Oncogenic KRAS signalling promotes the Wnt/β-catenin pathway through LRP6 in colorectal cancer

E Lemieux, S Cagnol, K Beaudry, J Carrier, N Rivard, E Lemieux, S Cagnol, K Beaudry, J Carrier, N Rivard

Abstract

Aberrant regulation of the Wnt/β-catenin signaling pathway is one of the major causes of colorectal cancer (CRC). Loss-of-function mutations in APC are commonly found in CRC, leading to inappropriate activation of canonical Wnt signaling. Conversely, gain-of-function mutations in KRAS and BRAF genes are detected in up to 60% of CRCs. Whereas KRAS/mitogen-activated protein kinase (MAPK) and canonical Wnt/β-catenin pathways are critical for intestinal tumorigenesis, mechanisms integrating these two important signaling pathways during CRC development are unknown. Results herein demonstrate that transformation of normal intestinal epithelial cells (IECs) by oncogenic forms of KRAS, BRAF or MEK1 was associated with a marked increase in β-catenin/TCF4 and c-MYC promoter transcriptional activities and mRNA levels of c-Myc, Axin2 and Lef1. Notably, expression of a dominant-negative mutant of T-Cell Factor 4 (ΔNTCF4) severely attenuated IEC transformation induced by oncogenic MEK1 and markedly reduced their tumorigenic and metastatic potential in immunocompromised mice. Interestingly, the Frizzled co-receptor LRP6 was phosphorylated in a MEK-dependent manner in transformed IECs and in human CRC cell lines. Expression of LRP6 mutant in which serine/threonine residues in each particular ProlineProlineProlineSerine/ThreonineProline motif were mutated to alanines (LRP6-5A) significantly reduced β-catenin/TCF4 transcriptional activity. Accordingly, MEK inhibition in human CRC cells significantly diminished β-catenin/TCF4 transcriptional activity and c-MYC mRNA and protein levels without affecting β-catenin expression or stability. Lastly, LRP6 phosphorylation was also increased in human colorectal tumors, including adenomas, in comparison with healthy adjacent normal tissues. Our data indicate that oncogenic activation of KRAS/BRAF/MEK signaling stimulates the canonical Wnt/β-catenin pathway, which in turn promotes intestinal tumor growth and invasion. Moreover, LRP6 phosphorylation by ERK1/2 may provide a unique point of convergence between KRAS/MAPK and Wnt/β-catenin signalings during oncogenesis.

Figures

Figure 1
Figure 1
Oncogenic KRAS and activated MEK1 induce EMT and perturb β-catenin localization. (a) Representative phase-contrast microscopy images of IEC-6 cells expressing pBABE (empty vector), KRASG12V, wtMEK or caMEK, and treated or not with 20 μM U0126 during 24 h. (b) Equal amounts of whole-cell lysates were separated by 10% SDS–PAGE and proteins analyzed by western blotting with specific antibodies against E-cadherin, phosphorylated ERK1/2, ERK2 and HA tag. (c) IEC-6 cells stably expressing wtMEK (panels 1–3) or caMEK (panels 4–6), pBABE (panels 7–9) or KRASG12V (panels 10–12) were fixed for immunofluorescence and stained for β-catenin protein (red) and DAPI (blue). Panels 3, 6, 9 and 12. Full overlap of the fluorescence signals (yellow). Representative immunofluorescence images are shown. Bars: 25 μm.
