SciELO - Scientific Electronic Library Online

 
vol.35 issue2Extracellular signals, cell interactions and transcription factors involved in the induction of the neural crest cellsT-kininogen inhibits kinin-mediated activation of ERK in endothelial cells author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

Related links

Share


Biological Research

Print version ISSN 0716-9760

Biol. Res. vol.35 no.2 Santiago  2002

http://dx.doi.org/10.4067/S0716-97602002000200019 

Biol Res 35: 277-286, 2002

Wnt signaling: A complex issue

BART HENDRIKS AND ERNST REICHMANN*

Fetal and Pediatric Surgical Research Unit, Department of Surgery
University Children's Hospital Zürich. Steinwiesstr. 75, CH-8032 Zürich, Switzerland

ABSTRACT

The development of tissues and organs in multicellular organisms is controlled by the interplay of several signaling pathways that cross-talk to provide positional information and induce cell fate specification. Together with other families of secreted factors such as TGFßs, FGFs, Hedgehog and Notch proteins, Wnt growth factors are crucially implicated in these processes. Here, we will first discuss molecular mechanisms and then consider some biological consequences of Wnt signaling.

Key terms: Wnts, frizzled, LRP 5/6, ß-catenin, APC, TCF, stem cells, cancer

INTRODUCTION

The first Wnt gene, mouse Wnt-1 (Int-1), was discovered in 1982 as a proto oncogene in mammary tumors activated by integration of the mouse mammary tumor virus (Nusse and Varmus, 1982). As a consequence, the potential involvement of Wnt genes in cancer became a major area of research in the 1980s (Nusse and Varmus, 1992). With the molecular identification of the Drosophila segment polarity gene wingless (wg) as the orthologue of Wnt-1 (Cabrera et al., 1987; Rijsewijk et al., 1987) and the phenotypic analysis of Wnt-1 mutations in the mouse (McMahon and Bradley, 1990; Thomas and Capecchi, 1990), it became clear that Wnt genes are important regulators of many developmental decisions (Parr and McMahon, 1994; Cadigan and Nusse, 1997) At present, close to 100 Wnt genes have been isolated from species ranging from human to the nematode Caenorhabditis elegans and the simple metazoan Hydra. Presumably, all Wnt proteins are secreted from cells and act through cell surface receptors either on the producing cell or on adjacent cells to determine cell fate or other differentiation parameters.

Within target cells, Wnt signaling involves the control of several dynamically interacting protein complexes. In that context, the protein ß-catenin is of particular importance since this multitude of protein-protein interactions is employed to control ß-catenin levels via phosphorylation and degradation. Wnt signaling, in essence, liberates ß-catenin from these complexes thereby favoring translocation to the nucleus, where ß-catenin associates with the T cell factor (TCF) to activate transcription of target genes.

OLECULAR MECHANISMS THAT UNDERLIE WNT SIGNALING

Wnt-genes and growth factors

The Wnt genes encode a large family of secreted protein growth factors that have been identified in animals from hydra to humans. The name Wnt (pronounced `wint') denotes the relationship of this family to the Drosophila segment polarity gene `wingless' and to its vertebrate ortholog, int-1, a mouse protooncogene. Transcription of Wnt family genes appears to be developmentally regulated in a precise temporal and spatial manner (see also Aybar et al., 2002). All of the known vertebrate Wnt genes encode for 38- to 43-kDa cysteine-rich putative glycoproteins, which have features typical of secreted growth factors: a hydrophobic signal sequence, a conserved asparagine-linked oligosaccharide consensus sequence, and 22 conserved cysteine residues whose relative spacing is maintained. In humans, 19 WNT proteins have been identified that share 27% to 83% amino-acid sequence identity and a conserved pattern of 23 or 24 cysteine residues. During development, Wnts have diverse roles in governing cell fate, proliferation, migration, polarity, and death. In adults, Wnts function in homeostasis, and inappropriate activation of the Wnt pathway is implicated in a variety of cancers.

Wnt-Receptors

Wnt ligands signal via seven transmembrane spanning receptors of the Frizzled family (ten members in the human genome) together with the recently identified LRP5 and LRP6 coreceptors, which are members of the low-density lipoprotein receptor-related protein family (LRP) (Wodarz and Nusse, 1998; Tamai et al., 2000). LRP6 associates with Frizzled in a Wnt-dependent manner (Fig. 1). A mutant of LRP6 lacking the intracellular domain blocks signaling by Wnt in Xenopus embryos. Deletion of the LRP6 gene in mice results in developmental defects that are strikingly similar to those caused by mutations of individual Wnt genes. Wnt stimulation of LRP5 has been shown to induce interaction with Axin and subsequent activation of Wnt target genes (Mao et al., 2001).

