Skip to main content
Stem Cells International logoLink to Stem Cells International
. 2017 Apr 12;2017:5295286. doi: 10.1155/2017/5295286

Wnt5a Signaling in Normal and Cancer Stem Cells

Yan Zhou 1, Thomas J Kipps 1,2,*, Suping Zhang 1,2,*
PMCID: PMC5405594  PMID: 28491097

Abstract

Wnt5a is involved in activating several noncanonical Wnt signaling pathways, which can inhibit or activate canonical Wnt/β-catenin signaling pathway in a receptor context-dependent manner. Wnt5a signaling is critical for regulating normal developmental processes, including stem cell self-renewal, proliferation, differentiation, migration, adhesion, and polarity. Moreover, the aberrant activation or inhibition of Wnt5a signaling is emerging as an important event in cancer progression, exerting both oncogenic and tumor suppressive effects. Recent studies show the involvement of Wnt5a signaling in regulating normal and cancer stem cell self-renewal, cancer cell proliferation, migration, and invasion. In this article, we review recent findings regarding the molecular mechanisms and roles of Wnt5a signaling in stem cells in embryogenesis and in the normal or neoplastic breast or ovary, highlighting that Wnt5a may have different effects on target cells depending on the surface receptors expressed by the target cell.

1. Introduction

Stem cells, including embryonic stem cells and stem cells identified in adult tissues, have an ability to self-renew and to generate more differentiated progeny. The embryonic stem cell is derived from the inner cell mass of the blastocyst, is pluripotent, and can thus generate all the tissues of the body [1]. Stem cells in adult tissues reside in specialized niches, where they integrate various environmental and intrinsic signaling inputs that determine cell fate and maintain tissue homeostasis.

Wnt factors are a group of these signaling molecules that act on stem cells within the stem cell niche to help maintain their capacity for self-renewal. Wnt factors are known to bind to different receptors to transduce the canonical signaling pathway or noncanonical signaling pathway(s) that regulate diverse biological activities [2, 3]. The canonical Wnt signaling pathway is initiated when Wnt factors bind to Frizzled family receptors (Fzd) and low-density lipoprotein receptor-related protein 5/6 coreceptors (LRP5/6) to form complexes. This results in the recruitment of Axin and dishevelled (Dvl) to the plasma membrane and disruption of the β-catenin degradation complex, leading to accumulation of β-catenin in the cytoplasm, which then translocates to the nucleus, where it binds to T-cell factor (TCF)/lymphoid enhancer factor (LEF) family and activates the transcription of canonical Wnt target genes [4, 5]. In contrast, several Wnt factors activate β-catenin-independent pathways (noncanonical Wnt signaling pathways), known as the Wnt-Ca2+ pathway or planar cell polarity (PCP) pathway, which act to direct cell migration during embryogenesis [68].

Nineteen secreted cysteine-rich Wnt family glycoproteins have been identified in mice and humans. During earlier mammalian embryogenesis or maintenance of adult tissue homeostasis, these Wnt factors and their receptors are dynamically expressed to activate appropriate signaling, to ensure the right balance between proliferation and differentiation. Perturbation of Wnt signaling with aberrant expression of Wnt factors, their receptors, or downstream signaling molecules may lead to the development of several human cancers [6, 9, 10]. Because the tumor initiation capacity seems to be restricted to a small population of tumor cells that are endowed with the capacity for self-renewal and differentiation, they often are referred to as cancer stem cells (CSCs, or tumor initiation cells) [1114]. More importantly, the biology of embryonic stem cells, normal stem cells, and CSCs is highly interrelated. This is evident from the fact that molecular signals (e.g., Wnt signaling) that define and maintain embryonic stem cell or adult normal stem cells are often aberrantly activated in tumor cells.

A major focus of this review is the role of Wnt5a signaling via different surface receptors in the regulation of stem cells in embryogenesis and in the normal or neoplastic breast or ovary, as there already are several excellent reviews that cover aspects of Wnt signaling in colon cancer [14, 15]. We summarized the latest advances on the signaling pathways that are activated by Wnt5a and that pose a conundrum for the rational design of drugs aimed at depleting the CSCs (or tumor initiation cells) within tumors while sparing the function of normal tissues.