Figure 2
Figure 2
Induction of β-catenin/TCF complex transcriptional activity in IECs transformed by oncogenic KRAS or MEK1. (a, b) IEC-6 cells stably expressing pBABE, KRASG12V, wtMEK or caMEK were transfected with 0.3 μg of TOPFLASH/FOPFLASH reporter genes (a) or c-myc/c-mut (4 × TBE2-wt/4 × TBE2-mut) luciferase reporters (b). Thirty-six hours after transfection, cells were lysed and luciferase activity was measured. The increase in luciferase activity was calculated relative to the level observed in pBABE-expressing cells, which was set at 1. Values were also normalized with Renilla-luciferase vector. Results are the mean±s.e. of at least three separate experiments. Significantly different from respective control at *P<0.05; **P<0.01; or ***P<0.001 (Student's t-test). (c, d) Cells expressing pBABE, KRASG12V, wtMEK or caMEK were treated or not with 20 μM U0126 during 24 h. Thereafter, cells were lyzed and mRNA were analyzed with quantitative real-time PCR for expression of c-Myc (c) and proteins were analyzed by western blotting for the expression of c-Myc, phosphorylated ERK1/2 and total ERK2 (d). (e, f) Cells expressing pBABE, KRASG12V, wtMEK or caMEK were treated or not with 20 μM U0126 during 24 h. Thereafter, cells were lyzed and mRNA analyzed with quantitative real-time PCR for expression of Axin2 and Lef1.
Figure 3
Figure 3
Attenuation of caMEK-driven morphological transformation of IECs occurs upon interference with the β-catenin/TCF4 complex. (a) Subconfluent IEC-6 wtMEK or caMEK cells stably expressing a dominant-negative form of TCF4 (ΔNTCF4) or the empty vector (EV) were transfected with 0.3 μg of the TOPFLASH/FOPFLASH reporter genes and c-myc/c-mut (4 × TBE2-wt/4 × TBE2-mut) luciferase reporters. Thirty-six hours after transfection, cells were lysed and luciferase activity was measured. The luciferase activity was calculated relative to the level observed in EV-expressing cells, which was set at 100%. Values were also normalized with Renilla-luciferase vector. Results are the mean±s.e. of at least three separate experiments. Significantly different from respective control at *P<0.05 or **P<0.01 (Student's t-test). (b) Equal amounts of lysates from IEC-6 wtMEK or caMEK cells stably expressing ΔNTCF4 or E.V. were separated by SDS–PAGE, and proteins analyzed by western blotting with specific antibodies against Tcf4, c-Myc, Fra-1, E-cadherin and total ERK2. (c) Representative phase-contrast microscopy images of IEC-6 caMEK expressing ΔNTCF4 or E.V. (as control). Bars: 50 μm. (d) Representative phase contrast microscopy images of IEC-6 caMEK that were treated or not with 7.5 μM ICG-001 during 36 h. Bars: 25 μm.
Figure 4
Figure 4
Expression of ΔNTCF4 inhibits proliferative, tumoral and invasive properties of cells transformed by activated MEK1. (a) IEC-6 caMEK cells stably expressing ΔNTCF4 or E.V. (as control) were seeded and the number of cells counted during 7 days. (b) IEC-6 caMEK cells stably expressing ΔNTCF4 or E.V. were cultured in soft agarose for 3 weeks before 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) staining. The number of colonies was calculated using the Image J software. (c) Invasion capacity of IEC-6 caMEK cells stably expressing ΔNTCF4 or E.V. through Matrigel was studied using Matrigel-coated Transwells during 48 h. Thereafter, cells were fixed and stained with 0.5% crystal violet solution. (d) Migration of IEC-6 caMEK cells stably expressing ΔNTCF4 or E.V. to the undersurface of the polycarbonate membrane of Boyden chambers was evaluated 24 h after seeding, in presence of 20 μM hydroxyurea. The number of cells in c, d was determined in 10 fields, the experiments performed in duplicate and the number of E.V.-expressing cells, which had migrated was set at 100%. Significantly different from respective control at ***P<0.001 (Student's t-test). (e) Tumor growth over time was measured after subcutaneous injection of 2 × 106 of IEC-6 caMEK cells stably expressing ΔNTCF4 or E.V. The results represent the mean tumor volume obtained from at least six mice injected for each cell line. Independent experiments were performed twice. (f) Representative digital images of mouse lungs 21 days after tail vein injection of 106 IEC-6 caMEK cells expressing E.V. or ΔNTCF4. Similar results were obtained in two independent experiments.
