Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Nov 1.
Published in final edited form as: Bone. 2015 Nov;80:60–66. doi: 10.1016/j.bone.2015.02.009

Wnt Signaling in Bone and Muscle

Michael A Rudnicki 1, Bart O Williams 2
PMCID: PMC4600531  NIHMSID: NIHMS664751  PMID: 26453496

Introduction

Wnt signaling plays central roles in many aspects of development and alterations in the pathway are commonly associated with human disease. The first Wnt gene was identified by Nusse and Varmus in 1982 based on its location in a chromosomal region enriched for integration sites in mouse mammary tumor virus (MMTV)-induced mammary adenocarcinomas [1]. It was originally referred to as Integration site-1 (int-1) because of this relationship with MMTV and inferred to be a secreted protein based on the presence of a signal peptide. The protein proved difficult to purify in biologically active form [2], so the initial identification of downstream signaling components was based on genetic systems. A key system in which this was examined was the development of embryonic segment polarity in Drosophila melanogaster. This was based on the fact that the Drosophila Wingless gene was found to be a close homolog of the mouse int-1 gene [3]. The observation that Wingless was a component of the well-defined segment polarity pathway allowed several other genes to be functionally linked to the Wingless/int-1 gene (reviewed in [4]). Because of the homology between int1 and Wingless, the gene was renamed as Wnt1 (Wingless + int1) [5] and was eventually recognized as the founding member a large Wnt gene family encoded by 19 different genes in humans and mice. Wnt proteins share characteristics as cysteine-rich glycoproteins dependent on the addition of a lipid modification for function [6].

In this review, we will briefly describe the components of signaling pathways induced by Wnt ligands and then describe the current state of research as this applies to aspects of development and disease as it relates to skeletal muscle and bone. We will conclude with a discussion of the parallels and differences in Wnt signaling in these two contexts and how these pathways are being (or could potentially be) targeted for therapeutic treatment of musculoskeletal diseases.

Wnt-Induced Signaling Pathways

The requirement for Wnt signaling in the establishment of Drosophila embryonic segment polarity helped define the core components of one signaling pathway initiated by Wnt ligands during the early to mid-1990s. This specific Wnt pathway, dependent on the β-catenin protein (armadillo in Drosophila), is referred to as the “canonical” pathway (reviewed in [7, 8]) (Fig. 1). In this pathway, a Wnt ligand engages a receptor complex that includes a member of the Frizzled family of seven transmembrane receptor proteins and either the Low density lipoprotein related receptors 5 and 6 (Lrp5/6). In the absence of this receptor complex, the cytoplasmic and nuclear levels of the β-catenin protein are reduced or absent due to Glycogen synthase kinase 3 (GSK3)-dependent phosphorylation of residues near its N-terminus which targets the protein for ubiquitin-dependent proteolysis. GSK3 has numerous putative substrates, but in the context of canonical Wnt signaling its activity is regulated by the formation of a multiprotein complex which includes the Adenomatous polyposis coli (APC) protein and Axin. Upon Wnt binding to the LRP5/6;Frizzled receptor complex, the cytoplasmic tail of the Lrp5/6 component becomes phosphorylated creating a binding site for Axin. Recruitment of Axin to this site results inhibits GSK3-mediated phosphorylation of β-catenin, allowing β-catenin levels to increase in the cytoplasm. In some cell types, additional pathways downstream of Wnt ligands may be required for nuclear localization of β-catenin. Upon entering the nucleus, β-catenin interacts with members of the LEF/TCF family of DNA binding proteins to bind to and activate target gene transcription.

Figure 1. Overview of Wnt Signaling Pathways.

Figure 1

A. Canonical signaling pathways are typically defined as those that are dependent on the stabilization of the β-catenin protein. In this context, Wnt ligands bind to a receptor complex that includes a member of the Frizzled family of seven transmembrane proteins and either Lrp5 or Lrp6. The engagement of this receptor complex leads to the phosphorylation of serine residues in the cytoplasmic tail of Lrp5/6 creating a binding site for the Axin protein. Axin is a component of a multi-protein complex which also includes the adenomatous polyposis coli (APC) protein and the serine/threonine protein kinase, Glycogen synthase kinase 3 (GSK3). In the absence of an upstream Wnt signal, this multiprotein complex facilitates the GSK3-dependent phosphorylation of β-catenin, targeting it for ubiquitin-dependent proteolysis. When Axin is recruited to the plasma membrane via its association with phosphorylated Lrp5/6, GSK3-mediated phosphorylation of β-catenin is inhibited, leading to increased levels of cytoplasmic β-catenin. β-catenin then can undergo nuclear translocation and lead to target gene activation. B. Non-canonical signaling pathways refer to the group of pathways induced by Wnt ligands that act via β-catenin-independent mechanisms. These include a pathway that activates the small GTPases, RhoA and Rac1, resulting in cytoskeletal rearrangement and activation of Jun N terminal kinase (JNK) and a pathway that induces PI3K and subsequent activation of Akt. Adapted from [106].

The historical focus on the “canonical” Wnt/β-catenin pathway has resulted in relatively less attention being played to the myriad of other signaling pathways activated by Wnt ligands. The shared characteristic of these pathways is that they signal in a β-catenin-independent manner [9] (Fig. 1). These pathways can be induced by activation of several alternative Wnt receptors such as Ror2 and Ryk, but may also be initiated by engagement of Frizzled receptors by Wnts, often in the absence of co-engagement of Lrp5/6. One of the best known of these pathways is referred to as the planar cell polarity (PCP) pathway. This pathway, like the “canonical” pathway, was originally characterized in Drosophila melanogaster. Here frizzled activation signals through the small GTPase, RhoA, and Jun N-terminal kinase (JNK) to regulate cell-cell communication required for the proper orientation of wing hair and the ommatidia of the compound eye [10]. The PCP pathway also influences cytoskeletal remodeling, cell adhesion, and motility in mammalian systems [11]. Other β-catenin-independent pathways induced by Wnts include those that activate Protein Kinase C [12], Protein Kinase A [13], mTOR [14], and PI3K/Akt [15].

The activation of receptor complexes by Wnts is also under complex regulatory control. For example, the four members of the Secreted family of frizzled proteins (sFRPs), can inhibit Wnt-induced signaling by preventing the interaction between Wnts and the Frizzled component of the co-receptor [16]. In addition, proteins such as members of the Dickkopf family [17] and Sclerostin [18] inhibit β-catenin-dependent, canonical signaling by preventing Wnt ligands from binding to Lrp5/6. An interesting recent development in this context is the demonstration that Lrp4 acts within the local microenvironment to bind Sclerostin and present it to Lrp5/6 to facilitate inhibition of Wnt signaling [19].

Recently, the processes involved in the production and secretion of Wnts have become more defined. Again, the foundation for this work was based on Drosophila genetics which identified the requirement for the gene, Porcupine, in Wnt-producing cells for establishment of embryonic segment polarity [20]. The protein encoded by Porcupine was subsequently identified as a member of the membrane-bound O-acyl transferase family required to acylate Wnt ligands [21]. Lipid-modified Wnts are then bound by a protein referred to as Wntless (also known as GPR177 or Sprinter) to allow transit to the plasma membrane and subsequent secretion [22]. All naturally occurring Wnt proteins are thought to require lipid modification by Porcupine and Wntless-mediated transport for secretion.

