Open access peer-reviewed chapter

Wnt Signaling and Genetic Bone Diseases

Written By

Yanqin Lu and Jinxiang Han

Submitted: 04 June 2018 Reviewed: 22 August 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.81070

From the Edited Volume

Osteogenesis and Bone Regeneration

Edited by Haisheng Yang

Chapter metrics overview

1,008 Chapter Downloads

View Full Metrics

Abstract

The Wnt signal transduction plays a vital role in regulating development throughout the animal kingdom. The Wnt signal transduction is complex, including Wnt ligands, receptors, coreceptors, transducers, transcription factors, antagonists, agonists and their modulators, and target genes. It is classified into β-catenin-dependent canonical and independent non-canonical Wnt (mainly planar cell polarity and Wnt/Ca2+) signaling pathways. Wnt signaling pathway is causative to multiple human diseases. Gene mutations from the components of WNT signaling machinery have been identified to relate with low or high bone mass diseases, such as osteogenesis imperfecta, Robinow syndrome, osteoporosis-pseudoglioma syndrome, and sclerosteosis. In this review, we provide an update of the Wnt signaling pathway and the bone diseases caused by the aberrant components of the pathways.

Keywords

  • Wnt
  • Wnt signaling pathway
  • genetic bone diseases

1. Introduction

The Wnt1 gene (originally named Int1) was identified in 1982 as a gene activated by integration of mouse mammary tumor virus (MMTV) proviral DNA in virally induced breast tumors [1]. The Int1 proto-oncogene is highly conserved in many species, the fly wingless (Wg) gene in Drosophila, functions in controlling segment polarity during larval development and also activated in cancer, was found to be a homolog of Wnt1 [2]. Later, McMahon and Moon found that ectopic expression of Int1 in Xenopus leads to dual axis formation, when mouse Int1 RNA was injected into Xenopus embryos. Duplication of axial structures was abolished by substitution of a single, conserved cysteine residue of Int1 [3]. Later, more and more Wnt family members were identified.

Advertisement

2. Wnt and its secretion

2.1 Wnt proteins and their structure

Till now, Wnt family currently includes 19 secreted lipid-modified glycoproteins in most mammalian genomes, including the human genome. They fall into 12 conserved Wnt subfamilies, of which at least 11 of these occur in the genome of a Cnidaria, highlighting the vital role of Wnt family members in the process of organismal patterning throughout the animal kingdom [4]. In humans, Wnt1 and Wnt10b are located adjacent to each other on chromosome 12, and they are transcribed in opposite directions. Wnt6 and Wnt10a are located adjacent to each other on chromosome 2 and transcribed from the same strand of DNA. Other Wnt genes are prone to be clustered within the human genome also, including Wnt2 and Wnt16, Wnt3a and Wnt14, and Wnt3 and Wnt15 [5]. Wnt1-Wnt6-Wnt10 is an ancient cluster of Wnt genes in a common ancestor of vertebrates and arthropods and this cluster was duplicated leading to Wnt1-Wnt6-Wnt10a and Wnt1-Wnt6-Wnt10b cluster in vertebrates [5]. Based on their ability to induce transformation of the epithelial cell line C57MG, Wnt family are classified into highly transformation members, which includes Wnt1, Wnt2, Wnt3, and Wnt3a, and nontransformation members including Wnt4, Wnt5a, Wnt5b, and Wnt7b. High transformation members are related to Wnt/β-catenin canonical pathway and nontransformation members are related to noncanonical Wnt pathways. Wnt6 and Wnt7a are categorized as intermediate transformation members, leading to weak morphological changes [6].

Wnt genes encode proteins of ~350–400 residues in length, with molecular weight of about 40 kDa in size. Little is known about the structure of Wnts for their highly hydrophobic characteristics. In 2012, the 3D structure of Xenopus Wnt8 protein as bound to mouse Frizzled-8 cysteine-rich domain (CRD) was solved. XWnt8 is consist of an N-terminal α-helical domain (NTD) that includes the lipid-modified thumb and a C-terminal cysteine-rich region (CTD). They resemble the extended thumb and index fingers to project into a pocket in the opposite side of Fzd-CRD [7].

2.2 Posttranslational modifications of Wnts in the ER and Golgi apparatus

Wnt proteins share some features in common. They have an amino-terminal signal peptide that targets them to the ER and undergo a series of posttranslational modifications in the secretory pathway before transporting into the extracellular space. Wnts contain several charged residues and 23–25 cysteines on average, and some of them participate in inter- and intramolecular disulfide bonds, leading to Wnt folding and multimerization [7, 8]. All Wnt proteins (except Drosophila WntD) undergo posttranslational acylation and glycosylation [9]. There are two conserved residues of fatty acylation reported till now. The first acylation is palmitate attached to a conserved cysteine residue 77 in murine Wnt3a through a thioester linkage. The second lipid modification was identified at the position of serine 209 in murine Wnt3a protein. This conserved residue is modified by a monounsaturated fatty acid, palmitoleic acid [10, 11, 12]. This lipid posttranslational modification leads to extremely hydrophobicity of Wnts and restrict Wnt proteins to membranes by injecting into the lipid bilayer [9, 11]. Cys77 mutant leads to the loss of Wnt3a activity without affecting secretion, while Wnt3a Ser209Ala mutant is retained in the ER and secretion is blocked [10, 11]. Crystal structure of XWnt8 discovered that only conserved serine (corresponding to serine 209 in murine Wnt3a) is acylated. Cys77 is involved in the formation of disulfide bond with a second conserved cysteine [7]. Till now, Drosophila WntD is the only nonlipidated member of Wnt family [13]. Monoacylation is further corroborated by the lack of Cys77 palmitoylation study [14, 15]. This serine acylation is essential for Wnt binding to the coreceptor Frizzled, Wnt secretion and binding to the chaperone Wntless [7, 16, 17].

The attachment of palmitoleate to Wnt’s conserved serine is mediated through substrate specificity by acyltransferase Porcupine, which is homologous to the superfamily of acyltransferase enzymes localized to the endoplasmic reticulum (ER). Mutation of Porcupine impeded Wnt acylation activity in vitro [18]. Wnt palmitoylation is reversible and it can be removed by Notum, the serine hydrolase, and this deacylase activity is specific for Wnt proteins [19, 20]. Hence, Notum’s inhibitors have potential for treating degenerative diseases by targeting Wnt signaling [21].

N-Glycosylation is another common posttranslational modification of Wnt ligands, and nitrogen atom of multiple asparagine residues of Wnts is attached to oligosaccharide. This modification precedes palmitoylation and is independent of it [22, 23]. The number and position of N-glycosylation vary in different Wnt members [24]. The role of Wnt protein’s N-glycosylation is unclear, but usually, it influences secretion, but not folding and structure [9]. For Wg protein, which has two known N-glycosylation, Asn103 and Asn414, Wg mutant can activate downstream signaling in both autocrine and paracrine signaling, despite reduced secretion ability. Loss of N-glycosylation of Wnt1 impairs paracrine signaling. For Wnt3a and Wnt5a, N-glycosylation is essential for secretion, but not for the activity of Wnt5a protein [23, 25]. Porcupine plays an important role in both lipid and glycosylated modifications of Wnts and its mutant displayed a decreased N-glycosylation activity [8, 9, 10, 26].

Besides acylation and N-glycosylation of Wnt proteins, several other modifications are included in the posttranslational modification. Posttranslational tyrosine sulfation of Wnt5a and Wnt11 is essential for the formation of Wnt5a/Wnt11 complexes, which induce the efficient signaling in the context of Xenopus axis formation [27]. Wnt1 is attached to glycosylphosphatidylinositol (GPI) anchor on the leaflet of the plasma membrane by the glycolipid tail. PGAP1 gene participates in this modification by creating a hydrophobic Wnt1 that is retained in the ER [28].

2.3 Secretion and release of Wnt proteins

After posttranslational palmitoylation and N-glycosylation, mature Wnt proteins are then transported from the Golgi to the plasma membrane for secretion by the conserved multipass transmembrane Wntless (Wls) receptor (known as GPR177 in mammals) [29]. Wnt secretion could not proceed with the absence of Wls, but other signaling proteins are not influenced by the removal of Wls [30, 31, 32]. Wls knockout mice exhibit impairment of body axis formation, and a phenotype mimics the deficiency of Wnt3. Wls is activated by β-catenin and LEF/TCF-dependent transcription and its mutants impede Wnt secretion and signaling [33]. Wls is essential for Wnt signaling, and tissue-specific knockouts of Wls impede varieties of processes including bone mass, skin homeostasis, peripheral lung differentiation, and pulmonary vascular development [34, 35, 36].

Endogenous Wls contains a carboxy-terminal ER-targeting signal, which directs Wls localizing predominantly in the ER, where it binds with acylated Wnt proteins [16, 37]. P24 protein family, which acts as cargo receptor for Wnt in the early secretory pathway, is essential for proper export of Wg from the ER [38, 39, 40]. Sec22 is packaged together with Wg and p24 during the early secretory phase of Wg and it functions as the vesicle SNARE (soluble NSF attachment protein receptor) [40].

The detailed mechanisms for Wnt secretion are not clear. Wnts-Wls complex transport from ER to plasma membrane is COPII vesicles dependent. Once arriving at the plasm membrane, Wnt is then released from plasm membrane and binds to lipoprotein particles or heparin sulfate proteoglycans (HSPGs) [41, 42]. The other theory supports that Wnt-Wls complex keeps together and internalizes at plasm membrane and dissociates from each other in endosomes. Then, Wnts is released through a recycling endosomal pathway and Wls is transported back to TGN through a retromer-dependent pathway [42, 43, 44]. Dpy23 and Vps35 are reported to regulate recycling of C. elegans Mig-14, which is the homolog of Wls. Wls is restricted to the plasma membrane with the Dyp23 mutant [45]. Retromer complex consists of Vps35, Vps29, Vps26, Vps10, Vps5, and Vps17 in yeast [46, 47, 48]. Vps35, Vps29, and Vps26 subcomplexes mediate cargo recognition and retrieve Vps10p from endosomes to the Golgi [47]. Vps35 mutant has no influence on the transportation of Wls to plasm membrane and endocytosis, but the retromer-dependent shuttle to the Golgi is inaccessible, and endocytosed Wls progresses to MVBs and lysosomes for degradation [43, 44, 49]. Vps5 and Vps17 are membrane-bound subcomplexes of retromer, and they are sorting nexins (SNX) with a phosphoinositide-binding SNX-phox homology (SNX-PX) domain [50]. Nexins SNX1, SNX2, SNX5, and SNX6 are SNX-BAR coat complex that interact with cargo-selective Vps35-Vps29-Vps26 complex. They are needed for most of the retromer cargo proteins, but not for the process of Wls recycling [50, 51]. Wls recycling specifically relies on SNX3, the retromer without BAR domain [51, 52]. SNX3 cointeracts with Wls and Vps26 on early endosomes and helps the association of the cargo-selective complex to Wls [51]. Wls recycled in Golgi further retrogrades transport to ER, which is mediated by the conserved C terminal sequence of Wls targeting ER. This process is currently COPI dependent and requires ER-Golgi intermediate compartment ERGIC2, though retrieval mechanisms need further investigation [37, 53]. Recently, miR-307a is found to inhibit Wg secretion by targeting Wls, and its overexpression induces ER stress specifically in the Wg-expressing cells. KKVY motif of Wg is responsible for its retrieval and ER stress [53].

Wnts are classic morphogens, which play an important role in tissue patterning by activating their target genes in a concentration-dependent manner and act in short and long range way [14, 54]. Various carriers have been identified that associate with extracellular Wnts, which include exovesicles [55], exosomes [56, 57], lipoprotein [41, 58], cytonemes (filopodia-like protrusions) [59, 60, 61], and Swim (secreted Wnt-interacting molecule) belonging to lipocalin family of protein [62]. These secreted Wnts associate to specific receptors on target cells to activate either canonical Wnt/β-catenin pathway or noncanonical Wnt/Ca2+ pathway.

Advertisement

3. Wnt signaling pathway

3.1 The canonical Wnt signaling pathway

The Wnt signaling pathway serves many important functions in body axis patterning, embryonic development, cell proliferation, and differentiation. In the absence of Wnt signaling, β-catenin is phosphorylated and ubiquitinated to keep low level by forming β-catenin destruction complex. The complex includes β-catenin, axin, casein kinase-1 (CK1), glycogen synthase kinase-3β (GSK-3β), and the adenomatous polyposis coli (APC) [63, 64]. PP2A and HSP105 are also involved in this complex. HSP105 recruits the phosphatase PP2A to the degradation complex to antagonize the phosphorylation of β-catenin, thus keeping the balance of phosphorylation-dephosphorylation [65]. Maintaining a phosphostatus balance of the β-catenin protein leads to its accumulation or degradation based on the signaling cues. The complex binds and phosphorylates β-catenin, leading to the ubiquitination by β-transducin repeat-containing protein (β-TrCP) ubiquitin ligase and subsequent proteasomal degradation [66].

In the presence of Wnt ligands, Wnt ligands bind to the specific receptor including Frizzled (Fzd) family member and subsequent LRP5/6 coreceptor. Axin is dephosphorylated and sequestered at the membrane. The binding triggers the recruitment of phosphoprotein disheveled (Dsh/Dvl) to form the LRP/Fzd/Dsh complexes, inducing the phosphorylation of LRP by CK1Υ and GSK3; as a consequence, axin is then dephosphorylated and sequestered at the membrane and destruction complex is inactivated. The signalosome composed of Fzd, LRP5/6, Dvl, axin, GSK3, and CK1 destroys the β-catenin destruction complex [67, 68]. Hence, cytosolic β-catenin accumulates and localizes to the nucleus, where it interacts with TCF/LEF family members and recruits other transcriptional coactivators, such as CBP, TBP, and BRG-1, to induce target gene expression [69, 70].

Axin is a scaffold protein and acts as an anchor for other four proteins in the complex. In addition, axin participates in the LRP6 phosphorylation on the PPPSPxS motifs, which in turn cause the accumulation of axin in the destruction complex and then lead to the initiation of β-catenin signaling [71]. Recently, axin was found to be fully phosphorylated in the state of Wnt-off and partly phosphorylated in the state of Wnt-on mediated by GSK-3β [72].

The role of APC in Wnt signaling is complex and multiple. APC acts as a carrier for GSK-3β and axin that promotes phosphorylation and consequent ubiquitin-dependent degradation of β-catenin [73]. It binds to β-catenin by 15 or 20-mer amino acid repeats. APC promotes export of β-catenin from nucleus, and hence the expression and transcriptional activity of nuclear β-catenin are reduced indirectly [74]. Meanwhile, APC downregulates the β-catenin/TCF transcription by directly interacting with transcriptional repressor C-terminal binding protein (CtBP) [75, 76]. APC may also serve as a positive regulator for Wnt signaling through downregulation of axin [77]. APC is vital for the phosphorylation of axin in both Wnt-off and Wnt-on states, the association with activated phospho-LRP6 and the rapid transition in axin activity [72]. Phosphorylated β-catenin requires APC for its targeting to ubiquitin ligase and protection from dephosphorylation mediated by protein phosphatase 2A (PP2A) [78]. Recently, APC was found to impede clathrin-dependent signalosome formation in the absence of ligand [79].

GSK3-β and CK1 are both serine/threonine kinases that phosphorylate the N-terminal portion of cytosolic β-catenin, and phosphorylation of β-catenin begins at Ser45 by CK1α and then phosphorylation of residues Thr41, Ser37, and Ser33 [80, 81]. Meanwhile, CK1, perhaps also GSK3β, phosphorylates APC on the 20-mer repeats. Phosphorylation of APC increases the binding affinity to β-catenin, and β-catenin disassociates from axin [63]. Phosphorylated β-catenin is then recognized by β-TrCP1, an F-box protein component of an Skp1/Cul1/F-box (SCF)-type ubiquitin ligase complex [82], followed by recruitment of E3 ubiquitin ligase and degraded by the 26S proteasome [83].

PP2A is a cellular heterotrimeric serine-threonine protein phosphatase consisting of a structural (A), a regulatory (B), and a catalytic subunit (C) [84]. PP2A has a dual opposite regulation role for Wnt signaling. PP2A is regarded as one of the members of β-catenin degradation complex [85]. PP2A dephosphorylates GSK3β through recruitment of DNAJB6 (DnaJ homolog subfamily B member 6) and HSPA8 (heat-shock cognate protein, HSC70) [86]. The B56 subunit of PP2A interacts with N-terminal of APC and decreases the amount of β-catenin and inhibits transcription of its target genes [87, 88]. Also, B56ε is required for Wnt/β-catenin signaling downstream of the Wnt ligand and upstream of Dsh [89]. PR61 β regulates Wnt signaling by inhibiting Dvl- and β-catenin–dependent T-cell factor activation, or suppressing the downstream target genes [90]. PR55α subunit of PP2A acts as the positive regulator for Wnt signaling. It interacts with β-catenin directly and controls dephosphorylation and degradation of β-catenin. Knockdown of PR55α increases β-catenin phosphorylation and decreases Wnt signaling, whereas is the same as PR55α overexpression [91, 92]. PP2C also upregulates Wnt signaling through the dephosphorylation of axin [93]. Meanwhile, many subunits of PP2A, such as PR55α, A, C, B56α, and PR61β and Υ, are reported to interact with axin [87, 90, 91].

PR61ε subunit of PP2A is involved in the initiation of the Wnt pathway. PR61ε binds to Fzd receptor, and binding of Wnt ligands promote the interaction of LRP5/6-associated CK1ε and PR61ε. The latter dephosphatases CK1ε, leading to recruitment of Dvl-2 to the receptor complex and the initiation of the Wnt signaling [94].

3.2 The noncanonical Wnt pathway

Due to varieties of both Wnts and their receptors and coreceptors, Wnt pathways are multiple and complex. There are multiple branches of β-catenin–independent Wnt signaling pathways. One is the Wnt/Ca2+ pathway, modulating intracellular Ca2+ level. The second is the Wnt/planar cell polarity (PCP) pathway, utilizing small Rho-like GTPases [95].

3.2.1 Wnt/PCP pathway

Polarization is a global property of cells and tissues. In addition to the ubiquitous epithelial apical-basal axis, many multicellular tissues also have planar cell polarity, orthology to apico-basal polarity [96, 97]. Compared with canonical Wnt signaling, various cell surface receptors have been involved in PCP signaling. PCP is composed of core protein complexes and Fat/Dachsous (Ds)/Fj (four-jointed) group. The latter is reported to act upstream of PCP to provide a directional information [98, 99]. Core protein complexes are composed of Frizzled, Flamingo (Fmi/Celsr), Van Gogh (Drosophila Vang or Stb/mammalian Vang), disheveled (Dsh/Dvl), Diego, and Prickle (Pk) [100]. The core complex within puncta is predominately stable than elsewhere in the junctions and highly asymmetrically organized, while core protein stoichiometry in both puncta and nonpuncta region is similar. The core protein is assembled around a stoichiometric Fz-Fmi nucleus. The amount of Fz and Stb is maintained relative to their binding partners for normal asymmetry [101]. In many cancers, Wnt/PCP signaling is upregulated and it contributes to cancer malignancy by enhancing the proliferation and migration, priming metastasis niches, and causing resistance to therapy [102, 103].

3.2.2 Wnt/Ca2+ signaling pathway

Wnt5a is the most common ligand for noncanonical Wnt signal transducer. It activates calcium signaling pathway by binding to receptor Fz2, 3, 4, 5, and Fz6, as well as coreceptor Ror1/2, which is the membrane-bound receptor tyrosine kinase [104, 105, 106, 107]. Dvl, axin, and GSK organize the complex and GSK phosphorylates Ror coreceptor [108]. Wnt/Fz/Ror then activates phospholipase C (PLC), leading to the generation of diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3) from membrane-bound phospholipid phosphatidyl inositol 4,5-bisphosphate (PIP2). Recently, SEC1413/the Sec14-like protein acts as GTPase proteins to mediate specific Wnt-Fz-Dvl complex signals downstream to phospholipase C δ4a (PLCδ4a). The binding of SEC141 to Wnt-Fz-Dvl complexes induces its translocation of SEC1413 to the plasma membrane, and then further binds to and activates PLCδ4a. In turn, PLCδ4a acts as a GTPase-activating protein to promote the hydrolysis of Sec14l3-bound GTP to GDP [109]. IP3 promotes the concentration of intracellular Ca2+, which activates calcineurin, phospho-serine/threonine specific protein phosphatase and calcium calmodulin-dependent protein kinase II (CaMKII). In turn, nuclear factor associated with T cells (NFAT) and regulatory proteins NFkB are activated. DAG activates protein kinase C (PKC), which further activates NFkB and CREB. Meanwhile, Wnt/Fz interaction may activate phosphodiesterase 6 (PDE6) in a calcium-dependent manner, leading to a decrease in cyclic guanosine monophosphate (cGMP) [110].

Advertisement

4. Wnt signaling in genetic bone diseases

Both bone modeling and remodeling are regulated by Wnt signaling, and mutation of Wnt signaling components is linked to various genetic bone diseases. Table 1lists human genetic bone diseases caused by Wnt signaling disorders. Genotypic and phenotypic heterogeneity of genetic bone diseases–related Wnt signaling pathways is obvious. Tooth agenesis is caused by Wnt 10a, Wnt10b, and LRP6 by either autosomal dominant (AR) or autosomal recessive (AR) inheritance form [111, 112, 113]. Tetraamelia syndrome is skeletally characterized by limb agenesis or complete absence of limbs, bilateral cleft lip/palate, ankyloglossia, and mandibular hypoplasia with the pathogenic gene of Wnt3 and RSPO2 [114, 115]. Other limb deficiency diseases in Wnt signaling includes Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome (AARRS) (MIM 276820) and split-hand/foot malformation 6 (MIM 225300), with pathogenic gene of Wnt7a and Wnt10b, respectively [116, 117].

PhenotypePhenotype MIM numberInheritanceGeneGene MIM numberReference
Wnt ligands
Osteogenesis imperfecta, type XV615220ARWnt1164820Pyott et al. [125]
Osteoporosis, early-onset, susceptibility to, autosomal dominant615221Wnt1Laine et al. [127]
Tetraamelia syndrome 1273395ARWnt3165330Niemann et al. [115]
Robinow syndrome, autosomal dominant 1180700ADWnt5a164975Person et al. [138]
Fuhrmann syndrome228930ARWnt7a601570Woods et al. [116]
Ulna and fibula, absence of, with severe limb deficiency276820ARWnt7a601570Woods et al. [116]
Odontoonychodermal dysplasia257980ARWnt10a606268Adaimy et al. [139]
Schopf-Schulz-Passarge syndrome224750ARWnt10a606268Bohring et al. [140]
Tooth agenesis, selective, 4150400AR, ADWnt10a606268Kantaputra and Sripathomsawat [111]
Split-hand/foot malformation 6225300ARWnt10b601906Ugur and Tolun [117]
Tooth agenesis, selective, 8617073ADWnt10b601906Yu et al. [112]
Receptor/coreceptor
Robinow syndromeARFZD2600667White et al. [141]
Nail disorder, nonsyndromic congenital, 10614157ARFZD6603409Frojmark et al. [142]
Cenani-Lenz syndactyly syndrome212780ARLRP4604270Li et al. [143]
Sclerosteosis 2614305AR, ADLRP4604270Leupin et al. [132]
Osteopetrosis, autosomal dominant 1607634ADLRP5603506Van Wesenbeeck et al. [122], Van Hul et al. [123]
Osteoporosis-pseudoglioma syndrome259770ARLRP5603506Gong et al. [124]
Osteosclerosis144750ADLRP5603506Van Wesenbeeck et al. [122]
Hyperostosis, endosteal144750ADLRP5603506Van Wesenbeeck et al. [122]
Van Buchem disease, type 2607636ADLRP5603506Van Wesenbeeck et al. [122], Little et al. [130]
Bone mineral density variability 1601884ADLRP5603506Nguyen et al. [131]
Osteoporosis166710ADLRP5603506Estrada et al. [121]
Tooth agenesis, selective, 7616724ADLRP6603507Massink et al. [113]
Brachydactyly, type B1113000ADROR2602337Oldridge et al. [144]
Robinow syndrome, autosomal recessive268310ARROR2602337van Bokhoven et al. [145], Afzal et al. [146]
Simpson-Golabi-Behmel syndrome, type 1312870XLRGPC3300037Pilia et al. [147]
Omodysplasia 1258315ARGPC6604404Campos-Xavier et al. [148]
Fetal akinesia deformation sequence208150ARMUSK601296Tan-Sindhunata et al. [149]
Antagonist
Osteoarthritis susceptibility 1165720MuSFRP3605083Loughlin et al. [150]
Pyle disease265900ARSFRP4606570Kiper et al. [129]
Craniodiaphyseal dysplasia, autosomal dominant122860ADSOST605740Kim et al. [135]
Sclerosteosis 1269500ARSOST605740Brunkow et al. [134]
Van Buchem disease239100ARSOST605740Balemans et al. [133]
Agonists
Robinow syndrome, autosomal dominant 2616331ADDVL1601365White et al. [151]
Robinow syndrome, autosomal dominant 3616894ADDVL3601368White et al. [152]
Bone mineral density, low, susceptibility to615311LGR4606666Styrkarsdottir et al. [128]
Palmoplantar hyperkeratosis with squamous cell carcinoma of skin and sex reversal610644ARRSPO1609595Parma et al. [153]
Humerofemoral hypoplasia with radiotibial ray deficiency618022RSPO2610575Szenker-Ravi et al. [114]
Tetraamelia syndrome 2618021RSPO2610575Szenker-Ravi et al. [114]
Anonychia congenita206800ARRSPO4610573Blaydon et al. [154]

Table 1.

Wnt signaling and human genetic bone diseases.

Robinow syndrome (RS) is characterized by facial features, orodental abnormalities, and hypoplastic genitalia [118]. All autosomal-dominant (DRS) and recessive (RRS) genes including Wnt5a, Dvl1, Dvl3, Fzd2, and ROR2 are involved in the Wnt/PCP pathways. This pathway plays an important role in the patterning and formation of the limb-bud outgrowth and growth plate in skeletal formation [119, 120].

Wnt signaling pathways are related to bone diseases with osteoporosis or high bone mass density (BMD) diseases. LRP5 gene is responsible for osteoporosis. Loss of function of LRP5 mutation causes osteoporosis (MIM 166710, 607634) and osteoporosis pseudoglioma syndrome (MIM 259770) [121, 122, 123, 124]. Meanwhile, osteoporosis genes in Wnt signaling components include Wnt1, LGR4, and SFRP4. Wnt1 is the pathogenic gene for osteogenesis imperfect type XV (with bilateral mutations) and early onset osteoporosis (with heterozygous mutation) [125, 126, 127]. For LGR4, nonsense variation of c.376C-T is strongly associated with low bone mass density and osteoporotic fractures [128]. SFRP4 is the pathogenic gene for Pyle disease characterized by both osteoporosis and expanded trabecular metaphyses [129].

LRP5 is also the pathogenic gene for diseases with high BMD, Van Buchem syndrome type 2 (MIM 607636), bone mineral density variability (MIM 601884), osteosclerosis, and hyperostosis, endosteal (MIM 144750) [122, 130, 131]. LRP4 mutations lead to type I sclerosteosis (MIM 614305), which is also the disease with high BMD [132]. Sclerosteosis (SOST) gene mutation causes the high BMD diseases of Van Buchem syndrome (MIM 239100), sclerosteosis 1 (MIM 269500), and craniodiaphyseal dysplasia (MIM 122860) [133, 134, 135]. Sclerostin encoded by SOST gene is the endogenous Wnt signaling inhibitor, which interacts with LRP receptors [136]. Nowadays, monoclonal antibody of sclerostin is being tested in human clinical trials [137].

In all, the components of Wnt signaling including Wnt ligands, their receptors, coreceptors, antagonists, and agonists can cause different types of genetic bone diseases, which are related to both canonical and noncanonical Wnt signaling pathways. Study of Wnt signaling in genetic bone diseases and other human diseases provides promises for translational medicine.

Advertisement

5. Conclusions

We review the current status of Wnt signaling, including the secretion of Wnt ligands, and how Wnts binding to surface receptors trigger different intracellular response and transcription of different downstream target genes. However, the interactions among each components and the mechanisms of these interactions still need further study. Meanwhile, the cross talk network between canonical and noncanonical Wnt signaling, Wnt signaling, and other signaling pathways remains unsolved fully. Mutations in the components of Wnt signaling pathways lead to various genetic bone diseases and other genetic diseases. Genotypic and phenotypic heterozygosis is common in these genetic bone diseases. For the vital role of Wnt signaling components in bone diseases, potential drugs based on Wnt signaling is useful for treating different bone diseases.

Advertisement

Acknowledgments

The study was supported by the grants from the Shandong Provincial Natural Science Foundation of China (2015ZRC03171), Shandong Key Research and Development Plan (2016GSF201222).

Advertisement

Conflict of interest

No conflict of interest.

References

  1. 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
  2. 2. 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
  3. 3. McMahon AP, Moon RT. Ectopic expression of the proto-oncogene int-1 in Xenopus embryos leads to duplication of the embryonic axis. Cell. 1989;58:1075-1084
  4. 4. Kusserow A, Pang K, Sturm C, Hrouda M, Lentfer J, Schmidt HA, et al. Unexpected complexity of the Wnt gene family in a sea anemone. Nature. 2005;433:156-160
  5. 5. Nusse R. An ancient cluster of Wnt paralogues. Trends in Genetics. 2001;17:443
  6. 6. Shimizu H, Julius MA, Giarre M, Zheng Z, Brown AM, Kitajewski J. Transformation by Wnt family proteins correlates with regulation of beta-catenin. Cell Growth & Differentiation. 1997;8:1349-1358
  7. 7. Janda CY, Waghray D, Levin AM, Thomas C, Garcia KC. Structural basis of Wnt recognition by Frizzled. Science. 2012;337:59-64
  8. 8. Tanaka K, Kitagawa Y, Kadowaki T. Drosophila segment polarity gene product porcupine stimulates the posttranslational N-glycosylation of wingless in the endoplasmic reticulum. The Journal of Biological Chemistry. 2002;277:12816-12823
  9. 9. Coudreuse D, Korswagen HC. The making of Wnt: New insights into Wnt maturation, sorting and secretion. Development. 2007;134:3-12
  10. 10. Willert K, Brown JD, Danenberg E, Duncan AW, Weissman IL, Reya T, et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature. 2003;423:448-452
  11. 11. Takada R, Satomi Y, Kurata T, Ueno N, Norioka S, Kondoh H, et al. Monounsaturated fatty acid modification of Wnt protein: Its role in Wnt secretion. Developmental Cell. 2006;11:791-801
  12. 12. Franch-Marro X, Wendler F, Griffith J, Maurice MM, Vincent JP. In vivo role of lipid adducts on Wingless. Journal of Cell Science. 2008;121:1587-1592
  13. 13. Ching W, Hang HC, Nusse R. Lipid-independent secretion of a Drosophila Wnt protein. The Journal of Biological Chemistry. 2008;283:17092-17098
  14. 14. Parchure A, Vyas N, Mayor S. Wnt and Hedgehog: Secretion of lipid-modified morphogens. Trends in Cell Biology. 2018;28:157-170
  15. 15. Rios-Esteves J, Haugen B, Resh MD. Identification of key residues and regions important for porcupine-mediated Wnt acylation. The Journal of Biological Chemistry. 2014;289:17009-17019
  16. 16. Coombs GS, Yu J, Canning CA, Veltri CA, Covey TM, Cheong JK, et al. WLS-dependent secretion of WNT3A requires Ser209 acylation and vacuolar acidification. Journal of Cell Science. 2010;123:3357-3367
  17. 17. Herr P, Basler K. Porcupine-mediated lipidation is required for Wnt recognition by Wls. Developmental Biology. 2012;361:392-402
  18. 18. Stabley JN, Towler DA. Arterial calcification in diabetes mellitus: Preclinical models and translational implications. Arteriosclerosis, Thrombosis, and Vascular Biology. 2017;37:205-217
  19. 19. De Gregorio R, Pulcrano S, De Sanctis C, Volpicelli F, Guatteo E, von Oerthel L, et al. miR-34b/c regulates Wnt1 and enhances mesencephalic dopaminergic neuron differentiation. Stem Cell Reports. 2018;10:1237-1250
  20. 20. Gross JC, Zelarayan LC. The mingle-mangle of Wnt signaling and extracellular vesicles: Functional implications for heart research. Frontiers in Cardiovascular Medicine. 2018;5:10
  21. 21. Peghaire C, Bats ML, Sewduth R, Jeanningros S, Jaspard B, Couffinhal T, et al. Fzd7 (Frizzled-7) expressed by endothelial cells controls blood vessel formation through Wnt/beta-catenin canonical signaling. Arteriosclerosis, Thrombosis, and Vascular Biology. 2016;36:2369-2380
  22. 22. Komekado H, Yamamoto H, Chiba T, Kikuchi A. Glycosylation and palmitoylation of Wnt-3a are coupled to produce an active form of Wnt-3a. Genes to Cells. 2007;12:521-534
  23. 23. Doubravska L, Krausova M, Gradl D, Vojtechova M, Tumova L, Lukas J, et al. Fatty acid modification of Wnt1 and Wnt3a at serine is prerequisite for lipidation at cysteine and is essential for Wnt signalling. Cellular Signalling. 2011;23:837-848
  24. 24. Tang X, Wu Y, Belenkaya TY, Huang Q, Ray L, Qu J, et al. Roles of N-glycosylation and lipidation in Wg secretion and signaling. Developmental Biology. 2012;364:32-41
  25. 25. Kurayoshi M, Yamamoto H, Izumi S, Kikuchi A. Post-translational palmitoylation and glycosylation of Wnt-5a are necessary for its signalling. The Biochemical Journal. 2007;402:515-523
  26. 26. Ching W, Nusse R. A dedicated Wnt secretion factor. Cell. 2006;125:432-433
  27. 27. Nadanaka S, Kinouchi H, Kitagawa H. Histone deacetylase-mediated regulation of chondroitin 4-O-sulfotransferase-1 (Chst11) gene expression by Wnt/beta-catenin signaling. Biochemical and Biophysical Research Communications. 2016;480:234-240
  28. 28. Kinoshita T, Maeda Y, Fujita M. Transport of glycosylphosphatidylinositol-anchored proteins from the endoplasmic reticulum. Biochimica et Biophysica Acta. 2013;1833:2473-2478
  29. 29. Das S, Yu S, Sakamori R, Stypulkowski E, Gao N. Wntless in Wnt secretion: Molecular, cellular and genetic aspects. Frontiers in Biology (Beijing). 2012;7:587-593
  30. 30. Langton PF, Kakugawa S, Vincent JP. Making, exporting, and modulating Wnts. Trends in Cell Biology. 2016;26:756-765
  31. 31. Banziger C, Soldini D, Schutt C, Zipperlen P, Hausmann G, Basler K. Wntless, a conserved membrane protein dedicated to the secretion of Wnt proteins from signaling cells. Cell. 2006;125:509-522
  32. 32. Bartscherer K, Pelte N, Ingelfinger D, Boutros M. Secretion of Wnt ligands requires Evi, a conserved transmembrane protein. Cell. 2006;125:523-533
  33. 33. Fu J, Jiang M, Mirando AJ, Yu HM, Hsu W. Reciprocal regulation of Wnt and Gpr177/mouse Wntless is required for embryonic axis formation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:18598-18603
  34. 34. Zhong Z, Zylstra-Diegel CR, Schumacher CA, Baker JJ, Carpenter AC, Rao S, et al. Wntless functions in mature osteoblasts to regulate bone mass. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:E2197-E2204
  35. 35. Augustin I, Gross J, Baumann D, Korn C, Kerr G, Grigoryan T, et al. Loss of epidermal Evi/Wls results in a phenotype resembling psoriasiform dermatitis. The Journal of Experimental Medicine. 2013;210:1761-1777
  36. 36. Cornett B, Snowball J, Varisco BM, Lang R, Whitsett J, Sinner D. Wntless is required for peripheral lung differentiation and pulmonary vascular development. Developmental Biology. 2013;379:38-52
  37. 37. Yu J, Chia J, Canning CA, Jones CM, Bard FA, Virshup DM. WLS retrograde transport to the endoplasmic reticulum during Wnt secretion. Developmental Cell. 2014;29:277-291
  38. 38. Buechling T, Chaudhary V, Spirohn K, Weiss M, Boutros M. p24 proteins are required for secretion of Wnt ligands. EMBO Reports. 2011;12:1265-1272
  39. 39. Port F, Hausmann G, Basler K. A genome-wide RNA interference screen uncovers two p24 proteins as regulators of Wingless secretion. EMBO Reports. 2011;12:1144-1152
  40. 40. Li X, Wu Y, Shen C, Belenkaya TY, Ray L, Lin X. Drosophila p24 and Sec22 regulate Wingless trafficking in the early secretory pathway. Biochemical and Biophysical Research Communications. 2015;463:483-489
  41. 41. Panakova D, Sprong H, Marois E, Thiele C, Eaton S. Lipoprotein particles are required for Hedgehog and Wingless signalling. Nature. 2005;435:58-65
  42. 42. Lorenowicz MJ, Korswagen HC. Sailing with the Wnt: Charting the Wnt processing and secretion route. Experimental Cell Research. 2009;315:2683-2689
  43. 43. Port F, Kuster M, Herr P, Furger E, Banziger C, Hausmann G, et al. Wingless secretion promotes and requires retromer-dependent cycling of Wntless. Nature Cell Biology. 2008;10:178-185
  44. 44. Yang PT, Lorenowicz MJ, Silhankova M, Coudreuse DY, Betist MC, Korswagen HC. Wnt signaling requires retromer-dependent recycling of MIG-14/Wntless in Wnt-producing cells. Developmental Cell. 2008;14:140-147
  45. 45. Pan CL, Baum PD, Gu M, Jorgensen EM, Clark SG, Garriga G. C. elegans AP-2 and retromer control Wnt signaling by regulating mig-14/Wntless. Developmental Cell. 2008;14:132-139
  46. 46. Hausmann G, Banziger C, Basler K. Helping Wingless take flight: How WNT proteins are secreted. Nature Reviews. Molecular Cell Biology. 2007;8:331-336
  47. 47. Seaman MN. Recycle your receptors with retromer. Trends in Cell Biology. 2005;15:68-75
  48. 48. Eaton S. Retromer retrieves wntless. Developmental Cell. 2008;14:4-6
  49. 49. Yuasa T, Otani T, Koike T, Iwamoto M, Enomoto-Iwamoto M. Wnt/beta-catenin signaling stimulates matrix catabolic genes and activity in articular chondrocytes: Its possible role in joint degeneration. Laboratory Investigation. 2008;88:264-274
  50. 50. Cullen PJ. Endosomal sorting and signalling: An emerging role for sorting nexins. Nature Reviews. Molecular Cell Biology. 2008;9:574-582
  51. 51. Harterink M, Port F, Lorenowicz MJ, McGough IJ, Silhankova M, Betist MC, et al. A SNX3-dependent retromer pathway mediates retrograde transport of the Wnt sorting receptor Wntless and is required for Wnt secretion. Nature Cell Biology. 2011;13:914-923
  52. 52. Johannes L, Wunder C. The SNXy flavours of endosomal sorting. Nature Cell Biology. 2011;13:884-886
  53. 53. Zhang P, Zhou L, Pei C, Lin X, Yuan Z. Dysfunction of Wntless triggers the retrograde Golgi-to-ER transport of Wingless and induces ER stress. Scientific Reports. 2016;6:19418
  54. 54. Ashe HL, Briscoe J. The interpretation of morphogen gradients. Development. 2006;133:385-394
  55. 55. Korkut C, Ataman B, Ramachandran P, Ashley J, Barria R, Gherbesi N, et al. Trans-synaptic transmission of vesicular Wnt signals through Evi/Wntless. Cell. 2009;139:393-404
  56. 56. Gross JC, Chaudhary V, Bartscherer K, Boutros M. Active Wnt proteins are secreted on exosomes. Nature Cell Biology. 2012;14:1036-1045
  57. 57. Beckett K, Monier S, Palmer L, Alexandre C, Green H, Bonneil E, et al. Drosophila S2 cells secrete wingless on exosome-like vesicles but the wingless gradient forms independently of exosomes. Traffic. 2013;14:82-96
  58. 58. Neumann S, Coudreuse DY, van der Westhuyzen DR, Eckhardt ER, Korswagen HC, Schmitz G, et al. Mammalian Wnt3a is released on lipoprotein particles. Traffic. 2009;10:334-343
  59. 59. Bischoff M, Gradilla AC, Seijo I, Andres G, Rodriguez-Navas C, Gonzalez-Mendez L, et al. Cytonemes are required for the establishment of a normal Hedgehog morphogen gradient in Drosophila epithelia. Nature Cell Biology. 2013;15:1269-1281
  60. 60. Stanganello E, Scholpp S. Role of cytonemes in Wnt transport. Journal of Cell Science. 2016;129:665-672
  61. 61. Stanganello E, Hagemann AI, Mattes B, Sinner C, Meyen D, Weber S, et al. Filopodia-based Wnt transport during vertebrate tissue patterning. Nature Communications. 2015;6:5846
  62. 62. Mulligan KA, Fuerer C, Ching W, Fish M, Willert K, Nusse R. Secreted Wingless-interacting molecule (Swim) promotes long-range signaling by maintaining Wingless solubility. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:370-377
  63. 63. Kimelman D, Xu W. Beta-catenin destruction complex: Insights and questions from a structural perspective. Oncogene. 2006;25:7482-7491
  64. 64. Behrens J, Jerchow BA, Wurtele M, Grimm J, Asbrand C, Wirtz R, et al. Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta. Science. 1998;280:596-599
  65. 65. Yu N, Kakunda M, Pham V, Lill JR, Du P, Wongchenko M, et al. HSP105 recruits protein phosphatase 2A to dephosphorylate beta-catenin. Molecular and Cellular Biology. 2015;35:1390-1400
  66. 66. Aberle H, Bauer A, Stappert J, Kispert A, Kemler R. Beta-catenin is a target for the ubiquitin-proteasome pathway. The EMBO Journal. 1997;16:3797-3804
  67. 67. Bilic J, Huang YL, Davidson G, Zimmermann T, Cruciat CM, Bienz M, et al. Wnt induces LRP6 signalosomes and promotes dishevelled-dependent LRP6 phosphorylation. Science. 2007;316:1619-1622
  68. 68. McGough IJ, Vincent JP. APC moonlights to prevent Wnt signalosome assembly. Developmental Cell. 2018;44:535-537
  69. 69. Stadeli R, Hoffmans R, Basler K. Transcription under the control of nuclear Arm/beta-catenin. Current Biology. 2006;16:R378-R385
  70. 70. Barker N, Clevers H. Mining the Wnt pathway for cancer therapeutics. Nature Reviews. Drug Discovery. 2006;5:997-1014
  71. 71. Zeng X, Huang H, Tamai K, Zhang X, Harada Y, Yokota C, et al. Initiation of Wnt signaling: Control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development. 2008;135:367-375
  72. 72. Tacchelly-Benites O, Wang Z, Yang E, Benchabane H. Axin phosphorylation in both Wnt-off and Wnt-on states requires the tumor suppressor APC. PLoS Genetics. 2018;14:e1007178
  73. 73. Rubinfeld B, Albert I, Porfiri E, Fiol C, Munemitsu S, Polakis P. Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science. 1996;272:1023-1026
  74. 74. Neufeld KL, Zhang F, Cullen BR, White RL. APC-mediated downregulation of beta-catenin activity involves nuclear sequestration and nuclear export. EMBO Reports. 2000;1:519-523
  75. 75. Sierra J, Yoshida T, Joazeiro CA, Jones KA. The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes & Development. 2006;20:586-600
  76. 76. Aoki K, Taketo MM. Adenomatous polyposis coli (APC): A multi-functional tumor suppressor gene. Journal of Cell Science. 2007;120:3327-3335
  77. 77. Takacs CM, Baird JR, Hughes EG, Kent SS, Benchabane H, Paik R, et al. Dual positive and negative regulation of wingless signaling by adenomatous polyposis coli. Science. 2008;319:333-336
  78. 78. Su Y, Fu C, Ishikawa S, Stella A, Kojima M, Shitoh K, et al. APC is essential for targeting phosphorylated beta-catenin to the SCFbeta-TrCP ubiquitin ligase. Molecular Cell. 2008;32:652-661
  79. 79. Saito-Diaz K, Benchabane H, Tiwari A, Tian A, Li B, Thompson JJ, et al. APC inhibits ligand-independent Wnt signaling by the clathrin endocytic pathway. Developmental Cell. 2018;44:566-581.e8
  80. 80. Amit S, Hatzubai A, Birman Y, Andersen JS, Ben-Shushan E, Mann M, et al. Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: A molecular switch for the Wnt pathway. Genes & Development. 2002;16:1066-1076
  81. 81. Liu C, Li Y, Semenov M, Han C, Baeg GH, Tan Y, et al. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002;108:837-847
  82. 82. Wu G, Xu G, Schulman BA, Jeffrey PD, Harper JW, Pavletich NP. Structure of a beta-TrCP1-Skp1-beta-catenin complex: Destruction motif binding and lysine specificity of the SCF(beta-TrCP1) ubiquitin ligase. Molecular Cell. 2003;11:1445-1456
  83. 83. Kitagawa M, Hatakeyama S, Shirane M, Matsumoto M, Ishida N, Hattori K, et al. An F-box protein, FWD1, mediates ubiquitin-dependent proteolysis of beta-catenin. The EMBO Journal. 1999;18:2401-2410
  84. 84. Virshup DM, Shenolikar S. From promiscuity to precision: Protein phosphatases get a makeover. Molecular Cell. 2009;33:537-545
  85. 85. Li X, Yost HJ, Virshup DM, Seeling JM. Protein phosphatase 2A and its B56 regulatory subunit inhibit Wnt signaling in Xenopus. The EMBO Journal. 2001;20:4122-4131
  86. 86. Mitra A, Menezes ME, Pannell LK, Mulekar MS, Honkanen RE, Shevde LA, et al. DNAJB6 chaperones PP2A mediated dephosphorylation of GSK3beta to downregulate beta-catenin transcription target, osteopontin. Oncogene. 2012;31:4472-4483
  87. 87. Seeling JM, Miller JR, Gil R, Moon RT, White R, Virshup DM. Regulation of beta-catenin signaling by the B56 subunit of protein phosphatase 2A. Science. 1999;283:2089-2091
  88. 88. Eichhorn PJ, Creyghton MP, Bernards R. Protein phosphatase 2A regulatory subunits and cancer. Biochimica et Biophysica Acta. 2009;1795:1-15
  89. 89. Yang J, Wu J, Tan C, Klein PS. PP2A:B56epsilon is required for Wnt/beta-catenin signaling during embryonic development. Development. 2003;130:5569-5578
  90. 90. Yamamoto H, Hinoi T, Michiue T, Fukui A, Usui H, Janssens V, et al. Inhibition of the Wnt signaling pathway by the PR61 subunit of protein phosphatase 2A. The Journal of Biological Chemistry. 2001;276:26875-26882
  91. 91. Zhang W, Yang J, Liu Y, Chen X, Yu T, Jia J, et al. PR55 alpha, a regulatory subunit of PP2A, specifically regulates PP2A-mediated beta-catenin dephosphorylation. The Journal of Biological Chemistry. 2009;284:22649-22656
  92. 92. Hein AL, Seshacharyulu P, Rachagani S, Sheinin YM, Ouellette MM, Ponnusamy MP, et al. PR55alpha subunit of protein phosphatase 2A supports the tumorigenic and metastatic potential of pancreatic cancer cells by sustaining hyperactive oncogenic signaling. Cancer Research. 2016;76:2243-2253
  93. 93. Strovel ET, Wu D, Sussman DJ. Protein phosphatase 2Calpha dephosphorylates axin and activates LEF-1-dependent transcription. The Journal of Biological Chemistry. 2000;275:2399-2403
  94. 94. Vinyoles M, Del Valle-Perez B, Curto J, Padilla M, Villarroel A, Yang J, et al. Activation of CK1varepsilon by PP2A/PR61varepsilon is required for the initiation of Wnt signaling. Oncogene. 2017;36:429-438
  95. 95. Schlessinger K, Hall A, Tolwinski N. Wnt signaling pathways meet Rho GTPases. Genes & Development. 2009;23:265-277
  96. 96. Tepass U. The apical polarity protein network in Drosophila epithelial cells: Regulation of polarity, junctions, morphogenesis, cell growth, and survival. Annual Review of Cell and Developmental Biology. 2012;28:655-685
  97. 97. Zallen JA. Planar polarity and tissue morphogenesis. Cell. 2007;129:1051-1063
  98. 98. Casal J, Struhl G, Lawrence PA. Developmental compartments and planar polarity in Drosophila. Current Biology. 2002;12:1189-1198
  99. 99. Zeidler MP, Perrimon N, Strutt DI. Multiple roles for four-jointed in planar polarity and limb patterning. Developmental Biology. 2000;228:181-196
  100. 100. Simons M, Mlodzik M. Planar cell polarity signaling: From fly development to human disease. Annual Review of Genetics. 2008;42:517-540
  101. 101. Strutt H, Gamage J, Strutt D. Robust asymmetric localization of planar polarity proteins is associated with organization into signalosome-like domains of variable stoichiometry. Cell Reports. 2016;17:2660-2671
  102. 102. VanderVorst K, Hatakeyama J, Berg A, Lee H, Carraway III KL. Cellular and molecular mechanisms underlying planar cell polarity pathway contributions to cancer malignancy. Seminars in Cell & Developmental Biology. 2018;81:78-87
  103. 103. Daulat AM, Borg JP. Wnt/planar cell polarity signaling: New opportunities for cancer treatment. Trends in Cancer. 2017;3:113-125
  104. 104. Kuhl M, Sheldahl LC, Malbon CC, Moon RT. Ca(2+)/calmodulin-dependent protein kinase II is stimulated by Wnt and Frizzled homologs and promotes ventral cell fates in Xenopus. The Journal of Biological Chemistry. 2000;275:12701-12711
  105. 105. Pereira C, Schaer DJ, Bachli EB, Kurrer MO, Schoedon G. Wnt5A/CaMKII signaling contributes to the inflammatory response of macrophages and is a target for the antiinflammatory action of activated protein C and interleukin-10. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28:504-510
  106. 106. Blumenthal A, Ehlers S, Lauber J, Buer J, Lange C, Goldmann T, et al. The Wingless homolog WNT5A and its receptor Frizzled-5 regulate inflammatory responses of human mononuclear cells induced by microbial stimulation. Blood. 2006;108:965-973
  107. 107. Nishita M, Enomoto M, Yamagata K, Minami Y. Cell/tissue-tropic functions of Wnt5a signaling in normal and cancer cells. Trends in Cell Biology. 2010;20:346-354
  108. 108. De A. Wnt/Ca2+ signaling pathway: A brief overview. Acta Biochimica et Biophysica Sinica. 2011;43:745-756
  109. 109. Gong B, Shen W, Xiao W, Meng Y, Meng A, Jia S. The Sec14-like phosphatidylinositol transfer proteins Sec14l3/SEC14L2 act as GTPase proteins to mediate Wnt/Ca(2+) signaling. eLife 2017;6:e26362. DOI: 10.7554/eLife.26362
  110. 110. Wang H, Lee Y, Malbon CC. PDE6 is an effector for the Wnt/Ca2+/cGMP-signalling pathway in development. Biochemical Society Transactions. 2004;32:792-796
  111. 111. Kantaputra P, Sripathomsawat W. WNT10A and isolated hypodontia. American Journal of Medical Genetics. Part A. 2011;155a:1119-1122
  112. 112. Yu P, Yang W, Han D, Wang X, Guo S, Li J, et al. Mutations in WNT10B are identified in individuals with oligodontia. American Journal of Human Genetics. 2016;99:195-201
  113. 113. Massink MP, Creton MA, Spanevello F, Fennis WM, Cune MS, Savelberg SM, et al. Loss-of-function mutations in the WNT co-receptor LRP6 cause autosomal-dominant oligodontia. American Journal of Human Genetics. 2015;97:621-626
  114. 114. Szenker-Ravi E, Altunoglu U, Leushacke M, Bosso-Lefevre C, Khatoo M, Thi Tran H, et al. RSPO2 inhibition of RNF43 and ZNRF3 governs limb development independently of LGR4/5/6. Nature. 2018;557:564-569
  115. 115. Niemann S, Zhao C, Pascu F, Stahl U, Aulepp U, Niswander L, et al. Homozygous WNT3 mutation causes tetra-amelia in a large consanguineous family. American Journal of Human Genetics. 2004;74:558-563
  116. 116. Woods CG, Stricker S, Seemann P, Stern R, Cox J, Sherridan E, et al. Mutations in WNT7A cause a range of limb malformations, including Fuhrmann syndrome and Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome. American Journal of Human Genetics. 2006;79:402-408
  117. 117. Ugur SA, Tolun A. Homozygous WNT10b mutation and complex inheritance in split-hand/foot malformation. Human Molecular Genetics. 2008;17:2644-2653
  118. 118. Bain MD, Winter RM, Burn J. Robinow syndrome without mesomelic 'brachymelia': A report of five cases. Journal of Medical Genetics. 1986;23:350-354
  119. 119. Li Y, Dudley AT. Noncanonical frizzled signaling regulates cell polarity of growth plate chondrocytes. Development. 2009;136:1083-1092
  120. 120. Yamaguchi TP, Bradley A, McMahon AP, Jones S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development. 1999;126:1211-1223
  121. 121. Estrada K, Styrkarsdottir U, Evangelou E, Hsu YH, Duncan EL, Ntzani EE, et al. Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fractureNature Genetics. 2012;44:491-501
  122. 122. Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Benichou O, Scopelliti D, 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
  123. 123. Van Hul E, Gram J, Bollerslev J, Van Wesenbeeck L, Mathysen D, Andersen PE, et al. Localization of the gene causing autosomal dominant osteopetrosis type I to chromosome 11q12-13. Journal of Bone and Mineral Research. 2002;17:1111-1117
  124. 124. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, et al. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513-523
  125. 125. Pyott SM, Tran TT, Leistritz DF, Pepin MG, Mendelsohn NJ, Temme RT, et al. WNT1 mutations in families affected by moderately severe and progressive recessive osteogenesis imperfecta. American Journal of Human Genetics. 2013;92:590-597
  126. 126. Keupp K, Beleggia F, Kayserili H, Barnes AM, Steiner M, Semler O, et al. Mutations in WNT1 cause different forms of bone fragility. American Journal of Human Genetics. 2013;92:565-574
  127. 127. Laine CM, Joeng KS, Campeau PM, Kiviranta R, Tarkkonen K, Grover M, et al. WNT1 mutations in early-onset osteoporosis and osteogenesis imperfecta. The New England Journal of Medicine. 2013;368:1809-1816
  128. 128. Styrkarsdottir U, Thorleifsson G, Sulem P, Gudbjartsson DF, Sigurdsson A, Jonasdottir A, et al. Nonsense mutation in the LGR4 gene is associated with several human diseases and other traits. Nature. 2013;497:517-520
  129. 129. Kiper POS, Saito H, Gori F, Unger S, Hesse E, Yamana K, et al. Cortical-bone fragility—Insights from sFRP4 deficiency in Pyle's disease. The New England Journal of Medicine. 2016;374:2553-2562
  130. 130. Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, 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
  131. 131. Nguyen TV, Livshits G, Center JR, Yakovenko K, Eisman JA. Genetic determination of bone mineral density: Evidence for a major gene. The Journal of Clinical Endocrinology and Metabolism. 2003;88:3614-3620
  132. 132. Leupin O, Piters E, Halleux C, Hu S, Kramer I, Morvan F, et al. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. The Journal of Biological Chemistry. 2011;286:19489-19500
  133. 133. Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, et al. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. Journal of Medical Genetics. 2002;39:91-97
  134. 134. Brunkow ME, Gardner JC, Van Ness J, Paeper BW, Kovacevich BR, Proll S, et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. American Journal of Human Genetics. 2001;68:577-589
  135. 135. Kim SJ, Bieganski T, Sohn YB, Kozlowski K, Semenov M, Okamoto N, et al. Identification of signal peptide domain SOST mutations in autosomal dominant craniodiaphyseal dysplasia. Human Genetics. 2011;129:497-502
  136. 136. Chang MK, Kramer I, Huber T, Kinzel B, Guth-Gundel S, Leupin O, et al. Disruption of Lrp4 function by genetic deletion or pharmacological blockade increases bone mass and serum sclerostin levels. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E5187-E5195
  137. 137. Padhi D, Jang G, Stouch B, Fang L, Posvar E. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. Journal of Bone and Mineral Research. 2011;26:19-26
  138. 138. Person AD, Beiraghi S, Sieben CM, Hermanson S, Neumann AN, Robu ME, et al. WNT5A mutations in patients with autosomal dominant Robinow syndrome. Developmental Dynamics. 2010;239:327-337
  139. 139. Adaimy L, Chouery E, Megarbane H, Mroueh S, Delague V, Nicolas E, et al. Mutation in WNT10A is associated with an autosomal recessive ectodermal dysplasia: The odonto-onycho-dermal dysplasia. American Journal of Human Genetics. 2007;81:821-828
  140. 140. Bohring A, Stamm T, Spaich C, Haase C, Spree K, Hehr U, et al. WNT10A mutations are a frequent cause of a broad spectrum of ectodermal dysplasias with sex-biased manifestation pattern in heterozygotes. American Journal of Human Genetics. 2009;85:97-105
  141. 141. White JJ, Mazzeu JF, Coban-Akdemir Z, Bayram Y, Bahrambeigi V, Hoischen A, et al. WNT signaling perturbations underlie the genetic heterogeneity of Robinow syndrome. American Journal of Human Genetics. 2018;102:27-43
  142. 142. Frojmark AS, Schuster J, Sobol M, Entesarian M, Kilander MBC, Gabrikova D, et al. Mutations in frizzled 6 cause isolated autosomal-recessive nail dysplasia. American Journal of Human Genetics. 2011;88:852-860
  143. 143. Li Y, Pawlik B, Elcioglu N, Aglan M, Kayserili H, Yigit G, et al. LRP4 mutations alter Wnt/beta-catenin signaling and cause limb and kidney malformations in Cenani-Lenz syndrome. American Journal of Human Genetics. 2010;86:696-706
  144. 144. Oldridge M, Fortuna AM, Maringa M, Propping P, Mansour S, Pollitt C, et al. Dominant mutations in ROR2, encoding an orphan receptor tyrosine kinase, cause brachydactyly type B. Nature Genetics. 2000;24:275-278
  145. 145. van Bokhoven H, Celli J, Kayserili H, van Beusekom E, Balci S, Brussel W, et al. Mutation of the gene encoding the ROR2 tyrosine kinase causes autosomal recessive Robinow syndrome. Nature Genetics. 2000;25:423-426
  146. 146. Afzal AR, Rajab A, Fenske CD, Oldridge M, Elanko N, Ternes-Pereira E, et al. Recessive Robinow syndrome, allelic to dominant brachydactyly type B, is caused by mutation of ROR2. Nature Genetics. 2000;25:419-422
  147. 147. Pilia G, Hughes-Benzie RM, MacKenzie A, Baybayan P, Chen EY, Huber R, et al. Mutations in GPC3, a glypican gene, cause the Simpson-Golabi-Behmel overgrowth syndrome. Nature Genetics. 1996;12:241-247
  148. 148. Campos-Xavier AB, Martinet D, Bateman J, Belluoccio D, Rowley L, Tan TY, et al. Mutations in the heparan-sulfate proteoglycan glypican 6 (GPC6) impair endochondral ossification and cause recessive omodysplasia. American Journal of Human Genetics. 2009;84:760-770
  149. 149. Tan-Sindhunata MB, Mathijssen IB, Smit M, Baas F, de Vries JI, van der Voorn JP, et al. Identification of a Dutch founder mutation in MUSK causing fetal akinesia deformation sequence. European Journal of Human Genetics. 2015;23:1151-1157
  150. 150. Loughlin J, Dowling B, Mustafa Z, Southam L, Chapman K. Refined linkage mapping of a hip osteoarthritis susceptibility locus on chromosome 2q. Rheumatology (Oxford). 2002;41:955-956
  151. 151. White J, Mazzeu JF, Hoischen A, Jhangiani SN, Gambin T, Alcino MC, et al. DVL1 frameshift mutations clustering in the penultimate exon cause autosomal-dominant Robinow syndrome. American Journal of Human Genetics. 2015;96:612-622
  152. 152. White JJ, Mazzeu JF, Hoischen A, Bayram Y, Withers M, Gezdirici A, et al. DVL3 alleles resulting in a −1 frameshift of the last exon mediate autosomal-dominant Robinow syndrome. American Journal of Human Genetics. 2016;98:553-561
  153. 153. Parma P, Radi O, Vidal V, Chaboissier MC, Dellambra E, Valentini S, et al. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nature Genetics. 2006;38:1304-1309
  154. 154. Blaydon DC, Ishii Y, O'Toole EA, Unsworth HC, Teh MT, Ruschendorf F, et al. The gene encoding R-spondin 4 (RSPO4), a secreted protein implicated in Wnt signaling, is mutated in inherited anonychia. Nature Genetics. 2006;38:1245-1247

Written By

Yanqin Lu and Jinxiang Han

Submitted: 04 June 2018 Reviewed: 22 August 2018 Published: 05 November 2018