Abstract
The balance between bone formation and bone resorption controls postnatal bone homeostasis. Research over the last decade has provided a vast amount of evidence that WNT signaling plays a pivotal role in regulating this balance. Therefore, understanding how the WNT signaling pathway regulates skeletal development and homeostasis is of great value for human skeletal health and disease.
1. Overview of Bone Homeostasis
The balance of two activities maintains bone homeostasis: bone formation by mesenchymal stem cell (MSC)-derived osteoblasts, and bone resorption by hematopoietic stem cell (HSC)-derived osteoclasts. Under pathologic conditions, this balance is disrupted. High osteoclast activity or low osteoblast activity leads to low bone mass (osteopenia), while low osteoclast activity or high osteoblast activity leads to high bone mass (osteopetrosis). Several signaling pathways are important for maintaining this delicate balance including those activated by WNT, BMP, PTH/PTHrP, Notch, and Hedgehog. In this review, we focus on the role of Wnt signaling in this process. We will first briefly describe the cellular mechanisms of Wnt signal transduction and then discuss the observations associated with alterations of this pathway in human skeletal disease that focused attention on this pathway. This will be followed by the discussion of studies utilizing genetically engineered mouse models to gain insights into the specific requirements for β-catenin, WNT ligands, and WNT receptors in osteoblast differentiation and function. We conclude with a discussion of the current status of therapeutic development of WNT-based strategies to treat human diseases associated with low bone mass.
2. Overview of Bone Development
Bone is formed by either intramembranous ossification or endochondral ossification (Figure 1). During intramembranous ossification, mesenchymal progenitors differentiate directly into osteoblasts 1. This process underlies the formation of cranial bones, a portion of the mandible, and the clavicle. Intramembranous ossification is initiated by a condensation of mesenchymal stem cells. As the condensation enlarges, osteoblast differentiation occurs within the interior and osteoid matrix is secreted. The matrix is then mineralized, encasing osteoblasts within the matrix to form osteocytes. Osteoblasts also then line the bone underneath the periosteum. This matrix is then vascularized to support long term cell survival.
Figure 1. Overview of Bone Formation.
During intramembranous ossification, new bone forms directly from a condensation of mesenchymal stem cells. In contrast, endochondral ossification occurs when a cartilaginous template is replaced by mineralized osteoid. In both cases, osteoblasts become encased in mineralized matrix creating osteocytes. Please refer to the text for more detail.
The key difference between intramembranous ossification and endochondral ossification is that in the latter bone forms as a result of replacement of a previously deposited cartilaginous template 2, 3. This is the process by which the long bones of the body form. After formation of the template (composed of hyaline cartilage), chondrocytes in the area that will become the primary ossification center begin secreting alkaline phosphatase which promotes calcification of the cartilaginous matrix and then undergo apoptosis 4. The bone collar then forms in the mid-portion of a future long bone via a process similar to intramembranous ossification. The bone collar then facilitates the penetration of blood vessels, providing a path for the introduction of pre-osteoblasts into the area which then begin depositing osteoid and subsequently mineralize the matrix in the primary ossification center. In addition, the distal ends of each long bone (epiphyses) form secondary ossification centers by a process similar to that outlined for the primary ossification center. The cartilage that remains between the primary and secondary ossification centers is referred to as the epiphyseal plate and continues to deposit new cartilage matrix to serve as a scaffold for additional endochondral ossification allowing the bone to grow in length. The hyaline cartilage that remains on the distal surface of the bone is referred to as articular cartilage.
3. WNT/β-catenin Signaling
Wnts are a family of 19 secreted glycolipoproteins in mammals and can broadly be classified as signaling through β-Catenin-dependent (canonical WNT signaling) or β-Catenin-independent (noncanonical WNT signaling) pathways. WNT pathways play fundamentally important roles in regulating cell fate decision, cell polarity, and cell proliferation; and alterations in components of this pathway are among the most common events associated with human disease 5.
The first mammalian Wnt gene, Int1 (later referred as Wnt1), was identified in 1982 as an insertion target of mouse mammary tumor virus (MMTV) associated with the formation of mammary gland tumors 6. Shortly thereafter, the Drosophila Wingless gene was recognized as a close homolog of Int1 7. The extensive characterization of the genetic requirements for proper specification of Drosophila embryonic segment polarity allowed for identification of downstream components of this pathway. In 1991, a new nomenclature was proposed to rename the original Int1 as Wnt1 (a combination of Wingless and Int1) and served as the basis for the naming of all other genes in this family 8.
Decades of work have defined the components of the WNT/β-catenin pathway 9, which is summarized briefly here (Figure 2). In the absence of Wnt, glycogen synthase kinase-3 (GSK-3) phosphorylates β-catenin, marking it for ubiquitin-dependent proteolysis. When Wnt binds to a member of the Frizzled family of seven-transmembrane receptors and either LRP5 or LRP6 10, 11, GSK-3 activity is down-regulated. Inactivation of GSK-3 increases β-catenin in the cytosol and nucleus, allowing β-catenin to interact with TCFs and LEF 12. These complexes modulate the transcriptional activity of target promoters 12. Frizzled-related proteins (sFRPs), as well as other secreted inhibitors like Dickkopf (DKK) and Sclerostin (SOST), regulate signaling at the level of the Wnt/Frizzled/Lrp interaction 13–19, while many proteins including Axin/Axin2 and APC control the pathway intracellularly 11, 12. Non-canonical pathways induced by Wnt ligands include those that activate Protein kinase C, Protein kinase A, mTOR, PI3K/AKT, and the planar cell polarity pathway which modulates cytoskeletal structure via regulation of small GTPases such as RHOB. These pathways can be induced by interaction of WNTs with Frizzled-receptors, putatively without the co-engagement of LRP5/6, but can also involve alterative receptors such as the receptor tyrosine kinase, ROR2 20.