Role of Wnt Signaling in Bone Development and Homeostasis

The intense interest in understanding the role of Wnt signaling in regulating bone development and homeostasis can be traced to three seminal manuscripts in the early 2000s. The first reported that the underlying cause of Osteoporosis pseudoglioma (OPPG), a syndrome in which affected patients have extremely low bone mass, was homozygosity for loss of function mutations in LRP5 [23]. Shortly after this, two groups independently reported that a point mutation in LRP5 (G171V) was the underlying cause of a dominantly inherited high bone mass (HBM) trait [24, 25]. The mechanistic explanation for high bone mass is that the G171V mutation produces an LRP5 protein that can no longer interact with inhibitors such as DKK1 and Sclerostin [26-30] (as well as other proteins such as MeSD [31, 32]). While some individuals with HBM suffer from complications such as headaches [33], the fact that they do not appear to be predisposed to carcinogenesis spurred a great deal of interest on the part of biotechnology and pharmaceutical companies in developing agents that mimicked the results of this alteration. Subsequent studies have reported additional mutations in the LRP5 gene that increase or decrease human bone mass (for example see [34]).

Mouse models in which Lrp5 was globally deleted demonstrated reduced bone mass [35-37], further validating that loss of LRP5 was the underlying cause of OPPG. In addition, osteoblast-specific expression of LRP5G171V resulted in the predicted high bone mass [38]. The link between LRP5 and β-catenin lead to an examination of the roles of other components of the canonical Wnt pathway in bone development and disease. For example, conditional deletion of Ctnnb1 (which encodes β-catenin) in osteoblasts or osteocytes resulted in severely low bone mass, while conditional activation of β-catenin by either cre-mediated expression of a non-degradable form of β-catenin or via deletion of Apc lead to dramatically increased bone mass [39-41]. In both cases, further work linked alterations in β-catenin signaling to dysregulation of the OPG/RANKL signaling axis. Specifically, OPG was shown to be a direct transcriptional target of β-catenin [39]. Thus, changes in bone mass seen in the context of altered Wnt signaling within osteoblasts, at least in part, may be due to alterations in osteoclast function or differentiation caused by changes in RANKL-induced signaling. There are several additional mechanisms by which Wnts derived from osteoblasts or other cells in the skeletal microenvironment influence osteoclast function. For example, osteoblast-derived Wnt5a can enhance osteoclast differentiation and activated via activation of the ROR2 receptor [42]. Also, the demonstration that dynamic regulation of β-catenin is required for proper osteoclast differentiation suggests that Wnt ligands may have stage-specific effects during osteoclastogenesis [43].

Osteoblast-specific deletion of the Lrp5 and Lrp6 genes also lead to significant reductions in bone mass [44], consistent with a role for canonical Wnt signaling within the osteoblast lineage in controlling bone mass. However, an alternative explanation in which Lrp5-mediated regulation of serotonin production by the enterochromaffin cells of the intestine has also been proposed [45-47]. At early stages of osteoblast differentiation, canonical Wnt signaling is required for commitment to the osteoblast lineage. Deletion of either Ctnnb1 [48-50] or Lrp5 and Lrp6 [51] results in a failure to efficiently form bone.

Alterations in known negative regulators of the canonical signaling pathway have also been clearly linked to changes in bone mass in both humans and mice. For example, alterations in the function of the Sclerostin (SOST) locus, which encodes a protein that inhibits canonical Wnt signaling, are causally linked to two syndromes associated with high bone mass, Sclerosteosis and van Buchem’s disease [52, 53]. This association was also validated in mouse models. In addition, mouse models with impaired Dkk1 function, another negative regulator of the pathway, also develop high bone mass [54].

In addition to work on the roles of the Wnt receptor complex and signaling components downstream of its activation, recent work has revealed key roles for specific Wnt ligands and components regulating its secretion from Wnt-producing cells in establishing and/or maintaining normal bone mass. For example, a meta-analysis of SNPs associated with alterations in human bone mineral density identified the genomic region where Wntless/GPR177 is located as containing SNPs significantly associated with changes in bone mineral density [55]. The importance of GPR177 has been validated using several mouse models which demonstrate that loss of GPR177 results in low bone mass [56-58]. Finally, recent work on the genetic causes of Osteogenesis imperfecta, a disease characterized by the presence of brittle bones, identified putative causative mutations in WNT1 [59-61]. This establishes a key role for Wnt1 in establishing and maintaining human bone mass, however the precise mechanisms by which this occurs await further study.

Perhaps because OPPG, Sclerosteosis, and van Buchem’s disease were also associated with alterations in bone mass correlating with changes in β-catenin signaling, non-canonical Wnt pathways have received less attention in the context of bone development. However, it is clear that non-canonical pathways also contribute to this process. For example, mouse models demonstrate that Wnt5a secreted from cells of the osteoblast lineage activates ROR2 on osteoclasts to regulate their differentiation and function [42]. In addition, mice lacking Frizzled 9 have reduced bone mass due to alterations in signaling pathways that are independent of β-catenin [62, 63]. Also, Wnt signaling acting through PKCΔ promotes bone formation [64]. These are just some examples of the demonstrated roles for noncanonical Wnt signaling in bone development, and there are undoubtedly additional examples that will emerge as we learn more about the functions of these pathways in other systems. Nowhere is this more apparent than in our understanding of muscle cell regeneration.

Wnt Signaling in Myogenesis

Skeletal muscle is derived from somites during embryonic development [65], and myogenic specification in somatic cells is regulated by signals from the surrounding tissues (Fig. 2). Expression of Wnt-1, -3a, -4, -6, -7a and 11 from dorsal neural tube or ectoderm is critical for the induction, initiation and progression of myogenesis in the presomitic mesoderm and early somites [66, 67]. Within the embryonic myogenic progenitors, Wnts also regulate the expression of Pax3/7, MyoD and Myf5, key transcription factors involved in myogenesis [68-72]. Genetic studies have demonstrated the role of several Wnt molecules and β-catenin in the normal development of skeletal muscles [73, 74]. Therefore, canonical and non-canonical Wnt signaling pathways play multiple essential roles in embryonic myogenesis.

Figure 2. Wnt signaling and the embryonic development of skeletal muscle.

Figure 2

Wnt signaling from surrounding tissues influences the development of muscle in a spaciotemporal manner. The dorsal regions of the neural tube express Wnt1, Wnt3a and Wnt4 whereas the dorsal ectoderm expresses Wnt4, Wnt6 and Wnt7a. The epaxial dermomyotome expresses Wnt11.

The role of canonical Wnt/β-catenin signaling in the regulation of postnatal satellite cell function and skeletal muscle regeneration has been controversial. Satellite cells are muscle resident stem cells responsible for postnatal regeneration of injured muscles. Canonical Wnt activation has been suggested to induce satellite cell proliferation during skeletal muscle regeneration [75]. However, contradictory results showing that activation of canonical Wnt signaling is necessary to counteract Notch signaling to induce myogenic differentiation were also reported [76]. Furthermore, in the aged niche, elevated systemic Wnt molecules impede myogenic differentiation and facilitate satellite cell fate conversion to fibroblastic cell lineages [77]. Recently, a thorough genetic study in adult satellite cells demonstrated that it is not activation of Wnt/β-catenin signaling but rather silencing that is important for muscle regeneration [76].

