Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

WNT7A/β-catenin signaling induces FGF1 and influences sensitivity to niclosamide in ovarian cancer

Subjects

Abstract

We previously characterized the link between WNT7A and the progression of ovarian cancer. Other groups have identified FGF1 as a relevant risk factor in ovarian cancer. Here, we show a linkage between these two signaling pathways that may be exploited to improve treatment and prognosis of patients with ovarian cancer. High expression of WNT7A and FGF1 are correlated in ovarian carcinomas and poor overall patient survival. A chromatin immunoprecipitation assay demonstrated that WNT7A/β-catenin signaling directly regulates FGF1 expression via TCF binding elements in the FGF1-1C promoter locus. In vitro gene manipulation studies revealed that FGF1 is sufficient to drive the tumor-promoting effects of WNT7A. In vivo xenograft studies confirmed that the stable overexpression of WNT7A or FGF1 induced a significant increase in tumor incidence, whereas FGF1 knockdown in WNT7A overexpressing cells caused a significant reduction in tumor size. Niclosamide most efficiently abrogated WNT7A/β-catenin signaling in our model, inhibited β-catenin transcriptional activity and cell viability, and increased cell death. Furthermore, niclosamide decreased cell migration following an increase in E-cadherin subsequent to decreased levels of SLUG. The effects of niclosamide on cell functions were more potent in WNT7A-overexpressing cells. Oral niclosamide inhibited tumor growth and progression in an intraperitoneal xenograft mouse model representative of human ovarian cancer. Collectively, these results indicate that FGF1 is a direct downstream target of WNT7A/β-catenin signaling and this pathway has potential as a therapeutic target in ovarian cancer. Moreover, niclosamide is a promising inhibitor of this pathway and may have clinical relevance.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Siegel R, Ma J, Zou Z, Jemal A . Cancer statistics, 2014. CA Cancer J Clin 2014; 64: 9–29.

    Google Scholar 

  2. Goff BA, Mandel LS, Melancon CH, Muntz HG . Frequency of symptoms of ovarian cancer in women presenting to primary care clinics. JAMA 2004; 291: 2705–2712.

    Article  CAS  Google Scholar 

  3. Teo R, Mohrlen F, Plickert G, Muller WA, Frank U . An evolutionary conserved role of Wnt signaling in stem cell fate decision. Dev Biol 2006; 289: 91–99.

    Article  CAS  Google Scholar 

  4. Polakis P . Wnt signaling and cancer. Genes Dev 2000; 14: 1837–1851.

    CAS  Google Scholar 

  5. Liu W, Xing F, Iiizumi-Gairani M, Okuda H, Watabe M, Pai SK et al. N-myc downstream regulated gene 1 modulates Wnt-beta-catenin signalling and pleiotropically suppresses metastasis. EMBO Mol Med 2012; 4: 93–108.

    Article  Google Scholar 

  6. Polakis P . The many ways of Wnt in cancer. Curr Opin Genet Dev 2007; 17: 45–51.

    Article  CAS  Google Scholar 

  7. Gatcliffe TA, Monk BJ, Planutis K, Holcombe RF . Wnt signaling in ovarian tumorigenesis. Int J Gynecol Cancer 2008; 18: 954–962.

    Article  CAS  Google Scholar 

  8. Arend RC, Londono-Joshi AI, Straughn JM Jr, Buchsbaum DJ . The Wnt/beta-catenin pathway in ovarian cancer: a review. Gynecol Oncol 2013; 131: 772–779.

    Article  CAS  Google Scholar 

  9. Yoshioka S, King ML, Ran S, Okuda H, MacLean JA 2nd, McAsey ME et al. WNT7A regulates tumor growth and progression in ovarian cancer through the WNT/beta-catenin pathway. Mol Cancer Res 2012; 10: 469–482.

    Article  CAS  Google Scholar 

  10. Friesel RE, Maciag T . Molecular mechanisms of angiogenesis: fibroblast growth factor signal transduction. FASEB J 1995; 9: 919–925.

    Article  CAS  Google Scholar 

  11. Powers CJ, McLeskey SW, Wellstein A . Fibroblast growth factors, their receptors and signaling. Endocr Relat Cancer 2000; 7: 165–197.

    Article  CAS  Google Scholar 

  12. Birrer MJ, Johnson ME, Hao K, Wong KK, Park DC, Bell A et al. Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas. J Clin Oncol 2007; 25: 2281–2287.

    Article  CAS  Google Scholar 

  13. Meng QH, Xu E, Hildebrandt MA, Liang D, Lu K, Ye Y et al. Genetic variants in the fibroblast growth factor pathway as potential markers of ovarian cancer risk, therapeutic response, and clinical outcome. Clin Chem 2014; 60: 222–232.

    Article  CAS  Google Scholar 

  14. Smith G, Ng MT, Shepherd L, Herrington CS, Gourley C, Ferguson MJ et al. Individuality in FGF1 expression significantly influences platinum resistance and progression-free survival in ovarian cancer. Br J Cancer 2012; 107: 1327–1336.

