The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.

The choroid plexus (ChP) is a sheet of predominantly epithelial cells that produces cerebrospinal fluid (CSF), secretes factors important for brain development and forms a crucial brain barrier (Chau et al., 2015; Fame and Lehtinen, 2020; Ghersi-Egea et al., 2018; Lehtinen et al., 2011; Silva-Vargas et al., 2016). ChP tissue is specified during the early stages of brain development (Hunter and Dymecki, 2007), and forms at, or near, the dorsal midline in each brain ventricle (lateral ventricle, LV; third ventricle, 3V; fourth ventricle, 4V; Currle et al., 2005). As progenitor cells proliferate and mature into epithelial cells, the epithelial sheet extends in a conveyor belt-like manner into the ventricles (Dani et al., 2021; Liddelow et al., 2010). Interactions between the maturing epithelial cells and the surrounding cellular network of mesenchymal (Wilting and Christ, 1989) and vascular (Nielsen and Dymecki, 2010) cells transform the growing epithelium to adopt its mature form.

The ChP tissues are regionally patterned, and they harbor distinct transcriptomes resulting in ventricle-specific secretomes (Dani et al., 2021; Lun et al., 2015). These findings raise the possibility that local CSF environments are tailored to instruct development of adjacent brain areas. Indeed, the 4V ChP expresses high levels of WNT5A, which is released into the CSF, associates with lipoprotein particles and influences hindbrain morphogenesis (Kaiser et al., 2019). In mice, the ChP also appears morphologically distinct in different ventricles; the LV ChP consists of two nearly flat, leaf-like epithelial sheets, whereas in the 3V and 4V the ChP adopts a more complex, frond-like structure (Dani et al., 2021). Despite a general understanding of the key steps required for ChP formation, surprisingly little is known about the molecular mechanisms underlying differences in ChP morphogenesis in each of these ventricles.

WNT5A signaling represents one compelling candidate signaling pathway for regulating regional ChP development. Wnt5a transcription and production are enriched in distally located mature epithelial cells in 4V ChP (Kaiser et al., 2019), whereas mesenchymal Wnt5a expression is predominantly localized to the proximal region extending to the core of all ChP (Dani et al., 2021; Langford et al., 2020). Systemic Wnt5a deficiency was recently reported to impair ChP development in the LV, 3V and 4V ChP (Langford et al., 2020), but the specific role of WNT5A in ChP epithelial cells has not been studied. As such, the molecular mechanisms that regulate the regionalized expression of Wnt5a in the ChP and its time- and cell type-restricted functions remain to be elucidated.

Overall, WNT signaling encompasses highly conserved signaling pathways involved in the regulation of numerous physiological processes during embryonic development and in adulthood (Saito-Diaz et al., 2013). WNT proteins (WNTs) represent a large family of lipid-modified glycoproteins acting as extracellular ligands that activate either a canonical cascade, mediated by active β-catenin, or the non-canonical branches of the WNT pathway, which depend on ligand binding to a large repertoire of cognate receptors (Niehrs, 2012). Among the WNTs, WNT5A represents a prototypical non-canonical WNT ligand, predominantly linked to the activation of the planar cell polarity (WNT/PCP) pathway (Kumawat and Gosens, 2015), where it influences various aspects of tissue patterning and establishment of cell polarity (Humphries and Mlodzik, 2018). Notably, WNT5A mediates conserved roles in the outgrowth of body structures, including budding tentacles in Hydra or limbs and distal digits in mouse. In these tissues, WNT5A exhibits similar spatial patterns of expression with upregulation in the most distal regions of the outgrowing structures (Philipp et al., 2009; Yamaguchi et al., 1999). As such, WNT5A, with its expression mainly restricted to the epithelium of 4V ChP, is an ideal candidate factor for regulating the formation of the complex frond-like shape of the 4V ChP.

Here, we elucidate dual roles of WNT5A in 4V ChP development. First, at early stages of ChP development, Wnt5a temporal expression and dosage are essential for establishing the blueprint for ChP morphogenesis and size. Second, at later stages of ChP development, WNT5A expression and secretion are enriched at the distal tips of the 4V ChP epithelium where WNT5A acts either locally to activate components of the WNT/PCP pathway in an autocrine manner or is released into the CSF for long-range signaling. At this later developmental stage, MEIS1 binds to the Wnt5a proximal promoter to regulate Wnt5a expression in the 4V ChP. Collectively, our findings reveal two distinct developmental stages at which WNT5A plays important roles in establishing 4V ChP form and function.

Epithelial WNT5A expression is required for 4V ChP morphogenesis

First, we confirmed our previous observation showing that, during development [embryonic day (E) 14.5], Wnt5a expression is enriched in the mouse 4V ChP epithelium in contrast to the LV ChP (Kaiser et al., 2019; Lun et al., 2015) (Fig. 1A,B). Wnt5a expression was most prominent at the distal tips of epithelial villi, with protein signal also present in mesenchymal cells of the stromal space (Fig. 1C, filled and unfilled arrowheads; white dashed line demarcates the distal part of each ChP villus, red line demarcates base of the villi). In 4V ChP, we observed segregation of WNT5A signal to the apical cytoplasm, with aquaporin 1 (AQP1) staining highlighting epithelial membrane (Fig. 1D, top, arrowheads), and to the basolateral cell membrane (stained with collagen IV) and mesenchymal cells (Fig. S1A). Using a ChP epithelium-based transwell system, we confirmed bi-directional secretion of WNT5A apically and basally by embryonic 4V ChP epithelia (Fig. S1B). Analysis of human gestational week 9 (GW9) brain specimens confirmed WNT5A expression on the apical and basolateral sides of the 4V ChP epithelium (Fig. 1D, bottom, filled and unfilled arrowheads, respectively) suggesting that Wnt5a may have evolutionarily conserved roles in the ChP.

Fig. 1.

Wnt5a is expressed by 4V ChP epithelium and controls its morphogenesis. (A) Schematic of E14.5 LV and 4V ChP showing the apical side of the epithelial layer (AQP1+) facing the CSF. A, P, anteroposterior axis; D, V, dorsoventral axis; LRL, lower rhombic lip; URL, upper rhombic lip. (B) In situ analysis combined with immunofluorescence staining of E14.5 coronal sections shows Wnt5a expression restricted to the ChP epithelium of 4V ChP (filled arrowheads) with minimal expression in the LV ChP epithelium (arrows). Wnt5a expression is largely absent from the stromal compartment (unfilled arrowheads; red dashed line demarcates basal lamina). AQP1 is a marker of ChP epithelium. Scale bars: 20 µm. (C) In situ hybridization (ISH) analysis and immunofluorescence (IF) of a coronal section showing that Wnt5a expression and protein levels are enriched in the distal tips (white dashed line, distal portion of the epithelial villi; filled arrowheads, tip of the villi) of E16.5 4V ChP epithelium villi whereas Wnt5a expression level observed at their base (red line) is low. Foxj1 is a marker of differentiated epithelium. IF analysis reveals that the WNT5A signal is restricted predominantly to the distal tips of E16.5 4V ChP epithelium villi (filled arrowheads; white dashed line) as opposed to the base of the villi (unfilled arrowheads; red line). AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (D) Immunostaining showing WNT5A signal in both the apical (filled arrowheads) and basolateral (unfilled arrowhead) portion of E14.5 mouse (top) and 9 weeks gestation human (bottom) 4V ChP epithelium. AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (E) Immunostaining of the 4V ChP between Wnt5aWT versus Wnt5aKO showing the greatly reduced 4V ChP growth and altered morphology in Wnt5aKO embryo at E16.5. Scale bars: 100 µm. (F-H) Analysis of E16.5 4V ChP morphology indicates a significant decrease in total area (F), the number of villi (G) and average villus length (H) between Wnt5aWT and Wnt5aKO. Graphs show n=4-6 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. (I) Immunostaining of E16.5 4V ChP in Wnt5aWT or Wnt5acKO embryos with recombination induced at E11.5 (iE11.5) or E12.5 (iE12.5). AQP1 staining demonstrates a reduction in the size and alteration in the morphology of 4V ChP in Wnt5acKO embryos with recombination induced at E11.5 but no effect on branching morphology of 4V ChP in Wnt5acKO embryos with recombination induced at E12.5. Scale bars: 100 µm. (J-L) Analysis of E16.5 4V ChP morphology indicates significant decrease in total area (J), the number of villi (K) and average villus length (L) between Wnt5aWT and Wnt5acKO. Graphs show n=4 biologically independent samples; error bars represent s.d.; P-values from two-tailed unpaired Student's t-tests.

Fig. 1.

Wnt5a is expressed by 4V ChP epithelium and controls its morphogenesis. (A) Schematic of E14.5 LV and 4V ChP showing the apical side of the epithelial layer (AQP1+) facing the CSF. A, P, anteroposterior axis; D, V, dorsoventral axis; LRL, lower rhombic lip; URL, upper rhombic lip. (B) In situ analysis combined with immunofluorescence staining of E14.5 coronal sections shows Wnt5a expression restricted to the ChP epithelium of 4V ChP (filled arrowheads) with minimal expression in the LV ChP epithelium (arrows). Wnt5a expression is largely absent from the stromal compartment (unfilled arrowheads; red dashed line demarcates basal lamina). AQP1 is a marker of ChP epithelium. Scale bars: 20 µm. (C) In situ hybridization (ISH) analysis and immunofluorescence (IF) of a coronal section showing that Wnt5a expression and protein levels are enriched in the distal tips (white dashed line, distal portion of the epithelial villi; filled arrowheads, tip of the villi) of E16.5 4V ChP epithelium villi whereas Wnt5a expression level observed at their base (red line) is low. Foxj1 is a marker of differentiated epithelium. IF analysis reveals that the WNT5A signal is restricted predominantly to the distal tips of E16.5 4V ChP epithelium villi (filled arrowheads; white dashed line) as opposed to the base of the villi (unfilled arrowheads; red line). AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (D) Immunostaining showing WNT5A signal in both the apical (filled arrowheads) and basolateral (unfilled arrowhead) portion of E14.5 mouse (top) and 9 weeks gestation human (bottom) 4V ChP epithelium. AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (E) Immunostaining of the 4V ChP between Wnt5aWT versus Wnt5aKO showing the greatly reduced 4V ChP growth and altered morphology in Wnt5aKO embryo at E16.5. Scale bars: 100 µm. (F-H) Analysis of E16.5 4V ChP morphology indicates a significant decrease in total area (F), the number of villi (G) and average villus length (H) between Wnt5aWT and Wnt5aKO. Graphs show n=4-6 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. (I) Immunostaining of E16.5 4V ChP in Wnt5aWT or Wnt5acKO embryos with recombination induced at E11.5 (iE11.5) or E12.5 (iE12.5). AQP1 staining demonstrates a reduction in the size and alteration in the morphology of 4V ChP in Wnt5acKO embryos with recombination induced at E11.5 but no effect on branching morphology of 4V ChP in Wnt5acKO embryos with recombination induced at E12.5. Scale bars: 100 µm. (J-L) Analysis of E16.5 4V ChP morphology indicates significant decrease in total area (J), the number of villi (K) and average villus length (L) between Wnt5aWT and Wnt5acKO. Graphs show n=4 biologically independent samples; error bars represent s.d.; P-values from two-tailed unpaired Student's t-tests.

We analyzed the functional consequences of Wnt5a deficiency employing several different mouse models. Phenotypic analysis of Wnt5a null mutants (Wnt5aKO) (Yamaguchi et al., 1999) revealed severe impairment of 4V ChP development as demonstrated by decreased size and reduced branching morphology compared with wild-type control embryos (Wnt5aWT) (Fig. 1E-H; Fig. S1C), in agreement with others (Langford et al., 2020). In the most severe Wnt5aKO cases, the 4V ChP lacked its characteristic convoluted structure and resembled instead the simpler sheet-like shape of the LV ChP (Fig. S1D). We also confirmed the previously reported disruption of LV ChP morphogenesis in Wnt5aKO embryos (Langford et al., 2020), which was characterized by the collapsed growth and impaired protrusion of the tissue into the lumen of the ventricle (Fig. S1E) accompanied by general shortening of the tissue (Fig. S1F,G). We investigated whether the observed morphological differences in 4V ChP were due to altered proliferative or apoptotic activity. No differences were observed in the 5-ethynyl-2′-deoxyuridine (EdU) incorporation rate (delivered at E11.5 and analyzed 24 h later) of mature epithelial cells (AQP1+) in the 4V ChP epithelium of Wnt5aKO ChP (Fig. S1H,I). Similarly, no changes in apoptotic activity analyzed by cleaved caspase 3 (CASP3) staining were detected in the embryonic 4V ChP between Wnt5aKO and control littermates (Fig. S1J). This suggests that any possible contribution of altered proliferation and apoptosis to the observed phenotypes occurred earlier in development.