Adult skeletal muscles contain heterogeneous types of muscle fibers that can be broadly divided into slow- and fast-twitch myofibers. In the limb muscles, the slow-twitch myofibers express type I myosin heavy chain (MyHC), while the fast-twitch myofibers express type IIa, IIx and IIb MyHC isoforms [78]. During early stages of myofiber generation in chicks, Wnt11 promotes fast myofiber formation, whereas Wnt5 enhances slow myofiber generation [79]. Wnt4 similarly stimulates fast myofiber formation in chicks [80]. Activation of β-catenin induced myofiber hypertrophy followed by degeneration of fast myofibers in zebrafish [81]. In mice, depletion of β-catenin during myogenesis in Pax7-lineage cells led to reduced slow myofibers and overall reduction of muscle mass [82]. During fetal development in mice, Wnt/ β-catenin signaling induces BMP4 expression in myoblasts which in turn induces slow myofibrogenesis (Skelet Muscle. 2013 Mar 5;3(1):5. doi: 10.1186/2044-5040-3-5). Together, these studies suggest that Wnt signaling plays diverse roles in regulating slow-versus fast-twitch myofiber formation during development.

Role of Wnt Signaling in Muscle Growth and Regeneration

Satellite cells located beneath the basal lamina of myofibers are required for the growth and regeneration of skeletal muscle [83]. Molecular genetic studies in mice have established that a small subset of the satellite cell population are stem cells that are capable of reconstituting the satellite cell population following transplantation, of long-term self-renewal, and of giving rise to committed myogenic progenitors through asymmetric apical-basal cell divisions [84, 85]. Cre-LoxP lineage tracing in mice using Myf5-Cre and R26R-YFP alleles allows the discrimination between committed satellite myogenic cells that have expressed Myf5-Cre (YFP+), and a small subpopulation (<10 %) of satellite stem cells that have never expressed Myf5-Cre (YFP) [84].

Satellite stem cells also express high levels of Fzd7 and signaling through the Wnt7a/Fzd7 planar-cell-polarity (PCP) pathway drives the symmetric expansion of satellite stem cells to accelerate and augment muscle regeneration [86]. Closer examination showed that administration of recombinant Wnt7a markedly stimulated the symmetric expansion of satellite stem cells but did not affect the growth or differentiation of myoblasts. Conversely, silencing of Fzd7 abrogated Wnt7a binding and stimulation of stem cell expansion. Wnt7a signaling was shown to induce the polarized distribution of the planar cell polarity effector Vangl2, and silencing of Vangl2 abrogated Wnt7a action on satellite stem cell expansion. Wnt7a overexpression enhanced muscle regeneration and increased both the number of satellite cells and muscle mass. Conversely, mice lacking Wnt7a exhibited a decrease in satellite cell number following regeneration. Therefore, Wnt7a signaling through the PCP pathway controls the homeostatic level of satellite stem cells and hence regulates the regenerative potential of muscle.

Over-expression of Wnt7a results in a significant hypertrophic response of skeletal muscle [15]. Wnt7a/Fzd7 signalling in myofibers was found to directly activate the Akt/mTOR growth pathway thereby inducing hypertrophy. Notably, the Fzd7 receptor complex was associated with Gαs and PI3kinase and these components were required for Wnt7a to activate the Akt/mTOR growth pathway in myotubes. Surprisingly, Wnt7a/Fzd7 activation of this pathway was completely independent of IGF-receptor activation. Together, these experiments demonstrate that Wnt7a/Fzd7 activates distinct pathways at different developmental stages during myogenic lineage progression, and together identify a novel non-canonical anabolic signalling pathway for Wnt7a and its receptor Fzd7 in skeletal muscle [15].

The remodeling of the stem cell niche in muscle by satellite cells expressing fibronectin (FN) plays an unexpected role in Wnt7a signaling and satellite stem cell expansion [87]. Syndecan-4 (Sdc4) and Fzd7 form a co-receptor complex in satellite cells and binding of the ECM glycoprotein FN to Sdc4 stimulates the ability of Wnt7a to induce the symmetric expansion of satellite stem cells. Newly activated satellite cells dynamically remodel their niche by transient high-level expression of FN. Knockdown of FN in prospectively isolated satellite cells severely impaires their ability to repopulate the satellite cell niche. Conversely, in vivo over-expression of FN with Wnt7a dramatically enhances the expansion of satellite stem cells in regenerating muscle. Therefore, activating satellite cells remodel their niche through autologous expression of FN that provides feedback to stimulate Wnt7a signalling through the Fzd7/Sdc4 co-receptor complex. Thus, FN and Wnt7a together regulate the homeostatic levels of satellite stem cells and satellite myogenic cells during regenerative myogenesis [87].

Wnt7a/Fzd7 also acts at another level during muscle growth and repair to increase the polarity and directional migration of murine satellite cells and human myogenic progenitors through activation of Dvl2 and the small GTPase Rac1 [88]. Importantly, these effects can be exploited to potentiate the outcome of myogenic cell transplantation into dystrophic muscles. A short 3h Wnt7a treatment resulted in a marked stimulation of tissue dispersal and engraftment of transplanted satellite cells leading to significantly improved muscle function. Moreover, myofibers at distal sites that fused with Wnt7a-treated cells were hypertrophic suggesting that the transplanted cells deliver activated Wnt7a/Fzd7 signaling complexes to recipient myofibers. Interestingly, satellite cells and their daughter progenitors take up Wnt7a/Fzd7 complexes by clathrin-dependent endocytosis into long-lived intracellular vesicles and delivery of these complexes to myofibers after fusion of the migrating progenitors stimulates hypertrophy. Taken together, these findings describe a viable and effective ex vivo cell modulation process that profoundly enhances the efficacy of stem cell therapy for skeletal muscle [88].

Thus, Wnt7a acts at multiple levels as an intrinsic factor to positively regulate regeneration: First, Wnt7a stimulates the symmetric expansion of the satellite stem cell compartment via the non-canonical PCP-signaling pathway; Second, Wnt7 signaling simulates the polarity and motility of satellite cells and myogenic progenitors via non-canonical PCP-signaling; Third, Wnt7a directly induces myofiber hypertrophy by activating the AKT/mTOR growth pathway.

Wnt7a treatment of the tibialis anterior (TA) muscle in an mdx mouse resulted in a significant increase in strength, as determined by generation of specific force per unit mass, and markedly reduced levels of contractile damage, likely due to a shift towards slow-twitch fiber type [89]. Wnt7a induced myotube hypertrophy and a shift towards slow-twitch fiber type in human primary myotubes. Taken together, these findings suggest that Wnt7a is a promising candidate for development as an ameliorative treatment for patients with DMD.

Duchenne Muscular Dystrophy (DMD) is a devastating genetic muscular disorder of childhood manifested by progressive debilitating muscle weakness and wasting, and ultimately death in the second or third decade of life [90]. Wnt7a treatment was found to efficiently induce satellite cell expansion and myofiber hypertrophy in treated muscles of mdx mice [89]. Importantly, Wnt7a treatment resulted in a significant increase in strength of the muscle as determined by generation of specific force. Furthermore Wnt7a reduced the level of contractile damage likely by inducing a fiber type shift towards slow-twitch. Wnt7a similarly induced myotube hypertrophy and a shift in fiber type towards slow-twitch in human primary myotubes. Therefore, Wnt7a appears to be a promising candidate for development as an ameliorative treatment for muscular dystrophy [89].

