1,499
Views
10
CrossRef citations to date
0
Altmetric
Research Paper

Intrinsic disorder in spondins and some of their interacting partners

, , , , &
Article: e1255295 | Received 10 Oct 2016, Accepted 27 Oct 2016, Published online: 15 Dec 2016

ABSTRACT

Spondins, which are proteins that inhibit and promote adherence of embryonic cells so as to aid axonal growth are part of the thrombospondin-1 family. Spondins function in several important biological processes, such as apoptosis, angiogenesis, etc. Spondins constitute a thrombospondin subfamily that includes F-spondin, a protein that interacts with Aβ precursor protein and inhibits its proteolytic processing; R-spondin, a 4-membered group of proteins that regulates Wnt pathway and have other functions, such as regulation of kidney proliferation, induction of epithelial proliferation, the tumor suppressant action; M-spondin that mediates mechanical linkage between the muscles and apodemes; and the SCO-spondin, a protein important for neuronal development. In this study, we investigated intrinsic disorder status of human spondins and their interacting partners, such as members of the LRP family, LGR family, Frizzled family, and several other binding partners in order to establish the existence and importance of disordered regions in spondins and their interacting partners by conducting a detailed analysis of their sequences, finding disordered regions, and establishing a correlation between their structure and biological functions.

Introduction

Until recently, it was believed that all proteins have well-defined 3 dimensional structures crucial for their unique biological functions. However, with recent advances in protein science, it has been proven that some genes in the eukaryotic genomes encode entire proteins or large segments of proteins that lack a well-structured 3-dimensional fold.Citation1-14 These intrinsically disordered proteins (IDPs) and intrinsically disordered protein regions (IDPRs) have important biological roles and are crucial for many cellular processes, such as signaling, transcriptional and translational activities.Citation3,4,8,10-12,15-20 Some IDPRs function as linkers between ordered domains, many of them frequently serve as sites of posttranslational modifications, or as regions affected by disease-related mutations, gene truncations or translocations.Citation21-28

Thrombospondins (TSPs) are multimeric multidomain secreted glycoproteins with antiangiogenic functions found in the extracellular matrix (ECM).Citation29,30 This family of the matricellular glycoproteins includes 5 different members, namely, TSP1, TSP2, TSP3, TSP4, and TSP5, which is further subdivided to groups A (TSP1 and TSP2) and B (TSP3, TSP4, and TSP5). Just like many other ECM proteins, TSPs are large modular proteins (whose length in humans ranges from 757 to 1,170 residues) that contain series of repeated domains of different type. For example, members of the group A, TSP1 (UniProt ID P07996) TSP2 (UniProt ID P35442), being the longest members of the TSP family with 1,170 and 1,171 residues, respectively, contain an N-terminal laminin G-like domain, a VWFC domain, 3 TSP1 repeats, 3 epidermal growth factor-like repeats (EGF-like, also known as TSP2 repeats), 8 aspartic acid-rich TSP3 repeats, and a TSP C-terminal domain. The group B members, TSP3 (UniProt ID: P49746), TSP4 (UniProt ID: P35443), and TSP5 (also known as cartilage oligomeric matrix protein (COMP), UniProt ID: P49747), are noticeably shorter (there are 956, 961, and 757 residues in TSP3, TSP4, and COMP, respectively) and have domain structure that is different from the domain organization of the group A TSPs. Namely, they contain unique N-terminal laminin G-like domain, lack the VWFC domain and TSP1 repeats, contain 4 copies of EGF-like domains, and 8 copies of the TSP3 repeats. Furthermore, group A and B members have different oligomeric structures, with the group A TSPs being assembled as homo-trimer and the group B TSPs existing as homo-pentamers.Citation31 These modular proteins act by bringing together cytokines, growth factors, other matrix components, membrane receptors, and extracellular proteases.Citation29-31

Spondins are a group of proteins from the thrombospondin superfamily. They are found in eukaryotic organisms and are grouped into different families, such as the R-spondin, the subcommissural organ (SCO)-spondin, the M-spondin (mindin), and the F-spondin. Spondins are engaged in various vital biological functions, such as regulators of Wnt signaling (R-spondins),Citation32-40 regulation of the developing skeleton, limb formation, and the maintenance of adult bone mass (R-spondins),Citation39,41 regulation of stem cells (R-spondin),Citation37,42,43 neuron/glia interaction and neuronal differentiation and development (SCO-spondin),Citation44,45 interaction with the β-amyloid precursor protein (APP) and its controlled proteolysis (F-spondin),Citation46,47 regulation of the accurate path-finding of embryonic axons (F-spondin),Citation48 and promotion of the neurite outgrowth and inhibition of the angiogenesis (F-spondin and mindin).Citation48 Being the members of the TSP family, these proteins have complex modular structures. Multifunctionality of these proteins, their ability to interact with numerous partners, their modular structure, and the presence of multiple aspartic acid-rich repeats suggest that spondins might belong to the family of hybrid proteins containing ordered domains and functional IDPRs.

The goal of this study was to establish the existence and importance of IDPRs in spondins and their interacting partners in human, and to conduct a detailed analysis of their sequences, find disordered regions, and establish a correlation between their structure and biological functions. Although most of the entities analyzed in this study are extracellular proteins possessing signal peptides that are removed following passage through the membrane, these secretion signals were included in the analysis. In other words, we analyzed entire protein sequences as reported in the corresponding UniProt entries.

R-spondin family

In human, there are 4 R-spondin proteins, which are secreted agonist of the canonical Wnt/β-catenin signaling pathway.Citation32-40 These proteins have molecular masses of approximately 35 kDa and are characterized by the presence of 2 N-terminal furin-like repeats, which are necessary for Wnt signaling. R-spondins can enhance responses to low-dose Wnt protein and also serves as activators of a canonical Wnt signaling pathway, where they act as ligands for the LGR4-6 receptors. Being potent stimulators of adult stem cells proliferation in vivo and in vitro, R-spondins have strong potential for therapeutic use in regenerative medicine.

R-spondin 1

R-spondin 1 is also referred to as Roof plate-specific spondin-1. This protein is encoded by RSPO1 gene located at the position 1p34.3 of the chromosome one, and is present as 3 isoforms in humans, a full-length canonical form (or isoform #1; UniProt ID: Q2MKA7-1) with sequence length of 263 residues, an isoform #2 (UniProt ID: Q2MKA7-2) that is characterized by MRLGLCVVALVLSWTHLTISSRGIKGKRQRRI → MIFRV substitution within the N-terminal region (residues 1–32), and an isoform #3 (UniProt ID: Q2MKA7-3) that misses residues 146–208. There are 5 functional domains in the canonical form of this protein, a signal peptide sequence at the N-terminus for secretion (residues 1–20), 2 cysteine-rich furin-like repeat domains (domains Fu1 and Fu2, residues 34–85 and 91–135, respectively), a TSP1 repeat domain (TSR, residues 147–207), and a basic amino acid-rich (BR) domain at the C-terminus (residues 208–263). Therefore, although alternative splicing does not affect the R-spondin 1 (Rspo1) N-terminal region with Fu1 and Fu2 domains, entire TSP type-1 domain is absent in its isoform #3, suggesting that this Rspo1 proteoform cannot interact with heparin sulfate proteoglycans (see below), and a signal peptide is removed in isoform #2, suggesting that this proteoform cannot be exported.

Rspo1 is known to strongly promote proliferation of the Wnt-dependent intestinal-crypt stem cell compartment,Citation49,50 with this activity being primarily attributed to the Fu1 and Fu2 domains of this protein,Citation51 which are involved in direct physical interaction with the members of the leucine-rich repeat-containing G protein-coupled receptors 4–6 (LGR4–LGR6).Citation52-54 High affinity binding of R-spondins to LGR5 (as well as its homologs LGR4 and LGR6) mediates R-spondin contribution to the canonical Wnt pathway.Citation52-54 The TSR domain is responsible for interaction with heparin sulfate proteoglycans (HSPGs).Citation55

Besides interaction with the LGR4-6 receptors, Rspo1 regulates the canonical Wnt/β-catenin dependent pathway and non-canonical Wnt signaling by acting as an inhibitor of a transmembrane E3 ubiquitin ligase, zinc and ring finger 3 (ZnRF3), and the E3 ubiquitin-protein ligase RING finger protein 43 (RNF43), which are critical negative feedback regulators of the Wnt pathway, which function through promoting ubiquitination and degradation of Wnt receptors, and thereby playing a crucial role in the control of cancer development.Citation38 Therefore, Rspo1 serves as an important member of the R-spondin-ZnRF3/RNF43 signaling module that plays a crucial role in the control of Wnt signaling.Citation38 In fact, RSpo1 can simultaneously bind to the extracellular domains of ZnRF3/RNF43 and LGR4/5.Citation56 Formation of this complex induces auto-ubiquitination and membrane clearance of ZnRF3/RNF43, leading to the increased levels of frizzled (Fzd). This Rspo1-based regulation of the Fzd turnover allows R-spondin to control both Wnt/β-catenin and Wnt/PCP signaling pathways. The formation of the tripartite ZnRF3/RNF43-Rspo1-LGR4/5 complex is explained by the fact that Rspo1 utilizes its Fu2 domain for interaction with LGR4/5 and binds to ZnRF3/RNF43 via its Fu1 domain.Citation38 This tripartite complex represents a complex functional state of Rspo1, with LGR4/5 and ZnRF3/RNF43 acting as the engagement and the efficacy receptors for Rspo1, respectively.Citation38 Furthermore, Rspo1 can be engaged in the physical binary interactions with the extracellular domains of frizzled-8 (Fzd8)Citation57 and low-density lipoprotein receptor-related protein 6 (LRP6),Citation58 which serves as the Wnt co-receptor, without formation of a ternary complex with these 2 proteins.Citation57 Furthermore, Rspo1 partake in various biological processes including canonical male meiosis, positive regulation of protein phosphorylation, regulation of gene expression, regulation of male germ cell proliferation, regulation of receptor internalization.Citation36,39

Structural information is available for the Fu1/Fu2-containing region of Rspo1 (residues 40–127; e.g., see PDB ID: 4BSP).Citation59 Although longer regions of Rspo1 are typically used in crystallization studies (residues 31–146), no coordinates have been determined for their N- (residues 31–39) and C-terminal regions (residues 128–146). Recent structural analysis revealed that in its unbound form, the Fu1/Fu2 region of Rspo1 is characterized by the non-equivalent Fu domains, where the Fu1 domain has 3 cysteine-knotted β-hairpins, whereas there are 2 such hairpins in the Fu2 domain, with the C-terminal hairpin being missing from the crystal structure, likely due to the high conformational flexibility.Citation59 Unbound Fu1/Fu2 region is organized in such a way that the sets of Fu1 and Fu2 β-hairpins are oriented at ∼90° to each other, with each Fu domain forming a leaflet consisting of 3 (Fu1) or 2 (Fu2) disulfide bond-connected β-hairpins.Citation59 However, binding to the LGR5 induced noticeable changes in the Fu1/Fu2 region, especially in its Ru2 part. shows that the overall shape of the LGR5-bound TSR was flattened because the 2 sets of β-hairpins from the Ru1 and Ru2 domains were aligned due to a twist caused by the rotation around the TSR longitudinal axis. Furthermore, the C-terminal part of the Fu1/Fu2 region became visible upon its binding to LGR-5, indicating dramatic binding-induced reduction in the conformational flexibility of this region.Citation59

Figure 1. Structural characterization of the Fu1/Fu2-containing region of Rspo1 in its unbound (PDB ID: 4BSP-A, red ribbon and 4BSO-A, blue ribbon) and bound forms (PDB ID: 4BSR-C, yellow ribbon and 4BSR-D, cyan ribbon).Citation59 Results of multiple structure alignment are shown in the middle of the plot, whereas individual chains are located around. To show location of each individual chain within the aligned structural ensemble, the remaining structures are made transparent. Multiple structural alignment was conducted using the MultiProt tool (http://bioinfo3d.cs.tau.ac.il/MultiProt/).Citation198 Resulting structures are drawn using the visual molecular dynamic tool VMD.Citation199

Figure 1. Structural characterization of the Fu1/Fu2-containing region of Rspo1 in its unbound (PDB ID: 4BSP-A, red ribbon and 4BSO-A, blue ribbon) and bound forms (PDB ID: 4BSR-C, yellow ribbon and 4BSR-D, cyan ribbon).Citation59 Results of multiple structure alignment are shown in the middle of the plot, whereas individual chains are located around. To show location of each individual chain within the aligned structural ensemble, the remaining structures are made transparent. Multiple structural alignment was conducted using the MultiProt tool (http://bioinfo3d.cs.tau.ac.il/MultiProt/).Citation198 Resulting structures are drawn using the visual molecular dynamic tool VMD.Citation199

represents the results of intrinsic disorder analysis in human Rspo1 (UniProt ID: Q2MKA7) and shows that this protein is expected to have several functional disordered regions. For example, a linker connecting signaling peptide to the rest of Rspo1 is disordered, likely to facilitate the accessibility of site of a proteolytic attack leading to the release of the signaling peptide and generating mature Rspo1 (residues 21–263). Although both Fu1 and Fu2 domains are mostly ordered, both a TSR and a BR domains are predicted to be mostly disordered. BR domain is predicted to have 2 potential disorder-based protein binding sites identified by the ANCHOR algorithm, that acts on the hypothesis that long regions of disorder include localized potential binding sites which are not capable of folding on their own due to not being able to form enough favorable intrachain interactions, but can obtain the energy to stabilize ordered conformation via interaction with a globular protein partner.Citation60,61 Furthermore, it is known that human Rspo1 can be subjected to several posttranslational modifications (PTMs), with sites of these PTMs being located within the IDPRs of this protein. For example, it was shown that the N-glycosylation of human Rspo1 at Asn137 (which is a residue located at the C-terminus of the Fu2 domain of this protein and is predicted to be disordered, being characterized by the mean disorder score of 0.53) is required for the efficient secretion and stability of this protein, but does not play a role in interaction of Rspo1 with heparin sulfate proteoglycans.Citation62 A single phosphorylation site (Thr253) is located within the disordered C-tail of human Rspo1. Since this site is positioned within one of the MoRFs, it is likely that it plays a role in regulation of the disorder-based interactivity of this protein.

Figure 2. Multifactorial computational disorder analysis of in human Rspo1. (A) Intrinsic disorder profile of Rspo1 (UniProt ID: Q2MKA7) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. (B) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo1 by the D2P2 database (http://d2p2.pro/).Citation200 Here, complementary disorder evaluations together with some disorder-related functional information are shown. To this end, the D2P2 database uses outputs of IUPred,Citation179 PONDR® VLXT,Citation201 PrDOS,Citation202 PONDR® VSL2B,Citation183,184 PV2,Citation200 and ESpritz.Citation180 Positions of disorder-based interactions sites (MoRFs) and sites of curated posttranslational modifications are also shown.

Figure 2. Multifactorial computational disorder analysis of in human Rspo1. (A) Intrinsic disorder profile of Rspo1 (UniProt ID: Q2MKA7) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. (B) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo1 by the D2P2 database (http://d2p2.pro/).Citation200 Here, complementary disorder evaluations together with some disorder-related functional information are shown. To this end, the D2P2 database uses outputs of IUPred,Citation179 PONDR® VLXT,Citation201 PrDOS,Citation202 PONDR® VSL2B,Citation183,184 PV2,Citation200 and ESpritz.Citation180 Positions of disorder-based interactions sites (MoRFs) and sites of curated posttranslational modifications are also shown.

shows the Rspo1 interactome evaluated by Search Tool for the Retrieval of Interacting Genes; STRING, http://string-db.org/, which generates a network of predicted associations based on predicted and experimentally-validated information on the interaction partners of a protein of interest.Citation63 In the corresponding network, the nodes correspond to proteins, whereas the edges show predicted or known functional associations. Seven types of evidence are used to build the corresponding network, where they are indicated by the differently colored lines: a green line represents neighborhood evidence; a red line - the presence of fusion evidence; a purple line - experimental evidence; a blue line – co-occurrence evidence; a light blue line - database evidence; a yellow line – text mining evidence; and a black line – co-expression evidence.Citation63 In our analysis, the most stringent criteria were used for selection of interacting proteins by choosing the highest cut-off of 0.9 as the minimal required confidence level. According to these criteria, human Rspo1 interacts with other members of the R-spondin family (Rspo2, Rspo3, and Rspo4), leucine-rich repeat containing G protein-coupled receptors 4, 5, and 6 (LGR4, LGR5, and LGR6), low density lipoprotein receptor-related protein 6 (LRP6), zinc and ring finger 3 (ZnRF3), ring finger protein 43 (RNF43), ribosomal protein S27a (RPS27A), ubiquitin A-52 residue ribosomal protein fusion product 1 (UBA52), ubiquitin B (UBB), ubiquitin C (UBC), dickkopf 1 homolog (DKK1), wingless-type MMTV integration site family, member 4 (WNT4), SRY (sex determining region Y)-box 9 (SOX9), and forkhead box L2 (FOXL2) (see ).

Figure 3. Interactome generated by Search Tool for the Retrieval of Interacting Genes; STRING, http://string-db.org/ for: (A) Human Rspo1 (UniProt ID: Q2MKA7); (B) Human Rspo2 (UniProt ID: Q6UXX9); (C) Human Rspo3 (UniProt ID: Q9BXY4); and (D) Human Rspo4 (UniProt ID: Q2I0M5).

Figure 3. Interactome generated by Search Tool for the Retrieval of Interacting Genes; STRING, http://string-db.org/ for: (A) Human Rspo1 (UniProt ID: Q2MKA7); (B) Human Rspo2 (UniProt ID: Q6UXX9); (C) Human Rspo3 (UniProt ID: Q9BXY4); and (D) Human Rspo4 (UniProt ID: Q2I0M5).

R-spondins 2, 3, and 4

Rspo2. Protein R-spondin 2 (Rspo2) is encoded by the RSPO2 gene located at the 8q23.1 position of the chromosome eight. Rspo2 is also known as roof plate-specific spondin-2. Similar to Rspo1, this protein is involved in a wide range of biological processes, such as bone mineralization, dopaminergic neuron differentiation, embryonic forelimb morphogenesis, embryonic hind-limb morphogenesis, epithelial tube branching involved in lung morphogenesis, lung growth, negative regulation of odontogenesis of dentin-containing tooth, osteoblast differentiation, and trachea cartilage morphogenesis.Citation36 Rspo2 also plays key role in regulation of the canonical Wnt pathway by passing signal into a cell through cell surface receptors, causing an accumulation of β-catenin in the cytoplasm and its eventual translocation into the nucleus.Citation35

Rspo2 has a signal peptide (residues 1–21), 2 Fu domains (residues 39–89 and 90–134, respectively), a TSR1 domain (residues 144–204), and a C-terminal basic amino acid rich domain (residues 205–243). Rspo2 (UniProt ID: Q6UXX9) has 3 proteoforms generated by alternative splicing. The full-length canonical isoform #1 (UniProt ID: Q6UXX9-1) has 243 residues. Isoform #2 (UniProt ID: Q6UXX9-2) is characterized by missing entire N-terminal domain (residues 1–67), whereas in the isoform #3 (UniProt ID: Q6UXX9-3) the 32–95 region is shrunk to one glycine residue and a residue 143 is missing.

Rspo3. R-spondin 3 protein (Rspo3) is encoded by the RSPO3 gene located at the 6q22.33 position of the chromosome six. RSPO3 has a 5-exon organization defining the presence of alternatively spliced isoforms and the domain structure of the protein. It is another member of the 4 vertebrate proteins that are secreted as agonists of the canonical Wnt/β-catenin signaling pathway (plays a role in cellular proliferation, differentiation and stem cell maintenance) and belongs to the superfamily of thrombospondin type 1 repeat (TSR1)-containing proteins.Citation64 It is approximately 35 KDa in length and characterized by the presence of 2 N-terminal furin-like repeats that are needed for Wnt signal potentiation and are present in important receptors for growth factors, such as EGF, HGF, and insulin; there are 41 proteins that contains TSR1 domains in the human genome.Citation65 As other members of the spondin family, this protein is characterized by the presence of the TSR1 domain that can be found in secreted proteins or extracellular portion of the transmembrane proteins. In Rspo3, TSR1 is located toward to the C-terminus and binds glycosaminoglycan and/or proteoglycans.Citation66 Rspo3 acts an upstream of Wnt pathway and in studying of the vasculogenesis and angiogenesis signals, the spondin-targeted disruption was shown to lead to severe vascular defects in the placenta.Citation67 Lspo3 may also serve as a negative regulator of the TGF-β pathway,Citation40,54,68 and as a key regulator of angiogenesis by activating the non-canonical Wnt signaling pathway in endothelial cells needed for the control of vascular stability and pruning.Citation67

The amino acid sequence of Rspo3 starts with a signal peptide (residues 1–21), followed by 2 Fu domains (residues 35–86 and 92–135, respectively), a TSR1 domain (residues 147–207), and a C-terminal basic amino acid rich domain (residues 208–272). Rspo3 (UniProt ID: Q9BXY4) is present in 2 alternatively splice isoforms. The full-length canonical isoform #1 (UniProt ID: Q9BXY4-1) has 272 residues. In isoform #2 (UniProt ID: Q9BXY4-1), the C-terminal region VSVSTVH (residues 266–272) is changed to GIEVTLAEGLTSVSQRTQPTPCRRRYL.

Rspo4. RSPO4 gene located at the position 20p13 on chromosome 20 encodes human R-spondin 4 protein (Rspo4, also called Roof plate-specific spondin-4). Rspo4 is a ligand for the LGR4, LGR5, and LGR6 receptors that activate the canonical Wnt signaling pathway. LGR4-6 interact with phosphorylated LRP6 and frizzled receptors that are activated by extracellular Wnt receptors leading to the activation of the canonical Wnt signaling pathway to increase expression of target genes. By acting as an inhibitor of ZnRF3, which is an important regulator of the Wnt signaling pathway, Rspo4 regulates both the canonical Wnt/β-catenin-dependent pathway and non-canonical Wnt signaling.Citation36,53,54

Domain organization of Rspo4 is similar to that of other members of the spondin family, and this protein has a signal peptide (residues 1–19), followed by 2 Fu domains (residues 37–85 and 85–128), a TSR1 domain (residues 138–197), and a positively charged C-tail (residues 198–234). There are 234 residues in the full-length canonical form of Rspo4 (UniProt ID: Q2I0M5-1), whereas the isoform #2 generated by the alternative splicing (UniProt ID: Q2I0M5-2) is characterized by missing the 137–198 region.

According to STRING analysis using the highest cut-off of 0.9 as the minimal required confidence level, Rspo2, Rspo3 and Rspo4 interact with each other, as well as bind to Rspo1 and interact with many binding partners of Rspo1, such as LGR4, LGR5 and LGR6, ZNFR3, UBA52, UBB, UBC, and RPS27A. In addition to this basic set, Rspo3 is known to interact with frizzled family receptor 8 (Fzd8) (see ). The ability to interact with similar partners is likely to be due to some sequence and structural similarity of the R-spondin family members. shows that despite being encoded by different genes located at different chromosomes, these proteins share noticeable level of sequence similarity, with sequence identity ranging from 37.5% to 44.45%. These proteins also show rather similar disorder profiles (see ), indicating that the peculiarities of disorder distribution within their sequences is conserved, providing indirect support to the idea that intrinsic disorder is needed at least for some of their functions. , 4D, and 4E further illustrate high levels of intrinsic disorder in the C-terminal halves of these proteins and also show that this disordered tail can be used for interactions with some binding partners of Rspo2, Rspo3, and Rspo4. Curiously, despite rather high sequence identity, close similarity of disorder profiles, highly overlapping interactomes, and important roles played in regulation and control of Wnt signaling, the biological activities of the members of the R-spondin family have very different outputs.

Figure 4. (A) Multiple sequence alignment of human Rspo1, Rspo2, Rspo3, and Rspo4 conducted by Clustal Omega (http://www.ebi.ac.uk/Tools/msa/clustalo/). The corresponding similarity matrix is shown in the bottom of this figure legend. (B) Superposition of the consensus disorder profiles calculated for Rspo1 (black curve), Rspo2 (red curve), Rspo3 (green line), and Rspo4 (yellow line) illustrating remarkable similarity of the distribution of disorder propensities within sequences of these proteins. (C) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo2 by the D2P2 database. (D) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo3 by the D2P2 database. (E) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo4 by the D2P2 database. Percent Identity Matrix - created by Clustal2.1. Q2MKA7|RSPO1_HUMAN 100.00 40.93 44.79 38.96. Q6UXX9|RSPO2_HUMAN 40.93 100.00 45.45 41.05. Q9BXY4|RSPO3_HUMAN 44.79 45.45 100.00 37.50. Q2I0M5|RSPO4_HUMAN 38.96 41.05 37.50 100.00.

Figure 4. (A) Multiple sequence alignment of human Rspo1, Rspo2, Rspo3, and Rspo4 conducted by Clustal Omega (http://www.ebi.ac.uk/Tools/msa/clustalo/). The corresponding similarity matrix is shown in the bottom of this figure legend. (B) Superposition of the consensus disorder profiles calculated for Rspo1 (black curve), Rspo2 (red curve), Rspo3 (green line), and Rspo4 (yellow line) illustrating remarkable similarity of the distribution of disorder propensities within sequences of these proteins. (C) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo2 by the D2P2 database. (D) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo3 by the D2P2 database. (E) Intrinsic disorder propensity and some important disorder-related functional information generated for human Rspo4 by the D2P2 database. Percent Identity Matrix - created by Clustal2.1. Q2MKA7|RSPO1_HUMAN 100.00 40.93 44.79 38.96. Q6UXX9|RSPO2_HUMAN 40.93 100.00 45.45 41.05. Q9BXY4|RSPO3_HUMAN 44.79 45.45 100.00 37.50. Q2I0M5|RSPO4_HUMAN 38.96 41.05 37.50 100.00.

Finally, in order to illustrate evolutionary conservation of peculiarities of disorder distribution in R-spondins of different origin, represents disorder profiles of Rspo1 (see ), Rspo2 (see ), Rspo3 (see ), and Rspo4 (see ) from fish, frog, lizard, bird, and human and shows that specific disorder patterns are generally well preserved in these proteins, further inferring the functional importance of the predicted IDPRs. In fact, similar to Rspo1, human Rspo2 can undergo N-glycosylation, which is important for secretion and stability of this protein.Citation62 Curiously, although N-glycosylation of Rspo2 was shown to occur at Asn160, a position near the N-terminus of TSR1 domain, it did not affect the heparin binding capability of this protein,Citation62 a rather unexpected outcome since the TSR domain is known to be responsible for interaction with heparin sulfate proteoglycans (HSPGs).Citation55 Therefore, it seems that similar to the functional importance of long IDPRs on the intracellular side for mediating protein-protein interactions and/or PTMs, extracellular proteins might use their long IDPRs to carry PTM sites and sites of protein-protein interactions.

Figure 5. Analysis of the evolutionary conservation of intrinsic disorder propensity in Rspo1 (A), Rspo2 (B), Rspo3 (C), and Rspo4 (D). Disorder profiles were generated by PONDR-FIT for proteins from fish (black curves), frog (red curves), lizard (green curves), bird (yellow curves), and human (blue curves). These plots indicate the presence of rather close resemblance of the peculiarities of disorder distribution within orthologues, suggesting that intrinsic disorder might be of at least some functional importance.

Figure 5. Analysis of the evolutionary conservation of intrinsic disorder propensity in Rspo1 (A), Rspo2 (B), Rspo3 (C), and Rspo4 (D). Disorder profiles were generated by PONDR-FIT for proteins from fish (black curves), frog (red curves), lizard (green curves), bird (yellow curves), and human (blue curves). These plots indicate the presence of rather close resemblance of the peculiarities of disorder distribution within orthologues, suggesting that intrinsic disorder might be of at least some functional importance.

Spondin family

F-spondin (spondin-1)

Protein spondin-1, also known as F-spondin, is encoded by the SPON1 gene located on chromosome 11. Although it was believed that F-spondin is preferentially expressed in the embryonic floor plate of vertebrates,Citation69-73 F-spondin transcripts were shown to be widely distributed in fetal and adult human tissues. For example, in normal adult tissue, F-spondin is expressed in various organs including the lung, ovary, small intestine, and kidney. An especially high expression of F-spondin is found in the adult human ovary, where it serves as a major factor for vascular smooth muscle cell (SMC) proliferation.Citation74

F-spondin is an extracellular matrix glycoprotein playing major role in the central nervous system (CNS), during development and later in life. In vitro, it participates in cell adhesion and can stimulate the sensory neuron and spinal cord cells attachment and outgrowth of neurites.Citation75 During early development, F-spondin also plays a role in the growth of axons in both the spinal cord and the peripheral nervous system (PNS). It is believed that F-spondin defines the axonal trajectory in the spinal cord by promoting the outgrowth of commissural axonsCitation73 and inhibiting the outgrowth of motor axons.Citation76 In adult vertebrate, F-spondin displays broad distribution.Citation77 In the periphery, the expression of F-spondin is abundant in enteric neurons and in tissues regenerating following trauma. In the brain, it is associated with structures forming long neuronal tracts, such as the retina, the olfactory bulb, the habenula, and the nucleus of the medial longitudinal fasciculus (nMLF). F-spondin is expressed in the neurogenic niches of adult brain and abundant in CSF-contacting secretory neurons, especially those in the hypothalamus. Importantly, this protein can also play a role in neuronal regeneration,Citation78 and was shown to bind to the Aβ precursor protein (APP) and prevent its cleavage.Citation79

The total length of the human F-spondin (UniProt ID: Q9HCB6) is 807 amino acids. Like many large ECM proteins, F-spondin has a complex multidomain structure. This protein possesses N-terminally located reelin_N and F-spondin (FS) domains (residues 29–194 and 195–388, respectively), which are related to the corresponding domains found in the proteins Reelin and Mindin, and 6 C-terminal TSR1 domains (residues 442–495 (TSR1-1), 501–555 (TSR1-2), 558–611 (TSR1-3), 614–666 (TSR1-4), 668–721 (TSR1-5), and 754–807 (TSR1-6)). TSR1-2, TSR1-3, and TSR1-4 contain specific sequence motif (CSVTCG), which is responsible for the CD36 binding.Citation80 All these domains likely to play a role in the inhibition of the APP cleavage, where F-spondin is engaged in the formation of a heterotrimer on the membrane with APP and apolipoprotein E receptor 2 (apoEr2). This trimer is formed via the reelin_N domain and the FS domain interaction with APP and TSRs interaction with apoEr2.Citation46,81 Being a glycoprotein, F-spondin has several C-mannosylation and O-fucosylation sites,Citation82,83 with the first 5 TSR domains containing conserved tryptophan residues that have been identified as sites for C-mannosylation.Citation82

Crystal structures of the reelin-N (PDB ID: 3COO)Citation84,85 and spondin domains (PDB ID: 3Q13)Citation81 of human F-spondin were solved. The reelin-N domain of human F-spondin has a sequence identity of 30% to the N-terminal domain of human reelin.Citation85 Structure of the reelin-N domain can be described as a variant immunoglobulin fold consisting of 2 β-sheets, where β-strands A, B, E, and D form one β-sheet, and β-strands A′ (or A0), G, F, C, D″ and D′ form the opposite β-sheet.Citation85 The protein is crystallized as a dimer, 2 chemically identical molecules of which are characterized by the substantial conformational differences. In fact, although the core structure of the domain of 2 monomers (Thr59-Glu100, A111-S162, total 92 Cα atoms from each monomer) can be superimposed with an RMSD value of 0.88 Å, the N-terminal region (up to Tyr58), CD loop and the C-terminal region (after S162) are totally out of the structural alignment with RMSD values of 4.97 Å, 2.03 Å, and 2.92 Å, respectively.Citation85

The F-spondin domain (FS domain) contains 2 distinctive conserved sequence repeats known as “FS1-domain” and “FS2-domain” that can also be found in other spondin family members, including mindin 1 and 2, and M-spondin.Citation85 Analysis of crystal structure of the FS domain (residues 191–434, PDB ID: 3Q13) revealed that it is characterized by the topology typical for the C2 domain β-sandwich folds,Citation86 where core of the FS domain has an 8-stranded β-sandwich fold, with β-strands β4, β1, β8, and β7 forming one β-sheet and β-strands β3, β2, β5 and β6 forming the other β-sheet.Citation81

Although no structural information is available for any of the 6 TSR domains of human F-spondin, structure of TSR1-2 and TSR1-3 of the rat protein, along with the linker between the 2 domains, have been determined by X-ray crystallography,Citation87 and solution structures were solved for the TSR1-1 and TSR1-4 of rat F-spondin by the heteronuclear NMR spectroscopy combined with automated NOESY assignment and structure calculation,Citation88 all of which are characterized by the antiparallel 3-stranded β-sheet fold stabilized by 3 disulfide bonds. In solution structures, the first strand has a rippled conformation, whereas the 2 other strands form an antiparallel β-sheet (residues 462–467 and 484–489 in TSR1-1, and 634–640 and 657–663 in TSR1-4).Citation88

Results of the computational evaluation of the abundance of predicted intrinsic disorder in F-spondin is shown in , which illustrate that this protein contains several IDPRs, albeit being not as disordered as R-spondins discussed in the preceding sections. Potential functional applications of disorder in F-spondin is outlined in the subsequent section dedicated to mindin.

Figure 6. (A) Aligned structures of FS domains of human F-spondin (235 residues, blue structure; PDB ID: 3Q13) and mindin (211 residues, red structure; PDB ID: 3D34). (B) Superposition of the consensus disorder profiles calculated for human F-spondin (blue curve; UniProt ID: Q9HCB6) and human mindin (red curve, UniProt ID: Q9BUD6). (C) Intrinsic disorder profile of human F-spondin (UniProt ID: Q9HCB6) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. (D) Intrinsic disorder profile of human mindin (UniProt ID: Q9BUD6) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors.

Figure 6. (A) Aligned structures of FS domains of human F-spondin (235 residues, blue structure; PDB ID: 3Q13) and mindin (211 residues, red structure; PDB ID: 3D34). (B) Superposition of the consensus disorder profiles calculated for human F-spondin (blue curve; UniProt ID: Q9HCB6) and human mindin (red curve, UniProt ID: Q9BUD6). (C) Intrinsic disorder profile of human F-spondin (UniProt ID: Q9HCB6) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. (D) Intrinsic disorder profile of human mindin (UniProt ID: Q9BUD6) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors.

shows remarkable evolutionary conservation of the disorder pattern in F-spondins of different origin. Obviously, high similarity of disorder profiles of F-spondins from fish, frog, lizard, bird, and human represents a strong indication that the peculiarities of distribution of the intrinsic disorder propensity within the amino acid sequences are of at least some functional importance for these proteins.

Figure 7. Analysis of the evolutionary conservation of intrinsic disorder propensity in F-spondin (A), mindin (B), and SCO-spondin (C). Disorder profiles were generated by PONDR-FIT for proteins from fish (black curves), frog (red curves), lizard (green curves), bird (yellow curves), and human (blue curves). These plots indicate the presence of rather close resemblance of the peculiarities of disorder distribution within orthologues, suggesting that intrinsic disorder might be of at least some functional importance.

Figure 7. Analysis of the evolutionary conservation of intrinsic disorder propensity in F-spondin (A), mindin (B), and SCO-spondin (C). Disorder profiles were generated by PONDR-FIT for proteins from fish (black curves), frog (red curves), lizard (green curves), bird (yellow curves), and human (blue curves). These plots indicate the presence of rather close resemblance of the peculiarities of disorder distribution within orthologues, suggesting that intrinsic disorder might be of at least some functional importance.

Mindin (spondin-2 or DIL-1)

Spondin-2, also known as differentially expressed in cancerous and non-cancerous lung cells 1 (DIL-1), is a second member of the spondin family. It is encoded by SPON2 gene located on chromosome 4. The DIL-1 name was given to this protein when it was cloned from noncancerous lung cells and found to be downregulated in cancerous lung cells.Citation89 SPON2 expression is significantly associated with the stages of the colorectal cancer (CRC), and the upregulated spondin-2 can be used as a predictor of the poor survival prognosis of CRC patients.Citation90 High levels of SPON2 and spondin-2 protein expression are also observed in Barrett's adenocarcinoma,Citation91 breast cancer,Citation92 gastric cancer,Citation93 liver cancer,Citation94,95 ovarian cancer,Citation96,97 pancreatic cancer,Citation98 and prostate cancer.Citation99,100 It was also found that levels of this protein undergo a linear and significant increase in patients with type 2 diabetes (T2D) as the stage of their diabetic nephropathy (DN) increased.Citation101

Spondin-2 is a cell adhesion protein that stimulates hippocampal embryonic neurons adhesion and growth.Citation102 It functions as an opsonin for macrophage phagocytosis of bacteria by binding to bacteria and their components.Citation103 It plays a significant role in the initiation of the innate immune response and acts as a unique pattern-recognition molecule in the ECM for microbial pathogens providing defense responses against fungus and viruses.Citation103 It also binds bacterial lipopolysaccharides (LPSs).Citation103 Mindin participates in integrin-dependent trafficking of eosinophils.Citation104 In an experimental stroke model, mindin was shown to serve as a critical mediator of ischemic brain injury, most likely via mediation of/by Akt signaling.Citation105 It is specifically involved in the positive regulation of interleukin-6 production, macrophage cytokine production, and tumor necrosis factor production.Citation103

The total length of the human mindin (UniProt ID: Q9BUD6) is 331 amino acids. Mindin has a signal peptide (residues 1–26), a F-spondin domain (FS, residues 31–221), structure of which was solved (PDB ID: 3D34),Citation106 and a single C-terminally located TSP type-1 domain (residues 277–331). Similar to F-spondin, the FS of mindin is calcium-binding protein with Ca2+ being ligated by Asp160, Asp188, and Asp192.Citation106 The structure of the FS domain of mindin is characterized by the 8-stranded antiparallel β-sandwich topology containing 2 4-stranded β-sheets, β4β1β8β7 and β6β5β2β3 with 2 α-helices packed on one side of the β-sandwich.Citation106 represents aligned structures of FS domains of human F-spondin (235 residues, blue structure) and mindin (211 residues, red structure) and show close structural similarity of the core regions of these 2 domains illustrated by the facts that the 200 structurally aligned residues are characterized by the RMSD value of 1.15 Å and that the structural dissimilarity is mostly found in loops and tail regions. This close structural similarity is only in part can be attributed to the sequence similarity between these 2 proteins which are characterized by sequence identity of 32.51% (see supplementary materials). show that F-spondin and mindin are predicted to have less intrinsic disorder in comparison with the members of R-spondin family. Since some similarity is present in disorder profiles of F-spondin and mindin (see ), one can assume that the peculiarities of disorder distribution within the sequences of FS domains could be of some functional importance. For example, it was pointed out that in mindin, residues Lys42, Glu122, and Glu141 can be engaged in interaction with the metal ion-dependent adhesion site (MIDAS) of the integrin,Citation106 and 2 of these residues, Glu122 and Glu141, are predicted to be highly flexible, possessing the mean disorder scores of 0.48 ± 0.07 and 0.48 ± 0.05, respectively.

Similar to F-spondins, intrinsic disorder profiles of mindins of different origin are characterized by high similarity levels, indicative of the important functional roles of intrinsic disorder in these proteins. In fact, clearly shows that the distributions of the intrinsic disorder propensity within the amino acid sequences of mindins from fish, frog, lizard, bird, and human are very similar.

SCO-spondin

With length of 5,147 residues in its canonical form, SCO-spondin is the longest member of the group of human spondins considered in this article. It is encoded by the SSPO gene located at the position 7q36.1 on chromosome 7. As other spondins, SCO-spondin is a member of the trombospondin family that is involved in neuronal development by modulating cell aggregative mechanisms. It is an extracellular matrix glycoprotein that is secreted and expressed early in development by the subcommissural organ (SCO), an ependymal differentiation located in the roof of the Sylvian aqueduct.Citation107 SCO-spondin also makes part of the Reissner's fiber (RF), a thread-like structure present in the central canal of the spinal cord, and may also be involved in posterior commissure formation and spinal cord differentiation during ontogenesis of the central nervous system.Citation45 The role of this protein in adult brain, and that of the Reissner's fiber it forms, remains largely unknown.

The potent sites for protein-protein interaction include 26 thrombospondin type 1 repeats (TSR), 9 low-density lipoprotein receptor (LDLR) type A domains, 2 epidermal growth factor (EGF)-like domains, and N- and C-terminal von Willebrand factor (VWF) cysteine-rich domains.Citation108 Besides the full-length canonical form, SCO-spondin has an alternatively spliced isoform #2 (UniProt ID: A2VEC9-2) whose shortened sequence (1,314 residues) originates from removal of long N- and C-terminal regions (residues 1–1122 and 2315–5147, respectively), changes of Ala1639 to ACVEAPAPPA…DCPQGEDELD, changes of Leu1671 to LVRVGVGGGGGSAMLPPSTRALTPLPPQ, and by substituting the 2179–2314 region LFPRNWDDLD…ETEHWPPGQE to VSPAQGRWGQ…RSGRNQSVLC.

The domain structure of human SCO-spondin is complex, which is not too surprising taking into account its length. The protein starts with the EMI domain (residues 18–802) followed by the von Willebrand factor type D domain-1 (VWFD-1, residues 194–408), TIL-1 domain (residues 469–924), VWFD-2 (residues 563–373), TIL-2 (residues 827–779), von Willebrand factor type C domain-1 (VWFC-1, residues 880–039), VWFD-3 (residues 1013–3219), TIL-3 (residues 1275–5331), LDL-receptor class A domain-1 (LDLRA-1, residues 1375–5412), LDLRA-2 (residues 1415–5450), LDLRA-3 (residues 1451–1487), LDLRA-4 (residues 1491–1529), LDLRA-5 (residues 1564–4600), LDLRA-6 (residues 1602–2641), LDLRA-7 (residues 1655–5693), TSR1-1 (residues 1694–4748), TSR1-2 (residues 1750–0808), EGF-like domain-1 (EGFL-1, residues 1824–4863), EGFL-2 (residues 1864–4901), TSR1-3 (residues 1909–9965), VWFC-2 (residues 1965–5025), F5/8 type C domain (residues 2065–5224), LDLRA-8 (residues 2233–3269), LDLRA-9 (residues 2390–0426), LDLRA-10 (residues 2463–3499), TSR1-4 (residues 2500–0553), TSR1-5 (residues 2555–5610), TIL-4 (2633–3675), TSR1-6 (residues 2715–5769), TSR1-7 (residues 2772–2828), TSR1-8 (residues 2830–0883), TSR1-9 (residues 2985–5040), TSR1-10 (residues 3041–1083), TSR1-11 (residues 3183–3250), TSR1-12 (residues 3252–2307), TIL-5 (residues 3311–1365), TSR1-13 (residues 3408–8470), TSR1-14 (residues 3472–2527), TSR1-15 (residues 3645–5693), TSR1-16 (residues 3811–1932), TSR1-17 (residues 3946–6002), TSR1-18 (residues 4004–4059), TSR1-19 (residues 4159–9212), TSR1-20 (residues 4253–3305), TSR1-21 (residues 4307–7363), TSR1-22 (residues 4365–5419), TSR1-23 (residues 4615–5665), TIL-6 (residues 4667–7723), TSR1-23 (residues 4763–3816), VWFC-3 (residues 4984–4042), and the C-terminal cystine knot (CTCK) domain (residues 5041–1140).

No structural information is available for any part of SCO-spondin. represents results of intrinsic disorder analysis in human SCO-spondin (UniProt ID: A2VEC9) and shows that this protein is expected to contain significant amount of IDPRs. In fact, according to different computational tools, the mean disorder score SCO-spondin ranges from 0.153 ± 0 .002 to 0.634 ± 0 .004 according to IUPred_long and PONDR® VSL2, respectively (the mean disorder score evaluated by averaging the results of 6 predictors shown in is 0.358 ± 0 .002), whereas its overall disorder content (i.e., percentage of residues with disorder scores exceeding the threshold of 0.5) ranges from 71.2 % to 5.4 % as evaluated by the same predictors (the averaged disorder content is 20.3%). According to ANCHOR analysis, there are 6 disorder-based binding sites in this protein, residues 2051–2057, 2069–2076, 2269–2274, 2278–2309, 2328–2368, and 2429–2443. The protein is expected to have 101 disulfide bonds and 43 glycosylation sites.

Figure 8. Intrinsic disorder profile of human SCO-spondin (UniProt ID: A2VEC9) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. Due to large length of this protein, the corresponding disorder profile is split on 5 segments to better show the peculiarities of intrinsic disorder distribution within this protein.

Figure 8. Intrinsic disorder profile of human SCO-spondin (UniProt ID: A2VEC9) generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors. Due to large length of this protein, the corresponding disorder profile is split on 5 segments to better show the peculiarities of intrinsic disorder distribution within this protein.

shows that despite their very large length, SPO-spondins from different organisms (fish, frog, lizard, bird, and human) still share some levels of similarity in their disorder profiles. This is in line with an idea that similar to other members of spondin family, intrinsic disorder is of functional importance for SPO-spondins.

Spondin-interacting proteins

Frizzled-4

Frizzled 4 (Fzd4) is a protein in humans (UniProt ID: Q9ULV1) encoded by the FZD4 gene on chromosome 11q14-q21 that belongs to the frizzled family of genes which encodes 7 transmembrane domain proteins. Frizzled are receptors linked to the canonical β-catenin (CTNNB1) signaling pathway resulting in the activation of disheveled proteins (Dvl), inhibition of GSK-3 kinase, nuclear accumulation of CTNNB1, and activation of Wnt target genes. Fzd4 is the only member of the frizzled family that binds strongly to a secreted retinal growth factor with angiogenic and neuroprotective properties, norrin, also known as the Norrie disease protein or X-linked exudative vitreoretinopathy 2 protein.Citation109 Fzd4 plays a positive role in regulating Wingless type MMTV integration site signaling pathway and the signals of this protein induced by norrin regulates vascular development of vertebrate retina and controls important blood vessels in the ear.Citation110 Wnt signaling pathway is crucial for the nervous system development and maintenance, and this cascade is initiated when Wnt binds with the frizzled transmembrane receptor family and results in activation of pathways like the β-catenin-dependent pathway or the planar cell polarity- (PCP-) dependent pathway, both of which are involved in early neural crest development.Citation111 Frizzled-4 also plays a role in regulating stem cell maintenance, cell migration and neuro protection in central nervous system.Citation112 R-spondins are considered as alternative Fzd ligands.Citation113

Fzd4 contains a frizzled (FZ) cysteine-rich domain (residues 40–161) located within the N-terminal extracellular domain of this transmembrane protein and crystal structure of which is known (e.g., PDB ID: 5BPB),Citation114 a Lys-Thr-X-X-X-Trp motif mediating the interaction with the PDZ domain of Dvl family members (residues 499–504), and a PDZ-binding motif (residues 535–537), both located within the C-terminal cytoplasmic domain, and 7 transmembrane helices (residues 223–243, 255–275, 303–323, 345–365, 390–410, 437–457, and 478–498). Although in agreement with its transmembrane nature, Fzd4 is predicted to be mostly ordered, it has 4 IDPRs (residues 1–44, 141–184, 419–429, and 513–537). Furthermore, the functional motifs needed for Fzd4 interaction with Dvl and PDZ are located within the C-terminal disordered tail (see and Supplementary Materials Figure S1A). Results of the STRING-based analysis of the Fzd4 interactivity are shown in Figure S2A that clearly indicates high binding promiscuity of this protein.

Figure 9. Intrinsic disorder profiles of: (A) Human Fzd4 (UniProt ID: Q9ULV1); (B) Human Fzd8 (UniProt ID: Q9H461); (C) Human ZnRF3 (UniProt ID: Q9ULT6); and (D) Human RNF43 UniProt ID: Q68DV7). These disorder profiles were generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors.

Figure 9. Intrinsic disorder profiles of: (A) Human Fzd4 (UniProt ID: Q9ULV1); (B) Human Fzd8 (UniProt ID: Q9H461); (C) Human ZnRF3 (UniProt ID: Q9ULT6); and (D) Human RNF43 UniProt ID: Q68DV7). These disorder profiles were generated by the superposition of the outputs of PONDR® VLXT, PONDR® FIT, PONDR® VL3, PONDR® VSL2, IUPred_short and IUPred_long and a consensus disorder profile calculated by averaging disorder profiles of individual predictors.

Frizzled-8

The gene that codes for frizzled-8 (Fzd8) is FZD8 located on chromosome 10p11.2.Citation115 The protein functions as receptor for Wnt proteins,Citation116 and serves as a component of the Wnt-Fzd-LRP5-LRP6 complex that induces association of the receptor-ligand complexes into ribosome-sized signalosomes resulting in the initiation of the β-catenin canonical signaling pathway that leads to the inhibition of GSK-3 kinase, activation of disheveled proteins, accumulation of β-catenin inside the nucleus, and activation of Wnt target genes. Fzd8 may be involved in transduction and intercellular transmission of polarity information during tissue morphogenesis and/or in differentiated tissues. This protein serves as co-receptor of Wnt proteins, such as Wnt1.Citation115 The extracellular domains of Fzd8 were shown to interact with Rspo1 and Rspo3.Citation57

Human Fzd8 (UniProt ID: Q9H461) is a 694 residue-long proteins that has a signaling peptide (residues 1–27) and N-terminally located FZ domain (residues 30–151), which is a part of the extracellular domain (residues 28–275). Similar to other members of the frizzled family, this protein has 7 transmembrane helices (276–296, 313–333, 397–417, 440–460, 484–504, 533–553, and 585–605) and a cytoplasmic C-terminal tail (residues 606–694). Regions 95–100 and 147–152 of Fzd8 are involved in Wnt binding, motif Lys-Thr-X-X-X-Trp located at 608–613 region mediates interaction with the PDZ domain of Dvl family members, and a PDZ-binding motif is located the very end of C-terminus (residues 692–694). shows that Fzd8 is predicted to have several IDPRs (residues 1–33, 156–249, 340–380, 516–526, 574–580, and 625–694) 4 disorder-based potential binding sites (residues 148–160, 196–210, 666–679, and 687–694), and several phosphorylation sites. Two functional motifs/regions of Fzd8 (one of the Dvl binding motifs (residues 147–152) and C-terminal PDZ-binding motif) are located within the disordered regions that are expected to undergo binding-induced disorder-to-order transitions, clearly indicating that intrinsic disorder is important for the functionality of this transmembrane protein (see and Supplementary Materials Figure S1B). Figure S2B represents the results of the STRING-based analysis of the Fzd8 interactivity and shows that this protein is involved in a wide range of protein-protein interactions.

E3 ubiquitin-protein ligase ZnRF3

E3 ubiquitin-protein ligase is encoded by gene ZNRF3 located on chromosome 22. This proteins is also known as RING finger protein 203 and Zinc/RING finger protein 3 (ZnRF3). ZnRF3-driven ubiquitination and subsequent degradation of Wnt receptor complex components, Frizzled and LRP6, defines the involvement of this E3 ubiquitin-protein ligase in negative regulation of both canonical and non-canonical Wnt signaling pathways. It is also involved in the tumor suppressor process in the intestinal stem cell zone by inhibiting the Wnt signaling pathway which leads to size limitation of the intestinal stem cell zone.Citation117 Overexpression of ZnRF3 was shown to negatively regulate both the Wnt and Hedgehog proliferative pathways (and thereby to negatively regulate cancer progression) via dramatic reduction of the levels of LGR5 and Gli1, which are component of the Wnt and Hedgehog signaling pathways, respectively.Citation118

R-spondin proteins, such as Rspo1, are responsible for the negative regulation of ZNRF3, since indirect association between ZnRF3 and LGR4 mediated by Rspo1 promotes membrane clearance of ZnRF3.Citation117 Interactions between the extracellular region of RNF43 and ZnRF3 provides a direct linkage between the extracellular recognition and E3 ligase activity needed for the modulation of cell surface signaling.Citation119 This E3 ubiquitin ligase serves as an important component of the Rspo-LGR4/5-ZnRF3/RNF43 module that acts as a regulator of the Wnt/β-catenin-mediated metabolic liver zonation and controls hepatic growth and size during development, homeostasis, and regeneration.Citation120

Human ZnRF3 (UniProt ID: Q9ULT6) is a single-pass transmembrane protein containing N-terminal signal peptide (residues 1–55), extracellular domain (residues 56–219), transmembrane helix (residues 220–240), and a cytoplasmic domain (residues 241–936), where the zinc finger domain (RING-type, residues 293–334) is embedded. This protein exists in 2 isoforms, the full-length canonical form (936 residues) and alternatively spliced isoform #2 that differs from the canonical form by missing first 100 residues. show that, despite being a transmembrane protein, ZnRF3 is predicted to contain significant level of intrinsic disorder (>50 %), especially at its cytoplasmic domain, which seems to be mostly disordered. There are 16 disorder-based binding regions in this protein (residues 37–48, 59–66, 427–434, 507–512, 593–599, 658–670, 685–694, 696–702, 705–713, 728–741, 774–784, 801–808, 828–842, 877–892, 897–911, and 918–926) and several phosphorylation sites (see also Figure S1C). High levels of intrinsic disorder in ZnRF3 are in line with known high predisposition of protein ubiquitin E3 ligases for intrinsic disorder,Citation121 and together with large number of disorder-based binding sites defines the ability of this protein to be engaged in numerous protein-protein interactions (see Figure S2C).

E3 ubiquitin-protein ligase RNF43

E3 ubiquitin-protein ligase RNF43 or RING finger protein 43 is encoded by the RNF43 gene located on the 17q23.2 chromosome. RNF43 is one of the interacting partners of spondins and functions as a negative regulator of both canonical and non-canonical Wnt signaling pathways by mediating the ubiquitination, endocytosis and subsequent degradation of Wnt receptor complex components, such as Frizzled.Citation38,117 Therefore, similar to ZnRF3, RNF43, which is considered as a stem-cell E3 ligase, reduces Wnt signals by selectively ubiquitinating frizzled receptors and targeting surface-expressed frizzled receptors to lysosomes for degradation.Citation122 RNF43, cancer-associated RING finger protein, can be found in the endoplasmic reticulum (ER) and in the nuclear envelope.Citation123 It was suggested that RNF43 may be involved in cell growth control through the interaction with a nuclear protein HAP95.Citation123

Human RNF43 exists as 4 alternative splicing-generated proteoforms. Isoforms #2 and #3 (UniProt ID: Q68DV7-2 and Q68DV7-3) are different from the canonical form by missing region 85–125 and 1–125, respectively, whereas in the isoform #4 (UniProt ID: Q68DV7-4), the C-terminal tail region SEEELEELCEQAV (residues 771–783) is changed to EFSEGSGCGRERRLQ LNISGQVKSANKGLMEAEKDTAEMTTKILNHRDSVSCWLECRNTPPLPGATPLVGRSQGGPREVLVWLRHQKGTWKAGCDGSCL. Similar to ZnRF3, human RNF43 is a single-pass transmembrane protein that contains signal peptide (residues 1–23), extracellular domain (residues 24–197), transmembrane helix (residues 198–218), and a cytoplasmic domain (residues 219–783), with the zinc finger domain (RING-type, residues 272–313). It has 3 regions with the compositional bias, serine-rich (residues 443–503), histidine-rich (residues 547–557), and proline-rich (residues 569–760).

Crystal structure of the extracellular protease-associated (PA) domain (residues 44–198) of RNF43 in a complex with the CRD Rspo1 and the LGR5 ectodomain (ECD) was solved (PDB ID: 4KNG).Citation56 PA domains that function as ligand recognition motifs and play regulatory roles are commonly found in proteases and receptors.Citation124 shows that the RNF43 PA domain consists of 7 β-strands that form a twisted β sheet, with the curved β3 defining a shallow binding groove used for binding of the β-hairpin protrusion of the Rspo1, and 3 peripheral α-helices.Citation124 Since there is no physical contact between LGR6 and RNF5, clearly illustrates the scaffolding role of Rspo1 that assembles the tripartite LGR3/Rspo1/RNF43 complex. Cytoplasmic domain is predicted to be mostly disordered (see ) and contain 11 disorder-based binding motifs (residues 349–365, 386–396, 400–436, 451–460, 480–490, 497–512, 531–568, 597–650, 674–690, 723–730, and 777–783) and several phosphorylation sites (see Figure S1D). This high intrinsic disorder level and presence of multiple disorder-based binding sites makes RNF43 a promiscuous binder (see Figure S2D).

Figure 10. Evaluating disorder predisposition and some important disorder-related functional information evaluated for human LRP5 (UniProt ID: O75197, plot (A) and human LRP6 (UniProt ID: O75581, plot (B) by the D2P2 database.

Figure 10. Evaluating disorder predisposition and some important disorder-related functional information evaluated for human LRP5 (UniProt ID: O75197, plot (A) and human LRP6 (UniProt ID: O75581, plot (B) by the D2P2 database.

Low-density lipoprotein receptor-related proteins 5 and 6 (LRP5 and LRP6)

Low-density lipoprotein receptor-related proteins 5 and 6 are encoded by the LRP5 and LRP6 genes located on the 11q13.4 and 12p13.2 chromosome respectively. Being located on the cell surface, LRP5 and LRP6 serve as co-receptors of Wnt/β-catenin signaling and are involved in the formation of bones. These two proteins are engaged in the formation of the Wnt-Fzd-LRP5-LRP6 complex responsible for triggering the β-catenin signaling pathway via promotion of the assembly of receptor-ligand complexes into signalsomes.Citation125 After the Fzd/LRP6 co-receptor complex is induced by Wnt, it recruits Dvl1 (segment polarity protein dishevelled homolog-1) to the plasma membrane leading to the recruitment of the Axin1/GSK3β-complex to the cell surface. This results in the formation of signalsomes and leads to the inhibition of the Axin1/GSK3-mediated phosphorylation and destruction of β-catenin.Citation126 LRP5 is also involved in the norrin-mediated signal transduction.Citation127

Functionality of these proteins is regulated by various PTMs. For example, dual phosphorylation of cytoplasmic PPPSP motifs sequentially by GSK3 and CK1 is required for Axin1-binding, and subsequent stabilization and activation of β-catenin via preventing GSK3-mediated phosphorylation of this protein. In in vitro experiments, LRP6 can be phosphorylated by GRK5/6 within and outside the PPPSP motifs. Phosphorylation at Ser-1490 by CDK14 during G2/M phase leads to regulation of the Wnt signaling pathway during the cell cycle. Phosphorylation by GSK3β is induced by RPSO1 binding and inhibited by DKK1. Negative regulation of LRP6 function by casein kinase I epsilon phosphorylation may induce a negative feedback loop by phosphorylation of sites on LRP5/6 that modulate axin binding and hence β-catenin degradation.Citation128

LRP6 undergoes gamma-secretase-dependent regulated intramembrane proteolysis (RIP).Citation129 The extracellular domain is first released by shedding, and then, through the action of gamma-secretase, the intracellular domain (ICD) is released into the cytoplasm where it is free to bind to GSK3β and to activate canonical Wnt signaling.Citation129 Palmitoylation on the 2 sites near the transmembrane domain leads to release of LRP6 from the endoplasmic reticulum, whereas mono-ubiquitination retains LRP6 in the endoplasmic reticulum.Citation130

Both proteins can be ubiquitinated by ZnRF3, which leads to their degradation via the proteasome.Citation131 As it was already pointed out, ZnRF3 (which is a cell-surface transmembrane E3 ubiquitin ligase) is associated with the Wnt receptor complex. The role of this protein in the inhibition of the Wnt signaling relies on its ability to promote the turnover of frizzled and LRP6. It was also shown that the Wnt/β-catenin signaling can be enhanced via the ZnRF3 inhibition which also disrupts the Wnt/planar cell polarity signaling in vivo.Citation38,117

Finally, N-glycosylation of LRP6 is required for its cell surface location. The maturation and plasma membrane localization of this protein can be blocked by the expression of Mest/Peg1 (mesoderm-specific transcript/paternally expressed gene 1) via the Mest/Peg1-controlled inhibition of the LRP6 glycosylation.Citation132

Although LRP5 and LRP6 have many overlapping activities and rather similar domain organization (see below), these proteins are not completely redundant and also have some unique functions during development and adult tissue homeostasis, with LRP6 playing a dominant role in embryogenesis.Citation133 This conclusion can be further illustrated by non-overlapping sets of diseases linked to abnormalities of these proteins. In fact, mutations in the human LRP5 gene are associated with several diseases, such as exudative vitreoretinopathy 1,Citation127 exudative vitreoretinopathy 4,Citation134 osteoporosis,Citation135,136 osteopetrosis, autosomal dominant 1,Citation137 osteoporosis-pseudoglioma syndrome,Citation138-142 high bone mass trait,Citation143-147 Worth type of endosteal hyperostosis,Citation137 and Van Buchem disease 2,Citation137 whereas mutations in the human LRP6 gene are known to cause autosomal dominant coronary artery disease 2,Citation148,149 and selective tooth agenesis 7.Citation150

Human LRP5 (UniProt ID: O75197) is a large (mature form has 1584 residues after removal of the N-terminally located 31 residue-long signal peptide), single-path transmembrane protein that have an N-terminal extracellular ectodomain (residues 32–1384), a transmembrane helix (residues 1385–1407) and a C-terminal cytoplasmic domain (residues 1408–1615). There are 4 β-propeller regions in this protein (residues 32–288, 341–602, 644–903, and 945–1212) connected by EGF-like domains (residues 295–337, 601–641, and 902–942). This cassette is connected via another EGF-like domain (residues 1213–1254) to a series of 3 LDL-receptor class A domains (residues 1258–1296, 1297–1333, and 1335–1371). It also has multiple repeats of different nature, such as 20 LDL-receptor class B repeats (residues 75–119, 120–162, 163–206, 207–247, 248–290, 385–427, 428–470, 471–514, 515–557, 558–600, 687–729, 730–772, 773–815, 816–855, 856–898, 989–1035, 1036–1078, 1079–1123, 1124–1164, and 1165–1207) and 11 YWTD repeats (residues 78–81, 123–126, 166–169, 251–254, 388–391, 431–434, 474–477, 559–562, 690–693, 819–822, and 859–862). Finally, C-terminal domain contains a proline-rich region (residues 1495–1610) that includes 5 PPPSP motifs (residues 1500–1506, 1538–1545, 1574–1581, 1591–1596, and 1605–1612). Although no structural information is available for human LRP5 protein, by prediction, it is expected to have rather disordered C-terminal tail (see ), where most PTM sites and disorder-based binding sites are concentrated.

After removal of the N-terminal signal peptide (residues 1–19), the amino acid sequence of human LRP6 (UniProt ID: O75581) consists of 1594 residues. This is another single-path transmembrane protein that has an N-terminal extracellular ectodomain (residues 20–1370), a transmembrane helix (residues 1371–1393) and a C-terminal cytoplasmic domain (residues 1394–1613). Similar to LRP5, LRP6 has 4 β-propeller regions (residues 20–275, 328–589, 631–890, and 933–1202) connected by EGF-like domains (residues 282–324, 588–628, and 889–930). The fourth EGF-like domain (residues 1203–1244) links β-propeller containing region to a set of 3 LDL-receptor class A domains (residues 1248–1286, 1287–1323, and 1325–1361). This protein also has 20 LDL-receptor class B repeats (residues 63–106, 107–149, 150–193, 194–236, 237–276, 372–414, 415–457, 458–501, 502–542, 543–584, 674–716, 717–759, 760–802, 803–842, 843–885, 977–1025, 1026–1068, 1069–1113, 1114–1156, and 1157–1198) and its C-terminal domain contains 5 PPPSP motifs (residues 1487–1493, 1527–1534, 1568–1575, 1588–1593, and 1603–1610).

Structural information is available for the β-propeller containing region, residues 20–635 containing first 2 β-propeller domains and first 2 EGF-like domains (PDB ID: 4DG6) and residues 629–1244 containing last 2 β-propeller domains and 2 EGF-like domains (PDB ID: 4A0P),Citation151 as well as for the 2 phosphorylated PPPSP motifs, residues 1568–1575 (PDB ID: 4NM5) and 1603–1610 (PDB ID: 4NM7) bound to the GSK3/Axin complex.Citation152 The structures of the individual β-propellers are typical of the YWTD class β-propeller domains (i.e., domains containing the N-terminal Tyr-Trp-Thr-Asp (YWTD) motif), with 6 blades of 4-stranded antiparallel β-sheets being symmetrically arranged around a central channel.Citation151 Since the linker between β-propeller and EGF domain crosses the base of the β-propeller, the EGF domain is positioned to form a predominantly hydrophobic contact with the second and third blades.Citation151 Obviously, proline-rich motifs are present in their complexes with GSK3/Axin in mostly irregular structure.Citation152 In agreement with this structural characterization and similar to LRP5, LRP6 is predicted to have mostly ordered N-terminal ectodomain and rather disordered C-terminal cytoplasmic domain that includes all the disorder-based binding sites and which is heavily decorated with PTMs (see ).

It was pointed out that the majority of the Wnt- and antagonist-binding sites of LRP5 and LRP6 are located within the 4 tandem β-propeller–EGF-like domain (PE) pairs (P1E1–P4E4), suggesting that they serve as major functional recognition modules of the ectodomain.Citation133 In fact, functional analysis of LRP6 and LRP5 revealed that their 4 tandem PE pairs might represent 2 independent functional units, P1E1P2E2 and P3E3P4E4, where P1E1P2E2 serves as the primary binding domain for Wnt9b,Citation153 whereas P3E3P4E4 is engaged in interaction with Wnt3aCitation153 and Dkk1.Citation154 It was also noted that P1E1P2E2 can contribute to Dkk1 binding.Citation153,155,156

Complexity of both LRP5 and LRP6 is further increased by the fact that these 2 proteins operate as disulfide-linked homodimers. Combined with abundant intrinsic disorder in their C-terminal regions, presence of multiple PTMs and several disorder-based binding sites, this overall complexity defines the ability of these proteins to be engaged in multiple protein-protein interactions (see Fig. S3).

Leucine-rich repeat-containing G-protein-coupled receptors 4, 5, and 6 (LGR4, LGR5, and LGR6)

Leucine-rich repeat-containing G-protein-coupled receptors (LGRs) belong the superfamily of G protein-coupled receptors (GPCRs) includes at least 800 7 transmembrane receptors participating in a multitude of physiological and pathological functions.Citation157 Various physiological and pathological roles of GPCRs rely on the ability of these receptors to transduce extracellular signals to various intracellular pathways via their activation mediated by binding to a broad range of ligands, such as eicosanoids, organic compounds, peptides, and proteins.Citation157 Until quite recently, LGRs were considered as orphan GPCRs, since their endogenous ligands remained unidentified. However, it is known now that LGRs belong to the Rhodopsin subfamily of GPCRs, being typically considered as classical GPCRs in terms of their structure and signal transduction.Citation157 Based on their natural ligands, LGRs 4–8 can be grouped into 2 classes, with R-spondins being the ligands for the LGRs 4, 5, and 6,Citation52 and with the relaxin family peptide (RXFP) serving as a ligand for the LGRs 7 and 8.Citation158 The LGR 4–6 proteins are known to interact with Wnt receptors, mediate R-spondin signaling, and lead directly to the activation of Wnt canonical pathway through the Frizzled and LRP proteins.Citation159

Despite their sequence and structural similarities (see below), these 3 LGRs have rather different functions. For example, LGR4 is expressed in proliferating cells of diverse tissues, including adult stem cells and progenitor cell.Citation160 Its expression was reported in adrenal gland, cartilage, eyes, kidney, nervous system cells, and reproductive tracts, where it engaged in a broad range of important physiological functions.Citation161,162 The loss of this protein causes abnormal renal development, defective development of the gall bladder and cystic ducts,Citation163 defective postnatal development of the male reproductive tract,Citation164 developmental defects in bone formation and remodeling dysfunction,Citation165 impaired hair placode formation,Citation166 intrauterine growth retardation associated with embryonic and perinatal lethality,Citation167 and ocular anterior segment dysgenesis.Citation160 On the other hand, LGR5 serves as a marker of stem cells of hair follicleCitation168 and gastrointestinal tract.Citation169 In mice, total neonatal lethality accompanied by ankyloglossia and gastrointestinal distension was reported when LGR5 gene was knockout.Citation170 Although LGR6 also marks stem cells in hair follicles, all the cell lineages of the skin (including those of the hair follicle, sebaceous gland, and interfollicular dermis) were shown to be generated by the LGR6-positive stem cells.Citation171 Increased phosphorylation of LRP6 allows this protein to interact with R-spondin 1–3 with high affinity leading to the enhancement of the Wnt signaling.Citation68

Finally, high level of LGR4 and LGR5 expression were reported in several types of cancers. For example, cervical and colon cancer cell invasiveness and metastasis are enhanced by the LGR4 overexpression,Citation172 whereas human colon and ovarian tumors are characterized by the LGR5 up-regulation which promotes cell proliferation and tumor formation in basal cell carcinoma.Citation173,174 Although the LGR6 has a positive effect on the Wnt signaling pathway, it can also function as a tumor suppressor,Citation68 functioning via protein binding, transmembrane receptor activation and regulation of the Fzd proteins through ubiquitination.Citation175

With their almost 50% sequence identities, LGR4 (UniProt ID: Q9BXB1, 951 residues, also known as G-protein coupled receptor 48, GPR48), LGR5 (UniProt ID: O75473, 907 residues, also known as G-protein coupled receptor 49, GPR49), and LGR6 (UniProt ID: Q9HBX8, 967 residues) are closely related to each other and are characterized by similar domain organization. In fact, the N-terminal extracellular ectodomains domains (ECDs) of these 7 transmembrane receptors are composed of 17 leucine rich repeats (LRR) flanked by cysteine-rich sequences at both the N- and C-termini.Citation176 Their ECDs are substantially longer than the cytoplasmic C-terminal domains.

All three LGRs have several isoforms produced by alternative splicing. For example, the 62–85 region is missing in the isoform #2 of the LGR4, whereas isoforms #2 and #3 of the LGR5 are different from the canonical form of this protein by missing regions 263–286 and 143–214, respectively. In the isoform #2 of LGR6, the 1–71 region MPSPPGLRAL…GDLDPLTAYL is substituted to a shorter N-tail MRLEGEGRSARAGQNLSRAGSARRGAPR and the 144–239 region is missing, whereas isoform #3 of this protein differs from the canonical form possessing even shorter N-tail, MGRPRLTLVCQVSIIISAR.

Structural information is available for the ECDs of human LGR4 (residues 26–502; PDB ID: 4KT1),Citation177 and LGR5 (residues 22–543, PDB ID: 4BST).Citation59 In these structures, ectodomains of LGR4 and LGR5 were shown to possess a typical horseshoe-shaped structure consisting of the 17 leucine-rich repeat (LRR) units. illustrates that these 3 LGRs are characterized by rather similar disorder propensities, with their cytoplasmic C-terminal domains being mostly disordered. Interactomes of LGRs 4–6 are shown in Figure S4 that clearly illustrates their high binding promiscuity.

Figure 11. Evaluating disorder predisposition and some important disorder-related functional information evaluated for human LGR4 (UniProt ID: Q9BXB1, plot A), LGR5 (UniProt ID: O75473, plot B), and LGR6 (UniProt ID: Q9HBX8, plot C) by the D2P2 database.

Figure 11. Evaluating disorder predisposition and some important disorder-related functional information evaluated for human LGR4 (UniProt ID: Q9BXB1, plot A), LGR5 (UniProt ID: O75473, plot B), and LGR6 (UniProt ID: Q9HBX8, plot C) by the D2P2 database.

Concluding remarks

Our study shows that all human spondins and their major interacting partners are predicted to have significant levels of intrinsic disorder and possess a number of functionally important IDPRs. In fact, according to PONDR® VSL2 (which is a rather accurate stand-alone disorder predictor) all proteins considered in this article were predicted to have noticeable levels of disordered residues: 74.8% (ZnRF3), 71.2% (SCO-spondin), 69.2% (RNF43), 63.5% (Rspo1), 50.10% (F-spondin), 50.0% (Rspo4), 49.6% (Rspo3), 46.0% (mindin), 44.4% (Rspo2), 39.2% (Fzd8), 26.2% (LRP6), 25.3% (LRP5), 23.2% (LGR6), 19.9% (LGR5), 19.2% (Fzd4), and 16.0% (LGR4). To gain information on the average disorder propensity of these proteins, the MobiDB database (http://mobidb.bio.unipd.it/) was utilized.Citation178,179 Since MobiDB generates consensus disorder scores by aggregating the output from 10 predictors, such as 2 versions of IUPred,Citation180 2 versions of ESpritz,Citation181 2 versions of DisEMBL,Citation182 JRONN,Citation183 PONDR® VSL2B,Citation184,185 and GlobPlot,Citation186 it is likely that this database provides the most conservative estimates of intrinsic disorder in a query protein. However, even according to their MobiDB disorder consensus scores, spondins and their major interactors ranges from 1.3% to 48.8% and were arranged as follows: 48.8% (RNF43), 45.6% (ZnRF3), 40.1% (Rspo3), 34.4% (Rspo1), 29.5% (Fzd8), 25.2% (Rspo4), 16.9% (Rspo2), 12.9% (F-spondin), 12.7% (mindin), 12.1% (Fzd4), 11.3% (LRP6), 9.8% (SCO-spondin), 9.0% (LGR6), 8.6% (LRP5), 5.2% (LGR4), and 1.3% (LGR5). In other words, on average, spondins and their binding partners contain 20.3 ± 3 .8% (MobiDB) or 43.7 ± 5 .1% (PONDR® VSL2) disordered residues. Furthermore, these proteins are characterized by relatively low structural coverage of 24.6 ± 7 .1%, which is the percent of residues with known structure, with the structure being unknown for 8 of 16 proteins analyzed in this study. In fact, according to their structural coverage, spondins and their partners forms the following series: 75.9% (LRP6), 67.3% (mindin), 57.6% (LGR5), 52.8% (LGR4), 50.3% (F-spondin), 44.0% (Rspo1), 25.7% (Fzd4), 19.8% (RNF43), 0% (Rspo2), 0% (Rspo3), 0% (Rspo4), 0% (SCO-spondin), 0% (Fzd8), 0% (ZnRF3), 0% (LRP5), and 0% (LGR6)). Even if proteins with unknown structure would be excluded from consideration, the structural coverage of the remaining proteins is of 49.2 ± 6 .8%.

One can imagine multiple reasons for the lack of structural information about a query protein, ranging from the absence of attempts to solve its structure that reflects the absence of the overall interest in this protein, to the presence of some challenges associated with its crystallization. However, the “lack of interest” argument can be excluded for a protein with a partial structural coverage, since structure was determined for a part of protein sequence. It is known that the presence of intrinsic disorder can serve as one of the bottlenecks in structural characterization of a target protein that can affect several processes of the structure determination pipeline, such as expression and stability,Citation187,188 as well as solubility,Citation189,190 and predisposition for crystallization.Citation189,191-196 In application to spondins and their partners, this hypothesis is supported by the observation that proteins with more predicted disorder typically have lower structure coverage.

Data presented in our study revealed that spondins and their binding partners contain considerable levels of disorder and intrinsic disorder might be related to functionality of these proteins. Although presence of high levels of intrinsic disorder was previously reported for many proteins from the extracellular matrix, including collagens,Citation197,198 our analysis of spondins and their interactors represents the first detailed analysis of functional intrinsic disorder in these proteins. Furthermore, the abundance and functional roles of intrinsic disorder are not too often compared for intracellular vs. extracellular proteins involved in a particular biological process, and our study represents one of the first attempts to cover this gap. Structural disorder is typically associated with intracellular proteins or with intracellular regions of transmembrane proteins. We show here that similar to other signaling systems and regulatory pathways (which are mostly intracellular), intrinsic disorder is crucial for various functional aspects of extracellular spondins. In fact, their IDPRs are utilized in protein-protein interactions and serve as sites of various posttranslational modifications. It is tempting to hypothesize that the proteins covered in this study can serve as representatives of all extracellular proteins, meaning that structural disorder in the extracellular space is as general as observed here.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Supplemental material

Supplemental_materials.zip

Download Zip (1.3 MB)

Funding

This work was supported in part by Chaikin-Wile foundation (IVZh).

References

  • Dunker AK, Garner E, Guilliot S, Romero P, Albrecht K, Hart J, Obradovic Z, Kissinger C, Villafranca JE. Protein disorder and the evolution of molecular recognition: theory, predictions and observations. Pac Symp Biocomput 1998:473-84; PMID:9697205
  • Wright PE, Dyson HJ. Intrinsically unstructured proteins: re-assessing the protein structure-function paradigm. J Mol Biol 1999; 293:321-31; PMID:10550212; http://dx.doi.org/10.1006/jmbi.1999.3110
  • Uversky VN, Gillespie JR, Fink AL. Why are “natively unfolded” proteins unstructured under physiologic conditions? Proteins 2000; 41:415-27; PMID:11025552; http://dx.doi.org/10.1002/1097-0134(20001115)41:3%3c415::AID-PROT130%3e3.0.CO;2-7
  • Dunker AK, Lawson JD, Brown CJ, Williams RM, Romero P, Oh JS, Oldfield CJ, Campen AM, Ratliff CM, Hipps KW, et al. Intrinsically disordered protein. J Mol Graph Model 2001; 19:26-59; PMID:11381529; http://dx.doi.org/10.1016/S1093-3263(00)00138-8
  • Tompa P. Intrinsically unstructured proteins. Trends Biochem Sci 2002; 27:527-33; PMID:12368089; http://dx.doi.org/10.1016/S0968-0004(02)02169-2
  • Daughdrill GW, Pielak GJ, Uversky VN, Cortese MS, Dunker AK. Natively disordered proteins. In: Buchner J, Kiefhaber T, eds. Handbook of Protein Folding. Weinheim, Germany: Wiley-VCH, Verlag GmbH & Co. KGaA 2005:271-353
  • Uversky VN, Dunker AK. Understanding protein non-folding. Biochim Biophys Acta 2010; 1804:1231-64; PMID:20117254; http://dx.doi.org/10.1016/j.bbapap.2010.01.017
  • Uversky VN, Dunker AK. The case for intrinsically disordered proteins playing contributory roles in molecular recognition without a stable 3D structure. F1000 Biol Reports 2013; 5:1; PMID:23361308; http://dx.doi.org/10.3410/B5-1
  • Tompa P. Intrinsically disordered proteins: a 10-year recap. Trends Biochem Sci 2012; 37:509-16; PMID:22989858; http://dx.doi.org/10.1016/j.tibs.2012.08.004
  • Uversky VN. A decade and a half of protein intrinsic disorder: biology still waits for physics. Protein Sci 2013; 22:693-724; PMID:23553817; http://dx.doi.org/10.1002/pro.2261
  • van der Lee R, Buljan M, Lang B, Weatheritt RJ, Daughdrill GW, Dunker AK, Fuxreiter M, Gough J, Gsponer J, Jones DT, et al. Classification of intrinsically disordered regions and proteins. Chem Rev 2014; 114:6589-631; PMID:24773235; http://dx.doi.org/10.1021/cr400525m
  • Habchi J, Tompa P, Longhi S, Uversky VN. Introducing protein intrinsic disorder. Chem Rev 2014; 114:6561-88; PMID:24739139; http://dx.doi.org/10.1021/cr400514h
  • Oldfield CJ, Dunker AK. Intrinsically disordered proteins and intrinsically disordered protein regions. Annual Rev Biochem 2014; 83:553-84; PMID:24606139; http://dx.doi.org/10.1146/annurev-biochem-072711-164947
  • Uversky VN. Dancing protein clouds: The strange biology and chaotic physics of intrinsically disordered proteins. J Biol Chem 2016; 291:6681-8; PMID:26851286; http://dx.doi.org/10.1074/jbc.R115.685859
  • Dunker AK, Brown CJ, Lawson JD, Iakoucheva LM, Obradovic Z. Intrinsic disorder and protein function. Biochemistry 2002; 41:6573-82; PMID:12022860; http://dx.doi.org/10.1021/bi012159
  • Dunker AK, Brown CJ, Obradovic Z. Identification and functions of usefully disordered proteins. Adv Protein Chem 2002; 62:25-49; PMID:12418100; http://dx.doi.org/10.1016/S0065-3233(02)62004-2
  • Uversky VN. Natively unfolded proteins: a point where biology waits for physics. Protein Sci 2002; 11:739-56; PMID:11910019; http://dx.doi.org/10.1110/ps.4210102
  • Uversky VN. What does it mean to be natively unfolded? Eur J Biochem 2002; 269:2-12; PMID:11784292; http://dx.doi.org/10.1046/j.0014-2956.2001.02649.x
  • Uversky VN. Protein folding revisited. A polypeptide chain at the folding-misfolding-nonfolding cross-roads: which way to go? Cell Mol Life Sci 2003; 60:1852-71; PMID:14523548; http://dx.doi.org/10.1007/s00018-003-3096-6
  • Uversky VN. Functional roles of transiently and intrinsically disordered regions within proteins. FEBS J 2015; 282:1182-9; PMID:25631540; http://dx.doi.org/10.1111/febs.13202
  • Iakoucheva LM, Radivojac P, Brown CJ, O'Connor TR, Sikes JG, Obradovic Z, Dunker AK. The importance of intrinsic disorder for protein phosphorylation. Nucleic Acids Res 2004; 32:1037-49; PMID:14960716; http://dx.doi.org/10.1093/nar/gkh253
  • Pejaver V, Hsu WL, Xin F, Dunker AK, Uversky VN, Radivojac P. The structural and functional signatures of proteins that undergo multiple events of post-translational modification. Protein Sci 2014; 23:1077-93; PMID:24888500; http://dx.doi.org/10.1002/pro.2494
  • Romero PR, Zaidi S, Fang YY, Uversky VN, Radivojac P, Oldfield CJ, Cortese MS, Sickmeier M, LeGall T, Obradovic Z, et al. Alternative splicing in concert with protein intrinsic disorder enables increased functional diversity in multicellular organisms. Proc Natl Acad Sci U S A 2006; 103:8390-5; PMID:16717195; http://dx.doi.org/10.1073/pnas.0507916103
  • Buljan M, Chalancon G, Dunker AK, Bateman A, Balaji S, Fuxreiter M, Babu MM. Alternative splicing of intrinsically disordered regions and rewiring of protein interactions. Curr Opin Struct Biol 2013; 23:443-50; PMID:23706950; http://dx.doi.org/10.1016/j.sbi.2013.03.006
  • Buljan M, Chalancon G, Eustermann S, Wagner GP, Fuxreiter M, Bateman A, Babu MM. Tissue-specific splicing of disordered segments that embed binding motifs rewires protein interaction networks. Mol Cell 2012; 46:871-83; PMID:22749400; http://dx.doi.org/10.1016/j.molcel.2012.05.039
  • Uversky VN, Oldfield CJ, Dunker AK. Intrinsically disordered proteins in human diseases: introducing the D2 concept. Annu Rev Biophys 2008; 37:215-46; PMID:18573080; http://dx.doi.org/10.1146/annurev.biophys.37.032807.125924
  • Uversky VN, Dave V, Iakoucheva LM, Malaney P, Metallo SJ, Pathak RR, Joerger AC. Pathological unfoldomics of uncontrolled chaos: intrinsically disordered proteins and human diseases. Chem Rev 2014; 114:6844-79; PMID:24830552; http://dx.doi.org/10.1021/cr400713r
  • Uversky VN. Wrecked regulation of intrinsically disordered proteins in diseases: pathogenicity of deregulated regulators. Front Mol Biosci 2014; 1:6; PMID:25988147; http://dx.doi.org/10.3389/fmolb.2014.00006
  • Bornstein P. Diversity of function is inherent in matricellular proteins - an Appraisal of Thrombospondin-1. J Cell Biol 1995; 130:503-6; PMID:7542656; http://dx.doi.org/10.1083/jcb.130.3.503
  • Chen H, Herndon ME, Lawler J. The cell biology of thrombospondin-1. Matrix Biology 2000; 19:597-614; PMID:11102749; http://dx.doi.org/10.1016/S0945-053X(00)00107-4
  • Adams JC. Thrombospondins: multifunctional regulators of cell interactions. Annu Rev Cell Dev Biol 2001; 17:25-51; PMID:11687483; http://dx.doi.org/10.1146/annurev.cellbio.17.1.25
  • Jin YR, Yoon JK. The R-spondin family of proteins: emerging regulators of WNT signaling. Int J Biochem Cell Biol 2012; 44:2278-87; PMID:22982762; http://dx.doi.org/10.1016/j.biocel.2012.09.006
  • Niehrs C. The complex world of WNT receptor signalling. Nat Rev Mol Cell Biol 2012; 13:767-79; PMID:23151663; http://dx.doi.org/10.1038/nrm3470
  • Cruciat CM, Niehrs C. Secreted and transmembrane wnt inhibitors and activators. Cold Spring Harb Perspect Biol 2013; 5:a015081; PMID:23085770; http://dx.doi.org/10.1101/cshperspect.a015081
  • Biason-Lauber A. WNT4, RSPO1, and FOXL2 in sex development. Semin Reprod Med 2012; 30:387-95; PMID:23044875; http://dx.doi.org/10.1055/s-0032-1324722
  • de Lau WB, Snel B, Clevers HC. The R-spondin protein family. Genome Biol 2012; 13:242; PMID:22439850; http://dx.doi.org/10.1186/gb-2012-13-3-242
  • Mah AT, Yan KS, Kuo CJ. Wnt pathway regulation of intestinal stem cells. J Physiol 2016; 594:4837-47; PMID:27581568; http://dx.doi.org/10.1113/JP271754
  • Hao HX, Jiang X, Cong F. Control of wnt receptor turnover by R-spondin-ZNRF3/RNF43 signaling module and its dysregulation in cancer. Cancers (Basel) 2016; 8:54; PMID:27338477; http://dx.doi.org/10.3390/cancers8060054
  • Knight MN, Hankenson KD. R-spondins: novel matricellular regulators of the skeleton. Matrix Biol 2014; 37:157-61; PMID:24980904; http://dx.doi.org/10.1016/j.matbio.2014.06.003
  • de Lau W, Peng WC, Gros P, Clevers H. The R-spondin/Lgr5/Rnf43 module: regulator of Wnt signal strength. Genes Dev 2014; 28:305-16; PMID:24532711; http://dx.doi.org/10.1101/gad.235473.113
  • Rodriguez-Leon J, Tomas AR, Johnson A, Kawakami Y. Recent advances in the study of limb development: the emergence and function of the apical ectodermal ridge. J Stem Cells 2013; 8:79-98; PMID:24698985
  • Fujii M, Sato T. Culturing intestinal stem cells: applications for colorectal cancer research. Front Genet 2014; 5:169; PMID:24926316; http://dx.doi.org/10.3389/fgene.2014.00169
  • Koo BK, Clevers H. Stem cells marked by the R-spondin receptor LGR5. Gastroenterology 2014; 147:289-302; PMID:24859206; http://dx.doi.org/10.1053/j.gastro.2014.05.007
  • Meiniel A, Meiniel R, Goncalves-Mendes N, Creveaux I, Didier R, Dastugue B. The thrombospondin type 1 repeat (TSR) and neuronal differentiation: roles of SCO-spondin oligopeptides on neuronal cell types and cell lines. Int Rev Cytol 2003; 230:1-39; PMID:14692680; http://dx.doi.org/10.1016/S0074-7696(03)30001-4
  • Meiniel A. SCO-spondin, a glycoprotein of the subcommissural organ/Reissner's fiber complex: evidence of a potent activity on neuronal development in primary cell cultures. Microsc Res Tech 2001; 52:484-95; PMID:11241859; http://dx.doi.org/10.1002/1097-0029(20010301)52:5%3c484::AID-JEMT1034%3e3.0.CO;2-0
  • Hoe HS, Rebeck GW. Functional interactions of APP with the apoE receptor family. J Neurochem 2008; 106:2263-71; PMID:18554321; http://dx.doi.org/10.1111/j.1471-4159.2008.05517.x
  • Hoe HS, Rebeck GW. Regulated proteolysis of APP and ApoE receptors. Mol Neurobiol 2008; 37:64-72; PMID:18415033; http://dx.doi.org/10.1007/s12035-008-8017-0
  • Feinstein Y, Klar A. The neuronal class 2 TSR proteins F-spondin and Mindin: a small family with divergent biological activities. Int J Biochem Cell Biol 2004; 36:975-80; PMID:15094111; http://dx.doi.org/10.1016/j.biocel.2004.01.002
  • Kim KA, Kakitani M, Zhao JS, Oshima T, Tang T, Binnerts M, Liu Y, Boyle B, Park E, Emtage P, et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science 2005; 309:1256-9; PMID:16109882; http://dx.doi.org/10.1126/science.1112521
  • Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459:262-U147; PMID:19329995; http://dx.doi.org/10.1038/nature07935
  • Kim KA, Wagle M, Tran K, Zhan XM, Dixon MA, Liu SC, Gros D, Korver W, Yonkovich S, Tomasevic N, et al. R-Spondin family members regulate the Wnt pathway by a common mechanism. Mol Biol Cell 2008; 19:2588-96; PMID:18400942; http://dx.doi.org/10.1091/mbc.E08-02-0187
  • Carmon KS, Gong X, Lin QS, Thomas A, Liu QY. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2011; 108:11452-7; PMID:21693646; http://dx.doi.org/10.1073/pnas.1106083108
  • de Lau W, Barker N, Low TY, Koo BK, Li VSW, Teunissen H, Kujala P, Haegebarth A, Peters PJ, van de Wetering M, et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 2011; 476:293-U57; PMID:21727895; http://dx.doi.org/10.1038/nature10337
  • Glinka A, Dolde C, Kirsch N, Huang YL, Kazanskaya O, Ingelfinger D, Boutros M, Cruciat CM, Niehrs C. LGR4 and LGR5 are R-spondin receptors mediating Wnt/β-catenin and Wnt/PCP signalling. EMBO Rep 2011; 12:1055-61; PMID:21909076; http://dx.doi.org/10.1038/embor.2011.175
  • Chen H, Sottile J, Strickland DK, Mosher DF. Binding and degradation of thrombospondin-1 mediated through heparan sulphate proteoglycans and low-density-lipoprotein receptor-related protein: Localization of the functional activity to the trimeric N-terminal heparin-binding region of thrombospondin-1. Biochem J 1996; 318:959-63; PMID:8836144; http://dx.doi.org/10.1042/bj3180959
  • Chen PH, Chen XY, Lin ZH, Fang DY, He XL. The structural basis of R-spondin recognition by LGR5 and RNF43. Genes Dev 2013; 27:1345-50; PMID:23756651; http://dx.doi.org/10.1101/gad.219915.113
  • Nam JS, Turcotte TJ, Smith PF, Choi S, Yoon JK. Mouse cristin/R-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate β-catenin-dependent gene expression. J Biol Chem 2006; 281:13247-57; PMID:16543246; http://dx.doi.org/10.1074/jbc.M508324200
  • Wei Q, Yokota C, Semenov MV, Doble B, Woodgett J, He X. R-spondin1 is a high affinity ligand for LRP6 and induces LRP6 phosphorylation and β-catenin signaling. J Biol Chem 2007; 282:15903-11; PMID:17400545; http://dx.doi.org/10.1074/jbc.M701927200
  • Peng WC, de Lau W, Forneris F, Granneman JC, Huch M, Clevers H, Gros P. Structure of stem cell growth factor R-spondin 1 in complex with the ectodomain of its receptor LGR5. Cell Rep 2013; 3:1885-92; PMID:23809763; http://dx.doi.org/10.1016/j.celrep.2013.06.009
  • Meszaros B, Simon I, Dosztanyi Z. Prediction of protein binding regions in disordered proteins. PLoS Comput Biol 2009; 5:e1000376; PMID:19412530; http://dx.doi.org/10.1371/journal.pcbi.1000376
  • Dosztanyi Z, Meszaros B, Simon I. ANCHOR: web server for predicting protein binding regions in disordered proteins. Bioinformatics 2009; 25:2745-6; PMID:19717576; http://dx.doi.org/10.1093/bioinformatics/btp518
  • Chang CF, Hsu LS, Weng CY, Chen CK, Wang SY, Chou YH, Liu YY, Yuan ZX, Huang WY, Lin H, et al. N-Glycosylation of human R-Spondin 1 is required for efficient secretion and stability but not for its heparin binding ability. Int J Mol Sci 2016; 17:937; PMID:27314333; https://doi.org/10.3390/ijms17060937
  • Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, et al. The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res 2011; 39:D561-8; PMID:21045058; http://dx.doi.org/10.1093/nar/gkq973
  • Baenziger NL, Brodie GN, Majerus PW. A thrombin-sensitive protein of human platelet membranes. Proc Natl Acad Sci U S A 1971; 68:240-3; PMID:5276296; http://dx.doi.org/10.1073/pnas.68.1.240
  • Li SJ, Yen TY, Endo Y, Klauzinska M, Baljinnyam B, Macher B, Callahan R, Rubin JS. Loss-of-function point mutations and two-furin domain derivatives provide insights about R-spondin2 structure and function. Cell Signal 2009; 21:916-25; PMID:19385064; http://dx.doi.org/10.1016/j.cellsig.2009.02.001
  • Guo NH, Krutzsch HC, Negre E, Vogel T, Blake DA, Roberts DD. Heparin- and sulfatide-binding peptides from the type I repeats of human thrombospondin promote melanoma cell adhesion. Proc Natl Acad Sci U S A 1992; 89:3040-4; PMID:1557410; http://dx.doi.org/10.1073/pnas.89.7.3040
  • Kazanskaya O, Ohkawara B, Heroult M, Wu W, Maltry N, Augustin HG, Niehrs C. The Wnt signaling regulator R-spondin 3 promotes angioblast and vascular development. Development 2008; 135:3655-64; PMID:18842812; http://dx.doi.org/10.1242/dev.027284
  • Gong X, Carmon KS, Lin Q, Thomas A, Yi J, Liu Q. LGR6 is a high affinity receptor of R-spondins and potentially functions as a tumor suppressor. PLoS One 2012; 7:e37137; PMID:22615920; http://dx.doi.org/10.1371/journal.pone.0037137
  • Ruiz i Altaba A, Cox C, Jessell TM, Klar A. Ectopic neural expression of a floor plate marker in frog embryos injected with the midline transcription factor Pintallavis. Proc Natl Acad Sci U S A 1993; 90:8268-72; PMID:8367492; http://dx.doi.org/10.1073/pnas.90.17.8268
  • Klar A, Jessell TM, Ruiz i Altaba A. Control of floor plate identity and function in the embryonic nervous system. Cold Spring Harb Symp Quant Biol 1992; 57:473-82; PMID:1339683; http://dx.doi.org/10.1101/SQB.1992.057.01.052
  • Klar A, Baldassare M, Jessell TM. F-spondin: a gene expressed at high levels in the floor plate encodes a secreted protein that promotes neural cell adhesion and neurite extension. Cell 1992; 69:95-110; PMID:1555244; http://dx.doi.org/10.1016/0092-8674(92)90121-R
  • Higashijima S, Nose A, Eguchi G, Hotta Y, Okamoto H. Mindin/F-spondin family: novel ECM proteins expressed in the zebrafish embryonic axis. Dev Biol 1997; 192:211-27; PMID:9441663; http://dx.doi.org/10.1006/dbio.1997.8760
  • Burstyn-Cohen T, Tzarfaty V, Frumkin A, Feinstein Y, Stoeckli E, Klar A. F-Spondin is required for accurate pathfinding of commissural axons at the floor plate. Neuron 1999; 23:233-46; PMID:10399931; http://dx.doi.org/10.1016/S0896-6273(00)80776-X
  • Miyamoto K, Morishita Y, Yamazaki M, Minamino N, Kangawa K, Matsuo H, Mizutani T, Yamada K, Minegishi T. Isolation and characterization of vascular smooth muscle cell growth promoting factor from bovine ovarian follicular fluid and its cDNA cloning from bovine and human ovary. Arch Biochem Biophys 2001; 390:93-100; PMID:11368520; http://dx.doi.org/10.1006/abbi.2001.2367
  • Schubert D, Klar A, Park M, Dargusch R, Fischer WH. F-spondin promotes nerve precursor differentiation. J Neurochem 2006; 96:444-53; PMID:16300627; http://dx.doi.org/10.1111/j.1471-4159.2005.03563.x
  • Tzarfati-Majar V, Burstyn-Cohen T, Klar A. F-spondin is a contact-repellent molecule for embryonic motor neurons. Proc Natl Acad Sci U S A 2001; 98:4722-7; PMID:11287656; http://dx.doi.org/10.1073/pnas.081062398
  • Akle V, Guelin E, Yu L, Brassard-Giordano H, Slack BE, Zhdanova IV. F-spondin/spon1b expression patterns in developing and adult zebrafish. PLoS One 2012; 7:e37593; PMID:22768035; http://dx.doi.org/10.1371/journal.pone.0037593
  • Burstyn-Cohen T, Frumkin A, Xu YT, Scherer SS, Klar A. Accumulation of F-spondin in injured peripheral nerve promotes the outgrowth of sensory axons. J Neurosci 1998; 18:8875-85; PMID:9786993
  • Ho A, Sudhof TC. Binding of F-spondin to amyloid-β precursor protein: a candidate amyloid-β precursor protein ligand that modulates amyloid-β precursor protein cleavage. Proc Natl Acad Sci U S A 2004; 101:2548-53; PMID:14983046; http://dx.doi.org/10.1073/pnas.0308655100
  • Asch AS, Silbiger S, Heimer E, Nachman RL. Thrombospondin sequence motif (CSVTCG) is responsible for CD36 binding. Biochem Biophys Res Commun 1992; 182:1208-17; PMID:1371676; http://dx.doi.org/10.1016/0006-291X(92)91860-S
  • Tan K, Lawler J. The structure of the Ca(2)+-binding, glycosylated F-spondin domain of F-spondin - A C2-domain variant in an extracellular matrix protein. BMC Struct Biol 2011; 11:22; PMID:21569239; http://dx.doi.org/10.1186/1472-6807-11-22
  • Gonzalez de Peredo A, Klein D, Macek B, Hess D, Peter-Katalinic J, Hofsteenge J. C-mannosylation and o-fucosylation of thrombospondin type 1 repeats. Mol Cell Proteomics 2002; 1:11-8; PMID:12096136; http://dx.doi.org/10.1074/mcp.M100011-MCP200
  • Hofsteenge J, Huwiler KG, Macek B, Hess D, Lawler J, Mosher DF, Peter-Katalinic J. C-mannosylation and O-fucosylation of the thrombospondin type 1 module. J Biol Chem 2001; 276:6485-98; PMID:11067851; http://dx.doi.org/10.1074/jbc.M008073200
  • Nagae M, Nishikawa K, Yasui N, Yamasaki M, Nogi T, Takagi J. Structure of the F-spondin reeler domain reveals a unique β-sandwich fold with a deformable disulfide-bonded loop. Acta Crystallogr D Biol Crystallogr 2008; 64:1138-45; PMID:19020352; http://dx.doi.org/10.1107/S0907444908028308
  • Tan K, Duquette M, Liu JH, Lawler J, Wang JH. The crystal structure of the heparin-binding reelin-N domain of f-spondin. J Mol Biol 2008; 381:1213-23; PMID:18602404; http://dx.doi.org/10.1016/j.jmb.2008.06.045
  • Rizo J, Sudhof TC. C2-domains, structure and function of a universal Ca2+-binding domain. J Biol Chem 1998; 273:15879-82; PMID:9632630; http://dx.doi.org/10.1074/jbc.273.26.15879
  • Tan K, Duquette M, Liu JH, Dong Y, Zhang R, Joachimiak A, Lawler J, Wang JH. Crystal structure of the TSP-1 type 1 repeats: a novel layered fold and its biological implication. J Cell Biol 2002; 159:373-82; PMID:12391027; http://dx.doi.org/10.1083/jcb.200206062
  • Paakkonen K, Tossavainen H, Permi P, Rakkolainen H, Rauvala H, Raulo E, Kilpelainen I, Guntert P. Solution structures of the first and fourth TSR domains of F-spondin. Proteins 2006; 64:665-72; PMID:16736493; http://dx.doi.org/10.1002/prot.21030
  • Manda R, Kohno T, Matsuno Y, Takenoshita S, Kuwano H, Yokota J. Identification of genes (SPON2 and C20orf2) differentially expressed between cancerous and noncancerous lung cells by mRNA differential display. Genomics 1999; 61:5-14; PMID:10512675; http://dx.doi.org/10.1006/geno.1999.5939
  • Zhang Q, Wang XQ, Wang J, Cui SJ, Lou XM, Yan B, Qiao J, Jiang YH, Zhang LJ, Yang PY, et al. Upregulation of spondin-2 predicts poor survival of colorectal carcinoma patients. Oncotarget 2015; 6:15095-110; PMID:25945835; http://dx.doi.org/10.18632/oncotarget.3822
  • Razvi MH, Peng D, Dar AA, Powell SM, Frierson HF, Jr, Moskaluk CA, Washington K, El-Rifai W. Transcriptional oncogenomic hot spots in Barrett's adenocarcinomas: serial analysis of gene expression. Genes Chromosomes Cancer 2007; 46:914-28; PMID:17636545; http://dx.doi.org/10.1002/gcc.20479
  • Ellsworth RE, Seebach J, Field LA, Heckman C, Kane J, Hooke JA, Love B, Shriver CD. A gene expression signature that defines breast cancer metastases. Clin Exp Metastasis 2009; 26:205-13; PMID:19112599; http://dx.doi.org/10.1007/s10585-008-9232-9
  • Rajkumar T, Vijayalakshmi N, Gopal G, Sabitha K, Shirley S, Raja UM, Ramakrishnan SA. Identification and validation of genes involved in gastric tumorigenesis. Cancer Cell Int 2010; 10:45; PMID:21092330; http://dx.doi.org/10.1186/1475-2867-10-45
  • Liao CH, Yeh SC, Huang YH, Chen RN, Tsai MM, Chen WJ, Chi HC, Tai PJ, Liao CJ, Wu SM, et al. Positive regulation of spondin 2 by thyroid hormone is associated with cell migration and invasion. Endocr Relat Cancer 2010; 17:99-111; PMID:19903741; http://dx.doi.org/10.1677/ERC-09-0050
  • Luo JH, Ren B, Keryanov S, Tseng GC, Rao UN, Monga SP, Strom S, Demetris AJ, Nalesnik M, Yu YP, et al. Transcriptomic and genomic analysis of human hepatocellular carcinomas and hepatoblastomas. Hepatology 2006; 44:1012-24; PMID:17006932; http://dx.doi.org/10.1002/hep.21328
  • Anderson GL, McIntosh M, Wu L, Barnett M, Goodman G, Thorpe JD, Bergan L, Thornquist MD, Scholler N, Kim N, et al. Assessing lead time of selected ovarian cancer biomarkers: a nested case-control study. J Natl Cancer Inst 2010; 102:26-38; PMID:20042715; http://dx.doi.org/10.1093/jnci/djp438
  • Simon I, Liu Y, Krall KL, Urban N, Wolfert RL, Kim NW, McIntosh MW. Evaluation of the novel serum markers B7-H4, Spondin 2, and DcR3 for diagnosis and early detection of ovarian cancer. Gynecol Oncol 2007; 106:112-8; PMID:17490732; http://dx.doi.org/10.1016/j.ygyno.2007.03.007
  • Badea L, Herlea V, Dima SO, Dumitrascu T, Popescu I. Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology 2008; 55:2016-27; PMID:19260470
  • Barbieri CE. Evolution of novel biomarkers for detection of prostate cancer. J Urol 2013; 190:1970-1; PMID:24045221; http://dx.doi.org/10.1016/j.juro.2013.09.018
  • Lucarelli G, Rutigliano M, Bettocchi C, Palazzo S, Vavallo A, Galleggiante V, Trabucco S, Di Clemente D, Selvaggi FP, Battaglia M, et al. Spondin-2, a secreted extracellular matrix protein, is a novel diagnostic biomarker for prostate cancer. J Urol 2013; 190:2271-7; PMID:23665271; http://dx.doi.org/10.1016/j.juro.2013.05.004
  • Kahvecioglu S, Guclu M, Ustundag Y, Gul CB, Dogan I, Dagel T, Esen B, Esen SA, Celik H, Esen I. Evaluation of Serum Spondin 2 Levels in the Different Stages of Type 2 Diabetic Nephropathy. Nephrology (Carlton) 2015; 20:721-6; PMID:25973958; https://doi.org/10.1111/nep.12507
  • Feinstein Y, Borrell V, Garcia C, Burstyn-Cohen T, Tzarfaty V, Frumkin A, Nose A, Okamoto H, Higashijima S, Soriano E, et al. F-spondin and mindin: two structurally and functionally related genes expressed in the hippocampus that promote outgrowth of embryonic hippocampal neurons. Development 1999; 126:3637-48; PMID:10409509
  • He YW, Li H, Zhang J, Hsu CL, Lin E, Zhang N, Guo J, Forbush KA, Bevan MJ. The extracellular matrix protein mindin is a pattern-recognition molecule for microbial pathogens. Nat Immunol 2004; 5:88-97; PMID:14691481; http://dx.doi.org/10.1038/ni1021
  • Li Z, Garantziotis S, Jia W, Potts EN, Lalani S, Liu Z, He YW, Foster WM, Hollingsworth JW. The extracellular matrix protein mindin regulates trafficking of murine eosinophils into the airspace. J Leukoc Biol 2009; 85:124-31; PMID:18818374; http://dx.doi.org/10.1189/jlb.0208135
  • Wang L, Lu Y, Zhang X, Zhang Y, Jiang D, Dong X, Deng S, Yang L, Guan Y, Zhu L, et al. Mindin is a critical mediator of ischemic brain injury in an experimental stroke model. Exp Neurol 2013; 247:506-16; PMID:23360804; http://dx.doi.org/10.1016/j.expneurol.2013.01.022
  • Li Y, Cao C, Jia W, Yu L, Mo M, Wang Q, Huang Y, Lim JM, Ishihara M, Wells L, et al. Structure of the F-spondin domain of mindin, an integrin ligand and pattern recognition molecule. EMBO J 2009; 28:286-97; PMID:19153605; http://dx.doi.org/10.1038/emboj.2008.288
  • Gobron S, Monnerie H, Meiniel R, Creveaux I, Lehmann W, Lamalle D, Dastugue B, Meiniel A. SCO-spondin: a new member of the thrombospondin family secreted by the subcommissural organ is a candidate in the modulation of neuronal aggregation. J Cell Sci 1996; 109(Pt 5):1053-61; PMID:8743952
  • Gobron S, Creveaux I, Meiniel R, Didier R, Herbet A, Bamdad M, El Bitar F, Dastugue B, Meiniel A. Subcommissural organ/Reissner's fiber complex: characterization of SCO-spondin, a glycoprotein with potent activity on neurite outgrowth. Glia 2000; 32:177-91; PMID:11008217; http://dx.doi.org/10.1002/1098-1136(200011)32:2%3c177::AID-GLIA70%3e3.0.CO;2-V
  • Shen G, Ke J, Wang Z, Cheng Z, Gu X, Wei Y, Melcher K, Xu HE, Xu W. Structural basis of the Norrin-Frizzled 4 interaction. Cell Res 2015; 25:1078-81; PMID:26227961; http://dx.doi.org/10.1038/cr.2015.92
  • Kirikoshi H, Sagara N, Koike J, Tanaka K, Sekihara H, Hirai M, Katoh M. Molecular cloning and characterization of human Frizzled-4 on chromosome 11q14-q21. Biochem Biophys Res Commun 1999; 264:955-61; PMID:10544037; http://dx.doi.org/10.1006/bbrc.1999.1612
  • Sasselli V, Boesmans W, Vanden Berghe P, Tissir F, Goffinet AM, Pachnis V. Planar cell polarity genes control the connectivity of enteric neurons. J Clin Invest 2013; 123:1763-72; PMID:23478408; http://dx.doi.org/10.1172/JCI66759
  • Abbruzzese G, Gorny AK, Kaufmann LT, Cousin H, Kleino I, Steinbeisser H, Alfandari D. The Wnt receptor Frizzled-4 modulates ADAM13 metalloprotease activity. J Cell Sci 2015; 128:1139-49; PMID:25616895; http://dx.doi.org/10.1242/jcs.163063
  • Hendrickx M, Leyns L. Non-conventional Frizzled ligands and Wnt receptors. Dev Growth Differ 2008; 50:229-43; PMID:18366384; http://dx.doi.org/10.1111/j.1440-169X.2008.01016.x
  • Chang TH, Hsieh FL, Zebisch M, Harlos K, Elegheert J, Jones EY. Structure and functional properties of Norrin mimic wnt for signalling with Frizzled4, Lrp5/6, and proteoglycan. Elife 2015; 4:e06554; PMID:26158506; http://dx.doi.org/10.7554/elife.06554
  • Saitoh T, Hirai M, Katoh M. Molecular cloning and characterization of human Frizzled-8 gene on chromosome 10p11.2. Int J Oncol 2001; 18:991-6; PMID:11295046
  • 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. Arterioscler Thromb Vasc Biol 2008; 28:504-10; PMID:18174455; http://dx.doi.org/10.1161/ATVBAHA.107.157438
  • Hao HX, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, Lei H, Mickanin C, Liu D, Ruffner H, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 2012; 485:195-200; PMID:22575959; http://dx.doi.org/10.1038/nature11019
  • Qin H, Cai A, Xi H, Yuan J, Chen L. ZnRF3 induces apoptosis of gastric cancer cells by antagonizing Wnt and Hedgehog signaling. Panminerva Med 2015; 57:167-75; PMID:25923840
  • Zebisch M, Jones EY. ZNRF3/RNF43–A direct linkage of extracellular recognition and E3 ligase activity to modulate cell surface signalling. Prog Biophys Mol Biol 2015; 118:112-8; PMID:25937466; http://dx.doi.org/10.1016/j.pbiomolbio.2015.04.006
  • Planas-Paz L, Orsini V, Boulter L, Calabrese D, Pikiolek M, Nigsch F, Xie Y, Roma G, Donovan A, Marti P, et al. The RSPO-LGR4/5-ZNRF3/RNF43 module controls liver zonation and size. Nat Cell Biol 2016; 18:467-79; PMID:27088858; http://dx.doi.org/10.1038/ncb3337
  • Boomsma W, Nielsen SV, Lindorff-Larsen K, Hartmann-Petersen R, Ellgaard L. Bioinformatics analysis identifies several intrinsically disordered human E3 ubiquitin-protein ligases. PeerJ 2016; 4:e1725; PMID:26966660; http://dx.doi.org/10.7717/peerj.1725
  • Koo BK, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, van Es JH, Mohammed S, Heck AJ, Maurice MM, et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 2012; 488:665-9; PMID:22895187; http://dx.doi.org/10.1038/nature11308
  • Sugiura T, Yamaguchi A, Miyamoto K. A cancer-associated RING finger protein, RNF43, is a ubiquitin ligase that interacts with a nuclear protein, HAP95. Exp Cell Res 2008; 314:1519-28; PMID:18313049; http://dx.doi.org/10.1016/j.yexcr.2008.01.013
  • Luo X, Hofmann K. The protease-associated domain: a homology domain associated with multiple classes of proteases. Trends Biochem Sci 2001; 26:147-8; PMID:11246007; http://dx.doi.org/10.1016/S0968-0004(00)01768-0
  • Feng Q, Gao N. Keeping wnt signalosome in check by vesicular traffic. J Cell Physiol 2015; 230:1170-80; PMID:25336320; http://dx.doi.org/10.1002/jcp.24853
  • Tamai K, Semenov M, Kato Y, Spokony R, Liu C, Katsuyama Y, Hess F, Saint-Jeannet JP, He X. LDL-receptor-related proteins in wnt signal transduction. Nature 2000; 407:530-5; PMID:11029007; http://dx.doi.org/10.1038/35035117
  • Zhang L, Yang Y, Li S, Tai Z, Huang L, Liu Y, Zhu X, Di Y, Qu C, Jiang Z, et al. Whole exome sequencing analysis identifies mutations in LRP5 in indian families with familial exudative vitreoretinopathy. Genet Test Mol Biomarkers 2016; 20:346-51; PMID:27228167; http://dx.doi.org/10.1089/gtmb.2015.0322
  • Swiatek W, Kang H, Garcia BA, Shabanowitz J, Coombs GS, Hunt DF, Virshup DM. Negative regulation of LRP6 function by casein kinase I epsilon phosphorylation. J Biol Chem 2006; 281:12233-41; PMID:16513652; http://dx.doi.org/10.1074/jbc.M510580200
  • Mi K, Johnson GV. Regulated proteolytic processing of LRP6 results in release of its intracellular domain. J Neurochem 2007; 101:517-29; PMID:17326769; http://dx.doi.org/10.1111/j.1471-4159.2007.04447.x
  • Abrami L, Kunz B, Iacovache I, van der Goot FG. Palmitoylation and ubiquitination regulate exit of the wnt signaling protein LRP6 from the endoplasmic reticulum. Proc Natl Acad Sci U S A 2008; 105:5384-9; PMID:18378904; http://dx.doi.org/10.1073/pnas.0710389105
  • Jiang X, Cong F. Probing wnt receptor turnover: A critical regulatory point of wnt pathway. Methods Mol Biol 2016; 1481:39-48; PMID:27590150; http://dx.doi.org/10.1007/978-1-4939-6393-5_5
  • Jung H, Lee SK, Jho EH. Mest/Peg1 inhibits Wnt signalling through regulation of LRP6 glycosylation. Biochem J 2011; 436:263-9; PMID:21375506; http://dx.doi.org/10.1042/BJ20101512
  • He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in Wnt/β-catenin signaling: arrows point the way. Development 2004; 131:1663-77; PMID:15084453; http://dx.doi.org/10.1242/dev.01117
  • Jiao X, Ventruto V, Trese MT, Shastry BS, Hejtmancik JF. Autosomal recessive familial exudative vitreoretinopathy is associated with mutations in LRP5. Am J Hum Genet 2004; 75:878-84; PMID:15346351; http://dx.doi.org/10.1086/425080
  • Hartikka H, Makitie O, Mannikko M, Doria AS, Daneman A, Cole WG, Ala-Kokko L, Sochett EB. Heterozygous mutations in the LDL receptor-related protein 5 (LRP5) gene are associated with primary osteoporosis in children. J Bone Miner Res 2005; 20:783-9; PMID:15824851; http://dx.doi.org/10.1359/JBMR.050101
  • Mizuguchi T, Furuta I, Watanabe Y, Tsukamoto K, Tomita H, Tsujihata M, Ohta T, Kishino T, Matsumoto N, Minakami H, et al. LRP5, low-density-lipoprotein-receptor-related protein 5, is a determinant for bone mineral density. J Hum Genet 2004; 49:80-6; PMID:14727154; http://dx.doi.org/10.1007/s10038-003-0111-6
  • 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. Am J Hum Genet 2003; 72:763-71; PMID:12579474; http://dx.doi.org/10.1086/368277
  • Streeten EA, McBride D, Puffenberger E, Hoffman ME, Pollin TI, Donnelly P, Sack P, Morton H. Osteoporosis-pseudoglioma syndrome: description of 9 new cases and beneficial response to bisphosphonates. Bone 2008; 43:584-90; PMID:18602879; http://dx.doi.org/10.1016/j.bone.2008.04.020
  • Barros ER, Dias da Silva MR, Kunii IS, Hauache OM, Lazaretti-Castro M. A novel mutation in the LRP5 gene is associated with osteoporosis-pseudoglioma syndrome. Osteoporos Int 2007; 18:1017-8; PMID:17437160; http://dx.doi.org/10.1007/s00198-007-0360-x
  • Cheung WM, Jin LY, Smith DK, Cheung PT, Kwan EY, Low L, Kung AW. A family with osteoporosis pseudoglioma syndrome due to compound heterozygosity of two novel mutations in the LRP5 gene. Bone 2006; 39:470-6; PMID:16679074; http://dx.doi.org/10.1016/j.bone.2006.02.069
  • Ai M, Heeger S, Bartels CF, Schelling DK, Osteoporosis-Pseudoglioma Collaborative G. Clinical and molecular findings in osteoporosis-pseudoglioma syndrome. Am J Hum Genet 2005; 77:741-53; PMID:16252235; http://dx.doi.org/10.1086/497706
  • 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; 107:513-23; PMID:11719191; http://dx.doi.org/10.1016/S0092-8674(01)00571-2
  • Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, Wu D, Insogna K, Lifton RP. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 2002; 346:1513-21; PMID:12015390; http://dx.doi.org/10.1056/NEJMoa013444
  • 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. Am J Hum Genet 2002; 70:11-9; PMID:11741193; http://dx.doi.org/10.1086/338450
  • Johnson ML. The high bone mass family–the role of Wnt/Lrp5 signaling in the regulation of bone mass. J Musculoskelet Neuronal Interact 2004; 4:135-8; PMID:15615112
  • Rickels MR, Zhang X, Mumm S, Whyte MP. Oropharyngeal skeletal disease accompanying high bone mass and novel LRP5 mutation. J Bone Miner Res 2005; 20:878-85; PMID:15824861; http://dx.doi.org/10.1359/JBMR.041223
  • Balemans W, Devogelaer JP, Cleiren E, Piters E, Caussin E, Van Hul W. Novel LRP5 missense mutation in a patient with a high bone mass phenotype results in decreased DKK1-mediated inhibition of Wnt signaling. J Bone Miner Res 2007; 22:708-16; PMID:17295608; http://dx.doi.org/10.1359/jbmr.070211
  • Singh R, Smith E, Fathzadeh M, Liu W, Go GW, Subrahmanyan L, Faramarzi S, McKenna W, Mani A. Rare nonconservative LRP6 mutations are associated with metabolic syndrome. Hum Mutat 2013; 34:1221-5; PMID:23703864; http://dx.doi.org/10.1002/humu.22360
  • Mani A, Radhakrishnan J, Wang H, Mani A, Mani MA, Nelson-Williams C, Carew KS, Mane S, Najmabadi H, Wu D, et al. LRP6 mutation in a family with early coronary disease and metabolic risk factors. Science 2007; 315:1278-82; PMID:17332414; http://dx.doi.org/10.1126/science.1136370
  • Massink MP, Creton MA, Spanevello F, Fennis WM, Cune MS, Savelberg SM, Nijman IJ, Maurice MM, van den Boogaard MJ, van Haaften G. Loss-of-Function Mutations in the WNT Co-receptor LRP6 Cause Autosomal-Dominant Oligodontia. Am J Hum Genet 2015; 97:621-6; PMID:26387593; http://dx.doi.org/10.1016/j.ajhg.2015.08.014
  • Chen S, Bubeck D, MacDonald BT, Liang WX, Mao JH, Malinauskas T, Llorca O, Aricescu AR, Siebold C, He X, et al. Structural and functional studies of LRP6 ectodomain reveal a platform for Wnt signaling. Dev Cell 2011; 21:848-61; PMID:22000855; http://dx.doi.org/10.1016/j.devcel.2011.09.007
  • Stamos JL, Chu ML, Enos MD, Shah N, Weis WI. Structural basis of GSK-3 inhibition by N-terminal phosphorylation and by the Wnt receptor LRP6. Elife 2014; 3:e01998; PMID:24642411; http://dx.doi.org/10.7554/eLife.01998
  • Bourhis E, Tam C, Franke Y, Bazan JF, Ernst J, Hwang J, Costa M, Cochran AG, Hannoush RN. Reconstitution of a frizzled8.Wnt3a.LRP6 signaling complex reveals multiple Wnt and Dkk1 binding sites on LRP6. J Biol Chem 2010; 285:9172-9; PMID:20093360; http://dx.doi.org/10.1074/jbc.M109.092130
  • Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, Niehrs C. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature 2001; 411:321-5; PMID:11357136; http://dx.doi.org/10.1038/35077108
  • Zhang Y, Wang Y, Li X, Zhang J, Mao J, Li Z, Zheng J, Li L, Harris S, Wu D. The LRP5 high-bone-mass G171V mutation disrupts LRP5 interaction with Mesd. Mol Cell Biol 2004; 24:4677-84; PMID:15143163; http://dx.doi.org/10.1128/MCB.24.11.4677-4684.2004
  • Binnerts ME, Tomasevic N, Bright JM, Leung J, Ahn VE, Kim KA, Zhan X, Liu S, Yonkovich S, Williams J, et al. The first propeller domain of LRP6 regulates sensitivity to DKK1. Mol Biol Cell 2009; 20:3552-60; PMID:19477926; http://dx.doi.org/10.1091/mbc.E08-12-1252
  • Tang XL, Wang Y, Li DL, Luo J, Liu MY. Orphan G protein-coupled receptors (GPCRs): biological functions and potential drug targets. Acta Pharmacol Sin 2012; 33:363-71; PMID:22367282; http://dx.doi.org/10.1038/aps.2011.210
  • Kong RC, Shilling PJ, Lobb DK, Gooley PR, Bathgate RA. Membrane receptors: structure and function of the relaxin family peptide receptors. Mol Cell Endocrinol 2010; 320:1-15; PMID:20138959; http://dx.doi.org/10.1016/j.mce.2010.02.003
  • MacDonald BT, Semenov MV, Huang H, He X. Dissecting molecular differences between Wnt coreceptors LRP5 and LRP6. PLoS One 2011; 6:e23537; PMID:21887268; http://dx.doi.org/10.1371/journal.pone.0023537
  • Mazerbourg S, Bouley DM, Sudo S, Klein CA, Zhang JV, Kawamura K, Goodrich LV, Rayburn H, Tessier-Lavigne M, Hsueh AJ., Leucine-rich repeat-containing, G protein-coupled receptor 4 null mice exhibit intrauterine growth retardation associated with embryonic and perinatal lethality. Mol Endocrinol 2004; 18:2241-2254; http://dx.doi.org/10.1210/me.2004-0133
  • Weng J, Luo J, Cheng X, Jin C, Zhou X, Qu J, Tu L, Ai D, Li D, Wang J, et al. Deletion of G protein-coupled receptor 48 leads to ocular anterior segment dysgenesis (ASD) through down-regulation of Pitx2. Proc Natl Acad Sci U S A 2008; 105:6081-6; PMID:18424556; http://dx.doi.org/10.1073/pnas.0708257105
  • Shpakov AO. Structural-functional organization of polypeptide hormones receptors containing LRR-repeats and their interaction with heterotrimeric G proteins. Tsitologiia 2009; 51:637-49; PMID:19799348
  • Yamashita R, Takegawa Y, Sakumoto M, Nakahara M, Kawazu H, Hoshii T, Araki K, Yokouchi Y, Yamamura K. Defective development of the gall bladder and cystic duct in Lgr4- hypomorphic mice. Dev Dyn 2009; 238:993-1000; PMID:19301403; http://dx.doi.org/10.1002/dvdy.21900
  • Mendive F, Laurent P, Van Schoore G, Skarnes W, Pochet R, Vassart G. Defective postnatal development of the male reproductive tract in LGR4 knockout mice. Dev Biol 2006; 290:421-34; PMID:16406039; http://dx.doi.org/10.1016/j.ydbio.2005.11.043
  • Luo J, Zhou W, Zhou X, Li D, Weng J, Yi Z, Cho SG, Li C, Yi T, Wu X, et al. Regulation of bone formation and remodeling by G-protein-coupled receptor 48. Development 2009; 136:2747-56; PMID:19605502; http://dx.doi.org/10.1242/dev.033571
  • Mohri Y, Kato S, Umezawa A, Okuyama R, Nishimori K. Impaired hair placode formation with reduced expression of hair follicle-related genes in mice lacking Lgr4. Dev Dyn 2008; 237:2235-42; PMID:18651655; http://dx.doi.org/10.1002/dvdy.21639
  • Mazerbourg S, Bouley DM, Sudo S, Klein CA, Zhang JV, Kawamura K, Goodrich LV, Rayburn H, Tessier-Lavigne M, Hsueh AJ. Leucine-rich repeat-containing, G protein-coupled receptor 4 null mice exhibit intrauterine growth retardation associated with embryonic and perinatal lethality. Mol Endocrinol 2004; 18:2241-54; PMID:15192078; http://dx.doi.org/10.1210/me.2004-0133
  • Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, Toftgard R. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet 2008; 40:1291-9; PMID:18849992; http://dx.doi.org/10.1038/ng.239
  • Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007; 449:1003-7; PMID:17934449; http://dx.doi.org/10.1038/nature06196
  • Morita H, Mazerbourg S, Bouley DM, Luo CW, Kawamura K, Kuwabara Y, Baribault H, Tian H, Hsueh AJ. Neonatal lethality of LGR5 null mice is associated with ankyloglossia and gastrointestinal distension. Mol Cell Biol 2004; 24:9736-43; PMID:15509778; http://dx.doi.org/10.1128/MCB.24.22.9736-9743.2004
  • Snippert HJ, Haegebarth A, Kasper M, Jaks V, van Es JH, Barker N, van de Wetering M, van den Born M, Begthel H, Vries RG, et al. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science 2010; 327:1385-9; PMID:20223988; http://dx.doi.org/10.1126/science.1184733
  • Gao Y, Kitagawa K, Hiramatsu Y, Kikuchi H, Isobe T, Shimada M, Uchida C, Hattori T, Oda T, Nakayama K, et al. Upregulation of GPR48 induced by down-regulation of p27Kip1 enhances carcinoma cell invasiveness and metastasis. Cancer Res 2006; 66:11623-31; PMID:17178856; http://dx.doi.org/10.1158/0008-5472.CAN-06-2629
  • McClanahan T, Koseoglu S, Smith K, Grein J, Gustafson E, Black S, Kirschmeier P, Samatar AA. Identification of overexpression of orphan G protein-coupled receptor GPR49 in human colon and ovarian primary tumors. Cancer Biol Ther 2006; 5:419-26; PMID:16575208; http://dx.doi.org/10.4161/cbt.5.4.2521
  • Tanese K, Fukuma M, Yamada T, Mori T, Yoshikawa T, Watanabe W, Ishiko A, Amagai M, Nishikawa T, Sakamoto M. G-protein-coupled receptor GPR49 is up-regulated in basal cell carcinoma and promotes cell proliferation and tumor formation. Am J Pathol 2008; 173:835-43; PMID:18688030; http://dx.doi.org/10.2353/ajpath.2008.071091
  • Abo A, Clevers H. Modulating Wnt receptor turnover for tissue repair. Nat Biotechnol 2012; 30:835-6; PMID:22965053; http://dx.doi.org/10.1038/nbt.2361
  • Barker N, Clevers H. Leucine-rich repeat-containing G-protein-coupled receptors as markers of adult stem cells. Gastroenterology 2010; 138:1681-96; PMID:20417836; http://dx.doi.org/10.1053/j.gastro.2010.03.002
  • Wang D, Huang B, Zhang S, Yu X, Wu W, Wang X. Structural basis for R-spondin recognition by LGR4/5/6 receptors. Genes Dev 2013; 27:1339-44; PMID:23756652; http://dx.doi.org/10.1101/gad.219360.113
  • Di Domenico T, Walsh I, Martin AJ, Tosatto SC. MobiDB: a comprehensive database of intrinsic protein disorder annotations. Bioinformatics 2012; 28:2080-1; PMID:22661649; http://dx.doi.org/10.1093/bioinformatics/bts327
  • Potenza E, Domenico TD, Walsh I, Tosatto SC. MobiDB 2.0: an improved database of intrinsically disordered and mobile proteins. Nucleic Acids Res 2015; 43:D315-20; PMID:25361972; http://dx.doi.org/10.1093/nar/gku982
  • Dosztanyi Z, Csizmok V, Tompa P, Simon I. IUPred: web server for the prediction of intrinsically unstructured regions of proteins based on estimated energy content. Bioinformatics 2005; 21:3433-4; PMID:15955779; http://dx.doi.org/10.1093/bioinformatics/bti541
  • Walsh I, Martin AJ, Di Domenico T, Tosatto SC. ESpritz: accurate and fast prediction of protein disorder. Bioinformatics 2012; 28:503-9; PMID:22190692; http://dx.doi.org/10.1093/bioinformatics/btr682
  • Linding R, Jensen LJ, Diella F, Bork P, Gibson TJ, Russell RB. Protein disorder prediction: implications for structural proteomics. Structure 2003; 11:1453-9; PMID:14604535; http://dx.doi.org/10.1016/j.str.2003.10.002
  • Yang ZR, Thomson R, McNeil P, Esnouf RM. RONN: the bio-basis function neural network technique applied to the detection of natively disordered regions in proteins. Bioinformatics 2005; 21:3369-76; PMID:15947016; http://dx.doi.org/10.1093/bioinformatics/bti534
  • Obradovic Z, Peng K, Vucetic S, Radivojac P, Dunker AK. Exploiting heterogeneous sequence properties improves prediction of protein disorder. Proteins 2005; 61 Suppl 7:176-82; PMID:16187360; http://dx.doi.org/10.1002/prot.20735
  • Peng K, Radivojac P, Vucetic S, Dunker AK, Obradovic Z. Length-dependent prediction of protein intrinsic disorder. Bmc Bioinformatics 2006; 7:208; PMID:16618368; http://dx.doi.org/10.1186/1471-2105-7-208
  • Linding R, Russell RB, Neduva V, Gibson TJ. GlobPlot: exploring protein sequences for globularity and disorder. Nucleic Acids Res 2003; 31:3701-8; PMID:12824398; http://dx.doi.org/10.1093/nar/gkg519
  • Reeves R, Nissen MS. Purification and assays for high mobility group HMG-I(Y) protein function. Methods Enzymol 1999; 304:155-88; PMID:10372360; http://dx.doi.org/10.1016/S0076-6879(99)04011-2
  • Stewart AA, Ingebritsen TS, Cohen P. The protein phosphatases involved in cellular regulation. Five. Purification and properties of a Ca2+/calmodulin-dependent protein phosphatase (2B) from rabbit skeletal muscle. Eur J Biochem 1983; 132:289-95; PMID:6301828; http://dx.doi.org/10.1111/j.1432-1033.1983.tb07361.x
  • Bandaru V, Cooper W, Wallace SS, Doublie S. Overproduction, crystallization and preliminary crystallographic analysis of a novel human DNA-repair enzyme that recognizes oxidative DNA damage. Acta Crystallogr D Biol Crystallogr 2004; 60:1142-4; PMID:15159582; http://dx.doi.org/10.1107/S0907444904007929
  • Petros AM, Medek A, Nettesheim DG, Kim DH, Yoon HS, Swift K, Matayoshi ED, Oltersdorf T, Fesik SW. Solution structure of the antiapoptotic protein bcl-2. Proc Natl Acad Sci USA2001; 98:3012-7; PMID:11248023; http://dx.doi.org/10.1073/pnas.041619798
  • Oldfield CJ, Xue B, Van YY, Ulrich EL, Markley JL, Dunker AK, Uversky VN. Utilization of protein intrinsic disorder knowledge in structural proteomics. Biochim Biophys Acta 2012; 1834:487-98; PMID:23232152; http://dx.doi.org/10.1016/j.bbapap.2012.12.003
  • Harauz G, Ishiyama N, Hill CM, Bates IR, Libich DS, Fares C. Myelin basic protein-diverse conformational states of an intrinsically unstructured protein and its roles in myelin assembly and multiple sclerosis. Micron 2004; 35:503-42; PMID:15219899; http://dx.doi.org/10.1016/j.micron.2004.04.005
  • Bailey RW, Dunker AK, Brown CJ, Garner EC, Griswold MD. Clusterin, a binding protein with a molten globule-like region. Biochemistry 2001; 40:11828-40; PMID:11570883; http://dx.doi.org/10.1021/bi010135x
  • Iakoucheva LM, Kimzey AL, Masselon CD, Bruce JE, Garner EC, Brown CJ, Dunker AK, Smith RD, Ackerman EJ. Identification of intrinsic order and disorder in the DNA repair protein XPA. Protein Sci 2001; 10:560-71; PMID:11344324; http://dx.doi.org/10.1110/ps.29401
  • Daughdrill GW, Chadsey MS, Karlinsey JE, Hughes KT, Dahlquist FW. The C-terminal half of the anti-sigma factor, FlgM, becomes structured when bound to its target, sigma 28. Nat Struct Biol 1997; 4:285-91; PMID:9095196; http://dx.doi.org/10.1038/nsb0497-285
  • Cary PD, King DS, Crane-Robinson C, Bradbury EM, Rabbani A, Goodwin GH, Johns EW. Structural studies on two high-mobility-group proteins from calf thymus, HMG-14 and HMG-20 (ubiquitin), and their interaction with DNA. Eur J Biochem 1980; 112:577-80; PMID:6257511; http://dx.doi.org/10.1111/j.1432-1033.1980.tb06123.x
  • Smithers B, Oates ME, Tompa P, Gough J. Three reasons protein disorder analysis makes more sense in the light of collagen. Protein Sci 2016; 25:1030-6; PMID:26941008; http://dx.doi.org/10.1002/pro.2913
  • Peysselon F, Xue B, Uversky VN, Ricard-Blum S. Intrinsic disorder of the extracellular matrix. Mol Biosyst 2011; 7:3353-65; PMID:22009114; http://dx.doi.org/10.1039/c1mb05316g
  • Shatsky M, Nussinov R, Wolfson HJ. A method for simultaneous alignment of multiple protein structures. Proteins 2004; 56:143-56; PMID:15162494; http://dx.doi.org/10.1002/prot.10628
  • Humphrey W, Dalke A, Schulten K. VMD: visual molecular dynamics. J Mol Graphics 1996; 14:33-8, 27–8; http://dx.doi.org/10.1016/0263-7855(96)00018-5
  • Oates ME, Romero P, Ishida T, Ghalwash M, Mizianty MJ, Xue B, Dosztanyi Z, Uversky VN, Obradovic Z, Kurgan L, et al. D(2)P(2): database of disordered protein predictions. Nucleic Acids Res 2013; 41:D508-16; PMID:23203878; http://dx.doi.org/10.1093/nar/gks1226
  • Romero P, Obradovic Z, Li X, Garner EC, Brown CJ, Dunker AK. Sequence complexity of disordered protein. Proteins 2001; 42:38-48; PMID:11093259; http://dx.doi.org/10.1002/1097-0134(20010101)42:1%3c38::AID-PROT50%3e3.0.CO;2-3
  • Ishida T, Kinoshita K. PrDOS: prediction of disordered protein regions from amino acid sequence. Nucleic Acids Res 2007; 35:W460-4; PMID:17567614; http://dx.doi.org/10.1093/nar/gkm363

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.