The Wnt Gene Family in Mammary Gland Development


The purpose of this review is to consider the involvement of Wnt proteins in mammary gland development. Advances in Wnt biology that are derived from studies of other systems are summarised in the reviews indicated.

Historical Perspective

The prototype member of the Wnt gene family, Wnt-1 (formerly known as int-1) was first isolated as a common site of integration by the mouse mammary tumour virus (MMTV) and was later shown to be homologous to the Drosophila segment polarity gene wingless [1-3] . In mouse mammary epithelial cells, MMTV integration activates Wnt-1 by increasing transcription of Wnt-1 mRNA leading to increased (tumourigenic) levels of the normal Wnt-1 protein. Wnt-1 has been shown to be a member of a large gene family that is highly conserved through evolution (reviewed in [4-6] ) and Wnt gene products have been implicated in the control of numerous developmental processes such as axis formation in Xenopus embryos [7] , murine hindbrain and kidney development [8-11] . Biochemical studies of Wnt proteins in cell culture and in Drosophila have shown that Wnt polypeptides are glycosylated, secreted and associate with the cell surface/extracellular matrix [12-16] . Microinjection analyses in Xenopus have shown functional differences between Wnt family members and have shown that the determinants of Wnt-specificity are localised to the C-terminal half of the protein [17] . Genetic studies of wingless in Drosophila, have identified genes with related phenotypes and have ordered them into a Wingless signal transduction pathway (reviewed in [18] ).

Evidence for the involvement of Wnts in mammary development

Although MMTV expression of Wnt-1 and Wnt-3 drives the formation of mouse mammary tumours [19-21] , neither gene is expressed at detectable levels in the mammary gland, making it unlikely that they play a role in postnatal mammary development. By contrast, several other members of the Wnt gene family are expressed. However definitive proof of the requirement for any Wnt gene in the mammary gland is currently lacking. What follows is a summary of the evidence that strongly points to a role for Wnt gene products in mammary development.

a. Correlations
Northern analysis of mouse mammary gland RNA at different stages of postnatal development has so far shown the presence of mouse Wnt-2, Wnt-4, Wnt-5a, Wnt-5b, Wnt-6, Wnt-7b and Wnt-10b messages. Studies of human breast material from reduction mammoplasties have additionally shown the presence of Wnt-3 and Wnt-7a. The following table summarises developmental timecourse data [20-22, 42] .


table

The exact timecourse of Wnt expression varies for each gene, allowing large numbers of 'potential' correlations to be drawn with phases of mammary development / changes in systemic hormones. Wnt-2, Wnt-5a, Wnt-7b and Wnt-10b are expressed during ductal development at the earliest times examined (5-6 weeks of age). The time at which expression of these genes is initiated is currently unknown and will require analyses of earlier (possibly fetal) timepoints. By contrast, the increase in expression of Wnt-4, Wnt-5b and Wnt-6 during pregnancy suggests an involvement of these genes in lobular development. During lactation, expression of most Wnt messages is down-regulated, while some but not all genes are re-induced during involution. Localisation studies show Wnt expression in both the stromal and epithelial compartments of the mammary gland, raising the possibility that Wnts could be involved in both stromal-epithelial and epithelial-stromal interactions [22, 23] . Expression of some Wnt genes is reduced in ovariectomised animals, but it is not clear whether these changes are the direct consequences of hormonal regulation or if the changes in Wnt expression mediate hormonal regulation of mammary development.

b. Investigating Function Through Ectopic Expression
Cell culture studies
C57MG mammary epithelial cells and 10T1/2 fibroblasts have been used to compare the consequences of ectopic expression of different members of the murine Wnt gene family. Expression of Wnt-1 in C57MG and 10T1/2 cells causes cells to 'morphologically transform' ie. to elongate and overgrow [24, 25] . In C57MG cells, expression of Wnt-1, Wnt-2, Wnt-3A, Wnt-5B, Wnt-7A and Wnt-7B induced morphological transformation (with different efficiencies), while Wnt-4, Wnt-5A and Wnt-6 were non-transforming. In 10T1/2 fibroblasts, expression of Wnt-1, Wnt-6 and Wnt-7B caused 'morphological transformation', while expression of Wnt-4 and Wnt-5B had no obvious effect. These data show that cell lines differ in their sensitivity to Wnts and that Wnts can be divided into at least two classes: transforming and non-transforming. C57MG cells respond to Wnt-1 expression by down-regulating endogenous Wnt-4 and Wnt-5a mRNA levels and altering levels of ß-catenin [26-28] . A potential role for Wnt-5A in mammary morphogenesis was implied by the observation that Wnt-5A message levels are strongly down-regulated in human luminal epithelial cells cultured in a collagen matrix in the presence of HGF/scatter factor [29].