Opportunities for Therapeutics

The Wnt pathway has emerged as a therapeutic target for many diseases [91]. In the context of bone and muscle, therapies are being developed and evaluated that aim to increase the activity of the pathway. This is contrast to therapeutic programs in the oncology space which aim to treat several tumor types by blocking Wnt signaling activity. For example, Porcupine inhibitors are currently in clinical trials to treat several types of cancer [92]. It would not be surprising based on mouse models in which Wnt secretion from osteoblasts has been blocked by deletion of Wntless/GPR177 [58] if patients being treated with these inhibitors are susceptibility to side effects associated with decreasing bone mass. Conversely, clinicians should be appropriately cognizant of the potential for Wnt activating therapies to predispose to inappropriate cellular growth. Thankfully, this has not been a problem to date perhaps due to the specificity of some of the therapies. For example, Sclerostin is primarily expressed from mature osteocytes and is thought to act in the local environment, so the effects of anti-sclerostin therapies would be predicted to be limited to the skeleton [18].

Several agents which upregulate canonical Wnt signaling are being developed. These include strategies to block the function of Sclerostin or DKK1 which are currently being tested in human clinical trials [93]. In addition, recent studies characterizing the activity of antibodies designed to modulate Lrp6-mediated activation of β-catenin signaling have potential future promise [94, 95]. In addition, the direct application of liposomal vesicles packaged with purified Wnt3a stimulates bone regeneration and engraftment [96-98]. Finally, treatment with lithium chloride, which stabilizes β-catenin via its inhibition of GSK-3 activity [99], can also enhance fracture healing [100, 101].

An obstacle to the use of Wnt7a in recombinant protein therapy is its hydrophobicity caused by the palmitoylation of the protein during post-translational processing prior to secretion. The recently solved crystal structure of a Wnt/Frizzled complex was consistent with the concept that the lipid-modification of Wnt was necessary for function as the Frizzled protein contains a cleft that specifically interacts with this lipid modification [102]. This lipid modification is also consistent with the difficulty in purifying biologically active Wnt proteins as the purification protocols resulted in Wnt proteins lacking the Wnt modification. This lipid modification also complicates the development of Wnt proteins for potential therapeutic use as the associated hydrophobicity creates challenges for delivery.

Recently, however, one of the authors of this review developed a variant of Wnt7a that is comprised of the final carboxyl-terminal 137 amino acids (Wnt7aCT), and entirely lacks lipidation sites [103]. Wnt7aCT exhibits similar or superior effect on muscle regeneration relative to full length Wnt7a, is readily expressed to high levels, and exhibits improved dispersal in tissue, and bioavailability. This work is significant because it will potentially transform the utility of Wnts for biotherapeutic applications.

Understanding how Wnt signaling controls the growth and repair of bone and muscle tissue is clearly highly relevant to understanding the regenerative processes that occur in patients with degenerative diseases such as osteogenesis imperfecta and muscular dystrophy. Without question, mechanistic studies are providing novel insights into the biology of bone and muscle regeneration and this knowledge is being actively translated towards having clinical impact. Such mechanistic insights are key to the development of new modalities of therapeutic intervention that will include protein biologics but also small drugs that target these pathways.

Recent work has focused increased attention on the mechanisms by which muscle and bone interact to maintain musculoskeletal health [104]. Emerging information suggests that Wnt ligands may mediate some aspects of the interaction between these tissues. For example, Wnt3a secreted from osteocytes may support myogenesis and maintenance of muscle function. Muscle tissue appears to secrete factors that can inhibit osteocyte apoptosis in association with elevated levels of β-catenin [105]. Further evaluation of how Wnt signaling may mediate muscle-bone crosstalk, as well as assessment as to how applicable Wnt signaling mechanisms identified in either myocytes and osteoblasts are to other cell types will provide additional opportunities for insights into this critical signaling pathway.

Highlights.

Canonical Wnt signaling is critical for normal bone and muscle development

Non-canonical Wnt pathways are key to muscle repair processes associated with satellite cells Understanding of these processes will be critical for developing effective therapeutic strategies