    Article  CAS  Google Scholar 

  15. Lu W, Lin C, Roberts MJ, Waud WR, Piazza GA, Li Y . Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/beta-catenin pathway. PLoS ONE 2011; 6: e29290.

    Article  CAS  Google Scholar 

  16. Mook RA Jr, Chen M, Lu J, Barak LS, Lyerly HK, Chen W . Small molecule modulators of Wnt/beta-catenin signaling. Bioorg Med Chem Lett 2013; 23: 2187–2191.

    Article  CAS  Google Scholar 

  17. Osada T, Chen M, Yang XY, Spasojevic I, Vandeusen JB, Hsu D et al. Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations. Cancer Res 2011; 71: 4172–4182.

    Article  CAS  Google Scholar 

  18. Sack U, Walther W, Scudiero D, Selby M, Kobelt D, Lemm M et al. Novel effect of antihelminthic Niclosamide on S100A4-mediated metastatic progression in colon cancer. J Natl Cancer Inst 2011; 103: 1018–1036.

    Article  CAS  Google Scholar 

  19. Wieland A, Trageser D, Gogolok S, Reinartz R, Hofer H, Keller M et al. Anticancer effects of niclosamide in human glioblastoma. Clin Cancer Res 2013; 19: 4124–4136.

    Article  CAS  Google Scholar 

  20. Chen M, Wang J, Lu J, Bond MC, Ren XR, Lyerly HK et al. The anti-helminthic niclosamide inhibits Wnt/Frizzled1 signaling. Biochemistry 2009; 48: 10267–10274.

    Article  CAS  Google Scholar 

  21. Hendrix ND, Wu R, Kuick R, Schwartz DR, Fearon ER, Cho KR . Fibroblast growth factor 9 has oncogenic activity and is a downstream target of Wnt signaling in ovarian endometrioid adenocarcinomas. Cancer Res 2006; 66: 1354–1362.

    Article  CAS  Google Scholar 

  22. Aletti GD, Gallenberg MM, Cliby WA, Jatoi A, Hartmann LC . Current management strategies for ovarian cancer. Mayo Clin Proc 2007; 82: 751–770.

    Article  Google Scholar 

  23. Parkin DM, Bray F, Ferlay J, Pisani P . Global cancer statistics, 2002. CA Cancer J Clin 2005; 55: 74–108.

    Article  Google Scholar 

  24. Chotani MA, Chiu IM . Differential regulation of human fibroblast growth factor 1 transcripts provides a distinct mechanism of cell-specific growth factor expression. Cell Growth Differ 1997; 8: 999–1013.

    CAS  Google Scholar 

  25. Madiai F, Hackshaw KV, Chiu IM . Characterization of the entire transcription unit of the mouse fibroblast growth factor 1 (FGF-1) gene. Tissue-specific expression of the FGF-1.A mRNA. J Biol Chem 1999; 274: 11937–11944.

    Article  CAS  Google Scholar 

  26. Barker N, Clevers H . Mining the Wnt pathway for cancer therapeutics. Nat Rev 2006; 5: 997–1014.

    CAS  Google Scholar 

  27. Chen W, Chen M, Barak LS . Development of small molecules targeting the Wnt pathway for the treatment of colon cancer: a high-throughput screening approach. Am J Physiol Gastrointest Liver Physiol 2010; 299: G293–G300.

    Article  CAS  Google Scholar 

  28. Dodge ME, Lum L . Drugging the cancer stem cell compartment: lessons learned from the hedgehog and Wnt signal transduction pathways. Annu Rev Pharmacol Toxicol 2011; 51: 289–310.

    Article  CAS  Google Scholar 

  29. Meijer L, Flajolet M, Greengard P . Pharmacological inhibitors of glycogen synthase kinase 3. Trends Pharmacol Sci 2004; 25: 471–480.

    Article  CAS  Google Scholar 

  30. Steele IA, Edmondson RJ, Bulmer JN, Bolger BS, Leung HY, Davies BR . Induction of FGF receptor 2-IIIb expression and response to its ligands in epithelial ovarian cancer. Oncogene 2001; 20: 5878–5887.

    Article  CAS  Google Scholar 

  31. Zaid TM, Yeung TL, Thompson MS, Leung CS, Harding T, Co NN et al. Identification of FGFR4 as a potential therapeutic target for advanced-stage, high-grade serous ovarian cancer. Clin Cancer Res 2013; 19: 809–820.

    Article  CAS  Google Scholar 

  32. Chamorro MN, Schwartz DR, Vonica A, Brivanlou AH, Cho KR, Varmus HE . FGF-20 and DKK1 are transcriptional targets of beta-catenin and FGF-20 is implicated in cancer and development. EMBO J 2005; 24: 73–84.

    Article  CAS  Google Scholar 

  33. Curtin JC, Lorenzi MV . Drug discovery approaches to target Wnt signaling in cancer stem cells. Oncotarget 2010; 1: 563–577.

    Google Scholar 

  34. Huang SM, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009; 461: 614–620.

    Article  CAS  Google Scholar 

  35. Yo YT, Lin YW, Wang YC, Balch C, Huang RL, Chan MW et al. Growth inhibition of ovarian tumor-initiating cells by niclosamide. Mol Cancer Ther 2012; 11: 1703–1712.

