Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2012 Mar 2;287(10):7026-38.
doi: 10.1074/jbc.M111.276311. Epub 2012 Jan 12.

Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP)

Affiliations

Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP)

Beiyun Zhou et al. J Biol Chem. .

Abstract

Interactions between transforming growth factor-β (TGF-β) and Wnt are crucial to many biological processes, although specific targets, rationale for divergent outcomes (differentiation versus block of epithelial proliferation versus epithelial-mesenchymal transition (EMT)) and precise mechanisms in many cases remain unknown. We investigated β-catenin-dependent and transforming growth factor-β1 (TGF-β1) interactions in pulmonary alveolar epithelial cells (AEC) in the context of EMT and pulmonary fibrosis. We previously demonstrated that ICG-001, a small molecule specific inhibitor of the β-catenin/CBP (but not β-catenin/p300) interaction, ameliorates and reverses pulmonary fibrosis and inhibits TGF-β1-mediated α-smooth muscle actin (α-SMA) and collagen induction in AEC. We now demonstrate that TGF-β1 induces LEF/TCF TOPFLASH reporter activation and nuclear β-catenin accumulation, while LiCl augments TGF-β-induced α-SMA expression, further confirming co-operation between β-catenin- and TGF-β-dependent signaling pathways. Inhibition and knockdown of Smad3, knockdown of β-catenin and overexpression of ICAT abrogated effects of TGF-β1 on α-SMA transcription/expression, indicating a requirement for β-catenin in these Smad3-dependent effects. Following TGF-β treatment, co-immunoprecipitation demonstrated direct interaction between endogenous Smad3 and β-catenin, while chromatin immunoprecipitation (ChIP)-re-ChIP identified spatial and temporal regulation of α-SMA via complex formation among Smad3, β-catenin, and CBP. ICG-001 inhibited α-SMA expression/transcription in response to TGF-β as well as α-SMA promoter occupancy by β-catenin and CBP, demonstrating a previously unknown requisite TGF-β1/β-catenin/CBP-mediated pro-EMT signaling pathway. Clinical relevance was shown by β-catenin/Smad3 co-localization and CBP expression in AEC of IPF patients. These findings suggest a new therapeutic approach to pulmonary fibrosis by specifically uncoupling CBP/catenin-dependent signaling downstream of TGF-β.

