Niclosamide Suppresses Cancer Cell Growth By Inducing Wnt Co-Receptor LRP6 Degradation and Inhibiting the Wnt/β-Catenin Pathway
Abstract
The Wnt/b-catenin signaling pathway is important for tumor initiation and progression. The low density lipoprotein receptor-related protein-6 (LRP6) is an essential Wnt co-receptor for Wnt/b-catenin signaling and represents a promising anticancer target. Recently, the antihelminthic drug, niclosamide was found to inhibit Wnt/b-catenin signaling, although the mechanism was not well defined. We found that niclosamide was able to suppress LRP6 expression and phosphorylation, block Wnt3A-induced b-catenin accumulation, and inhibit Wnt/b-catenin signaling in HEK293 cells. Furthermore, the inhibitory effects of niclosamide on LRP6 expression/phosphorylation and Wnt/b-catenin signaling were conformed in human prostate PC-3 and DU145 and breast MDA-MB-231 and T-47D cancer cells. Moreover, we showed that the mechanism by which niclosamide suppressed LRP6 resulted from increased degradation as evident by a shorter half-life. Finally, we demonstrated that niclosamide was able to induce cancer cell apoptosis, and displayed excellent anticancer activity with IC50 values less than 1 mM for prostate PC-3 and DU145 and breast MDA-MB-231 and T-47D cancer cells. The IC50 values are comparable to those shown to suppress the activities of Wnt/b-catenin signaling in prostate and breast cancer cells. Our data indicate that niclosamide is a unique small molecule Wnt/b-catenin signaling inhibitor targeting the Wnt co-receptor LRP6 on the cell surface, and that niclosamide has a potential to be developed a novel chemopreventive or therapeutic agent for human prostate and breast cancer.Citation
PLoS One. 2011 Dec 16; 6(12):e29290ae974a485f413a2113503eed53cd6c53
10.1371/journal.pone.0029290
Scopus Count
Related articles
- Rottlerin induces Wnt co-receptor LRP6 degradation and suppresses both Wnt/β-catenin and mTORC1 signaling in prostate and breast cancer cells.
- Authors: Lu W, Lin C, Li Y
- Issue date: 2014 Jun
- Silibinin inhibits Wnt/β-catenin signaling by suppressing Wnt co-receptor LRP6 expression in human prostate and breast cancer cells.
- Authors: Lu W, Lin C, King TD, Chen H, Reynolds RC, Li Y
- Issue date: 2012 Dec
- Niclosamide sensitizes triple-negative breast cancer cells to ionizing radiation in association with the inhibition of Wnt/β-catenin signaling.
- Authors: Yin L, Gao Y, Zhang X, Wang J, Ding D, Zhang Y, Zhang J, Chen H
- Issue date: 2016 Jul 5
- Salinomycin suppresses LRP6 expression and inhibits both Wnt/β-catenin and mTORC1 signaling in breast and prostate cancer cells.
- Authors: Lu W, Li Y
- Issue date: 2014 Oct
- Gigantol inhibits Wnt/β-catenin signaling and exhibits anticancer activity in breast cancer cells.
- Authors: Yu S, Wang Z, Su Z, Song J, Zhou L, Sun Q, Liu S, Li S, Li Y, Wang M, Zhang GQ, Zhang X, Liu ZJ, Lu D
- Issue date: 2018 Feb 14
Related items
Showing items related by title, author, creator and subject.
-
Forced Notch Signaling Inhibits Commissural Axon Outgrowth in the Developing Chick Central Nerve SystemShi, Ming; Liu, Zhirong; Lv, Yonggang; Zheng, Minhua; Du, Fang; Zhao, Gang; Huang, Ying; Chen, Jiayin; Han, Hua; Ding, Yuqiang; et al. (2011-01-21)Background: A collection of in vitro evidence has demonstrated that Notch signaling plays a key role in the growth of neurites in differentiated neurons. However, the effects of Notch signaling on axon outgrowth in an in vivo condition remain largely unknown.
-
The Influence of Porphyromonas gingivalis Fimbrial Expression on Human Dendritic Cell Signaling and Innate Immune Homeostatic FunctionsMeghil, Mohamed; Biomedical Sciences (Augusta University, 05-2019)Perturbation of fundamental processes of immune homeostasis, such as apoptosis and autophagy, can result in dire consequences such as autoimmune diseases. Periodontitis is an inflammatory oral disease that is characterized by oral microbial dysbiosis, and deregulation of the host immune response. The aim of this study was to elucidate the underlying mechanisms by which Porphyromonas gingivalis (P. gingivalis) manipulates dendritic cell (DC) signaling to perturb immune homeostasis. Using a combination of Western blotting, flow cytometry, qRT-PCR and immunofluorescence analysis, we show a pivotal role for the minor (Mfa1) fimbriae of P. gingivalis in nuclear/cytoplasmic shuttling of Akt and FOXO1. Mfa1-induced Akt nuclear localization and activation ultimately induced mTOR. The upregulated Akt/mTOR axis was shown to downregulate the intracellular levels of LC3II, an autophagy protein also designated Atg8, required for autophagosome formation and maturation. Further studies utilizing the allosteric panAkt inhibitor MK2206 and rapamycin an mTOR inhibitor confirmed that the activation of Akt/mTOR signaling by P. gingivalis inhibited autophagy in DCs. Concomitant with inhibiting autophagy, we show a pivotal role for Mfa1 fimbriated P. gingivalis in induction of the antiapoptotic protein BCL2, decreased caspase-3 cleavage and decreased expression of pro-apoptotic proteins Bax and Bim in infected DCs, ultimately blocking programmed death of infected DC cells. Importantly, we show that, by using ABT-199 peptide to disrupt interaction of antiapoptotic BCL2 and its proapoptotic partners BAK/BAX, we can restore programmed cell death to P. gingivalis-infected DC cells. In summary, we have identified the underlying mechanism utilized by P. gingivalis to promote the survival of its host immune cells while preventing its own autophagic elimination.
-
Signaling Mechanism of Blood-Retinal Barrier Regulation: Role of Mitogen-ActivatedYang, Jinling; Department of Cellular Biology and Anatomy (2011-03)Breakdown of the blood-retinal barrier (BRB) is an early hallmark of diabetic retinopathy. A critical component in retinal vascular hyper-permeability is increased production of vascular endothelial growth factor (VEGF). VEGF is a potent permeability factor that activates mitogen-activated protein (MAP) kinases. Pigment epithelium-derived factor (PEDF), an endogenous anti-permeability factor, blocks VEGF-induced vascular permeability increase. However, the mechanisms underlying the actions of VEGF and PEDF in regulating endothelial permeability are not yet clear. Previous studies in our laboratory have shown that VEGF induces paracellular permeability via beta-catenin nuclear translocation/transcriptional activation and subsequent upregulation of urokinase plasminogen activator receptor (uPAR). This current study tests the role of two MAP kinases, p38 and extracellular-signal regulated kinase (ERK), in regulating VEGFinduced beta-catenin signaling, uPAR expression and BRB breakdown. We also evaluate the effects of PEDF on this VEGF permeability inducing pathway. The role of MAP kinase in this VEGF permeability inducing pathway was first evaluated using inhibitors of p38 and ERK. These inhibitors preserve the endothelial barrier function upon VEGF treatment. In confluent endothelial cells, cytosolic beta-catenin is phosphorylated by glycogen synthase kinase (GSK) then ubiquitinated and degraded. With VEGF treatment, GSK is phosphorylated/inactive followed by beta-catenin cytosolic accumulation, nuclear translocation and subsequent uPAR expression. These effects were blocked by MAP kinases inhibitors. This indicates p38 and ERK as mediators of VEGF-induced beta-catenin signaling, uPAR expression and endothelial barrier breakdown. Next, it was found that PEDF not only blocks VEGF-induced endothelial permeability increase and MAP kinase activation but also prevents the activation of GSK/beta-catenin signaling as well as uPAR expression. However, PEDF did not block VEGF receptor-2 (VEGFR-2) phosphorylation suggesting that PEDF acts downstream of VEGFR-2 and upstream of MAP kinase level. To further evaluate the role of p38 in regulating VEGF-induced permeability, adenovirusmediated delivery of p38alpha mutants was used. One p38alpha mutant has an altered ATP-binding site thus looses its activity. It is more efficient in blocking VEGF-induced GSK/beta-catenin signaling, uPAR expression and paracellular permeability increase. This study identifies p38alpha and ERK as mediators of VEGF permeability-inducing signaling. They could also serve as potential therapeutic targets for diseases featured by blood-retinal barrier dysfunction.