Figure 5
Figure 5
Inhibition of MEK activity in human CRC cell lines significantly reduces β-catenin/TCF complex activity. (a) Subconfluent DLD-1 and HT-29 cells were transfected with 0.3 μg of the TOPFLASH/FOPFLASH luciferase reporter vectors. Twelve hours after transfection, cells were treated or not with 20 μM U0126 during 24 h after which luciferase activity was measured. The luciferase activity was calculated relative to the level observed in dimethylsulphoxide-treated cells, which was set at 1. The luciferase activity was also normalized with Renilla-luciferase vector. Results are the mean±s.e. of at least three separate experiments. Significantly different from untreated cells at *P<0.05; **P<0.01 or ***P<0.0001 (Student's t-test). (b, c) HT-29 and DLD-1 cells were treated or not with 20 μM U0126 during 16 h after which c-myc mRNA levels were evaluated using quantitative real-time PCR, whereas proteins were analyzed by western blotting with specific antibodies against c-MYC, β-catenin, E-cadherin, phosphorylated ERK1/2 and total ERK2. (d) HT-29 cells were treated during 16 h with 20 μM U0126. Thereafter, cells were fixed for immunofluorescence and stained for β-catenin protein (red) and DAPI (blue). (e, f) Cells were treated during 16 h with 20 μM U0126. Thereafter, 800 μg of cell lysates were immunoprecipitated with nontarget IgG (negative control), anti-TCF4 (e) or anti-E-cadherin (f) antibodies. Proteins from immunoprecipitates were solubilized in Laemmli's buffer, separated by 7.5% SDS–PAGE and analyzed by western blotting to determine β-catenin association. IP: immunoprecipitation.
Figure 6
Figure 6
LRP6 is phosphorylated in a MEK-dependent manner in human CRC cells and in IEC-6 expressing oncogenic KRAS, BRAF or MEK1. (a, b) DLD-1 and HT-29 cells were treated or not (DMSO) with 20 μM U0126 during 16 h and equal amounts of cell lysates were separated by SDS–PAGE. In a, proteins were analyzed by western blotting for expression of β-catenin phosphorylated on serine-552, tyrosine-86, tyrosine-654 and tyrosine-142 with phospho-specific antibodies. In addition, β-catenin unphosphorylated on serine-37 and threonine-41 was also analyzed by a specific antibody as well as phosphorylated ERK1/2 and total ERK2. In b, proteins were analyzed by western blotting for expression of total LRP6 and LRP6 phosphorylated on serine-1490 and threonine-1572 as well as phosphorylated ERK1/2 and total ERK2. (c) Equal amounts of lysates from IEC-6 pBABE, KRASG12V, wtMEK and caMEK expressing cells treated or not with 20 μM U0126 during 24 h were analyzed by western blotting for the expression of total ERK2, phosphorylated ERK1/2, total Lrp6 and Lrp6 phosphorylated on serine-1490 and threonine-1572. (d) IEC-6 BRAFV600EER cells were stimulated or not with 250 nM 4-OH tamoxifen in presence or absence of MEK inhibitors (20 μM U0126; 2 μM PD184352) at the indicated times. Proteins were analyzed by western blotting for the expression of total ERK2, phosphorylated ERK1/2, c-Myc, β-actin, total Lrp6 and Lrp6 phosphorylated on threonine-1572 or serine-1490. (e) Mucosal enrichments from 4-week-old BRafIEC-KO and control murine colons were analyzed by western blotting for the expression of phosphorylated ERK1/2 (pERK), ERK2, total Lrp6 and Lrp6 phosphorylated on threonine-1572 or serine-1490. Five mice per group were analyzed and representative western blot analysis of two mice per group is shown.