PubMed Disclaimer

Figures

FIGURE 1.
FIGURE 1.
TGF-β1 activates β-catenin-dependent signaling. A, representative Western blots (upper panel) and quantitative analysis of α-SMA (lower panel) following treatment of RLE-6TN cells with TGF-β1 (2.5 ng/ml) and/or LiCl (7.5 mm) for 6 days. Controls include media only, NaCl (7.5 mm) and TGF-β vehicle. Lamin A/C is used as a loading control (n = 4, *, p < 0.05 compared with vehicle). B, RLE-6TN cells were transfected with a LEF/TCF TOPFLASH reporter or its mutant, FOPFLASH, followed by treatment with TGF-β1 (2.5 ng/ml) for 24 h. Luciferase activity was normalized to Renilla luciferase activity (n = 3, * = p < 0.05 compared with vehicle). C, ICAT expression plasmid pCS2/ICAT and TOPFLASH reporter were co-transfected in RLE-6TN cells followed by TGF-β1 treatment. Reporter activity was determined 48 h after transfection and normalized to Renilla luciferase activity (n = 3, * = p < 0.05 (significantly different from pCS2 in the absence of TGF-β1)). D, representative Western blot (n = 3) for β-catenin in nuclear and cytoplasmic fractions harvested from RLE-6TN cells treated with TGF-β1 for indicated times. Lamin A/C and GAPDH are used to verify purity of nuclear and cytosolic fractions, respectively. Treatment with Wnt3a is used as a positive control. Cell lysates from RLE-6TN cells treated with TGF-β1 for 6 h were immunoprecipitated using anti-active-β-catenin (E) or anti-p-Tyr654-β-catenin (F) Abs. Immunoprecipitated active-β-catenin, dephosphorylated on Ser-37 and Thr-41 (E) or p-Tyr654-β-catenin (F), were detected by Western blot (n = 2). Representative Western blot (G) and quantitative analysis of total β-catenin (H) and α-SMA (I) in lysate from RLE-6TN cells transfected with β-catenin or control siRNA followed by TGF- β treatment for 48 h. GAPDH is used as a loading control (n = 3; *, p < 0.05 compared with control siRNA; **, p < 0.05 compared with control siRNA in the absence of TGF-β1). J, α-SMA reporter was transfected into RLE-6TN cells transduced with lentiviral vector expressing ICAT or control vector expressing GFP. Firefly luciferase activity was measured following treatment with TGF-β1 and normalized to Renilla luciferase activity. Expression of Myc-tagged ICAT was detected by Western blotting using anti-myc Ab (inset). β-Actin is used as loading control (n = 3, *, p < 0.05 compared with cells transduced with GFP control vector treated with TGF-β1).
FIGURE 2.
FIGURE 2.
ICG-001 inhibits TGF-β1-induced α-SMA induction and EMT. Representative Western blot (A) and quantitative analysis (B) of α-SMA protein in RLE-6TN cells treated with TGF-β1 (0.5 ng/ml) ± ICG-001 for 2 days. Lamin A/C is used as loading control. C, RLE-6TN cells were transfected with 764-bp α-SMA reporter, followed by treatment with TGF-β1 (2.5 ng/ml) for 24 h in the presence and absence of ICG-001 (5 μm). Luciferase activity was normalized to Renilla luciferase activity (n = 5, *, p < 0.05 compared with other conditions). D, co-immunoprecipitation of β-catenin with CBP (n = 3) in RLE-6TN cells treated with TGF-β1 ± ICG-001 for 24 h. NE is RLE-6TN nuclear extract which was used as positive control for β-catenin. E, representative immunofluorescence image (n = 3) of staining for α-SMA and phalloidin in RLE-6TN cells treated with TGF-β1 ± ICG-001 for 4 days. RLE-6TN cells treated with TGF-β vehicle are shown as control. Scale bar = 20 μm.
FIGURE 3.
FIGURE 3.
TGF-β-induced α-SMA induction and transcription in RLE-6TN cells is Smad-dependent. Representative Western blot (A) and quantitative analysis (B) of α-SMA protein in RLE-6TN cells treated with TGF-β1 (or vehicle DMSO) ± SIS3. M denotes medium. Lamin A/C is used as a loading control (n = 3, *, p < 0.05 (significantly different from DMSO). Western blot (C) and quantitative analysis of Smad3 (D) and α-SMA (E) protein using cell lysate from RLE-6TN cells transduced with lentivirus expressing shRNA for Smad3 or control pGIPZ nonsilencing shRNA followed by TGF-β1 treatment. Lamin A/C and GAPDH were used as loading controls (n = 3, *, p < 0.05, significantly different from control shRNA). F, RLE-6TN cells were transfected with 764-bp α-SMA reporter, followed by treatment with TGF-β1 (2.5 ng/ml) for 24 h in the presence and absence of SIS3 (6 μm). Luciferase activity was normalized to Renilla luciferase activity (n = 5, *, p < 0.05 compared with TGF-β1 vehicle and TGF-β1/SIS3 together). G, co-transfection of RLE-6TN cells with rat α-SMA promoter reporter, together with Smad3 expression plasmid pRK-5F/Smad3 or empty vector pRK-5F, followed by TGF-β1 treatment for 48 h. Firefly luciferase activity is normalized to Renilla luciferase activity (n = 3, *, p < 0.05, significantly different from the pRK-5F). H, co-transfection of RLE-6TN cells with wild type α-SMAp-Luc and SBE1 and SBE2 mutants α-SMAp-Luc-SBEm1 and α-SMAp-Luc-SBEm2, followed by TGF-β1 treatment for 15 h. Firefly luciferase activity is normalized to Renilla luciferase activity (n = 8, *, p < 0.05; NS, not significantly different).
FIGURE 4.
FIGURE 4.