Next, we conditionally deleted Wnt5a (Wnt5acKO) in ChP epithelial cells using an inducible Foxj1 promoter-driven CREERT2 system (Kaiser et al., 2019) (Fig. S2A,B). In line with the reported properties of the Cre-lox recombination system (Nakamura et al., 2006), we observed the loss of WNT5A protein approximately 24 h following tamoxifen injection (Fig. S2C). When tamoxifen was delivered at E12.5, tissue-specific WNT5A ablation in 4V ChP epithelium of Wnt5acKO versus control embryos persisted at least until E16.5 (Fig. S2D). WNT5A was readily detected in the lower rhombic lip (LRL) domain adjacent to the 4V ChP where Foxj1 is not expressed in both E16.5 Wnt5aWT and Wnt5acKO embryos (Fig. S2E). We also observed loss of WNT5A immunoreactivity in the stromal compartment at the tips of distal villi (Fig. S2D) as well as in the mesenchyme of more proximal regions (Fig. S2E) in the Wnt5acKO 4V ChP. As such, WNT5A signal detected in the mesenchymal cells located at the distal tip of ChP villi likely represents predominantly epithelial-derived WNT5A. We failed to observe disrupted AQP1 localization in 4V ChP epithelial cells, as suggested in other studies (Langford et al., 2020).

The specific timing of Wnt5a ablation had strikingly different effects on the developing 4V ChP. Induction of Cre recombination by tamoxifen injection at E11.5 profoundly disrupted the overall size and branching morphology of the 4V ChP in Wnt5acKO (Fig. 1I-L), when analyzed at E16.5. In contrast, induction of Cre recombination only 24 h later, at E12.5, failed to induce a detectable phenotype in the E16.5 4V ChP (Fig. 1I-L). The morphological defects observed following E11.5 Cre-recombination were regionalized within the 4V ChP, being most pronounced in the ventral part of the 4V ChP, adjacent to the LRL region compared with the more lateral region of the ChP adjacent to the upper rhombic lip (URL) region (Fig. S2F, filled arrowheads). Consistent with the systemic Wnt5aKO embryos and previous studies (Langford et al., 2020), no differences in either proliferation (EdU+ cells) or apoptotic cell death (cleaved CASP3) markers were observed at E12.5, despite the disrupted 4V ChP morphogenesis (Fig. S2G-I). In contrast to findings in systemic Wnt5aKO embryos (Fig. S1E-G), we failed to detect morphological changes in the developing LV ChP in Wnt5acKO embryos (Fig. S2J-L). Taken together, these data demonstrate that Wnt5a expression between E11.5 and E12.5 is essential for establishing the blueprint for 4V ChP morphogenesis and size, and Wnt5a deficiency at this early stage has profound, long-lasting consequences on ChP morphology.

Wnt5a overexpression during late embryogenesis results in defective morphogenesis of embryonic ChPs

Wnt5a dosage, demonstrated by both ablation and ectopic overexpression studies, disrupts the morphogenesis of the small intestine (Cervantes et al., 2009; van Amerongen et al., 2012), a simple columnar epithelium with similarities to the ChP (Grosse et al., 2011). To determine the consequences of supplemental WNT5A on 4V ChP, we employed a previously established model for Wnt5a overexpression (Wnt5aOE) (van Amerongen et al., 2012). In these mice, despite being driven by the Rosa26 promoter, higher levels of FLAG-WNT5A were detected in embryonic 4V ChP compared with the surrounding regions (Fig. 2A, arrowheads). We also observed FLAG-WNT5A expression in other brain regions that typically express Wnt5a, including in close proximity to the cortical hem (Fig. 2B, arrowheads; Fig. S3A, arrowheads). In the 4V ChP epithelium, FLAG-WNT5A localized to the cytoplasm of epithelial cells and revealed limited local spreading within the surrounding 4V ChP extracellular space (Fig. 2C, filled and unfilled arrowheads). The reasons for spatially restricted overexpression of WNT5A despite using the Rosa26 promoter remain unknown but this has been observed in other tissues (personal communication, R.v.A.). These observations may result from post-transcriptional as well as post-translational regulation of WNT5A in cells that either express or do not express Wnt5a endogenously.

Fig. 2.

Ectopic Wnt5a expression impairs morphogenesis of LV ChP and 4V ChP. (A) Immunostaining of coronal sections showing an induction of Flag-tagged WNT5A (FLAG-WNT5A) overexpression in 4V ChP, but not in the adjacent regions (arrowheads), in Wnt5aOE embryos compared with Wnt5aWT littermates. AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (B) Immunostaining of coronal sections showing induction of FLAG-WNT5A overexpression in LV ChP in Wnt5aOE embryos compared with Wnt5aWT littermates. Induction of ectopic FLAG-WNT5A protein is also observed in the adjacent cortical hem (arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (C) Higher magnification view showing signal for the ectopic FLAG-WNT5A, which is mostly restricted to the cytoplasm of cells producing FLAG-WNT5A in 4V ChP epithelium (arrowheads); no FLAG-WNT5A is detected in the basal lamina or stroma compared with endogenous WNT5A (unfilled arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (D) Immunostaining of coronal sections demonstrates altered 4V ChP morphology in Wnt5aOE compared with Wnt5aWT at E14.5. Higher magnification images show a detailed view of the 4V ChP (dashed box) in both Wnt5aWT and Wnt5aOE embryos. AQP1 is a marker of ChP epithelium. Scale bars: 300 µm (main panels); 100 µm (higher magnification panels). (E-G) Analysis of E14.5 4V ChP morphology indicates that there is a significant decrease in total area (E), the number of villi (F) and average villus length (G) between Wnt5aWT and Wnt5aOE. Graphs show n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. Biological replicates are indicated in the graph. (H) Higher magnification view shows that ectopic production of FLAG-WNT5A in Wnt5aOE embryos results in detection of the WNT5A signal in LV ChP epithelial cells (filled arrowheads) that normally display very low levels of WNT5A signal as observed in Wnt5aWT LV ChP epithelium (unfilled arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (I) Immunostaining of coronal sections demonstrates altered LV ChP morphology between Wnt5aWT and Wnt5aOE at E14.5. AQP1 is a marker of ChP epithelium. Scale bars: 100 µm. (J,K) Analysis of E14.5 LV ChP morphology indicates that there is a less robust decrease (compared with that observed for 4V ChP) in total area (J) and average length (K) between Wnt5aWT and Wnt5aOE. Graphs show n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. Biological replicates are indicated in the graph.

Fig. 2.

Ectopic Wnt5a expression impairs morphogenesis of LV ChP and 4V ChP. (A) Immunostaining of coronal sections showing an induction of Flag-tagged WNT5A (FLAG-WNT5A) overexpression in 4V ChP, but not in the adjacent regions (arrowheads), in Wnt5aOE embryos compared with Wnt5aWT littermates. AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (B) Immunostaining of coronal sections showing induction of FLAG-WNT5A overexpression in LV ChP in Wnt5aOE embryos compared with Wnt5aWT littermates. Induction of ectopic FLAG-WNT5A protein is also observed in the adjacent cortical hem (arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (C) Higher magnification view showing signal for the ectopic FLAG-WNT5A, which is mostly restricted to the cytoplasm of cells producing FLAG-WNT5A in 4V ChP epithelium (arrowheads); no FLAG-WNT5A is detected in the basal lamina or stroma compared with endogenous WNT5A (unfilled arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (D) Immunostaining of coronal sections demonstrates altered 4V ChP morphology in Wnt5aOE compared with Wnt5aWT at E14.5. Higher magnification images show a detailed view of the 4V ChP (dashed box) in both Wnt5aWT and Wnt5aOE embryos. AQP1 is a marker of ChP epithelium. Scale bars: 300 µm (main panels); 100 µm (higher magnification panels). (E-G) Analysis of E14.5 4V ChP morphology indicates that there is a significant decrease in total area (E), the number of villi (F) and average villus length (G) between Wnt5aWT and Wnt5aOE. Graphs show n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. Biological replicates are indicated in the graph. (H) Higher magnification view shows that ectopic production of FLAG-WNT5A in Wnt5aOE embryos results in detection of the WNT5A signal in LV ChP epithelial cells (filled arrowheads) that normally display very low levels of WNT5A signal as observed in Wnt5aWT LV ChP epithelium (unfilled arrowheads). AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (I) Immunostaining of coronal sections demonstrates altered LV ChP morphology between Wnt5aWT and Wnt5aOE at E14.5. AQP1 is a marker of ChP epithelium. Scale bars: 100 µm. (J,K) Analysis of E14.5 LV ChP morphology indicates that there is a less robust decrease (compared with that observed for 4V ChP) in total area (J) and average length (K) between Wnt5aWT and Wnt5aOE. Graphs show n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-tests. Biological replicates are indicated in the graph.

Wnt5a overexpression had detrimental consequences on the formation of the 4V ChP that were characterized by considerable reduction of its overall size and decreased complexity of its branched, villous structure (Fig. 2D-G). These effects likely stemmed from the overactivation of WNT5A signaling early in differentiating AQP1+ epithelial cells (Fig. S3B). As with knockout experiments, we failed to observe any changes in Ki67 or CASP3 immunoreactivity in the E14.5 4V ChP of Wnt5aOE embryos compared with Wnt5aWT littermate controls (Fig. S3C,D, arrowheads). We also noted increased FLAG-WNT5A expression in LV ChP epithelial cells of Wnt5aOE embryos compared with controls (Fig. 2H, filled and unfilled arrowheads). The effects of Wnt5a overexpression on LV ChP size and length were similar to 4V ChP but slightly weaker (Fig. 2I-K). Taken together, our data demonstrate that 4V ChP morphogenesis relies on tightly regulated WNT5A dosage, likely stipulated at the earliest stages of ChP development.

WNT5A promotes activation of non-canonical WNT signaling in 4V ChP

At E16.5, WNT5A localizes to the distal tips of the 4V ChP epithelium (Fig. 1C) and can be released apically into the CSF for long-range signaling to instruct hindbrain development (Kaiser et al., 2019). In addition, WNT5A localization suggests that it can be released basally (Fig. 1B-D; Fig. S1A,B) for potential local signaling within the ChP. To address the extent of WNT5A signaling activation, we performed biochemical analyses of LV and 4V ChP tissue lysates. WNT5A activation leads to phosphorylation of several downstream signaling pathway components; namely, the WNT5A receptors ROR1 and ROR2 (Grumolato et al., 2010; Ho et al., 2012; Yamamoto et al., 2007) and a key signaling mediator, dishevelled 2 (DVL2) (Bryja et al., 2007). This activation can be detected by western blotting as a phosphorylation-dependent mobility shift. Using this approach, we discovered considerably stronger activation of WNT signaling in the embryonic 4V ChP than in LV ChP during late embryogenesis (Fig. 3A; Fig. S4A). Wnt5a deficiency in HEK293 cells mimicked the differences between 4V and LV ChP, suggesting that activation of these readouts depended on WNT5A (Fig. S4B). Indeed, Wnt5a ablation in both Wnt5aKO and Wnt5acKO ChPs resulted in the loss of induction of non-canonical WNT pathway components, thus demonstrating that their activation was mediated exclusively by 4V ChP epithelium-derived WNT5A (Fig. 3B-D; Fig. S4C). Conversely, we observed that LV ChP, which is normally devoid of endogenous WNT5A signaling (Kaiser et al., 2019), exhibited relatively higher activation of various markers of canonical WNT signaling, including upregulation of the downstream target genes Axin2 (Jho et al., 2002) and Lef1 (Hovanes et al., 2001) or phosphorylation of LRP6 (Tamai et al., 2000) compared with 4V ChP (Fig. 3E-G; Fig. S4D-F). We attribute these effects to the capacity of WNT5A to suppress the canonical WNT pathway during development (Topol et al., 2003). In support of this model, ablation of Wnt5a using both Wnt5aKO and Wnt5acKO mouse models resulted in a partial shift in the balance of WNT signaling from non-canonical to canonical signaling (Fig. 3H-J; Fig. S4G). The progressive decrease of the canonical WNT signaling target genes Axin2 and Lef1 expression along the proximal-distal (P-D) axis was correlated with gradually increasing expression levels of Wnt5a within 4V ChP epithelium (Fig. S4H, filled and unfilled arrowheads). The regionally restricted activation of non-canonical signaling supports a role of WNT5A-mediated signaling that is specific for embryonic development of 4V ChP.

Fig. 3.

WNT5A activates non-canonical WNT signaling in 4V ChP. (A) Western blot analysis of LV ChP and 4V ChP at E14.5 and E17.5 shows induction of the non-canonical WNT signaling pathway specific for 4V ChP compared with the LV ChP. Phosphorylation-dependent shift in the readouts of WNT5A-triggered signaling (ROR1, ROR2 and DVL2) is indicated by the arrowheads. β-actin serves as a loading control. (B) Western blot analysis of E14.5 LV ChP and 4V ChP isolated from Wnt5aWT and Wnt5aKO embryos indicates loss of non-canonical WNT signaling component (ROR1, ROR2 and DVL2) activation upon ablation of WNT5A. Phosphorylation-dependent shift in the readouts of WNT5A-triggered signaling is indicated by the arrowheads. β-actin serves as a loading control. (C,D) Western blot analysis of Wnt5aWT and either E11.5-induced (C) or E12.5-induced (D) Wnt5acKO embryos highlights the absence of non-canonical WNT pathway activation upon loss of WNT5A-mediated signaling, specifically in 4V ChP epithelium. β-actin serves as a loading control. (E) Western blot analysis of LV ChP and 4V ChP at E14.5 and E17.5 shows LV ChP-specific activation of the canonical WNT signaling pathway as demonstrated by the increase of phosphorylated LRP6 (pLRP6) protein level. β-actin serves as a loading control. (F) qPCR of the WNT canonical target gene Axin2 expression in LV ChP and 4V ChP at E14.5. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (G) In situ hybridization analysis of coronal sections confirming increased Axin2 expression in E14.5 LV ChP epithelium compared with 4V ChP epithelium. Scale bars: 10 µm. (H) Western blot analysis of E14.5 LV ChP and 4V ChP isolated from Wnt5aWT and Wnt5aKO embryos highlights activation of canonical WNT signaling in Wnt5aKO 4V ChP as measured by the increase in pLRP6 protein level. β-actin serves as a loading control. (I,J) Western blot analysis of Wnt5aWT and either E11.5-induced (I) or E12.5-induced (J) Wnt5acKO embryos shows minor activation of canonical WNT signaling upon 4V ChP epithelium-specific ablation of WNT5A as indicated by the increase of overall pLRP6 protein levels.

Fig. 3.

WNT5A activates non-canonical WNT signaling in 4V ChP. (A) Western blot analysis of LV ChP and 4V ChP at E14.5 and E17.5 shows induction of the non-canonical WNT signaling pathway specific for 4V ChP compared with the LV ChP. Phosphorylation-dependent shift in the readouts of WNT5A-triggered signaling (ROR1, ROR2 and DVL2) is indicated by the arrowheads. β-actin serves as a loading control. (B) Western blot analysis of E14.5 LV ChP and 4V ChP isolated from Wnt5aWT and Wnt5aKO embryos indicates loss of non-canonical WNT signaling component (ROR1, ROR2 and DVL2) activation upon ablation of WNT5A. Phosphorylation-dependent shift in the readouts of WNT5A-triggered signaling is indicated by the arrowheads. β-actin serves as a loading control. (C,D) Western blot analysis of Wnt5aWT and either E11.5-induced (C) or E12.5-induced (D) Wnt5acKO embryos highlights the absence of non-canonical WNT pathway activation upon loss of WNT5A-mediated signaling, specifically in 4V ChP epithelium. β-actin serves as a loading control. (E) Western blot analysis of LV ChP and 4V ChP at E14.5 and E17.5 shows LV ChP-specific activation of the canonical WNT signaling pathway as demonstrated by the increase of phosphorylated LRP6 (pLRP6) protein level. β-actin serves as a loading control. (F) qPCR of the WNT canonical target gene Axin2 expression in LV ChP and 4V ChP at E14.5. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (G) In situ hybridization analysis of coronal sections confirming increased Axin2 expression in E14.5 LV ChP epithelium compared with 4V ChP epithelium. Scale bars: 10 µm. (H) Western blot analysis of E14.5 LV ChP and 4V ChP isolated from Wnt5aWT and Wnt5aKO embryos highlights activation of canonical WNT signaling in Wnt5aKO 4V ChP as measured by the increase in pLRP6 protein level. β-actin serves as a loading control. (I,J) Western blot analysis of Wnt5aWT and either E11.5-induced (I) or E12.5-induced (J) Wnt5acKO embryos shows minor activation of canonical WNT signaling upon 4V ChP epithelium-specific ablation of WNT5A as indicated by the increase of overall pLRP6 protein levels.

Embryonic ChP epithelium expresses WNT/PCP components

We analyzed in greater detail the spatial distribution of WNT pathway signaling components in the ChP. Expression of genes encoding non-canonical WNT pathway components, including Dvl2 and both Ror1 and Ror2, was mostly restricted to the epithelial cell layer with only low expression in the stromal compartment of the LV and 4V ChP (Fig. 4A,B; Fig. S5A-B′). Given the central role of WNT5A in the WNT/PCP pathway, we also inspected the expression pattern of Vangl2, a downstream mediator of WNT5A-dependent signaling in the WNT/PCP pathway (Gao et al., 2011). Vangl2 expression exhibited a distinct spatial pattern with higher levels detected in the embryonic LV ChP than the 4V ChP (Fig. 4C). In addition, Vangl2 expression was enriched in ChP epithelial cells compared with the stroma, in agreement with our observations regarding expression of genes encoding other non-canonical WNT components (Fig. 4D; Fig. S5B″). VANGL2 antibody was validated in Vangl2−/− knockout animals (Vangl2KO) (Fig. S5C) and a Vangl2 knockout cell line (Mentink et al., 2018). With this antibody, we observed that VANGL2 expression was enriched in epithelial cells in both mouse and human embryonic LV ChP (Fig. 4E-G; Fig. S5D). In contrast to a recent report (Langford et al., 2020), we found that VANGL2 exhibited a strong basolateral distribution typical for its role in mediating cell-to-cell signaling within the developing epithelium (Sittaramane et al., 2013). We also showed overlap of VANGL2 with the dedicated VANGL2 binding partner Scrib (SCRIBBLE; Kallay et al., 2006) in the ChP epithelium during mouse and human embryonic development (Fig. S5E,F; arrowheads). Taken together, our data support a model in which VANGL2 is involved in the control of the WNT/PCP signaling in embryonic ChP.

Fig. 4.

WNT/PCP components are expressed in embryonic ChP epithelium. (A,B) In situ hybridization analysis of coronal sections showing gene expression of the non-canonical WNT pathway components Ror1 (A) and Ror2 (B) that is restricted to both E14.5 LV and 4V ChP epithelium. Wnt5a expression highlights ChP epithelium. Scale bars: 5 µm. (C) qPCR of Vangl2 expression in LV ChP and 4V ChP at E14.5. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (D) In situ hybridization analysis of coronal sections demonstrating a higher expression level of Vangl2 at E14.5 in LV ChP compared with 4V ChP epithelium. Wnt5a expression highlights ChP epithelium. Scale bars: 20 µm. (E) Western blot analysis of VANGL2 protein levels in LV ChP and 4V ChP, isolated at E14.5, showing higher VANGL2 protein abundance in the LV ChP compared with the 4V ChP at E14.5. β-actin serves as a loading control. (F) Immunostaining of sagittal sections of VANGL2 protein levels between E15.5 LV ChP and 4V ChP epithelium with increased signal observed in LV ChP epithelium, especially in its most distal portion (inset). VANGL2 signal segregates to the apical portion of the basolateral compartment of the epithelial cells (insets). Claudin-1 (CLA-1) is a marker of ChP epithelium. Insets show magnifications of the boxed areas. Scale bars: 200 µm (main panels); 20 µm (insets). (G) Immunostaining of sagittal sections demonstrates differential VANGL2 protein levels between 9 weeks gestation human LV ChP and 4V ChP epithelium with increased signal in LV ChP epithelium, especially in its most distal portion as shown in the magnified view (inset). AQP-1 is a marker of ChP epithelium. Insets show magnifications of the boxed areas. Scale bars: 200 µm (main panels); 50 µm (insets).

Fig. 4.

WNT/PCP components are expressed in embryonic ChP epithelium. (A,B) In situ hybridization analysis of coronal sections showing gene expression of the non-canonical WNT pathway components Ror1 (A) and Ror2 (B) that is restricted to both E14.5 LV and 4V ChP epithelium. Wnt5a expression highlights ChP epithelium. Scale bars: 5 µm. (C) qPCR of Vangl2 expression in LV ChP and 4V ChP at E14.5. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (D) In situ hybridization analysis of coronal sections demonstrating a higher expression level of Vangl2 at E14.5 in LV ChP compared with 4V ChP epithelium. Wnt5a expression highlights ChP epithelium. Scale bars: 20 µm. (E) Western blot analysis of VANGL2 protein levels in LV ChP and 4V ChP, isolated at E14.5, showing higher VANGL2 protein abundance in the LV ChP compared with the 4V ChP at E14.5. β-actin serves as a loading control. (F) Immunostaining of sagittal sections of VANGL2 protein levels between E15.5 LV ChP and 4V ChP epithelium with increased signal observed in LV ChP epithelium, especially in its most distal portion (inset). VANGL2 signal segregates to the apical portion of the basolateral compartment of the epithelial cells (insets). Claudin-1 (CLA-1) is a marker of ChP epithelium. Insets show magnifications of the boxed areas. Scale bars: 200 µm (main panels); 20 µm (insets). (G) Immunostaining of sagittal sections demonstrates differential VANGL2 protein levels between 9 weeks gestation human LV ChP and 4V ChP epithelium with increased signal in LV ChP epithelium, especially in its most distal portion as shown in the magnified view (inset). AQP-1 is a marker of ChP epithelium. Insets show magnifications of the boxed areas. Scale bars: 200 µm (main panels); 50 µm (insets).

MEIS1 regulates expression of Wnt5a in 4V ChP epithelium

We next sought to identify the mechanisms that control the expression of Wnt5a in the 4V ChP epithelium. The MEIS family of transcriptional co-activators has been previously linked to the patterning of vertebrate limb or hindbrain, which are also developmentally regulated by WNT5A-mediated signaling (Dibner et al., 2001; Kaiser et al., 2019; Mercader et al., 2009; Yamaguchi et al., 1999). Moreover, several studies have established a direct interaction between MEIS factors and the WNT signaling pathway in the nearby regions of developing 4V ChP, such as hindbrain (Elkouby et al., 2012; Stephens et al., 2010). Importantly, MEIS1 is implicated in the regulation of Wnt5a expression in branchial arches (Amin et al., 2015) and is upregulated in the 4V ChP compared with LV ChP during development (Lun et al., 2015). We thus decided to investigate the possible role of MEIS1 in the regulation of Wnt5a expression in the 4V ChP.

We confirmed Meis1 expression in 4V ChP during late embryogenesis (Fig. 5A) with Meis1 transcripts being restricted to ChP epithelial cells marked by Ttr, a signature gene of ChP epithelial cells (Fig. 5B) (Kato et al., 1986). We confirmed the presence of MEIS1 protein in 4V ChP from E14.5 to E17.5 (Fig. 5C,D) using validated antibodies against MEIS1 (Fig. S6A,B). We identified the potential MEIS1-binding site(s) upstream of Wnt5a by analyzing chromatin immunoprecipitation followed by sequencing (ChIP-seq) on E18.5 4V ChP. MEIS1 binding was enriched at the genomic region containing the proximal Wnt5a promoter (Fig. 5E-G). We confirmed MEIS1 binding to the Wnt5a promoter by ChIP-qPCR (Fig. 5H). Consistent with these data, the spatial distribution of Meis1 expression partially overlapped with Wnt5a expression in 4V ChP epithelial cells (Fig. 5I,J). Further supporting these findings, Meis1 was co-expressed with Wnt5a from very early stages of embryonic 4V ChP development (Fig. S6C-E). At the protein level, MEIS1-positive cells also displayed strong immunoreactivity for WNT5A in both embryonic mouse and human 4V ChP epithelium (Fig. 5K,L), particularly at the distal tips of ChP villi (Fig. 5M,N, filled and unfilled arrowheads). At later embryonic and early postnatal stages, the expression of both MEIS1 and WNT5A progressively decreased in the 4V ChP (Fig. S6F-H).

Fig. 5.

Meis1 expression pattern overlaps with Wnt5a expression in embryonic 4V ChP. (A) qPCR of Meis1 expression in LV ChP and 4V ChP at E14.5. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (B) In situ hybridization analysis of coronal sections showing that Meis1 expression is restricted to the 4V ChP epithelium and absent from the LV ChP epithelium at E14.5. Ttr is a marker of ChP epithelium. Arrowhead indicates cortical hem region. Scale bars: 10 µm. (C) Immunostaining of MEIS1 in the LV ChP and 4V ChP at E14.5 and E17.5. Comparison with the AQP1, a marker of ChP epithelium, shows absence of MEIS1 in the LV ChP, whereas MEIS1 can be detected in the 4V ChP epithelial layer. Scale bars: 10 µm. (D) Western blot analysis of MEIS1 protein in tissue lysates of LV ChP and 4V ChP at E14.5 and E17.5. β-actin serves as a loading control. (E) Line plot showing the distribution of MEIS1 ChIP and Input signal over genebodies. The x-axis is the relative position of genebody. Each gene length from the transcription start site (TSS) to the transcription end site (TES) was scaled to 0-100. The y-axis is the normalized tag density, which is the average MEIS1 ChIP and Input signals over all scaled Refseq genes. (F) Line plot illustrating the distribution of MEIS1 ChIP and Input signals around promoters. The x-axis is the position relative to the TSS from −5 kb upstream to +5 kb downstream. The y-axis is the normalized tag density, which is the average MEIS1 ChIP and Input signals in all Refseq genes. The Refseq gene annotation was obtained from the UCSC website. (G) A screenshot from the UCSC genome browser showing MEIS1 signal enriched at the Wnt5a promoter. (H) The enrichment of MEIS1 binding of Wnt5a promoter (prom1) in E18.5 4V ChP with Cd19k36 enhancer used as control. Results are shown as relative fold enrichment over input in three independent experiments. Graph shows n=3 biologically independent samples; error bars represent s.e.m.; P-value from two-tailed unpaired Student's t-test. (I) Sagittal sections showing similar Meis1 and Wnt5a expression pattern in 4V ChP epithelium at E13.5. Image credit: Allen Institute. Scale bars: 100 µm. (J) In situ hybridization analysis of coronal section showing an overlap of expression of Meis1 and Wnt5a within 4V ChP epithelium at E14.5. Scale bar: 20 µm. (K) Immunostaining of MEIS1 and WNT5A in 4V ChP epithelium at E14.5 and E17.5. AQP1 is a marker of ChP epithelium. Scale bar: 10 µm. (L) Immunostaining showing that MEIS1 detection is restricted to 4V ChP epithelium compared with LV ChP in human embryos at 9 weeks gestation. AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (M) In situ hybridization analysis of E16.5 coronal sections showing that expression of Meis1 and Wnt5a within the 4V ChP epithelium is restricted mostly to the tips of 4V ChP epithelium villi (filled arrowheads) but mostly missing from proximally located regions of the villi (unfilled arrowhead). Foxj1 is a marker of ChP epithelium. Scale bar: 50 µm. (N) Immunostaining showing the correlation of WNT5A signal intensity with MEIS1 signal within 4V ChP epithelium at E14.5 (filled arrowheads showing the distal portion of 4V ChP and unfilled arrowheads indicating the more proximal part of ChP). Scale bar: 50 µm.

Fig. 5.

Meis1 expression pattern overlaps with Wnt5a expression in embryonic 4V ChP. (A) qPCR of Meis1 expression in LV ChP and 4V ChP at E14.5. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-value from two-tailed paired Student's t-test. (B) In situ hybridization analysis of coronal sections showing that Meis1 expression is restricted to the 4V ChP epithelium and absent from the LV ChP epithelium at E14.5. Ttr is a marker of ChP epithelium. Arrowhead indicates cortical hem region. Scale bars: 10 µm. (C) Immunostaining of MEIS1 in the LV ChP and 4V ChP at E14.5 and E17.5. Comparison with the AQP1, a marker of ChP epithelium, shows absence of MEIS1 in the LV ChP, whereas MEIS1 can be detected in the 4V ChP epithelial layer. Scale bars: 10 µm. (D) Western blot analysis of MEIS1 protein in tissue lysates of LV ChP and 4V ChP at E14.5 and E17.5. β-actin serves as a loading control. (E) Line plot showing the distribution of MEIS1 ChIP and Input signal over genebodies. The x-axis is the relative position of genebody. Each gene length from the transcription start site (TSS) to the transcription end site (TES) was scaled to 0-100. The y-axis is the normalized tag density, which is the average MEIS1 ChIP and Input signals over all scaled Refseq genes. (F) Line plot illustrating the distribution of MEIS1 ChIP and Input signals around promoters. The x-axis is the position relative to the TSS from −5 kb upstream to +5 kb downstream. The y-axis is the normalized tag density, which is the average MEIS1 ChIP and Input signals in all Refseq genes. The Refseq gene annotation was obtained from the UCSC website. (G) A screenshot from the UCSC genome browser showing MEIS1 signal enriched at the Wnt5a promoter. (H) The enrichment of MEIS1 binding of Wnt5a promoter (prom1) in E18.5 4V ChP with Cd19k36 enhancer used as control. Results are shown as relative fold enrichment over input in three independent experiments. Graph shows n=3 biologically independent samples; error bars represent s.e.m.; P-value from two-tailed unpaired Student's t-test. (I) Sagittal sections showing similar Meis1 and Wnt5a expression pattern in 4V ChP epithelium at E13.5. Image credit: Allen Institute. Scale bars: 100 µm. (J) In situ hybridization analysis of coronal section showing an overlap of expression of Meis1 and Wnt5a within 4V ChP epithelium at E14.5. Scale bar: 20 µm. (K) Immunostaining of MEIS1 and WNT5A in 4V ChP epithelium at E14.5 and E17.5. AQP1 is a marker of ChP epithelium. Scale bar: 10 µm. (L) Immunostaining showing that MEIS1 detection is restricted to 4V ChP epithelium compared with LV ChP in human embryos at 9 weeks gestation. AQP1 is a marker of ChP epithelium. Scale bars: 10 µm. (M) In situ hybridization analysis of E16.5 coronal sections showing that expression of Meis1 and Wnt5a within the 4V ChP epithelium is restricted mostly to the tips of 4V ChP epithelium villi (filled arrowheads) but mostly missing from proximally located regions of the villi (unfilled arrowhead). Foxj1 is a marker of ChP epithelium. Scale bar: 50 µm. (N) Immunostaining showing the correlation of WNT5A signal intensity with MEIS1 signal within 4V ChP epithelium at E14.5 (filled arrowheads showing the distal portion of 4V ChP and unfilled arrowheads indicating the more proximal part of ChP). Scale bar: 50 µm.

We adopted an adeno-associated viral (AAV) approach using a serotype with tropism for ChP epithelial cells (Cui et al., 2020; Haddad et al., 2013; Xu et al., 2021) to test whether supplemental Meis1 expression would be sufficient to influence 4V ChP morphology (Fig. S7A,B). We validated efficient intracerebroventricular delivery and protein induction by robust induction of GFP signal in the epithelium of both LV and 4V ChP (Fig. S7C,D). We then successfully applied this approach to overexpress MEIS1 at E10.5 in developing 4V ChP (Fig. S7E,F) to test the effect of MEIS1 enrichment on WNT5A signaling during the crucial developmental time window. qPCR analysis revealed marked upregulation of Wnt5a expression in E16.5 4V ChP following Meis1 overexpression induced at E10.5, which we further confirmed by in situ hybridization (Fig. 6A,B; filled and unfilled arrowheads). Meis1 overexpression also resulted in higher levels of WNT5A protein in the 4V ChP epithelium at E16.5 (Fig. 6C,D) and in 4V ChP tissue lysate (Fig. 6E). Nevertheless, we did not observe any change in the overall morphology of the tissue (Fig. 6F). These results suggest that MEIS1-driven WNT5A upregulation was not sufficient to induce changes in gross morphology, possibly because it was unable to induce activation of the non-canonical WNT pathway monitored by phosphorylation of ROR1 or DVL2 (Fig. S7G). MEIS1 overexpression in embryonic LV ChP epithelium did result in a small increase of Wnt5a expression (Fig. S7H-J) that, however, did not translate into a detectable increase in WNT5A protein level or activation of the non-canonical WNT pathway (Fig. S7K,L).

Fig. 6.

Meis1 regulates expression in 4V ChP epithelium. (A) qPCR analysis of Wnt5a expression in E10.5 AAV transduced E16.5 4V ChP. Graph shows n=3-6 biologically independent samples; error bars represent s.d.; P-value from two-tailed unpaired Student's t-test. (B) In situ hybridization analysis of coronal sections showing Wnt5a expression in E16.5 4V ChP cells overexpressing Meis1 (filled arrowheads) and low Wnt5a expression in cells transfected with empty vector (unfilled arrowheads). Endogenous GFP staining shows successfully transfected cells. Scale bars: 10 µm. (C) Immunostaining of MEIS1 and WNT5A signal in either empty control or Meis1-overexpressing E16.5 4V ChP epithelium shows upregulation of MEIS1 levels in 4V ChP epithelium that is associated with increased WNT5A signal. AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (D) Quantification of mean WNT5A signal intensity difference between E16.5 AAV-GFP and AAV-Meis1-GFP transduced 4V ChP epithelium. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-value from two-tailed unpaired Student's t-test. (E) Western blot analysis of MEIS1 and WNT5A protein levels in tissue lysates from AAV-GFP or AAV-Meis1-GFP transduced E16.5 4V ChP. Overexpression of Meis1 results in an increase of both MEIS1 and WNT5A protein levels in 4V ChP compared with control vector. Vinculin serves as a loading control. (F) Quantification of the difference in total area between E16.5 AAV-GFP and AAV-Meis1-GFP transduced 4V ChP. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-values from two-tailed unpaired Student's t-test. (G) Immunostaining of an embryonic sagittal section demonstrating a decrease of WNT5A signal in the E14 4V ChP epithelium of Meis1KO embryos compared with Meis1WT embryonic 4V ChP (insets; arrowheads). Inset images correspond to the highlighted boxed areas. Claudin-1 (CLA-1) and AQP1 are markers of ChP epithelium. Scale bars: 50 µm (main panels); 10 µm (insets). (H) Quantification of mean WNT5A signal intensity difference between E14 Meis1WT and Meis1cKO 4V ChP epithelium. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (I) Quantification of the difference in total area between E14 Meis1WT and Meis1cKO 4V ChP epithelium. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (J) In situ hybridization analysis of coronal sections showing Meis2 expression being restricted to 4V ChP epithelium and absent from LV ChP epithelium at E14.5, as observed for Meis1. Scale bars: 10 µm. (K) Immunostaining of embryonic sagittal sections demonstrating the difference in WNT5A levels (insets) and morphology between E14 Meis1WT, Meis1cKO and Meis1/2dKO 4V ChP. AQP1 is a marker of ChP epithelium. Scale bars: 100 µm (main panels); 20 µm (insets). (L) Quantification of mean WNT5A signal intensity difference between E14 Meis1WT and Meis1/2dKO 4V ChP. Graph shows n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (M) Quantification of the difference in total area between E14 Meis1WT and Meis1/2dKO 4V ChP. Graph shows n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test.

Fig. 6.

Meis1 regulates expression in 4V ChP epithelium. (A) qPCR analysis of Wnt5a expression in E10.5 AAV transduced E16.5 4V ChP. Graph shows n=3-6 biologically independent samples; error bars represent s.d.; P-value from two-tailed unpaired Student's t-test. (B) In situ hybridization analysis of coronal sections showing Wnt5a expression in E16.5 4V ChP cells overexpressing Meis1 (filled arrowheads) and low Wnt5a expression in cells transfected with empty vector (unfilled arrowheads). Endogenous GFP staining shows successfully transfected cells. Scale bars: 10 µm. (C) Immunostaining of MEIS1 and WNT5A signal in either empty control or Meis1-overexpressing E16.5 4V ChP epithelium shows upregulation of MEIS1 levels in 4V ChP epithelium that is associated with increased WNT5A signal. AQP1 is a marker of ChP epithelium. Scale bars: 50 µm. (D) Quantification of mean WNT5A signal intensity difference between E16.5 AAV-GFP and AAV-Meis1-GFP transduced 4V ChP epithelium. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-value from two-tailed unpaired Student's t-test. (E) Western blot analysis of MEIS1 and WNT5A protein levels in tissue lysates from AAV-GFP or AAV-Meis1-GFP transduced E16.5 4V ChP. Overexpression of Meis1 results in an increase of both MEIS1 and WNT5A protein levels in 4V ChP compared with control vector. Vinculin serves as a loading control. (F) Quantification of the difference in total area between E16.5 AAV-GFP and AAV-Meis1-GFP transduced 4V ChP. Graph shows n=4 biologically independent samples; error bars represent s.d.; P-values from two-tailed unpaired Student's t-test. (G) Immunostaining of an embryonic sagittal section demonstrating a decrease of WNT5A signal in the E14 4V ChP epithelium of Meis1KO embryos compared with Meis1WT embryonic 4V ChP (insets; arrowheads). Inset images correspond to the highlighted boxed areas. Claudin-1 (CLA-1) and AQP1 are markers of ChP epithelium. Scale bars: 50 µm (main panels); 10 µm (insets). (H) Quantification of mean WNT5A signal intensity difference between E14 Meis1WT and Meis1cKO 4V ChP epithelium. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (I) Quantification of the difference in total area between E14 Meis1WT and Meis1cKO 4V ChP epithelium. Graph shows n=7 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (J) In situ hybridization analysis of coronal sections showing Meis2 expression being restricted to 4V ChP epithelium and absent from LV ChP epithelium at E14.5, as observed for Meis1. Scale bars: 10 µm. (K) Immunostaining of embryonic sagittal sections demonstrating the difference in WNT5A levels (insets) and morphology between E14 Meis1WT, Meis1cKO and Meis1/2dKO 4V ChP. AQP1 is a marker of ChP epithelium. Scale bars: 100 µm (main panels); 20 µm (insets). (L) Quantification of mean WNT5A signal intensity difference between E14 Meis1WT and Meis1/2dKO 4V ChP. Graph shows n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test. (M) Quantification of the difference in total area between E14 Meis1WT and Meis1/2dKO 4V ChP. Graph shows n=3 biologically independent samples; error bars represent s.d.; P-values from two-tailed paired Student's t-test.

To extend the functional analysis of the link between Meis1 and Wnt5a expression, we used the Wnt1-Cre2 mouse line (Lewis et al., 2013) to conditionally delete Meis1 (Meis1cKO) in the 4V ChP epithelium (Fig. S8A,B). We confirmed efficient reduction of Meis1 expression in 4V ChP epithelial cells (Fig. S6B) but not in adjacent regions where Wnt1 was not expressed (Fig. S8A,C, filled and unfilled arrowheads). Importantly, immunofluorescence analysis of Meis1cKO embryos revealed a prominent reduction of WNT5A production in 4V ChP epithelial cells and reduced size of the 4V ChP compared with Meis1WT littermates (Fig. 6G-I). Meis2, another member of the MEIS transcription factor family, is also expressed in the 4V ChP epithelium (Lun et al., 2015). To test for possible redundancy between Meis1 and Meis2, we first confirmed that Meis2 expression was restricted to the developing 4V ChP epithelium similar to Meis1 (Fig. 6J). Next, we conditionally deleted both Meis1 and Meis2 (Meis1/2dKO) in the 4V ChP. We were unable to obtain more than three Meis1/2dKO embryos, but even this limited sample set indicated that Meis2 deficiency further decreased WNT5A expression in Meis1/2dKO embryos compared with Meis1 knockout alone (Fig. 6K,L). Notably, analysis of Meis1/2dKO embryos was also suggestive of further impairment in the morphogenesis and decreased overall size of the 4V ChP compared with wild-type and Meis1KO littermates (Fig. 6M). Collectively, these findings demonstrate a direct regulation of Wnt5a expression by MEIS1 in the developing 4V ChP and suggest redundancy of MEIS1 and MEIS2 in this process.

Despite large overlap in their cellular architecture and secretory capacity, ChPs adopt dramatically different morphologies during development. Here, we report a novel role for epithelium-derived WNT5A in the regulation of tissue branching morphology within the 4V ChP. WNT5A-mediated signaling, established as an important regulator of morphogenesis in various tissues (Yamaguchi et al., 1999), has been recently recognized to play an important role during ChP embryogenesis (Kaiser et al., 2019; Langford et al., 2020). Using temporally controlled conditional deletion of Wnt5a directed to the ChP epithelium, we identified two roles of WNT5A, separated in time and space. Our data agree and expand upon recently published findings showing a crucial role of WNT5A in the development of embryonic ChPs (Langford et al., 2020).

By integrating information about WNT5A expression domains and functional data from various Wnt5a mouse mutants, we propose a model in which WNT5A has two distinct roles in mouse ChP formation (Fig. 7). Early on, Wnt5a is expressed in the precursor domains that are directly adjacent to and feed the growth of 4V, 3V and LV ChP epithelium (Langford et al., 2020; Dani et al., 2021). As development proceeds, Wnt5a is expressed in the maturing ChP epithelium in a spatially restricted fashion enriched in the 4V ChP (Kaiser et al., 2019). Systemic Wnt5a deletion impairs development of all ChPs (Langford et al., 2020). In contrast, conditional deletion of Wnt5a in time and space within Foxj1-expressing epithelial cells (Wnt5acKO) restricts the growth phenotype to the 4V and not LV ChP. However, we are unable to comment on 3V ChP, which was not examined in this present study. The effect is age-dependent: strong when recombination is induced at E11.5 and non-detectable when induced at later stages. Our data show that the tamoxifen injection at E11.5 results in the loss of WNT5A protein at E12.5. Thus, WNT5A appears to be essential during this developmental period when branching of the 4V ChP begins. A similar temporal requirement for Otx2 expression has also been described (Johansson et al., 2013).

Fig. 7.

WNT5A signaling in the embryonic ChP epithelium. Schematic summarizing the key aspects of WNT5A signaling in embryonic LV and 4V ChP epithelium. Wnt5a is expressed in a region adjacent to both developing ChPs (CH for LV ChP and LRL for 4V ChP) with Wnt5a expression being restricted to 4V ChP epithelium and not LV ChP epithelium. Meis1 expression is also spatially restricted to the 4V ChP epithelium and not the LV ChP. Foxj1 is a marker gene for embryonic ChP epithelium. Epithelial WNT5A, production of which is under direct regulatory control of the transcription co-factor MEIS1, stimulates in an autocrine manner the transmembrane receptors ROR1 and ROR2. Their activation leads to the activation of DVL2 and DVL3. Ultimately, WNT5A signaling contributes to the suppression of WNT/canonical signaling, which is re-activated upon loss of WNT5A. Key components of the WNT/PCP signaling pathway, namely VANGL2 and its interacting partner SCRIBBLE also participate in the proper establishment of the ChP epithelium.

Fig. 7.

WNT5A signaling in the embryonic ChP epithelium. Schematic summarizing the key aspects of WNT5A signaling in embryonic LV and 4V ChP epithelium. Wnt5a is expressed in a region adjacent to both developing ChPs (CH for LV ChP and LRL for 4V ChP) with Wnt5a expression being restricted to 4V ChP epithelium and not LV ChP epithelium. Meis1 expression is also spatially restricted to the 4V ChP epithelium and not the LV ChP. Foxj1 is a marker gene for embryonic ChP epithelium. Epithelial WNT5A, production of which is under direct regulatory control of the transcription co-factor MEIS1, stimulates in an autocrine manner the transmembrane receptors ROR1 and ROR2. Their activation leads to the activation of DVL2 and DVL3. Ultimately, WNT5A signaling contributes to the suppression of WNT/canonical signaling, which is re-activated upon loss of WNT5A. Key components of the WNT/PCP signaling pathway, namely VANGL2 and its interacting partner SCRIBBLE also participate in the proper establishment of the ChP epithelium.

We identify MEIS1 as one transcription factor that participates in the control of Wnt5a expression in the 4V ChP epithelium. Our data further suggest that MEIS1 and MEIS2 together regulate Wnt5a expression and 4V ChP development. Regionalized and conserved expression of Wnt5a, Meis1 and Meis2 has previously been reported to occur during both human and mouse ChP embryogenesis (Kaiser et al., 2019; Lun et al., 2015). Nevertheless, despite our identification of this MEIS-WNT5A axis, the full transcriptional factor network that defines 4V ChP epithelium cell fate remains elusive. Potential co-regulation with HOXA2, which is also enriched in the 4V ChP epithelium (Awatramani et al., 2003; Lun et al., 2015), may be possible. HOXA2 binds the Wnt5a promoter (Donaldson et al., 2012), and in the branchial arches, MEIS1 and HOXA2 synergize to positively regulate Wnt5a expression (Amin et al., 2015). Future studies may reveal whether a similar evolutionarily conserved mechanism contributes to the development of the ChP.

WNT5A controls cell polarity (Humphries and Mlodzik, 2018) as well as morphogenesis and tissue outgrowth in many developing tissues, including mammary gland (Kessenbrock et al., 2017), kidney (Pietilä et al., 2016), lung (Li et al., 2005) and prostate gland (Huang et al., 2009), which all exhibit complex branched morphology. The role of Wnt5a in regulating the morphogenesis of distal regions of branched structures appears to also be evolutionarily conserved as it can be observed in various developmental contexts, including lung and prostate (Huang et al., 2009; Li et al., 2005), embryonic digits (Yamaguchi et al., 1999) and budding tentacles in Hydra (Philipp et al., 2009). Basolateral secretion of WNT5A by epithelial cells has also been shown to participate in the process of lumen formation in kidney epithelium (Yamamoto et al., 2015). These findings point to a more universal role of epithelial WNT5A in the regulation of branching and outgrowth of epithelial sheets. Importantly, WNT5A appears to be necessary for the initial stages of ChP development, but it is not essential after E12.5 despite continuing 4V ChP tissue growth and folding that progress with development.

WNT5A is known to contribute to the balance between WNT/β-catenin (canonical) and non-canonical WNT signaling, which is essential for proper tissue morphogenesis and homeostasis (Alexander et al., 2012). Crosstalk between WNT5A and other WNTs may differ for the two expression domains of Wnt5a in the developing ChP (see Fig. 7). As Wnt5a expression domains are located adjacent to all embryonic ChPs (Langford et al., 2020) and overlap with the expression domains of WNT ligands that activate β-catenin pathway, it is possible that WNT5A acts in concert with other WNT genes, for example the typical canonical ligand WNT2b, which is known to be an important patterning factor orchestrating proper development of ChPs in the cortical hem (Grove et al., 1998). Importantly, the Wnt5a domain also overlaps with an R-spondin (R-SPO)-positive progenitor domain (Dani et al., 2021). R-SPO acts as an amplifier of WNT/β-catenin signaling, which suggests that in the progenitor domain WNT5A can synergize or coordinate with WNT/β-catenin signals. In contrast, in the 4V ChP epithelium WNT5A seems to be a signaling factor suppressing canonical WNT signaling, as reported in mammary gland development (Roarty et al., 2009). Higher WNT/β-catenin signaling (as shown by upregulation of target genes Axin2 or Lef1 or phosphorylation of LRP6) was detected in the embryonic LV ChP epithelium with low/undetectable Wnt5a expression or upon deletion of Wnt5a in the 4V ChP. Notably, in comparison with Wnt5a, both Axin2 and Lef1 show a reverse pattern of expression along the P-D axis in the embryonic 4V ChP. This observation suggests that the spatial control of WNT/β-catenin signaling is achieved by increasing levels of WNT5A within the ChP epithelium (Pourreyron et al., 2012; Sato et al., 2010). Canonical and non-canonical WNT pathways have been implicated in regulating different aspects of epithelial morphogenesis, such as apical constriction, which is crucial for inducing branching in epithelial monolayers (Choi and Sokol, 2009; Fumoto et al., 2017). The striking difference observed between mouse LV and 4V ChP morphologies, together with distinct activation of WNT pathway signaling, raises the possibility that tight regulation of canonical versus non-canonical WNT signaling contributes to induction or modulation of epithelial folding, as has been recently proposed for developing Drosophila wing disc epithelium or in mouse embryonic submandibular gland (Gou et al., 2021; Sui and Dahmann, 2020). Although Wnt5a contributions to 4V ChP morphogenesis may be conserved, it is important to note that the process of ChP morphogenesis will likely prove to involve multiple pathways, as human LV ChP, in contrast to rodent LV ChP, exhibits a branched structure.

WNT5A has also been implicated in the regulation of endothelial cell migration and microvasculature assembly (Carvalho et al., 2019; Ramakrishnan et al., 2016). Thus, WNT5A production at the distal tips of 4V ChP epithelium may contribute to the interplay between the ChP epithelium and the stromal compartment, where it may facilitate vascular outgrowth within the villi as has been shown for SHH, another morphogen specifically produced in the developing 4V ChP (Nielsen and Dymecki, 2010). Given the lack of detected changes in proliferation or cell death upon Wnt5a manipulation, these newly revealed biological functions of WNT5A represent avenues for future research to elucidate further the mechanisms underlying disrupted 4V ChP morphogenesis upon loss of WNT5A.

In summary, our data reveal the complex functions of WNT5A in regulating ChP development. From a clinical perspective, rare neoplasms of the ChP have higher incidence in children and display regional specificity (Sun et al., 2014). Thus, elucidating the regionalized signaling pathways underlying ChP development may expose molecular mechanisms that lend one ventricle more or less susceptible to cancer. Because the ChP represents a tissue amenable to genetic manipulation (Chen et al., 2020), and it adopts distinct, ventricle-specific morphologies owing to inherent differences in activation of WNT5A-driven PCP signaling, the ChP provides a tractable model for future research investigating the mechanistic basis of WNT5A signaling in tissue morphogenesis.

Mouse strains

Conditional Meis1fl/fl mice were generated from the embryonic stem cell clone HEPD0632_4_H07 purchased from EUCOMM. The Frt-flanked lacZ/neo cassette was removed by ACTFLPe (strain #005703). LoxP sites flank exon ENSMUSE00000655363 encoding the homeobox region of the Meis1 gene. Genotyping primers CTGCGCTTCCTACATCACTG and CACTTCAGCGTCACTTGGAA produce a 227-bp fragment for the wild-type allele and a 262-bp band for the floxed allele.

Conditional Meis2fl/fl strain with loxP sites around exons 2-6 was described previously (Machon et al., 2015). Genotyping primers GCAAGGGTGCTGAGGTTAAA and TCAGACCCAGGAATTTGAGG produce a 235-bp fragment in the wild-type allele and a 324-bp fragment in the floxed allele.

The Wnt1-Cre2 mouse strain was purchased from The Jackson Laboratory (strain #022137) and it was used for specific deletion of the Meis1fl/fl gene in 4V ChP (referred to as Meis1cKO in this article). Genotyping primers GCATTTCTGGGGATTGCTTA and CCCGGCAAAACAGGTAGTTA amplify a 241-bp Cre fragment.

The reporter line mT/mG was purchased from The Jackson Laboratory (strain #007676).

The Wnt5a−/− (Wnt5aKO) mouse strain used in this article corresponds to Wnt5atm1Amc (Yamaguchi et al., 1999). Mouse strain Wnt5atm1.1Krvl/J (referred to as Wnt5aflox/flox in this article) (Ryu et al., 2013) was purchased from Jackson Laboratories; Foxj1tm1.1(cre/ERT2/GFP)Htg (referred to as Foxj1-creERT2 in this article) (Muthusamy et al., 2014) and Gt(ROSA)26Sortm14(CAG−tdTomato)Hze (referred to as tdTomato in this article) (Madisen et al., 2010) were shared with the Karolinska Institutet, Sweden through a collaboration agreement. All mouse strains were housed, bred and treated in Czech Centre for Phenogenomics (Institute of Molecular Genetics, CAS) in accordance with protocols approved by the animal work committee of the Institute of Molecular Genetics, CAS and Central Commission for Animal Welfare of Ministry of Agriculture Czech Republic (PP-90-2015, PP-64-2018). Induction of conditional knockout or the tdTomato reporter was induced by single dose of tamoxifen (Sigma-Aldrich) intraperitoneal injection of pregnant female mice at a concentration of 4.5 mg of tamoxifen dissolved in sterile sunflower oil per 20 g weight of mouse.

Overexpression of Wnt5a (Wnt5aOE) was induced in embryos carrying both an inducible Wnt5a transgene and a Rosa26rtTA driver as described earlier [van Amerongen et al., 2012; strains are available from Jackson Laboratories: FVB/N-Tg(tetO-Wnt5a) 17Rva/J, stock number 022938; B6.Cg-Gt(ROSA)26Sortm1(rtTA*M2)Jae/J, stock number 006965]. Wnt5aOE was induced by administering doxycycline to pregnant mice from E10.5 onwards by dissolving doxycycline in the drinking water (1-2 mg/ml) ad libitum. Ror2; Vangl2 mice were obtained from Dr Yingzi Yang (Gao et al., 2011). Mice were crossed as double heterozygotes (Ror2+/−; Vangl2+/−) due to homozygote lethality. All mice were used according to the rules and regulations of the local ethical committee (Stockholm Norra Djurförsöksetisks Nämd: N273/11, N326/12, N158/15; and the Animal Welfare Committee of the University of Amsterdam).

Reagents used are listed in Table S1.

In utero injection

All AAV injection in utero experiments were performed under protocols approved by the IACUC of Boston Children's Hospital (BCH). Timed-pregnant CD-1 dams were obtained from Charles River Laboratories, were anesthetized with isoflurane inhalation, and laparotomy was performed at E10.5 or E13.5. AAV-GFP or AAV-Meis1 (1 µl containing 1∼5×1012 gc/ml) was injected into a lateral ventricle using a pulled glass pipette (Drummond Scientific Company). Analgesic (meloxicam 5 mg/kg) was provided subcutaneously following surgery. For E10.5 injections, uterine horns were bathed in sterile PBS for improved visualization of ventricles by ultrasound. E16.5 embryos were harvested and drop-fixed in 4% paraformaldehyde (PFA; Millipore) before tissue processing or snap-freezing for qPCR analysis.

The murine Meis1 gene was obtained from the MEIS1A-MIY (Addgene, plasmid #21013) by EcoRI digestion and ligated into an AAV-CMV-IRES-hrGFP (Agilent) vector digested with EcoRI. AAV5-GFP and AAV5-Meis1 were produced by the viral core at Boston Children's Hospital (BCH).

Reagents used are listed in Table S1. Constructs used are listed in Table S2.

ChIP-sequencing assays

ChIP-seq protocol was adapted from Laurent et al. (2015). Briefly, 4V ChP from E18.5 embryos were dissected into cold HBSS. Tissues were crosslinked in 1% formaldehyde at room temperature for 10 min while rotating. Glycine (Sigma-Aldrich) was added to a final concentration of 125 mM and incubated at room temperature for 5 min. Tissues were centrifuged at 4°C at 2000 g and washed twice with 1×PBS with protease inhibitors (Thermo Fisher Scientific). Tissues were snap-frozen in liquid nitrogen and stored at −80°C.

Frozen tissues were resuspended in lysis buffer with protease inhibitors (50 mM Tris pH 8.1, 10 mM EDTA pH 8.0, 1% SDS). Tissues were homogenized using an insulin syringe and then fragmented with an ultrasonic sonicator (Qsonica). After sonication, dilution buffer (0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris-HCl pH 8.0 and 167 mM NaCl) with protease inhibitors were added to give a final SDS concentration of 0.1%.

Chromatin samples were pre-cleared with Protein A beads (Thermo Fisher Scientific) under rotation at 4°C for 1 h prior to incubation overnight with anti-Meis1 or anti-IgG antibody (15 µg of antibody per sample). Protein A beads were added to precipitate antibody complexes and rotated for 1 h at 4°C and then washed with low salt buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.0, 150 mM NaCl), high salt buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.0, 500 mM NaCl), LiCl buffer (0.25 M LiCl, 1% IGEPAL-CA630, 1% deoxycholic acid, 1 mM EDTA, 10 mM Tris pH 8.0) and TE buffer (10 mM Tris-HCl, 1 mM EDTA). Antibody complexes were eluted with 1% SDS in 100 mM NaHCO3, and crosslinks were reversed with NaCl and proteinase K (New England Biolabs). DNA was recovered by phenol-chloroform extraction and ethanol precipitation and quantified with Qubit (Invitrogen).

ChIP-seq libraries were prepared using NEBNext DNA library preparation reagents (E6000) as described in the Illumina Multiplex ChIP-seq DNA sample Prep Kit. Libraries were indexed using a single indexed PCR primer. Libraries were quantified by Qubit (Invitrogen) and sequenced using a HiSeq 2500 (Illumina) to generate 50 bp single-end reads. Reagents used are listed in Table S1.

ChIP-seq analysis

ChIP-seq samples were sequenced in single-end on the Illumina HiSeq2500 platform. Raw reads were mapped to mouse reference genomes (mm9) downloaded from the UCSC website (www.genome.ucsc.edu). Bowtie (v1.0.0) (Langmead et al., 2009) was used for alignment with ‘–m 1’ and other default parameters. The ‘–m 1’ allows reads uniquely aligned to the genome. MACS (v2.0.10) (Zhang et al., 2008) was used to identify the enriched regions with default parameters. These enriched regions were annotated with an R package, ChIPpeakAnno (v3.6.5) (Zhu et al., 2010), in Bioconductor. The promoters were defined as 2 kb from up- to downstream of transcriptional start sites. Reagents used are listed in Table S1.

ChIP-qPCR

ChIP-qPCR was performed on immunoprecipitated DNA after amplification with NEB Next DNA library preparation (New England Biolabs). qPCR was performed with SYBR green (Roche) for detection on a LightCycler 480 system (Roche) according to the manufacturer's instructions. The results were calculated as relative fold enrichment over the input. Reagents used are listed in Table S1. Primers are listed in Table S5.

Choroid plexus epithelial cell primary culture

ChP tissue was collected from E14.5 embryos isolated from sacrificed pregnant CD1 mice and choroid plexus epithelial cells (CPECs) were isolated from LV ChP and 4V ChP. During isolation, extracted tissue was kept at room temperature (RT) in HBSS solution (Sigma-Aldrich). After isolation, extracted tissue was briefly centrifuged (200 g, 10 s at RT). Following aspiration of supernatant, 500 µl of 2 mg/ml solution of Pronase (Sigma-Aldrich) was added to the extracted tissue and incubated for 5 min at 37°C. The solution was then transferred to DMEM/F-12 medium containing 10% fetal bovine serum (FBS; Thermo Fisher Scientific) and centrifuged (300 g, 2 min at RT). Tissue was transferred to complete culture medium consisting of DMEM/F-12 supplemented with 10% FBS, 10 ng/ml EGF (Invitrogen), 20 µM cytosine arabinoside (Sigma-Aldrich) 50 U/ml penicillin and 50 U/ml streptomycin. Cells were mechanically dissociated through a 21-gauge needle for six to eight forced passages, followed by gentle repeated re-suspension with a 200 µl pipette. Finally, cells were seeded onto Transwell-0.4 µm thick clear filter inserts (Sigma-Aldrich). Inserts were pre-coated on their upper side with laminin (Sigma-Aldrich) as described by the manufacturer. To achieve higher purity of epithelial cells, an adhering-off method was applied to reduce fibroblast contamination. After the initial seeding, supernatant containing unadhered cells was transferred to a new laminin-coated well thus removing from culture fibroblasts characterized by a higher adherence affinity.

In order to produce conditioned medium (CM), CPEC primary cultures were maintained in complete culture medium. CM was collected every 48 h up to 10 days after seeding. Supernatant was subjected to sequential centrifugation steps of 200 g for 5 min (to remove viable cells), 1500 g for 10 min (to remove dead cells) and 6000 g for 15 min (to remove cell debris). Reagents used are listed in Table S1.

Fetal tissue section

Ethical approval allowing human fetal tissue acquisition and analysis was provided by the National Research Ethics Service Committee East of England–Cambridge Central, UK (ethics number 96/085).

Cell culture and transfection

HEK293T cells were seeded in complete DMEM medium containing 10% FBS, 2 mM L-glutamine, 50 U/ml penicillin and 50 U/ml streptomycin (Thermo Fisher Scientific) on 10 cm dishes 24 h prior to transfection. The cells were transfected with a total of 5 µg of DNA at ∼40% confluency in DMEM medium only. The transfection reaction mixture was prepared using OptiMEM (Thermo Fisher Scientific) and Lipofectamine 2000 (Thermo Fisher Scientific), at a ratio of 1 µg DNA:2 µl Lipofectamine 2000, followed by incubation with cells for 4-6 h. Afterwards, the transfection medium was exchanged for the complete medium. Reagents used are listed in Table S1.

CRISPR/Cas9 generation of WNT5A-only HEK293 T-REx cells

Plasmid-encoding guide RNA targeting the human WNT5A gene was used. The gRNA sequence was cloned into plasmids pSpCas9(BB)-2A-GFP (PX458) (Addgene plasmid #41815) or pU6-(BbsI) CBh-Cas9-T2A-mCherry (Addgene plasmid #64324). T-REx-293 cells (Invitrogen) were cultured according to the manufacturer's instructions and transfected by Lipofectamine 2000 DNA Transfection Reagent (Thermo Fisher Scientific) with plasmids encoding guide RNAs targeting human WNT5A gene. Next day, transfected cells were single cell sorted and grown as single colonies. Selection of WNT5A knockout (KO) clones was carried out by PCR. Genomic DNA was isolated by DirectPCR Lysis Reagent (Viagen Biotech) and then the fragment of genomic DNA was amplified by PCR using DreamTaq DNA Polymerase (Thermo Fisher Scientific). Then, the PCR product was cut by TaqI (Thermo Fisher Scientific). WNT5A ablation efficiency was assessed by western blot analysis using WNT5A antibody (MAB645, R&D Systems).

Reagents used are listed in Table S1. Constructs used are listed in Table S2. Primers are listed in Table S5.

Western blotting

Samples were subjected to SDS-PAGE, electrotransferred onto a Hybond-P membrane (GE Healthcare), immunodetected using appropriate primary and secondary antibodies and visualized by ECL (Millipore) or Supersignal Femto solution (Thermo Fisher). Signal intensities were calculated using ImageJ. Briefly, the area of the peak intensity for the protein of interest was divided by the corresponding values of peak intensity obtained for the control protein.

Reagents used are listed in Table S1. Antibodies used are listed in Table S3.

MicroCT

Embryos were fixed for 1 week in 4% PFA and stained with Lugol's Iodine solution for 2 weeks or longer. Stock solution (10 g KI and 5 g I2 in 100 ml H2O) was diluted to a 25% working solution in H2O. Stained specimens were removed from contrast agent, rinsed with PBS and embedded in 2.5% low gelling temperature agarose dissolved in water. Scanning was performed using a SkyScan 1272 high-resolution microCT (Bruker, Belgium), with voxel size set up for 3 µm.

Immunofluorescence and EdU staining

For mouse embryo immunofluorescence analysis and in situ hybridization mice were dissected and isolated embryos were transferred into ice-cold PBS, fixed in 4% PFA in PBS for several hours followed by several washes in ice-cold PBS and finally cryoprotected by sequential incubation in 15% and then 30% sucrose solutions. Embryos were next frozen in optimum cutting temperature (OCT) compound (Sakura FineTek) on dry ice. Serial 14-µm-thick coronal sections were used for immunofluorescence analysis. Human fetal tissue for cryosectioning was immersion-fixed overnight in 4% PFA at 4°C, then cryoprotected in sucrose before embedding in OCT compound and then 14-µm-thick sections were cut using a Leica CM1850 cryostat. Human fetal tissue samples were then processed using an identical immunofluorescence protocol as for the mouse samples.

For immunofluorescence analysis, all the sections underwent antigen retrieval by direct boiling for 1 min at 550 W in the microwave followed by 10 min incubation at 85°C in a water bath, using antigen retrieval solution (Dako). Sections were washed in PBT (PBS with 0.5% Tween-20) and blocked in PBTA (PBS, 5% donkey serum, 0.3% Triton X-100, 1% bovine serum albumin). Samples were incubated overnight at 4°C with primary antibodies diluted in PBTA. Following washes in PBT, samples were incubated with corresponding Alexa Fluor secondary antibodies (Invitrogen, Abcam) for 1 h at RT, followed by 5 min incubation at RT with DAPI (Thermo Fisher). Finally, samples were mounted in DAKO mounting solution (Dako).

EdU (Life Technologies) was injected 24 or 72 h before the embryos were harvested at a concentration of 65 mg/g. Cells with incorporated EdU were visualized using a Click-iT EdU Alexa Fluor 555 Imaging Kit (Life Technologies).

For immunofluorescence analysis of ChP primary culture, cells grown on laminin-coated cover slips were first washed several times in ice-cold PBS, followed by fixation for 15 min in ice-cold 4% PFA. Later, cells were washed several times in PBT, blocked with PBTA for 30 min and incubated overnight at 4°C with primary antibodies. Following repeated washing in PBT, cells were incubated for 1 h at RT with appropriate secondary antibodies, DAPI for 5 min and mounted in DAKO mounting medium (Dako).

Reagents used are listed in Table S1. Antibodies used are listed in Table S3.

Immunofluorescence quantification

Quantitative analysis of immunofluorescence signal was carried out in a randomized single-blinded manner. Only the AQP1+ area of embryonic 4V ChP was used for the signal intensity analysis. Embryos from the same litters were used for the comparative analysis and all the samples were processed using the same immunofluorescence protocol (described above; see Table S3) and same settings (for laser wavelength 561: gain 800 V, laser intensity 1.2%; for laser wavelength 640: gain 800 V, laser intensity 0.5%) of the confocal microscope (LSM880, Zeiss). Images were not further post-processed or enhanced. Signal was analyzed from three consecutive sections obtained from each embryonic 4V ChP. Normalization was carried out by averaging WNT5A signal intensity from each individual control AAV-GFP embryonic 4V ChP as indicated in the corresponding graphs (Fig. 6D,H).

For morphological analysis of embryonic LV and 4V ChP, only the AQP1+ ChP area was included in the analysis.

In situ analysis

In situ analysis of the gene expression was performed on 14-µm cryosections of embryos at various stages of embryonic development isolated from CD1 mice. After isolation, embryos were immediately transferred and kept in fresh 4% PFA for 2 h, washed briefly in ice cold PBS, incubated for 6 h in 30% sucrose solution at 4°C, and frozen at −80°C. Transcripts were detected using an adapted protocol for the RNAscope 2.0 assay for fixed frozen tissue (Advanced Cell Diagnostics). Staining was performed using the RNAscope Fluorescent Multiplex Reagent Kit (Advanced Cell Diagnostics).

Indicated in situ images were adopted from Allen Institute for Brain Science: Allen Developing Mouse Brain Atlas (Lein et al., 2006) (http://developingmouse.brain-map.org) or from the Eurexpress database (Diez-Roux et al., 2011) (http://www.eurexpress.org/ee/).

Reagents used are listed in Table S1. Probes used are listed in Table S4.

Real-time qPCR

RNA was isolated from WT CD1 embryos collected at different developmental stages using RNAeasy kit (Qiagen). Samples were treated with DNase (Qiagen) to prevent contamination with genomic DNA. The specificity of primers was determined by BLAST run of the primer sequences. Annealing temperature was 57°C for all primers.

qPCR reactions were performed once for every gene/sample in triplicate. PCR was performed according to the manufacturer's protocol using Lightcycler 480 SYBR Green 1 Master Mix (Roche). The following thermo cycling program parameters were used for qPCR analysis: incubation step at 95°C for 5 min, then 45 cycles 95°C for 10 s, 57°C for 10 s and 72°C for 10 s. qPCR analysis was carried out using a LightCycler 480 Instrument II (Roche).

ΔCp values were calculated in every sample for each gene of interest with Actb as the reporter gene. Relative change of expression level for the analyzed gene (ΔCp) was performed by subtraction of the gene expression levels in the LV ChP or 4V ChP from the gene expression level of the housekeeping gene (Actb). Next, the ratio of the gene expression level of Actb (Figs 3F, 4F, 5A and Fig. S4F), or of Rn18s for qPCRs with Taqman probes (Fig. 6A, Fig. S7E,I,J), and the gene of interest in either 4V ChP or LV ChP was calculated using the following formula: 2^−ΔCp.

Reagents used are listed in Table S1. Primers are listed in Table S5.

Statistics

Gene expression data

Replicates represent independent experiments. Data in Figs 3F, 4C, 5A, 6A and Fig. S4E and S7E,I,J are represented in columns showing the mean with standard deviation (s.d.). Significance was measured using paired or unpaired two-tailed Student's t-test with unequal sample variance. Biological replicates per condition are indicated in the corresponding graphs. Data in Fig. 5H are represented as columns showing the standard error of the mean (s.e.m.).

Confocal images

Data in Figs 1F-H,J-L, 2E-G,J,K, 6D,F,H,I,K,L and Figs S1F,G,I and S2H,K,L are expressed as columns showing the mean with s.d. Significance was measured using unpaired or paired two-tailed Student's t-test with unequal sample variance. Biological replicates used per condition are indicated in the corresponding graphs.

Western blots

Images used for quantitative analysis reported in Figs S4A,C,D,G and S5D are expressed as columns showing the mean with s.d. Significance was measured using paired two-tailed Student's t-test with unequal sample variance. Biological replicates per condition are indicated in the corresponding graph.

Except for Fig. 1D (human fetal sample), Fig. 4G,K and Figs S2C and S6A, all the displayed immunostaining images and western blots are representative of at least three independent experiments.

We thank members of Bryja and Lehtinen labs for their help and suggestions; Dr Marcela Buchtová and Mgr Marie Šulcová (Masaryk University) for their valuable assistance with RNAscope hybridization; Dr Sean Li (Boston Children's Hospital) for sharing Wnt5a knockout mice with the Lehtinen lab; and the BCH Viral Core. We are also grateful to Nikodém Zezula (Masaryk University) for his assistance with the figure graphic design and Mgr Monika Novákova (Masaryk University) for help with the animal work. We thank MEYS CR for support to the following core facilities: CELLIM of CEITEC supported by the Czech-BioImaging large RI project (LM2018129 funded by MEYS CR), Czech Centre for Phenogenomics (LM2018126), Higher quality and capacity of transgenic model breeding (by MEYS and ERDF, OP RDI CZ.1.05/2.1.00/19.0395), Czech Centre for Phenogenomics: developing towards translation research (by MEYS and ESIF, OP RDE CZ.02.1.01/0.0/0.0/16_013/0001789), BCH viral core for AAV production.

Author contributions

Conceptualization: K.K., M.K.L., V.B.; Methodology: K.K., A.J., M.P.L., B.L., R.M.F., S.G., F.W.; Formal analysis: B.L., F.W.; Investigation: K.K., A.J., P.K., M.P.L.; Resources: J.P., O.M., M.P., D.G., R.v.A., R.A.B., I.B., R.S., Z.K., E.A., M.K.L.; Data curation: A.J., N.D., B.L., F.W.; Writing - original draft: K.K., M.K.L., V.B.; Writing - review & editing: K.K.; Visualization: K.K., P.K., N.D.; Supervision: K.K., M.K.L., V.B.; Project administration: K.K., M.K.L., V.B.; Funding acquisition: K.K., M.K.L., V.B.

Funding

The collaboration between Masarykova Univerzita and the Karolinska Institutet (KI-MU program), was co-financed by the European Social Fund and the state budget of the Czech Republic (CZ.1.07/2.3.00/20.0180). Funding to the V.B. lab was obtained from Neuron Fund for Support of Science (Neuron Nadační Fond Na Podporu Vědy; 23/2016) and the Czech Science Foundation (Grantová Agentura České Republiky; GA17-16680S). This work was supported by European Structural and Investment Funds, Operational Programme Research, Development and Education [‘Preclinical Progression of New Organic Compounds with Targeted Biological Activity’ (Preclinprogress) – CZ.02.1.01/0.0/0.0/16_025/0007381]. Work in the E.A. lab was supported by the Swedish Research Council (Vetenskapsrådet; VR projects: DBRM, 2011-3116, 2011-3318 and 2016-01526), the Swedish Foundation for Strategic Research (Stiftelsen för Strategisk Forskning; SRL program and SLA SB16-0065), the European Commission (NeuroStemcellRepair), the Karolinska Institutet (SFO Strat Regen, Senior grant 2018), Hjärnfonden (FO2015:0202 and FO2017-0059) and Cancerfonden (CAN 2016/572). K.K. was supported by Masarykova Univerzita (MUNI/E/0965/2016). R.v.A. acknowledges funding support from the Universiteit van Amsterdam (MacGillavry fellowship), the Dutch Cancer Society (KWF Kankerbestrijding; career development award ANW 2013-6057) and the Netherlands Science Foundation (NWO, Nederlandse Organisatie voor Wetenschappelijk Onderzoek; VIDI 864.13.002). Work in the O.M. lab is supported by the Czech Science Foundation (Grantová Agentura České Republiky; 18-00514S). Z.K. acknowledges funding support from GACR (Grantová Agentura České Republiky; 18-20759S). R.A.B. is supported by the NIHR Cambridge Biomedical Research Centre at Cambridge University Hospital and the Wellcome Trust/Medical Research Council Cambridge Stem Cell Institute. The M.K.L. laboratory was supported by a Reagan Sloane Shanley Research Internship (to N.D., the Boston Children's Hospital Office of Faculty Development (OFD) Fellowship Award (to R.M.F. and N.D.), the National Institutes of Health R01 NS088566 (to M.K.L.), the New York Stem Cell Foundation (M.K.L.) and Boston Children's Hospital Intellectual and Developmental Disabilities Research Center (1U54HD090255). M.K.L. is a New York Stem Cell Foundation Robertson Investigator. Deposited in PMC for release after 12 months.

Data availability

ChIP-Seq data have been deposited in NCBI Gene Expression Omnibus under accession number GSE166529.

The peer review history is available online at https://journals.biologists.com/dev/article-lookup/doi/10.1242/dev.192054

