process is required for epithelial cell proliferation and glandular morphogenesis. In the present study, we sought to identify additional candidate transcriptional regulators of the Lef-1 gene during glandular morphogenesis. To this end, we found that Sox17 expression is dramatically downregulated in early glandular progenitor cells that induce Lef-1 expression. Wnt stimulation of undifferentiated primary airway epithelial cells induced similar changes in Sox17 and Lef-1 expression. Reporter assays revealed that ectopic expression of Sox17 suppresses Wnt3A/β-catenin activation of the Lef-1 promoter in cell lines. EMSA and ChIP analyses defined several Sox17- and TCF4-binding sites that collaborate in transcriptional control of the Lef-1 promoter. More specifically, Sox17 bound to four sites in the Lef-1 promoter, either directly or indirectly through TCF complexes. The DNA- or β-catenin-binding domains of Sox17 controlled context-specific binding of Sox17/TCF complexes on the Lef-1 promoter. Combinatorial site-directed mutagenesis of Sox17- or TCF-binding sites in the Lef-1 promoter demonstrated that these sites control Wnt/β-catenin-mediated induction and/or repression. These findings demonstrate for the first time that Sox17 can directly regulate Wnt/β-catenin-dependent transcription of the Lef-1 promoter and reveal new context-dependent binding sites in the Lef-1 promoter that facilitate protein-protein interactions between Sox17 and TCF4.

Free full text 


Logo of ajplungLink to Publisher's site
Am J Physiol Lung Cell Mol Physiol. 2010 Nov; 299(5): L694–L710.
Published online 2010 Aug 27. https://doi.org/10.1152/ajplung.00140.2010
PMCID: PMC2980392
PMID: 20802155

Sox17 modulates Wnt3A/β-catenin-mediated transcriptional activation of the Lef-1 promoter

Abstract

Wnt/β-catenin-dependent activation of lymphoid enhancer factor 1 (Lef-1) plays an important role in numerous developmental processes. In this context, transcription of the Lef-1 gene is increased by Wnt-mediated TCF4/β-catenin activation on the Lef-1 promoter through mechanisms that remain poorly defined. In mouse airway submucosal gland progenitor cells, Wnt3A transiently induces Lef-1 gene expression, and this process is required for epithelial cell proliferation and glandular morphogenesis. In the present study, we sought to identify additional candidate transcriptional regulators of the Lef-1 gene during glandular morphogenesis. To this end, we found that Sox17 expression is dramatically downregulated in early glandular progenitor cells that induce Lef-1 expression. Wnt stimulation of undifferentiated primary airway epithelial cells induced similar changes in Sox17 and Lef-1 expression. Reporter assays revealed that ectopic expression of Sox17 suppresses Wnt3A/β-catenin activation of the Lef-1 promoter in cell lines. EMSA and ChIP analyses defined several Sox17- and TCF4-binding sites that collaborate in transcriptional control of the Lef-1 promoter. More specifically, Sox17 bound to four sites in the Lef-1 promoter, either directly or indirectly through TCF complexes. The DNA- or β-catenin-binding domains of Sox17 controlled context-specific binding of Sox17/TCF complexes on the Lef-1 promoter. Combinatorial site-directed mutagenesis of Sox17- or TCF-binding sites in the Lef-1 promoter demonstrated that these sites control Wnt/β-catenin-mediated induction and/or repression. These findings demonstrate for the first time that Sox17 can directly regulate Wnt/β-catenin-dependent transcription of the Lef-1 promoter and reveal new context-dependent binding sites in the Lef-1 promoter that facilitate protein-protein interactions between Sox17 and TCF4.

Keywords: lymphoid enhancer factor 1, submucosal glands, airway, development, T cell factor

the development of numerous bud-forming epithelial organs is regulated through epithelial/mesenchymal interactions that stimulate signal transduction cascades and the transcription of genes important to epithelial cell proliferation, migration, and differentiation. The Wnt/β-catenin/lymphoid enhancer factor 1 (Lef-1) signaling pathway is one of the most extensively studied transcription cascades in this regard (9, 34, 44). In this context, canonical Wnt proteins bind to transmembrane-frizzled receptors and activate signaling cascades by stabilizing β-catenin and its interactions with transcription factors of the TCF/Lef-1 family. Following translocation to the nucleus, β-catenin/Lef-1 and β-catenin/TCF complexes modulate target-gene transcription by assembling multiprotein complexes (9, 51).

It is widely recognized that spatial and temporal control of TCF/Lef-1 activation during both organogenesis and carcinogenesis is influenced by multiple signals (Sox, E-cadherin, TGF-β, Notch, SHH, and Dkk1) that positively or negatively control the action of secreted Wnts. In this context, Lef-1-mediated Wnt signaling is required for the morphogenesis of numerous bud-forming epithelial organs such as hair follicles, teeth, mammary glands, and submucosal glands (7, 911, 14, 20, 21, 27, 32, 44). Although the most widely studied level of Lef-1 regulation by Wnt proteins involves posttranscriptional control by activated β-catenin, it is recognized that Wnts also mediate transcriptional control of the Lef-1 promoter both during developmental processes such as submucosal gland formation and under pathological conditions such as colon cancer (10, 11, 17, 24, 30, 32, 46). In this context, a Wnt-responsive element (WRE) and TCF binding sites in the Lef-1 promoter play important roles in Wnt-mediated transcriptional activation (5, 6, 11, 17, 30). The Lef-1 promoter WRE is required for Wnt3A responsiveness in cell lines and also confers temporal and spatial control of expression in developing vibrissa/hair follicles and submucosal glands in mice (10, 11, 17, 32). Additionally, several TCF binding sites residing upstream of WRE in the Lef-1 promoter have been suggested to be critical in β-catenin-dependent activation of the Lef-1 promoter in colon cancers (3, 5, 18, 23, 30).

Submucosal glands (SMG) in the conducting airways play important roles in both normal lung function and innate immunity. These structures may also serve as a stem/progenitor cell niche in the proximal airways (8, 15, 31). Wnt3A-mediated transcriptional induction of Lef-1 gene expression in glandular progenitor cells is required for proliferative signals that facilitate glandular morphogenesis (10, 11, 13, 14, 16, 17, 31). Transgenic mice harboring a 2.5-kb Lef-1 promoter segment controlling expression of a reporter have demonstrated that transcriptional induction of the promoter within glandular progenitor cells requires a 110-bp WRE and Wnt3A (10, 11, 17).

The mechanism that controls transcription of the Lef-1 promoter in gland progenitor cells remains unclear; however, studies in other trophic units of the lung have lent insights into the potential mechanism. For example, Sox17 (an SRY-related HMG box transcription factor) has been shown to influence both proliferation and differentiation of bronchiolar and respiratory epithelial progenitor cells in the distal airways (29, 36, 37). In other organ systems, Sox17 can either activate or inhibit Wnt signals through its interactions with β-catenin and TCF family members (28, 41, 42). Wnt3A-mediated activation of the Lef-1 promoter in cell lines appears to involve derepression at the WRE (17), suggesting that antagonists of Wnt signaling might be involved; Sox17 is well recognized as an antagonist of Wnt signaling, and in the context of breast and colorectal cancers, Sox17 inactivation leads to elevated Wnt/TCF/Lef-1 signaling and proliferation (18, 41, 53). Aberrant activation of Lef-1 gene transcription is also known to play an important role in colorectal cancers (23, 45, 46). Given these functional relationships among Sox17, Wnt, and TCF/Lef-1, we sought to investigate whether Sox17 might act as a modulator of Wnt-mediated activation of the Lef-1 promoter. Importantly, candidate Sox consensus binding sequences have previously been identified within the Lef-1 promoter (17).

Localization of Sox17 and Lef-1 expression in developing SMGs revealed that Sox17 expression was downregulated in glandular progenitors that induced Lef-1 expression. With the hypothesis that Sox17 represses transcription from the Lef-1 promoter to modulate Wnt inducibility, we went on to demonstrate that Sox17 expression inhibited Wnt3A/β-catenin-mediated activation of the Lef-1 promoter in each of several cell lines tested, and that this inhibition required the Sox17 HMG domain. Sox17 associates with four sites in the Lef-1 promoter. On three of these sites, Sox17 directly binds to DNA via its HMG domain, in one case competing for occupancy with TCF and in another case forming a ternary complex with TCF/β-catenin through the Sox17 β-catenin binding domain. Sox17 indirectly binds the fourth site via its interactions with TCF/β-catenin. Mutational analysis of Sox17/TCF binding sites within the Lef-1 promoter suggested that multiple sites coordinate both baseline and Wnt/β-catenin inducible activity of the promoter. Based on these data, we propose a model whereby the ratio of Sox17 vs. TCF binding to the Lef-1 promoter controls its transcription in a β-catenin-dependent and -independent fashion. Such a mechanism might be important for the modulation of Lef-1 expression in the context of Wnt/β-catenin signaling during development and/or disease.

MATERIALS AND METHODS

Lef-1 and Sox17 immunostaining.

Immunofluorescence staining was conducted on 6-μm frozen sections of ferret trachea using a rabbit anti-mouse Lef-1 antibody (10) and a goat anti-human Sox17 antibody (R&D Systems, Minneapolis, MN). The sections were fixed in 4% paraformaldehyde at room temperature for 20 min, followed by incubation in 0.2% Triton X-100 PBS solution at room temperature for 20 min. The sections were then incubated with blocking buffer (5% donkey serum in PBS) at room temperature for 1 h before the mixture of primary antibodies was applied (1:1,000 dilution for the anti-Lef-1 antibody and 1:200 dilution for the anti-Sox17 antibody) in blocking buffer at 4°C overnight. Following washing, a mixture of secondary antibodies (FITC-labeled donkey anti-goat IgG and Texas Red-labeled donkey anti-rabbit IgG; Jackson ImmunoResearch Laboratories, West Grove, PA; in blocking buffer, each diluted 1:250) was applied at room temperature for 2 h. The sections were then washed and mounted in Vectashield Mounting Medium containing DAPI (H-1200; Vector Laboratories, Burlingame, CA). Similar procedures were also used to immunohistochemically stain paraffin sections for Lef-1 and/or Sox17. Paraffin sections were deparaffinized, followed by incubation in methanol containing 0.3% H2O2 for 30 min to inactivate endogenous peroxidase. Sections were then rehydrated, and antigen retrieval was performed by boiling in citrate buffer for 20 min followed by slow cooling. The sections were then blocked with blocking buffer (5% serum in PBS) at room temperature for 2 h before incubating with rabbit anti-Lef-1 (clone C12A5, Cell Signaling Technology, 1:250) and/or guinea pig anti-Sox17 (29) (a kind gift from Dr. Whitsett, 1:1,000) in blocking buffer at 4°C overnight. Following washing, a mixture of biotinylated donkey anti-rabbit (Vector Labs, 1:250) and/or peroxidase-labeled anti-guinea pig (Jackson Laboratories, 1:500) secondary antibodies was applied at room temperature for 2 h. The Lef-1 protein was detected with ABC or Elite ABC kits (Vector Labs) and developed with Alkaline phosphatase Red substrate (for double staining) or a DAB peroxidase substrate kit (for single staining). The Sox17 protein was detected with a DAB peroxidase substrate kit (Vector Labs).

Plasmid constructs.

The Wnt3A expression plasmids were a generous gift from Dr. R. Nusse. The Lef-1 promoter-luciferase reporter, LF−2700/−200-Luc, was previously described (1, 43) and encompassed −2,700 to −200 bp of the promoter relative to the translational start site of the Lef-1 protein (17). The S37A-β-catenin and mouse Sox17 (wild-type; TAD, β-catenin-binding domain deleted; t-Sox17, HMG domain deleted; Sox17mut1, HMG domain mutant M76A; and Sox17mut2, HMG domain mutant G103R) expression plasmids were previously described (41). The plasmid pUseTCF4 and the reporter TCF-responsive TOP-flash were purchased from Upstate Biotech. The TOP-flash reporter consists of tandem TCF/LEF-1 binding sites upstream of a minimal cfos promoter. The Lef-1-expressing plasmid was previously described by our laboratory (14).