Figure 7
Figure 7
Oncogenic KRAS signaling triggers β-catenin/TCF4 complex activation via LRP6 phosphorylation. (a) IEC-6 KRASG12Vcells were co-transfected with 0.3 μg of luciferase TOPFLASH/FOPFLASH reporters and increasing concentrations of plasmids expressing or not (E.V., empty vector), wild-type LRP6 or LRP6-5A mutant. Twenty-four hours after transfection, cells were lysed and luciferase activity was measured. The increase in luciferase activity was calculated relative to the level observed in E.V.-expressing cells, which was set at 1. Values were also normalized with Renilla-luciferase vector. Results are the mean±s.e. of at least three separate experiments. (b) IEC-6 BRAFV600EER were co-transfected with 0.3 μg of luciferase TOPFLASH/FOPFLASH reporters and 0.4 μg of plasmids expressing or not (E.V., empty vector) wild-type LRP6 or LRP6-5A mutant. Twenty-four hours after transfection, cells were stimulated or not with 250 nM tamoxifen for an additional 24 h after which luciferase activity was measured as described in a. Significantly different from untreated cells at *P<0.05; **P<0.01 or ***P<0.0001 (Student's t-test). (c) caMEK-expressing cells were stably infected with lentiviruses encoding for a control shRNA (scrambled sequence, shControl) or encoding Lrp6-specific shRNAs (shLrp6A, B or C). After selection, stable table cell populations were lysed and protein lysates were analyzed by western blot for Lrp6 and β-actin protein expression. (d) Cell populations were cultured in soft agarose for 3 weeks before MTT staining. The number of colonies was determined using the ImageJ software. Results are the mean±s.e. of at least three independent experiments. ***, significantly different from shControl cells at P<0.05 (Student's t-test). ****P<0.005.
Figure 8
Figure 8
LRP6 phosphorylation on serine-1490 and threonine-1572 is increased in colorectal adenomas and adenocarcinomas. (a) Expression of total ERK2, LRP6 and phosphorylated LRP6 on serine-1490 and threonine-1572 was investigated by western blotting in seven paired colorectal adenomas (M: normal margins and A: adenomas). (b) Levels of phosphorylated LRP6 were normalized to the levels of total LRP6 levels in each tissue specimen. Tumor-relative phosphorylated/total LRP6 ratios were matched as reference to its normal samples (set at 1) resulting in a dimensionless value (arbitrary units (AU)). Analyzed by paired t-test and * indicates significantly different from normal margins at P⩽0.05. (c) Expression of LRP6 and phosphorylated LRP6 on serine-1490 and threonine-1572 was further investigated in a series of 53 paired specimens (M: resection margins and AC: primary adenocarcinomas) by western blot. Expression levels of phosphorylated LRP6 on serine 1490 and 1572 were normalized to the intensity β-actin expression and to a reference sample, resulting in a dimensionless value (AU). Densitometry of LRP6 phosphorylation in tumor tissues relative to their matched normal samples was analyzed by paired t-test. Significantly different from healthy resection margins **P⩽0.005 and ***P⩽0.001. (d) Representative immunoblot analysis of total LRP6 and LRP6 phosphorylated on threonine 1572 and serine 1490 performed on protein extracts from eight paired resection margins and advanced adenocarcinomas (AC). Tubulin expression is shown as a control of protein loading.

References

    1. 1Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990; 61: 759–767.
    1. 2Gregorieff A, Clevers H. Wnt signalling in the intestinal epithelium: from endoderm to cancer. Genes Dev 2005; 19: 877–890.
    1. 3Taketo MM. Mouse models of gastrointestinal tumors. Cancer Sci 2006; 97: 355–361.
    1. 4Ashton GH, Morton JP, Myant K, Phesse TJ, Ridgway RA, Marsh V et al. Focal adhesion kinase is required for intestinal regeneration and tumorigenesis downstream of Wnt/c-Myc signalling. Dev Cell 2010; 19: 259–269.
    1. 5Sansom OJ, Meniel VS, Muncan V, Phesse TJ, Wilkins JA, Reed KR et al. Myc deletion rescues Apc deficiency in the small intestine. Nature 2007; 446: 676–679.
    1. 6Worthley DL, Leggett BA. Colorectal cancer: molecular features and clinical opportunities. Clin Biochem Rev 2010; 31: 31–38.
    1. 7Pretlow TP, Pretlow TG. Mutant KRAS in aberrant crypt foci (ACF): initiation of colorectal cancer? Biochim Biophys Acta 2005; 1756: 83–96.
    1. 8Smakman N, Borel Rinkes IH, Voest EE, Kranenburg O. Control of colorectal metastasis formation by K-Ras. Biochim Biophys Acta 2005; 1756: 103–114.