Figure 2. Canonical Wnt Signaling Pathway.
In the absence of an upstream ligand, receptor Frizzled (FZD) and co-receptor LRP5/6 are inactive (1). The cytoplasmic β-catenin will be recruited into a “destruction complex” consisting of Axin, APC, casein kinase 1a (CK1) and glycogen synthase kinase 3 (GSK3) (2). This “destruction complex” facilitates the phosphorylation of β-catenin by GSK3 and subsequent ubiquitinylation (Ub) by β-TrCP, an E3 ligase, targeting β-catenin for proteasome-mediated degradation. In this context, the transcription repressor Groucho occupies T-Cell Factor/Lymphoid enhancer factor (TCF/LEF) DNA binding sites (7). This inactive state could also be caused by lack of co-receptors availability due to effectors (DKK1/WISE/SOST) binding to LRP5/6 and preventing its association with Wnt (3), or by other effectors associating with Wnts and blocking their ability to interact with the co-receptor complex (4). When a Wnt ligand engages the receptor complex (5), the C-terminus of LRP5/6 is phosphorylated, creating a binding site for Axin, resulting in inhibition of the destruction complex (6). This inhibits GSK3 activity, allowing cytoplasmic levels of β-catenin to increase (8). β-catenin subsequently translocates into the nucleus and complexes with LEF/TCF proteins and other co-factors to activate transcription of target genes (9).
4. Wnt/β-catenin Signaling in Human Skeletal Diseases
Wnt signaling was first linked to skeletal development in 1994, when Wnt3a-deficient embryos were found to exhibit axial defects 21. However, the identification of loss-of-function mutations in LRP5 in human osteoporosis pseudoglioma (OPPG), a rare syndrome associated with dramatic reductions in bone mass, intensified interest in this area 22. Shortly after this, two groups independently reported that point mutations in LRP5, which prevent Sclerostin (SOST)- and Dickkopf (DKK1-mediated) binding and inhibition, caused high bone mass in two different families 23, 24. Subsequent studies have identified numerous mutations in in LRP5 to be associated with alterations in human bone mass 25, 26. In addition, loss-of-function mutations in the highly related LRP6 gene were also associated with osteoporosis in humans 27. Finally, loss of function mutations in LRP4, an antagonist of Wnt/β-catenin signaling which shares homology with LRP5/6, were identified as causes of sclerosteosis in human patients 28.
Additional insight into the role of WNT signaling in human disease was obtained from studies of two related human conditions associated with high bone mass. Sclerostin (SOST), a Wnt antagonist, was found to be causally dysregulated in Sclerosteosis and Van Buchem’s Disease, rare high-bone-mass genetic disorders 29–32. SOST is a Wnt/β-catenin signaling antagonist which binds LRP5 and LRP6 to prevent their association with Wnt ligands 33. Recent work has definitively shown that interactions of SOST with both LRP5 and LRP6 are necessary for full activity 34, 35. Given that SOST is primarily expressed in osteocytes 33, inhibiting SOST to prevent it from normally downregulating LRP5 and LRP6 to treat osteoporosis may cause less unintended side effects 31, 36 because blocking SOST function would not be expected to affect regulation of any tissues besides the skeleton.
Several other examples of Wnt pathway-associated mutations causing skeletal phenotypes in humans have emerged. For example, heterozygous nonsense mutation in AXIN2 that leads to Wnt signaling activation causes both severe permanent tooth agenesis and colorectal neoplasia 37, and a homozygous WNT3 loss-of-function mutation causes defects in limb formation and craniofacial and urogenital development 38. Finally, several high-profile recent reports have linked mutations in the WNT1 gene to Osteogenesis Imperfecta 39–42, a disease characterized by the presence of brittle bone which is easily fractured.
5. Mouse Models to Assess the Role of β-catenin in the Osteoblast Lineage
Mouse models using different cell-specific promoters to manipulate Wnt components have been generated and characterized 43. We focus on insights generated by assessment of genetically engineered mice with alterations in β-catenin (encoded by the Ctnnb1 gene), WNT ligands, and WNT receptors.
5.1 β-catenin
Ectopic activation of β-catenin in osteochondral progenitors at a stage before full commitment to either the chondrocyte or osteoblast lineage promotes osteoblast differentiation and inhibits chondrogenesis 44–46, while conditional inactivation of Wnt/β-catenin signaling in progenitor cells has the opposite effect 44, 47–49. The developing bones with WNT/β-catenin signaling inactivation were capable of expressing early markers of the osteoblast lineage, but failed to express markers of osteoblast commitment.
Loss of β-catenin in mature osteoblasts and/or osteocytes causes severe osteopenia associated with increased osteoclastogenesis, whereas constitutive activation of β-catenin within osteoblasts impairs osteoclast formation 50, 51. Osteoblasts and osteocytes secrete two proteins, macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL), required for osteoclast differentiation and maturation 52 (Figure 3). RANKL stimulates osteoclast maturation through interacting with its receptor, RANK, expressed at the surface of osteoclast precursors. The RANKL-RANK interaction is also regulated by another secreted glycoprotein, osteoprotegerin (OPG), which can also be derived from osteoblasts/osteocytes 53. OPG is a soluble TNFα receptor that acts as a decoy receptor for RANKL, and it inhibits osteoclast maturation and protects bone from both normal osteoclast remodeling and ovariectomy-associated bone loss 54. Importantly, elevated OPG levels were associated with ectopic activation of β-catenin within osteoblasts 50, 51 and OPG was shown to be direct transcriptional target of β-catenin 51, consistent with the elevated osteoclastogenesis seen in mouse models with loss of β-catenin signaling in osteoblasts (Figure 3).
Figure 3. Regulation of RANKL Signaling.
The Receptor activator of nuclear factor κβ (RANK) is expressed on osteoclasts and upon binding from its cognate ligand, RANKL, stimulates osteoclast differentiation and activation. RANKL is expressed by cells of the osteoblast lineage, which also express another protein referred to as Osteoprotegerin (OPG). OPG binds to RANKL, prevents interaction between RANKL with RANK with the end result being inhibition of osteoclast differentiation and activation. OPG expression can also be activation at the transcriptional level via the binding of β-catenin/TCF complexes to the OPG promoter.
Importantly, understanding the mechanisms of OPG/RANKL interactions has facilitated the development of a novel treatment for osteoporosis, denosumab, which has demonstrated efficacy in increasing bone mass in human clinical trials 55, 56.
5.2 WNT Ligands in Bone
WNT requires a lipid modification to facilitate secretion and function, which complicated their purification and structural analysis 57. This lipid modification, the addition of a palmitoleic acid to a serine residue, is mediated by the ER-resident protein, Porcupine (encoded by the PORCN gene), a member of the membrane-bound O-acyltransferase family 58. Lipid-modified Wnt is then recognized by another ER-resident protein, Wntless/GPR177, which is required to escort Wnt ligands to the cell surface for secretion 58. Further confirmation of the importance of lipid modification in Wnt function was provided when the crystal structure of the Xenopus Wnt8 in complex with cysteine-rich domain (CRD) of mouse FZD8 was reported revealing that the palmitoleic acid binds to specific pocket on the surface of the Frizzled protein 59.
Several groups, including our own, have taken advantage of the pivotal role of Porcupine or GPR177 to all WNTs, to address questions related to the requirement for Wnt family ligands derived from a single cell type within bone to mediate bone development and/or homeostasis. Using this approach, mature osteoblasts were shown to be a critical source of Wnt ligands for postnatal skeletal maintenance 60, 61 Furthermore, when Gpr177 was deleted in both osteoblasts and chondrocytes, cartilage development was affected and a more severe skeletal phenotype was observed 62, 63. Much work is still needed to elucidate the role of Wnts from different tissues, and finding a way to distinguish the consequence from inhibiting the effects of canonical or noncanonical Wnt would be invaluable.
5.2a Canonical Wnts in bone
Mice homozygous for germline deficient mutations of several Wnt genes exhibit skeletal phenotypes (reviewed in 64). One such example is that Wnt10b-deficient mice have slightly increased trabecular bone at a young age, but progressively lose bone as they age. This phenotype in Wnt10b-null mice is presumably due to progressive reduction of osteochondral progenitors 65, 66. Consistent with a role for Wnt10b in bone homeostasis, transgenic mice ectopically expressing Wnt10b in brown adipose and bone marrow showed increased bone mass 65. The recent finding that mutations in WNT1 were causally associated with some types of Osteogenesis Imperfecta also emphasizes the importance of Wnt proteins in normal bone development and homeostasis 39–42, 67. In addition, WNT16 was also found to influence bone mineral density, and specifically cortical bone thickness, but more work needs to be done to evaluate which downstream pathways mediate these effects 68, 69.
5.2b Noncanonical WNTs in bone
Some WNTs preferentially signal through β-catenin-independent mechanisms including Ca2+ flux, Protein kinase C activation, and planar cell polarity (PCP) pathway activation 70. Among noncanonical WNTs, WNT5A is the most extensively studied and plays a critical role in embryonic bone development 71. WNT5A binds to the extracellular cysteine-rich domain (CRD) of ROR2 (Receptor tyrosine kinase-like orphan receptor 2) 72. A close functional relationship between ROR2 and WNT5A is consistent with the fact that mice deficient for either gene develop similar phenotypes including dwarfism, facial abnormalities, and shortened limbs and tails, 73. Recently, osteoblast-derived Wnt5a was shown to enhance osteoclastogenesis 74 by engaging ROR2 on the surface of osteoclast precursors to activate Jun N-terminal kinase (JNK) which then increased Receptor activator of nuclear factor κβ (RANK) expression. However, WNT5A may exert its effects in cell-context dependent manner. For example, a reduction in WNT5A expression was correlated with tail defects and spinal bifida associated with decreased cell proliferation within the ventral caudal region 75. A role for another noncanonical Wnt pathway required for bone formation, activated by WNT7B has also been demonstrated 76, 77. A link between WNT ligands, LRP receptors, and activation of metabolic pathways has also been established and will undoubtedly be the subject of intense study in upcoming years 77, 78.
5.3 Wnt Co-receptors in Bone
5.3a. LRP5/6
The finding that both loss-of-function and gain-of-function mutations in LRP5 and LRP6 alter bone homeostasis in humans stimulated studies on the role of Wnt signaling in this process. Global inactivation of Lrp5 in mice recapitulates the phenotypes seen in human OPPG patients 79–83, and germline mutations in Lrp6 in mice also decrease bone mass 79, 84, 85. Several recent studies demonstrate that osteoblast- or osteocyte-specific alterations of Lrp5 and/or Lrp6 result in significant changes in bone mass 86, 87.
It is important to note that an alternative model postulates that LRP5 functions through regulating serotonin secretion from the gut to affect osteoblast activity 88, 89. In this model, loss of LRP5 in the enterochromaffin cells of the intestine leads to elevated production and secretin of serotonin. Increased levels of serum serotonin then function via binding to the HTR1B receptor to inhibit osteoblast proliferation. However, data from several other laboratories is not consistent with this model 87, 90. The reasons for these discrepancies are currently unclear.
In terms of earlier stages of osteoblast differentiation, inactivation of both Lrp5 and Lrp6 in mouse osteochondral progenitors results in a failure in differentiation 91, phenocopying what is seen in embryos in which β-catenin (Ctnnb1) is conditionally deleted in the same lineage 44, 47–49.
5.3b Other LDL Receptor Family Members
Several other members of the LDL receptor family (Figure 4) have putative roles in skeletal development and bone homeostasis. For example, loss-of-function mutations in LRP4 are found in patients with bone overgrowth and skeletal abnormalities 92–95. LRP4 contains an extracellular motif that interacts with SOST supporting a model by which LRP4 is required to facilitate the activities of SOST 92, 93, 96. The interactions of LRP4 with the Wise protein may also contribute to the observed phenotypes 97. LRP8, also known as Apolipoprotein E Receptor 2 (APOER2), may also play a role in maintaining normal bone as siRNA-mediated knockdown of LRP8 in C2C12 mybolast cells or KS483 pre-osteoblast cells reduced β-catenin-induced transcriptional reporter activity and interfered with osteoblast differentiation in vitro. Finally, there are also reports that the LRP1 protein can modulate the activity of the WNT pathway, although further validation of the importance of this interaction in skeletal development is required 98, 99.
Figure 4. Low-density Lipoprotein (LDL) Receptor Family Members with Putative Roles in Bone Development.
The Low-density lipoprotein related receptors are single pass Type I transmembrane proteins which share several structural motifs. These include β-propellers (each containing several individual EGF repeats) and ligand-binding repeats (which mediate interactions with apolipoproteins. The relative sizes of LRP1, LRP4, LRP5/6, and LRP8 are represented. LRP5 and LRP6 form a novel subclass of this family with the cytoplasmic tail of these proteins containing PPPS/TP motifs which are targets of phosphorylation and act to stimulate downstream signaling upon activation by WNT ligands.
6. Therapeutic Considerations
Because Wnt/β-catenin signaling is elevated in many human tumors, there is significant interest in identifying agents that inhibit the pathway to treat several types of cancer 100. The same properties of WNT/β-catenin signaling to stimulate proliferation and/or regulate cell differentiation in the context of cancer cells are also what make it an attractive target for activation in the setting of regenerative medicine to treat diseases such as osteoporosis. It will be critical to remain cognizant of these properties in targeting this pathway in both settings.
One example of targeting the pathway for inhibition of tumor cell growth is an ongoing Phase I clinical trial in which a chemical inhibitor of Porcupine is being tested for efficacy in several types of cancer 101. There is a strong rationale for these trials. However, clinicians monitoring patients participating in these trials should be cognizant of potential side effects related to the normal functions of WNT signaling in mediating self-renewal of stem cell compartments. For example, in the context of bone biology one would be concerned, given that inhibition of WNT secretion disrupts normal bone homeostasis 60, 62, that patients being treated with Porcupine inhibitors might be susceptible to increased bone loss. If this were the case, perhaps this could be partially mitigated by concurrent treatment with agents like bisphosphonates that block bone resorption.
In the case of anabolic therapies designed to treat conditions associated with low bone mass, the potential for agents which activate the WNT pathway to enhance cell proliferation should always be considered. One potentially reassuring aspect for the use of therapies that activate WNT/β-catenin signaling in anabolic therapies is the experience with lithium chloride (LiCl) 102. A major mechanism for LiCl action is inhibition of GSK3 activity, thus leading to stabilization of β-catenin. LiCl has been used for over 60 years as a treatment for bipolar disorder 102. Despite this widespread use, there appears to be no reported cancer predisposition in these patients. Another way to diminish the potential for side effects in therapies designed to activate Wnt/β-catenin is to use strategies that limit the targets to specific tissues. In this respect, the specific enrichment of Sclerostin expression from osteocytes is a significant advantage.
Acknowledgments
The authors gratefully acknowledge support from the Van Andel Research Institute and NIAMS/NIH (R01 AR053293). We thank David Nadziejka for editorial support.
References
- 1.Franz-Odendaal TA. Induction and patterning of intramembranous bone. Front Biosci (Landmark Ed) 2011;16:2734–2746. doi: 10.2741/3882. [DOI] [PubMed] [Google Scholar]
- 2.Karaplis AC. Embryonic development of bone and the molecular regulation of intramembranous and endochondral bone formation. In: Bilezikian JP, Raisz L, Rodan GA, editors. Principles of Bone Biology. 2. Vol. 1. San Diego, CA: Academic Press; 2002. pp. 33–58. [Google Scholar]
- 3.Recker R. Embryology, anatomy, and microstructure of bone. In: Coe FL, Favus MJ, editors. Disorders of Bone and Mineral Metabolism. New York, NY: Raven Press; 1992. [Google Scholar]
- 4.Farnum CE, Wilsman NJ. Morphologic stages of the terminal hypertrophic chondrocyte of growth plate cartilage. Anat Rec. 1987;219:221–232. doi: 10.1002/ar.1092190303. [DOI] [PubMed] [Google Scholar]
- 5.Joiner DM, Ke J, Zhong Z, Xu HE, Williams BO. LRP5 and LRP6 in development and disease. Trends Endocrinol Metab. 2013;24:31–39. doi: 10.1016/j.tem.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.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]
- 7.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;50:649–657. doi: 10.1016/0092-8674(87)90038-9. [DOI] [PubMed] [Google Scholar]
- 8.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]
- 9.Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149:1192–1205. doi: 10.1016/j.cell.2012.05.012. [DOI] [PubMed] [Google Scholar]
- 10.He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way. Development. 2004;131:1663–1677. doi: 10.1242/dev.01117. [DOI] [PubMed] [Google Scholar]
- 11.Nusse R. The Wnt Gene Homepage. 2006 http://www.stanford.edu/~rnusse/wntwindow.html.
- 12.Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and beta-catenin signalling: diseases and therapies. Nat Rev Genet. 2004;5:691–701. doi: 10.1038/nrg1427. [DOI] [PubMed] [Google Scholar]
- 13.Ellies DL, Viviano B, McCarthy J, Rey JP, Itasaki N, Saunders S, Krumlauf R. Bone Density Ligand, Sclerostin, Directly Interacts With LRP5 but Not LRP5(G171V) to Modulate Wnt Activity. J Bone Miner Res. 2006;21:1738–1749. doi: 10.1359/jbmr.060810. [DOI] [PubMed] [Google Scholar]
- 14.Glass DA, 2nd, Karsenty G. Molecular bases of the regulation of bone remodeling by the canonical Wnt signaling pathway. Curr Top Dev Biol. 2006;73:43–84. doi: 10.1016/S0070-2153(05)73002-7. [DOI] [PubMed] [Google Scholar]
- 15.Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005;280:19883–19887. doi: 10.1074/jbc.M413274200. [DOI] [PubMed] [Google Scholar]
- 16.Ott SM. Sclerostin and Wnt signaling--the pathway to bone strength. J Clin Endocrinol Metab. 2005;90:6741–6743. doi: 10.1210/jc.2005-2370. [DOI] [PubMed] [Google Scholar]
- 17.Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005;280:26770–26775. doi: 10.1074/jbc.M504308200. [DOI] [PubMed] [Google Scholar]
- 18.Semenov MV, He X. LRP5 mutations linked to high bone mass diseases cause reduced LRP5 binding and inhibition by SOST. J Biol Chem. 2006 doi: 10.1074/jbc.M609509200. [DOI] [PubMed] [Google Scholar]
- 19.Winkler DG, Sutherland MS, Ojala E, Turcott E, Geoghegan JC, Shpektor D, Skonier JE, Yu C, Latham JA. Sclerostin inhibition of Wnt-3a-induced C3H10T1/2 cell differentiation is indirect and mediated by bone morphogenetic proteins. J Biol Chem. 2005;280:2498–2502. doi: 10.1074/jbc.M400524200. [DOI] [PubMed] [Google Scholar]
- 20.Green JL, Kuntz SG, Sternberg PW. Ror receptor tyrosine kinases: orphans no more. Trends Cell Biol. 2008;18:536–544. doi: 10.1016/j.tcb.2008.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Takada S, Stark KL, Shea MJ, Vassileva G, McMahon JA, McMahon AP. Wnt-3a regulates somite and tailbud formation in the mouse embryo. Genes Dev. 1994;8:174–189. doi: 10.1101/gad.8.2.174. [DOI] [PubMed] [Google Scholar]
- 22.Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–523. doi: 10.1016/s0092-8674(01)00571-2. [DOI] [PubMed] [Google Scholar]
- 23.Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, Manning SP, Swain PM, Zhao SC, Eustace B, et al. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. American journal of human genetics. 2002;70:11–19. doi: 10.1086/338450. [DOI] [PMC free article] [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. The New England journal of medicine. 2002;346:1513–1521. doi: 10.1056/NEJMoa013444. [DOI] [PubMed] [Google Scholar]
- 25.Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, Key L, Renton T, Bartels C, Gong Y, et al. Six novel missense mutations in the LDL receptor-related protein 5 (LRP5) gene in different conditions with an increased bone density. American journal of human genetics. 2003;72:763–771. doi: 10.1086/368277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Johnson ML, Summerfield DT. Parameters of LRP5 from a structural and molecular perspective. Crit Rev Eukaryot Gene Expr. 2005;15:229–242. doi: 10.1615/critreveukargeneexpr.v15.i3.50. [DOI] [PubMed] [Google Scholar]
- 27.Mani A, Radhakrishnan J, Wang H, Mani MA, Nelson-Williams C, Carew KS, Mane S, Najmabadi H, Wu D, Lifton RP. LRP6 mutation in a family with early coronary disease and metabolic risk factors. Science. 2007;315:1278–1282. doi: 10.1126/science.1136370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Leupin O, Piters E, Halleux C, Hu S, Kramer I, Morvan F, Bouwmeester T, Schirle M, Bueno-Lozano M, Fuentes FJ, et al. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. The Journal of biological chemistry. 2011;286:19489–19500. doi: 10.1074/jbc.M110.190330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Balemans W, Ebeling M, Patel N, Van Hul E, Olson P, Dioszegi M, Lacza C, Wuyts W, Van Den Ende J, Willems P, et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST) Human molecular genetics. 2001;10:537–543. doi: 10.1093/hmg/10.5.537. [DOI] [PubMed] [Google Scholar]
- 30.Williams BO. Insights into the mechanisms of sclerostin action in regulating bone mass accrual. J Bone Miner Res. 2014;29:24–28. doi: 10.1002/jbmr.2154. [DOI] [PubMed] [Google Scholar]
- 31.Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, Dioszegi M, Dikkers FG, Hildering P, Willems PJ, et al. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002;39:91–97. doi: 10.1136/jmg.39.2.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Staehling-Hampton K, Proll S, Paeper BW, Zhao L, Charmley P, Brown A, Gardner JC, Galas D, Schatzman RC, Beighton P, et al. 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;110:144–152. doi: 10.1002/ajmg.10401. [DOI] [PubMed] [Google Scholar]
- 33.van Bezooijen RL, Roelen BA, Visser A, van der Wee-Pals L, de Wilt E, Karperien M, Hamersma H, Papapoulos SE, ten Dijke P, Lowik CW. Sclerostin is an osteocyte-expressed negative regulator of bone formation, but not a classical BMP antagonist. The Journal of experimental medicine. 2004;199:805–814. doi: 10.1084/jem.20031454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kedlaya R, Veera S, Horan DJ, Moss RE, Ayturk UM, Jacobsen CM, Bowen ME, Paszty C, Warman ML, Robling AG. Sclerostin inhibition reverses skeletal fragility in an Lrp5-deficient mouse model of OPPG syndrome. Sci Transl Med. 2013;5:211ra158. doi: 10.1126/scitranslmed.3006627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Chang MK, Kramer I, Keller H, Gooi JH, Collett C, Jenkins D, Ettenberg SA, Cong F, Halleux C, Kneissel M. Reversing LRP5-dependent osteoporosis and SOST-deficiency induced sclerosing bone disorders by altering WNT signaling activity. J Bone Miner Res. 2013 doi: 10.1002/jbmr.2059. [DOI] [PubMed] [Google Scholar]
- 36.Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med. 2013;19:179–192. doi: 10.1038/nm.3074. [DOI] [PubMed] [Google Scholar]
- 37.Lammi L, Arte S, Somer M, Jarvinen H, Lahermo P, Thesleff I, Pirinen S, Nieminen P. Mutations in AXIN2 cause familial tooth agenesis and predispose to colorectal cancer. American journal of human genetics. 2004;74:1043–1050. doi: 10.1086/386293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Niemann S, Zhao C, Pascu F, Stahl U, Aulepp U, Niswander L, Weber JL, Muller U. Homozygous WNT3 mutation causes tetra-amelia in a large consanguineous family. American journal of human genetics. 2004;74:558–563. doi: 10.1086/382196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Faqeih E, Shaheen R, Alkuraya FS. WNT1 mutation with recessive osteogenesis imperfecta and profound neurological phenotype. J Med Genet. 2013;50:491–492. doi: 10.1136/jmedgenet-2013-101750. [DOI] [PubMed] [Google Scholar]
- 40.Laine CM, Joeng KS, Campeau PM, Kiviranta R, Tarkkonen K, Grover M, Lu JT, Pekkinen M, Wessman M, Heino TJ, et al. WNT1 mutations in early-onset osteoporosis and osteogenesis imperfecta. N Engl J Med. 2013;368:1809–1816. doi: 10.1056/NEJMoa1215458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pyott SM, Tran TT, Leistritz DF, Pepin MG, Mendelsohn NJ, Temme RT, Fernandez BA, Elsayed SM, Elsobky E, Verma I, et al. WNT1 mutations in families affected by moderately severe and progressive recessive osteogenesis imperfecta. Am J Hum Genet. 2013;92:590–597. doi: 10.1016/j.ajhg.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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;50:345–348. doi: 10.1136/jmedgenet-2013-101567. [DOI] [PubMed] [Google Scholar]
- 43.Bone marrow stromal cells influence myeloma progression via Dkk1. Bonekey Rep. 2012;1:104. doi: 10.1038/bonekey.2012.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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;8:739–750. doi: 10.1016/j.devcel.2005.03.016. [DOI] [PubMed] [Google Scholar]
- 45.Rudnicki JA, Brown AM. Inhibition of chondrogenesis by Wnt gene expression in vivo and in vitro. Dev Biol. 1997;185:104–118. doi: 10.1006/dbio.1997.8536. [DOI] [PubMed] [Google Scholar]
- 46.Reinhold MI, Kapadia RM, Liao Z, Naski MC. The Wnt-inducible transcription factor Twist1 inhibits chondrogenesis. J Biol Chem. 2006;281:1381–1388. doi: 10.1074/jbc.M504875200. [DOI] [PubMed] [Google Scholar]
- 47.Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell. 2005;8:727–738. doi: 10.1016/j.devcel.2005.02.013. [DOI] [PubMed] [Google Scholar]
- 48.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]
- 49.Rodda SJ, McMahon AP. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development. 2006;133:3231–3244. doi: 10.1242/dev.02480. [DOI] [PubMed] [Google Scholar]
- 50.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;280:21162–21168. doi: 10.1074/jbc.M501900200. [DOI] [PubMed] [Google Scholar]
- 51.Glass DA, 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, McMahon AP, Lang RA, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–764. doi: 10.1016/j.devcel.2005.02.017. [DOI] [PubMed] [Google Scholar]
- 52.Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003;423:337–342. doi: 10.1038/nature01658. [DOI] [PubMed] [Google Scholar]
- 53.Khosla S. Minireview: the OPG/RANKL/RANK system. Endocrinology. 2001;142:5050–5055. doi: 10.1210/endo.142.12.8536. [DOI] [PubMed] [Google Scholar]
- 54.Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89:309–319. doi: 10.1016/s0092-8674(00)80209-3. [DOI] [PubMed] [Google Scholar]
- 55.Miyazaki T, Tokimura F, Tanaka S. A review of denosumab for the treatment of osteoporosis. Patient Prefer Adherence. 2014;8:463–471. doi: 10.2147/PPA.S46192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Silva-Fernandez L, Rosario MP, Martinez-Lopez JA, Carmona L, Loza E. Denosumab for the treatment of osteoporosis: a systematic literature review. Reumatol Clin. 2013;9:42–52. doi: 10.1016/j.reuma.2012.06.007. [DOI] [PubMed] [Google Scholar]
- 57.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]
- 58.Port F, Basler K. Wnt trafficking: new insights into Wnt maturation, secretion and spreading. Traffic. 2010;11:1265–1271. doi: 10.1111/j.1600-0854.2010.01076.x. [DOI] [PubMed] [Google Scholar]
- 59.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]
- 60.Zhong Z, Zylstra-Diegel CR, Schumacher CA, Baker JJ, Carpenter AC, Rao S, Yao W, Guan M, Helms JA, Lane NE, et al. Wntless functions in mature osteoblasts to regulate bone mass. Proc Natl Acad Sci U S A. 2012;109:E2197–2204. doi: 10.1073/pnas.1120407109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wan Y, Lu C, Cao J, Zhou R, Yao Y, Yu J, Zhang L, Zhao H, Li H, Zhao J, et al. Osteoblastic Wnts differentially regulate bone remodeling and the maintenance of bone marrow mesenchymal stem cells. Bone. 2013;55:258–267. doi: 10.1016/j.bone.2012.12.052. [DOI] [PubMed] [Google Scholar]
- 62.Lu C, Wan Y, Cao J, Zhu X, Yu J, Zhou R, Yao Y, Zhang L, Zhao H, Li H, et al. Wnt-mediated reciprocal regulation between cartilage and bone development during endochondral ossification. Bone. 2013;53:566–574. doi: 10.1016/j.bone.2012.12.016. [DOI] [PubMed] [Google Scholar]
- 63.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;28:1150–1159. doi: 10.1002/jbmr.1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Maupin KA, Droscha CJ, Williams BO. A Comprehensive Overview of Skeletal Phenotypes Associated with Alterations in Wnt/β-catenin Signaling in Humans and Mice. Bone Research. 2013;1:27–71. doi: 10.4248/BR201301004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bennett CN, Longo KA, Wright WS, Suva LJ, Lane TF, Hankenson KD, MacDougald OA. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc Natl Acad Sci U S A. 2005;102:3324–3329. doi: 10.1073/pnas.0408742102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Stevens JR, Miranda-Carboni GA, Singer MA, Brugger SM, Lyons KM, Lane TF. Wnt10b deficiency results in age-dependent loss of bone mass and progressive reduction of mesenchymal progenitor cells. J Bone Miner Res. 2010;25:2138–2147. doi: 10.1002/jbmr.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Keupp K, Beleggia F, Kayserili H, Barnes AM, Steiner M, Semler O, Fischer B, Yigit G, Janda CY, Becker J, et al. Mutations in WNT1 cause different forms of bone fragility. Am J Hum Genet. 2013;92:565–574. doi: 10.1016/j.ajhg.2013.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Garcia-Ibarbia C, Perez-Nunez MI, Olmos JM, Valero C, Perez-Aguilar MD, Hernandez JL, Zarrabeitia MT, Gonzalez-Macias J, Riancho JA. Missense polymorphisms of the WNT16 gene are associated with bone mass, hip geometry and fractures. Osteoporos Int. 2013;24:2449–2454. doi: 10.1007/s00198-013-2302-0. [DOI] [PubMed] [Google Scholar]
- 69.Zheng HF, Tobias JH, Duncan E, Evans DM, Eriksson J, Paternoster L, Yerges-Armstrong LM, Lehtimaki T, Bergstrom U, Kahonen M, et al. WNT16 influences bone mineral density, cortical bone thickness, bone strength, and osteoporotic fracture risk. PLoS Genet. 2012;8:e1002745. doi: 10.1371/journal.pgen.1002745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 2003;5:367–377. doi: 10.1016/s1534-5807(03)00266-1. [DOI] [PubMed] [Google Scholar]
- 71.Yang Y, Topol L, Lee H, Wu J. Wnt5a and Wnt5b exhibit distinct activities in coordinating chondrocyte proliferation and differentiation. Development. 2003;130:1003–1015. doi: 10.1242/dev.00324. [DOI] [PubMed] [Google Scholar]
- 72.Grumolato L, Liu G, Mong P, Mudbhary R, Biswas R, Arroyave R, Vijayakumar S, Economides AN, Aaronson SA. Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors. Genes Dev. 2010;24:2517–2530. doi: 10.1101/gad.1957710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Oishi I, Suzuki H, Onishi N, Takada R, Kani S, Ohkawara B, Koshida I, Suzuki K, Yamada G, Schwabe GC, et al. The receptor tyrosine kinase Ror2 is involved in non-canonical Wnt5a/JNK signalling pathway. Genes Cells. 2003;8:645–654. doi: 10.1046/j.1365-2443.2003.00662.x. [DOI] [PubMed] [Google Scholar]
- 74.Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T, Kikuchi Y, Takada I, Kato S, Kani S, et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med. 2012;18:405–412. doi: 10.1038/nm.2653. [DOI] [PubMed] [Google Scholar]
- 75.Gofflot F, Hall M, Morriss-Kay GM. Genetic patterning of the posterior neuropore region of curly tail mouse embryos: deficiency of Wnt5a expression. Int J Dev Biol. 1998;42:637–644. [PubMed] [Google Scholar]
- 76.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;12:113–127. doi: 10.1016/j.devcel.2006.11.00. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Chen J, Tu X, Esen E, Joeng KS, Lin C, Arbeit JM, Ruegg MA, Hall MN, Ma L, Long F. WNT7B promotes bone formation in part through mTORC1. PLoS Genet. 2014;10:e1004145. doi: 10.1371/journal.pgen.1004145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Esen E, Chen J, Karner CM, Okunade AL, Patterson BW, Long F. WNT-LRP5 signaling induces Warburg effect through mTORC2 activation during osteoblast differentiation. Cell Metab. 2013;17:745–755. doi: 10.1016/j.cmet.2013.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Holmen SL, Giambernardi TA, Zylstra CR, Buckner-Berghuis BD, Resau JH, Hess JF, Glatt V, Bouxsein ML, Ai M, Warman ML, et al. Decreased BMD and limb deformities in mice carrying mutations in both Lrp5 and Lrp6. J Bone Miner Res. 2004;19:2033–2040. doi: 10.1359/JBMR.040907. [DOI] [PubMed] [Google Scholar]
- 80.Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA, 2nd, Hartmann C, Li L, Hwang TH, Brayton CF, et al. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–314. doi: 10.1083/jcb.200201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Fujino T, Asaba H, Kang MJ, Ikeda Y, Sone H, Takada S, Kim DH, Ioka RX, Ono M, Tomoyori H, et al. Low-density lipoprotein receptor-related protein 5 (LRP5) is essential for normal cholesterol metabolism and glucose-induced insulin secretion. Proc Natl Acad Sci U S A. 2003;100:229–234. doi: 10.1073/pnas.0133792100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.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]
- 83.Clement-Lacroix P, Ai M, Morvan F, Roman-Roman S, Vayssiere B, Belleville C, Estrera K, Warman ML, Baron R, Rawadi G. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci U S A. 2005;102:17406–17411. doi: 10.1073/pnas.0505259102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kubota T, Michigami T, Sakaguchi N, Kokubu C, Suzuki A, Namba N, Sakai N, Nakajima S, Imai K, Ozono K. Lrp6 hypomorphic mutation affects bone mass through bone resorption in mice and impairs interaction with Mesd. J Bone Miner Res. 2008;23:1661–1671. doi: 10.1359/jbmr.080512. [DOI] [PubMed] [Google Scholar]
- 85.Kokubu C, Heinzmann U, Kokubu T, Sakai N, Kubota T, Kawai M, Wahl MB, Galceran J, Grosschedl R, Ozono K, et al. Skeletal defects in ringelschwanz mutant mice reveal that Lrp6 is required for proper somitogenesis and osteogenesis. Development. 2004;131:5469–5480. doi: 10.1242/dev.01405. [DOI] [PubMed] [Google Scholar]
- 86.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]
- 87.Cui Y, Niziolek PJ, MacDonald BT, Zylstra CR, Alenina N, Robinson DR, Zhong Z, Matthes S, Jacobsen CM, Conlon RA, et al. Lrp5 functions in bone to regulate bone mass. Nat Med. 2011;17:684–691. doi: 10.1038/nm.2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yadav VK, Ryu JH, Suda N, Tanaka KF, Gingrich JA, Schutz G, Glorieux FH, Chiang CY, Zajac JD, Insogna KL, et al. Lrp5 controls bone formation by inhibiting serotonin synthesis in the duodenum. Cell. 2008;135:825–837. doi: 10.1016/j.cell.2008.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Inose H, Zhou B, Yadav VK, Guo XE, Karsenty G, Ducy P. Efficacy of serotonin inhibition in mouse models of bone loss. J Bone Miner Res. 2011;26:2002–2011. doi: 10.1002/jbmr.439. [DOI] [PubMed] [Google Scholar]
- 90.Lee GS, Simpson C, Sun BH, Yao C, Foer D, Sullivan B, Matthes S, Alenina N, Belsky J, Bader M, et al. Measurement of plasma, serum, and platelet serotonin in individuals with high bone mass and mutations in LRP5. J Bone Miner Res. 2014;29:976–981. doi: 10.1002/jbmr.2086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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;359:222–229. doi: 10.1016/j.ydbio.2011.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Leupin O, Piters E, Halleux C, Hu S, Kramer I, Morvan F, Bouwmeester T, Schirle M, Bueno-Lozano M, Fuentes FJ, et al. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. J Biol Chem. 2011;286:19489–19500. doi: 10.1074/jbc.M110.190330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Choi HY, Dieckmann M, Herz J, Niemeier A. Lrp4, a novel receptor for Dickkopf 1 and sclerostin, is expressed by osteoblasts and regulates bone growth and turnover in vivo. PLoS One. 2009;4:e7930. doi: 10.1371/journal.pone.0007930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Li Y, Pawlik B, Elcioglu N, Aglan M, Kayserili H, Yigit G, Percin F, Goodman F, Nurnberg G, Cenani A, et al. LRP4 mutations alter Wnt/beta-catenin signaling and cause limb and kidney malformations in Cenani-Lenz syndrome. Am J Hum Genet. 2010;86:696–706. doi: 10.1016/j.ajhg.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ohazama A, Porntaveetus T, Ota MS, Herz J, Sharpe PT. Lrp4: A novel modulator of extracellular signaling in craniofacial organogenesis. Am J Med Genet A. 2010;152A:2974–2983. doi: 10.1002/ajmg.a.33372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kumar J, Swanberg M, McGuigan F, Callreus M, Gerdhem P, Akesson K. LRP4 association to bone properties and fracture and interaction with genes in the Wnt- and BMP signaling pathways. Bone. 2011;49:343–348. doi: 10.1016/j.bone.2011.05.018. [DOI] [PubMed] [Google Scholar]
- 97.Ohazama A, Johnson EB, Ota MS, Choi HY, Porntaveetus T, Oommen S, Itoh N, Eto K, Gritli-Linde A, Herz J, et al. Lrp4 modulates extracellular integration of cell signaling pathways in development. PLoS One. 2008;3:e4092. doi: 10.1371/journal.pone.0004092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Terrand J, Bruban V, Zhou L, Gong W, El Asmar Z, May P, Zurhove K, Haffner P, Philippe C, Woldt E, et al. LRP1 controls intracellular cholesterol storage and fatty acid synthesis through modulation of Wnt signaling. J Biol Chem. 2009;284:381–388. doi: 10.1074/jbc.M806538200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zilberberg A, Yaniv A, Gazit A. The low density lipoprotein receptor-1, LRP1, interacts with the human frizzled-1 (HFz1) and down-regulates the canonical Wnt signaling pathway. J Biol Chem. 2004;279:17535–17542. doi: 10.1074/jbc.M311292200. [DOI] [PubMed] [Google Scholar]
- 100.Rey JP, Ellies DL. Wnt modulators in the biotech pipeline. Dev Dyn. 2010;239:102–114. doi: 10.1002/dvdy.22181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lum L, Clevers H. Cell biology. The unusual case of Porcupine. Science. 2012;337:922–923. doi: 10.1126/science.1228179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Phiel CJ, Klein PS. Molecular targets of lithium action. Annu Rev Pharmacol Toxicol. 2001;41:789–813. doi: 10.1146/annurev.pharmtox.41.1.789. [DOI] [PubMed] [Google Scholar]