Acknowledgements

This work from the laboratory of M.A.R. was supported by grants from the Canadian Institutes for Health Research, the Muscular Dystrophy Association, the National Institutes of Health, the Canadian Stem Cell Network, the Ontario Ministry of Research and Innovation, the Canada Research Chair Program. M.A.R. is a founding scientist with Fate Therapeutics, who are developing Wnt7a as a therapeutic agent. The work from the laboratory of B.O.W. is supported by the Van Andel Research Institute and by the NIH/NIAMS (R01 AR053293). B.O.W. was also the recipient of a research grant from Genentech and is a consultant for Amgen.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Nusse R, Varmus HE. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell. 1982;31:99–109. doi: 10.1016/0092-8674(82)90409-3. [DOI] [PubMed] [Google Scholar]
  • [2].Willert K, Nusse R. Wnt proteins. Cold Spring Harb Perspect Biol. 2012;4:a007864. doi: 10.1101/cshperspect.a007864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Rijsewijk F, Schuermann M, Wagenaar E, Parren P, Weigel D, Nusse R. The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell. 1987;50:649–57. doi: 10.1016/0092-8674(87)90038-9. [DOI] [PubMed] [Google Scholar]
  • [4].Gonsalves FC, DasGupta R. Function of the wingless signaling pathway in Drosophila. Methods Mol Biol. 2008;469:115–25. doi: 10.1007/978-1-60327-469-2_10. [DOI] [PubMed] [Google Scholar]
  • [5].Nusse R, Brown A, Papkoff J, Scambler P, Shackleford G, McMahon A, Moon R, Varmus H. A new nomenclature for int-1 and related genes: the Wnt gene family. Cell. 1991;64:231. doi: 10.1016/0092-8674(91)90633-a. [DOI] [PubMed] [Google Scholar]
  • [6].Ke J, Xu HE, Williams BO. Lipid modification in Wnt structure and function. Curr Opin Lipidol. 2013;24:129–33. doi: 10.1097/MOL.0b013e32835df2bf. [DOI] [PubMed] [Google Scholar]
  • [7].Joiner DM, Ke J, Zhong Z, Xu HE, Williams BO. LRP5 and LRP6 in development and disease. Trends Endocrinol Metab. 2013;24:31–9. doi: 10.1016/j.tem.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149:1192–205. doi: 10.1016/j.cell.2012.05.012. [DOI] [PubMed] [Google Scholar]
  • [9].Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 2003;5:367–77. doi: 10.1016/s1534-5807(03)00266-1. [DOI] [PubMed] [Google Scholar]
  • [10].Singh J, Mlodzik M. Planar cell polarity signaling: coordination of cellular orientation across tissues. Wiley Interdiscip Rev Dev Biol. 2012;1:479–99. doi: 10.1002/wdev.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Gao B, Yang Y. Planar cell polarity in vertebrate limb morphogenesis. Curr Opin Genet Dev. 2013;23:438–44. doi: 10.1016/j.gde.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Kuhl M, Sheldahl LC, Park M, Miller JR, Moon RT. The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet. 2000;16:279–83. doi: 10.1016/s0168-9525(00)02028-x. [DOI] [PubMed] [Google Scholar]
  • [13].Chen AE, Ginty DD, Fan CM. Protein kinase A signalling via CREB controls myogenesis induced by Wnt proteins. Nature. 2005;433:317–22. doi: 10.1038/nature03126. [DOI] [PubMed] [Google Scholar]
  • [14].Inoki K, Ouyang H, Zhu T, Lindvall C, Wang Y, Zhang X, Yang Q, Bennett C, Harada Y, Stankunas K, Wang CY, He X, MacDougald OA, You M, Williams BO, Guan KL. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell. 2006;126:955–68. doi: 10.1016/j.cell.2006.06.055. [DOI] [PubMed] [Google Scholar]
  • [15].von Maltzahn J, Bentzinger CF, Rudnicki MA. Wnt7a-Fzd7 signalling directly activates the Akt/mTOR anabolic growth pathway in skeletal muscle. Nat Cell Biol. 2012;14:186–91. doi: 10.1038/ncb2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Surana R, Sikka S, Cai W, Shin EM, Warrier SR, Tan HJ, Arfuso F, Fox SA, Dharmarajan AM, Kumar AP. Secreted frizzled related proteins: Implications in cancers. Biochim Biophys Acta. 2014;1845:53–65. doi: 10.1016/j.bbcan.2013.11.004. [DOI] [PubMed] [Google Scholar]
  • [17].Niehrs C. Function and biological roles of the Dickkopf family of Wnt modulators. Oncogene. 2006;25:7469–81. doi: 10.1038/sj.onc.1210054. [DOI] [PubMed] [Google Scholar]
  • [18].Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med. 2013;19:179–92. doi: 10.1038/nm.3074. [DOI] [PubMed] [Google Scholar]
  • [19].Chang MK, Kramer I, Huber T, Kinzel B, Guth-Gundel S, Leupin O, Kneissel M. Disruption of Lrp4 function by genetic deletion or pharmacological blockade increases bone mass and serum sclerostin levels. Proc Natl Acad Sci U S A. 2014 doi: 10.1073/pnas.1413828111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Kadowaki T, Wilder E, Klingensmith J, Zachary K, Perrimon N. The segment polarity gene porcupine encodes a putative multitransmembrane protein involved in Wingless processing. Genes Dev. 1996;10:3116–28. doi: 10.1101/gad.10.24.3116. [DOI] [PubMed] [Google Scholar]
  • [21].Takada R, Satomi Y, Kurata T, Ueno N, Norioka S, Kondoh H, Takao T, Takada S. Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Dev Cell. 2006;11:791–801. doi: 10.1016/j.devcel.2006.10.003. [DOI] [PubMed] [Google Scholar]
  • [22].Ching W, Nusse R. A dedicated Wnt secretion factor. Cell. 2006;125:432–3. doi: 10.1016/j.cell.2006.04.018. [DOI] [PubMed] [Google Scholar]
  • [23].Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, Sabatakos G, Apte S, Adkins WN, Allgrove J, Arslan-Kirchner M, Batch JA, Beighton P, Black GC, Boles RG, Boon LM, Borrone C, Brunner HG, Carle GF, Dallapiccola B, De Paepe A, Floege B, Halfhide ML, Hall B, Hennekam RC, Hirose T, Jans A, Juppner H, Kim CA, Keppler-Noreuil K, Kohlschuetter A, LaCombe D, Lambert M, Lemyre E, Letteboer T, Peltonen L, Ramesar RS, Romanengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Sullivan B, Superti-Furga A, Swoboda W, van den Boogaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia T, Baron R, Olsen BR, Warman ML. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–23. doi: 10.1016/s0092-8674(01)00571-2. [DOI] [PubMed] [Google Scholar]
  • [24].Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, Wu D, Insogna K, Lifton RP. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med. 2002;346:1513–21. doi: 10.1056/NEJMoa013444. [DOI] [PubMed] [Google Scholar]
  • [25].Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, Manning SP, Swain PM, Zhao SC, Eustace B, Lappe MM, Spitzer L, Zweier S, Braunschweiger K, Benchekroun Y, Hu X, Adair R, Chee L, FitzGerald MG, Tulig C, Caruso A, Tzellas N, Bawa A, Franklin B, McGuire S, Nogues X, Gong G, Allen KM, Anisowicz A, Morales AJ, Lomedico PT, Recker SM, Van Eerdewegh P, Recker RR, Johnson ML. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet. 2002;70:11–9. doi: 10.1086/338450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Ai M, Holmen SL, Van Hul W, Williams BO, Warman ML. Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone mass-associated missense mutations in LRP5 affect canonical Wnt signaling. Mol Cell Biol. 2005;25:4946–55. doi: 10.1128/MCB.25.12.4946-4955.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Balemans W, Devogelaer JP, Cleiren E, Piters E, Caussin E, Van Hul W. Novel LRP5 missense mutation in a patient with a high bone mass phenotype results in decreased DKK1-mediated inhibition of Wnt signaling. J Bone Miner Res. 2007;22:708–16. doi: 10.1359/jbmr.070211. [DOI] [PubMed] [Google Scholar]