    1. 9Saucier C, Rivard N. Epithelial cell signalling in colorectal cancer metastasis. In: Beauchemin N, Huot J (eds) Metastasis of Colorectal Cancer vol.14. Springer: Netherlands, 2010, pp 205–241.
    1. 10Dasari A, Messersmith WA. New strategies in colorectal cancer: biomarkers of response to epidermal growth factor receptor monoclonal antibodies and potential therapeutic targets in phosphoinositide 3-kinase and mitogen-activated protein kinase pathways. Clin Cancer Res 2010; 16: 3811–3818.
    1. 11Dienstmann R, Tabernero J. BRAF as a target for cancer therapy. Anticancer Agents Med Chem 2011; 11: 285–295.
    1. 12Ikenoue T, Kanai F, Hikiba Y, Obata T, Tanaka Y, Imamura J et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res 2005; 65: 4562–4567.
    1. 13Deng G, Bell I, Crawley S, Gum J, Terdiman JP, Allen BA et al. BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer. Clin Cancer Res 2004; 10: 191–195.
    1. 14Li WQ, Kawakami K, Ruszkiewicz A, Bennett G, Moore J, Iacopetta B. BRAF mutations are associated with distinctive clinical, pathological and molecular features of colorectal cancer independently of microsatellite instability status. Mol Cancer 2006; 5: 2.
    1. 15Yuen ST, Davies H, Chan TL, Ho JW, Bignell GR, Cox C et al. Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res 2002; 62: 6451–6455.
    1. 16Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 2002; 418: 934.
    1. 17Ramos JW. The regulation of extracellular signal-regulated kinase (ERK) in mammalian cells. Int J Biochem Cell Biol 2008; 40: 2707–2719.
    1. 18Aliaga JC, Deschenes C, Beaulieu JF, Calvo EL, Rivard N. Requirement of the MAP kinase cascade for cell cycle progression and differentiation of human intestinal cells. Am J Physiol 1999; 277: G631–G641.
    1. 19Rivard N, Boucher MJ, Asselin C, L'Allemain G. MAP kinase cascade is required for p27 downregulation and S phase entry in fibroblasts and epithelial cells. Am J Physiol 1999; 277: C652–C664.
    1. 20Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol 2005; 6: 322–327.
    1. 21Eggstein S, Franke M, Kutschka I, Manthey G, von Specht BU, Ruf G et al. Expression and activity of mitogen activated protein kinases in human colorectal carcinoma. Gut 1999; 44: 834–838.
    1. 22Lee SH, Lee JW, Soung YH, Kim SY, Nam SW, Park WS et al. Colorectal tumors frequently express phosphorylated mitogen-activated protein kinase. APMIS 2004; 112: 233–238.
    1. 23Boucher MJ, Jean D, Vezina A, Rivard N. Dual role of MEK/ERK signalling in senescence and transformation of intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2004; 286: G736–G746.
    1. 24Lemieux E, Bergeron S, Durand V, Asselin C, Saucier C, Rivard N. Constitutively active MEK1 is sufficient to induce epithelial-to-mesenchymal transition in intestinal epithelial cells and to promote tumor invasion and metastasis. Int J Cancer 2009; 125: 1575–1586.
    1. 25Komatsu K, Buchanan FG, Katkuri S, Morrow JD, Inoue H, Otaka M et al. Oncogenic potential of MEK1 in rat intestinal epithelial cells is mediated via cyclooxygenase-2. Gastroenterology 2005; 129: 577–590.
    1. 26Voisin L, Julien C, Duhamel S, Gopalbhai K, Claveau I, Saba-El-Leil MK et al. Activation of MEK1 or MEK2 isoform is sufficient to fully transform intestinal epithelial cells and induce the formation of metastatic tumors. BMC Cancer 2008; 8: 337–2407-8-337.
    1. 27Sebolt-Leopold JS, Dudley DT, Herrera R, Van Becelaere K, Wiland A, Gowan RC et al. Blockade of the MAP kinase pathway suppresses growth of colon tumors in vivo. Nat Med 1999; 5: 810–816.