It has been shown that Wnt signaling is inhibited by the secreted protein Dickkopf 1 (Dkk1), a member of a multigene family, which induces head formation in amphibian embryos (Glinka et al., 1998) (Fig. 1). Dkk1 has been demonstrated to inhibit Wnt signaling by binding to and antagonizing LRP5/6 (Mao et al., 2001). There is recent evidence that the transmembrane proteins Kremen 1 and Kremen2 are high-affinity Dkk1 receptors that functionally cooperate with Dkk1 to block Wnt signaling (Fig. 1). Kremen 2 forms a ternary complex with Dkk1 and LRP6, and induces rapid endocytosis and removal of the LRP6 from the plasma membrane. From these data it is concluded that Kremen 1 and Kremen 2 are components of a membrane complex which modulates canonical Wnt signaling through LRP6 in vertebrates (Mao et al., 2002).


Fig. 1. Diversification of the Wnt signaling pathway and the dual role role of ß-catenin. See text for details.
This scheme is adapted from Figure 1 in Current Opinion in Genetics & Development 2001, 11:547-553 by J. Huelsken and W. Birchmeier

The canonical Wnt pathway

The canonical Wnt pathway (ß-catenin pathway) leads to activation of target genes in the nucleus (Fig.1). In this pathway ß-Catenin/Armadillo is stabilized by preventing its degradation in proteasomes. Activated Frizzled receptors signal through a conserved motif (Umbhauer et al., 2000) to Dishevelled but the mechanism of Dishevelled activation is still ill-defined. Casein kinase Ie has recently been identified as an essential positive regulator that acts downstream of Dishevelled and regulates ß-catenin stability (Sakanaka et al., 1999; Peters et al., 1999). Axin/Conductin, in cooperation with the tumor suppressor gene product APC, promote ß-catenin degradation (Fig. 1). This involves serine-threonine phosphorylation of the amino-terminus of ß-catenin by GSK3ß (Fig. 1) and subsequent ubiquitination (Behrens et al., 1998; Polakis, 2001). Stabilized ß-catenin accumulates in the cytoplasm and is translocated to the nucleus, where it interacts with members of the LEF/TCF family of transcription factors and activates gene expression (Behrens et al., 1996; Molenaar et al., 1996) (Fig. 1).

It has recently been shown that the Drosophila segment polarity gene legless (lgs), a homolog of human BCL 9, functions as an adaptor protein to physically link Pygo to the ß-catenin-TCF complex (Fig. 1). This recruitment of Pygo is required for ß-catenin to function as a transcriptional coactivator and hence to enable the Wnt/Wg system to induce target genes (Kramps et al., 2002) (Fig. 1).

LEF/TCF can prevent transcriptional activation when bound to transcriptional co-repressors like CREB-binding protein (CBP), CtBP or members of the Groucho family (see Fig. 1 and Sharpe et al., 2001 for a review). Other results indicate that chromatin remodeling may be an important aspect of Wnt-induced gene regulation (Sharpe et al., 2001; Collins and Treisman, 2000; Sawa et al., 2000).

THE TCF/LEF FAMILY OF TRANSCRIPTION FACTORS

The founding members of the TCF/LEF family of transcription factors, TCF-1 and LEF-1, were identified in screens for T cell-specific transcription factors. TCF-1 was identified by its ability to bind to the CD3e enhancer, whereas LEF-1 was found in a screen for proteins binding to the TCRa enhancer and a site in the HIV LTR (Oosterwegel et al., 1991; van de Wetering et al., 1991; Travis et al., 1991). In more recent years, two additional family members were identified in mammals: TCF-3 and TCF-4 (Korinek et al., 1998). The Drosophila genome only contains one TCF gene, called dtcf or pangolin (Brunner et al., 1997; van de Wetering et al., 1997). Similarly, a single gene, Pop-1, resides in the genome of the nematode Caenorhabditis elegans (Lin et al., 1995). Proteins of the TCF/LEF family contain an 80-amino-acid high mobility group (HMG) box. HMG boxes bind DNA as monomers and can do so in a sequence-specific manner (Grosschedl et al., 1994; Laudet et al., 1993). However, the HMG box not only recognizes specific DNA sequences, but also induces a dramatic bend in the DNA structure (Giese et al., 1992; Dooijes et al., 1993). In doing so, HMG boxes may coordinate the binding of other transcription factors (Grosschedl et al., 1994; Giese et al., 1995; Bianchi and Beltrame, 1998).

This "DNA-bending" activity of TCF/LEFs, in conjunction with the observation that these factors cannot directly activate transcription in reporter assays, suggests that TCF/LEF family members primarily serve an architectural function. LEF-1 appears unique in that it contains a context-dependent activation domain (CAD) (Carlsson et al., 1993), which can activate transcription in the presence of the coactivator ALY (Bruhn et al., 1997). The other TCF family members do not appear to contain a CAD domain (Van de Wetering et al., 1996).

TCF/Lef mRNAs undergo extensive alternative splicing. In addition, the best studied gene of this family, TCF-1, also exhibits alternative promoter usage. Over 100 TCF-1 isoforms may theoretically be produced. However, Western blotting has revealed the predominant expression of only eight different TCF-1 isoforms (Van de Wetering et al., 1996). LEF-1 exhibits two splice variants (Waterman et al., 1991) and also possesses two promoters (Hovanes et al., 2001). Several C-terminal splice variants of TCF-4 exist that exhibit strong sequence homology to similar TCF-1 mRNAs (Korinek et al., 1997; Duval et al., 2000) (Fig. 1). While the functional relevance of alternative splice products from TCF/Lef genes is currently unclear, alternative promoter usage of both genes generates protein isoforms that either contain or lack the N-terminal ß-catenin interaction domain.

Target genes of Wnt signaling

In most models, Wnt signaling is predominantly implicated in control of cell fate by altering the transcriptional program of target cells in an instructive fashion. Consistent with these models, mutation of Wnt genes or inappropriate expression of Wnts usually lead to changes in cell fate as a consequence of altered gene expression. Not surprisingly, many genes regulated directly or indirectly by Wnts are transcription factors or secreted signaling molecules thought to represent key players in the plethora of genes controlling development. Among the best-studied examples are members of the homeobox gene family, including engrailed (en), ultrabithorax (ubx) and siamois. For two of these genes, ubx and siamois, there is evidence for direct transcriptional activation by binding of an Arm/ß-catenin complex together with HMG-box proteins to specific sites in the target gene promoter region (Brannon et al., 1997; Riese et al., 1997). Both these studies found that transcriptional activation by Wnt signaling alone is not sufficient to explain the normal expression pattern of target genes. Other sources of patterning information appear to be integrated at the level of the target gene promoter, giving rise to the final transcriptional pattern. Such combinatorial gene activation may explain how Wnt signaling can regulate a large number of different target genes and participate in apparently unrelated processes depending on the cellular context in which signaling takes place. One example for tissue-specific differences in target gene regulation is the effect of Wg on the expression of achaete (ac), a proneural gene. In the wing imaginal disc, Wg activates ac expression (Couso et al., 1994), whereas in the eye imaginal disc, Wg acts as a repressor of ac (Cadigan and Nusse, 1996). Cooperation with other signaling pathways at the level of target gene promoters may also explain why ubiquitous expression of Wg does not lead to ubiquitous expression of Wg target genes (Noordermeer et al., 1992; Sampedro et al., 1993; Baylies et al., 1995).

The generation of transcriptionally active TCF/ß-catenin complexes in tumor cells results in the inappropriate activation of TCF target genes, including c-myc and cyclin D1, (He et al., 1998; Tetsu and McCormick, 1999) which may ultimately lead to cancer. The expression of c-myc was found to be changed after induction of wild-type APC in an APC-deficient colon carcinoma cell line. Subsequent analysis showed that the c-myc promoter was activated by TCF-4/ß-catenin via specific binding sites, and that of endogenous c-myc could be blocked by a dominant-negative TCF-4 (He et al., 1998). Because c-myc overexpression previously had been linked to colorectal tumorigenesis, it is possible that TCF-4/ß-catenin induces neoplastic growth via activation of c-myc. Furthermore, this study indicates that several other genes associated with tumorigenesis are up- or down-regulated by APC. Components of the c-jun family and the metalloproteinase matrilysin were also shown to be targets of TCF/ß-catenin (Mann et al., 1999; Crawford et al., 1999).

Diversification of the Wnt signal

More recently, at least four different branches are being distinguished within the Wnt pathway. These include the canonical Wnt pathway, the planar cell polarity pathway, the spindle orientation and asymmetric cell division pathway and the Wnt/Ca2+ pathway. These branches will be discussed in the order indicated. The canonical Wnt pathway has been described above.

In the planar cell polarity pathway Wnt signaling activates Jun-N-terminal kinase (JNK) and directs asymmetric cytoskeletal organization and coordinated polarization of cell morphology within the plane of epithelial sheets. Examples of Wnt-mediated JNK activation are seen during eye and wing development of Drosophila and gastrulation movements in Zebrafish and Xenopus (Mlodzik, 2000; Heisenberg et al., 2000). This pathway branches at the level of Dishevelled (Fig. 1) and involves downstream components like the small guanosine triphosphatase Rho and a kinase cascade including Misshapen (a Ste20 homologue), JNK kinase and JNK (Mlodzik, 2000; Wallingford et al., 2000). Drosophila Rho-associated kinase (Drok) was identified recently as a target of the pathway that regulates the activity of myosin and hence provides a direct link to the cytoskeleton (Winter et al., 2001).

The mechanism underlying Wnt signaling divergence into either the canonical or the planar cell polarity pathway is presently the subject of intense investigation. Particular Wnts and Frizzled receptors, like Wnt11 or Frizzled 7, specifically activate the JNK pathway, as described for convergent extension movements of the mesoderm during Xenopus and zebrafish gastrulation (Heisenberg et al., 2000). The Wnt target gene naked/naked cuticle was identified recently as an antagonist for Wnt signaling. Naked protein binds to Dishevelled and blocks ß-catenin but stimulates the JNK pathway (Zeng et al., 2000; Yan et al., 2001; Rousset et al., 2001).

Several members of the Wnt-signaling pathway like GSK3ß or APC have been implicated in the pathway which regulates spindle orientation and asymmetric cell division in C. elegans and Drosophila. In the central nervous system of Drosophila, neuroepithelial cells divide symmetrically along the planar axis. Neuroblasts originate from this layer by asymmetric divisions and undergo further differentiation (Lu et al., 2001). The integrity of adherens junctions in the neuroepithelium was shown to provide a planar polarity cue required for symmetrical division, and APC associated with adherens junctions as well as microtubule-associated EB1 are necessary for this process (Lu et al., 2001).

The Wnt/Ca2+ pathway leads to release of intracellular calcium, possibly via a G-protein mediated process (Fig. 1). This pathway involves activation of phospholipase C, protein kinase C and calmodulin-dependent kinase II and is implicated in Xenopus ventralization and in the regulation of convergent extension movements (Kuhl et al., 2000; Wallingford et al., 2001).

BIOLOGICAL ASPECTS OF WNT SIGNALING

Wnt signaling in body axis formation and mesoderm patterning

ß-Catenin and other components of the canonical Wnt pathway have previously been found to be essential for body axis formation in Xenopus and Zebrafish (Moon and Kimelman, 1998; Solnica-Krezel, 1999). In Xenopus, accumulation of ß-catenin on the dorso-anterior side of the embryo is the earliest sign of axis formation and precedes gastrulation (Schneider et al., 1996; Larabell et al., 1997). Overexpression of ß-catenin induces formation of an additional embryonic axis (Heasman et al., 1994). Interestingly, members of the Wnt-signaling cascade have also been implicated recently in establishing the body axis in Hydra, a member of the evolutionary ancient metazoan phylum Cnidaria (Hobmayer et al., 2000). This suggests that Wnt signaling is an evolutionarily conserved process important for axial differentiation in all multicellular animals.

Wnt signaling in cell fate specification and stem cell control

Stem cells are pluripotent cells found in many tissues, that can adopt various fates. When exposed to particular growth factors and cytokines, they generate progenitors that proliferate transiently and then withdraw from the cell cycle and terminally differentiate (Fuchs and Segre, 2000). Currently, a great deal of research is directed towards identifying molecules that maintain stem cells and that control their commitment to particular lineages. Recently, the Wnt/ß-catenin signaling pathway was implicated in the control of stem cell specification in the skin (Huelsken et al., 2001). Skin stem cells reside in the bulges of hair follicles and are bipotent, as they give rise to both keratinocytes of the hair follicle and the interfollicular epidermis (Cotsarelis et al., 1999; Taylor et al., 2000; Jensen et al., 1999). These bulge stem cells represent the reservoir that allows both self-renewal of the epidermis and cyclic regeneration of hair follicles. Keratin 14-Cre mediated conditional ablation of the ß-catenin gene in the skin of mice blocked the differentiation of stem cells into the follicular lineages, since keratinocytes could only adopt the epidermal fate (Huelsken et al., 2001). In wild-type skin, ß-catenin might cooperate in the stem cells with LEF-1 and/or TCF-3 transcription factors to control these decisions over cell fate (van Genderen et al., 1994; DasGupta and Fuchs, 1999; Merrill et al., 2001). Detailed analysis revealed that, in the absence of ß-catenin, skin stem cells fail to initiate follicular morphogenesis but do give rise to dermal cysts that exhibit features of epidermal differentiation, while lacking markers of follicular keratinocytes. These cysts and the associated stem cells remain observable for many weeks in the skin of knock out mice. The data imply that ß-catenin is necessary for instructing stem cells to form follicular keratinocytes. ß-catenin alone, however, may be insufficient to induce follicular differentiation of keratinocytes, since additional mesenchymal signals are likely to be required.

Wnt signaling in cancer

Wnts do not appear to play a dominant role in human cancer. Rather, mutations in downstream components of the Wnt cascade are a major cause of several types of cancer. APC is mutated at both alleles in 80% of all colon carcinomas (Polakis et al., 1999). These mutations almost invariably lead to a truncation of APC, removing its interaction domains for ß-catenin and axin. As a consequence, the activity of the destruction complex is impaired and cytosolic ß-catenin is no longer phosphorylated and degraded. Thus accumulated ß-catenin in turn inappropriately activates one of the TCF/LEF family members, TCF4, that is specifically expressed in the intestinal epithelium (Korinek et al., 1997). In a significant fraction of sporadic colon tumors lacking APC mutations, mutations were found in the CTNNB1 (ß-catenin) gene. These mutations occur in or around the third exon, which encodes the putative phosphorylation sites for GSK-3ß and remove the target residues of this kinase (Morin et al., 1997; Polakis et al., 1999). Mutations in ß-catenin are not only found in intestinal tumors. The CTNNB1 gene has been shown to be extensively mutated in various other tumors. As the exception that perhaps confirms the rule, mutations in Axin/Conduction have rarely been found in colon tumors and other types of malignancies (Webster et al., 2000).

Taken together, mutations in APC, axin, and ß-catenin share a common denominator, in that they all promote formation of ß- catenin/TCF complexes. This implies that inappropriate activation of TCF target genes represents the primary event in cellular transformation of epithelial cells (reviewed in Polakis, 2000; Fodde and Smits, 2001).

PERSPECTIVES

Despite the impressive recent advances that have substantially improved our understanding of Wnt signaling, many gaps remain to be filled before we can hope to get a glimpse of the complete picture. Open questions include:

a) What determines the specificity of interactions between Wnts and Frizzled receptors?
b) How do Frizzled receptors activate downstream signaling components?
c) What is the function of Dsh?
d) How do Wnts affect cell polarity and cell adhesion?
e) What is the role of Wnt signaling in cancer?

Given the importance of the signaling pathway and the multitude of experimental systems being used to study Wnt signaling, answers to remaining questions can be expected to come from many different directions at an accelerated pace.

ACKNOWLEDGEMENTS

Our research is supported by the Swiss National Science Foundation and the Swiss Cancer League.

REFERENCES

AYBAR MJ, GLAVIC A, MAYOR R (2002) Extracellular signals, cell interactions and transcription factors involved in the induction of the neural crest cells. Biol Res 35: 267-275

BAYLIES MK, MARTINEZ ARIAS A, BATE M (1995) Wingless is required for the formation of a subset of muscle founder cells during Drosophila embryogenesis. Development 121: 3829-3837

BEHRENS J, VON KRIES JP, KUHL M, BRUHN L, WEDLICH D, GROSSCHEDL R, BIRCHMEIER W (1996) Functional interaction of beta-catenin with the transcription factor LEF-1. Nature 382: 638-642

BEHRENS J, JERCHOW BA, WURTELE M, GRIMM J, ASBRAND C, WIRTZ R, KUHL M, WEDLICH D, BIRCHMEIER W (1998) Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science 280: 596-599

BIANCHI ME, BELTRAME M (1998) Flexing DNA: HMG-box proteins and their partners. Am J Hum Genet 63: 1573-1577

BRANNON M, GOMPERTS M, SUMOY L, MOON RT, KIMELMAN D (1997) A beta-catenin/XTcf-3 complex binds to the siamois promoter to regulate dorsal axis specification in Xenopus. Genes Dev 11: 2359-2370

BRUHN L, MUNNERLYN A, GROSSCHEDL R (1997) ALY, a context-dependent coactivator of LEF-1 and AML-1, is required for TCRalpha enhancer function. Genes Dev 11: 640-653

BRUNNER E, PETER O, SCHWEIZER L, BASLER K (1997) Pangolin encodes a Lef-1 homologue that acts downstream of Armadillo to transduce the Wingless signal in Drosophila. Nature 385: 829-833

CABRERA CV, ALONSO MC, JOHNSTON P, PHILLIPS RG, LAWRENCE PA (1987) Phenocopies induced with antisense RNA identify the wingless gene. Cell 50: 659-663

CADIGAN KM, NUSSE R (1996) Wingless signaling in the Drosophila eye and embryonic epidermis. Development 122: 2801-2812

CADIGAN KM, NUSSE R (1997) Wnt signaling: a common theme in animal development. Genes Dev 11: 3286-3305

CARLSSON P, WATERMAN ML, JONES KA (1993) The hLEF/TCF-1 alpha HMG protein contains a context-dependent transcriptional activation domain that induces the TCR alpha enhancer in T cells. Genes Dev 7: 2418-2430

COLLINS RT, TREISMAN JE (2000) Osa-containing Brahma chromatin remodeling complexes are required for the repression of wingless target genes. Genes Dev 14: 3140-3152

COTSARELIS G, KAUR P, DHOUAILLY D, HENGGE U, BICKENBACH J (1999) Epithelial stem cells in the skin: definition, markers, localization and functions. Exp Dermatol 8: 80-88

COUSO JP, BISHOP SA, MARTINEZ ARIAS A (1994) The wingless signaling pathway and the patterning of the wing margin in Drosophila. Development 120: 621-636

CRAWFORD HC, FINGLETON BM, RUDOLPH-OWEN LA, GOSS KJ, RUBINFELD B, POLAKIS P, MATRISIAN LM (1999) The metalloproteinase matrilysin is a target of beta-catenin transactivation in intestinal tumors. Oncogene 18: 2883-2891

DASGUPTA R, FUCHS E (1999) Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development 126: 4557-4568

DOOIJES D, VAN DE WETERING M, KNIPPELS L, CLEVERS H (1993) The Schizosaccharomyces pombe mating-type gene mat-Mc encodes a sequence-specific DNA-binding high mobility group box protein. J Biol Chem 268: 24813-24817

DUVAL A, ROLLAND S, TUBACHER E, BUI H, THOMAS G, HAMELIN R (2000) The human T-cell transcription factor-4 gene: structure, extensive characterization of alternative splicings, and mutational analysis in colorectal cancer cell lines. Cancer Res 60: 3872-3879

FODDE R, SMITS R (2001) Disease model: familial adenomatous polyposis. Trends Mol Med 7: 369-373

FUCHS E, SEGRE JA (2000) Stem cells: a new lease on life. Cell 100: 143-155

GIESE K, COX J, GROSSCHEDL R (1992) The HMG domain of lymphoid enhancer factor 1 bends DNA and facilitates assembly of functional nucleoprotein structures. Cell 69: 185-195

GIESE K, KINGSLEY C, KIRSHNER JR, GROSSCHEDL R (1995) Assembly and function of a TCR alpha enhancer complex is dependent on LEF-1-induced DNA bending and multiple protein-protein interactions. Genes Dev 9: 995-1008

GLINKA A, WU W, DELIUS H, MONAGHAN AP, BLUMENSTOCK C, NIEHRS C (1998) Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 391: 357-362

GROSSCHEDL R, GIESE K, PAGEL J (1994) HMG domain proteins: architectural elements in the assembly of nucleoprotein structures. Trends Genet 10: 94-100

HE TC, SPARKS AB, RAGO C, HERMEKING H, ZAWEL L, DA COSTA LT, MORIN PJ, VOGELSTEIN B, KINZLER KW (1998) Identification of c-MYC as a target of the APC pathway. Science 281: 1509-1512

HEASMAN J, CRAWFORD A, GOLDSTONE K, GARNER-HAMRICK P, GUMBINER B, MCCREA P, KINTNER C, NORO CY, WYLIE C (1994) Overexpression of cadherins and underexpression of beta-catenin inhibit dorsal mesoderm induction in early Xenopus embryos. Cell 79: 791-803

HEISENBERG CP, TADA M, RAUCH GJ, SAUDE L, CONCHA ML, GEISLER R, STEMPLE DL, SMITH JC, WILSON SW (2000) Silberblick/Wnt11 mediates convergent extension movements during zebrafish gastrulation. Nature 405: 76-81

HOBMAYER B, RENTZSCH F, KUHN K, HAPPEL CM, VON LAUE CC, SNYDER P, ROTHBACHER U, HOLSTEIN TW (2000) WNT signalling molecules act in axis formation in the diploblastic metazoan Hydra. Nature 407: 186-189

HOVANES K, LI TW, MUNGUIA JE, TRUONG T, MILOVANOVIC T, LAWRENCE MARSH J, HOLCOMBE RF, WATERMAN ML (2001) Beta-catenin-sensitive isoforms of lymphoid enhancer factor-1 are selectively expressed in colon cancer. Nat Genet 28: 53-57

HUELSKEN J, VOGEL R, ERDMANN B, COTSARELIS G, BIRCHMEIER W (2001) Beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell 105: 533-545

JENSEN UB, LOWELL S, WATT FM (1999) The spatial relationship between stem cells and their progeny in the basal layer of human epidermis: a new view based on whole-mount labelling and lineage analysis. Development 126: 2409-2418

KORINEK V, BARKER N, MORIN PJ, VAN WICHEN D, DE WEGER R, KINZLER KW, VOGELSTEIN B, CLEVERS H (1997) Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science 275: 1784-1787

KORINEK V, BARKER N, WILLERT K, MOLENAAR M, ROOSE J, WAGENAAR G, MARKMAN M, LAMERS W, DESTREE O, CLEVERS H (1998) Two members of the Tcf family implicated in Wnt/beta-catenin signaling during embryogenesis in the mouse. Mol Cell Biol 18: 1248-1256

KRAMPS T, PETER O, BRUNNER E, NELLEN D, FROESCH B, CHATTERJEE S, MURONE M, ZULLIG S, BASLER K (2002) Wnt/wingless signaling requires BCL9/legless-mediated recruitment of pygopus to the nuclear beta-catenin-TCF complex. Cell 109: 47-60

KUHL M, SHELDAHL LC, PARK M, MILLER JR, MOON RT (2000) The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet 16: 279-283

LARABELL CA, TORRES M, ROWNING BA, YOST C, MILLER JR, WU M, KIMELMAN D, MOON RT (1997) Establishment of the dorso-ventral axis in Xenopus embryos is presaged by early asymmetries in beta-catenin that are modulated by the Wnt signaling pathway. J Cell Biol 136: 1123-1136

LAUDET V, STEHELIN D, CLEVERS H (1993) Ancestry and diversity of the HMG box superfamily. Nucleic Acids Res 21: 2493-2501

LIN R, THOMPSON S, PRIESS JR (1995) Pop-1 encodes an HMG box protein required for the specification of a mesoderm precursor in early C. elegans embryos. Cell 83: 599-609

LU B, ROEGIERS F, JAN LY, JAN YN (2001) Adherens junctions inhibit asymmetric division in the Drosophila epithelium. Nature 409: 522-525

MANN B, GELOS M, SIEDOW A, HANSKI ML, GRATCHEV A, ILYAS M, BODMER WF, MOYER MP, RIECKEN EO, BUHR HJ, HANSKI C (1999) Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci U S A 96: 1603-1608

MAO B, WU W, DAVIDSON G, MARHOLD J, LI M, MECHLER BM, DELIUS H, HOPPE D, STANNEK P, WALTER C, GLINKA A, NIEHRS C (2002) Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin signalling. Nature 417: 664-667

MAO J, WANG J, LIU B, PAN W, FARR GH, 3RD, FLYNN C, YUAN H, TAKADA S, KIMELMAN D, LI L, WU D (2001) Low-density lipoprotein receptor-related protein-5 binds to Axin and regulates the canonical Wnt signaling pathway. Mol Cell 7: 801-809

MCMAHON AP, BRADLEY A (1990) The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell 62: 1073-1085

MERRILL BJ, GAT U, DASGUPTA R, FUCHS E (2001) Tcf3 and Lef1 regulate lineage differentiation of multipotent stem cells in skin. Genes Dev 15: 1688-1705

MLODZIK M (2000) Spiny legs and prickled bodies: new insights and complexities in planar polarity establishment. Bioessays 22: 311-315

MOLENAAR M, VAN DE WETERING M, OOSTERWEGEL M, PETERSON-MADURO J, GODSAVE S, KORINEK V, ROOSE J, DESTREE O, CLEVERS H (1996) XTcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell 86: 391-399

MOON RT, KIMELMAN D (1998) From cortical rotation to organizer gene expression: toward a molecular explanation of axis specification in Xenopus. Bioessays 20: 536-545

MORIN PJ, SPARKS AB, KORINEK V, BARKER N, CLEVERS H, VOGELSTEIN B, KINZLER KW (1997) Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275: 1787-1790

NOORDERMEER J, JOHNSTON P, RIJSEWIJK F, NUSSE R, LAWRENCE PA (1992) The consequences of ubiquitous expression of the wingless gene in the Drosophila embryo. Development 116: 711-719

NUSSE R, VARMUS HE (1982) Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 31: 99-109

NUSSE R, VARMUS HE (1992) Wnt genes. Cell 69: 1073-1087

OOSTERWEGEL M, VAN DE WETERING M, DOOIJES D, KLOMP L, WINOTO A, GEORGOPOULOS K, MEIJLINK F, CLEVERS H (1991) Cloning of murine TCF-1, a T cell-specific transcription factor interacting with functional motifs in the CD3-epsilon and T cell receptor alpha enhancers. J Exp Med 173: 1133-1142

PARR BA, MCMAHON AP (1994) Wnt genes and vertebrate development. Curr Opin Genet Dev 4: 523-528

PETERS JM, MCKAY RM, MCKAY JP, GRAFF JM (1999) Casein kinase I transduces Wnt signals. Nature 401: 345-350

POLAKIS P (2000) Wnt signaling and cancer. Genes Dev 14: 1837-1851

POLAKIS P (2001) More than one way to skin a catenin. Cell 105: 563-566

POLAKIS P, HART M, RUBINFELD B (1999) Defects in the regulation of beta-catenin in colorectal cancer. Adv Exp Med Biol 470: 23-32

RIESE J, YU X, MUNNERLYN A, ERESH S, HSU SC, GROSSCHEDL R, BIENZ M (1997) LEF-1, a nuclear factor coordinating signaling inputs from wingless and decapentaplegic. Cell 88: 777-787

RIJSEWIJK F, SCHUERMANN M, WAGENAAR E, PARREN P, WEIGEL D, NUSSE R (1987) The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell 50: 649-657

ROUSSET R, MACK JA, WHARTON KA, JR., AXELROD JD, CADIGAN KM, FISH MP, NUSSE R, SCOTT MP (2001) Naked cuticle targets dishevelled to antagonize Wnt signal transduction. Genes Dev 15: 658-671

SAKANAKA C, LEONG P, XU L, HARRISON SD, WILLIAMS LT (1999) Casein kinase epsilon in the wnt pathway: regulation of beta-catenin function. Proc Natl Acad Sci U S A 96: 12548-12552

SAMPEDRO J, JOHNSTON P, LAWRENCE PA (1993) A role for wingless in the segmental gradient of Drosophila? Development 117: 677-687

SAWA H, KOUIKE H, OKANO H (2000) Components of the SWI/SNF complex are required for asymmetric cell division in C. elegans. Mol Cell 6: 617-624

SCHNEIDER S, STEINBEISSER H, WARGA RM, HAUSEN P (1996) Beta-catenin translocation into nuclei demarcates the dorsalizing centers in frog and fish embryos. Mech Dev 57: 191-198

SHARPE C, LAWRENCE N, MARTINEZ ARIAS A (2001) Wnt signalling: a theme with nuclear variations. Bioessays 23: 311-318

SOLNICA-KREZEL L (1999) Pattern formation in zebrafish—fruitful liaisons between embryology and genetics. Curr Top Dev Biol 41: 1-35

TAMAI K, SEMENOV M, KATO Y, SPOKONY R, LIU C, KATSUYAMA Y, HESS F, SAINT-JEANNET JP, HE X (2000) LDL-receptor-related proteins in Wnt signal transduction. Nature 407: 530-535

TAYLOR G, LEHRER MS, JENSEN PJ, SUN TT, LAVKER RM (2000) Involvement of follicular stem cells in forming not only the follicle but also the epidermis. Cell 102: 451-461

TETSU O, MCCORMICK F (1999) Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398: 422-426

THOMAS KR, CAPECCHI MR (1990) Targeted disruption of the murine int-1 proto-oncogene resulting in severe abnormalities in midbrain and cerebellar development. Nature 346: 847-850

TRAVIS A, AMSTERDAM A, BELANGER C, GROSSCHEDL R (1991) LEF-1, a gene encoding a lymphoid-specific protein with an HMG domain, regulates T-cell receptor alpha enhancer function [corrected]. Genes Dev 5: 880-894

UMBHAUER M, DJIANE A, GOISSET C, PENZO-MENDEZ A, RIOU JF, BOUCAUT JC, SHI DL (2000) The C-terminal cytoplasmic Lys-Thr-X-X-X-Trp motif in frizzled receptors mediates Wnt/beta-catenin signalling. EMBO J 19: 4944-4954

VAN DE WETERING M, OOSTERWEGEL M, DOOIJES D, CLEVERS H (1991) Identification and cloning of TCF-1, a T lymphocyte-specific transcription factor containing a sequence-specific HMG box. EMBO J 10: 123-132

VAN DE WETERING M, CASTROP J, KORINEK V, CLEVERS H (1996) Extensive alternative splicing and dual promoter usage generate Tcf-1 protein isoforms with differential transcription control properties. Mol Cell Biol 16: 745-752

VAN DE WETERING M, CAVALLO R, DOOIJES D, VAN BEEST M, VAN ES J, LOUREIRO J, YPMA A, HURSH D, JONES T, BEJSOVEC A, PEIFER M, MORTIN M, CLEVERS H (1997) Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF. Cell 88: 789-799

VAN GENDEREN C, OKAMURA RM, FARINAS I, QUO RG, PARSLOW TG, BRUHN L, GROSSCHEDL R (1994) Development of several organs that require inductive epithelial-mesenchymal interactions is impaired in LEF-1-deficient mice. Genes Dev 8: 2691-2703

WALLINGFORD JB, ROWNING BA, VOGELI KM, ROTHBACHER U, FRASER SE, HARLAND RM (2000) Dishevelled controls cell polarity during Xenopus gastrulation. Nature 405: 81-85

WALLINGFORD JB, EWALD AJ, HARLAND RM, FRASER SE (2001) Calcium signaling during convergent extension in Xenopus. Curr Biol 11: 652-661

WATERMAN ML, FISCHER WH, JONES KA (1991) A thymus-specific member of the HMG protein family regulates the human T cell receptor C alpha enhancer. Genes Dev 5: 656-669

WEBSTER MT, ROZYCKA M, SARA E, DAVIS E, SMALLEY M, YOUNG N, DALE TC, WOOSTER R (2000) Sequence variants of the axin gene in breast, colon, and other cancers: an analysis of mutations that interfere with GSK3 binding. Genes Chromosomes Cancer 28: 443-453

WINTER CG, WANG B, BALLEW A, ROYOU A, KARESS R, AXELROD JD, LUO L (2001) Drosophila Rho-associated kinase (Drok) links Frizzled-mediated planar cell polarity signaling to the actin cytoskeleton. Cell 105: 81-91

WODARZ A, NUSSE R (1998) Mechanisms of Wnt signaling in development. Annu Rev Cell Dev Biol 14: 59-88

YAN D, WALLINGFORD JB, SUN TQ, NELSON AM, SAKANAKA C, REINHARD C, HARLAND RM, FANTL WJ, WILLIAMS LT (2001) Cell autonomous regulation of multiple Dishevelled-dependent pathways by mammalian Nkd. Proc Natl Acad Sci USA 98: 3802-3807

ZENG W, WHARTON KA, JR., MACK JA, WANG K, GADBAW M, SUYAMA K, KLEIN PS, SCOTT MP (2000) Naked cuticle encodes an inducible antagonist of Wnt signalling. Nature 403: 789-795

Corresponding author: Ernst Reichmann. tel.: +41 1 266 7493 (7111). fax: +41 1 266 7170. e-mail: Ernst.Reichmann@kispi.unizh.ch

Received: July 02, 2002. In revised form: July 09, 2002. Accepted: July 16, 2002

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License