  • [28].Balemans W, Piters E, Cleiren E, Ai M, Van Wesenbeeck L, Warman ML, Van Hul W. The binding between sclerostin and LRP5 is altered by DKK1 and by high-bone mass LRP5 mutations. Calcif Tissue Int. 2008;82:445–53. doi: 10.1007/s00223-008-9130-9. [DOI] [PubMed] [Google Scholar]
  • [29].Semenov MV, He X. LRP5 mutations linked to high bone mass diseases cause reduced LRP5 binding and inhibition by SOST. J Biol Chem. 2006;281:38276–84. doi: 10.1074/jbc.M609509200. [DOI] [PubMed] [Google Scholar]
  • [30].Ellies DL, Viviano B, McCarthy J, Rey JP, Itasaki N, Saunders S, Krumlauf R. Bone density ligand, Sclerostin, directly interacts with LRP5 but not LRP5G171V to modulate Wnt activity. J Bone Miner Res. 2006;21:1738–49. doi: 10.1359/jbmr.060810. [DOI] [PubMed] [Google Scholar]
  • [31].Hsieh JC, Lee L, Zhang L, Wefer S, Brown K, DeRossi C, Wines ME, Rosenquist T, Holdener BC. Mesd encodes an LRP5/6 chaperone essential for specification of mouse embryonic polarity. Cell. 2003;112:355–67. doi: 10.1016/s0092-8674(03)00045-x. [DOI] [PubMed] [Google Scholar]
  • [32].Zhang Y, Wang Y, Li X, Zhang J, Mao J, Li Z, Zheng J, Li L, Harris S, Wu D. The LRP5 high-bone-mass G171V mutation disrupts LRP5 interaction with Mesd. Mol Cell Biol. 2004;24:4677–84. doi: 10.1128/MCB.24.11.4677-4684.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, Key L, Renton T, Bartels C, Gong Y, Warman ML, De Vernejoul MC, Bollerslev J, Van Hul W. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. Am J Hum Genet. 2003;72:763–71. doi: 10.1086/368277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Johnson ML, Summerfield DT. Parameters of LRP5 from a structural and molecular perspective. Crit Rev Eukaryot Gene Expr. 2005;15:229–42. doi: 10.1615/critreveukargeneexpr.v15.i3.50. [DOI] [PubMed] [Google Scholar]
  • [35].Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA, 2nd, Hartmann C, Li L, Hwang TH, Brayton CF, Lang RA, Karsenty G, Chan L. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–14. doi: 10.1083/jcb.200201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Holmen SL, Giambernardi TA, Zylstra CR, Buckner-Berghuis BD, Resau JH, Hess JF, Glatt V, Bouxsein ML, Ai M, Warman ML, Williams BO. Decreased BMD and limb deformities in mice carrying mutations in both Lrp5 and Lrp6. J Bone Miner Res. 2004;19:2033–40. doi: 10.1359/JBMR.040907. [DOI] [PubMed] [Google Scholar]
  • [37].Iwaniec UT, Wronski TJ, Liu J, Rivera MF, Arzaga RR, Hansen G, Brommage R. PTH stimulates bone formation in mice deficient in Lrp5. J Bone Miner Res. 2007;22:394–402. doi: 10.1359/jbmr.061118. [DOI] [PubMed] [Google Scholar]
  • [38].Akhter MP, Wells DJ, Short SJ, Cullen DM, Johnson ML, Haynatzki GR, Babij P, Allen KM, Yaworsky PJ, Bex F, Recker RR. Bone biomechanical properties in LRP5 mutant mice. Bone. 2004;35:162–9. doi: 10.1016/j.bone.2004.02.018. [DOI] [PubMed] [Google Scholar]
  • [39].Glass DA, 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, Karsenty G. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–64. doi: 10.1016/j.devcel.2005.02.017. [DOI] [PubMed] [Google Scholar]
  • [40].Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO. Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem. 2005;280:21162–8. doi: 10.1074/jbc.M501900200. [DOI] [PubMed] [Google Scholar]
  • [41].Kramer I, Halleux C, Keller H, Pegurri M, Gooi JH, Weber PB, Feng JQ, Bonewald LF, Kneissel M. Osteocyte Wnt/beta-catenin signaling is required for normal bone homeostasis. Mol Cell Biol. 2010;30:3071–85. doi: 10.1128/MCB.01428-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T, Kikuchi Y, Takada I, Kato S, Kani S, Nishita M, Marumo K, Martin TJ, Minami Y, Takahashi N. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med. 2012;18:405–12. doi: 10.1038/nm.2653. [DOI] [PubMed] [Google Scholar]
  • [43].Wei W, Zeve D, Suh JM, Wang X, Du Y, Zerwekh JE, Dechow PC, Graff JM, Wan Y. Biphasic and dosage-dependent regulation of osteoclastogenesis by beta-catenin. Mol Cell Biol. 2011;31:4706–19. doi: 10.1128/MCB.05980-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Riddle RC, Diegel CR, Leslie JM, Van Koevering KK, Faugere MC, Clemens TL, Williams BO. Lrp5 and Lrp6 exert overlapping functions in osteoblasts during postnatal bone acquisition. PLoS One. 2013;8:e63323. doi: 10.1371/journal.pone.0063323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Yadav VK, Ryu JH, Suda N, Tanaka KF, Gingrich JA, Schutz G, Glorieux FH, Chiang CY, Zajac JD, Insogna KL, Mann JJ, Hen R, Ducy P, Karsenty G. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell. 2008;135:825–37. doi: 10.1016/j.cell.2008.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Cui Y, Niziolek PJ, MacDonald BT, Alenina N, Matthes S, Jacobsen CM, Conlon RA, Brommage R, Powell DR, He X, Bader M, Williams BO, Warman ML, Robling AG. Reply to Lrp5 regulation of bone mass and gut serotonin synthesis. Nat Med. 2014;20:1229–30. doi: 10.1038/nm.3697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Kode A, Obri A, Paone R, Kousteni S, Ducy P, Karsenty G. Lrp5 regulation of bone mass and serotonin synthesis in the gut. Nat Med. 2014;20:1228–9. doi: 10.1038/nm.3698. [DOI] [PubMed] [Google Scholar]
  • [48].Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–50. doi: 10.1016/j.devcel.2005.03.016. [DOI] [PubMed] [Google Scholar]
  • [49].Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell. 2005;8:727–38. doi: 10.1016/j.devcel.2005.02.013. [DOI] [PubMed] [Google Scholar]
  • [50].Hu H, Hilton MJ, Tu X, Yu K, Ornitz DM, Long F. Sequential roles of Hedgehog and Wnt signaling in osteoblast development. Development. 2005;132:49–60. doi: 10.1242/dev.01564. [DOI] [PubMed] [Google Scholar]
  • [51].Joeng KS, Schumacher CA, Zylstra-Diegel CR, Long F, Williams BO. Lrp5 and Lrp6 redundantly control skeletal development in the mouse embryo. Dev Biol. 2011;359:222–9. doi: 10.1016/j.ydbio.2011.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, Dioszegi M, Dikkers FG, Hildering P, Willems PJ, Verheij JB, Lindpaintner K, Vickery B, Foernzler D, Van Hul W. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002;39:91–7. doi: 10.1136/jmg.39.2.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Staehling-Hampton K, Proll S, Paeper BW, Zhao L, Charmley P, Brown A, Gardner JC, Galas D, Schatzman RC, Beighton P, Papapoulos S, Hamersma H, Brunkow ME. A 52-kb deletion in the SOST-MEOX1 intergenic region on 17q12-q21 is associated with van Buchem disease in the Dutch population. Am J Med Genet. 2002;110:144–52. doi: 10.1002/ajmg.10401. [DOI] [PubMed] [Google Scholar]