2. Wnt5a Signaling Pathway

Wnt5a is a noncanonical Wnt ligand that is evolutionarily conserved and plays an important role in development. Homozygous Wnt5a knockout mice have perinatal lethality due to developmental defects [16]. Previous studies showed that Wnt5a can interact with Fzd2 and receptor tyrosine kinase-like orphan receptor 2 (ROR2) to activate Rac1 in a β-catenin-independent pathway. More recently, we demonstrated that Wnt5a induced heterooligermization of receptor tyrosine kinase-like orphan receptor 1/2 (ROR1/ROR2), which recruited and activated guanine exchange factors (GEFs), which in turn activated RhoA and Rac1, respectively, enhancing leukemia-cell chemotaxis and proliferation [17]. On the other hand, as another action of Wnt5a, it competed with Wnt3a for binding to Fzd2, thereby inhibiting Wnt3a-dependent LRP6 phosphorylation and β-catenin accumulation in vitro and in intact cells, inhibiting the capacity of Wnt3a to induce accumulation of β-catenin, and thereby inhibiting β-catenin-dependent Wnt signaling [18].

However, Wnt5a also could activate β-catenin-dependent pathway and induce secondary axis formation in Xenopus embryos that coexpressed the Fzd5 receptor [19, 20]. Subsequently, another study found that Wnt5a could inhibit canonical Wnt/β-catenin signaling in cells that expressed ROR2, but also could induce canonical Wnt/β-catenin signaling in cells that expressed Fzd4 and LRP5 [21]. Thus, a single Wnt5a ligand can have disparate effects on cells depending on receptor availability. Therefore, the cellular context dictates the effect of Wnt5a. This might account to the observation that the Wnt5a could exert either oncogenic or tumor suppressive effects in different cancers [18, 2123].

3. Wnt5a Signaling in Embryonic Stem Cells

The Wnt pathway (triggered either by Wnt3a, Wnt5a, or Wnt6) also could be involved in the short-term maintenance of pluripotency of both mouse and human ES cells [24]. The first evidence for this was provided by a study of a pharmacological inhibitor of glycogen synthase kinase-3 (GSK3) that could modulate GSK3 activity, leading to activation of β-catenin canonical signaling and increased stability of c-Myc [25, 26]. Wnt5a and Wnt6 subsequently were found to be produced by embryonic fibroblast feeder cells, which could inhibit mouse ES cell differentiation in a serum-dependent manner. Direct activation of β-catenin, using constitutively active β-catenin (S33Y) without disturbing the upstream components of the Wnt/β-catenin, fully recapitulated the effect of Wnts on ES cells. Importantly, Wnt5a also is a potent inhibitor of β-catenin phosphorylation, thereby stabilizing β-catenin [5]. These data suggest that Wnt5a signals to stabilize β-catenin to further activate Wnt/β-catenin canonical signaling in mouse ES cells. Finally, the Wnt/β-catenin pathway can upregulate the mRNA for STAT3, a known regulator of ES cell self-renewal [27], suggesting a molecular mechanism whereby the Wnt/β-catenin pathway acts to prevent ES cell differentiation through convergence on the LIF/JAK-STAT pathway at the level of STAT3 [28]. However, what surface receptors are involved in the activation of Wnt5a/β-catenin signaling in ES cells and whether noncanonical Wnt signaling also could be activated by Wnt5a in ES cells remains unclear.

4. Wnt5a Signaling on Stem Cells in Normal Mammary Gland and Ovarian Tissues

A line of evidence supports the importance of Wnt in maintaining of mammary stem cells in mammary gland [2931]. However, Wnt5a does not induce β-catenin/TCF transactivation activity in mammary gland tissue. The cells of the mammary gland are hyperproliferative in Wnt5a knockout mice, whereas in the presence of ectopic Wnt5a, ductal extension is inhibited [32]. These phenotypes are the inverse of the activation of canonical Wnt/β-catenin signaling. Therefore, Wnt5a may limit and control mammary gland growth by suppressing Wnt/β-catenin signaling. It is puzzling then that this inhibition of mammary gland growth was not observed in transgenic MMTV-Wnt5a strains [33].

In the Drosophila germarium, altered Wingless (Wg; the fly Wnt homologue) signaling occurs in ovarian germline stem cells (GSCs) through removal of its regulators such as dishevelled, armadillo, Axin, and shaggy; this causes GSC loss, influences follicle cell proliferation, and induces differentiation [34, 35]. Using conditional gene targeting for Wnt5a in ovarian granulosa cells (GC) results in the female subfertility associated with increased follicular atresia and decreased rates of ovulation. Further study found that Wnt5a regulates its target genes not by signaling via the Wnt/Ca2+ or planar cell polarity pathway, but rather by inhibiting the canonical pathway, causing both β-catenin and cAMP responsive element binding (CREB) protein levels to decrease via a glycogen synthase kinase-3β-dependent pathway in ovarian granulosa cells (GC). These data indicate that Wnt5a is required for normal ovarian follicle development and can antagonize gonadotropin responsiveness in granulosa cells by suppressing canonical Wnt/β-catenin signaling [36].

Collectively, Wnt5a is most likely to suppress β-catenin signaling in normal stem cells of mammary gland and ovarian tissues, although Wnt5a is able to activate β-catenin signaling in embryonic stem cells. Whether this difference is due to receptors that are differentially expressed in embryonic stem cells versus stem cells of normal mammary gland or the ovary needs to be further investigated.

5. Wnt5a Signaling in Breast Cancer Stem Cells

Wnt factors may affect various mammary epithelial cells and induce an expansion of stem cells during tumor progression [3742]. Moreover, aberrant β-catenin expression was associated with basal, triple-negative breast cancers and poor clinical outcome [43, 44]. The occurrence of aberrant Wnt signaling in specific breast cancer subtypes is less likely dictated by activation caused by somatic mutations. Mutations have not been identified in CTNNB1, which encodes β-catenin, and mutations within APC have been identified in less than 20% of breast cancer patient samples [45]. Further investigation is needed to determine which Wnt ligands and receptors cause this aberrant Wnt/β-catenin signaling.

Recently, a study on MMTV-Wnt1 mouse primary cells found that both recombinant Wnt3a and recombinant Wnt5a could enhance formation of mammospheres in vitro. Wnt5a-induced formation of mammospheres was not caused by an increase in canonical Wnt/β-catenin signaling, but was instead mediated by noncanonical Wnt signaling requiring the receptor tyrosine kinase ROR2 and the activity of the Jun N-terminal kinase, JNK, in mouse breast cancer cells [46]. This is consistent with the observation that patients with breast cancers that express ROR2 had a significantly shorter overall survival than those with tumors lacking expression of ROR2 [47]. However, silencing ROR2 in human breast cancer cell lines decreased both β-catenin-dependent and β-catenin-independent targets [47], suggesting that ROR2 may be involved in both β-catenin-dependent and β-catenin-independent Wnt signaling pathways. ROR1 predominately seems to be expressed by less well-differentiated tumors that have high potential for relapse and metastases and that also express markers associated with epithelial–mesenchymal transition (EMT) [48, 49]. Conversely, silencing ROR1 in human breast cancer cell lines could attenuate expression of genes associated with EMT and impair their migration/invasion capacity in vitro and their metastatic potential in vivo [49]. Very recently, Chien et al. reported that ROR1 expression might be an independent adverse prognostic factor in patients with triple-negative breast cancers [50]. Whether ROR1 can have a similar effect with ROR2 on breast cancer stem cell and what signaling pathway(s) are activated by ROR1 in breast cancer stem cells remains unknown.

A study on a mouse model of ErbB2-induced breast cancer found conflicting evidence on the effect of Wnt5a on human breast cancer stem cells. During ErbB2-induced mammary tumorigenesis, basal tumor-initiating cells (TIC), which exhibited enhanced tumorigenic capacity compared with the corresponding luminal progenitors, preferentially lost Wnt5a expression, as determined by transcript profiling analysis. Moreover, Wnt5a heterozygosity promoted tumor multiplicity and pulmonary metastasis. As a TGFβ substrate, luminal cell-produced Wnt5a induced a feed-forward loop that activated SMAD2 in a RYK- and TGFβR1-dependent manner to limit the expansion of basal TIC in a paracrine fashion, a potential explanation for the suppressive effect of Wnt5a on mammary tumorigenesis [51]. In this mouse model, it remains uncertain whether canonical Wnt/β-catenin signaling was activated or if Wnt5a also inhibited canonical β-catenin signaling to suppress basal TIC expansion.

The Weinberg laboratory studied immortalized human mammary epithelial cells (mMECs) and found that both the canonical and noncanonical Wnt pathways cooperated with TGFβ signaling in not only the maintenance, but intriguingly also the induction, of stem cell properties [52]. Moreover, only noncanonical Wnt ligands Wnt5a and Wnt16 were found upregulated in stem-like cells relative to non-stem-like cells [52]. Therefore, it is possible that Wnt5a or Wnt16 may activate either the canonical or noncanonical Wnt signaling pathway in a receptor context-dependent manner in human breast cancer stem cells.

6. Wnt5a Signaling in Ovarian Cancer Stem Cells

Similar to breast cancer, Wnt5a effect on ovarian cancer is also controversial. An early study of primary ovarian tumors (n = 130) that showed low levels of Wnt5a in ovarian cancer relative to normal ovary is predictive of a poor outcome [53]. Ectopic expression of Wnt5a inhibits the proliferation of human ovarian cancer cell line OVCAR5 both in vitro and in vivo orthotopic ovarian cancer mouse models. Mechanistically, ectopic expression of Wnt5a in OVCAR5 antagonizes canonical Wnt/β-catenin signaling and induces cellular senescence by activating the histone repressor A (HIRA)/promyelocytic leukemia (PML) senescence pathway [53].

In contrast, studies involving a large number of patients found that upregulation of Wnt5a was associated with a relatively poor prognosis [5458]. Compared with the frequency of Wnt1 expression in ovarian cancer, Wnt5a was more frequently found in malignant epithelial ovarian cancer patients (80% out of a total 38) [54]. Of note, patients with ovarian cancers that express high levels of both Wnt1 and Wnt5a had a significantly lower probability of long-term survival than patients with ovarian cancers that did not express Wnt1 or Wnt5a. Furthermore, Wnt5a is prevalent in ascites fluid obtained from women with ovarian cancer [55], suggesting that it contributes to the ovarian tumor microenvironment. This is consistent with the observation that high levels of Wnt5a are associated with increased risk for metastasis [55].

Activating mutations in the canonical Wnt pathway is rare in ovarian cancer, with the exception of endometrioid ovarian cancers [59]. The contribution of canonical Wnt signaling to progression of ovarian cancer remains unknown. Conversely, studies on noncanonical Wnt/β-catenin signaling show that Wnt5a may regulate ovarian cancer EMT, migration, or metastasis via noncanonical signaling pathways [57, 60]. Consistent with this notion, recent studies have found that expression of ROR1, a receptor of Wnt5a, was highly expressed by high-grade and less-differentiated ovarian cancers and associated with a relatively short disease-free survival and overall survival compared to ovarian cancers that did not express ROR1 [6163]. Ovarian cancers that express high levels of ROR1 had gene expression signatures associated with ovarian CSCs [62]. Furthermore, tumor-cell expression of ROR1 apparently correlates with the expression of ALDH1 and the capacity to form tumor spheroids in vitro (both markers of CSC). Treating primary ovarian patient-derived xenograft (PDX) tumor cells, which express high levels of ROR1 with an anti-ROR1 mAb (UC-961 or cirmtuzumab), inhibited spheroid formation and migration in vitro and engraftment and re-engraftment in immune-deficient mice, indicating that ROR1 may influence ovarian cancer stem cell self-renewal. Further studies are needed to determine if Wnt5a is responsible for the influence that ROR1 apparently has on the biology of ovarian cancer stem cells.

7. Concluding Remarks

Wnt5a may play an important role in embryonic stem cell and organ development. However, the role of Wnt5a in cancer stem cells is varied and complex. It may suppress or promote tumor progression. To elucidate molecular mechanisms that drive altered cellular behavior, additional research on tissue-specific expression of specific receptors and coreceptors is needed. In particular, a more detailed understanding of the complex cross-talk between Wnt5a and specific receptors that are expressed in embryonic stem cells and that may be re-expressed or reactivated in cancer stem cells, but not in normal somatic tissues (e.g., ROR1 and ROR2), may enable development of specific inhibitors that block aberrant signaling and thereby favorably impact patient survival.

Acknowledgments

This study was supported by the Breast Cancer Research Foundation (NIH Grant P01-CA081534), the California Institute for Regenerative Medicine, the National Natural Science Foundation of China (NSFC, Grant no. 2016YFC009159), and the Science and Technology Foundation of Shenzhen, China (Shenzhen Peacock Innovation Team Project, Grant no. KQTD20140630100658078).

Conflicts of Interest

The authors declare that there are no conflicts of interest regarding the publication of this paper.

References

  • 1.Reya T., Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843–850. doi: 10.1038/nature03319. [DOI] [PubMed] [Google Scholar]
  • 2.Wodarz A., Nusse R. Mechanisms of Wnt signaling in development. Annual Review of Cell and Developmental Biology. 1998;14(1):59–88. doi: 10.1146/annurev.cellbio.14.1.59. [DOI] [PubMed] [Google Scholar]
  • 3.Logan C. Y., Nusse R. The Wnt signaling pathway in development and disease. Annual Review of Cell and Developmental Biology. 2004;20:781–810. doi: 10.1146/annurev.cellbio.20.010403.113126. [DOI] [PubMed] [Google Scholar]
  • 4.Baarsma H. A., Konigshoff M., Gosens R. The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacology & Therapeutics. 2013;138(1):66–83. doi: 10.1016/j.pharmthera.2013.01.002. [DOI] [PubMed] [Google Scholar]
  • 5.Liu C., Li Y., Semenov M., et al. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002;108(6):837–847. doi: 10.1016/S0092-8674(02)00685-2. [DOI] [PubMed] [Google Scholar]
  • 6.Kikuchi A., Yamamoto H. Tumor formation due to abnormalities in the beta-catenin-independent pathway of Wnt signaling. Cancer Science. 2008;99(2):202–208. doi: 10.1111/j.1349-7006.2007.00675.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Veeman M. T., Axelrod J. D., Moon R. T. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Developmental Cell. 2003;5(3):367–377. doi: 10.1016/S1534-5807(03)00266-1. [DOI] [PubMed] [Google Scholar]
  • 8.Kohn A. D., Moon R. T. Wnt and calcium signaling: beta-catenin-independent pathways. Cell Calcium. 2005;38(3-4):439–446. doi: 10.1016/j.ceca.2005.06.022. [DOI] [PubMed] [Google Scholar]
  • 9.Moon R. T., Kohn A. D., De Ferrari G. V., Kaykas A. WNT and beta-catenin signalling: diseases and therapies. Nature Reviews. Genetics. 2004;5(9):691–701. doi: 10.1038/nrg1427. [DOI] [PubMed] [Google Scholar]
  • 10.Polakis P. The many ways of Wnt in cancer. Current Opinion in Genetics & Development. 2007;17(1):45–51. doi: 10.1016/j.gde.2006.12.007. [DOI] [PubMed] [Google Scholar]
  • 11.Vermeulen L., Sprick M. R., Kemper K., Stassi G., Medema J. P. Cancer stem cells—old concepts, new insights. Cell Death and Differentiation. 2008;15(6):947–958. doi: 10.1038/cdd.2008.20. [DOI] [PubMed] [Google Scholar]
  • 12.Simons B. D., Clevers H. Strategies for homeostatic stem cell self-renewal in adult tissues. Cell. 2011;145(6):851–862. doi: 10.1016/j.cell.2011.05.033. [DOI] [PubMed] [Google Scholar]
  • 13.Visvader J. E. Cells of origin in cancer. Nature. 2011;469(7330):314–322. doi: 10.1038/nature09781. [DOI] [PubMed] [Google Scholar]
  • 14.de Sousa E. M. F., Vermeulen L. Wnt signaling in cancer stem cell biology. Cancer. 2016;8(7) doi: 10.3390/cancers8070060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Asem M. S., Buechler S., Wates R. B., Miller D. L., Stack M. S. Wnt5a signaling in cancer. Cancer. 2016;8(9):p. 79. doi: 10.3390/cancers8090079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yamaguchi T. P., Bradley A., McMahon A. P., Jones S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development. 1999;126(6):1211–1223. doi: 10.1242/dev.126.6.1211. [DOI] [PubMed] [Google Scholar]
  • 17.Yu J., Chen L., Cui B., et al. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. The Journal of Clinical Investigation. 2016;126(2):585–598. doi: 10.1172/JCI83535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sato A., Yamamoto H., Sakane H., Koyama H., Kikuchi A. Wnt5a regulates distinct signalling pathways by binding to Frizzled2. The EMBO Journal. 2010;29(1):41–54. doi: 10.1038/emboj.2009.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.He X., Saint-Jeannet J. P., Wang Y., Nathans J., Dawid I., Varmus H. A member of the frizzled protein family mediating axis induction by Wnt-5A. Science. 1997;275(5306):1652–1654. doi: 10.1126/science.275.5306.1652. [DOI] [PubMed] [Google Scholar]
  • 20.Ishikawa T., Tamai Y., Zorn A. M., et al. Mouse Wnt receptor gene Fzd5 is essential for yolk sac and placental angiogenesis. Development. 2001;128(1):25–33. doi: 10.1242/dev.128.1.25. [DOI] [PubMed] [Google Scholar]
  • 21.Mikels A. J., Nusse R. Purified Wnt5a protein activates or inhibits beta-catenin-TCF signaling depending on receptor context. PLoS Biology. 2006;4(4) doi: 10.1371/journal.pbio.0040115.e115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Torres M. A., Yang-Snyder J. A., Purcell S. M., DeMarais A. A., McGrew L. L., Moon R. T. Activities of the Wnt-1 class of secreted signaling factors are antagonized by the Wnt-5A class and by a dominant negative cadherin in early Xenopus development. The Journal of Cell Biology. 1996;133(5):1123–1137. doi: 10.1083/jcb.133.5.1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.McDonald S. L., Silver A. The opposing roles of Wnt-5a in cancer. British Journal of Cancer. 2009;101(2):209–214. doi: 10.1038/sj.bjc.6605174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sato N., Meijer L., Skaltsounis L., Greengard P., Brivanlou A. H. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nature Medicine. 2004;10(1):55–63. doi: 10.1038/nm979. [DOI] [PubMed] [Google Scholar]
  • 25.Bain J., McLauchlan H., Elliott M., Cohen P. The specificities of protein kinase inhibitors: an update. The Biochemical Journal. 2003;371(Part 1):199–204. doi: 10.1042/BJ20021535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cartwright P., McLean C., Sheppard A., Rivett D., Jones K., Dalton S. LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development. 2005;132(5):885–896. doi: 10.1242/dev.01670. [DOI] [PubMed] [Google Scholar]
  • 27.Raz R., Lee C. K., Cannizzaro L. A., d'Eustachio P., Levy D. E. Essential role of STAT3 for embryonic stem cell pluripotency. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(6):2846–2851. doi: 10.1073/pnas.96.6.2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hao J., Li T. G., Qi X., Zhao D. F., Zhao G. Q. WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Developmental Biology. 2006;290(1):81–91. doi: 10.1016/j.ydbio.2005.11.011. [DOI] [PubMed] [Google Scholar]
  • 29.Constantinou T., Baumann F., Lacher M. D., Saurer S., Friis R., Dharmarajan A. SFRP-4 abrogates Wnt-3a-induced beta-catenin and Akt/PKB signalling and reverses a Wnt-3a-imposed inhibition of in vitro mammary differentiation. Journal of Molecular Signaling. 2008;3(1):p. 10. doi: 10.1186/1750-2187-3-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Badders N. M., Goel S., Clark R. J., et al. The Wnt receptor, Lrp5, is expressed by mouse mammary stem cells and is required to maintain the basal lineage. PloS One. 2009;4(8) doi: 10.1371/journal.pone.0006594.e6594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zeng Y. A., Nusse R. Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell. 2010;6(6):568–577. doi: 10.1016/j.stem.2010.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Roarty K., Serra R. Wnt5a is required for proper mammary gland development and TGF-beta-mediated inhibition of ductal growth. Development. 2007;134(21):3929–3939. doi: 10.1242/dev.008250. [DOI] [PubMed] [Google Scholar]
  • 33.Baxley S. E., Jiang W., Serra R. Misexpression of wingless-related MMTV integration site 5A in mouse mammary gland inhibits the milk ejection response and regulates connexin43 phosphorylation. Biology of Reproduction. 2011;85(5):907–915. doi: 10.1095/biolreprod.111.091645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Forbes A. J., Spradling A. C., Ingham P. W., Lin H. The role of segment polarity genes during early oogenesis in drosophila. Development. 1996;122(10):3283–3294. doi: 10.1242/dev.122.10.3283. [DOI] [PubMed] [Google Scholar]
  • 35.Song X., Xie T. Wingless signaling regulates the maintenance of ovarian somatic stem cells in drosophila. Development. 2003;130(14):3259–3268. doi: 10.1242/dev.00524. [DOI] [PubMed] [Google Scholar]
  • 36.Abedini A., Zamberlam G., Lapointe E., et al. WNT5a is required for normal ovarian follicle development and antagonizes gonadotropin responsiveness in granulosa cells by suppressing canonical WNT signaling. FASEB Journal. 2016;30(4):1534–1547. doi: 10.1096/fj.15-280313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rijsewijk F., Schuermann M., Wagenaar E., Parren P., Weigel D., Nusse R. The drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell. 1987;50(4):649–657. doi: 10.1016/0092-8674(87)90038-9. [DOI] [PubMed] [Google Scholar]
  • 38.Tsukamoto A. S., Grosschedl R., Guzman R. C., Parslow T., Varmus H. E. Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice. Cell. 1988;55(4):619–625. doi: 10.1016/0092-8674(88)90220-6. [DOI] [PubMed] [Google Scholar]
  • 39.Rosner A., Miyoshi K., Landesman-Bollag E., et al. Pathway pathology: histological differences between ErbB/Ras and Wnt pathway transgenic mammary tumors. The American Journal of Pathology. 2002;161(3):1087–1097. doi: 10.1016/S0002-9440(10)64269-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Teissedre B., Pinderhughes A., Incassati A., Hatsell S. J., Hiremath M., Cowin P. MMTV-Wnt1 and -DeltaN89beta-catenin induce canonical signaling in distinct progenitors and differentially activate hedgehog signaling within mammary tumors. PloS One. 2009;4(2) doi: 10.1371/journal.pone.0004537.e4537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Li Y., Hively W. P., Varmus H. E. Use of MMTV-Wnt-1 transgenic mice for studying the genetic basis of breast cancer. Oncogene. 2000;19(8):1002–1009. doi: 10.1038/sj.onc.1203273. [DOI] [PubMed] [Google Scholar]
  • 42.Vaillant F., Asselin-Labat M. L., Shackleton M., Forrest N. C., Lindeman G. J., Visvader J. E. The mammary progenitor marker CD61/beta3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. Cancer Research. 2008;68(19):7711–7717. doi: 10.1158/0008-5472.CAN-08-1949. [DOI] [PubMed] [Google Scholar]
  • 43.Lindvall C., Zylstra C. R., Evans N., et al. The Wnt co-receptor Lrp6 is required for normal mouse mammary gland development. PloS One. 2009;4(6) doi: 10.1371/journal.pone.0005813.e5813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Khramtsov A. I., Khramtsova G. F., Tretiakova M., Huo D., Olopade O. I., Goss K. H. Wnt/beta-catenin pathway activation is enriched in basal-like breast cancers and predicts poor outcome. The American Journal of Pathology. 2010;176(6):2911–2920. doi: 10.2353/ajpath.2010.091125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Geyer F. C., Lacroix-Triki M., Savage K., et al. Beta-catenin pathway activation in breast cancer is associated with triple-negative phenotype but not with CTNNB1 mutation. Modern Pathology. 2011;24(2):209–231. doi: 10.1038/modpathol.2010.205. [DOI] [PubMed] [Google Scholar]
  • 46.Many A. M., Brown A. M. Both canonical and non-canonical Wnt signaling independently promote stem cell growth in mammospheres. PloS One. 2014;9(7) doi: 10.1371/journal.pone.0101800.e101800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Henry C., Quadir A., Hawkins N. J., et al. Expression of the novel Wnt receptor ROR2 is increased in breast cancer and may regulate both beta-catenin dependent and independent Wnt signalling. Journal of Cancer Research and Clinical Oncology. 2015;141(2):243–254. doi: 10.1007/s00432-014-1824-y. [DOI] [PubMed] [Google Scholar]
  • 48.Zhang S., Chen L., Cui B., et al. ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth. PloS One. 2012;7(3) doi: 10.1371/journal.pone.0031127.e31127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cui B., Zhang S., Chen L., et al. Targeting ROR1 inhibits epithelial-mesenchymal transition and metastasis. Cancer Research. 2013;73(12):3649–3660. doi: 10.1158/0008-5472.CAN-12-3832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chien H. P., Ueng S. H., Chen S. C., et al. Expression of ROR1 has prognostic significance in triple negative breast cancer. Virchows Archiv. 2016;468(5):589–595. doi: 10.1007/s00428-016-1911-3. [DOI] [PubMed] [Google Scholar]
  • 51.Borcherding N., Kusner D., Kolb R., et al. Paracrine WNT5A signaling inhibits expansion of tumor-initiating cells. Cancer Research. 2015;75(10):1972–1982. doi: 10.1158/0008-5472.CAN-14-2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Scheel C., Eaton E. N., Li S. H., et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 2011;145(6):926–940. doi: 10.1016/j.cell.2011.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bitler B. G., Nicodemus J. P., Li H., et al. Wnt5a suppresses epithelial ovarian cancer by promoting cellular senescence. Cancer Research. 2011;71(19):6184–6194. doi: 10.1158/0008-5472.CAN-11-1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Badiglian Filho L., Oshima C. T., De Oliveira Lima F., et al. Canonical and noncanonical Wnt pathway: a comparison among normal ovary, benign ovarian tumor and ovarian cancer. Oncology Reports. 2009;21(2):313–320. doi: 10.3892/or_00000223. [DOI] [PubMed] [Google Scholar]
  • 55.Barbolina M. V., Burkhalter R. J., Stack M. S. Diverse mechanisms for activation of Wnt signalling in the ovarian tumour microenvironment. The Biochemical Journal. 2011;437(1):1–12. doi: 10.1042/BJ20110112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Peng C., Zhang X., Yu H., Wu D., Zheng J. Wnt5a as a predictor in poor clinical outcome of patients and a mediator in chemoresistance of ovarian cancer. International Journal of Gynecological Cancer. 2011;21(2):280–288. doi: 10.1097/IGC.0b013e31820aaadb. [DOI] [PubMed] [Google Scholar]
  • 57.Qi H., Sun B., Zhao X., et al. Wnt5a promotes vasculogenic mimicry and epithelial-mesenchymal transition via protein kinase Calpha in epithelial ovarian cancer. Oncology Reports. 2014;32(2):771–779. doi: 10.3892/or.2014.3229. [DOI] [PubMed] [Google Scholar]
  • 58.Henry C., Llamosas E., Knipprath-Meszaros A., et al. Targeting the ROR1 and ROR2 receptors in epithelial ovarian cancer inhibits cell migration and invasion. Oncotarget. 2015;6(37):40310–40326. doi: 10.18632/oncotarget.5643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kim H., Wu R., Cho K. R., et al. Comparative proteomic analysis of low stage and high stage endometrioid ovarian adenocarcinomas. Proteomics. Clinical Applications. 2008;2(4):571–584. doi: 10.1002/prca.200780004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ford C. E., Punnia-Moorthy G., Henry C. E., et al. The non-canonical Wnt ligand, Wnt5a, is upregulated and associated with epithelial to mesenchymal transition in epithelial ovarian cancer. Gynecologic Oncology. 2014;134(2):338–345. doi: 10.1016/j.ygyno.2014.06.004. [DOI] [PubMed] [Google Scholar]
  • 61.Zhang H., Qiu J., Ye C., et al. ROR1 expression correlated with poor clinical outcome in human ovarian cancer. Scientific Reports. 2014;4:p. 5811. doi: 10.1038/srep05811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhang S., Cui B., Lai H., et al. Ovarian cancer stem cells express ROR1, which can be targeted for anti-cancer-stem-cell therapy. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(48):17266–17271. doi: 10.1073/pnas.1419599111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Winter W. E., 3rd, Maxwell G. L., Tian C., et al. Prognostic factors for stage III epithelial ovarian cancer: a Gynecologic Oncology Group study. Journal of Clinical Oncology. 2007;25(24):3621–3627. doi: 10.1200/JCO.2006.10.2517. [DOI] [PubMed] [Google Scholar]

Articles from Stem Cells International are provided here courtesy of Wiley

RESOURCES