    1. 28Ikenoue T, Hikiba Y, Kanai F, Aragaki J, Tanaka Y, Imamura J et al. Different effects of point mutations within the B-Raf glycine-rich loop in colorectal tumors on mitogen-activated protein/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase and nuclear factor kappaB pathway and cellular transformation. Cancer Res 2004; 64: 3428–3435.
    1. 29Cagnol S, Rivard N. Oncogenic KRAS and BRAF activation of the MEK/ERK signalling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition. Oncogene 2013; 32: 564–576.
    1. 30Nandan MO, McConnell BB, Ghaleb AM, Bialkowska AB, Sheng H, Shao J et al. Kruppel-like factor 5 mediates cellular transformation during oncogenic KRAS-induced intestinal tumorigenesis. Gastroenterology 2008; 134: 120–130.
    1. 31Brondello JM, Brunet A, Pouyssegur J, McKenzie FR. The dual specificity mitogen-activated protein kinase phosphatase-1 and -2 are induced by the p42/p44MAPK cascade. J Biol Chem 1997; 272: 1368–1376.
    1. 32Brunet A, Pages G, Pouyssegur J. Growth factor-stimulated MAP kinase induces rapid retro-phosphorylation and inhibition of MAP kinase kinase (MEK1). FEBS Lett 1994; 346: 299–303.
    1. 33Gopalbhai K, Meloche S. Repression of mitogen-activated protein kinases ERK1/ERK2 activity by a protein tyrosine phosphatase in rat fibroblasts transformed by upstream oncoproteins. J Cell Physiol 1998; 174: 35–47.
    1. 34Tian X, Liu Z, Niu B, Zhang J, Tan TK, Lee SR et al. E-cadherin/beta-catenin complex and the epithelial barrier. J Biomed Biotechnol 2011; 2011: 567305.
    1. 35Yochum GS, Cleland R, Goodman RH. A genome-wide screen for beta-catenin binding sites identifies a downstream enhancer element that controls c-Myc gene expression. Mol Cell Biol 2008; 28: 7368–7379.
    1. 36Clevers H. Wnt/beta-catenin signalling in development and disease. Cell 2006; 127: 469–480.
    1. 37Naishiro Y, Yamada T, Takaoka AS, Hayashi R, Hasegawa F, Imai K et al. Restoration of epithelial cell polarity in a colorectal cancer cell line by suppression of beta-catenin/T-cell factor 4-mediated gene transactivation. Cancer Res 2001; 61: 2751–2758.
    1. 38van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A et al. The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell 2002; 111: 241–250.
    1. 39Emami KH, Nguyen C, Ma H, Kim DH, Jeong KW, Eguchi M et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription. Proc Natl Acad Sci USA 2004; 101: 12682–12687.
    1. 40Rowan AJ, Lamlum H, Ilyas M, Wheeler J, Straub J, Papadopoulou A et al. APC mutations in sporadic colorectal tumors: A mutational "hotspot" and interdependence of the "two hits". Proc Natl Acad Sci USA 2000; 97: 3352–3357.
    1. 41Yang J, Zhang W, Evans PM, Chen X, He X, Liu C. Adenomatous polyposis coli (APC) differentially regulates beta-catenin phosphorylation and ubiquitination in colon cancer cells. J Biol Chem 2006; 281: 17751–17757.
    1. 42Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H et al. The F-box protein beta-TrCP associates with phosphorylated beta-catenin and regulates its activity in the cell. Curr Biol 1999; 9: 207–210.
    1. 43Piedra J, Miravet S, Castano J, Palmer HG, Heisterkamp N, Garcia de Herreros A et al. p120 Catenin-associated Fer and Fyn tyrosine kinases regulate beta-catenin Tyr-142 phosphorylation and beta-catenin-alpha-catenin Interaction. Mol Cell Biol 2003; 23: 2287–2297.
    1. 44Brembeck FH, Schwarz-Romond T, Bakkers J, Wilhelm S, Hammerschmidt M, Birchmeier W. Essential role of BCL9-2 in the switch between beta-catenin's adhesive and transcriptional functions. Genes Dev 2004; 18: 2225–2230.
    1. 45Roura S, Miravet S, Piedra J, Garcia de Herreros A, Dunach M. Regulation of E-cadherin/Catenin association by tyrosine phosphorylation. J Biol Chem 1999; 274: 36734–36740.
    1. 46Taurin S, Sandbo N, Qin Y, Browning D, Dulin NO. Phosphorylation of beta-catenin by cyclic AMP-dependent protein kinase. J Biol Chem 2006; 281: 9971–9976.
    1. 47Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H et al. Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem 2007; 282: 11221–11229.
    1. 48He XC, Yin T, Grindley JC, Tian Q, Sato T, Tao WA et al. PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nat Genet 2007; 39: 189–198.
    1. 49Krejci P, Aklian A, Kaucka M, Sevcikova E, Prochazkova J, Masek JK et al. Receptor tyrosine kinases activate canonical WNT/beta-catenin signalling via MAP kinase/LRP6 pathway and direct beta-catenin phosphorylation. PLoS ONE 2012; 7: e35826.
    1. 50Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE J et al. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat Genet 2009; 41: 544–552.
    1. 51Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov 2013; 3: 742–750.
    1. 52Casalino L, De Cesare D, Verde P. Accumulation of Fra-1 in ras-transformed cells depends on both transcriptional autoregulation and MEK-dependent posttranslational stabilization. Mol Cell Biol 2003; 23: 4401–4415.
    1. 53Liu J, Pan S, Hsieh MH, Ng N, Sun F, Wang T et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc Natl Acad Sci USA 2013; 110: 20224–20229.
    1. 54Proffitt KD, Virshup DM. Precise regulation of porcupine activity is required for physiological Wnt signalling. J Biol Chem 2012; 287: 34167–34178.
    1. 55Fernandez-Medarde A, Santos E. Ras in cancer and developmental diseases. Genes Cancer 2011; 2: 344–358.
    1. 56Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer 2003; 3: 459–465.
    1. 57Niault TS, Baccarini M. Targets of Raf in tumorigenesis. Carcinogenesis 2010; 31: 1165–1174.
    1. 58Daugherty RL, Gottardi CJ. Phospho-regulation of Beta-catenin adhesion and signalling functions. Physiology (Bethesda) 2007; 22: 303–309.
    1. 59Bienz M. beta-Catenin: a pivot between cell adhesion and Wnt signalling. Curr Biol 2005; 15: R64–R67.
    1. 60Gumbiner BM. Regulation of cadherin adhesive activity. J Cell Biol 2000; 148: 399–404.
    1. 61Harris TJ, Peifer M. Decisions, decisions: beta-catenin chooses between adhesion and transcription. Trends Cell Biol 2005; 15: 234–237.
    1. 62Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science 2004; 303: 1483–1487.
    1. 63Perez-Moreno M, Fuchs E. Catenins: keeping cells from getting their signals crossed. Dev Cell 2006; 11: 601–612.
    1. 64Singh A, Sweeney MF, Yu M, Burger A, Greninger P, Benes C et al. TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers. Cell 2012; 148: 639–650.
    1. 65Horst D, Chen J, Morikawa T, Ogino S, Kirchner T, Shivdasani RA. Differential WNT activity in colorectal cancer confers limited tumorigenic potential and is regulated by MAPK signalling. Cancer Res 2012; 72: 1547–1556.
    1. 66Phelps RA, Chidester S, Dehghanizadeh S, Phelps J, Sandoval IT, Rai K et al. A two-step model for colon adenoma initiation and progression caused by APC loss. Cell 2009; 137: 623–634.
    1. 67Wang C, Zhao R, Huang P, Yang F, Quan Z, Xu N et al. APC loss-induced intestinal tumorigenesis in Drosophila: roles of Ras in Wnt signalling activation and tumor progression. Dev Biol 2013; 378: 122–140.
    1. 68Bennecke M, Kriegl L, Bajbouj M, Retzlaff K, Robine S, Jung A et al. Ink4a/Arf and oncogene-induced senescence prevent tumor progression during alternative colorectal tumorigenesis. Cancer Cell 2010; 18: 135–146.
    1. 69Rad R, Cadinanos J, Rad L, Varela I, Strong A, Kriegl L et al. A genetic progression model of Braf(V600E)-induced intestinal tumorigenesis reveals targets for therapeutic intervention. Cancer Cell 2013; 24: 15–29.
    1. 70Carragher LAS, Snell KR, Giblett SM, Aldridge VSS, Patel B, Cook SJ et al. V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol Med 2010; 2: 458–471.
    1. 71Zeller E, Hammer K, Kirschnick M, Braeuning A. Mechanisms of RAS/beta-catenin interactions. Arch Toxicol 2013; 87: 611–632.
    1. 72Fodde R, Tomlinson I. Nuclear beta-catenin expression and Wnt signalling: in defence of the dogma. J Pathol 2010; 221: 239–241.
    1. 73Tamai K, Zeng X, Liu C, Zhang X, Harada Y, Chang Z et al. A mechanism for Wnt coreceptor activation. Mol Cell 2004; 13: 149–156.
    1. 74Wolf J, Palmby TR, Gavard J, Williams BO, Gutkind JS. Multiple PPPS/TP motifs act in a combinatorial fashion to transduce Wnt signalling through LRP6. FEBS Lett 2008; 582: 255–261.
    1. 75Wu X, Tu X, Joeng KS, Hilton MJ, Williams DA, Long F. Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signalling. Cell 2008; 133: 340–353.
    1. 76Janssen KP, Alberici P, Fsihi H, Gaspar C, Breukel C, Franken P et al. APC and oncogenic KRAS are synergistic in enhancing Wnt signalling in intestinal tumor formation and progression. Gastroenterology 2006; 131: 1096–1109.
    1. 77Luo F, Brooks DG, Ye H, Hamoudi R, Poulogiannis G, Patek CE et al. Mutated K-ras(Asp12) promotes tumourigenesis in Apc(Min) mice more in the large than the small intestines, with synergistic effects between K-ras and Wnt pathways. Int J Exp Pathol 2009; 90: 558–574.
    1. 78Haigis KM, Kendall KR, Wang Y, Cheung A, Haigis MC, Glickman JN et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet 2008; 40: 600–608.
    1. 79Lee SH, Hu LL, Gonzalez-Navajas J, Seo GS, Shen C, Brick J et al. ERK activation drives intestinal tumorigenesis in Apc(min/+) mice. Nat Med 2010; 16: 665–670.
    1. 80Obrador-Hevia A, Chin SF, Gonzalez S, Rees J, Vilardell F, Greenson JK et al. Oncogenic KRAS is not necessary for Wnt signalling activation in APC-associated FAP adenomas. J Pathol 2010; 221: 57–67.
    1. 81Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov 2014; 13: 513–532.
    1. 82Ettenberg SA, Charlat O, Daley MP, Liu S, Vincent KJ, Stuart DD et al. Inhibition of tumorigenesis driven by different Wnt proteins requires blockade of distinct ligand-binding regions by LRP6 antibodies. Proc Natl Acad Sci USA 2010; 107: 15473–15478.
    1. 83Bergeron S, Lemieux E, Durand V, Cagnol S, Carrier JC, Lussier JG et al. The serine protease inhibitor serpinE2 is a novel target of ERK signalling involved in human colorectal tumorigenesis. Mol Cancer 2010; 9: 271–4598-9-271.
    1. 84Simoneau M, Coulombe G, Vandal G, Vezina A, Rivard N. SHP-1 inhibits beta-catenin function by inducing its degradation and interfering with its association with TATA-binding protein. Cell Signal 2011; 23: 269–279.
    1. 85Langlois MJ, Bergeron S, Bernatchez G, Boudreau F, Saucier C, Perreault N et al. The PTEN phosphatase controls intestinal epithelial cell polarity and barrier function: role in colorectal cancer progression. PLoS ONE 2010; 5: e15742.
    1. 86Madison BB, Dunbar L, Qiao XT, Braunstein K, Braunstein E, Gumucio DL. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J Biol Chem 2002; 277: 33275–33283.

Source: PubMed

3
Suscribir