  • [54].MacDonald BT, Joiner DM, Oyserman SM, Sharma P, Goldstein SA, He X, Hauschka PV. Bone mass is inversely proportional to Dkk1 levels in mice. Bone. 2007;41:331–9. doi: 10.1016/j.bone.2007.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Rivadeneira F, Styrkarsdottir U, Estrada K, Halldorsson BV, Hsu YH, Richards JB, Zillikens MC, Kavvoura FK, Amin N, Aulchenko YS, Cupples LA, Deloukas P, Demissie S, Grundberg E, Hofman A, Kong A, Karasik D, van Meurs JB, Oostra B, Pastinen T, Pols HA, Sigurdsson G, Soranzo N, Thorleifsson G, Thorsteinsdottir U, Williams FM, Wilson SG, Zhou Y, Ralston SH, van Duijn CM, Spector T, Kiel DP, Stefansson K, Ioannidis JP, Uitterlinden AG. Twenty bone-mineral-density loci identified by large-scale meta-analysis of genome-wide association studies. Nat Genet. 2009;41:1199–206. doi: 10.1038/ng.446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Wan Y, Lu C, Cao J, Zhou R, Yao Y, Yu J, Zhang L, Zhao H, Li H, Zhao J, Zhu X, He L, Liu Y, Yao Z, Yang X, Guo X. Osteoblastic Wnts differentially regulate bone remodeling and the maintenance of bone marrow mesenchymal stem cells. Bone. 2013;55:258–67. doi: 10.1016/j.bone.2012.12.052. [DOI] [PubMed] [Google Scholar]
  • [57].Maruyama T, Jiang M, Hsu W. Gpr177, a novel locus for bone mineral density and osteoporosis, regulates osteogenesis and chondrogenesis in skeletal development. J Bone Miner Res. 2013;28:1150–9. doi: 10.1002/jbmr.1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Zhong Z, Zylstra-Diegel CR, Schumacher CA, Baker JJ, Carpenter AC, Rao S, Yao W, Guan M, Helms JA, Lane NE, Lang RA, Williams BO. Wntless functions in mature osteoblasts to regulate bone mass. Proc Natl Acad Sci U S A. 2012;109:E2197–204. doi: 10.1073/pnas.1120407109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Laine CM, Joeng KS, Campeau PM, Kiviranta R, Tarkkonen K, Grover M, Lu JT, Pekkinen M, Wessman M, Heino TJ, Nieminen-Pihala V, Aronen M, Laine T, Kroger H, Cole WG, Lehesjoki AE, Nevarez L, Krakow D, Curry CJ, Cohn DH, Gibbs RA, Lee BH, Makitie O. WNT1 mutations in early-onset osteoporosis and osteogenesis imperfecta. N Engl J Med. 2013;368:1809–16. doi: 10.1056/NEJMoa1215458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Keupp K, Beleggia F, Kayserili H, Barnes AM, Steiner M, Semler O, Fischer B, Yigit G, Janda CY, Becker J, Breer S, Altunoglu U, Grunhagen J, Krawitz P, Hecht J, Schinke T, Makareeva E, Lausch E, Cankaya T, Caparros-Martin JA, Lapunzina P, Temtamy S, Aglan M, Zabel B, Eysel P, Koerber F, Leikin S, Garcia KC, Netzer C, Schonau E, Ruiz-Perez VL, Mundlos S, Amling M, Kornak U, Marini J, Wollnik B. Mutations in WNT1 cause different forms of bone fragility. Am J Hum Genet. 2013;92:565–74. doi: 10.1016/j.ajhg.2013.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Fahiminiya S, Majewski J, Mort J, Moffatt P, Glorieux FH, Rauch F. Mutations in WNT1 are a cause of osteogenesis imperfecta. J Med Genet. 2013;50:345–8. doi: 10.1136/jmedgenet-2013-101567. [DOI] [PubMed] [Google Scholar]
  • [62].Heilmann A, Schinke T, Bindl R, Wehner T, Rapp A, Haffner-Luntzer M, Nemitz C, Liedert A, Amling M, Ignatius A. The Wnt serpentine receptor Frizzled-9 regulates new bone formation in fracture healing. PLoS One. 2013;8:e84232. doi: 10.1371/journal.pone.0084232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Albers J, Schulze J, Beil FT, Gebauer M, Baranowsky A, Keller J, Marshall RP, Wintges K, Friedrich FW, Priemel M, Schilling AF, Rueger JM, Cornils K, Fehse B, Streichert T, Sauter G, Jakob F, Insogna KL, Pober B, Knobeloch KP, Francke U, Amling M, Schinke T. Control of bone formation by the serpentine receptor Frizzled-9. J Cell Biol. 2011;192:1057–72. doi: 10.1083/jcb.201008012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Tu X, Joeng KS, Nakayama KI, Nakayama K, Rajagopal J, Carroll TJ, McMahon AP, Long F. Noncanonical Wnt signaling through G protein-linked PKCdelta activation promotes bone formation. Dev Cell. 2007;12:113–27. doi: 10.1016/j.devcel.2006.11.00. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Buckingham M, Vincent SD. Distinct and dynamic myogenic populations in the vertebrate embryo. Curr Opin Genet Dev. 2009;19:444–53. doi: 10.1016/j.gde.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • [66].Cossu G, Borello U. Wnt signaling and the activation of myogenesis in mammals. EMBO J. 1999;18:6867–72. doi: 10.1093/emboj/18.24.6867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].von Maltzahn J, Chang NC, Bentzinger CF, Rudnicki MA. Wnt signaling in myogenesis. Trends Cell Biol. 2012;22:602–9. doi: 10.1016/j.tcb.2012.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Abu-Elmagd M, Robson L, Sweetman D, Hadley J, Francis-West P, Munsterberg A. Wnt/Lef1 signaling acts via Pitx2 to regulate somite myogenesis. Dev Biol. 2010;337:211–9. doi: 10.1016/j.ydbio.2009.10.023. [DOI] [PubMed] [Google Scholar]
  • [69].Tajbakhsh S, Borello U, Vivarelli E, Kelly R, Papkoff J, Duprez D, Buckingham M, Cossu G. Differential activation of Myf5 and MyoD by different Wnts in explants of mouse paraxial mesoderm and the later activation of myogenesis in the absence of Myf5. Development. 1998;125:4155–62. doi: 10.1242/dev.125.21.4155. [DOI] [PubMed] [Google Scholar]
  • [70].Borello U, Berarducci B, Murphy P, Bajard L, Buffa V, Piccolo S, Buckingham M, Cossu G. The Wnt/beta-catenin pathway regulates Gli-mediated Myf5 expression during somitogenesis. Development. 2006;133:3723–32. doi: 10.1242/dev.02517. [DOI] [PubMed] [Google Scholar]
  • [71].Ikeya M, Takada S. Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome. Development. 1998;125:4969–76. doi: 10.1242/dev.125.24.4969. [DOI] [PubMed] [Google Scholar]
  • [72].Otto A, Schmidt C, Patel K. Pax3 and Pax7 expression and regulation in the avian embryo. Anat Embryol (Berl) 2006;211:293–310. doi: 10.1007/s00429-006-0083-3. [DOI] [PubMed] [Google Scholar]
  • [73].Aoki K, Taketo MM. Tissue-specific transgenic, conditional knockout and knock-in mice of genes in the canonical Wnt signaling pathway. Methods Mol Biol. 2008;468:307–31. doi: 10.1007/978-1-59745-249-6_24. [DOI] [PubMed] [Google Scholar]
  • [74].van Amerongen R, Berns A. Knockout mouse models to study Wnt signal transduction. Trends Genet. 2006;22:678–89. doi: 10.1016/j.tig.2006.10.001. [DOI] [PubMed] [Google Scholar]
  • [75].Otto A, Schmidt C, Luke G, Allen S, Valasek P, Muntoni F, Lawrence-Watt D, Patel K. Canonical Wnt signalling induces satellite-cell proliferation during adult skeletal muscle regeneration. J Cell Sci. 2008;121:2939–50. doi: 10.1242/jcs.026534. [DOI] [PubMed] [Google Scholar]
  • [76].Murphy MM, Keefe AC, Lawson JA, Flygare SD, Yandell M, Kardon G. Transiently active Wnt/beta-catenin signaling is not required but must be silenced for stem cell function during muscle regeneration. Stem Cell Reports. 2014;3:475–88. doi: 10.1016/j.stemcr.2014.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007;317:807–10. doi: 10.1126/science.1144090. [DOI] [PubMed] [Google Scholar]
  • [78].Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles. Physiol Rev. 2011;91:1447–531. doi: 10.1152/physrev.00031.2010. [DOI] [PubMed] [Google Scholar]