Transgenic mice and the 'Transgenic' fat pad
Ectopic expression of Wnt-1 in vivo has been achieved using transgenic mice and recombinant retroviruses. In both systems, expression of Wnt-1 caused the formation of a characteristic hyperplasia with a feathery morphology that occupied the majority of the mammary fat pad in virgin animals [30, 31] . The molecular mechanisms which generated the Wnt-1 hyperplastic phenotype are not understood, but indirect arguments suggest that Wnt-1 enhances the proliferation of the mammary epithelial cells in vivo. Recombinant retroviruses have recently been used to show that Wnt-4 induces a 'pregnancy-like' hyperplasia in the glands of virgin mice, By contrast, retroviruses containing Wnt-5B, Wnt-6 and Wnt-7B cDNAs failed to give rise to gross morphological changes although failure of viral transfer could not be excluded as an alternative reason for the lack of phenotype ( [32] ,; Unpublished data J. Bradbury, PAW Edwards, T. C. Dale).

Knock out mice
Knockout mice have been described for the Wnt-1, Wnt-3A, Wnt-4 and Wnt-7A genes [8, 11, 33-35] . Of these, only Wnt-4 is normally expressed in the mammary gland and might be expected to give a mammary phenotype. Unfortunately the role of Wnt-4 in mammary development cannot be easily assessed as Wnt-4 mutant mice suffer from defective kidney tubulogenesis and die before birth. The phenotypes of further Wnt null phenotypes are eagerly awaited.


Implications for Mammary Development and Tumourigenesis

Wnt expression is maximal during periods of morphological change and is lost during the terminal differentiation that accompanies lactation, suggesting Wnts may control developmental and morphological changes in the gland. An apparent problem with this hypothesis is that expression of some genes remains high in the mature virgin gland after ductal end bud development has ceased. However, these observations may not be inconsistent because significant levels of cell proliferation (and presumably apoptosis) occur in the virgin gland [36, 37] . The developmental patterns of expression TGFß, FGF, EGF and HGF/Scatter mRNAs show a similar range to that of Wnt family members [38-41] , suggesting that both Wnt factors may co-operate with members of other ligand families during mammary development. This contention is supported by the observation that basic FGF can regulate the expression of Wnt-4 and Wnt-5A in C57MG cells [28] , while several members of the FGF family co-operate with MMTV-Wnt-1 in the generation of mouse mammary tumours [42-45].

The best current model to explain Wnt induced neoplasia is the theory that Wnt-1 and Wnt-3 functionally substitute for one or more endogenous Wnt genes and drive inappropriate cellular proliferation. As ectopic Wnt-1 expression decouples mammary growth from a dependence on estrogen and progesterone and the expression of some endogenous Wnts is dependent on ovarian function [23, 46] , the model for Wnt-1 function may be further refined:

image

The identity of the endogenous homologue(s) for Wnt-1/3 in the above model would be expected to include pregnancy-induced Wnts as the morphology of Wnt-1 hyperplasias mimic aspects of pregnancy development. Although ectopic expression of Wnt-4 also generates a pregnancy-like hyperplasia, the phenotype is clearly different from that induced by Wnt-1, suggesting that Wnt-1 does not simply substitute for the function of Wnt-4. Wnts that are normally expressed in the mammary gland may be involved in tumourigenesis. Studies have shown amplifications of the Wnt-2 gene in mammary tumours from GR mice [47] . Interestingly, we have recent data that suggests that a proportion of breast carcinomas express high levels of WNT-2 (T. C. Dale, S. Weber-Hall K.Smith, E. Huguet, B.A. Gusterson, A. Harris. In preparation). As murine Wnt-2 and human WNT-2 are both normally expressed in the stromal compartment, this suggests that tumourigenesis may involve WNT-2 switching compartments to the tumour epithelium, with the possible creation of an autocrine loop. Overexpression of Wnt-5A and Wnt-7B in benign and malignant mammary tumours has also been documented [48-50] . In the case of Wnt-5A, expression was increased by a mechanism that did not involve gene amplification. The overexpression of Wnt genes in human breast cancer raises the possibility that 'functional substitution' may also be involved in human tumourigenesis. More detailed studies of the role of Wnt function in vivo should shed light on these processes.

Future directions

In the long term, studies in the mammary gland should be able to identify the networks of gene products whose expression defines the ductal and lobular developmental motifs utilised in other organs (eg. lung and salivary gland). A particularly interesting aspect of these studies will be an understanding of how these processes are brought under hormonal control. In the short term, two technical approaches should be powerful in the analysis of Wnt function. The first is the characterisation of mammary development in 'knockout' mice. The second is the recent availability of the soluble Wnt factors which will allow the characterisation of Wnt signal transduction pathways in the mammalian context [51, 52].

Acknowledgements

Many thanks to Stuart Naylor and Dave Cook for reading the manusript before submission. My apologies to anyone who is missed or incorrectly quoted in this summary. To make the most of the electronic medium, I would be happy to include changes, comments and suggestions in the next revision.


References

1. Nusse, R. and H.E. Varmus, Many tumours induced by the mouse mammary t umour virus contain provirus integrated in the same region of the host genome. Cell, 1982. 31: p. 99-109.

2. van, O.A. and R. Nusse, Structure and nucleotide sequence of the putative mammary oncogene int-1; proviral insertions leave the protein-encoding domain i ntact. Cell, 1984. 39(1): p. 233-40.


3. Bender, W. and M. Peifer, Oncogenes take wing. Cell, 1987. 50(4): p. 519- 20.


4. McMahon, A.P., The Wnt family of developmental regulators. Trends in Genetics, 1992. 8(7): p. 236-242.

5. McMahon, A.P., A super family of putative developmental signaling molecules related to the Proto-oncogene Wnt-1/int-1. Adv. Dev. Biol, 1992. 1: p. 31-60.

6. Nusse, R. and H.E. Varmus, Wnt Genes. Cell, 1992. 69: p. 1073-1087.

7. Moon, R.T., et al., Dissecting Wnt signalling pathways and Wnt-sensitive developmental processes through transient misexpression analyses in embryos of Xenopus laevis. Development (Supplement), 1993. : p. 85-94.

8. McMahon, A.P. and A. Bradley, The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell, 1990. 62(6): p. 1073-85.

9. McMahon, A.P., et al., The midbrain-hindbrain phenotype of Wnt-1-Wnt1- Mice results from stepwise deletion of engrailed-expressing cells by 9.5 days postcoitum. Cell, 1992. 69: p. 581-595.

10. Thomas, K.R., et al., Swaying is a mutant allele of the proto-oncogene Wnt-1. Cell, 1991. 67: p. 969-976.

11. Stark, K., et al., Epthelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature, 1994. 372: p. 679-682.

12. Gonzalez, F., et al., Secretion and movement of wingless protein in the epidermis of the Drosophila embryo. Mech Dev, 1991. 35(1): p. 43-54.

13. Papkoff, J. and B. Schryver, Secreted int-1 protein is associated with the cell surface. Mol Cell Biol, 1990. 10(6): p. 2723-30.

14. van den Heuvel, M., et al., Distribution of the wingless gene product in Drosophila embryos: a protein involved in cell-cell communication. Cell, 1989. 59(4): p. 739-49.

15. Smolich, B.D., et al., Wnt family proteins are secreted and associated with the cell surface. Mol. Biol. Cell, 1994. 4(12): p. 1267-75.

16. Papkoff, J., Identification and biochemical characterization of secreted Wnt-1 protein from P19 embryonal carcinoma cells induced to differentiate along the neuroectodermal lineage. Oncogene, 1994. 9: p. 313-317.

17. Du, S.J., et al., Identification of distinct classes and functional domains of Wnts through expression of wild-type and chimeric proteins in Xenopus embryos. Mol Cell Biol, 1995. 15(5): p. 2625-34.

18. Klingensmith, J. and R. Nusse, Signaling by wingless in Drosophila. Dev Biol, 1994. 166(2): p. 396-414.

19. Roelink, H., et al., Wnt-3, a gene activated by proviral insertion in mouse mammary tumors, is homologous to int-1/Wnt-1 and is normally expressed i n mouse embryos and adult brain. Proc Natl Acad Sci U S A, 1990. 87(12): p. 4519-23.

20. Roelink, H. and R. Nusse, Expression of two members of the Wnt family during mouse development--restricted temporal and spatial patterns in the developing neural tube. Genes Dev, 1991. 5(3): p. 381-8.

21. Gavin, B.J. and A.P. McMahon, Differential regulation of the Wnt gene family during pregnancy and lactation suggests a role in postnatal development of the mammary gland. Mol Cell Biol, 1992. 12(5): p. 2418-23.

22. Buhler, T.A., et al., Localization and quantification of Wnt-2 gene expression i n mouse mammary development. Developmental Biology, 1993. 155: p. 87- 96.

23. Weber-Hall, S.J., et al., Developmental and hormonal regulation of Wnt gene expression in the mouse mammary gland. Differentiation, 1994. 57: p. 205- 214.

24. Bradbury, J.M., et al., Alterations of the growth characteristics of the fibroblast cell line C3H 10T1/2 by members of the Wnt gene family. Oncogene, 1994. 9: p. 2597-2603.

25. Wong, G.T., B.J. Gavin, and A.P. McMahon, Differential transformation of mammary epithelial cells by Wnt genes. Mol. Cell. Biol., 1994. 14(9): p. 6278-6286.

26. Hinck, L., W.J. Nelson, and J. Papkoff, Wnt-1 modulates cell-cell adhesion in mammalian cells by stabilizing ß-catenin binding to the cell adhesion protein cadherin. J. Cell Biol., 1994. 124(5): p. 729-741.

27. Hinck, L., et al., ß-catenin: a common target for the regulation of cell adhesion by Wnt-1 and Src signaling pathways. TIBS, 1994. 19: p. 538-542.

28. Olson, D.J. and J. Papkoff, Regulated expression of Wnt family members during proliferation of C57MG mammary cells. Cell Growth and Differentiation, 1994. 5: p. 197-206.

29. Huguet, E.L., et al., Regulation of Wnt5a mRNA expression in human mammary epithelial cells by cell shape, confluence, and hepatocyte growth factor. J. Biol. Chem., 1995. 270: p. 12851-12856.

30. Tsukamoto, A.S., et al., 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): p. 619-25.

31. Edwards, P.A.W., et al., Hyperplasia of mouse mammary epithelium induced by expression of the Wnt-1 (int-1) oncogene in reconstituted mammary gland. Oncogene, 1992. 7: p. 2041-2051.

32. Bradbury, J.M., et al., Wnt-4 expression induces a pregnancy-like growth pattern in reconstituted mammary glands in virgin mice. Dev. Biol., 1995. 170: p. 553-563.

33. Parr, B.A. and A.P. McMahon, Dorsalizing signal Wnt-7a required for normal polarity of D-V and A-P axes of mouse limb. Nature, 1995. 374: p. 350-353.

34. Takada, S., et al., Wnt-3a regulates somite and tailbud formation in the mouse embryo. Genes Dev, 1994. 8(2): p. 174-89.

35. Thomas, K.R. and M.R. Capecchi, Targeted disruption of the murine int-1 proto-oncogene resulting in severe abnormalities in midbrain and cerebellar development. Nature, 1990. 346(6287): p. 847-50.

36. Zwierzchowski, L., et al., Variation of DNA polymerase activities and DNA synthesis in Mouse Mammary Gland During Pregnancy and Early Lactation. Differentiation, 1984. 28: p. 179-185.

37. Joshi, K., et al., Cellular proliferation in the rat mammary gland during pregnancy and lactation. Lab Invest, 1986. 54(1): p. 52-61.

38. Robinson, S., G. Silberstein, and C.W. Daniel, Evidence supporting a role for TGF-beta Isoforms in growth regulation and functional differentiation of the mouse mammary gland., in Mechanisms Regulating Lactation and Infant Nutrient Utilization., M.E. Picciano and B. Lonnerdal, Editor. 1992, Wiley- Liss: New York. p. 43-52.

39. Coleman-Krnacik, S. and J.M. Rosen, Differential temporal and spatial gene expression of fibroblast growth factor family members during mouse mammary gland development. Mol. Endocrinol., 1994. 8(2): p. 218-229.

40. Snedeker, S.M., C.F. Brown, and R.P. DiAugustine, Expression and functional properties of transforming growth factor alpha and epidermal growth factor during mouse mammary gland ductal morphogenesis. Proc Natl Acad Sci U S A, 1991. 88(1): p. 276-80.

41. Niranjan, B., et al., HGF/SF: A potent cytokine for mammary growth, morphogenesis and development. Development, 1995. 121: p. 2897-2908.

42. MacArthur, C.A., D.B. Shankar, and G.M. Shackleford, Fgf-8, activated by proviral insertion, cooperates with the Wnt-1 transgene in murine mammary tumorigenesis. J Virol, 1995. 69(4): p. 2501-7.

43. Shackleford, G.M., et al., Mouse mammary tumor virus infection accelerates mammary carcinogenesis in Wnt-1 transgenic mice by insertional activation of int-2/Fgf-3 and hst/Fgf-4. Proc Natl Acad Sci U S A, 1993. 90(2): p. 740-4.

44. Lee, F.S., et al., Insertional mutagenesis identifies a member of the Wnt gene family as a candidate oncogene in the mammary epithelium of int-2/Fgf-3 transgenic mice. Proc. Natl. Acad. Sci, 1995. 92: p. 2268-2272.

45. Kwan, H., et al., Transgenes expressing the Wnt-1 and int-2 proto-oncogenes cooperate during mammary carcinogenesis in doubly transgenic mice. Mol Cell Biol, 1992. 12(1): p. 147-54.

46. Lin, T.P., et al., Role of endocrine, autocrine, and paracrine interactions in the development of mammary hyperplasia in Wnt-1 transgenic mice. Cancer Res, 1992. 52(16): p. 4413-9.

47. Nusse, R., The Wnt gene family in tumorigenesis and in normal development. J. Steroid Biochem. Molec. Biol., 1992. 43(1): p. 9-12.

48. Huguet, E.L., et al., Differential expression of human Wnt genes 2,3,4 and 7B in human breast cell lines and normal and disease states of human breast tissue. Cancer Research, 1994. 54: p. 2615-2621.

49. Lejeune, S., et al., Wnt5a cloning, expression and upregulation in human primary breast cancers. Clinical Cancer Research, 1995. 1: p. 215-222.

50. Iozzo, R.V., I. Eichstetter, and K.G. Danielson, Aberrant expression of the growth factor Wnt-5A in human malignancy. Cancer Res, 1995. 55(16): p. 3495-9.

51. Bradley, R.S. and A. Brown, A soluble form of Wnt-1 protein with mitogenic activity on mammary epithelial cells. Mol. Cell. Biol, 1995. 15: p. 4616-4622.

52. van Leeuwen, F., S.H. Samos, and R. Nusse, Biological activity of soluble wingless protein in cultured drosophila imaginal disc cells. Nature, 1994. 368: p. 342-344.


contributed by Trevor Dale
Institute of Cancer Research
Haddow Laboratories
15 Cotswold Rd.
Sutton, Surrey
SM2 5NG, UK

tel. 44-181-643-8901
FAX 44-181-643-0238
e-mail ( trevor@icr.ac.uk)


submitted: March 15, 1996 (revised June 1998)


back to the home page