Cell lines and transient transfection experiments.

The HEK293T, MCF-7, A549, SW480, and L cell cell lines were all purchased from American Type Culture Collection and grown in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin. Wnt3A conditioned medium and control medium were harvested from Wnt3A-expressing L cells and control L cells, using medium with and without FBS (50). Cells were transfected using Lipofectamine-LTX (Invitrogen Life Technologies) according to the manufacturer's instructions or by electroporation for larger-scale analyses. Cells were grown to ~50–70% confluency before transfection. Unless otherwise specified, transient transfection reporter assays were performed in 24-well plates and utilized 0.2 μg of the Lef-1 promoter-luciferase reporter and 0.01 μg of the pCMV-Renilla luciferase plasmid (Promega, Madison, WI) as an internal control for transfection efficiency. For the TOP-flash reporter experiments, 24-well plates were utilized, and cells were transfected with 0.05 μg of the TOP-flash reporter and 0.01 μg of the pCMV-Renilla luciferase plasmid (internal control for normalization of transfection efficiency). In both Lef-1 promoter-luciferase reporter and TOP-flash reporter experiments evaluating the effects of other expressed proteins (0.5 μg S37A-β-catenin and/or 0.2 μg Sox17/well of a 24-well plate), a third plasmid (pcDNA3.1; Invitrogen, Carlsbad, CA) was added to the cotransfection cocktail to normalize the total amount of DNA transfected in all experimental comparisons. The transfections were carried out in serum-containing medium, and cells were harvested for the firefly and renilla luciferase assays 24–30 h after transfection unless otherwise specified. Protein was prepared for the assays by washing the cells in PBS, followed by lysis in 1× Passive Reporter Lysis Buffer (Promega). Protein concentrations were determined using the Bradford method, and all lysates were normalized to the same protein concentration using the lysis buffer. Ten micrograms of total cellular lysate was used for measurement of the relative luciferase activity units (RLU), for both firefly and renilla luciferase, using the Dual Luciferase Assay kit (Promega). Transfection efficiencies were normalized by dividing the relative firefly luciferase units by the relative renilla luciferase units. Following normalization, values were represented as RLU.

Immunoprecipitation and Western blot analyses.

Nuclear extracts and nuclear protein complexes were prepared for Western blotting and immunoprecipitation studies using the NucBuster Protein Extraction kit (Novagen, EMD Bioscience) or the Nuclear complex Co-IP kit (Active Motif, Carlsbad, CA), respectively, according to the manufacturers' recommendations. For immunoprecipitation analyses, 1,000 μg of protein was mixed with the appropriate antibodies, followed by binding to protein A or protein G agarose beads, or Nickel Dynabeads in the case of His-tagged proteins. The bound proteins were then eluted in an SDS sample buffer and resolved on SDS-polyacrylamide gels. The gels were then transferred onto Nylon C Hybond membranes, and these were blotted with appropriate antibodies. For Western blot analyses, 100 μg of nuclear complex protein was resolved by SDS-PAGE and blotted with appropriate antibodies following protein transfer to membranes. The following antibodies and sources were used for immunoprecipitation and Western blotting: antibodies against total β-catenin, active β-catenin (antibody that recognizes only β-catenin dephosphorylated on Ser37 or Thr41), Lef-1 (clone 2D12), TCF4, and GSK3-βY214 were purchased from Upstate Biotechnology. Other antibodies used in this study were anti-V5 (Invitrogen), rabbit anti-cyclin D1 (Abcam, Cambridge, MA), and goat anti-Sox17 (R&D Systems). Blots were then washed and subsequently incubated with secondary antibody (anti-IgG conjugated to HRP). The immune complexes were detected by ECL chemiluminescence or with SuperSignal (Pierce Chemical, Rockford, IL).

Induction and cell fractionation of undifferentiated primary airway epithelial cells.

Recombinant adenoviruses expressing Wnt1 or Wnt3A were generated using methods previously described (2). These recombinant adenoviral vectors (Ad.Wnt1 and Ad.Wnt3A) were used to synthesize these Wnts in HCT-116 cells. The cells were infected with 100 particles/cell, conditioned medium was collected, and Wnts were concentrated by absorption onto Blue Sepharose beads. It was necessary to concentrate the Wnts in this fashion to remove serum, which would otherwise cause primary airway epithelial cells to differentiate. Ad.BglII (empty vector control)-infected cells were used to create mock-conditioned medium. The adenovirus-containing medium was removed 16 h after infection, and the cells were washed twice with PBS before being fed a small volume of fresh medium. Conditioned medium was collected after 24 h and filtered (0.22 μm) before absorption onto Blue Sepharose beads (Amersham). Blue Sepharose beads (0.5 ml) were washed in PBS and added to 50 ml of conditioned medium. After overnight rotation of the samples at 4°C, the beads were washed in 3× PBS/1% CHAPS and stored in this solution at 4°C until use. Primary human and ferret airway epithelial cells were isolated as previously described by our laboratory and grown on plastic in a proliferative undifferentiated state (16, 33). Cells were induced by adding mock-, Wnt1-, or Wnt3A-conditioned beads to cells in culture. Thirty-six hours after the bead addition, the cells were harvested and nuclear extracts (NE) and postnuclear supernatants (PNS) were prepared using a Nuclear Extract Kit (Active Motif). Briefly, cells in a 100-mm dish were removed by gentle scraping, followed by lysis with a proprietary hypotonic lysis buffer (Active Motif). The cytoplasmic fraction (PNS) was collected after centrifuging the suspension at 14,000 g at 4°C for 30 s. The nuclear pellet was resuspended in 50 μl of proprietary complete lysis buffer (Active Motif) and incubated on ice for 30 min on a rocking platform. The sample was then vortexed for 30 s and centrifuged at 14,000 g for 10 min, after which the supernatant (NE) was transferred to a prechilled tube. Although contamination of cellular plasma membrane proteins cannot be excluded from this type of preparation, the company's lysis buffers are designed to enrich for nuclear proteins. We have previously used this method with small samples and have succeeded in enriching for nuclear and cytoplasmic proteins at >95% purity, as determined by Western blotting for H1 (nuclear) and IκBα (cytoplasmic) (52). PNS and NE were quantified for protein concentration before analysis by Western blotting for Lef-1, β-catenin, TCF-4, and Sox17.

Chromatin immunoprecipitation assays.

Three 150-mm plates of A549 cells were transfected with 30 μg of plasmid expressing either mouse Sox17 or TCF4 (41). Cells were then fixed and harvested for chromatin immunoprecipitation (ChIP) at 36 h posttransfection. Formaldehyde cross-linked DNA-protein complexes were sheared by sonication, to an average size of 400–600 bp, diluted 1:10 with the ChIP dilution buffer included in the EZChIP kit (Upstate Biologicals), and then precleared with protein A agarose beads. The precleared DNA-protein mixture was used for the ChIP assays. Ten micrograms of anti-Sox17 (R&D Systems), anti-TCF4 (Upstate Biologicals), or nonimmune control IgG antibody was added to the ChIP reaction mixture. Immunoprecipitation and DNA recovery were performed according to the EZChIP kit protocol from Upstate Biologicals. The recovered DNA was detected by PCR and quantified by real-time PCR using 3 μl of the final DNA precipitate. Ten primer sets were used to survey the 2.5-kb human Lef-1 promoter locus (see Table 3). The PCR protocol was carried out for 30 cycles, each at 95°C for 15 s and 60°C for 45 s. Real-time quantitative PCR was performed using Bio-Rad IQ SYBR Green Supermix and iCycler iQ Detection System (Bio-Rad, Hercules, CA). The relative copy number for each ChIP DNA fragment was calculated as the fold increase of the relative copy number in the immunoprecipitated fraction (with the specific capture antibody) over the relative copy number in the IgG-negative control for each sample.

Table 3.

The sequences and positions of synthetic oligonucleotides used to generate mutants of Lef-1-luciferase reporter constructs

Mutated SiteLef-1 Promoter Position, bpOligonucleotide Sequence
TLS1−2693/−26875′-CCACACTGAATTCGGCGCAGCACagggtgcAAGTTTTAAAAATTGCTTCCTAGTAAATTCTTATTCCC-3′
TLS2−2427/−24215′-GGGAGATGAAGATACATTTCTTTATGTCagggtggTACTGTTCTGAAGTTTTACAGATGG-3′
TLS3−2254/−22485′-GCTGTAACTTTCACAAAGTATCTCCTGAAAACTCCTGTagggtgcCAAAATTCATCAAAGAGACAGTGGAGACTGGGG-3′
TLS4−2240/−22345′-CCTGAAAACTCCTGTCTTTGTACAAAATTCcgaccctAGACAGTGGAGACTGGGGAATTTTTG-3′
SXS5−2956/−19515′-GTACATCCCGTGGTGAGAACAGAATGAAAGATATcggtggTAAAAAGCAATAATTAAAATCTAGTCTTCAGTTCCTTCTT-3′
TLS6−921/−9155′-GCCAGGCTGAGAAACTCGAGCCGGGccaccctAGGGGTCGGACTCAGTGTGTGTGTCGG-3′

Six putative TCF/Lef-1 and/or Sox binding sites (1–6) within the human Lef-1 promoter were selected for mutation analysis, and the mutated sequences are shown in lowercase, italics, and bold. The base position of the mutations is based on the sequence of the 2.5-kb human Lef-1 promoter relative to the start translational methionine at +1 (17).

Purification of His-tagged TCF4, Lef-1, and Sox17 mouse proteins.

Mouse cDNAs for TCF4, Lef-1, and Sox17 wild-type and mutant proteins (41) were cloned in-frame into the pET151/D-TOPO vector backbone using the Champion pET Directional TOPO Expression Kit (Invitrogen). The resultant clones were confirmed by DNA sequencing and expressed NH2-terminal His-tagged fusion proteins in bacteria. The induction of correctly sized fusion proteins in bacteria was confirmed by Western blotting, using both anti-His antibody and antibodies against the protein backbone. The His-tagged fusion proteins were purified by HPLC using a HisTrap HP column (GE Healthcare Life Sciences), and the purity was confirmed by SDS-PAGE Coomassie staining and Western blotting. Concentrations of the purified proteins were normalized using the Bradford method, and aliquots were stored at −80°C until use. Purified GST and GST-β-catenin protein were purchased from Millipore (Billerica, MA).

Non-radioactive EMSA.

To test whether Sox17, TCF4, or Lef-1 protein physically binds to putative TCF/Lef-1 and/or Sox DNA binding sites in the Lef-1 promoter, we developed a non-radioactive EMSA. This approach assessed DNA/protein interactions using purified recombinant proteins and purified 5′-biotinylated double-stranded oligonucleotides (see Table 2). Five micrograms of purified His-fusion protein(s) was preincubated for 10 min at 4°C in binding buffer containing 0.005 units of poly(dG-dC) (Sigma), 0.005 units of poly(dI-dC) (Sigma), 0.05% Igepal-CA630 (Sigma), and 2 mg/ml BSA (19). After the preincubation, 2 pmol of purified double-stranded biotinylated oligonucleotide was added to the binding mixture (final volume of 20 μl) and incubated at room temperature for an additional 20–30 min. Annealed oligonucleotides were purified using a 7.5% polyacrylamide gel in 0.5× TBE buffer, and the concentration after purification was determined by slot blotting. The EMSA binding reaction mixture was resolved by electrophoresis in a 7.5% polyacrylamide gel in 0.5× TBE buffer at low voltage. After electrophoresis, the DNA was transferred to a Nylon membrane using TBE buffer at 40 V for 1–2 h. DNA was then cross-linked to the nylon membrane by UV (120 mJ/cm2) using the “autocrosslink” function on a Stratalinker. The biotinylated oligonucleotides were detected on the membrane using a NEB Phototope-Star Detection kit and following the manufacturer's instructions (New England Biolabs).

Table 2.