  • [79].Anakwe K, Robson L, Hadley J, Buxton P, Church V, Allen S, Hartmann C, Harfe B, Nohno T, Brown AM, Evans DJ, Francis-West P. Wnt signalling regulates myogenic differentiation in the developing avian wing. Development. 2003;130:3503–14. doi: 10.1242/dev.00538. [DOI] [PubMed] [Google Scholar]
  • [80].Takata H, Terada K, Oka H, Sunada Y, Moriguchi T, Nohno T. Involvement of Wnt4 signaling during myogenic proliferation and differentiation of skeletal muscle. Dev Dyn. 2007;236:2800–7. doi: 10.1002/dvdy.21327. [DOI] [PubMed] [Google Scholar]
  • [81].Tee JM, van Rooijen C, Boonen R, Zivkovic D. Regulation of slow and fast muscle myofibrillogenesis by Wnt/beta-catenin and myostatin signaling. PLoS One. 2009;4:e5880. doi: 10.1371/journal.pone.0005880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Hutcheson DA, Zhao J, Merrell A, Haldar M, Kardon G. Embryonic and fetal limb myogenic cells are derived from developmentally distinct progenitors and have different requirements for beta-catenin. Genes Dev. 2009;23:997–1013. doi: 10.1101/gad.1769009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84:209–38. doi: 10.1152/physrev.00019.2003. [DOI] [PubMed] [Google Scholar]
  • [84].Kuang S, Kuroda K, Le Grand F, Rudnicki MA. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell. 2007;129:999–1010. doi: 10.1016/j.cell.2007.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Wang YX, Rudnicki MA. Satellite cells, the engines of muscle repair. Nat Rev Mol Cell Biol. 2011;13:127–33. doi: 10.1038/nrm3265. [DOI] [PubMed] [Google Scholar]
  • [86].Le Grand F, Jones AE, Seale V, Scime A, Rudnicki MA. Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell. 2009;4:535–47. doi: 10.1016/j.stem.2009.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Bentzinger CF, Wang YX, von Maltzahn J, Soleimani VD, Yin H, Rudnicki MA. Fibronectin regulates Wnt7a signaling and satellite cell expansion. Cell Stem Cell. 2013;12:75–87. doi: 10.1016/j.stem.2012.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Bentzinger CF, von Maltzahn J, Dumont NA, Stark DA, Wang YX, Nhan K, Frenette J, Cornelison D, Rudnicki MA. Wnt7a stimulates myogenic stem cell motility and engraftment resulting in improved muscle strength. J Cell Biol. 2014 doi: 10.1083/jcb.201310035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].von Maltzahn J, Renaud JM, Parise G, Rudnicki MA. Wnt7a treatment ameliorates muscular dystrophy. Proc Natl Acad Sci U S A. 2012;109:20614–9. doi: 10.1073/pnas.1215765109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Emery AE. The muscular dystrophies. Bmj. 1998;317:991–5. doi: 10.1136/bmj.317.7164.991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Rey JP, Ellies DL. Wnt modulators in the biotech pipeline. Dev Dyn. 2010;239:102–14. doi: 10.1002/dvdy.22181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Lum L, Clevers H. Cell biology. The unusual case of Porcupine. Science. 2012;337:922–3. doi: 10.1126/science.1228179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Mason JJ, Williams BO. SOST and DKK: Antagonists of LRP Family Signaling as Targets for Treating Bone Disease. J Osteoporos. 2010:2010. doi: 10.4061/2010/460120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Gong Y, Bourhis E, Chiu C, Stawicki S, DeAlmeida VI, Liu BY, Phamluong K, Cao TC, Carano RA, Ernst JA, Solloway M, Rubinfeld B, Hannoush RN, Wu Y, Polakis P, Costa M. Wnt isoform-specific interactions with coreceptor specify inhibition or potentiation of signaling by LRP6 antibodies. PLoS One. 2010;5:e12682. doi: 10.1371/journal.pone.0012682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Ettenberg SA, Charlat O, Daley MP, Liu S, Vincent KJ, Stuart DD, Schuller AG, Yuan J, Ospina B, Green J, Yu Q, Walsh R, Li S, Schmitz R, Heine H, Bilic S, Ostrom L, Mosher R, Hartlepp KF, Zhu Z, Fawell S, Yao YM, Stover D, Finan PM, Porter JA, Sellers WR, Klagge IM, Cong F. Inhibition of tumorigenesis driven by different Wnt proteins requires blockade of distinct ligand-binding regions by LRP6 antibodies. Proc Natl Acad Sci U S A. 2010;107:15473–8. doi: 10.1073/pnas.1007428107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Minear S, Leucht P, Jiang J, Liu B, Zeng A, Fuerer C, Nusse R, Helms JA. Wnt proteins promote bone regeneration. Sci Transl Med. 2010;2 doi: 10.1126/scitranslmed.3000231. 29ra30. [DOI] [PubMed] [Google Scholar]
  • [97].Zhao L, Rooker SM, Morrell N, Leucht P, Simanovskii D, Helms JA. Controlling the in vivo activity of Wnt liposomes. Methods Enzymol. 2009;465:331–47. doi: 10.1016/S0076-6879(09)65017-5. [DOI] [PubMed] [Google Scholar]
  • [98].Morrell NT, Leucht P, Zhao L, Kim JB, ten Berge D, Ponnusamy K, Carre AL, Dudek H, Zachlederova M, McElhaney M, Brunton S, Gunzner J, Callow M, Polakis P, Costa M, Zhang XM, Helms JA, Nusse R. Liposomal packaging generates Wnt protein with in vivo biological activity. PLoS One. 2008;3:e2930. doi: 10.1371/journal.pone.0002930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Hedgepeth CM, Conrad LJ, Zhang J, Huang HC, Lee VM, Klein PS. Activation of the Wnt signaling pathway: a molecular mechanism for lithium action. Dev Biol. 1997;185:82–91. doi: 10.1006/dbio.1997.8552. [DOI] [PubMed] [Google Scholar]
  • [100].Chen Y, Alman BA. Wnt pathway, an essential role in bone regeneration. J Cell Biochem. 2009;106:353–62. doi: 10.1002/jcb.22020. [DOI] [PubMed] [Google Scholar]
  • [101].Silkstone D, Hong H, Alman BA. Beta-catenin in the race to fracture repair: in it to Wnt. Nat Clin Pract Rheumatol. 2008;4:413–9. doi: 10.1038/ncprheum0838. [DOI] [PubMed] [Google Scholar]
  • [102].Janda CY, Waghray D, Levin AM, Thomas C, Garcia KC. Structural basis of Wnt recognition by Frizzled. Science. 2012;337:59–64. doi: 10.1126/science.1222879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].von Maltzahn J, Zinoviev R, Chang NC, Bentzinger CF, Rudnicki MA. A truncated Wnt7a retains full biological activity in skeletal muscle. Nat Commun. 2013;4:2869. doi: 10.1038/ncomms3869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Bonewald LF, Kiel DP, Clemens TL, Esser K, Orwoll ES, O'Keefe RJ, Fielding RA. Forum on bone and skeletal muscle interactions: summary of the proceedings of an ASBMR workshop. J Bone Miner Res. 2013;28:1857–65. doi: 10.1002/jbmr.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Jahn K, Lara-Castillo N, Brotto L, Mo CL, Johnson ML, Brotto M, Bonewald LF. Skeletal muscle secreted factors prevent glucocorticoid-induced osteocyte apoptosis through activation of beta-catenin. Eur Cell Mater. 2012;24:197–209. doi: 10.22203/ecm.v024a14. discussion 209-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Bentzinger CF, von Maltzahn J, Dumont NA, Stark DA, Wang YX, Nhan K, Frenette J, Cornelison DD, Rudnicki MA. Wnt7a stimulates myogenic stem cell motility and engraftment resulting in improved muscle strength. J Cell Biol. 2014;205:97–111. doi: 10.1083/jcb.201310035. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES