Wnts are secreted proteins that are essential for a wide array of developmental and physiological processes. They signal across the plasma membrane by interacting with serpentine receptors of the Frizzled (Fz) family and members of the low-density-lipoprotein-related protein (LRP) family. Activation of Fz-LRP promotes the stability and nuclear localization of β-catenin by compromising the ability of a multiprotein complex containing axin, adenomatosis polyposis coli (APC) and glycogen synthase kinase 3 (GSK3) to target it for degradation and block its nuclear import. The Fz-LRP receptor complex probably accomplishes this by generating multiple signals in the cytoplasm. These involve activation of Dishevelled (Dsh), possibly through trimeric G proteins and LRP-mediated axin binding and/or degradation. However, individual Wnts and Fzs can activate both β-catenin-dependent and -independent pathways, and Fz co-receptors such as LRP probably provide some of this specificity. Additional, conflicting data concern the role of the atypical receptor tyrosine kinase Ryk, which might mediate Wnt signaling independently of Fz and/or function as a Fz co-receptor in some cells.

Wnts are secreted lipid-modified glycoproteins (Willert et al., 2003) found in all metozoans examined to date. During development, they are used by cells to influence the fate or behavior of neighboring cells (Cadigan and Nusse, 1997; Wodarz and Nusse, 1998; Logan and Nusse, 2004). In the mature organism, Wnts are implicated in maintaining stem-cell-like fates in the intestinal epithelium (Pinto and Clevers, 2005), skin (Lowry et al., 2005) and hemopoietic cells (Reya et al., 2003; Willert et al., 2003). Inappropriate activation of Wnt signaling contributes to numerous human cancers (Polakis, 2000; van Es et al., 2003; Gregorieff and Clevers, 2005) and reduced Wnt signaling has been implicated in osteoporosis (Koay and Brown, 2005; Levasseur et al., 2005).

A combination of genetic and molecular studies has provided a biochemical model for how Wnt molecules act. They are thought to signal through several different pathways. In this Commentary, we focus mainly on signaling through β-catenin, sometimes referred to as canonical Wnt signaling. During this branch of signaling, Wnt induces stabilization and nuclear localization of β-catenin, an intracellular signal normally held in check by a complex containing axin, adenomatosis polyposis coli (APC) and glycogen synthase kinase 3 (GSK3) that normally targets it for degradation (Fig. 1A,B). Reviews containing more details about the other branches of Wnt signaling can be found elsewhere (Veeman et al., 2003; Fanto and McNeill, 2004; Kohn and Moon, 2005).

Wnt/β-catenin signaling involves at least two types of receptors: Frizzled (Fz) and low-density-lipoprotein-related protein (LRP). Below, we discuss current models for how a Fz-LRP receptor complex might signal, emphasizing the possibility that it generates more than one signal. We examine these models in relation to the issue of signaling specificity of β-catenin-dependent and -independent pathways. Finally, we explore work implicating the kinase Ryk as a Wnt receptor and/or Fz co-receptor.

The initial connection between seven-transmembrane-span proteins of the Fz family and Wnt proteins came from studies in Drosophila cell culture. Transfection of Drosophila frizzled 2 (fz2) confers the ability to bind Wingless (Wg; a fly Wnt) and stabilize Armadillo (Arm; the fly β-catenin) upon cells that do not express Fz2 and are unresponsive to Wg (Bhanot et al., 1996). Additional evidence implicating Fzs in Wnt signaling came from mis-expression studies in Xenopus (Yang-Snyder et al., 1996; He et al., 1997) and the finding that a mutation in lin-17, which encodes a Fz, affects T-cell polarity in Caenorhabditis elegans (Sawa et al., 1996), which is controlled by LIN-44, a Wnt (Herman et al., 1995).

The extracellular portion of Fz proteins contains a cysteine-rich domain (CRD), which is thought to mediate direct binding to Wnt proteins. The CRDs of Fz1 and Fz2 in flies, and Fz8 in mice, have been shown to bind to Wnt proteins with nanomolar affinity (Hsieh et al., 1999b; Rulifson et al., 2000; Wu and Nusse, 2002). Expression of truncated forms of Fz1 and Fz2 containing the extracellular domains tethered to the membrane inhibits Wg signaling, which is consistent with direct binding of Wnts to the CRD (Cadigan et al., 1998; Zhang and Carthew, 1998).

Despite the abundant functional and physical data indicating Fz proteins are essential for Wnt signaling, this remains largely unproven for most family members, presumably owing to genetic redundancy (see Logan and Nusse, 2004). A notable exception is in flies, where loss of fz1 and fz2 results in a phenotype that is virtually indistinguishable from wg mutants (Bhat, 1998; Bhanot et al., 1999; Chen and Struhl, 1999; Muller et al., 1999). Both fz genes must be mutated to obtain a loss of Wg signaling. The relatively simple correlation of the wg and fz1,fz2 phenotypes makes it an ideal system for structure-function studies of these receptors.

Fig. 1.

Outline of Wnt/β-catenin signaling. (A) In the absence of Wnt, β-catenin (β-cat) is phosphorylated by a complex containing GSK3. This targets β-catenin for proteosomal degradation. In the nucleus, members of the T-cell factor (TCF) family of DNA-binding proteins repress Wnt targets, in concert with co-repressors such as Groucho (Gro). (B) Upon Wnt binding to Fz-LRP receptors, a combination of LRP-axin interaction and Dvl phosphorylation (P) blocks the APC-axin-GSK3 complex from phosphorylating β-catenin. The accumulated β-catenin then enters the nucleus, where it converts TCF into a transcriptional activator. See the Wnt homepage (http://www.stanford.edu/~rnusse/wntwindow.html) for a more complete description of Wnt signaling components.

Fig. 1.

Outline of Wnt/β-catenin signaling. (A) In the absence of Wnt, β-catenin (β-cat) is phosphorylated by a complex containing GSK3. This targets β-catenin for proteosomal degradation. In the nucleus, members of the T-cell factor (TCF) family of DNA-binding proteins repress Wnt targets, in concert with co-repressors such as Groucho (Gro). (B) Upon Wnt binding to Fz-LRP receptors, a combination of LRP-axin interaction and Dvl phosphorylation (P) blocks the APC-axin-GSK3 complex from phosphorylating β-catenin. The accumulated β-catenin then enters the nucleus, where it converts TCF into a transcriptional activator. See the Wnt homepage (http://www.stanford.edu/~rnusse/wntwindow.html) for a more complete description of Wnt signaling components.

There is also a compelling case for some LRPs transducing Wnt signals. LRPs are single-pass transmembrane proteins that have relatively large extracellular and short cytosolic domains [see He et al. (He et al., 2004), for more details concerning LRP structure]. Mutations in the fly LRP, Arrow (Arr), result in a mutant phenotype very similar to that of wg (Wehrli et al., 2000). Disruption of LRP6 in mice causes an embryonic phenotype that appears to be a composite of Wnt1, Wnt3a and Wnt7a mutants (Pinson et al., 2000). Mouse embryos lacking both LRP6 and LRP5 fail to form a primitive streak and lack mesoderm (Kelly et al., 2004), which is also observed in Wnt3- and β-catenin-knockouts (Liu et al., 1999; Huelsken et al., 2000). Complementing these loss-of-function data are studies demonstrating that overexpression of LRP5 or LRP6 lacking the intracellular domain can block Wnt signaling (Tamai et al., 2000). Truncations of either LRP lacking the extracellular domains but still tethered to the membrane can constitutively activate Wnt/β-catenin signaling in Xenopus and mammalian cell culture (Tamai et al., 2000; Mao et al., 2001b; Liu et al., 2003; Cong et al., 2004b; Gonzalez-Sancho et al., 2004; Tamai et al., 2004). These data all point towards an essential, positive role for LRP5, LRP6 and Arr in Wnt signaling.

The extracellular domains of Fz and LRPs can bind to Wnts in coprecipitation assays (Tamai et al., 2000; Kato et al., 2002; Itasaki et al., 2003; Liu et al., 2003; Cong et al., 2004b). However, the Wnt-LRP interactions appear to be weaker than the nanomolar binding observed with Wnt-Fz (He et al., 2004). Although no interaction between a native LRP and Fz has been reported, the soluble extracellular domains of LRP6 can form a complex with the murine Fz8/CRD in a Wnt1-dependent manner (Tamai et al., 2000; Semenov et al., 2001). Similar results have not been obtained for Arr, Fz2 and Wg in flies (Wu and Nusse, 2002). Nevertheless, the vertebrate results have led to the hypothesis that Wnt ligand induces association of Fz and LRP at the cell surface.

Direct evidence for Wnt stimulating the Fz-LRP association is lacking; thus, neither Wnt-dependent co-immunoprecipitation of native proteins nor fluorescence resonance energy transfer (FRET) studies with tagged Fz and LRP have been reported. However, two recent studies report that forced association of Fz and LRP6 can stimulate Wnt/β-catenin signaling independently of Wnt. One approach used LRP6 fused with the neurotrophic receptor TrkN and human Fz5 tagged with NT3, a ligand of TrkN. Co-expression of these fusion proteins in cultured Drosophila cells resulted in >20-fold activation of a Wnt reporter gene (Cong et al., 2004b). Impressive synergy is also observed when human HEK-293T cells are co-transfected with LRP6 and a fusion of Fz5 and DKK1 (Holmen et al., 2005). DKK1 is a Wnt antagonist that is a ligand for LRP6 (Mao et al., 2001a; Semenov et al., 2001); presumably, the DKK1 moiety on Fz5 recruits LRP6 to activate Wnt/β-catenin signaling in the absence of exogenously added Wnt.

Clouding the picture painted above is the finding that the CRD, thought to be necessary and sufficient for Wnt binding (Bhanot et al., 1996; Hsieh et al., 1999b; Rulifson et al., 2000; Wu and Nusse, 2002), is dispensable for Fz function. Transgenes that produce Fz1 or Fz2 lacking the CRD are able to completely rescue fly embryos or larval tissues lacking endogenous fz1, fz2 and fz3 (Chen et al., 2004). However, another study found that similar constructs signal less efficiently than wild-type proteins (Povelones and Nusse, 2005). Fusion of the human Wnt inhibitory factor (WIF) domain, which binds to Wnts with high affinity (Hsieh et al., 1999a), to Fz lacking the CRD restores full Wnt/β-catenin signaling activity. A protein in which Wnt is fused to Fz lacking the CRD (Wnt-FzΔCRD) also had full activity (Povelones and Nusse, 2005). These data support a model in which the CRD recruits Wnt to Fz, where it can then interact with other portions of Fz or other co-receptors (see also Bejsovec, 2005).

The finding that the CRD of Fz is dispensable for Wnt/β-catenin signaling is not incompatible with the model that Wnt stimulates assembly of a Fz-LRP oligomer but argues against the idea that Wnt acts as an adaptor between the two co-receptors. It is possible that binding of Wnt to LRP induces a conformational change that makes binding to Fz favorable. Constitutively active Wnt-FzΔCRD fusions could bind preferentially to LRP. Indeed, the Wg-FzΔCRD chimera requires Arr for its signaling activity (Povelones and Nusse, 2005). Clearly, more work examining the effects of Wnt binding on native Fz and LRP in the plasma membrane is needed.

Activation of the Wnt/β-catenin pathway results in the stabilization and nuclear translocation of β-catenin/Arm. Wnt stimulation inhibits the ability of the axin-APC-GSK3 complex to phosphorylate β-catenin, which targets the protein for ubiquitylation and proteosomal degradation (Kitagawa et al., 1999; Farr et al., 2000; Hinoi et al., 2000; Salic et al., 2000). The PDZ-containing protein Dishevelled (Dsh in flies; Dvl1/Dvl2/Dvl3 in mammals) is required for Wnt-dependent inhibition of this complex (Yanagawa et al., 1995; Li et al., 1999; Rothbacher et al., 2000; Salic et al., 2000; Kishida et al., 2001). There is also evidence that, in Drosophila, axin can retain Arm in the cytoplasm, and that this form of regulation is distinct from regulation of Arm turnover (Tolwinski and Wieschaus, 2001; Tolwinski et al., 2003; Tolwinski and Wieschaus, 2004). Recent work, summarized below, suggests that activation of Fz-LRP causes two distinct events: binding of axin to the intracellular domain of LRP and Fz-induced phosphorylation of Dsh/Dvl. These branches in the signaling cascade then converge on β-catenin stability and nuclear localization (Fig. 2).

The LRP/axin branch

Expression of constructs comprising the intracellular domain of LRP5 or LRP6 tethered to the membrane can constitutively stabilize β-catenin and activate Wnt reporter genes (Mao et al., 2001b; Li et al., 2002; Brennan et al., 2004; Cong et al., 2004b; Tamai et al., 2004). This appears to be independent of Fz, since it is observed in S2 cells in which all four Drosophila fzs are expressed at extremely low levels, i.e. <1 transcript/cell (Brennan et al., 2004). A similar truncated form of Arr does not activate Wg signaling in flies (Tolwinski et al., 2003). However, a chimera containing the intracellular domain of Arr fused to the C-terminus of Fz2 activates Wg targets independently of endogenous wg, fz1/fz2 or arr (Tolwinski et al., 2003). Perhaps mis-expression of the truncated Arr from a single-copy transgene is not sufficient to bypass the other (Fz-dependent) branch of Wnt signaling. Fusion of this domain to a Fz presumably activates both branches of the pathway, leading to Arm stabilization and nuclear translocation.

Fig. 2.

The two-signal model of Fz-LRP signaling to β-catenin showing the regulatory relationships between each component (A) and summarizing the probable physical interactions (B). In this model, Wnt stimulation promotes Fz-LRP oligomerization, which transduces two separate signals to the cytoplasm. The first signal is LRP phosphorylation, mediated by membrane-localized GSK3 and LRP-bound CKIγ. We speculate that neither kinase is activated by Wnt. Rather, the LPR-Wnt-Fz interaction allows LRP to become phosphorylatable, which then promotes the recruitment of axin to the plasma membrane, leading to its inactivation and/or degradation. The second signal is Fz-dependent Dsh/Dvl phosphorylation, which we tentatively propose involves trimeric G proteins. Activated Dvl then inhibits the APC-axin-GSK3 complex by a poorly understood mechanism. The dashed line in panel A reflects the fact that Dsh/Dvl also participates in recruitment of axin to the plasma membrane. Although hyperactivation of either branch by overexpression can stabilize β-catenin, both signals are required under physiological conditions.

Fig. 2.

The two-signal model of Fz-LRP signaling to β-catenin showing the regulatory relationships between each component (A) and summarizing the probable physical interactions (B). In this model, Wnt stimulation promotes Fz-LRP oligomerization, which transduces two separate signals to the cytoplasm. The first signal is LRP phosphorylation, mediated by membrane-localized GSK3 and LRP-bound CKIγ. We speculate that neither kinase is activated by Wnt. Rather, the LPR-Wnt-Fz interaction allows LRP to become phosphorylatable, which then promotes the recruitment of axin to the plasma membrane, leading to its inactivation and/or degradation. The second signal is Fz-dependent Dsh/Dvl phosphorylation, which we tentatively propose involves trimeric G proteins. Activated Dvl then inhibits the APC-axin-GSK3 complex by a poorly understood mechanism. The dashed line in panel A reflects the fact that Dsh/Dvl also participates in recruitment of axin to the plasma membrane. Although hyperactivation of either branch by overexpression can stabilize β-catenin, both signals are required under physiological conditions.

The ability of intracellular LRP to activate β-catenin strongly correlates with its ability to bind axin (Mao et al., 2001b; Cong et al., 2004b; Tamai et al., 2004). This depends upon the phosphorylation of several motifs containing Ser/Thr residues in the cytoplasmic domain of LRP6, which act redundantly to activate Wnt/β-catenin signaling (Tamai et al., 2004; Davidson et al., 2005). Each motif contains two clusters containing casein kinase I (CKI) consensus sites and a PPPSP site (Tamai et al., 2004; Davidson et al., 2005; Zeng et al., 2005). CKIγ has been shown to phosphorylate the CKI sites and is required for Wnt/β-catenin signaling (Davidson et al., 2005). The PPPSP sites are phosphorylated by GSK3, and this is required for the adjacent phosphorylation of the CKI sites (Zeng et al., 2005). Importantly, Wnt stimulation promotes rapid (10-15 minutes) phosphorylation at both GSK3 and CKI sites (Tamai et al., 2004; Davidson et al., 2005; Zeng et al., 2005) and FRET has shown Wnt-dependent binding of axin to LRP5 with similar kinetics (Mao et al., 2001b). These data support a model in which Wnt stimulation promotes phosphorylation of LRP by GSK3 and CKIγ, which recruits axin to the plasma membrane, where it is inactivated and/or targeted for degradation. Wnt stimulation can promote axin degradation in mammalian cell culture (Willert et al., 1999; Yamamoto et al., 1999; Mao et al., 2001b) and fly embryos (Tolwinski et al., 2003). This should promote β-catenin/Arm stability because axin is an essential component of the degradation complex (Lee et al., 2003). It should also promote β-catenin/Arm nuclear localization, owing to loss of cytoplasmic tethering (Tolwinski and Wieschaus, 2001; Tolwinski and Wieschaus, 2004).

Several research groups have demonstrated that truncated LRP5/LRP6 activates β-catenin/Arm independently of Dsh/Dvl (Li et al., 2002; Schweizer and Varmus, 2003; Cong et al., 2004b; Liu, G. et al., 2005,Liu, G. et al., 2005). This suggests that it can recruit axin to the plasma membrane independently of Dsh/Dvl. Although this might be true under some conditions, axin-GFP is recruited to the plasma membrane in a dsh-dependent manner in the fly embryo (Cliffe et al., 2003). In addition, overexpression of Dsh can recruit axin to the plasma membrane (Cliffe et al., 2003) and downregulate its levels (N. Tolwinski, personal communication). These results can be reconciled if one proposes that, under physiological conditions (i.e. Wnt stimulation of receptors), both LRP and Dsh/Dvl participate in recruitment of axin to the membrane and its degradation (Fig. 2).

There are several important gaps in our understanding of the regulation of axin by LRPs. The mechanism of axin degradation is not understood and it is also not clear whether axin degradation is a secondary consequence of its inhibition (see He et al., 2004).

The Fz/Dsh branch

Dsh/Dvl is essential for Wnt signaling in flies (Klingensmith et al., 1994; Noordermeer et al., 1994; Theisen et al., 1994) and mammalian cells (Li et al., 2002; Liu, G. et al., 2005,Liu, G. et al., 2005). Wg can promote membrane association of Dsh (Yanagawa et al., 1995; Cliffe et al., 2003), and expression of several Fz proteins causes Dsh/Dvl to be recruited to the plasma membrane (Axelrod et al., 1998; Boutros et al., 2000; Rothbacher et al., 2000; Umbhauer et al., 2000). Direct binding of the C-terminus of murine Fz7 to Dvl1 has been reported (Wong et al., 2003) and point mutations in the cytoplasmic loops or the C-terminus of human Fz5 and Xenopus Fz3 abolish binding to Dsh/Dvl (Umbhauer et al., 2000; Cong et al., 2004b). These Fz mutants cannot activate β-catenin, even though they reside on the cell surface, which suggests they fold properly and are trafficked correctly (Umbhauer et al., 2000; Cong et al., 2004b).

Fz can also promote Dsh/Dvl phosphorylation (Willert et al., 1997; Rothbacher et al., 2000; Umbhauer et al., 2000; Takada et al., 2005). This is thought to be important for Wnt/β-catenin signaling since Wnt causes hyperphosphorylation of Dsh/Dvl in vivo (Yanagawa et al., 1995; Gonzalez-Sancho et al., 2004). Although the phosphoacceptor sites on Dsh/Dvl required for activation have not been identified (these proteins are notoriously Ser/Thr rich), the strong correlation between Dsh/Dvl phosphorylation and its ability to activate β-catenin/Arm (Yanagawa et al., 1995; Capelluto et al., 2002) suggests that this is an important part of Wnt/β-catenin signal transduction.

Does Dsh/Dvl phosphorylation form the second branch of Fz-LRP signaling? A recent study found that dominant-negative LRP6 or Arr does not block Wnt-induced Dvl phosphorylation, despite blocking β-catenin stabilization (Gonzalez-Sancho et al., 2004). Likewise, the LRP antagonist Dkk1 blocks β-catenin stabilization without affecting Dvl phosphorylation. In addition, LRP or Arr lacking the extracellular domain activates β-catenin/Arm without apparent phosphorylation of Dsh/Dvl (Gonzalez-Sancho et al., 2004). These data argue that LRP and axin act independently of Dsh/Dvl phosphorylation.

How does Fz activate Dsh/Dvl phosphorylation? Several kinases can phosphorylate Dsh/Dvl, including CKIϵ and Par1, which also appear to be required for Wnt/β-catenin signaling (Peters et al., 1999; Sun et al., 2001; Gao et al., 2002; Hino et al., 2003; Cong et al., 2004a; Ossipova et al., 2005). CKIϵ has been reported to be activated by Wnt stimulation, although the mechanism is not clear (Swiatek et al., 2004).

Intriguing candidates for a link between the serpentine Fz receptors and Dsh/Dvl activation are trimeric G proteins. Pertussis toxin, which inactivates some Gα subunits, and antisense oligonucleotides have been used to show that Gαο and Gαq are required for Wnt/β-catenin signaling in cultured cells and Xenopus embryos (Liu et al., 2001; Liu et al., 2002; Malbon, 2004). Moreover, expression of GTPase-defective (i.e. constitutively active) forms of these Gα subunits activates a Wnt/β-catenin reporter gene (Liu et al., 2001). These results are consistent with Gαο/Gαq mediating Wnt/β-catenin signaling. Similarly, reduction of Gαο gene activity compromises Wg signaling in the developing fly wing (Katanaev et al., 2005). Overexpression of wild-type or constitutively active Gαο activates Wg targets, and epitasis experiments demonstrate that Gαο activates the pathway downstream of fz1/fz2 and upstream of dsh. These data are consistent with a model where Gαο is required for Fz to activate Dsh in a Wnt-dependent manner (Fig. 2). However, there is no biochemical confirmation of this relationship at present and the direct effectors of GTP-bound Gαo/Gαq remain to be identified.

Wnt proteins are known to activate several pathways through Fz receptors in addition to the β-catenin pathway (Fig. 3). Some Wnt and some Fz proteins tend to activate β-catenin signaling; others preferentially activate β-catenin-independent pathways. For example, Wnt1, Wnt3a and Wnt8 are often referred to as `canonical/β-catenin Wnts', whereas Wnt 5a and Wnt11 are considered `non-canonical Wnts' (Du et al., 1995; Shimizu et al., 1997; Kuhl et al., 2000). Although these preferences have largely been determined by misexpression studies in Xenopus embryos, they are also observed in other systems (e.g. Heisenberg et al., 2000; Gonzalez-Sancho et al., 2004; Liu, G. et al., 2005,Liu, G. et al., 2005; Takada et al., 2005).

Fig. 3.

Outline of several Wnt signaling pathways. The Wnt/β-catenin, PCP and Wnt/Ca2+ pathways are shown. All three require Fz-family members. Dsh/Dvl is required for both the β-catenin and PCP pathways; its role in Wnt/Ca2+ signaling is less clear (see Veeman et al., 2003). LRP is thought to be specific for Wnt/β-catenin signaling.

Fig. 3.

Outline of several Wnt signaling pathways. The Wnt/β-catenin, PCP and Wnt/Ca2+ pathways are shown. All three require Fz-family members. Dsh/Dvl is required for both the β-catenin and PCP pathways; its role in Wnt/Ca2+ signaling is less clear (see Veeman et al., 2003). LRP is thought to be specific for Wnt/β-catenin signaling.

Many studies blur the distinction between these two Wnt subclasses. The planar cell polarity (PCP) pathway is a noncanonical Wnt pathway that acts through Dsh/Dvl, Rho and Rho-associated kinase (Rho-kinase) (Strutt et al., 1997; Boutros and Mlodzik, 1999; Habas et al., 2001; Winter et al., 2001; Fanto and McNeill, 2004). However, stimulation with the canonical Wnt1 and Wnt3a leads to rapid (10 minutes) activation of Rho and Rho-kinase in several mammalian cell lines (Habas et al., 2001; Kishida et al., 2004). Conversely, the non-canonical Wnt5a can activate β-catenin in mammary epithelial cells (Civenni et al., 2003). Wnt11, which is required for convergent extension (a β-catenin-independent pathway) in frogs and fish (Heisenberg et al., 2000; Tada and Smith, 2000; Wallingford et al., 2000), is also required for the β-catenin-dependent specification of dorsal cell fate in the early Xenopus embryo (Tao et al., 2005).

Fz proteins also refuse to be pigeonholed. Fz7 acts in both β-catenin-dependent and non-canonical Wnt signaling (Djiane et al., 2000; Medina et al., 2000; Sumanas et al., 2000; Habas et al., 2003). In flies, Fz1 and Fz2 act redundantly in Wnt/β-catenin signaling (Bhat, 1998; Bhanot et al., 1999; Chen and Struhl, 1999a; Muller et al., 1999), whereas Fz1 is also required in PCP signaling (Adler, 2002; Vinson et al., 1989). Attempts to map the area of Fz1 required for PCP signaling through the use of Fz1-Fz2 chimeras reveal sequences in both the N- and C-termini of Fz1 (Strapps and Tomlinson, 2001; Wu et al., 2004).

One explanation for Fz signaling diversity is that Fz co-receptors provide specificity. Wnt11 signaling in dorsal specification in Xenopus embryos requires the EGF-CFC protein FRL1, and FRL1 and Wnt11 can be coimmunoprecipitated when overexpressed (Tao et al., 2005). Perhaps FRL1 enables the non-canonical Wnt11 to signal through the β-catenin pathway.

LRP might also regulate the signaling readout of Wnt-Fz interaction. Aaronson and colleagues (Liu, G. et al., 2005,Liu, G. et al., 2005) converted Wnt5a into a canonical Wnt by fusing it to the C-terminus of Dkk2 (Dkk2C), a ligand for LRP5/LRP6. In HEK-293T cells, Wnt5a cannot activate β-catenin, even when cotransfected with ten different Fz isoforms or Dkk2C (Liu, G. et al., 2005,Liu, G. et al., 2005). However, a Wnt5a-Dkk2C fusion protein efficiently activates β-catenin (Liu, G. et al., 2005,Liu, G. et al., 2005). The implication is that Wnt5a is less efficient at promoting Fz-LRP oligomerization than are other Wnt molecules. The ability of particular Wnt molecules to trigger Fz-LRP interaction might determine the likelihood of their activating β-catenin signaling.

Adding to the complicated relationship between Wnt and its receptors is the recent finding that the atypical receptor tyrosine kinase Ryk and its fly (Derailed; DRL) and worm (LIN-18) orthologs play important roles in Wnt signaling. These single-transmembrane-span proteins contain an extracellular domain similar to the secreted Wnt inhibitor WIF (Patthy, 2000), which can bind to Wnts with nanomolar affinity (Hsieh et al., 1999a). On the cytoplasmic side, Ryk contains a PDZ-binding domain and a tyrosine kinase domain lacking some residues thought to be important for catalysis (see Halford and Stacker, 2001).

Ryk-family members have been shown to be required for Wnt signaling in several contexts. In the fly embryonic nervous system, drl mutants display improper axonal guidance across the midline similar to that observed in Wnt5 mutants (Bonkowsky et al., 1999; Yoshikawa et al., 2003; Fradkin et al., 2004). In the mouse, the repulsion of corticospinal tract axons by Wnt1 and Wnt5a, and the inhibition of retinal ganglion cell axon outgrowth, is blocked by incubation with an antibody directed against the WIF domain of Ryk (Liu, Y. et al., 2005,Liu, Y. et al., 2005; Schmitt et al., 2005). Small interfering (si)RNA-mediated knockdown of mouse Ryk also inhibits Wnt3a-induced neurite outgrowth in dorsal root ganglion (DRG) explants (Lu et al., 2004). In C. elegans, loss of lin-18 alters the P7 vulval cell fate in a fashion similar to that seen in Wnt mutants (Inoue et al., 2004). Finally, knocking down Ryk dramatically reduces the ability of Wnt1 to activate a Wnt/β-catenin reporter gene in human HEK-293T cells (Lu et al., 2004).

There are several reports of physical interaction between Ryk and Wnt molecules. Ryk can co-immunoprecipitate with Wnt1 and Wnt3a when co-expressed (Lu et al., 2004) and an exogenously added extracellular domain of Drl (containing the WIF domain) precipitates endogenous Dwnt5 from fly embryonic extracts (Yoshikawa et al., 2003). The affinity of the Wnt1-Ryk interaction is in the nanomolar range (Liu, Y. et al., 2005,Liu, Y. et al., 2005). The loss-of-function data combined with the observed protein interaction argue strongly for Ryk acting as a Wnt receptor.

Does Ryk mediate Wnt signaling in concert with Fz-LRP or independently? The picture is not clear. The WIF domain of Ryk interacts with the CRD of Fz8, and the two proteins may form a ternary complex with Wnt1 (Lu et al., 2004). Several other reports argue that Ryk, LIN-18 or Drl act independently of Fz, but this idea is based largely on negative evidence. For example, a twofold reduction in fz1 and fz2 gene activity in flies has no effect on a Drl mis-expression phenotype (Yoshikawa et al., 2003). In the worm vulva, genetic studies argue that LIN-18 acts in parallel with the LIN-17 Fz receptor (Inoue et al., 2004), although it is not clear whether another Fz is involved. Ryk might thus act by Fz-dependent and -independent mechanisms (Fig. 4).

Downstream, Ryk and Dvl have been shown to interact in HEK-293T cells, in which Ryk is clearly required for β-catenin signaling (Lu et al., 2004). For the other examples mentioned, it is not clear what signaling pathway Ryk uses. In flies, Drl mis-expression is not affected by the loss of zygotic dsh (Yoshikawa et al., 2003), but this could easily be explained by the presence of maternally provided dsh. Although no requirement for β-catenin has been shown to be necessary for proper P7 cell fate (Eisenmann et al., 1998), there are at least four β-catenin genes in C. elegans (Kidd et al., 2005). This opens the possibility that several β-catenins act redundantly in this process downstream of Wnt/LIN-18.

Whatever the mechanism(s) by which Ryk mediates Wnt signaling, it cannot be obligatory for Wnt signaling in all cell types, since knockouts of the only family member in mice or worms do not display widespread defects in Wnt signaling (Halford et al., 2000; Inoue et al., 2004).

Fig. 4.

Three models for Ryk in Wnt signaling. (A) In Drosophila, Drl acts as a receptor for Wnt5 to mediate axonal guidance signaling (Yoshikawa et al., 2003). Whether Drl acts with a Fz and through β-catenin is not clear. (B) In C. elegans vulva development, a Ryk (LIN-18) and Wnt (MOM-2) are thought to act in parallel to a Wnt-Fz pair (LIN-44–LIN17) to specify P7 cell fate (Inoue et al., 2004). The downstream signaling mechanism is not clear. (C) In human HEK-293T cells, Ryk is thought to act as a co-receptor for Fz8 to mediate Wnt/β-catenin signaling (Lu et al., 2004).

Fig. 4.

Three models for Ryk in Wnt signaling. (A) In Drosophila, Drl acts as a receptor for Wnt5 to mediate axonal guidance signaling (Yoshikawa et al., 2003). Whether Drl acts with a Fz and through β-catenin is not clear. (B) In C. elegans vulva development, a Ryk (LIN-18) and Wnt (MOM-2) are thought to act in parallel to a Wnt-Fz pair (LIN-44–LIN17) to specify P7 cell fate (Inoue et al., 2004). The downstream signaling mechanism is not clear. (C) In human HEK-293T cells, Ryk is thought to act as a co-receptor for Fz8 to mediate Wnt/β-catenin signaling (Lu et al., 2004).

Additional components of Wnt signaling that interact with Wnt molecules or their receptors remain to be identified, and others must be more fully understood. Indeed, we have not discussed several secreted factors (e.g. Norrin) that can activate or repress Wnt/β-catenin signaling (Kawano and Kypta, 2003; Xu et al., 2004). Other surface proteins containing CRD-like domains – e.g. Ror (Oishi et al., 2003) and heparin sulfate proteoglycans (Lin, 2004) – have also been implicated in Wnt signaling. There is also evidence that intracellular trafficking of Wnt signaling components can regulate the sensitivity of cells to Wnt molecules. Maturation and trafficking of Fz are inhibited by an endoplasmic reticulum protein called Shisa (Yamamoto et al., 2005). Protein kinase C has been shown to act with Wnt5A to stimulate internalization of Fz4 through Dvl2 and β-arrestin2-dependent endocytosis, which could regulate Wnt signaling (Chen et al., 2003). Endocytosis has also been suggested to be important for Wnt transport between cells and perhaps for signaling activity (Seto and Bellen, 2004). Understanding how these various factors and processes modulate the various Wnt pathways and influence signal specificity will continue to occupy researchers in this field.

Much of our understanding about Wnts and their receptors comes from overexpression studies, often using proteins not found in nature. These are complemented by loss-of-function analyses (based on mutants or RNA interference) to ensure that the proteins being manipulated are physiologically relevant. Although this has been a successful formula for understanding the outlines of Wnt signaling, future research should increasingly focus on what happens to the endogenous receptors immediately after Wnt binding. Does Wnt stimulate the interaction or modification of endogenous Fz, LRP or Ryk? These experiments could be complemented by FRET-based assays, as has been done for LRP5 and axin (Mao et al., 2001b).

If we aim to understand the exact series of biochemical events involved in Wnt reception, can a functional receptor complex be reconstituted in artificial membranes? To date, several Wnts have been purified in biologically active form (Willert et al., 2003; Kishida et al., 2004; Povelones and Nusse, 2005). Whether Wnt receptors can be purified in a useful way remains to be seen. Since the ultimate goal is to learn what happens when Wnt signaling is activated by Wnt molecules (as opposed to overexpression of receptor components), the field will benefit from an increased focus on membrane protein biochemistry.

We thank Nick Tolwinski, Takao Inoue and Wange Lu for helpful discussions concerning their data. Y.I.L. was supported by a predoctoral fellowship from the American Heart Association (0315219Z) and by NIH grants RO1 GM59846 and CA95869 to K.M.C.

Adler, P. N. (
2002
). Planar signaling and morphogenesis in Drosophila.
Dev. Cell
2
,
525
-535.
Axelrod, J. D., Miller, J. R., Shulman, J. M., Moon, R. T. and Perrimon, N. (
1998
). Differential recruitment of Dishevelled provides signaling specificity in the planar cell polarity and Wingless signaling pathways.
Genes Dev.
12
,
2610
-2622.
Bejsovec, A. (
2005
). Wnt pathway activation: new relations and locations.
Cell
120
,
11
-14.
Bhanot, P., Brink, M., Samos, C. H., Hsieh, J. C., Wang, Y., Macke, J. P., Andrew, D., Nathans, J. and Nusse, R. (
1996
). A new member of the frizzled family from Drosophila functions as a Wingless receptor.
Nature
382
,
225
-230.
Bhanot, P., Fish, M., Jemison, J. A., Nusse, R., Nathans, J. and Cadigan, K. M. (
1999
). Frizzled and Dfrizzled-2 function as redundant receptors for Wingless during Drosophila embryonic development.
Development
126
,
4175
-4186.
Bhat, K. M. (
1998
). frizzled and frizzled 2 play a partially redundant role in wingless signaling and have similar requirements to wingless in neurogenesis.
Cell
95
,
1027
-1036.
Bonkowsky, J. L., Yoshikawa, S., O'Keefe, D. D., Scully, A. L. and Thomas, J. B. (
1999
). Axon routing across the midline controlled by the Drosophila Derailed receptor.
Nature
402
,
540
-544.
Boutros, M. and Mlodzik, M. (
1999
). Dishevelled: at the crossroads of divergent intracellular signaling pathways.
Mech. Dev.
83
,
27
-37.
Boutros, M., Mihaly, J., Bouwmeester, T. and Mlodzik, M. (
2000
). Signaling specificity by Frizzled receptors in Drosophila.
Science
288
,
1825
-1828.
Brennan, K., Gonzalez-Sancho, J. M., Castelo-Soccio, L. A., Howe, L. R. and Brown, A. M. (
2004
). Truncated mutants of the putative Wnt receptor LRP6/Arrow can stabilize beta-catenin independently of Frizzled proteins.
Oncogene
23
,
4873
-4884.
Cadigan, K. M. and Nusse, R. (
1997
). Wnt signaling: a common theme in animal development.
Genes Dev.
11
,
3286
-3305.
Cadigan, K. M., Fish, M. P., Rulifson, E. J. and Nusse, R. (
1998
). Wingless repression of Drosophila frizzled 2 expression shapes the Wingless morphogen gradient in the wing.
Cell
93
,
767
-777.
Capelluto, D. G., Kutateladze, T. G., Habas, R., Finkielstein, C. V., He, X. and Overduin, M. (
2002
). The DIX domain targets dishevelled to actin stress fibres and vesicular membranes.
Nature
419
,
726
-729.
Chen, C. M. and Struhl, G. (
1999
). Wingless transduction by the Frizzled and Frizzled2 proteins of Drosophila.
Development
126
,
5441
-5452.
Chen, C. M., Strapps, W., Tomlinson, A. and Struhl, G. (
2004
). Evidence that the cysteine-rich domain of Drosophila Frizzled family receptors is dispensable for transducing Wingless.
Proc. Natl. Acad. Sci. USA
101
,
15961
-15966.
Chen, W., ten Berge, D., Brown, J., Ahn, S., Hu, L. A., Miller, W. E., Caron, M. G., Barak, L. S., Nusse, R. and Lefkowitz, R. J. (
2003
), Dishevelled 2 recruits b-arrestin 2 to mediate Wnt5A-stimulated endocytosis of Frizzled 4.
Science
301
,
1391
-1394.
Civenni, G., Holbro, T. and Hynes, N. E. (
2003
). Wnt1 and Wnt5a induce cyclin D1 expression through ErbB1 transactivation in HC11 mammary epithelial cells.
EMBO Rep.
4
,
166
-171.
Cliffe, A., Hamada, F. and Bienz, M. (
2003
). A role of Dishevelled in relocating Axin to the plasma membrane during wingless signaling.
Curr. Biol.
13
,
960
-966.
Cong, F., Schweizer, L. and Varmus, H. (
2004a
). Casein kinase Iepsilon modulates the signaling specificities of dishevelled.
Mol. Cell. Biol.
24
,
2000
-2011.
Cong, F., Schweizer, L. and Varmus, H. (
2004b
). Wnt signals across the plasma membrane to activate the beta-catenin pathway by forming oligomers containing its receptors, Frizzled and LRP.
Development
131
,
5103
-5115.
Davidson, G., Wu, W., Shen, J., Bilic, J., Fenger, U., Stannek, P., Glinka, A. and Niehrs, C. (
2005
). Casein kinase 1g couples Wnt receptor activation to cytoplasmic signal transduction.
Nature
438
,
867
-872.
Djiane, A., Riou, J., Umbhauer, M., Boucaut, J. and Shi, D. (
2000
). Role of frizzled 7 in the regulation of convergent extension movements during gastrulation in Xenopus laevis.
Development
127
,
3091
-3100.
Du, S. J., Purcell, S. M., Christian, J. L., McGrew, L. L. and Moon, R. T. (
1995
). Identification of distinct classes and functional domains of Wnts through expression of wild-type and chimeric proteins in Xenopus embryos.
Mol. Cell. Biol.
15
,
2625
-2634.
Eisenmann, D. M., Maloof, J. N., Simske, J. S., Kenyon, C. and Kim, S. K. (
1998
). The beta-catenin homolog BAR-1 and LET-60 Ras coordinately regulate the Hox gene lin-39 during Caenorhabditis elegans vulval development.
Development
125
,
3667
-3680.
Fanto, M. and McNeill, H. (
2004
). Planar polarity from flies to vertebrates.
J. Cell Sci.
117
,
527
-533.
Farr, G. H., 3rd, Ferkey, D. M., Yost, C., Pierce, S. B., Weaver, C. and Kimelman, D. (
2000
). Interaction among GSK-3, GBP, axin, and APC in Xenopus axis specification.
J. Cell Biol.
148
,
691
-702.
Fradkin, L. G., van Schie, M., Wouda, R. R., de Jong, A., Kamphorst, J. T., Radjkoemar-Bansraj, M. and Noordermeer, J. N. (
2004
). The Drosophila Wnt5 protein mediates selective axon fasciculation in the embryonic central nervous system.
Dev. Biol.
272
,
362
-375.
Gao, Z. H., Seeling, J. M., Hill, V., Yochum, A. and Virshup, D. M. (
2002
). Casein kinase I phosphorylates and destabilizes the beta-catenin degradation complex.
Proc. Natl. Acad. Sci. USA
99
,
1182
-1187.
Gonzalez-Sancho, J. M., Brennan, K. R., Castelo-Soccio, L. A. and Brown, A. M. (
2004
). Wnt proteins induce dishevelled phosphorylation via an LRP5/6-independent mechanism, irrespective of their ability to stabilize beta-catenin.
Mol. Cell. Biol.
24
,
4757
-4768.
Gregorieff, A. and Clevers, H. (
2005
). Wnt signaling in the intestinal epithelium: from endoderm to cancer.
Genes Dev.
19
,
877
-890.
Habas, R., Kato, Y. and He, X. (
2001
). Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daam1.
Cell
107
,
843
-854.
Habas, R., Dawid, I. B. and He, X. (
2003
). Coactivation of Rac and Rho by Wnt/Frizzled signaling is required for vertebrate gastrulation.
Genes Dev.
17
,
295
-309.
Halford, M. M. and Stacker, S. A. (
2001
). Revelations of the RYK receptor.
BioEssays
23
,
34
-45.
Halford, M. M., Armes, J., Buchert, M., Meskenaite, V., Grail, D., Hibbs, M. L., Wilks, A. F., Farlie, P. G., Newgreen, D. F., Hovens, C. M. et al. (
2000
). Ryk-deficient mice exhibit craniofacial defects associated with perturbed Eph receptor crosstalk.
Nat. Genet.
25
,
414
-418.
He, X., Saint-Jeannet, J. P., Wang, Y., Nathans, J., Dawid, I. and Varmus, H. (
1997
). A member of the Frizzled protein family mediating axis induction by Wnt-5A.
Science
275
,
1652
-1654.
He, X., Semenov, M., Tamai, K. and Zeng, X. (
2004
). LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way.
Development
131
,
1663
-1677.
Heisenberg, C. P., Tada, M., Rauch, G. J., Saude, L., Concha, M. L., Geisler, R., Stemple, D. L., Smith, J. C. and Wilson, S. W. (
2000
). Silberblick/Wnt11 mediates convergent extension movements during zebrafish gastrulation.
Nature
405
,
76
-81.
Herman, M. A., Vassilieva, L. L., Horvitz, H. R., Shaw, J. E. and Herman, R. K. (
1995
). The C. elegans gene lin-44, which controls the polarity of certain asymmetric cell divisions, encodes a Wnt protein and acts cell nonautonomously.
Cell
83
,
101
-110.
Hino, S., Michiue, T., Asashima, M. and Kikuchi, A. (
2003
). Casein kinase I epsilon enhances the binding of Dvl-1 to Frat-1 and is essential for Wnt-3a-induced accumulation of beta-catenin.
J. Biol. Chem.
278
,
14066
-14073.
Hinoi, T., Yamamoto, H., Kishida, M., Takada, S., Kishida, S. and Kikuchi, A. (
2000
). Complex formation of adenomatous polyposis coli gene product and axin facilitates glycogen synthase kinase-3 beta-dependent phosphorylation of beta-catenin and downregulates beta-catenin.
J. Biol. Chem.
275
,
34399
-34406.
Holmen, S. L., Robertson, S. A., Zylstra, C. R. and Williams, B. O. (
2005
). Wnt-independent activation of beta-catenin mediated by a Dkk1-Fz5 fusion protein.
Biochem. Biophys. Res. Commun.
328
,
533
-539.
Hsieh, J. C., Kodjabachian, L., Rebbert, M. L., Rattner, A., Smallwood, P. M., Samos, C. H., Nusse, R., Dawid, I. B. and Nathans, J. (
1999a
). A new secreted protein that binds to Wnt proteins and inhibits their activities.
Nature
398
,
431
-436.
Hsieh, J. C., Rattner, A., Smallwood, P. M. and Nathans, J. (
1999b
). Biochemical characterization of Wnt-frizzled interactions using a soluble, biologically active vertebrate Wnt protein.
Proc. Natl. Acad. Sci. USA
96
,
3546
-3551.
Huelsken, J., Vogel, R., Brinkmann, V., Erdmann, B., Birchmeier, C. and Birchmeier, W. (
2000
). Requirement for beta-catenin in anterior-posterior axis formation in mice.
J. Cell Biol.
148
,
567
-578.
Inoue, T., Oz, H. S., Wiland, D., Gharib, S., Deshpande, R., Hill, R. J., Katz, W. S. and Sternberg, P. W. (
2004
). C. elegans LIN-18 is a Ryk ortholog and functions in parallel to LIN-17/Frizzled in Wnt signaling.
Cell
118
,
795
-806.
Itasaki, N., Jones, C. M., Mercurio, S., Rowe, A., Domingos, P. M., Smith, J. C. and Krumlauf, R. (
2003
). Wise, a context-dependent activator and inhibitor of Wnt signalling.
Development
130
,
4295
-4305.
Katanaev, V. L., Ponzielli, R., Semeriva, M. and Tomlinson, A. (
2005
). Trimeric G protein-dependent frizzled signaling in Drosophila.
Cell
120
,
111
-122.
Kato, M., Patel, M. S., Levasseur, R., Lobov, I., Chang, B. H., Glass, D. A., 2nd, Hartmann, C., Li, L., Hwang, T. H., Brayton, C. F. et al. (
2002
). Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor.
J. Cell Biol.
157
,
303
-314.
Kawano, Y. and Kypta, R. (
2003
). Secreted antagonists of the Wnt signalling pathway.
J. Cell Sci.
116
,
2627
-2634.
Kelly, O. G., Pinson, K. I. and Skarnes, W. C. (
2004
). The Wnt co-receptors Lrp5 and Lrp6 are essential for gastrulation in mice.
Development
131
,
2803
-2815.
Kidd, A. R., 3rd, Miskowski, J. A., Siegfried, K. R., Sawa, H. and Kimble, J. (
2005
). A beta-catenin identified by functional rather than sequence criteria and its role in Wnt/MAPK signaling.
Cell
121
,
761
-772.
Kishida, M., Hino, S., Michiue, T., Yamamoto, H., Kishida, S., Fukui, A., Asashima, M. and Kikuchi, A. (
2001
). Synergistic activation of the Wnt signaling pathway by Dvl and casein kinase I{epsilon}.
J. Biol. Chem.
276
,
33147
-33155.
Kishida, S., Yamamoto, H. and Kikuchi, A. (
2004
). Wnt-3a and Dvl induce neurite retraction by activating Rho-associated kinase.
Mol. Cell. Biol.
24
,
4487
-4501.
Kitagawa, M., Hatakeyama, S., Shirane, M., Matsumoto, M., Ishida, N., Hattori, K., Nakamichi, I., Kikuchi, A. and Nakayama, K. (
1999
). An F-box protein, FWD1, mediates ubiquitin-dependent proteolysis of beta-catenin.
EMBO J.
18
,
2401
-2410.
Klingensmith, J., Nusse, R. and Perrimon, N. (
1994
). The Drosophila segment polarity gene dishevelled encodes a novel protein required for response to the wingless signal.
Genes Dev.
8
,
118
-130.
Koay, M. A. and Brown, M. A. (
2005
). Genetic disorders of the LRP5-Wnt signalling pathway affecting the skeleton.
Trends Mol. Med.
11
,
129
-137.
Kohn, A. D. and Moon, R. T. (
2005
). Wnt and calcium signaling: beta-Catenin-independent pathways.
Cell Calcium
38
,
439
-446.
Kuhl, M., Sheldahl, L. C., Park, M., Miller, J. R. and Moon, R. T. (
2000
). The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape.
Trends Genet.
16
,
279
-283.
Lee, E., Salic, A., Kruger, R., Heinrich, R. and Kirschner, M. W. (
2003
). The roles of APC and Axin derived from experimental and theoretical analysis of the Wnt pathway.
PLoS Biol.
1
,
E10
.
Levasseur, R., Lacombe, D. and de Vernejoul, M. C. (
2005
). LRP5 mutations in osteoporosis-pseudoglioma syndrome and high-bone-mass disorders.
Joint Bone Spine
72
,
207
-214.
Li, L., Yuan, H., Weaver, C. D., Mao, J., Farr, G. H., 3rd, Sussman, D. J., Jonkers, J., Kimelman, D. and Wu, D. (
1999
). Axin and Frat1 interact with dvl and GSK, bridging Dvl to GSK in Wnt-mediated regulation of LEF-1.
EMBO J.
18
,
4233
-4240.
Li, L., Mao, J., Sun, L., Liu, W. and Wu, D. (
2002
). Second cysteine-rich domain of Dickkopf-2 activates canonical Wnt signaling pathway via LRP-6 independently of dishevelled.
J. Biol. Chem.
277
,
5977
-5981.
Lin, X. (
2004
). Functions of heparan sulfate proteoglycans in cell signaling during development.
Development
131
,
6009
-6021.
Liu, G., Bafico, A., Harris, V. K. and Aaronson, S. A. (
2003
). A novel mechanism for Wnt activation of canonical signaling through the LRP6 receptor.
Mol. Cell. Biol.
23
,
5825
-5835.
Liu, G., Bafico, A. and Aaronson, S. A. (
2005
). The mechanism of endogenous receptor activation functionally distinguishes prototype canonical and noncanonical Wnts.
Mol. Cell. Biol.
25
,
3475
-3482.
Liu, P., Wakamiya, M., Shea, M. J., Albrecht, U., Behringer, R. R. and Bradley, A. (
1999
). Requirement for Wnt3 in vertebrate axis formation.
Nat. Genet.
22
,
361
-365.
Liu, T., DeCostanzo, A. J., Liu, X., Wang, H., Hallagan, S., Moon, R. T. and Malbon, C. C. (
2001
). G protein signaling from activated rat frizzled-1 to the beta-catenin-Lef-Tcf pathway.
Science
292
,
1718
-1722.
Liu, T., Lee, Y. N., Malbon, C. C. and Wang, H. Y. (
2002
). Activation of the beta-catenin/Lef-Tcf pathway is obligate for formation of primitive endoderm by mouse F9 totipotent teratocarcinoma cells in response to retinoic acid.
J. Biol. Chem.
277
,
30887
-30891.
Liu, Y., Shi, J., Lu, C. C., Wang, Z. B., Lyuksyutova, A. I., Song, X. and Zou, Y. (
2005
). Ryk-mediated Wnt repulsion regulates posterior-directed growth of corticospinal tract.
Nat. Neurosci.
8
,
1151
-1159.
Logan, C. Y. and Nusse, R. (
2004
). The Wnt signaling pathway in development and disease.
Annu. Rev. Cell Dev. Biol.
20
,
781
-810.
Lowry, W. E., Blanpain, C., Nowak, J. A., Guasch, G., Lewis, L. and Fuchs, E. (
2005
). Defining the impact of beta-catenin/Tcf transactivation on epithelial stem cells.
Genes Dev.
19
,
1596
-1611.
Lu, W., Yamamoto, V., Ortega, B. and Baltimore, D. (
2004
). Mammalian Ryk is a Wnt coreceptor required for stimulation of neurite outgrowth.
Cell
119
,
97
-108.
Malbon, C. C. (
2004
). Frizzleds: new members of the superfamily of G-protein-coupled receptors.
Front. Biosci.
9
,
1048
-1058.
Mao, B., Wu, W., Li, Y., Hoppe, D., Stannek, P., Glinka, A. and Niehrs, C. (
2001a
). LDL-receptor-related protein 6 is a receptor for Dickkopf proteins.
Nature
411
,
321
-325.
Mao, J., Wang, J., Liu, B., Pan, W., Farr, G. H., 3rd, Flynn, C., Yuan, H., Takada, S., Kimelman, D., Li, L. et al. (
2001b
). Low-density lipoprotein receptor-related protein-5 binds to Axin and regulates the canonical Wnt signaling pathway.
Mol. Cell
7
,
801
-809.
Medina, A., Reintsch, W. and Steinbeisser, H. (
2000
). Xenopus frizzled 7 can act in canonical and non-canonical Wnt signaling pathways: implications on early patterning and morphogenesis.
Mech. Dev.
92
,
227
-237.
Muller, H., Samanta, R. and Wieschaus, E. (
1999
). Wingless signaling in the Drosophila embryo: zygotic requirements and the role of the frizzled genes.
Development
126
,
577
-586.
Noordermeer, J., Klingensmith, J., Perrimon, N. and Nusse, R. (
1994
). dishevelled and armadillo act in the wingless signalling pathway in Drosophila.
Nature
367
,
80
-83.
Oishi, I., Suzuki, H., Onishi, N., Takada, R., Kani, S., Ohkawara, B., Koshida, I., Suzuki, K., Yamada, G., Schwabe, G. C. et al. (
2003
). The receptor tyrosine kinase Ror2 is involved in non-canonical Wnt5a/JNK signalling pathway.
Genes Cells
8
,
645
-654.
Ossipova, O., Dhawan, S., Sokol, S. and Green, J. B. (
2005
). Distinct PAR-1 proteins function in different branches of Wnt signaling during vertebrate development.
Dev. Cell
8
,
829
-841.
Patthy, L. (
2000
). The WIF module.
Trends Biochem. Sci.
25
,
12
-13.
Peters, J. M., McKay, R. M., McKay, J. P. and Graff, J. M. (
1999
). Casein kinase I transduces Wnt signals.
Nature
401
,
345
-350.
Pinson, K. I., Brennan, J., Monkley, S., Avery, B. J. and Skarnes, W. C. (
2000
). An LDL-receptor-related protein mediates Wnt signalling in mice.
Nature
407
,
535
-538.
Pinto, D. and Clevers, H. (
2005
). Wnt, stem cells and cancer in the intestine.
Biol. Cell
97
,
185
-196.
Polakis, P. (
2000
). Wnt signaling and cancer.
Genes Dev.
14
,
1837
-1851.
Povelones, M. and Nusse, R. (
2005
). The role of the cysteine-rich domain of Frizzled in Wingless-Armadillo signaling.
EMBO J
.
24
,
3493
-3503.
Reya, T., Duncan, A. W., Ailles, L., Domen, J., Scherer, D. C., Willert, K., Hintz, L., Nusse, R. and Weissman, I. L. (
2003
). A role for Wnt signalling in self-renewal of haematopoietic stem cells.
Nature
423
,
409
-414.
Rothbacher, U., Laurent, M. N., Deardorff, M. A., Klein, P. S., Cho, K. W. and Fraser, S. E. (
2000
). Dishevelled phosphorylation, subcellular localization and multimerization regulate its role in early embryogenesis.
EMBO J.
19
,
1010
-1022.
Rulifson, E. J., Wu, C. H. and Nusse, R. (
2000
). Pathway specificity by the bifunctional receptor frizzled is determined by affinity for wingless.
Mol. Cell
6
,
117
-126.
Salic, A., Lee, E., Mayer, L. and Kirschner, M. W. (
2000
). Control of beta-catenin stability: reconstitution of the cytoplasmic steps of the wnt pathway in Xenopus egg extracts.
Mol. Cell
5
,
523
-532.
Sawa, H., Lobel, L. and Horvitz, H. R. (
1996
). The Caenorhabditis elegans gene lin-17, which is required for certain asymmetric cell divisions, encodes a putative seven-transmembrane protein similar to the Drosophila frizzled protein.
Genes Dev.
10
,
2189
-2197.
Schmitt, A. M., Shi, J., Wolf, A. M., Lu, C. C., King, L. A. and Zou, Y. (
2005
). Wnt-Ryk signalling mediates medial-lateral retinotectal topographic mapping.
Nature
439
,
31
-37.
Schweizer, L. and Varmus, H. (
2003
). Wnt/Wingless signaling through beta-catenin requires the function of both LRP/Arrow and frizzled classes of receptors.
BMC Cell Biol.
4
,
4
.
Semenov, M. V., Tamai, K., Brott, B. K., Kuhl, M., Sokol, S. and He, X. (
2001
). Head inducer Dickkopf-1 is a ligand for Wnt coreceptor LRP6.
Curr. Biol.
11
,
951
-961.
Seto, E. S. and Bellen, H. J. (
2004
). The ins and outs of Wingless signaling.
Trends Cell Biol.
14
,
45
-53.
Shimizu, H., Julius, M. A., Giarre, M., Zheng, Z., Brown, A. M. and Kitajewski, J. (
1997
). Transformation by Wnt family proteins correlates with regulation of beta-catenin.
Cell Growth Differ.
8
,
1349
-1358.
Strapps, W. R. and Tomlinson, A. (
2001
). Transducing properties of Drosophila Frizzled proteins.
Development
128
,
4829
-4835.
Strutt, D. I., Weber, U. and Mlodzik, M. (
1997
). The role of RhoA in tissue polarity and Frizzled signalling.
Nature
387
,
292
-295.
Sumanas, S., Strege, P., Heasman, J. and Ekker, S. C. (
2000
). The putative wnt receptor Xenopus frizzled-7 functions upstream of beta-catenin in vertebrate dorsoventral mesoderm patterning.
Development
127
,
1981
-1990.
Sun, T. Q., Lu, B., Feng, J. J., Reinhard, C., Jan, Y. N., Fantl, W. J. and Williams, L. T. (
2001
). PAR-1 is a Dishevelled-associated kinase and a positive regulator of Wnt signalling.
Nat. Cell Biol.
3
,
628
-636.
Swiatek, W., Tsai, I. C., Klimowski, L., Pepler, A., Barnette, J., Yost, H. J. and Virshup, D. M. (
2004
). Regulation of casein kinase I epsilon activity by Wnt signaling.
J. Biol. Chem.
279
,
13011
-13017.
Tada, M. and Smith, J. C. (
2000
). Xwnt11 is a target of Xenopus Brachyury: regulation of gastrulation movements via Dishevelled, but not through the canonical Wnt pathway.
Development
127
,
2227
-2238.
Takada, R., Hijikata, H., Kondoh, H. and Takada, S. (
2005
). Analysis of combinatorial effects of Wnts and Frizzleds on beta-catenin/armadillo stabilization and Dishevelled phosphorylation.
Genes Cells
10
,
919
-928.
Tamai, K., Semenov, M., Kato, Y., Spokony, R., Liu, C., Katsuyama, Y., Hess, F., Saint-Jeannet, J. P. and He, X. (
2000
). LDL-receptor-related proteins in Wnt signal transduction.
Nature
407
,
530
-535.
Tamai, K., Zeng, X., Liu, C., Zhang, X., Harada, Y., Chang, Z. and He, X. (
2004
). A mechanism for Wnt coreceptor activation.
Mol. Cell
13
,
149
-156.
Tao, Q., Yokota, C., Puck, H., Kofron, M., Birsoy, B., Yan, D., Asashima, M., Wylie, C. C., Lin, X. and Heasman, J. (
2005
). Maternal wnt11 activates the canonical wnt signaling pathway required for axis formation in Xenopus embryos.
Cell
120
,
857
-871.
Theisen, H., Purcell, J., Bennett, M., Kansagara, D., Syed, A. and Marsh, J. L. (
1994
). dishevelled is required during wingless signaling to establish both cell polarity and cell identity.
Development
120
,
347
-360.
Tolwinski, N. S. and Wieschaus, E. (
2001
). Armadillo nuclear import is regulated by cytoplasmic anchor Axin and nuclear anchor dTCF/Pan.
Development
128
,
2107
-2117.
Tolwinski, N. S. and Wieschaus, E. (
2004
). Rethinking WNT signaling.
Trends Genet.
20
,
177
-181.
Tolwinski, N. S., Wehrli, M., Rives, A., Erdeniz, N., DiNardo, S. and Wieschaus, E. (
2003
). Wg/Wnt signal can be transmitted through arrow/LRP5,6 and Axin independently of Zw3/Gsk3beta activity.
Dev. Cell
4
,
407
-418.
Umbhauer, M., Djiane, A., Goisset, C., Penzo-Mendez, A., Riou, J. F., Boucaut, J. C. and Shi, D. L. (
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 Es, J. H., Barker, N. and Clevers, H. (
2003
). You Wnt some, you lose some: oncogenes in the Wnt signaling pathway.
Curr. Opin. Genet. Dev.
13
,
28
-33.
Veeman, M. T., Axelrod, J. D. and Moon, R. T. (
2003
). A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling.
Dev. Cell
5
,
367
-377.
Vinson, C. R., Conover, S. and Adler, P. N. (
1989
). A Drosophila tissue polarity locus encodes a protein containing seven potential transmembrane domains.
Nature
338
,
263
-264.
Wallingford, J. B., Rowning, B. A., Vogeli, K. M., Rothbacher, U., Fraser, S. E. and Harland, R. M. (
2000
). Dishevelled controls cell polarity during Xenopus gastrulation.
Nature
405
,
81
-85.
Wehrli, M., Dougan, S. T., Caldwell, K., O'Keefe, L., Schwartz, S., Vaizel-Ohayon, D., Schejter, E., Tomlinson, A. and DiNardo, S. (
2000
). arrow encodes an LDL-receptor-related protein essential for Wingless signalling.
Nature
407
,
527
-530.
Willert, K., Brink, M., Wodarz, A., Varmus, H. and Nusse, R. (
1997
). Casein kinase 2 associates with and phosphorylates dishevelled.
EMBO J.
16
,
3089
-3096.
Willert, K., Shibamoto, S. and Nusse, R. (
1999
). Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex.
Genes Dev.
13
,
1768
-1773.
Willert, K., Brown, J. D., Danenberg, E., Duncan, A. W., Weissman, I. L., Reya, T., Yates, J. R., 3rd and Nusse, R. (
2003
). Wnt proteins are lipid-modified and can act as stem cell growth factors.
Nature
423
,
448
-452.
Winter, C. G., Wang, B., Ballew, A., Royou, A., Karess, R., Axelrod, J. D. and 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. and Nusse, R. (
1998
). Mechanisms of Wnt signaling in development.
Annu. Rev. Cell Dev. Biol.
14
,
59
-88.
Wong, H. C., Bourdelas, A., Krauss, A., Lee, H. J., Shao, Y., Wu, D., Mlodzik, M., Shi, D. L. and Zheng, J. (
2003
). Direct binding of the PDZ domain of Dishevelled to a conserved internal sequence in the C-terminal region of Frizzled.
Mol. Cell
12
,
1251
-1260.
Wu, C. H. and Nusse, R. (
2002
). Ligand receptor interactions in the Wnt signaling pathway in Drosophila.
J. Biol. Chem.
277
,
41762
-41769.
Wu, J., Klein, T. J. and Mlodzik, M. (
2004
). Subcellular localization of frizzled receptors, mediated by their cytoplasmic tails, regulates signaling pathway specificity.
PLoS Biol.
2
,
E158
.
Xu, Q., Wang, Y., Dabdoub, A., Smallwood, P. M., Williams, J., Woods, C., Kelley, M. W., Jiang, L., Tasman, W., Zhang, K. et al. (
2004
). Vascular development in the retina and inner ear: control by Norrin and Frizzled-4, a high-affinity ligand-receptor pair.
Cell
116
,
883
-895.
Yamamoto, H., Kishida, S., Kishida, M., Ikeda, S., Takada, S. and Kikuchi, A. (
1999
). Phosphorylation of axin, a Wnt signal negative regulator, by glycogen synthase kinase-3beta regulates its stability.
J. Biol. Chem.
274
,
10681
-10684.
Yamamoto, A., Nagano, T., Takehara, S., Hibi, M. and Aizawa, S. (
2005
). Shisa promotes head formation through the inhibition of receptor protein maturation for the caudalizing factors, Wnt and FGF.
Cell
120
,
223
-235.
Yanagawa, S., van Leeuwen, F., Wodarz, A., Klingensmith, J. and Nusse, R. (
1995
). The dishevelled protein is modified by wingless signaling in Drosophila.
Genes Dev.
9
,
1087
-1097.
Yang-Snyder, J., Miller, J. R., Brown, J. D., Lai, C. J. and Moon, R. T. (
1996
). A frizzled homolog functions in a vertebrate Wnt signaling pathway.
Curr. Biol.
6
,
1302
-1306.
Yoshikawa, S., McKinnon, R. D., Kokel, M. and Thomas, J. B. (
2003
). Wnt-mediated axon guidance via the Drosophila Derailed receptor.
Nature
422
,
583
-588.
Zeng, X., Tamai, K., Doble, B., Li, S., Huang, H., Habas, R., Okamura, H., Woodget, J. and He, X. (
2005
). A dual-kinase mechanism for Wnt coreceptor phosphorylation and activation.
Nature
438
,
873
-877.
Zhang, J. and Carthew, R. W. (
1998
). Interactions between Wingless and DFz2 during Drosophila wing development.
Development
125
,
3075
-3085.