Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Expression of Signaling Components in Embryonic Eyelid Epithelium

  • Qinghang Meng,

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

  • Chang Jin,

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

  • Yinglei Chen,

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

  • Jing Chen,

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

  • Mario Medvedovic,

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

  • Ying Xia

    ying.xia@uc.edu

    Affiliation Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America

Correction

26 Aug 2014: The PLOS ONE Staff (2014) Correction: Expression of Signaling Components in Embryonic Eyelid Epithelium. PLOS ONE 9(8): e106783. https://doi.org/10.1371/journal.pone.0106783 View correction

Abstract

Closure of an epithelium opening is a critical morphogenetic event for development. An excellent example for this process is the transient closure of embryonic eyelid. Eyelid closure requires shape change and migration of epithelial cells at the tip of the developing eyelids, and is dictated by numerous signaling pathways. Here we evaluated gene expression in epithelial cells isolated from the tip (leading edge, LE) and inner surface epithelium (IE) of the eyelid from E15.5 mouse fetuses by laser capture microdissection (LCM). We showed that the LE and IE cells are different at E15.5, such that IE had higher expression of muscle specific genes, while LE acquired epithelium identities. Despite their distinct destinies, these cells were overall similar in expression of signaling components for the “eyelid closure pathways”. However, while the LE cells had more abundant expression of Fgfr2, Erbb2, Shh, Ptch1 and 2, Smo and Gli2, and Jag1 and Notch1, the IE cells had more abundant expression of Bmp5 and Bmpr1a. In addition, the LE cells had more abundant expression of adenomatosis polyposis coli down-regulated 1 (Apcdd1), but the IE cells had high expression of Dkk2. Our results suggest that the functionally distinct LE and IE cells have also differential expression of signaling molecules that may contribute to the cell-specific responses to morphogenetic signals. The expression pattern suggests that the EGF, Shh and NOTCH pathways are preferentially active in LE cells, the BMP pathways are effective in IE cells, and the Wnt pathway may be repressed in LE and IE cells via different mechanisms.

Introduction

Formation of the eyelid is one of the last major morphogenetic events in mammalian prenatal development. Though for the most part data are scarce in humans, histological analyses of available embryos/fetuses have shown that eyelid development proceeds through four distinct phases, namely, lid formation, growth, fusion and re-opening [1], [2]. In mice, eyelid development follows similar steps but has been characterized in greater detail. Mouse eyelid formation begins at around embryonic day 11.5 (E11.5). At this time, the surface ectoderm adjacent to the developing cornea folds to form the lid buds, which are a simple structure consisting of loose periocular mesenchyme (POM) covered by undifferentiated ectoderm [3][6]. The eyelid buds grow from E12 onward, and they extend across the ocular surface, undergoing proliferation and differentiation. The eyelid at this stage is covered by epidermis, overlaid by periderm at the anterior surface and conjunctiva at the posterior surface. The epithelial margins of the superior and inferior lid fuse between E15 - E16. Lid fusion begins when the periderm cells become rounded and piled up at the leading edges of the eyelids, and then stream out across the corneal surface. The eyelids meet at the inner and outer canthi and temporarily fuse across the cornea [3], [4]. Once contact is established between the apposed eyelids, the cells at the fusion junction flatten and form a strip along the fusion line, and they slough off with the rest of the periderm [4], [7], [8]. Mouse eyelid remains closed between E16.5 and postnatal day 12–14. Cells at the eyelid fusion junction undergo desquamation and/or apoptosis, resulting in separation of the upper and lower eyelids at around postnatal day 14 [4], [9].

Much is known about the molecular factors involved in eyelid formation and fusion. This is because, although mice are normally born with a closed eyelid, a large number of genetic mutant strains display a distinct “eye open at birth” (EOB) phenotype. The Mouse Genome Informatics (MGI) (http://www.informatics.jax.org/) has a collection of >138 genotypes associated with the phenotype; the number is likely to increase with complete or partial knockout of new genes.

The majority of the EOB phenotype is caused by failure of eyelid fusion at E15–E16. One of the most significant findings made by the analysis of EOB mice is that multiple signaling pathways are involved in the regulation of eyelid closure. Some pathways, such as RA-RXR/RAR and PITX2-DKK2, and the FOXL and OAR2 transcription factors, seem to operate in the periocular mesenchyme [10][12]; others, such as the FGF10-FGFR and BMP-BMPR pathways, act through crosstalk between mesenchyme and epithelium [6], [13]. Furthermore, a number of pathways, including MAP3K1-JNK, EGFR, ROCK and PCP, are specifically effective in the eyelid epithelial cells [14][35]. There is also evidence for signal compartmentalization and spatial segregation, so that the signaling pathways are activated in distinct cell population in the developing eyelids [21], [36].

Though the outline of the pathways is more or less drawn, the role that the actual players involved in signal transduction has not been fully understood. Genetic knockout studies in mice have helped to elucidate the roles of some of the signaling molecules. Using this approach, it is shown that multiple EGFR ligands act additively to regulate eyelid morphogenesis. Thus, whereas the Hb-egf-null and Tgfα-null mice display occasionally “open-eye” phenotype, the compound mutants, i.e. Hb-egf(−/−)Tgfα(+/−) and Hb-egf(+/−)Tgfα(−/−), have a slightly increased penetrance, and the double homozygous null mice have a drastically increased penetrance of the phenotype. Furthermore, the triple null mice, lacking three of the EGFR ligand genes, Egf, Areg and Tgfα, exhibit a severe “eye-open” phenotype [37]. Similarly, by generating a series of genetic mutant strains, Huang, et. al. have shown the BMP signaling is specifically involved in eyelid closure. Mice lacking components of the TGFβ pathways have normal eyelid development, but those with impaired BMP signaling display an ‘eyelid open at birth’ phenotype [13].

The most remarkable feature of lid closure is the shape change and migration underwent by the epithelial cells at the “tip” of the eyelid. This is accompanied by activation of specific morphogenetic pathways. It is possible that the tip cells have unique surrounding tissues, i.e., microenvironments, which produce morphogens for specific activation of signaling pathways. Alternatively, the tip cells may have unique gene expression thereby acquiring new signaling and morphogenetic properties. Gene expression is a crucial facet of its function, and many genes essential for eyelid closure, such as Tgfα, Hb-egf, Activinβb and Map3k1, are indeed up-regulated in the developing eyelid epithelium [6], [20], [38], [39].

In the present work, we applied a global approach to compare gene expression profiles in epithelial cells isolated from the tip (leading edge, LE) and the inner surface (inner epithelium, IE) of the embryonic eyelid. We evaluated the relative abundance in expression of genes whose products might constitute the major “eyelid closure pathways”. Results may help to understand how signals are distinctly regulated in the LE cells and provide guidance for selecting “genes of interest” for expression and knockout studies.

Materials and Methods

Experimental animals

C57BL/6 fetuses were collected at E15.5. Euthanasia of the E15.5 fetuses was done by decapitation with surgical scissors, and genotypes were determined by PCR. Experiments conducted with these animals were carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of the University of Cincinnati (Protocol no. 06-04-19-01).

Tissue and cell preparation, RNA and cDNA generation and microarray

This process was done as previously described [40]. Briefly, the heads of E15.5 fetuses were embedded in Tissue-Tek OCT medium (Sakura Finetek USA) and stored in −80°C. Eight µm coronal sections were mounted on plain uncoated glass slides, dehydrated and stained with HistoGene LCM frozen section staining kit, and were used for LCM following the manufacturer's protocol (Molecular Devices). Cells from 4 sections were collected on one LCM cap and lysed for RNA harvesting. The lysates from each fetus were pooled and processed as one biological sample. It was estimated that 10 ng and 15 ng total RNA were obtained from LE and IE eyelid epithelium, respectively, per fetus.

RNA was analyzed by Agilent 2100 Bioanalyzer (Agilent, Santa Clara, CA) and samples with RNA Integrity Number (RIN) >5.5 were processed for cDNA amplification. cDNA amplification and biotinylation was done using Ovation Pico WTA System (NuGEN, San Carlos, CA) following the manufacturer's instructions. Specifically, RNA (10 ng) was processed into first strand cDNA, a DNA/RNA heteroduplex, and thereafter a linear isothermal amplified cDNA. The amplified cDNA was purified with a PCR Purification Kit (QIAGEN, Valencia, CA).

The cDNAs from each fetus were considered one biological sample and 3 samples were used for triplicate hybridization on the Affymetrix GeneChip Mouse Gene 1.0 ST array (P/N 901168, Affymetrix, Santa Clara, CA). The arrays were hybridized with 15 µg of fragmented aRNA. The hybridization, staining, and washing are carried out using the Affymetrix GeneChip Hybridization Wash and Stain Kit (P/N 900720) following the manufacturer's protocols. The arrays were hybridized for 16 hr at 45°C using Affymetrix Hybridization Oven 640 (P/N 800139). FS450-0001 protocol was used for staining and washing the GeneChips using the Affymetrix Fluidics Station 450 (P/N 00-0079). The GeneChips were scanned with Affymetrix GeneChip Scanner 3000 7G Plus using Affymetrix GeneChip Command Console 3.2.3.1515 software and Affymetrix preset settings.

Quantitative RT-PCR

Quantitative PCR was performed using an MX3000p thermal cycler system and SYBR Green QPCR Master Mix (Stratagene), using conditions optimized for each target gene primers with efficiency greater than 85%, cycles less than 29 and sample locations on the plates been randomized. The PCR products were subjected to melting curve analysis and the relative cycle differences in qRT-PCR were determined using ΔCt, as described [41]. The ΔCt value for each sample was determined using the cycle threshold (Ct) value of the specific gene normalized to that of Gapdh. The fold change was calculated based on the ratio between LE versus IE (control) samples, designated as 1. Data are based on triplicate reactions of at least 3 biological samples.

Statistical and bioinformatics analyses

Array data (GEO repository, accession no. GSE39240) were analyzed at gene level using statistical software R and the limma package of Bioconductor [42] with custom CDF downloaded from BrainArray [43]. Data pre-processing, including background correction and normalization, was performed using RMA. Array quality was assessed using the Array Quality Metrics package of Bioconductor [44]. Statistical significance of differential gene expression between LE and IE samples were established based on empirical Bayes linear model as implemented in limma package [42].

Functional enrichment analysis of differentially expressed genes was performed using the logistic regression based LRpath methodology [45] as implemented in the R package CLEAN [46]. The gene list used in the functional enrichment analysis came from genes associated with Gene Ontology terms [47]. The statistical significance of gene list enrichment was determined based on the False Discovery Rate (fdr) cut-off of 0.1. The statistical significance of deviations of average gene expression levels for genes within the same group were established by calculating gene specific z-statistics and comparing it to the standard Normal distribution. The z-statistic was calculated by subtracting the average of expression levels of all genes in the group from the expression level of the gene and dividing the difference by the standard deviation of the expression levels within the group.

Results and Discussion

Gene expression profiles in the developing eyelid epithelium

To identify the molecular signatures of eyelid closure, we collected mouse fetuses at E15.5, a developmental stage immediately before the eyelid beginning to close. We used laser capture microdissection (LCM) to isolate epithelial cells from the leading edge (LE) and inner surface epithelium (IE). The samples were used for expression array and gene expression signatures were analyzed as described [40].

To determine whether the LE and IE cells were different at E15.5, we analyzed the expression data by Gene Ontology (GO). The LE cells were enriched for genes involved in epidermis development, transcription factor activity, pattern specification and odontogenesis. By contrast, the IE cells were enriched for genes for muscle development, RNA splicing, microtubule organization and centrosomes (Table 1). The GO signatures suggest that the E15.5 LE and IE cells have already departed to distinct paths from their common origin - the ocular surface ectoderm.

Expression of signaling molecules in the FGF and EGF pathways

To evaluate whether the LE and IE cells had differential expression of signaling molecules, we examined genes involved in the FGF and EGF pathways, known to be involved in eyelid closure. The fibroblast growth factor (FGF) family has 22 ligands and four membrane-bound receptors, FGFR 1-4, with different ligand binding affinities [48], [49]. In LE and IE cells, the Fgfr2 was the most abundantly expressed receptor gene, and Fgf9 was the highly expressed ligand gene (Table 2). Between LE and IE, there was no major difference in the expression of genes belonging to the families of FGF ligands and receptors, except for Fgfr2 (Table 2). The level of Fgfr2 was 1.8-fold higher in LE cells, suggesting that the LE cells might be more responsive to FGF signals than the IE cells.

Previously, we have shown that FGF9 expression was decreased in LE cells of Map3k1 knockout fetuses corresponding to failure of eyelid closure [40]. FGF9 could act in an autocrine fashion to induce epithelial branching, or it could send signals to the mesenchyme to induce PITX2 and FGF10. FGF10 in turn could trans-activate FGFR in epithelial cells and stimulate epithelial budding [50], [51]. Genetic studies show that FGF10 is crucial for eyelid closure, but FGF9, though required for sex determination and reproductive system development, lung embryogenesis, and inner ear morphogenesis, is dispensable for eyelid development [52][54]. Since FGF10 was almost undetectable in LE and IE cells, it is possible that this ligand is produced by the underlying mesenchymal cells, responsible for activation of FGFR2 in the eyelid epithelium [5], [6].

The epidermal growth factor (EGF) pathway operates in an autocrine fashion, such that ligands produced by the epithelial cells can activate receptors on the same or nearby cells [6], [38], [55], [56]. The mammalian system has nine ligands, which are first expressed as transmembrane proteins comprising a signal sequence, a transmembrane domain and the EGF domain(s). The ligands are then activated by ectodomain shedding that releases the EGF domain from the membrane-bound precursors. This is carried out by members of disintegrin and metalloproteases (ADAMS) family of type I transmembrane Zn-dependent proteases. There are four EGF receptor tyrosine kinases, including EGFR/ERBB1, ERBB2, ERBB3 and ERBB4 [57]. Activation of the receptors is also facilitated by members of the leucine-rich repeat containing G-protein coupled receptor (LGR) and G protein-coupled receptor (GPCR) families.

In LE and IE cells, the Egfr and Erbb2, and several genes in the GPCR families, such as Lgr4, Gpr125, Gpr20, Gpr180, Gpr89 and Gpr3, were abundantly expressed (Tables 3 and 4). Expression of Adams10 was also abundant (Table 5). Expression of Gpr56 was relatively abundant in LE cells, whereas expression of Adam 17, Lgr4, Gpr107 and Gpr137b-ps was more abundant in IE cells. Compared to the IE cells, the LE cells had significantly higher expression of Erbb2 (1.8-fold) and Gpr56 (1.3-fold), but less expression of Adamts1 (-2.6-fold).

The ligands specific for ERBB2 are unknown, but ERBB2 can dimerize with EGFR. The heterodimers, similar to the EGFR homodimers, can be activated by amphiregulin (AREG), heparin-binding EGF-like growth factor (HB-EGF) and transforming growth factor α (TGFα) [58]. Activation of the EGFR signaling is essential for embryonic eyelid closure [59]. Based on the relative abundance of receptor gene expression, the EGFR/EGFR and EGFR/ERBB2 dimers are likely to form in the developing eyelid epithelium. Specifically, the EGFR/ERBB2 may be the dominant form in LE, whereas the EGFR/EGFR is likely to be the predominant form in IE cells.

ADAMS10 is important for the development of blood vessels and central nervous system, as well as in pathological conditions such as inflammation and cancer [60]. Recently, it was shown that ADAMS10 may be the sheddase of notch receptors, involved in the release of the extracellular domain and mediating skin development; however, its role in eyelid development has not been established. On the other hand, the Adams17 knockout mice exhibit the open eye phenotype [61]. ADAMS17 is the major sheddase of TGFα, amphiregulin, HB-EGF and epiregulin, and is essential for activation of EGFR during development [62], [63]. Of the Lgr/Gpcr families, only the Lgr4 (−/−) mice have defective keratinocyte motility and produce the EOB phenotype. The Lgr4, also known as Gpr48, was known to play a role in HB-EGF-induced EGFR activation [64], [65]. The expression of Adams17 and Lgr4 was both relatively abundant in the IE cells (Tables 4 and 5).

The most surprising observation made by the RNA array was that expression of EGFR ligands was scarce in the LE and IE cells (Table 3). This was in clear contrast to previous findings made by in situ hybridization and immunohistochemistry, which showed that expression of TGFα and HB-EGF was up-regulated in a group cells located at the tip of the developing eyelid [6], [38], [66]. The discrepancy could be explained if induction of ligand is a temporal-spatial event, taking place in a small number of cells and in a narrow window during embryogenesis. Hence, either ligand up-regulation was insignificant at E15.5, or the expression signals were masked or under-represented in the collectives of the LCM captured cells, exemplifying the limitations of this approach.

Taken together, the gene expression data confirm that many genetically identified “eyelid closure” factors, such as FGFR, EGFR, ADAMS17 and LGR4, are also relatively abundant in the LE and/or IE cells, but some highly expressed genes, including Fgf9 and Adam10, are not known to be involved in eyelid closure. In comparison to the IE cells, the LE cells have higher expression of Fgfr2 and Erbb2, which may contribute to differential signaling responses of these cells.

Expression of genes involved in the TGFβ signaling

The TGFβ superfamily consists of more than 30 structurally related ligands. They belong to the Bone Morphogenetic Proteins (BMPs), TGFβs and Activin/Inhibin subfamilies [67]. These ligands act selectively on seven type I and five type II receptors, resulting in receptor dimerization and activation. The receptors in turn activate two sets of so called R-SMAD. SMAD 1, 5, and 8 are substrates of Type I receptors for BMPs, whereas SMAD2 and 3 are substrates for Type I receptors for TGFβs and Activins. Once activated, R-SMADs assemble with SMAD4, also known as co-SMAD, and the heterodimer translocates into the nucleus to regulate responsive gene expression.

In LE and IE cells, the Acvr2a was the significantly expressed receptor gene, while Smad2 was the abundantly expressed gene for intracellular transmitter. In addition, expression of Bmp7 was relatively abundant in LE, and Growth differentiation factor 10 (Gdf10) was abundant in IE cells (Table 6). Furthermore, the IE cells had a slightly higher expression of inhibin beta-B, but much higher Bmp5, Bmpr1a and Acvr1.

Previous genetic studies in mice have implicated TGF β signaling in eyelid closure. Huang et. al. carried out a methodical gene knockout study, in which each TGFβ cascade was specifically inactivated in ocular surface epithelium [13]. The results showed that BMP, but not TGFβ or activin, signaling was required for eyelid closure. The EOB phenotype was observed in mice lacking the type I BMP receptor genes, Acvr1 and Bmpr1a, the R-Smad genes, Smad 1 and Smad5, and the Co-Smad gene, Smad 4, but not in mice lacking the type II TGFβ receptor gene Tgfbr2 and the activin/TGFβ-activated R-Smad genes, Smad2 and Smad3. Conditional deletion of Bmpr1a in the ectoderm and overexpression of the inhibitory SMAD7 in keratinocytes also led to an EOB phenotype [68], [69]. Our data showed that although the LE and IE cells had type II BMP receptor expression, only the IE cells expressed abundantly the type I receptor BMPR1A. Hence, activation of the BMP pathway can be carried out mainly in the IE cells.

Of the ligands highly expressed in IE cells, BMP5 is required for chondrocytic activity during endochondral ossification, and its deficiency leads to a number of skeletal defects [70]. GDF10 is expressed in skeletal muscles but is dispensable for fetal development [71]. Recently, it was shown that GDF10, similar to TGFβ, can activate Smad2/3 and counteract the BMP signals [72]. Of the ligands highly expressed in LE cells, BMP7 is required for eye development, but is dispensable for eyelid closure [73]. The inhibin βB is required for embryonic eyelid closure; however, it may do so through a mechanism independent of SMAD [13], [20], [39]. These observations seem to support the idea that activation of the BMP pathways for eyelid closure is initiated by BMP4 produced by the the mesenchymal cells, but not ligands produced in the epithelial cells [13]. Collectively, the gene expression pattern has identified differential expression of Bmpr1a, Inhbb and Bmp5 in the LE and IE cells, and suggests that the BMP pathways may be preferentially activated in the IE cells.

Expression of genes involved in the canonical Wnt pathways

The canonical Wnt pathway is activated by binding of ligands to the Frizzled (FZD) receptors, seven-transmembrane proteins with 10 family members (FZD 1–10), and co-receptors, such as the low-density lipoprotein-related receptor protein-5 or -6 (LRP5/6) [74], [75]. The receptor signal is transduced by the Dishevelled (DVL), which are scaffold proteins that interact with diverse proteins, including kinases, phosphatases and adaptor proteins. Intracellular transduction of the Wnt signal is carried out by stabilization and cytosolic accumulation of the critical mediator, β-catenin. The β-catenin then translocates to the nucleus, binds with members of the T-cell factor (TCF)/lymphocyte enhancer factor (Lef) family of transcription factors to regulate target gene expression [76].

Wnt ligands are a family of secreted signaling proteins, consisting of 19 members in mammals [77]. Their activities are antagonized by the Secreted frizzled-related proteins (SFRPs) and the dickkopf homologs (DKKs). The SFRP is a family of secreted glycoproteins that may antagonize Wnt-mediated signaling by direct competitive interaction with Wnt ligands or by formation of non-signaling complexes with Frizzled proteins [78], [79]. The DKKs, also secreted cysteine-rich proteins, interact with and inhibit the Wnt co-receptor Lrp5/6 [80].

The array data showed that the Fzd3 was the most abundant receptor and Ctnnb1 and Tcf4 were abundant intracellular transducers expressed in LE and IE cells. While Sfrp2 was highly expressed in LE and IE cells, Dkk2 and Sfrp1 were abundantly expressed in the IE cells, and Apcdd1 was abundant in the LE cells (Table 7). In addition, Dkk2 was 4-fold more abundant in the IE cells, conversely, Apcdd1 was 1.7-fold more abundant in the LE cells.

Among the receptors highly expressed, FZD9 is required for bone morphogenesis and is a receptor for non-canonical Wnt that activates JNK, while DVL3 is required for cardiac outflow tract development [81][84]. Neither, however, is known to be involved in eyelid closure. Conversely, FZD3, involved in axonal outgrowth, and FZD6, required for hair patterning, can collaborate on eyelid closure. Knocking out both Fzd3 and Fzd6 causes “unfused eyelids” in 10% of the offsprings [85], [86]. Likewise, the Lrp6(−/−) mice display multiple defects, including open eyes [87][89]. Although the nuclear factor TCF4 has not been implicated in eyelid closure, TCF3, through interactions with β-catenin, is shown to be crucial for eyelid closure [36], [90].

Using the Wnt reporter mice, it was shown that Wnt activity is repressed overall in eyelid epithelium [36]. The repression is likely to be mediated by the expression of Wnt antagonists. On the one hand, the retinoic acid (RA)-Pitx2 pathway can induce the expression of Wnt antagonists in the periocular mesenchyme; while on the other hand, the BMP and FGFR2 pathways can activate the expression of Wnt antagonists in ocular surface epithelium [13], [91]. Our results showed that antagonists could indeed be produced in the LE and IE cells. Of the antagonists, SFRP4 is dispensable for fetal development; SFRP1 and SFRP2 have redundant functions in regulating embryonic patterning, and DKK2 is required for epithelial differentiation and eyelid closure [12], [92][94]. In addition, APCDD1 is a membrane-bound glycoprotein that can interact with WNT3A and LRP5 and inhibit Wnt signaling in a cell-autonomous manner [95]. Our data also suggested that the LE and IE cells might use distinct antagonists for Wnt inhibition.

In the Wnt reporter mice, it is also shown that the canonical Wnt pathway is activated in restricted areas of the developing eyelids [36]. Specifically, Wnt activity is induced in a small group of epithelial cells positioned at the transition zone between the palpebral conjunctiva and eyelid tip epidermis, so called mucocutaneous junction (MCJ) [96], [97]. Repression of Wnt in the MCJ cells results in failure of eyelid closure [36]. Hence, Wnt may establish distinct morphogenetic fields within the developing eyelids, so that activation takes place in MCJ, but repression occurs elsewhere. Isolation of the MCJ cells and characterizing their molecular signatures may help to understand the developmental roles of the temporal-spatial Wnt activity.

Genes in the SHH, NOTCH and the PCP pathways

The Sonic Hedgehog ligands bind to the transmembrane receptor Patched (Ptch) to initiate pathway signaling [98]. In its inactive state, PTCH exerts an inhibitory effect on the signal transducer Smoothened (SMO), but upon ligand binding, the inhibition on SMO is released and downstream signaling occurs. This leads to the activation of the Gli transcription factors. We found that expression of Ptch1, Smo and Gli2, but not the ligand genes, was relatively abundant in IE and LE cells (Table 8). This is in agreement with the idea that activation of Shh pathway is dependent on Ptch1 expression induced by the FGFR signaling in the eyelid epithelial cells, and the SHH expression induced by FGF10 in the periocular mesenchyme [6], [13]. Furthermore, many of the genes were expressed slightly but significantly higher in LE than in IE cells, suggesting that this pathway may be differentially activated in these cells.

The NOTCH cascade consists of NOTCH, its ligands, and intracellular signal transmitters. Mammals possess four different notch receptors, including NOTCH 1–4, which are membrane-tethered transcription factors. They are activated by the ligands of the Delta, Serrate, Lag-2 families. In LE and IE cells, expression of NOTCH ligands and receptors was overall low, but Jag1 was 1.5-fold and Notch 1 was 1.5-fold more abundant in the LE than in the IE cells (Table 9). The role of NOTCH in eyelid development however has been inconclusive. On the one hand, constitutive activation of NOTCH in periocular mesenchyme leads to abnormalities in cranial facial development and incomplete eyelid closure; on the other hand, genetic ablation of NOTCH signaling in ocular surface epithelium does not cause an EOB phenotype [12], [13], [99][101].

The non-canonical Wnt/planar cell polarity (PCP) pathway regulates cell orientation within the plane of a cell sheet and is involved in convergent extension during development [28], [102]. WNT5A, WNT5B, and WNT11 are the non-canonical WNT ligands, and FZD 3/6 and DVL are the receptors, which transmit signals through the core PCP proteins. The core is composed of cytoplasmic Prickled (PK), the transmembrane protein Van Gogh, the cadherin Starry/Flamingo (STAN/FMI), and the Ankyrin repeat protein Diego (DGO) [103], [104]. In addition, SEC24B is a cargo-binding component of the COPII vesicle coat [105]. The COPII vesicles are the primary pathway for active transport of secretary proteins from the ER to the Golgi. Though SEC24B is not a PCP core component, it selectively sorts VANGL2 into COPII vesicles thereby controlling PCP assembly and activity.

Expression of non-canonical Wnt ligands and core receptors was overall low in LE and IE cells with a few exceptions (Table 10). While expression of Fzd3 and Dvl3 was relatively abundant in LE and IE cells, expression of naked cuticle 1 homolog (Nkd1) was higher in LE, and expression of Sec24b was higher in IE cells. Genetic inactivation of many PCP genes, including Fzd3/6, Dvl2, Vangl2, Scrb1, Ptk7 and Celsr1, as well as Sec24b, causes craniofacial developmental abnormalities, including open eyelids [27][33], [35], [106]. It is yet to be determined whether the eyelid defect is secondary to craniofacial abnormalities resulting from inactivation of the PCP pathways.

Validation of differential gene expression by qRT-PCR

Collectively, the microarray studies identified 20 genes of the morphogenetic signaling pathways were differentially expressed in the LE and IE cells (Fig. 1). To validate the results, we used qRT-PCR to examine 7 relatively abundant genes (Fig. 2A and 2B). Consistent with the array data, qRT-PCR showed that the LE cells had significantly more expression of Erbb2, Gli2 and Notch1, but significantly less expression of Adamts1, Bmpr1a and Dkk2 than the IE cells. Also consistent with the array data, qRT-PCR showed that the LE cells had a slight but insignificant decrease in expression of Tcf4 and Adam17 than the IE cells (Fig. 2C, Tables 4 and 7). Different from the array data, however, qRT-PCR detected no difference of Fgfr2 expression in LE and IE cells (Fig. 2A). Hence, most gene expression pattern observed by cDNA array can be validated by qRT-PCR.

thumbnail
Figure 1. Summary of the microarray analyses.

Genes differentially expressed in LE and IE cells. Statistical significant differential gene expression between LE and IE samples were summarized in (A) genes expressed more in LE than IE cells, and (B) genes expressed less in LE than IE cells. * p<0.05, **p<0.01 and ***p<0.001 are considered significant.

https://doi.org/10.1371/journal.pone.0087038.g001

thumbnail
Figure 2. Differential gene expression in LE and IE cells.

Total RNA isolated from LE and IE cells of fetuses at E15.5 was used for qRT-PCR for the expression of (A) Fgfr2, Errb2, Gli2 and Notch1, (B) Adamts1, Bmpr1a and Dkk2 and (C) Tcf4 and Adam17. Relative expression was calculated based on that of Gapdh in each sample, and compared to the expression in IE cells, set as 1. The results are shown as mean ± SD from at least 3 samples and triplicate PCR of each sample. Statistic analyses were done by Student t-test, ***p<0.001 is considered significant. (D) Figure depicting the LE and IE cells in the developing eyelid and expresison of signaling factors.

https://doi.org/10.1371/journal.pone.0087038.g002

Conclusions

The LE and IE cells have the same ontogenic origin, but different developmental fate. The fate divergence can be detected at E15.5, as the IE cells develop gene expression signatures towards the muscle lineage, while the LE cells express epidermal markers. The LE cells also undergo morphological changes and migrate at E15.5 to eventually form the closed eyelid. This morphogenetic event is thought to be dictated by specific activation of signaling pathways. Our results show that the LE and IE cells are overall quite similar in the compositions for the major “eyelid closure pathways”, but there are a few differences (Fig. 2D). The LE cells have a slight but significant increased expression of Erbb2 of the EGF pathway, Pach1and 2 and Gli2 of the Shh pathway, Jag1 and Notch 1 of the Notch pathway, and Nkd1 of the PCP pathway, but the IE cells have higher expression of Bmpr1a, Acvr1 and Bmp5 of the BMP pathway. In addition, we find higher expression of Apcdd1 in the LE cells, but higher expression of Dkk2 in the IE cells of the Wnt pathway. Differential expression of signaling molecules in the eyelid epithelium may be one of the mechanisms for ectopic activation of morphogenetic pathways. The contributions of the eyelid mesenchyme should also be crucial and can be evaluated using the similar approach. Combination of LCM, cDNA array and pathway analyses can serve as a preliminary screening tool for identifying critical developmental genes for further expression and knockout studie.

Author Contributions

Conceived and designed the experiments: YX MM. Performed the experiments: QM CJ. Analyzed the data: YC JC MM. Contributed reagents/materials/analysis tools: MM. Wrote the paper: QM YX MM.

References

  1. 1. Pearson AA (1980) The development of the eyelids. Part I. External features. J Anat 130: 33–42.
  2. 2. Byun TH, Kim JT, Park HW, Kim WK (2011) Timetable for upper eyelid development in staged human embryos and fetuses. Anat Rec (Hoboken) 294: 789–796.
  3. 3. Harris MJ, McLeod MJ (1982) Eyelid growth and fusion in fetal mice. A scanning electron microscope study. Anat Embryol (Berl) 164: 207–220.
  4. 4. Findlater GS, McDougall RD, Kaufman MH (1993) Eyelid development, fusion and subsequent reopening in the mouse. J Anat 183: 121–9.
  5. 5. Li C, Guo H, Xu X, Weinberg W, Deng CX (2001) Fibroblast growth factor receptor 2 (Fgfr2) plays an important role in eyelid and skin formation and patterning. Dev Dyn 222: 471–483.
  6. 6. Tao H, Ono K, Kurose H, Noji S, Ohuchi H (2006) Exogenous FGF10 can rescue an eye-open at birth phenotype of Fgf10-null mice by activating activin and TGFalpha-EGFR signaling. Dev Growth Differ 48: 339–346.
  7. 7. Harris MJ, Juriloff DM (1986) Eyelid development and fusion induced by cortisone treatment in mutant, lidgap-Miller, foetal mice. A scanning electron microscope study. J Embryol Exp Morphol 91: 1–18.
  8. 8. Juriloff DM, Harris MJ (1989) A scanning electron microscope study of fetal eyelid closure accelerated by cortisone in SWV/Bc mice. Teratology 40: 59–66.
  9. 9. Mohamed YH, Gong H, Amemiya T (2003) Role of apoptosis in eyelid development. Exp Eye Res 76: 115–123.
  10. 10. Matt N, Dupe V, Garnier JM, Dennefeld C, Chambon P, et al. (2005) Retinoic acid-dependent eye morphogenesis is orchestrated by neural crest cells. Development 132: 4789–4800.
  11. 11. Molotkov A, Molotkova N, Duester G (2006) Retinoic acid guides eye morphogenetic movements via paracrine signaling but is unnecessary for retinal dorsoventral patterning. Development 133: 1901–1910.
  12. 12. Gage PJ, Qian M, Wu D, Rosenberg KI (2008) The canonical Wnt signaling antagonist DKK2 is an essential effector of PITX2 function during normal eye development. Dev Biol 317: 310–324.
  13. 13. Huang J, Dattilo LK, Rajagopal R, Liu Y, Kaartinen V, et al. (2009) FGF-regulated BMP signaling is required for eyelid closure and to specify conjunctival epithelial cell fate. Development 136: 1741–1750.
  14. 14. Schaeper U, Vogel R, Chmielowiec J, Huelsken J, Rosario M, et al. (2007) Distinct requirements for Gab1 in Met and EGF receptor signaling in vivo. Proc Natl Acad Sci U S A 104: 15376–15381.
  15. 15. Qu CK, Yu WM, Azzarelli B, Feng GS (1999) Genetic evidence that Shp-2 tyrosine phosphatase is a signal enhancer of the epidermal growth factor receptor in mammals. Proc Natl Acad Sci U S A 96: 8528–33.
  16. 16. Crotty T, Cai J, Sakane F, Taketomi A, Prescott SM, et al. (2006) Diacylglycerol kinase delta regulates protein kinase C and epidermal growth factor receptor signaling. Proc Natl Acad Sci U S A 103: 15485–15490.
  17. 17. Wojnowski L, Stancato LF, Zimmer AM, Hahn H, Beck TW, et al. (1998) Craf-1 protein kinase is essential for mouse development. Mech Dev 76: 141–149.
  18. 18. Scholl FA, Dumesic PA, Barragan DI, Harada K, Bissonauth V, et al. (2007) Mek1/2 MAPK kinases are essential for Mammalian development, homeostasis, and Raf-induced hyperplasia. Dev Cell 12: 615–629.
  19. 19. Schwartzberg PL, Stall AM, Hardin JD, Bowdish KS, Humaran T, et al. (1991) Mice homozygous for the ablm1 mutation show poor viability and depletion of selected B and T cell populations. Cell 65: 1165–1175.
  20. 20. Zhang L, Wang W, Hayashi Y, Jester JV, Birk DE, et al. (2003) A role for MEK kinase 1 in TGF-beta/activin-induced epithelium movement and embryonic eyelid closure. Embo J 22: 4443–4454.
  21. 21. Takatori A, Geh E, Chen L, Zhang L, Meller J, et al. (2008) Differential transmission of MEKK1 morphogenetic signals by JNK1 and JNK2. Development 135: 23–32.
  22. 22. Weston CR, Wong A, Hall JP, Goad ME, Flavell RA, et al. (2003) JNK initiates a cytokine cascade that causes Pax2 expression and closure of the optic fissure. Genes Dev 17: 1271–1280.
  23. 23. Schramek D, Kotsinas A, Meixner A, Wada T, Elling U, et al. (2011) The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression. Nat Genet 43: 212–219.
  24. 24. Thumkeo D, Shimizu Y, Sakamoto S, Yamada S, Narumiya S (2005) ROCK-I and ROCK-II cooperatively regulate closure of eyelid and ventral body wall in mouse embryo. Genes Cells 10: 825–834.
  25. 25. Rice DS, Hansen GM, Liu F, Crist MJ, Newhouse MM, et al. (2012) Keratinocyte migration in the developing eyelid requires LIMK2. PLoS ONE 7: e47168.
  26. 26. Shimizu Y, Thumkeo D, Keel J, Ishizaki T, Oshima H, et al. (2005) ROCK-I regulates closure of the eyelids and ventral body wall by inducing assembly of actomyosin bundles. J Cell Biol 168: 941–953.
  27. 27. Kibar Z, Vogan KJ, Groulx N, Justice MJ, Underhill DA, et al. (2001) Ltap, a mammalian homolog of Drosophila Strabismus/Van Gogh, is altered in the mouse neural tube mutant Loop-tail. Nat Genet 28: 251–255.
  28. 28. Murdoch JN, Doudney K, Paternotte C, Copp AJ, Stanier P (2001) Severe neural tube defects in the loop-tail mouse result from mutation of Lpp1, a novel gene involved in floor plate specification. Hum Mol Genet 10: 2593–2601.
  29. 29. Hamblet NS, Lijam N, Ruiz-Lozano P, Wang J, Yang Y, et al. (2002) Dishevelled 2 is essential for cardiac outflow tract development, somite segmentation and neural tube closure. Development 129: 5827–5838.
  30. 30. Curtin JA, Quint E, Tsipouri V, Arkell RM, Cattanach B, et al. (2003) Mutation of Celsr1 disrupts planar polarity of inner ear hair cells and causes severe neural tube defects in the mouse. Curr Biol 13: 1129–1133.
  31. 31. Lu X, Borchers AG, Jolicoeur C, Rayburn H, Baker JC, et al. (2004) PTK7/CCK-4 is a novel regulator of planar cell polarity in vertebrates. Nature 430: 93–98.
  32. 32. Montcouquiol M, Rachel RA, Lanford PJ, Copeland NG, Jenkins NA, et al. (2003) Identification of Vangl2 and Scrb1 as planar polarity genes in mammals. Nature 423: 173–177.
  33. 33. Harris MJ, Juriloff DM (2007) Mouse mutants with neural tube closure defects and their role in understanding human neural tube defects. Birth Defects Res A Clin Mol Teratol 79: 187–210.
  34. 34. Rivera C, Simonson SJ, Yamben IF, Shatadal S, Nguyen MM, et al. (2013) Requirement for Dlgh-1 in planar cell polarity and skeletogenesis during vertebrate development. PLoS ONE 8: e54410.
  35. 35. Torban E, Patenaude AM, Leclerc S, Rakowiecki S, Gauthier S, et al. (2008) Genetic interaction between members of the Vangl family causes neural tube defects in mice. Proc Natl Acad Sci U S A 105: 3449–3454.
  36. 36. Wu CI, Hoffman JA, Shy BR, Ford EM, Fuchs E, et al. (2012) Function of Wnt/beta-catenin in counteracting Tcf3 repression through the Tcf3-beta-catenin interaction. Development 139: 2118–2129.
  37. 37. Luetteke NC, Qiu TH, Fenton SE, Troyer KL, Riedel RF, et al. (1999) Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. Development 126: 2739–2750.
  38. 38. Luetteke NC, Qiu TH, Peiffer RL, Oliver P, Smithies O, et al. (1993) TGF alpha deficiency results in hair follicle and eye abnormalities in targeted and waved-1 mice. Cell 73: 263–78.
  39. 39. Vassalli A, Matzuk MM, Gardner HA, Lee KF, Jaenisch R (1994) Activin/inhibin beta B subunit gene disruption leads to defects in eyelid development and female reproduction. Genes Dev 8: 414–27.
  40. 40. Jin C, Chen J, Meng Q, Carreira V, Tam NN, et al.. (2012) Deciphering gene expression program of MAP3K1 in mouse eyelid morphogenesis. Dev Biol.
  41. 41. Schnekenburger M, Peng L, Puga A (2007) HDAC1 bound to the Cyp1a1 promoter blocks histone acetylation associated with Ah receptor-mediated trans-activation Biochim Biophys Acta. 1769: 569–578.
  42. 42. Smyth GK (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3: Article3.
  43. 43. Dai M, Wang P, Boyd AD, Kostov G, Athey B, et al. (2005) Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res 33: e175.
  44. 44. Kauffmann A, Gentleman R, Huber W (2009) arrayQualityMetrics–a bioconductor package for quality assessment of microarray data. Bioinformatics 25: 415–416.
  45. 45. Sartor MA, Leikauf GD, Medvedovic M (2009) LRpath: a logistic regression approach for identifying enriched biological groups in gene expression data. Bioinformatics 25: 211–217.
  46. 46. Freudenberg JM, Joshi VK, Hu Z, Medvedovic M (2009) CLEAN: CLustering Enrichment ANalysis. BMC Bioinformatics 10: 234.
  47. 47. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, et al. (2000) Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25: 25–29.
  48. 48. Reuss B, von Bohlen und HO (2003) Fibroblast growth factors and their receptors in the central nervous system. Cell Tissue Res 313: 139–157.
  49. 49. Goetz R, Mohammadi M (2013) Exploring mechanisms of FGF signalling through the lens of structural biology. Nat Rev Mol Cell Biol 14: 166–180.
  50. 50. Al AD, Sala FG, Baptista S, Galzote R, Danopoulos S, et al. (2012) FGF9-Pitx2-FGF10 signaling controls cecal formation in mice. Dev Biol 369: 340–348.
  51. 51. Okada K, Noda M, Nogawa H (2013) Autocrine growth factors are involved in branching morphogenesis of mouse lung epithelium. Zoolog Sci 30: 1–6.
  52. 52. Colvin JS, Green RP, Schmahl J, Capel B, Ornitz DM (2001) Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell 104: 875–889.
  53. 53. Colvin JS, White AC, Pratt SJ, Ornitz DM (2001) Lung hypoplasia and neonatal death in Fgf9-null mice identify this gene as an essential regulator of lung mesenchyme. Development 128: 2095–2106.
  54. 54. Pirvola U, Zhang X, Mantela J, Ornitz DM, Ylikoski J (2004) Fgf9 signaling regulates inner ear morphogenesis through epithelial-mesenchymal interactions. Dev Biol 273: 350–360.
  55. 55. Zenz R, Scheuch H, Martin P, Frank C, Eferl R, et al. (2003) c-Jun Regulates Eyelid Closure and Skin Tumor Development through EGFR Signaling. Dev Cell 4: 879–889.
  56. 56. Li G, Gustafson-Brown C, Hanks SK, Nason K, Arbeit JM, et al. (2003) c-Jun Is Essential for Organization of the Epidermal Leading Edge. Dev Cell 4: 865–877.
  57. 57. Hynes NE, Lane HA (2005) ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5: 341–354.
  58. 58. Dhomen NS, Mariadason J, Tebbutt N, Scott AM (2012) Therapeutic targeting of the epidermal growth factor receptor in human cancer. Crit Rev Oncog 17: 31–50.
  59. 59. Miettinen PJ, Berger JE, Meneses J, Phung Y, Pedersen RA, et al. (1995) Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 376: 337–41.
  60. 60. Saftig P, Reiss K (2011) The “A Disintegrin And Metalloproteases” ADAM10 and ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol 90: 527–535.
  61. 61. Hassemer EL, Endres B, Toonen JA, Ronchetti A, Dubielzig R, et al.. (2012) ADAM17 Transactivates EGFR Signaling During Embryonic Eyelid Closure. Invest Ophthalmol Vis Sci.
  62. 62. Le Gall SM, Bobe P, Reiss K, Horiuchi K, Niu XD, et al. (2009) ADAMs 10 and 17 represent differentially regulated components of a general shedding machinery for membrane proteins such as transforming growth factor alpha, L-selectin, and tumor necrosis factor alpha. Mol Biol Cell 20: 1785–1794.
  63. 63. Scheller J, Chalaris A, Garbers C, Rose-John S (2011) ADAM17: a molecular switch to control inflammation and tissue regeneration. Trends Immunol 32: 380–387.
  64. 64. Kato S, Mohri Y, Matsuo T, Ogawa E, Umezawa A, et al. (2007) Eye-open at birth phenotype with reduced keratinocyte motility in LGR4 null mice. FEBS Lett 581: 4685–4690.
  65. 65. Wang Z, Jin C, Li H, Li C, Hou Q, et al. (2010) GPR48-Induced keratinocyte proliferation occurs through HB-EGF mediated EGFR transactivation. FEBS Lett 584: 4057–4062.
  66. 66. Mann GB, Fowler KJ, Gabriel A, Nice EC, Williams RL, et al. (1993) Mice with a null mutation of the TGF alpha gene have abnormal skin architecture, wavy hair, and curly whiskers and often develop corneal inflammation. Cell 73: 249–61.
  67. 67. Mueller TD, Nickel J (2012) Promiscuity and specificity in BMP receptor activation. FEBS Lett 586: 1846–1859.
  68. 68. He W, Li AG, Wang D, Han S, Zheng B, et al. (2002) Overexpression of Smad7 results in severe pathological alterations in multiple epithelial tissues EMBO J. 21: 2580–2590.
  69. 69. Andl T, Ahn K, Kairo A, Chu EY, Wine-Lee L, et al. (2004) Epithelial Bmpr1a regulates differentiation and proliferation in postnatal hair follicles and is essential for tooth development. Development 131: 2257–2268.
  70. 70. Bailon-Plaza A, Lee AO, Veson EC, Farnum CE, van der Meulen MC (1999) BMP-5 deficiency alters chondrocytic activity in the mouse proximal tibial growth plate. Bone 24: 211–216.
  71. 71. Zhao R, Lawler AM, Lee SJ (1999) Characterization of GDF-10 expression patterns and null mice. Dev Biol 212: 68–79.
  72. 72. Matsumoto Y, Otsuka F, Hino J, Miyoshi T, Takano M, et al. (2012) Bone morphogenetic protein-3b (BMP-3b) inhibits osteoblast differentiation via Smad2/3 pathway by counteracting Smad1/5/8 signaling. Mol Cell Endocrinol 350: 78–86.
  73. 73. Dudley AT, Lyons KM, Robertson EJ (1995) A requirement for bone morphogenetic protein-7 during development of the mammalian kidney and eye. Genes Dev 9: 2795–2807.
  74. 74. Bhanot P, Brink M, Samos CH, Hsieh JC, Wang Y, et al. (1996) A new member of the frizzled family from Drosophila functions as a Wingless receptor. Nature 382: 225–230.
  75. 75. Wehrli M, Dougan ST, Caldwell K, O'Keefe L, Schwartz S, et al. (2000) arrow encodes an LDL-receptor-related protein essential for Wingless signalling. Nature 407: 527–530.
  76. 76. van de WM, Cavallo R, Dooijes D, van BM, van EJ, et al. (1997) Armadillo coactivates transcription driven by the product of the Drosophila segment polarity gene dTCF. Cell 88: 789–799.
  77. 77. Takada R, Satomi Y, Kurata T, Ueno N, Norioka S, et al. (2006) Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Dev Cell 11: 791–801.
  78. 78. Dann CE, Hsieh JC, Rattner A, Sharma D, Nathans J, et al. (2001) Insights into Wnt binding and signalling from the structures of two Frizzled cysteine-rich domains. Nature 412: 86–90.
  79. 79. Bafico A, Gazit A, Pramila T, Finch PW, Yaniv A, et al. (1999) Interaction of frizzled related protein (FRP) with Wnt ligands and the frizzled receptor suggests alternative mechanisms for FRP inhibition of Wnt signaling. J Biol Chem 274: 16180–16187.
  80. 80. He X, Semenov M, Tamai K, Zeng X (2004) LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way. Development 131: 1663–1677.
  81. 81. Ranheim EA, Kwan HC, Reya T, Wang YK, Weissman IL, et al. (2005) Frizzled 9 knock-out mice have abnormal B-cell development. Blood 105: 2487–2494.
  82. 82. Heasley LE, Winn RA (2008) Analysis of Wnt7a-stimulated JNK activity and cJun phosphorylation in non-small cell lung cancer cells. Methods Mol Biol 468: 187–196.
  83. 83. Albers J, Schulze J, Beil FT, Gebauer M, Baranowsky A, et al. (2011) Control of bone formation by the serpentine receptor Frizzled-9. J Cell Biol 192: 1057–1072.
  84. 84. Etheridge SL, Ray S, Li S, Hamblet NS, Lijam N, et al. (2008) Murine dishevelled 3 functions in redundant pathways with dishevelled 1 and 2 in normal cardiac outflow tract, cochlea, and neural tube development. PLoS Genet 4: e1000259.
  85. 85. Wang Y, Thekdi N, Smallwood PM, Macke JP, Nathans J (2002) Frizzled-3 is required for the development of major fiber tracts in the rostral CNS. J Neurosci 22: 8563–8573.
  86. 86. Guo N, Hawkins C, Nathans J (2004) Frizzled6 controls hair patterning in mice. Proc Natl Acad Sci U S A 101: 9277–9281.
  87. 87. Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, et al. (2002) Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol 157: 303–314.
  88. 88. Zhou CJ, Molotkov A, Song L, Li Y, Pleasure DE, et al. (2008) Ocular coloboma and dorsoventral neuroretinal patterning defects in Lrp6 mutant eyes. Dev Dyn 237: 3681–3689.
  89. 89. Zhou CJ, Wang YZ, Yamagami T, Zhao T, Song L, et al. (2010) Generation of Lrp6 conditional gene-targeting mouse line for modeling and dissecting multiple birth defects/congenital anomalies. Dev Dyn 239: 318–326.
  90. 90. Korinek V, Barker N, Moerer P, van DE, Huls G, et al. (1998) Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet 19: 379–383.
  91. 91. Kumar S, Duester G (2010) Retinoic acid signaling in perioptic mesenchyme represses Wnt signaling via induction of Pitx2 and Dkk2. Dev Biol 340: 67–74.
  92. 92. Satoh W, Gotoh T, Tsunematsu Y, Aizawa S, Shimono A (2006) Sfrp1 and Sfrp2 regulate anteroposterior axis elongation and somite segmentation during mouse embryogenesis. Development 133: 989–999.
  93. 93. Christov M, Koren S, Yuan Q, Baron R, Lanske B (2011) Genetic ablation of sfrp4 in mice does not affect serum phosphate homeostasis. Endocrinology 152: 2031–2036.
  94. 94. Mukhopadhyay M, Gorivodsky M, Shtrom S, Grinberg A, Niehrs C, et al. (2006) Dkk2 plays an essential role in the corneal fate of the ocular surface epithelium. Development 133: 2149–2154.
  95. 95. Shimomura Y, Agalliu D, Vonica A, Luria V, Wajid M, et al. (2010) APCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex. Nature 464: 1043–1047.
  96. 96. Knop E, Korb DR, Blackie CA, Knop N (2010) The lid margin is an underestimated structure for preservation of ocular surface health and development of dry eye disease. Dev Ophthalmol 45: 108–122.
  97. 97. Riau AK, Barathi VA, Beuerman RW (2008) Mucocutaneous junction of eyelid and lip: a study of the transition zone using epithelial cell markers. Curr Eye Res 33: 912–922.
  98. 98. Rubin LL, de Sauvage FJ (2006) Targeting the Hedgehog pathway in cancer. Nat Rev Drug Discov 5: 1026–1033.
  99. 99. Zhang Y, Lam O, Nguyen MT, Ng G, Pear WS, et al. (2013) Mastermind-like transcriptional co-activator-mediated Notch signaling is indispensable for maintaining conjunctival epithelial identity. Development 140: 594–605.
  100. 100. Saravanamuthu SS, Le TT, Gao CY, Cojocaru RI, Pandiyan P, et al. (2012) Conditional ablation of the Notch2 receptor in the ocular lens. Dev Biol 362: 219–229.
  101. 101. Zhang Y, Kao WW, Pelosi E, Schlessinger D, Liu CY (2011) Notch gain of function in mouse periocular mesenchyme downregulates FoxL2 and impairs eyelid levator muscle formation, leading to congenital blepharophimosis. J Cell Sci 124: 2561–2572.
  102. 102. Wang Y, Nathans J (2007) Tissue/planar cell polarity in vertebrates: new insights and new questions. Development 134: 647–658.
  103. 103. Adler PN (2002) Planar signaling and morphogenesis in Drosophila. Dev Cell 2: 525–535.
  104. 104. Seifert JR, Mlodzik M (2007) Frizzled/PCP signalling: a conserved mechanism regulating cell polarity and directed motility. Nat Rev Genet 8: 126–138.
  105. 105. Miller EA, Beilharz TH, Malkus PN, Lee MC, Hamamoto S, et al. (2003) Multiple cargo binding sites on the COPII subunit Sec24p ensure capture of diverse membrane proteins into transport vesicles. Cell 114: 497–509.
  106. 106. Merte J, Jensen D, Wright K, Sarsfield S, Wang Y, et al. (2010) Sec24b selectively sorts Vangl2 to regulate planar cell polarity during neural tube closure. Nat Cell Biol 12: 41–46.