    Article  CAS  Google Scholar 

  36. Cooke PS, Spencer TE, Bartol FF, Hayashi K . Uterine glands: development, function and experimental model systems. Mol Hum Reprod 2013; 19: 547–558.

    Article  CAS  Google Scholar 

  37. Sassoon D . Wnt genes and endocrine disruption of the female reproductive tract: a genetic approach. Mol Cell Endocrinol 1999; 158: 1–5.

    Article  CAS  Google Scholar 

  38. Balgi AD, Fonseca BD, Donohue E, Tsang TC, Lajoie P, Proud CG et al. Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling. PLoS ONE 2009; 4: e7124.

    Article  Google Scholar 

  39. Fonseca BD, Diering GH, Bidinosti MA, Dalal K, Alain T, Balgi AD et al. Structure-activity analysis of niclosamide reveals potential role for cytoplasmic pH in control of mammalian target of rapamycin complex 1 (mTORC1) signaling. J Biol Chem 2012; 287: 17530–17545.

    Article  CAS  Google Scholar 

  40. Jin Y, Lu Z, Ding K, Li J, Du X, Chen C et al. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. Cancer Res 2010; 70: 2516–2527.

    Article  CAS  Google Scholar 

  41. Li R, Hu Z, Sun SY, Chen ZG, Owonikoko TK, Sica GL et al. Niclosamide overcomes acquired resistance to erlotinib through suppression of STAT3 in non-small cell lung cancer. Mol Cancer Ther 2013; 12: 2200–2212.

    Article  CAS  Google Scholar 

  42. You S, Li R, Park D, Xie M, Sica GL, Cao Y et al. Disruption of STAT3 by niclosamide reverses radioresistance of human lung cancer. Mol Cancer Ther 2014; 13: 606–616.

    Article  CAS  Google Scholar 

  43. Kim NG, Koh E, Chen X, Gumbiner BM . E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components. Proc Natl Acad Sci USA 2011; 108: 11930–11935.

    Article  CAS  Google Scholar 

  44. Lau MT, Klausen C, Leung PC . E-cadherin inhibits tumor cell growth by suppressing PI3K/Akt signaling via beta-catenin-Egr1-mediated PTEN expression. Oncogene 2011; 30: 2753–2766.

    Article  CAS  Google Scholar 

  45. Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G . A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 1998; 392: 190–193.

    Article  CAS  Google Scholar 

  46. Takeichi M . Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol 1993; 5: 806–811.

    Article  CAS  Google Scholar 

  47. Hayashi K, Erikson DW, Tilford SA, Bany BM, Maclean JA 2nd, Rucker EB 3rd et al. Wnt genes in the mouse uterus: potential regulation of implantation. Biol Reprod 2009; 80: 989–1000.

    Article  CAS  Google Scholar 

  48. Hayashi K, Burghardt RC, Bazer FW, Spencer TE . WNTs in the ovine uterus: potential regulation of periimplantation ovine conceptus development. Endocrinology 2007; 148: 3496–3506.

    Article  CAS  Google Scholar 

  49. Chauhan SC, Vannatta K, Ebeling MC, Vinayek N, Watanabe A, Pandey KK et al. Expression and functions of transmembrane mucin MUC13 in ovarian cancer. Cancer Res 2009; 69: 765–774.

    Article  CAS  Google Scholar 

  50. Singh AP, Moniaux N, Chauhan SC, Meza JL, Batra SK . Inhibition of MUC4 expression suppresses pancreatic tumor cell growth and metastasis. Cancer Res 2004; 64: 622–630.

    Article  CAS  Google Scholar 

  51. Maclean JA 2nd, Hu Z, Welborn JP, Song HW, Rao MK, Wayne CM et al. The RHOX homeodomain proteins regulate the expression of insulin and other metabolic regulators in the testis. J Biol Chem 2013; 288: 34809–34825.

    Article  CAS  Google Scholar 

  52. Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res 2008; 14: 5198–5208.

    Article  CAS  Google Scholar 

  53. Denkert C, Budczies J, Darb-Esfahani S, Gyorffy B, Sehouli J, Konsgen D et al. A prognostic gene expression index in ovarian cancer - validation across different independent data sets. J Pathol 2009; 218: 273–280.

    Article  Google Scholar 

  54. Bonome T, Levine DA, Shih J, Randonovich M, Pise-Masison CA, Bogomolniy F et al. A gene signature predicting for survival in suboptimally debulked patients with ovarian cancer. Cancer Res 2008; 68: 5478–5486.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Gail Isenberg for editing the manuscript. This work was supported by NIH/NCI CA179214 and ACS-IL 139038 (to KH), and NIH/NICHD HD065584 (to JAM).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to K Hayashi.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

King, M., Lindberg, M., Stodden, G. et al. WNT7A/β-catenin signaling induces FGF1 and influences sensitivity to niclosamide in ovarian cancer. Oncogene 34, 3452–3462 (2015). https://doi.org/10.1038/onc.2014.277

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2014.277

This article is cited by

Search

Quick links