The sequences of biotinylated oligonucleotides used for DNA/protein binding assays

Putative Binding SiteLef-1 Promoter Position, bpSynthetic Biotinylated Oligonucletide Sequence
TLS1 (S1)−2693/−2687Biotin-5′-GGGGATCctttgtaAAGTTTTAAAAATTGC-3′
TLS2 (S2)−2427/−2421Biotin-5′-CATTTCTTTATGTCctttgttTACTGTTCTG-3′
TLS3 (S3)−2254/−2248Biotin-5′-CTCCTGTctttgtaCAAAATTCATC-3′
TLS4 (S4)−2240/−2234Biotin-5′-CAAAATTCatcaaagAGACATGG-3′
SXS5 (S5)−2956/−1951Biotin-5′-GAAAGATATattgttTAAAAAGC-3′
TLS6 (S6)−921/−915Biotin-5′-CTCGGCCGGGaacaaagAGGGGTCGG-3′
TLS3/4 (S3/4)−2254/−2234Biotin-5′-CTCCTGTctttgtaCAAAATTCatcaaagAGACATGG-3′
TLS2mut−2427/−2421Biotin-5′-CATTTCTTTATGTCagggtggTACTGTTCTG-3′
TLS3/4mut−2254/−2234Biotin-5′-CTCCTGTagggtgcCAAAATTCcgaccctAGACATGG-3′
SXS5mut−2956/−1951Biotin-5′-AGAATGAAAGATATcggtggTAAAAAGCAA-3′
TLS6mut−921/−915Biotin-5′-CGAGCCGGGccacccctAGGGGTCGGACTCA-3′
TOP sitesNABiotin-5′-AAGatcaaagGGGGGTACCctttgatCTTAC-3′
SP1 siteNABiotin-5′-GCGTACTCTGCCCGCCCCCTCTGACTC-3′

NA, not related to Lef-1 promoter. Synthetic oligonucleotides containing the sequences of putative TCF/Lef-1 and/or Sox binding sites of the human Lef-1 promoter (Fig. 7A) were labeled at the 5′ end with biotin; the potential binding sequences are shown in lowercase, italics, and bold. The labeled oligonucleotides were annealed to the respective nonlabeled complementary synthetic strands, purified by 7% PAGE, and titrated by slot-blotting using NEB Phototope-star kit (New England Biolabs). The SP1 site served as negative TCF/Lef-1 and Sox17 binding controls. The TOP oligo was the consensus from Top-flash and served as TCF- and Lef-1-binding positive controls.

Detection of protein complexes bound to biotinylated double-stranded oligonucleotides.

To evaluate whether Sox17, TCF4, Lef-1, and β-catenin can form multiprotein complexes on DNA sequences from the Lef-1 promoter, we mixed 30 μg of purified His-tagged Sox17, TCF4, and LEF-1 proteins (~500 pmol of each) and 1.5 μg of GST-β-catenin (13 pmol) (Upstate Biologicals) in binding buffer containing 0.0025 units poly(dI-dC), 0.0025 units poly(dG-dC), 0.01 μg single-stranded (denatured) salmon sperm DNA, and 0.05% Igepal-CA630. After preincubation on ice for 10 min, 5 pmol of gel-purified biotinylated double-stranded oligonucleotides was added, and the total volume was adjusted to 200 μl with binding buffer. The molecular ratio of oligonucleotide:protein was ~1:100 for transcription factors and 1:2.5 for β-catenin. An excess of transcription factors was used to drive complex formation and allow for competitive DNA binding between transcription factors, which could only be achieved under conditions where binding was saturated. The mixture was then incubated on ice for 20–30 min, followed by the addition of 800 μl of binding buffer containing 20 μl of prewashed Streptavidin C1 DynaBeads (Invitrogen). The mixture was then gently agitated at room temperature for 30–60 min, followed by washing of the beads in a magnet with washing buffer (25 mM Tris-Cl, 150 mM NaCl, 0.1% BSA, 0.05% Tween 20, pH 7.2) five times for 3 min each. The beads were then resuspended in 100 μl 2× SDS-PAGE loading buffer and boiled for 5 min. Twenty-five microliters of the suspension was then loaded onto an SDS-PAGE gel and Western blotted with antibodies against Sox17, TCF4, Lef-1, or β-catenin.

Mutagenesis of Lef-1 promoter reporter constructs.

The QuikChange MultiSite-Directed Mutagenesis kit from Stratagene was used to generate a series of mutations in the LF−2700/−200-Luciferase Lef-1 promoter/reporter plasmids (1, 43). We systematically introduced 6- to 7-bp mutations at certain conserved binding sites for TCF/Lef-1 and/or Sox proteins by changing A→C, T→G, C→A, or G→T. A total of 11 LF−2700/−200-Luciferase mutated constructs containing six single binding-site mutations (site S1 to S6) and five combination mutations were generated. The corresponding mutations in the synthetic oligonucleotides were used to evaluate in vitro binding; the relative positions of these mutations within the human Lef-1 promoter are listed in Table 3. All mutations within LF−2700/−200-Luciferase plasmid backbone were confirmed by sequencing.

RESULTS

Expression of Lef-1 and Sox17 is dynamically regulated in airway epithelial cells during submucosal gland development.

Like humans, ferrets have an abundance of SMGs throughout their cartilaginous airways and are thus considered a good model for studying SMG development and function. Immunostaining of ferret tracheal sections for Lef-1 and Sox17 revealed dynamic changes in the expression of these two proteins during early stages of SMG morphogenesis (Fig. 1). As previously reported, Lef-1 protein expression was induced in the nuclei of cells in the gland bud, relative to the surface airway epithelium (SAE). This increase in Lef-1 expression at the gland bud stage was accompanied by a decrease in Sox17 expression (Fig. 1, A, D–H). At later stages of glandular tubulogensis, Lef-1 expression progressively declined while Sox17 expression rose (Fig. 1, B and C). These observations demonstrating reciprocal regulation of Sox17 and Lef-1 in glandular progenitor cells led to the hypothesis that a reduction in Sox17 expression may be functionally linked to Wnt induction of Lef-1 expression.

An external file that holds a picture, illustration, etc.
Object name is zh50111057590001.jpg

Lymphoid enhancer factor 1 (Lef-1) and Sox17 expression changes dynamically during the development of airway submucosal glands (SMGs). A–C: frozen sections of ferret trachea from postnatal day 1 (D1), day 2 (D2), and day 6 (D6) were stained with antibodies specific to Lef-1 (red) and Sox-17 (green). Combined and individual immunofluorescence channels are shown. DAPI was used to mark nuclei, and dashed lines indicate the basal lamina of SMG buds and tubules. D–H: using a different set of Sox17 and Lef-1 antibodies, paraffin sections of day 1 ferret tracheas were histochemically stained for Sox17 and Lef-1. Two methods of histochemical staining included staining for Sox17 and Lef-1 individually using DAB peroxidase (black) histochemistry (D, E) or costaining for Sox17 and Lef-1 using DAB peroxidase (black for Sox17) and alkaline phosphatase (red for Lef-1) histochemistry. As previously reported, Lef-1 is expressed in both the newly forming gland buds (GB) and growing edges of the cartilage (C). SAE, surface airway epithelium; GT, gland tubule.

Lef-1 expression in undifferentiated primary airway epithelial cells is induced by Wnt3A and Wnt1, and this induction correlates with a decline in expression of full-length Sox17.

Both Wnt1 and Wnt3A have the ability to induce the Lef-1 promoter in transformed cell lines (11, 17) and glandular progenitor cells (10). We evaluated whether beads conjugated to Wnt1 or Wnt3A proteins (Fig. 2A) could modulate Lef-1 and/or Sox17 expression in undifferentiated primary human and ferret tracheobronchial airway epithelial cells (AECs). Indeed, treatment with either Wnt1- or Wnt3A-conjugated beads resulted in higher levels of Lef-1 and TCF4 protein expression than did treatment with unconjugated (mock) beads (Fig. 2B). Similar findings of Wnt3A-mediated induction of Lef-1 and TCF4 were seen in both human and ferret undifferentiated primary AECs treated with conditioned medium from Wnt3A-producing L cells (Fig. 2C). In these studies, Lef-1 induction correlated with a rise in cyclin D1 expression, consistent with previous findings that Lef-1 binds to and induces the cyclin D1 promoter (40) and that Lef-1 knockout mice have defects in glandular progenitor cell proliferation including cyclin D1 expression (10). Subcellular fractionation experiments demonstrated that Wnt1 and Wnt3A induced Lef-1, but not β-catenin, in the nuclear fraction and that the ratios of full-length (F-Sox17) and HMG-deleted (t-Sox17, which does not bind DNA) isoforms of Sox17 changed in response to Wnt induction. F-Sox17 levels decreased in the presence of Wnt while t-Sox17 levels increased (Fig. 2D). Because the F-Sox17 isoform was only weakly expressed in this model system relative to t-Sox17, the migratory position of the F-Sox17 protein within the SDS-PAGE was confirmed by recombinant adenovirus overexpression of F-Sox17 in undifferentiated primary AECs (Fig. 2E). Cumulatively, these results demonstrate that Wnts can induce Lef-1 and cyclin D1 expression in primary airway epithelial cells while simultaneously reducing the expression of F-Sox17. This phenotype is similar to that observed in glandular progenitor cells at the time of bud formation (10) (Fig. 1).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590002.jpg

Wnt proteins stimulate Lef-1 expression in undifferentiated primary airway epithelial cells. A: HCT-116 cells were infected with the indicated recombinant adenoviral vectors expressing Wnt1 or Wnt3A (Ad.Wnt1 and Ad.Wnt3A) or with a control adenoviral vector (Ad.BglII) lacking a transgene. Conditioned HCT-116 medium was absorbed to Blue Sepharose beads, and 20 μl of the 50-ml conditioned medium (m) or 20 μl of the 500 μl of absorbed beads (b) were loaded in the indicated lanes and subjected to Western blotting with anti-Wnt3A or anti-Wnt1 antibodies as indicated. B: undifferentiated primary human bronchial airway epithelial cells (AECs) grown on plastic were treated with the indicated Wnts absorbed to beads. At 36 h following treatment, whole cell lysates were harvested for Western blotting with anti-Lef-1, anti-TCF-4, and anti-β-catenin antibodies (noted at right). C: conditioned medium harvested from Wnt3A-expressing transgenic L cells or from control L cells lacking the Wnt3A transgene was applied to undifferentiated primary human (left) or ferret (right) AECs for 72 h. Nuclear extracts were used for Western blotting with anti-Lef-1, anti-β-catenin, anti-TCF4, anti-cyclin D1, and anti-β-actin antibodies. D: undifferentiated primary AECs were treated with Wnt-conjugated beads. At 36 h following treatment, postnuclear supernatants (PNS) and nuclear extracts (NE) were generated and evaluated by Western blotting with anti-Lef-1, anti-Sox17, and anti-β-catenin antibodies. F-Sox17 (long isoform) and t-Sox17 (truncated short-form missing the HMG binding domain) are shown, at long and short exposure times, respectively. E: a recombinant adenoviral vector encoding the long form of Sox17 (F-Sox17) was used to demonstrate isoform migration. Lane 1, nuclear extract from mock-treated undifferentiated primary AECs (from D) for comparison of F-Sox17 and t-Sox17 expression (long exposure time); lanes 2 and 3, Ad.BglII-infected AECs; lanes 4 and 5, Ad.F-Sox17-infected AECs.

Sox17 modulates Wnt3A-mediated activation of the Lef-1 promoter.

We next tested whether Sox17 expression can modulate Wnt-mediated activation of the Lef-1 promoter, using transient transfection of 293 cells with a 2.5-kb Lef-1 promoter-driven luciferase reporter. As a control for Wnt/TCF-mediated activation, we also evaluated the well-characterized Wnt/TCF-responsive reporter TOP-flash, whose activation by β-catenin is inhibited by Sox17 (41, 54). Results from this analysis demonstrated that the Lef-1 promoter was dynamically induced by Wnt3A expression, with activity peaking at 24–48 h posttransfection and declining thereafter (Fig. 3A). Coexpression of Sox17 with Wnt3A significantly reduced Wnt3A-mediated activation of the Lef-1 promoter. Paradoxically, however, Sox17 expression alone resulted in levels of Lef-1 promoter activity above baseline (Fig. 3, A and C). In the case of the TOP-flash reporter, Sox17 expression inhibited Wnt3A-mediated activation, as previously reported for its activation by Wnt1 or dominant-activated β-catenin (41, 54); notably, the expression of Sox17 alone did not induce TOP-flash expression as had been observed for the Lef-1 promoter (Fig. 3D). Immunoblotting analysis revealed that Sox17 expression promoted the degradation of activated β-catenin (dephosphorylated on Ser37 or Thr41) in both the presence and absence of Wnt3A, without altering the level of tyrosine-phosphorylated PY-214-GSK-3β (Fig. 3B). These observations are consistent with previous findings that Sox17 can inhibit Wnt signaling by stimulating GSK3β-independent degradation of β-catenin protein in colon cancer cell lines (12, 41).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590003.jpg

Sox17 dynamically regulates the Lef-1 promoter in the presence and absence of Wnt3A. A: a 2.5-kb Lef-1 promoter-luciferase reporter (Lef-1-Luciferase) construct was used to evaluate the kinetics of promoter activation in the presence of Wnt3A and/or Sox17 expression. 293 Cells were cotransfected with 0.2 μg of Lef-1-luciferase reporter and 0.5 μg of Wnt3A and/or 0.2 μg of Sox17 expression plasmids using Lipofectamine LTX reagent. For all transfections, the total DNA concentration was identical and normalized using the empty-vector control plasmid pcDNA. All transfections also contained 0.01 μg of a pCMV-Renilla luciferase plasmid for normalization of transfection efficiency. Relative activity of the Lef-1 promoter is plotted as relative light units (RLU). B: Western blot analysis of changes in total β-catenin, active β-catenin, Sox17, and phosphorylated GSK3β at 24 h posttransfection, with the transfected plasmids indicated above the blot. C and D: comparison of Lef-1-luciferase reporter (0.2 μg) (C) and TOP-flash TCF-responsive reporter (0.05 μg) (D) activities in cells transfected with the indicated reporter and Wnt3A (0.5 μg) and/or Sox17 (0.2 μg) expression constructs. E and F: comparison of Lef-1-luciferase reporter (E) and TOP-flash TCF-responsive reporter (F) activities in cell transfected with the indicated reporters and dominant-active β-catenin-S37A (0.5 μg) and/or Sox17 (0.2 μg) expression constructs. Values in A and C–F represent the mean (± SE, n = 9) RLU normalized for Renilla luciferase expression.

To further investigate both the importance of activated β-catenin in regulating the Lef-1 promoter and the ability of Sox17 to inhibit this activation, we studied induction of the Lef-1 promoter in 293 cells expressing dominant-active S37A-β-catenin and/or Sox17. Like Wnt3A, S37A-β-catenin activated both the Lef-1 promoter and TOP-flash reporters (Fig. 3, E and F). However, the extent of Sox17-mediated repression was significantly less than that seen in cells transfected with Wnt3A (Fig. 3, C and D). These findings suggested that Sox17-mediated repression of the Wnt3A-activated Lef-1 and TOP-flash promoters likely involves additional Wnt3A-induced factors besides β-catenin.

Sox17 regulates Wnt3A-mediated activation of Lef-1 promoter in a cell type-dependent fashion.

To substantiate results in 293 cells, we performed promoter-reporter experiments in four additional cell lines: MCF7 (human mammary adenocarcinoma), SW480 (human colon adenocarcinoma), A549 (human lung carcinoma), and L cells (murine transformed fibroblast). In the MCF-7, SW480, and A549 cells, the ability of Sox17 to repress Wnt3A-mediated activation of the Lef-1 and TOP-flash promoters was similar to that seen in 293 cells (Fig. 4). In L cells, by contrast, Wnt3A failed to induce the Lef-1 promoter and only weakly induced the TOP-flash reporter; however, in the absence of a Wnt signal, Sox17 expression led to repressed baseline activities of both these promoters. Notably, none of the four cell lines demonstrated Sox17-mediated activation of the Lef-1 promoter in the absence of Wnt3A, as had been observed in 293 cells. These data suggest that the ability of Sox17 to inhibit and/or activate the Lef-1 promoter in the presence or absence of a Wnt signal is likely highly dependent on other cell type-specific factors (Fig. 4).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590004.jpg

Sox17 transcriptionally regulates Wnt3A-mediated activation of both the Lef-1 promoter and TCF-responsive reporter TOP-flash. MCF-7, SW480, A549, or L cells were cotransfected with Wnt3A- (0.5 μg) and/or Sox17- (0.2 μg) expressing plasmid(s), together with the Lef-1 promoter luciferase (0.2 μg) (left) or TOP-flash luciferase (0.05 μg) (right) reporter. The pcDNA empty plasmid was used as a negative control and for normalizing total DNA content in each transfection. Each transfection cocktail also contained 0.01 μg of CMV-Renilla luciferase as an internal control plasmid for the normalization of transfection efficiency. At 24 h posttransfection, cell lysates were prepared, and both firefly and renilla luciferase activities were determined. Bars depict the mean (± SE, n = 3) RLU normalized for Renilla luciferase expression.

Sox17 repression of Wnt-mediated Lef-1 promoter induction requires the Sox17 HMG domain.

There are multiple mechanisms by which Sox17 can modulate Wnt activity. Sox17 can interact with β-catenin on DNA to regulate transcription or it can interact with and repress the activity of β-catenin/TCF/Lef-1 independently of DNA binding (28, 41, 42, 54). We sought to determine if Sox17 directly binds to and regulates the Lef-1 promoter. We performed structure-function studies using forms of Sox17 with mutations that effect specific domain functions (Fig. 5) [i.e., β-catenin binding domain (BBD) and high-mobility group (HMG) DNA binding domain]. We analyzed the ability of four mutant forms of Sox17 (Fig. 5, A and B) to interact with Lef-1 promoter sequences, TCF/Lef-1, and β-catenin, and for their ability to regulate the Lef-1 promoter. Only forms of Sox17 that could bind to DNA were able to regulate the Lef-1 promoter, suggesting a direct effect.

An external file that holds a picture, illustration, etc.
Object name is zh50111057590005.jpg

Structural analysis of Sox17-mediated regulation of the Lef-1 promoter by Wnt3A. Various deletion and point mutants of mouse Sox17 were used to determine which domains of Sox17 are required to repress Wnt3A-mediated transcriptional induction of the Lef-1 promoter. A: schematic representation of constructs encoding the full-length Sox17 protein and various Sox17 mutants (see materials and methods). All of the constructs contained a V5-tag at the COOH terminus. B: 293 cells were transfected with each of these constructs, and expression was evaluated by Western blotting with anti-V5 antibody at 24 h posttransfection. C: anti-V5 antibody was used to immunoprecipitate Sox17 from 293 cells transfected with the expression constructs shown in A, followed by Western blotting with anti-Sox17, anti-Lef-1, anti-TCF4, and anti-β-catenin antibodies. Nuclear extracts (500 μg) were used for IP reactions at 48 h posttransfection. The input lane represents direct loading of nuclear extract from full-length Sox17-transfected cells, whereas the No V5 lane omitted the capture V5 antibody from the IP reaction for full-length Sox17-transfected cells. D: analysis of the functions of different Sox17 domains in Wnt3A-mediated activation of Lef-1 promoter (top) and TCF-responsive reporter TOP-flash (bottom). 293 Cells were cotransfected with the Wnt3A (0.5 μg) expression plasmid and either the Lef-1 promoter (0.2 μg) or TOP-flash (0.05 μg) reporter plasmid, together with one of the indicated Sox17 (0.2 μg) expression plasmids. All transfections also contained 0.01 μg of a pCMV-Renilla luciferase plasmid for normalization of transfection efficiency. Bars depict the mean (± SE, n = 4) RLU at 24 h posttransfection and were normalized for Renilla luciferase expression.

As previously reported (41), full-length F-Sox17 associated with both endogenous β-catenin and TCF4 following immunoprecipitation of Sox17, and each of these mutants exhibited the predicted defects in associating with β-catenin or TCF4 (Fig. 5C). The TAD mutant lacked the ability to bind β-catenin but bound Lef-1 and TCF4, whereas the t-Sox17, Mut1, and Mut2 HMG binding domain mutants failed to bind Lef-1 or TCF4 but bound β-catenin (Fig. 5C). These findings substantiate previous results demonstrating that Sox17 can bind β-catenin and also associate with TCF4/Lef-1 through its HMG domain (41, 42, 54).

We next sought to evaluate whether Sox17/β-catenin and/or Sox17/TCF4 interactions are important for the ability of Sox17 to repress Wnt3A induction of the Lef-1 promoter in 293 cells. To distinguish between direct effects of Sox17 on the Lef-1 promoter and indirect effects on Wnt signaling, we also evaluated the Wnt/TCF-responsive TOP-flash reporter. One key difference was that the HMG domain of Sox17 differentially affected activation of the Lef-1 and Top-Flash promoters in the presence of Wnt3A (Fig. 5D). Wnt3A-mediated transcriptional activation of the Lef-1 promoter was not repressed by either the t-Sox17 or Mut1 protein, and repression by Mut2 was marginal compared with that effected by F-Sox17. By contrast, Sox17 Mut1 and Mut2 both significantly enhance TOP-flash activation in the presence of Wnt3A. Although Mut2 was previously shown to effect a similar enhancement of β-catenin-S37A-mediated TOP-flash in COS cells (41), Mut1 completely repressed β-catenin-S37A-mediated TOP-flash activation under the same conditions (41). Sox17/β-catenin interactions also contributed to repression of Wnt3A activation of both the Lef-1 and Top-flash promoters to varying extents, as the TAD mutant retained only partial activity compared with F-Sox17. Thus, context-dependent differences in the function of the Sox17 HMG binding domain appear to influence Wnt/β-catenin activation of both the Lef-1 and TOP-flash promoters.

Sox17 and TCF4 physically bind to the endogenous Lef-1 promoter in A549 cells.

We next performed ChIP assays to identify the regions of Sox17 and TCF4 binding to the Lef-1 promoter (Fig. 6). We utilized a lung carcinoma cell line (A549 cells) since this cell type is most relevant to our interest of Lef-1 regulation in airway epithelia. A549 cells were transfected with plasmids encoding V5-tagged-Sox17 or TCF4 before ChIP analysis. Immunoprecipitation was carried out using anti-V5 and anti-TCF4 primary antibodies (or nonimmune isotype-matched IgG alone as a negative control), and 10 oligonucleotide primer sets spanning the 2.5-kb Lef-1 promoter sequence were used for PCR amplification from these samples (Table 1 and Fig. 6A). Results from this analysis indicated that the endogenous Lef-1 promoter contains multiple Sox17 and TCF4 binding sites (Fig. 6, B and C). The identified binding regions overlapped with candidate consensus sequences for either Sox binding sites (SXS) or TCF/Lef-1 binding sites (TLS) (termed TLS1, TLS2, TLS3/4, SXS5, and TLS6; sites are numbered in sequence from the 5′ → 3' end of the promoter) (Fig. 6A). Quantitative PCR results demonstrated that Sox17 strongly bound to regions P2 (containing TLS2), P4 (containing SXS5), and P7 [containing TLS6 and the previously identified WRE (17)] of the Lef-1 promoter, with weaker binding at P1 (TLS1) (Fig. 6, B and C); notably, only one of these regions (P4) contained a Sox consensus site (SXS5). By contrast, TCF4 bound most strongly to the P3 region (TLS3/4) of the Lef-1 promoter and more weakly to the P1 (TLS1) and P7 (TLS6) regions. These finding demonstrate that both Sox17 and TCF4 can physically interact with the Lef-1 promoter.

An external file that holds a picture, illustration, etc.
Object name is zh50111057590006.jpg

ChIP survey of Sox17 and TCF4 binding sites in the endogenous Lef-1 promoter of A549 cells. A: schematic representation of the 10 PCR fragments used to survey the 2.5-kb human Lef-1 promoter (Table 3). Each PCR fragment covered 250–400 bp of the promoter. B and C: A549 cells were transfected with mouse Sox17- or TCF4-expressing plasmid, and genomic DNA sheared to an average size of 400–600 bp before ChIP was performed with anti-Sox17 and TCF4 antibodies. The recovered DNA was then evaluated by either standard PCR (B) or real-time PCR (C). B: agarose gel images show PCR products following 30 cycles of amplification from DNA immunoprecipitated with anti-Sox17 antibody (top), anti-TCF4 antibody (middle), or nonimmune total IgG from the same species as the primary capture antibody (bottom; used as a negative control). C: quantitative PCR results from the ChIP assays. Values depict the fold increase in copies of each target sequence detected with the specific capture antibody over the number detected with the species-matched IgG control antibody alone. Data are representative of 3 independent experiments.

Table 1.

Sequences of primer sets for ChIP assays of the 2.5-kb human Lef-1 promoter

Primer SetLef-1 Promoter Region Amplified By PrimersPrimer Sequence
P1−2781 bp to −2444 bpFwd: 5′-CCTTCACCAACCCCTTAATTTCACCCC-3′
Rev: 5′-CTTCATCTCCCTTAAATCGAAAACAAGC-3′
P2−2584 bp to −2280 bpFwd: 5′-CTTTACCCATTCAATGTTCAGCAGCCTG-3′
Rev: 5′-GTGAAAGTTACAGCAGAAGGGAAGGCTG-3′
P3−2306 bp to −1991 bpFwd: 5′-CAGCTTCCCTTCTGCTGTAACTTTCAC-3′
Rev: 5′-GAGAGTTGTGCTAGGTCTCCAGTGGG-3′
P4−2017 bp to −1656 bpFwd: 5′-CCCACTGGAGACCTAGCACAACTCTC-3′
Rev: 5′-GCCGCTGCCGCTGTGATGGACACC-3′
P5−1679 bp to −1296 bpFwd: 5′-GGTGTCCATCACAGCGGCAGCGGC-3′
Rev: 5′-GAGAGGAGGTGGTGATTGAGGGC-3′
P6−1319 bp to −936 bpFwd: 5′-GCCCTCAATCACCACCTCCTTCTC-3′
Rev: 5′-CTCAGCCTGGCTCGCCGGCTCG-3′
P7−958 bp to −578 bpFwd: 5′-CGAGCCGGCGAGCCAGGCTGAG-3′
Rev: 5′-CGTCCACTTCCTGAAGGGTGGG-3′;
P8−599 bp to −233 bpFwd: 5′-CCCACCCTTCAGGAAGTGGACG-3′
Rev: 5′-GCGCCCGCCCCCGAGGCCG-3′
P9−256 bp to +10 bpFwd: 5′-CGGCCTCGGGGGCGGGCGC-3′
Rev: 5′-GTTGGGGCATCCCGGCGGCTCTG-3′
P10−13 bp to +436 bpFwd: 5′-CAGAGCCGCCGGGATGCCCCAAC-3′
Rev: 5′-CGCCTTCGTTCCCTTCCTCCCTC-3′

Base pair position is relative to the translational ATG start site in exon 1.

Sox17 directly binds to sequences in the Lef-1 promoter.

Based on the overlap in ChIP binding of Sox17 to Lef-1 promoter regions that contain TCF consensus sites, we performed a more extensive sequence analysis of the Lef-1 promoter for TCF and Sox candidate binding sites. Six sites in the Lef-1 promoter (TLS1, 2, 3, 4, and 6) matched two widely accepted consensus sequences for TCF (CTTTGWW and CWTTGWW) (4, 38, 39) (Table 4). Of note, a widely accepted SRY/Sox consensus sequence (WWCAAWG) (22, 25, 49) is identical to the reverse-strand TCF consensus sequence CWTTGWW (Fig. 7A). Additionally, one core Sox consensus sequence (AACAAT) in the Lef-1 promoter (SXS5) did not conform to a TCF consensus sequence. Given that TCF and SRY/Sox proteins can bind to similar sequences with different affinities (22), this analysis suggested potential for context-specific binding of TCF4 and Sox17 to the same sites in the Lef-1 promoter (Table 4).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590007.jpg

Evaluation of Sox17 and TCF binding sites in the Lef-1 promoter by EMSA. A: schematic representation of the 2.5-kb human Lef-1 promoter, with the positions and sequences of candidate TCF and Sox17 protein binding sites indicated (as determined based on the consensus sequences described in Table 4). The direction of arrows marked TCF and Sox17 indicate whether the consensus conforms to the forward or reverse stand of the DNA sequence. B: EMSA gels depicting protein/DNA complexes formed on the indicated biotinylated probes (Table 2) following binding by purified BSA (b), Sox17 (S), TCF4 (Tc), and Lef-1 (L). SP1 and TOP are negative and positive control biotinylated oligonucleotides for SP1 and TCF/Lef-1 (same consensus in TOP-flash) binding, respectively. C: EMSA gels depicting protein/DNA complexes formed when purified Sox17 protein was incubated with the indicated wild-type (W) or mutant (Mu) biotinylated probes (Table 2) for the 3 primary Sox17 binding sites. D: EMSA gels depicting protein/DNA complexes formed on the indicated biotinylated probes following binding of purified BSA (b), full-length Sox17 (S), Sox17-TAD (T), t-Sox17 (t), Sox17-mut1 (M1), or Sox17-mut2 (M2).

Table 4.

Candidate consensus binding sites for TCF and Sox within the human Lef-1 promoter

SiteLef-1 Promoter Location, bpForward-Strand SequenceReverse-Strand SequencePotential for TCF BindingPotential for Sox Binding
TCF C-1CTTTGWW*WWCAAAGYesNo
TCF C-2CWTTGWWWWCAAWGYesYes
Sox C-1WWCAAWGCWTTGWWYesYes
Sox C-2AACAATATTGTTNoYes
TLS1−2693/−2687tcCTTTGTAaattTACAAAGgaYes (C-1)Yes (C-1)
Yes (C-2)No (C-2)
TLS2−2427/−2421tcCTTTGTTtataAACAAAGgaYes (C-1)Yes (C-1)
Yes (C-2)No (C-2)
TLS3−2254/−2248gtCTTTGTAcatgTACAAAGacYes (C-1)Yes (C-1)
Yes (C-2)No (C-2)
TLS4−2240/−2234tcATCAAAGagctCTTTGATgaYes (C-1)Yes (C-1)
Yes (C-2)No (C-2)
SXS5−2956/−1951atATTGTTtataAACAATatNo (C-1)No (C-1)
No (C-2)Yes (C-2)
TLS6−921/−915ggAACAAAGagctCTTTGTTccYes (C-1)Yes (C-1)
Yes (C-2)No (C-2)

Two consensus sequences for TCF and Sox are given as C-1 and C-2. The position of the core consensus sequence (capital letters) in the Lef-1 promoter is given. This numbering is based on the location relative to the start translational methionine at +1 (17). The core consensus sequences are given in capital letters, and bases outside the core consensus sequence are given in lowercase letters.

*Consensus sequences were derived from the following manuscripts: TCF–CTTTGWW (4), TCF–CWTTGWW (38, 39), Sox17–ATTGTT (26), Sox–WWCAAWG (22, 25, 49).
The consensus sequences for TCF and Sox are identical but are shown as traditionally reported in the field for the coding-strand sequence.

To test the potential molecular interactions at these sites in the Lef-1 promoter, we performed EMSA using recombinant His-tagged Sox17, TCF4, and Lef-1 proteins purified from bacteria. Results from these studies demonstrated that Sox17 binds strongly to double-stranded oligonucleotides (Table 2) containing TLS2, SXS5, and TLS6 sequences, and that it binds weakly to TLS1 and TLS3/4 (Fig. 7B). TCF4 and Lef-1 bound strongly to TLS3/4 and/or TLS6, but relatively weakly to TLS1. The TOP consensus sequence, which is known to avidly bind TCF proteins, strongly bound to both TCF4 and Lef-1, but not to Sox17 (Fig. 7B). A negative control SP1 consensus demonstrated no binding to Sox17, TCF4, or Lef-1 protein. These findings support the hypothesis that the Lef-1 promoter harbors overlapping sites of binding for Sox17 and TCF proteins and that the affinities of these proteins for these sites are sequence specific.

To confirm the specificity of Sox17 binding to these sites, we performed EMSAs with mutated DNA sequences for TLS2, SXS5, and TLS6 sites (Table 2), those that demonstrated the strongest binding by ChIP and EMSA. As shown in Fig. 7C, Sox17 effectively bound to the wild-type, but not mutated, sequences. We next evaluated which functional domains of Sox17 are required for efficient DNA binding of Sox17 at these sites. These studies utilized Sox17 mutant proteins shown in Fig. 5A. As anticipated, the HMG binding domain mutant (t-Sox17) was unable to bind to any of these three sites (Fig. 7D). Additionally, mutations in the HMG domain either significantly reduced, or complete ablated, Sox17 binding to the TLS2, SXS5, and TLS6 sites. Notably, the Sox17 TAD mutant (deletion of only the BBD) bound the SXS5 and TLS6 sites much more weakly than did the wild-type F-Sox17 protein, but bound the TLS2 site with normal affinity (Fig. 7D). These findings suggested that binding of Sox17 to SXS5 and TLS6 sites is influenced by its BBD. Collectively, the results from these EMSA experiments suggest that multiple distinct and overlapping Sox17 and TCF binding sites exist within the Lef-1 promoter and that the affinity of Sox17 for certain sites is influenced by its BBD.

Sox17 interacts with β-catenin, TCF4, and Lef-1 to form stable protein/DNA complex on Lef-1 promoter elements.

Given the potential for overlap in the binding of TCF4, Lef-1, and Sox17 to specific sites in the Lef-1 promoter, it was important to establish whether these proteins form intermolecular complexes while bound to Lef-1 promoter elements. Indeed, coimmunoprecipitation (co-IP) of V5-tagged-Sox17, TCF4, and β-catenin-S37A from nuclear extracts demonstrated that these three proteins interact to varying extents in 293 and SW480 cells (Fig. 8A). To determine how interactions between Sox17, TCF/Lef-1, and β-catenin influence binding at specific TCF/Sox sites (TLS2, TLS3/4, SXS5, and TLS6) in the Lef-1 promoter, we developed a biotin/avidin-based DNA binding assay in which biotinylated oligonucleotides are used to capture complexes from a mixture of purified recombinant Sox17, TCF4, Lef-1, and β-catenin proteins (Table 2, Fig. 8B). As predicted by EMSA data, only Sox17 was captured with TLS2 oligonucleotide from this mixture of proteins (Fig. 8C). Additionally, no β-catenin was associated with the Sox17 bound to the TLS2 sequence. By contrast, Sox17, TCF4, Lef-1, and β-catenin proteins all bound to the TLS3/4, SXS5, and TLS6 sequences. The fact that Sox17, TCF4, and Lef-1 were each able to bind to the TLS3/4 and TLS6 sequences as a mixture of proteins substantiated our results from the EMSA assays, which had indicated each of these proteins could individually bind these sites to various extents (Fig. 7). However, the affinity of Sox17 for the TLS6 and TLS3/4 sequences appeared to be either reduced or enhanced, respectively, by the presence of TCF4, Lef-1, and β-catenin. These findings suggest that TCFs can influence Sox17 binding to the Lef-1 promoter in a sequence-specific manner through either competition for the binding site or intermolecular interactions. Of greatest interest was the finding that TCF4 and Lef-1 were associated with the SXS5 site only in the presence of Sox17 (Fig. 8C); neither TCF4 nor Lef-1 was observed to bind SXS5 when they were incubated in isolation with the DNA (Fig. 7B). These findings suggest that Sox17 binding to SXS5 promotes the formation of complexes with TCF4/Lef-1 and β-catenin. Similar observations were made for the TLS3/4 site for which the presence of TCF4/Lef-1/β-catenin enhanced Sox17 binding (Fig. 8C) over that seen in the presence of Sox17 alone (Fig. 7B).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590008.jpg

Sox17, TCF4, Lef-1, and β-catenin form intermolecular complexes on sequences within the Lef-1 promoter. A: the indicated cell lines were transfected with V5-tagged Sox17-, TCF4-, and β-catenin-expressing plasmids, and nuclear extracts were prepared at 48 h posttransfection. These extracts were then immunoprecipitated with antibodies against V5 (for Sox17), TCF4, or β-catenin (Bc) before being immunoblotted against the indicated antibodies. B: schematic representation of the strategy used in the experiments represented in C and D to detect protein/DNA complexes bound to biotinylated oligonucleotide sequences from the Lef-1 promoter. Purified recombinant proteins were prebound to the biotinylated oligonucleotides before they were captured on Avidin dynabeads and Western blotted with the appropriate antibodies. C: association of β-catenin, Sox17, TCF4, and Lef-1 with the indicated Lef-1 promoter sequences. An SP1 oligonucleotide served as negative control. D: association of β-catenin, TCF4, Lef-1, and Sox17 mutant (Sox17-TAD or t-Sox17) with the indicated Lef-1 promoter sequences.

To investigate whether certain Sox17 domains influence the formation of Sox17/TCF4/Lef-1/β-catenin complexes on the SXS5, TLS6, or TLS3/4 Lef-1 promoter sequences, we repeated the binding assays with the Sox17-TAD and t-Sox17 mutants. Interestingly, unlike full-length Sox17, Sox17-TAD was incapable of forming a complex on SXS5 with TCF4, Lef-1, or β-catenin (Fig. 8D vs. Fig. 8C), thus the Sox17 BBD domain is necessary for complex formation at SXS5. By contrast, enhanced Sox17 binding to the TLS3/4 site in the presence of TCF4/Lef-1/β-catenin did not require the Sox17 BBD (Fig. 8D). On the other hand, t-Sox17 lacking the HMG domain failed to bind DNA at any of the three sites analyzed in the presence of TCF4/Lef-1/β-catenin (Fig. 8D). The ability of Sox17-TAD, but not t-Sox17, to associate with TLS3/4 in the presence of TCF4/Lef-1/β-catenin (Fig. 8D) suggests that the interaction between the HMG domains of Sox17 and TCF4 stabilize the protein complex leading to increased DNA affinity.

Multiple Sox17 sites in the Lef-1 promoter control activation and repression in a β-catenin-dependent fashion.

To evaluate the functional importance of Sox17 and TCF binding sites in activation and repression of the Lef-1 promoter, we mutated these three binding sites (TLS2, SXS5, and TLS6) within the Lef-1 promoter-luciferase reporter, both individually and in combination, and assessed transcriptional activity in 293 cells expressing β-catenin-S37A or Sox17 (Fig. 9, A and B). Notably, mutation of each of these sites (TLS2, SXS5, TLS6) individually, and in one combination (TLS2/SXS5), significantly impaired β-catenin-mediated activation of the promoter. Although the baseline activity of the promoter was impaired by the triple mutation (3.6-fold), this mutant was significantly more responsive to β-catenin-S37A activation (6.2-fold) than the wild-type promoter (4.6-fold). We next evaluated which of the primary Sox17 binding sites were necessary for promoter induction in 293 cells by overexpressed Sox17 (Fig. 9B). We found that mutation of the TLS2 site significantly enhanced Sox17-mediated activation of the Lef-1 promoter, suggesting that this site may repress transcriptional activity when Sox17 is also bound to other sites in the promoter. In support of this hypothesis, the TLS2/TLS6/SXS5 mutant lost Sox17-dependent hyperactivation. The Sox17 BBD was also necessary for hyperactivation of the TLS2 mutant, since the Sox17-TAD mutant did not activate TLS2 mutant transcription above the wild-type promoter (Fig. 9B).

An external file that holds a picture, illustration, etc.
Object name is zh50111057590009.jpg

Transcriptional analysis of the Lef-1 promoter with mutated Sox17 and TCF4/Lef-1 binding sites. A: schematic representation of TCF/Lef-1 and Sox17 binding interactions on the Lef-1 promoter as determined by ChIP, EMSA, and DNA-affinity capture assays. The size of each arrowed box denotes the level of binding (i.e., smallest represents weakest binding). The presence of β-catenin (red) indicates that a particular factor is able to bind β-catenin and/or that β-catenin may have to interact with the BBD of Sox17 to mediate an intermolecular interaction (i.e., at SXS5). B: promoter activity when 293 cells were transfected with the indicated mutant or wild-type Lef-1 promoter-luciferase constructs (as indicated by color), together with plasmid expressing the protein(s) indicated on the x-axis. C: promoter activity when A549 cells were transfected with the indicated mutant or wild-type Lef-1 promoter-luciferase constructs on the x-axis, together with plasmid expressing Wnt3A and/or Sox17 (as indicated by color). Significant differences in expression levels as determined by Student's t-test (P < 0.05) are indicated by the connecting lines.

We next sought to evaluate which Lef-1 promoter mutations interfered with the ability of Sox17 to repress promoter activation by β-catenin-S37A. As previously observed (Fig. 3E), Sox17 expression led to significant inhibition of β-catenin-dependent activation of the wild-type Lef-1 promoter in 293 cells (Fig. 9B). By contrast, all seven Lef-1 promoter mutants lost Sox17-mediated repression in the presence β-catenin-S37A. These findings suggest that all Sox17 binding sites within the Lef-1 promoter are equally required for Sox17-mediated repression in the context of β-catenin-mediated activation. Also, promoter expression in the context of Sox17-TAD/β-catenin-S37A coexpression was indistinguishable from that produced by F-Sox17/β-catenin-S37A expression, regardless of which promoter construct (wild-type or any mutant) was tested. Thus, the BBD of Sox17 appears not to influence the transcriptional properties of Sox17 in the context of an activated β-catenin signal. In summary, these findings demonstrate that Sox17 can both activate and inhibit the Lef-1 promoter and that although the Sox17 BBD contributes to this activation, this region of the molecule is not required for promoter inhibition in the presence of a β-catenin signal.

Multiple Sox17 and TCF sites in the Lef-1 promoter influence its responsiveness to Wnt3A.

We next evaluated 10 Lef-1 promoter mutants in the A549 cell line (Fig. 9C) in which Wnt3A induction of the Lef-1 promoter is significantly repressed by Sox17 (Fig. 4). Results from these experiments demonstrate that mutation of the three primary Sox17 binding sites (TLS2, SXS5, and TLS6) either individually or in combination destroyed the ability of Sox17 to repress Wnt3A activation of the Lef-1 promoter (Fig. 9C). This was similar to the effect of Sox17 seen in 293 cells in the context of β-catenin-S37A activation. With the exception of the TLS2/TLS6/SXS5 triple mutant, all mutant combinations of these sites significantly elevated baseline activity of the promoter while reducing its induction by Wnt3A. This finding is consistent with previous work suggesting that Wnt3A-mediated induction of the promoter involves derepression (17). These findings suggest that Sox17 binding sites in the Lef-1 promoter act in concert to mediate repression in the presence and absence of a Wnt signal.

An additional set of mutants evaluated in A549 cells affected the TLS3 and TLS4 sites, which predominantly bind TCF4/Lef-1 directly (Fig. 7B), but for which Sox17 can also associate in an HMG domain-dependent manner (Fig. 8D) when TCF4/Lef-1/β-catenin are present. Notably, mutation of these two sites, either in combination or individually, also prevented Sox17-dependent repression of the Lef-1 promoter in the presence of Wnt3A. However, the double mutant TLS3/TLS4 also lost Wnt3A-mediated activation in conjunction with an elevated baseline activity, similar to that previously reported for WRE deletion (17). The pattern of expression from this TLS3/TLS4 mutant was strikingly similar to that of the SXS5 mutant. Given the similar phenotypes of these two mutants, we hypothesize that TCF4 or Sox17 protein bound to these distant Lef-1 promoter sites may interact to control activation and repression of the promoter. Although we found no evidence for direct binding of TCF4, Lef-1, or Sox17 to the WRE by EMSA (data not shown), deletion of this 110-bp region (−879 to −769 bp) significantly reduces Wnt3A responsiveness and raises baseline activity of the Lef-1 promoter (17). We hypothesize that internal deletion of the WRE alters the context of Sox17 and/or TCF binding at the TLS6 site, which is just upstream to the WRE. Thus, it appears to be inappropriate to call this 110-bp segment a Wnt-responsive element, since Wnt responsiveness is controlled by multiple distant sites across the Lef-1 promoter.

DISCUSSION

Canonical Wnt signaling plays roles in numerous developmental programs through β-catenin-dependent transcription factors of the TCF/Lef-1 family. Analysis of a transgenic Lef-1 promoter-driven reporter gene in Wnt3A knockout mice has demonstrated that the promoter is activated by Wnt3A signals in airway submucosal gland progenitor cells (10). Given that Sox17 expression is inversely regulated with Lef-1 expression in submucosal gland progenitor cells (Fig. 1), Sox17 appeared to be an interesting candidate for regulating the Lef-1 promoter. Sox17 is also known to influence the proliferation and differentiation of airway epithelial cells in mice (29, 36, 37) and also to inhibit Wnt signaling (41, 54). Indeed, Wnt stimulation of undifferentiated primary airway epithelial cells led to a dramatic increase in Lef-1 and cyclin D1 protein levels, with a concomitant decrease in the DNA binding form of Sox17 (F-Sox17) (Fig. 2). Given that Lef-1 induces the cyclin D1 promoter (40) and that glandular airway progenitors fail to express Lef-1 and cyclin D1 in the absence of Wnt3A (10), these studies support a role for Lef-1 in driving the proliferation of glandular progenitors. However, the increased proliferation of glandular progenitor cells that inhibit Sox17 and induced Lef-1 expression is inconsistent with a previous report demonstrating that overexpression of Sox17 in respiratory epithelial cells of transgenic mice promotes proliferation and differentiation (37). Thus, Sox17 may be capable of influencing different pathways that can either inhibit or promote proliferation in the proximal and respiratory airway epithelium, respectively. Such differences may be controlled by the context of the Wnt signal and responsiveness of the Lef-1 promoter in a given airway cell type.

Our initial experiments in five cell lines demonstrated that Sox17 inhibits Wnt3A/β-catenin activation of the Lef-1 promoter (Figs. 3 and and4).4). Several findings supported direct binding of Sox17 to the Lef-1 promoter as the mechanisms for inhibiting Wnt3A/β-catenin-mediated transcription. First, the DNA binding domain of Sox17 was required for its ability to inhibit Wnt3A-mediated activation of the Lef-1 promoter (Fig. 5). Second, ChIP and EMSA analysis demonstrated that three regions in the endogenous Lef-1 promoter bound Sox17 (TLS2, SXS5, and TLS6) (Fig. 6B and and7).7). Of significant interest was the finding that the Sox17 BBD is required for efficient binding to SXS5 and TLS6, but not to TLS2. This unexpected finding suggests that the structure of Sox17-bound DNA differs across the Lef-1 promoter. Given that Sox17 associates with TCF4/β-catenin complexes (Fig. 5C) (41) and that TCF4/β-catenin complexes are in part required for Wnt-mediated activation of the Lef-1 promoter (5, 6, 30), the finding that the Sox17 BBD influences binding to certain sites in the Lef-1 promoter is of significant interest.

The HMG domains of Sox proteins mediate DNA binding in a sequence-specific fashion. Like other Sox proteins, Sox17 imposes a widening of the minor groove of the DNA and bending of ~80° (35, 47, 48). This feature is uncommon among transcription factors, most of which target mainly the major groove. The differences in DNA binding topology between the Sox proteins and other transcription factors, such as TCFs/Lef-1, make it possible for Sox proteins to bind to DNA in close proximity to other transcription factors (47). This characteristic of Sox protein/DNA binding has suggested that members of the Sox family of proteins organize the local chromatin structure and assemble associated transcription factors into multiprotein complexes capable of modulating transcription. Our findings on TCF4/Lef-1 and Sox17 binding to Lef-1 promoter sequences support the notion that intermolecular complexes between TCF4/Lef-1, β-catenin, and Sox17 form at certain sites on the promoter (Fig. 10). A perfect example of this is Sox17 binding to SXS5; although TCF4/Lef-1 do not bind this site in isolation, Sox17 binding at SXS5 can lead to the recruitment of TCF4/Lef-1/β-catenin complexes, and this function is dependent on the Sox17 BBD (Fig. 8, C and D). This finding suggests that β-catenin binding to Sox17 may be necessary to form TCF complexes on SXS5. By contrast, TCF4/Lef-1 binding to the TLS3/4 site can recruit Sox17, and this requires the HMG, but not the BBD, domain of Sox17. Whether TCF4/Lef-1 binding at TLS3/4 increases the affinity of Sox17 for one of the two TCF/Sox consensuses at this site, or Sox17 associates with TCF4/Lef-1 through its HMG domain as previously reported (41), is currently unclear. However, given that purified Sox17 has a low affinity for TLS3/4, either scenario is plausible. Two other sites in the Lef-1 promoter (TLS6 and TLS2) bind to TCF/Lef-1 and/or Sox17 in unusual ways. TLS2 binds primarily to Sox17, and binding at this site does not associate with β-catenin (Figs. 6, ,7,7, ,8C).8C). By contrast, both Sox17 and TCF4/Lef-1 can associate with TLS6. Thus, Sox17 and TCF4/Lef-1 may compete for occupancy of TLS6. These results suggest that the ratio of bound Sox17 to bound TCF4/Lef-1 in the context of a Wnt/β-catenin signal could dynamically regulate transcription from the Lef-1 promoter.

An external file that holds a picture, illustration, etc.
Object name is zh50111057590010.jpg

Context-dependent interactions between Sox17 and TCF4/Lef-1 at specific sites on the Lef-1 promoter and working models for Lef-1 promoter regulation. A: a summary of Sox17 and TCF4/Lef-1 binding to sites on the Lef-1 promoter, and of the context-dependent interactions that depend on the BBD and HMG domains of Sox17. Based on consensus sequences, 6 TCF/Sox sites exist in the Lef-1 promoter (sites 1–6). Site 2 (TLS2) binds only Sox17. Sites 3/4 (TLS3/4) primarily bind TCF4/Lef-1, but can also recruit Sox17 in the presence of both β-catenin and the Sox17 HMG domain. Site 5 (SXS5) primarily binds Sox17, but can recruit TCF4/Lef-1 in the presence of both β-catenin and the Sox17 β-catenin binding domain (BBD). Site 6 (TLS6) can directly bind to both Sox17 and TCF4/Lef-1. Whether context-dependent interactions between Sox17 and TCF4/Lef-1 occur at this site could not be determined from this study. However, we hypothesize that binding of Sox17 and TCFs to this site may be competitive. B: working models for Lef-1 promoter regulation. In the presence of a Wnt signal where β-catenin rises and Sox17 falls (as in submucosal gland progenitor cells), we hypothesize that sites within the Lef-1 promoter are predominantly bound by TCF4/β-catenin, and this leads to increased transcription. In the presence of a sustained Wnt signal, we hypothesize that Sox17 expression is reactivated, leading to competitive occupancy of Sox17/TCF4 sites and complex formation. This may function to repress the promoter. Alternative transcriptional states, such as the cell-specific induction of the Lef-1 promoter by Sox17 overexpression in 293 cells, may be explained by intermolecular complex formation between Sox17 and TCFs at distant sites in the promoter. These interactions may also explain the complex transcriptional consequences of mutating one or more TCF/Sox17 sites within the promoter that are inconsistent with assigning a strictly inhibitor function to Sox17 binding.

The process by which multiple and overlapping Sox17/TCF/Lef-1 binding sites in the full-length 2.5-kb Lef-1 promoter coordinate transcription in a Wnt/β-catenin-dependent fashion has been challenging to dissect. However, several themes have emerged from mutagenesis of the promoter. First, in both 293 and A549 cells, simultaneous mutation of all three primary Sox17 binding sites (TLS2/SXS5/TLS6) significantly impaired baseline, as well as Wnt3A/β-catenin-dependent, transcription from the promoter (Fig. 9). This is perhaps not surprising since two of these sites associate with TCF4/Lef-1/β-catenin either directly (TLS6) or indirectly (SXS5) through Sox17 binding. Mutation of Sox17 sites individually (TLS2, SXS5, or TLS6) led to the greatest decline in β-catenin-dependent induction of the promoter in 293 cells without affecting baseline transcription. This finding in 293 cells suggests that Sox17, although capable of repressing β-catenin-dependent Lef-1 transcription, may also be required for the induction of Lef-1 transcription, through interactions with TCF4/Lef-1/β-catenin complexes. In A549 cells, mutation of the SXS5 site completely ablated Wnt3A-mediated induction, and did so by raising baseline activation of the promoter; this effect was less pronounced in the TLS2 and TLS6 mutants. This finding in A549 cells fits our model whereby Sox17 expression in airway epithelia represses transcription from the Lef-1 promoter, and Wnt3A derepresses the promoter at SXS5. However, the finding that the double (SXS5/TLS6) and triple (TLS2/SXS5/TLS6) mutants retained some level of Wnt3A induction in A549 cells suggests that SXS5 does not act in isolation to control Wnt3A-mediated promoter activation.

Of the cell lines evaluated, 293 cells appear to be unique in their ability to induce the Lef-1 promoter when Sox17 alone is overexpressed. Hyperactivation of the TLS2 mutant promoter in the setting of Sox17 overexpression suggests that this site may play a predominantly inhibitory role in the absence of β-catenin, in which case Sox17 occupancy of SXS5 and TLS6 likely predominates (Fig. 9B). Since Sox17-TAD mutant failed to hyperactivate the TLS2 mutant promoter, we hypothesize that the Sox17 BBD is required for activation of the Lef-1 promoter at the SXS5 and TLS6 sites. Such findings are consistent with the fact that the Sox17-TAD mutant binds much less efficiently to these sites than does F-Sox17 (Fig. 7D). However, assigning a strictly inhibitory function to TLS2 is not consistent with the finding that the SXS5/TLS6 mutant retains the ability to activate the Lef-1 promoter in the presence of overexpressed Sox17; in this case, Sox17 occupancy of TLS2 likely predominates. For these reasons, our current hypothesis is that the overall Sox17 occupancy on the Lef-1 promoter controls transcriptional activity, and that context-dependent changes in the formation of Sox17 complexes with TCF4/Lef-1 influences Lef-1 transcription.

The ability of Sox17 to suppress Wnt3A activation of both the Lef-1 and TOP-flash promoters is shared by multiple cell lines. However, given that Sox17 does not directly bind to the TOP-flash promoter, the mechanisms of action underlying this inhibition appear different. Previous studies have demonstrated that one mechanism by which Sox17 inhibits Wnt signaling (as assayed by TOP-flash) is proteasome-mediated degradation of TCF4 (41). This degradation requires the HMG binding domain of Sox17. Indeed, Sox17-mediated changes in TCF4 levels may be partly responsible for the ability of Sox17 to inhibit Wnt3A/β-catenin-mediated induction of the Lef-1 promoter. However, mutagenesis studies of the Sox17 binding site in the Lef-1 promoter suggest that direct Sox17 binding to the promoter is also involved (Fig. 9); mutation of any of the three major Sox17 binding sites in the Lef-1 promoter (TLS2, SXS5, or TLS6), alone or in combination, prevented repression of Wnt3A-induced promoter activity in the context of Sox17 overexpression in A549 cells (Fig. 9C). These findings suggest that the direct cumulative binding of Sox17 at all three binding sites in the Lef-1 promoter is required for the repression of Wnt3A activation; they cannot be explained by simply a posttranslational mechanism whereby Sox17 mediates the degradation of TCF4/β-catenin, as previously reported in colon cancers (41). Although we also found direct interactions among Sox17, TCF4, and β-catenin as previously described (41), we did not observe a dominant effect of Sox17-mediated degradation of TCF4 when these two proteins were coexpressed in cell lines (Fig. 8A). Thus, this function may be cell-type specific. During SMG development, TCF4 is also highly expressed in both the SAE and newly forming glandular buds that induce Lef-1 (10), despite differences in the level of Sox17 protein expression in these two cellular compartments. Therefore, decreased Sox17 expression during glandular development does not appear to influence the stability of TCF4 protein in glandular progenitor cells.

In summary, the experiments described here demonstrate several novel features of Sox17 and its ability to regulate the Lef-1 promoter (Fig. 10). First, we have shown that Sox17 binds directly to multiple sites in the Lef-1 promoter and controls its transcriptional activity, in both the presence and absence of Wnt/β-catenin signals. Second, we have shown that Sox17/TCF4/Lef-1/β-catenin complexes can be directed to certain sites in the Lef-1 promoter through the Sox17 BBD or HMG domains. Third, we have identified sites within the Lef-1 promoter that can bind to both Sox17 and TCF4/Lef-1, which may control Wnt/β-catenin-dependent transcription in a competitive manner. This work suggests that TCF and Sox proteins may act in concert to finely control transcriptional activation and repression of the Lef-1 promoter response to Wnt signals. Our findings provide important new insights into the duality of context-dependent HMG-transcription factors in the control of gene expression.

GRANTS

This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grants P30-DK-054759 and R37-DK-047967 (to J.F. Engelhardt), the Roy J. Carver Chair in Molecular Medicine (to J. F. Engelhardt), and Reproductive Scientists Development Program Grant 5K12HD00849 and Cancer Center Support Grant 3P30CA086862-10S4 (to M. J. Goodheart).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

ACKNOWLEDGMENTS

We thank Dr. Jeffrey Whitsett (Cincinnati Children's Hospital Research Foundation, Cincinnati, OH) for his kind gift of the guinea pig anti-Sox17 antibody. We also gratefully acknowledge Dr. Christine Blaumueller (University of Iowa, Iowa City, IA) for editorial contributions.

REFERENCES

1. Amen M, Liu X, Vadlamudi U, Elizondo G, Diamond E, Engelhardt JF, Amendt BA. PITX2 and beta-catenin interactions regulate Lef-1 isoform expression. Mol Cell Biol 27: 7560–7573, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
2. Anderson RD, Haskell RE, Xia H, Roessler BJ, Davidson BL. A simple method for the rapid generation of recombinant adenovirus vectors. Gene Ther 7: 1034–1038, 2000 [Abstract] [Google Scholar]
3. Arce L, Pate KT, Waterman ML. Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer 9: 159, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
4. Arce L, Yokoyama NN, Waterman ML. Diversity of LEF/TCF action in development and disease. Oncogene 25: 7492–7504, 2006 [Abstract] [Google Scholar]
5. Atcha FA, Munguia JE, Li TW, Hovanes K, Waterman ML. A new beta-catenin-dependent activation domain in T cell factor. J Biol Chem 278: 16169–16175, 2003 [Abstract] [Google Scholar]
6. Atcha FA, Syed A, Wu B, Hoverter NP, Yokoyama NN, Ting JH, Munguia JE, Mangalam HJ, Marsh JL, Waterman ML. A unique DNA binding domain converts T-cell factors into strong Wnt effectors. Mol Cell Biol 27: 8352–8363, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
7. Boras-Granic K, Chang H, Grosschedl R, Hamel PA. Lef1 is required for the transition of Wnt signaling from mesenchymal to epithelial cells in the mouse embryonic mammary gland. Dev Biol 295: 219–231, 2006 [Abstract] [Google Scholar]
8. Borthwick DW, Shahbazian M, Krantz QT, Dorin JR, Randell SH. Evidence for stem-cell niches in the tracheal epithelium. Am J Respir Cell Mol Biol 24: 662–670, 2001 [Abstract] [Google Scholar]
9. Clevers H. Wnt/beta-catenin signaling in development and disease. Cell 127: 469–480, 2006 [Abstract] [Google Scholar]
10. Driskell RR, Goodheart M, Neff T, Liu X, Luo M, Moothart C, Sigmund CD, Hosokawa R, Chai Y, Engelhardt JF. Wnt3a regulates Lef-1 expression during airway submucosal gland morphogenesis. Dev Biol 305: 90–102, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
11. Driskell RR, Liu X, Luo M, Filali M, Zhou W, Abbott D, Cheng N, Moothart C, Sigmund CD, Engelhardt JF. Wnt-responsive element controls Lef-1 promoter expression during submucosal gland morphogenesis. Am J Physiol Lung Cell Mol Physiol 287: L752–L763, 2004 [Abstract] [Google Scholar]
12. Du YC, Oshima H, Oguma K, Kitamura T, Itadani H, Fujimura T, Piao YS, Yoshimoto T, Minamoto T, Kotani H, Taketo MM, Oshima M. Induction and downregulation of Sox17 and its possible roles during the course of gastrointestinal tumorigenesis. Gastroenterology 137: 1346–1357, 2009 [Abstract] [Google Scholar]
13. Duan D, Sehgal A, Yao J, Engelhardt JF. Lef1 transcription factor expression defines airway progenitor cell targets for in utero gene therapy of submucosal gland in cystic fibrosis. Am J Respir Cell Mol Biol 18: 750–758, 1998 [Abstract] [Google Scholar]
14. Duan D, Yue Y, Zhou W, Labed B, Ritchie TC, Grosschedl R, Engelhardt JF. Submucosal gland development in the airway is controlled by lymphoid enhancer binding factor 1 (LEF1). Development 126: 4441–4453, 1999 [Abstract] [Google Scholar]
15. Engelhardt JF. Stem cell niches in the mouse airway. Am J Respir Cell Mol Biol 24: 649–652, 2001 [Abstract] [Google Scholar]
16. Engelhardt JF, Schlossberg H, Yankaskas JR, Dudus L. Progenitor cells of the adult human airway involved in submucosal gland development. Development 121: 2031–2046, 1995 [Abstract] [Google Scholar]
17. Filali M, Cheng N, Abbott D, Leontiev V, Engelhardt JF. Wnt-3A/beta-catenin signaling induces transcription from the LEF-1 promoter. J Biol Chem 277: 33398–33410, 2002. [Abstract] [Google Scholar]
18. Fu DY, Wang ZM, Li C, Wang BL, Shen ZZ, Huang W, Shao ZM. Sox17, the canonical Wnt antagonist, is epigenetically inactivated by promoter methylation in human breast cancer. Breast Cancer Res Treat 119: 601–612, 2010. [Abstract] [Google Scholar]
19. Garcia C, Calvo E, Nieto A. The transcription factor SOX17 is involved in the transcriptional control of the uteroglobin gene in rabbit endometrium. J Cell Biochem 102: 665–679, 2007 [Abstract] [Google Scholar]
20. Gebeshuber CA, Sladecek S, Grunert S. Beta-catenin/LEF-1 signalling in breast cancer–central players activated by a plethora of inputs. Cells Tissues Organs 185: 51–60, 2007 [Abstract] [Google Scholar]
21. Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem 281: 22429–22433, 2006 [Abstract] [Google Scholar]
22. Harley VR, Lovell-Badge R, Goodfellow PN. Definition of a consensus DNA binding site for SRY. Nucleic Acids Res 22: 1500–1501, 1994 [Europe PMC free article] [Abstract] [Google Scholar]
23. Hovanes K, Li TW, Munguia JE, Truong T, Milovanovic T, Lawrence Marsh J, Holcombe RF, Waterman ML. Beta-catenin-sensitive isoforms of lymphoid enhancer factor-1 are selectively expressed in colon cancer. Nat Genet 28: 53–57, 2001 [Abstract] [Google Scholar]
24. Hovanes K, Li TW, Waterman ML. The human LEF-1 gene contains a promoter preferentially active in lymphocytes and encodes multiple isoforms derived from alternative splicing. Nucleic Acids Res 28: 1994–2003, 2000 [Europe PMC free article] [Abstract] [Google Scholar]
25. Jenkins E, Moss JB, Pace JM, Bridgewater LC. The new collagen gene COL27A1 contains SOX9-responsive enhancer elements. Matrix Biol 24: 177–184, 2005 [Europe PMC free article] [Abstract] [Google Scholar]
26. Kanai Y, Kanai-Azuma M, Noce T, Saido TC, Shiroishi T, Hayashi Y, Yazaki K. Identification of two Sox17 messenger RNA isoforms, with and without the high mobility group box region, and their differential expression in mouse spermatogenesis. J Cell Biol 133: 667–681, 1996 [Europe PMC free article] [Abstract] [Google Scholar]
27. Katoh M. Cross-talk of WNT and FGF signaling pathways at GSK3beta to regulate beta-catenin and SNAIL signaling cascades. Cancer Biol Ther 5: 1059–1064, 2006. [Abstract] [Google Scholar]
28. Kormish JD, Sinner D, Zorn AM. Interactions between SOX factors and Wnt/beta-catenin signaling in development and disease. Dev Dyn 239: 56–68 [Europe PMC free article] [Abstract] [Google Scholar]
29. Lange AW, Keiser AR, Wells JM, Zorn AM, Whitsett JA. Sox17 promotes cell cycle progression and inhibits TGF-beta/Smad3 signaling to initiate progenitor cell behavior in the respiratory epithelium. PLoS One 4: e5711, 2009 [Europe PMC free article] [Abstract] [Google Scholar]
30. Li TW, Ting JH, Yokoyama NN, Bernstein A, van de Wetering M, Waterman ML. Wnt activation and alternative promoter repression of LEF1 in colon cancer. Mol Cell Biol 26: 5284–5299, 2006 [Europe PMC free article] [Abstract] [Google Scholar]
31. Liu X, Driskell RR, Engelhardt JF. Airway glandular development and stem cells. Curr Top Dev Biol 64: 33–56, 2004 [Abstract] [Google Scholar]
32. Liu X, Driskell RR, Luo M, Abbott D, Filali M, Cheng N, Sigmund CD, Engelhardt JF. Characterization of Lef-1 promoter segments that facilitate inductive developmental expression in skin. J Invest Dermatol 123: 264–274, 2004 [Europe PMC free article] [Abstract] [Google Scholar]
33. Liu X, Luo M, Zhang L, Ding W, Yan Z, Engelhardt JF. Bioelectric properties of chloride channels in human, pig, ferret, and mouse airway epithelia. Am J Respir Cell Mol Biol 36: 313–323, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
34. Nusse R. Wnt signaling in disease and in development. Cell Res 15: 28–32, 2005 [Abstract] [Google Scholar]
35. Palasingam P, Jauch R, Ng CK, Kolatkar PR. The structure of Sox17 bound to DNA reveals a conserved bending topology but selective protein interaction platforms. J Mol Biol 388: 619–630, 2009 [Abstract] [Google Scholar]
36. Park KS, Wells JM, Zorn AM, Wert SE, Laubach VE, Fernandez LG, Whitsett JA. Transdifferentiation of ciliated cells during repair of the respiratory epithelium. Am J Respir Cell Mol Biol 34: 151–157, 2006 [Europe PMC free article] [Abstract] [Google Scholar]
37. Park KS, Wells JM, Zorn AM, Wert SE, Whitsett JA. Sox17 influences the differentiation of respiratory epithelial cells. Dev Biol 294: 192–202, 2006 [Abstract] [Google Scholar]
38. Roose J, Clevers H. TCF transcription factors: molecular switches in carcinogenesis. Biochim Biophys Acta 1424: M23–M37, 1999 [Abstract] [Google Scholar]
39. Schwartz DR, Wu R, Kardia SL, Levin AM, Huang CC, Shedden KA, Kuick R, Misek DE, Hanash SM, Taylor JM, Reed H, Hendrix N, Zhai Y, Fearon ER, Cho KR. Novel candidate targets of beta-catenin/T-cell factor signaling identified by gene expression profiling of ovarian endometrioid adenocarcinomas. Cancer Res 63: 2913–2922, 2003 [Abstract] [Google Scholar]
40. Shtutman M, Zhurinsky J, Simcha I, Albanese C, D'Amico M, Pestell R, Ben-Ze'ev A. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA 96: 5522–5527, 1999 [Europe PMC free article] [Abstract] [Google Scholar]
41. Sinner D, Kordich JJ, Spence JR, Opoka R, Rankin S, Lin SC, Jonatan D, Zorn AM, Wells JM. Sox17 and Sox4 differentially regulate beta-catenin/T-cell factor activity and proliferation of colon carcinoma cells. Mol Cell Biol 27: 7802–7815, 2007 [Europe PMC free article] [Abstract] [Google Scholar]
42. Sinner D, Rankin S, Lee M, Zorn AM. Sox17 and beta-catenin cooperate to regulate the transcription of endodermal genes. Development 131: 3069–3080, 2004 [Abstract] [Google Scholar]
43. Vadlamudi U, Espinoza HM, Ganga M, Martin DM, Liu X, Engelhardt JF, Amendt BA. PITX2, beta-catenin and LEF-1 interact to synergistically regulate the LEF-1 promoter. J Cell Sci 118: 1129–1137, 2005 [Abstract] [Google Scholar]
44. van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development 136: 3205–3214, 2009 [Abstract] [Google Scholar]
45. Waterman ML. Expression of lymphoid enhancer factor/T-cell factor proteins in colon cancer. Curr Opin Gastroenterol 18: 53–59, 2002 [Abstract] [Google Scholar]
46. Waterman ML. Lymphoid enhancer factor/T cell factor expression in colorectal cancer. Cancer Metastasis Rev 23: 41–52, 2004 [Abstract] [Google Scholar]
47. Wegner M. From head to toes: the multiple facets of Sox proteins. Nucleic Acids Res 27: 1409–1420, 1999 [Europe PMC free article] [Abstract] [Google Scholar]
48. Werner MH, Burley SK. Architectural transcription factors: proteins that remodel DNA. Cell 88: 733–736, 1997 [Abstract] [Google Scholar]
49. Wiebe MS, Nowling TK, Rizzino A. Identification of novel domains within Sox-2 and Sox-11 involved in autoinhibition of DNA binding and partnership specificity. J Biol Chem 278: 17901–17911, 2003 [Abstract] [Google Scholar]
50. Willert K, Brown JD, Danenberg E, Duncan AW, Weissman IL, Reya T, Yates JR, 3rd, Nusse R. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423: 448–452, 2003 [Abstract] [Google Scholar]
51. Willert K, Nusse R. Beta-catenin: a key mediator of Wnt signaling. Curr Opin Genet Dev 8: 95–102, 1998 [Abstract] [Google Scholar]
52. Yan Z, Zak R, Zhang Y, Ding W, Godwin S, Munson K, Peluso R, Engelhardt JF. Distinct classes of proteasome-modulating agents cooperatively augment recombinant adeno-associated virus type 2 and type 5-mediated transduction from the apical surfaces of human airway epithelia. J Virol 78: 2863–2874, 2004 [Europe PMC free article] [Abstract] [Google Scholar]
53. Zhang W, Glockner SC, Guo M, Machida EO, Wang DH, Easwaran H, Van Neste L, Herman JG, Schuebel KE, Watkins DN, Ahuja N, Baylin SB. Epigenetic inactivation of the canonical Wnt antagonist SRY-box containing gene 17 in colorectal cancer. Cancer Res 68: 2764–2772, 2008 [Europe PMC free article] [Abstract] [Google Scholar]
54. Zorn AM, Barish GD, Williams BO, Lavender P, Klymkowsky MW, Varmus HE. Regulation of Wnt signaling by Sox proteins: XSox17 alpha/beta and XSox3 physically interact with beta-catenin. Mol Cell 4: 487–498, 1999 [Abstract] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Article citations


Go to all (24) article citations

Data 


Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

NCI NIH HHS (1)

NICHD NIH HHS (1)

NIDDK NIH HHS (4)