TGF-β1 induces CBP-dependent interaction between Smad3 and β-catenin and occupancy of the SBE1 region of the α-SMA promoter. A, co-IP was performed with anti-Smad3 Ab using nuclear extracts (NE) harvested from RLE-6TN cells treated with TGF-β1 vehicle and DMSO (lane 1), TGF-β1, and DMSO (lane 2) and TGF-β1 together with SIS3 (3 μm) (lane 3). IgG is used as control (lane 4). Associated β-catenin was analyzed by Western blotting. NE was used as WB control. B, ChIP assay was performed with anti-β-catenin Ab for pull-down using chromatin harvested from RLE-6TN cells in the presence or absence of TGF-β1 (0.5 ng/ml) ± SIS3 (3 μm), followed by amplification of the SBE1-containing region at the α-SMA promoter by qPCR (n = 3). IgG pull-down is used as control. C, ChIP-re-ChIP assay was performed first with mouse IgG (lane 4) or anti-β-catenin Ab (lanes 5, 6, and 7) and then with anti-Smad3 (lane 6) or rabbit IgG (lane 7) for pull-down using chromatin harvested from RLE-6TN cells treated with TGF-β1. Enrichment of SBE1-containing region at the α-SMA promoter was identified by PCR (n = 3). M and NT denote marker and no template, respectively. D, ChIP assay was performed with anti-β-catenin Ab for pull-down using chromatin harvested from RLE-6TN cells in the presence or absence of TGF-β1 (0.5 ng/ml) ± ICG-001 (7.5 μm), followed by amplification of SBE1-containing region at the α-SMA promoter by qPCR (n = 3). ChIP efficiency was calculated relative to untreated cells precipitated with anti-β-catenin Ab, which was set as 1. IgG pull-down is used as control.
FIGURE 5.
FIGURE 5.
TGF-β1 induces β-catenin and CBP occupancy of SBE1 region of the α-SMA promoter. A, ChIP assay was performed with anti-CBP Ab for pull-down using chromatin harvested from RLE-6TN cells treated with TGF-β1 (0.5 ng/ml) ± ICG-001 (7.5 μm), followed by amplification of SBE1-containing region of the α-SMA promoter by qPCR. Data were processed from two pull-downs with qPCR performed in quadruplicate. ChIP efficiency was calculated relative to untreated cells precipitated with anti-CBP Ab, which was set as 1. IgG pull-down is used as control. B, ChIP-re-ChIP assay was performed first with mouse IgG (lane 4) or anti-β-catenin Abs (lanes 5, 6, and 7) and then with anti-CBP (lane 6) or rabbit IgG (lane 7) Abs for pull-down using chromatin harvested from RLE-6TN cells treated with TGF-β1. Enrichment of SBE1-containing region at the α-SMA promoter was identified by PCR (n = 3). M and NT denote marker and no template, respectively.
FIGURE 6.
FIGURE 6.
Co-localization of β-catenin with Smad3 in hyperplastic AT2 cells of IPF lung. A, representative immunofluorescence staining for β-catenin (red) and Smad3 (green) in hyperplastic AT2 cells of IPF lung tissue with anti-β-catenin and anti-Smad3 Abs. Nuclei (blue) are stained with DAPI. B, immunofluorescence staining for β-catenin (red) and pro-SP-C (green, cytoplasmic) in hyperplastic AT2 cells of IPF lung tissue. Nuclei (blue) are stained with DAPI. Scale bar = 20 μm. C, immunofluorescence staining for Smad3 (green) and Nkx2.1 (red, nuclear) in hyperplastic AT2 cells of IPF lung tissue. Nuclei (blue) are stained with DAPI. Scale bar = 20 μm.
FIGURE 7.
FIGURE 7.
Expression of CBP in IPF lung. A, representative immunohistochemistry for staining of CBP (pink) in hyperplastic AT2 cells of IPF lung tissue using anti-CBP Ab. Nuclei (blue) are stained with hematoxylin. Scale bar = 20 μm. B, magnified views of the rectangles shown in A. C, negative control using IgG.
FIGURE 8.
FIGURE 8.
Model for transcriptional regulation of α-SMA by TGF-β1. In addition to initiating phosphorylation of Smad3 and nuclear translocation of p-Smad3, TGF-β1 induces nuclear accumulation of active β-catenin through phosphorylation at Tyr-654 and dephosphorylation at Ser-37 and Thr-41. In the nucleus, a multi-protein complex among p-Smad3, β-catenin, and CBP is formed which interacts with the Smad binding element (SBE) at the α-SMA promoter to regulate α-SMA expression during EMT.

Similar articles

Cited by

References

    1. Whitman M. (1998) Smads and early developmental signaling by the TGFβ superfamily. Genes Dev. 12, 2445–2462 - PubMed
    1. Labbé E., Lock L., Letamendia A., Gorska A. E., Gryfe R., Gallinger S., Moses H. L., Attisano L. (2007) Transcriptional cooperation between the transforming growth factor-β and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Res. 67, 75–84 - PubMed
    1. Nishita M., Hashimoto M. K., Ogata S., Laurent M. N., Ueno N., Shibuya H., Cho K. W. (2000) Interaction between Wnt and TGF-β signaling pathways during formation of Spemann's organizer. Nature 403, 781–785 - PubMed
    1. Labbé E., Letamendia A., Attisano L. (2000) Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-β and wnt pathways. Proc. Natl. Acad. Sci. U.S.A. 97, 8358–8363 - PMC - PubMed
    1. Furuhashi M., Yagi K., Yamamoto H., Furukawa Y., Shimada S., Nakamura Y., Kikuchi A., Miyazono K., Kato M. (2001) Axin facilitates Smad3 activation in the transforming growth factor β signaling pathway. Mol. Cell. Biol. 21, 5132–5141 - PMC - PubMed

Publication types

MeSH terms

-