Alexander
,
C. M.
,
Goel
,
S.
,
Fakhraldeen
,
S. A.
and
Kim
,
S.
(
2012
).
Wnt signaling in mammary glands: plastic cell fates and combinatorial signaling
.
Cold Spring Harb. Perspect. Biol.
4
,
a008037
.
Amin
,
S.
,
Donaldson
,
I. J.
,
Zannino
,
D. A.
,
Hensman
,
J.
,
Rattray
,
M.
,
Losa
,
M.
,
Spitz
,
F.
,
Ladam
,
F.
,
Sagerström
,
C.
and
Bobola
,
N.
(
2015
).
Hoxa2 selectively enhances meis binding to change a branchial arch ground state
.
Dev. Cell
32
,
265
-
277
.
Awatramani
,
R.
,
Soriano
,
P.
,
Rodriguez
,
C.
,
Mai
,
J. J.
and
Dymecki
,
S. M.
(
2003
).
Cryptic boundaries in roof plate and choroid plexus identified by intersectional gene activation
.
Nat. Genet.
35
,
70
-
75
.
Bryja
,
V.
,
Schulte
,
G.
,
Rawal
,
N.
,
Grahn
,
A.
and
Arenas
,
E.
(
2007
).
Wnt-5a induces Dishevelled phosphorylation and dopaminergic differentiation via a CK1-dependent mechanism
.
J. Cell Sci.
120
,
586
-
595
.
Carvalho
,
J. R.
,
Fortunato
,
I. C.
,
Fonseca
,
C. G.
,
Pezzarossa
,
A.
,
Barbacena
,
P.
,
Dominguez-Cejudo
,
M. A.
,
Vasconcelos
,
F. F.
,
Santos
,
N. C.
,
Carvalho
,
F. A.
and
Franco
,
C. A.
(
2019
).
Non-canonical Wnt signaling regulates junctional mechanocoupling during angiogenic collective cell migration
.
eLife
8
,
e45853
.
Cervantes
,
S.
,
Yamaguchi
,
T. P.
and
Hebrok
,
M.
(
2009
).
Wnt5a is essential for intestinal elongation in mice
.
Dev. Biol.
326
,
285
-
294
.
Chau
,
K. F.
,
Springel
,
M. W.
,
Broadbelt
,
K. G.
,
Park
,
H.
,
Topal
,
S.
,
Lun
,
M. P.
,
Mullan
,
H.
,
Maynard
,
T.
,
Steen
,
H.
,
LaMantia
,
A. S.
et al. 
(
2015
).
Progressive differentiation and instructive capacities of amniotic fluid and cerebrospinal fluid proteomes following neural tube closure
.
Dev. Cell
35
,
789
-
802
.
Chen
,
X.
,
He
,
Y.
,
Tian
,
Y.
,
Wang
,
Y.
,
Wu
,
Z.
,
Lan
,
T.
,
Wang
,
H.
,
Cheng
,
K.
and
Xie
,
P.
(
2020
).
Different serotypes of adeno-associated virus vector- and lentivirus-mediated tropism in choroid plexus by intracerebroventricular delivery
.
Hum. Gene Ther.
31
,
440
-
447
.
Choi
,
S.-C.
and
Sokol
,
S. Y.
(
2009
).
The involvement of lethal giant larvae and Wnt signaling in bottle cell formation in Xenopus embryos
.
Dev. Biol.
336
,
68
-
75
.
Cui
,
J.
,
Shipley
,
F. B.
,
Shannon
,
M. L.
,
Alturkistani
,
O.
,
Dani
,
N.
,
Webb
,
M. D.
,
Sugden
,
A. U.
,
Andermann
,
M. L.
and
Lehtinen
,
M. K.
(
2020
).
Inflammation of the embryonic choroid plexus barrier following maternal immune activation
.
Dev. Cell
55
,
617
-
628.e6
.
Currle
,
D. S.
,
Cheng
,
X.
,
Hsu
,
C.-M.
and
Monuki
,
E. S.
(
2005
).
Direct and indirect roles of CNS dorsal midline cells in choroid plexus epithelia formation
.
Development
132
,
3549
-
3559
.
Dani
,
N.
,
Herbst
,
R. H.
,
McCabe
,
C.
,
Green
,
G. S.
,
Kaiser
,
K.
,
Head
,
J. P.
,
Cui
,
J.
,
Shipley
,
F. B.
,
Jang
,
A.
,
Dionne
,
D.
et al. 
(
2021
).
A cellular and spatial map of the choroid plexus across brain ventricles and ages
.
Cell
S0092-8674
,
00438-4
.
Dibner
,
C.
,
Elias
,
S.
and
Frank
,
D.
(
2001
).
XMeis3 protein activity is required for proper hindbrain patterning in Xenopus laevis embryos
.
Development
128
,
3415
-
3426
.
Diez-Roux
,
G.
,
Banfi
,
S.
,
Sultan
,
M.
,
Geffers
,
L.
,
Anand
,
S.
,
Rozado
,
D.
,
Magen
,
A.
,
Canidio
,
E.
,
Pagani
,
M.
,
Peluso
,
I.
et al. 
(
2011
).
A high-resolution anatomical atlas of the transcriptome in the mouse embryo
.
PLoS Biol.
9
,
e1000582
.
Donaldson
,
I. J.
,
Amin
,
S.
,
Hensman
,
J. J.
,
Kutejova
,
E.
,
Rattray
,
M.
,
Lawrence
,
N.
,
Hayes
,
A.
,
Ward
,
C. M.
and
Bobola
,
N.
(
2012
).
Genome-wide occupancy links Hoxa2 to Wnt–β-catenin signaling in mouse embryonic development
.
Nucleic Acids Res.
40
,
3990
-
4001
.
Elkouby
,
Y. M.
,
Polevoy
,
H.
,
Gutkovich
,
Y. E.
,
Michaelov
,
A.
and
Frank
,
D.
(
2012
).
A hindbrain-repressive Wnt3a/Meis3/Tsh1 circuit promotes neuronal differentiation and coordinates tissue maturation
.
Development
139
,
1487
-
1497
.
Fame
,
R. M.
and
Lehtinen
,
M. K.
(
2020
).
Emergence and developmental roles of the cerebrospinal fluid system
.
Dev. Cell
52
,
261
-
275
.
Fumoto
,
K.
,
Takigawa-Imamura
,
H.
,
Sumiyama
,
K.
,
Kaneiwa
,
T.
and
Kikuchi
,
A.
(
2017
).
Modulation of apical constriction by Wnt signaling is required for lung epithelial shape transition
.
Development
144
,
151
-
162
.
Gao
,
B.
,
Song
,
H.
,
Bishop
,
K.
,
Elliot
,
G.
,
Garrett
,
L.
,
English
,
M. A.
,
Andre
,
P.
,
Robinson
,
J.
,
Sood
,
R.
,
Minami
,
Y.
et al. 
(
2011
).
Wnt signaling gradients establish planar cell polarity by inducing Vangl2 phosphorylation through Ror2
.
Dev. Cell
20
,
163
-
176
.
Ghersi-Egea
,
J.-F.
,
Strazielle
,
N.
,
Catala
,
M.
,
Silva-Vargas
,
V.
,
Doetsch
,
F.
and
Engelhardt
,
B.
(
2018
).
Molecular anatomy and functions of the choroidal blood-cerebrospinal fluid barrier in health and disease
.
Acta Neuropathol.
135
,
337
-
361
.
Gou
,
L.
,
Ren
,
X.
and
Ji
,
P.
(
2021
).
Canonical Wnt signaling regulates branching morphogenesis of submandibular gland by modulating levels of lama5
.
Int. J. Dev. Biol.
(in press)
.
Grosse
,
A. S.
,
Pressprich
,
M. F.
,
Curley
,
L. B.
,
Hamilton
,
K. L.
,
Margolis
,
B.
,
Hildebrand
,
J. D.
and
Gumucio
,
D. L.
(
2011
).
Cell dynamics in fetal intestinal epithelium: Implications for intestinal growth and morphogenesis
.
Development
138
,
4423
-
4432
.
Grove
,
E. A.
,
Tole
,
S.
,
Limon
,
J.
,
Yip
,
L.
and
Ragsdale
,
C. W.
(
1998
).
The hem of the embryonic cerebral cortex is defined by the expression of multiple Wnt genes and is compromised in Gli3-deficient mice
.
Development
125
,
2315
-
2325
.
Grumolato
,
L.
,
Liu
,
G.
,
Mong
,
P.
,
Mudbhary
,
R.
,
Biswas
,
R.
,
Arroyave
,
R.
,
Vijayakumar
,
S.
,
Economides
,
A. N.
and
Aaronson
,
S. A.
(
2010
).
Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors
.
Genes Dev.
24
,
2517
-
2530
.
Haddad
,
M. R.
,
Donsante
,
A.
,
Zerfas
,
P.
and
Kaler
,
S. G.
(
2013
).
Fetal brain-directed AAV gene therapy results in rapid, robust, and persistent transduction of mouse choroid plexus epithelia
.
Mol. Ther. Nucleic Acids
2
,
e101
.
Ho
,
H.-Y. H.
,
Susman
,
M. W.
,
Bikoff
,
J. B.
,
Ryu
,
Y. K.
,
Jonas
,
A. M.
,
Hu
,
L.
,
Kuruvilla
,
R.
and
Greenberg
,
M. E.
(
2012
).
Wnt5a-Ror-Dishevelled signaling constitutes a core developmental pathway that controls tissue morphogenesis
.
Proc. Natl. Acad. Sci. USA
109
,
1
-
8
.
Hovanes
,
K.
,
Li
,
T. W. H.
,
Munguia
,
J. E.
,
Truong
,
T.
,
Milovanovic
,
T.
,
Lawrence Marsh
,
J.
,
Holcombe
,
R. F.
and
Waterman
,
M. L.
(
2001
).
β -catenin – sensitive isoforms of lymphoid enhancer factor-1 are selectively expressed in colon cancer
.
Nat. Genet.
28
,
53
-
57
.
Huang
,
L.
,
Pu
,
Y.
,
Hu
,
W. Y.
,
Birch
,
L.
,
Luccio-Camelo
,
D.
,
Yamaguchi
,
T.
and
Prins
,
G. S.
(
2009
).
The role of Wnt5a in prostate gland development
.
Dev. Biol.
328
,
188
-
199
.
Humphries
,
A. C.
and
Mlodzik
,
M.
(
2018
).
From instruction to output: Wnt/PCP signaling in development and cancer
.
Curr. Opin. Cell Biol.
51
,
110
-
116
.
Hunter
,
N. L.
and
Dymecki
,
S. M.
(
2007
).
Molecularly and temporally separable lineages form the hindbrain roof plate and contribute differentially to the choroid plexus
.
Development
134
,
3449
-
3460
.
Jho
,
E.-H.
,
Zhang
,
T.
,
Domon
,
C.
,
Joo
,
C.-K.
,
Freund
,
J.-N.
and
Costantini
,
F.
(
2002
).
Wnt/β-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway
.
Mol. Cell. Biol.
22
,
1172
-
1183
.
Johansson
,
P. A.
,
Irmler
,
M.
,
Acampora
,
D.
,
Beckers
,
J.
,
Simeone
,
A.
and
Götz
,
M.
(
2013
).
The transcription factor Otx2 regulates choroid plexus development and function
.
Development
140
,
1055
-
1066
.
Kaiser
,
K.
,
Gyllborg
,
D.
,
Procházka
,
J.
,
Salašová
,
A.
,
Kompaníková
,
P.
,
Molina
,
F. L.
,
Laguna-Goya
,
R.
,
Radaszkiewicz
,
T.
,
Harnoš
,
J.
,
Procházková
,
M.
et al. 
(
2019
).
WNT5A is transported via lipoprotein particles in the cerebrospinal fluid to regulate hindbrain morphogenesis
.
Nat. Commun.
10
,
1
-
15
.
Kallay
,
L. M.
,
McNickle
,
A.
,
Brennwald
,
P. J.
,
Hubbard
,
A. L.
and
Braiterman
,
L. T.
(
2006
).
Scribble associates with two polarity proteins, Lgl2 and Vangl2, via distinct molecular domains
.
J. Cell. Biochem.
99
,
647
-
664
.
Kato
,
M.
,
Soprano
,
D. R.
,
Makover
,
A.
,
Kato
,
K.
,
Herbert
,
J.
and
Goodman
,
D. W. S.
(
1986
).
Localization of immunoreactive transthyretin (prealbumin) and of transthyretin mRNA in fetal and adult rat brain
.
Differentiation
31
,
228
-
235
.
Kessenbrock
,
K.
,
Smith
,
P.
,
Steenbeek
,
S. C.
,
Pervolarakis
,
N.
,
Kumar
,
R.
,
Minami
,
Y.
,
Goga
,
A.
,
Hinck
,
L.
and
Werb
,
Z.
(
2017
).
Diverse regulation of mammary epithelial growth and branching morphogenesis through noncanonical Wnt signaling
.
Proc. Natl. Acad. Sci. USA
114
,
3121
-
3126
.
Kumawat
,
K.
and
Gosens
,
R.
(
2015
).
WNT-5A: signaling and functions in health and disease
.
Cell. Mol. Life Sci.
73
,
567
-
587
.
Langford
,
M. B.
,
O'Leary
,
C. J.
,
Veeraval
,
L.
,
White
,
A.
,
Lanoue
,
V.
and
Cooper
,
H. M.
(
2020
).
WNT5a regulates epithelial morphogenesis in the developing choroid plexus
.
Cereb. Cortex
30
,
3617
-
3631
.
Langmead
,
B.
,
Trapnell
,
C.
,
Pop
,
M.
and
Salzberg
,
S. L.
(
2009
).
Ultrafast and memory-efficient alignment of short DNA sequences to the human genome
.
Genome Biol.
10
,
R25
.
Laurent
,
B.
,
Ruitu
,
L.
,
Murn
,
J.
,
Hempel
,
K.
,
Ferrao
,
R.
,
Xiang
,
Y.
,
Liu
,
S.
,
Garcia
,
B. A.
,
Wu
,
H.
,
Wu
,
F.
et al. 
(
2015
).
A specific LSD1/KDM1A isoform regulates neuronal differentiation through H3K9 demethylation
.
Mol. Cell
57
,
957
-
970
.
Lehtinen
,
M. K.
,
Zappaterra
,
M. W.
,
Chen
,
X.
,
Yang
,
Y. J.
,
Hill
,
A. D.
,
Lun
,
M.
,
Maynard
,
T.
,
Gonzalez
,
D.
,
Kim
,
S.
,
Ye
,
P.
et al. 
(
2011
).
The cerebrospinal fluid provides a proliferative niche for neural progenitor cells
.
Neuron
69
,
893
-
905
.
Lein
,
E. S.
,
Hawrylycz
,
M. J.
,
Ao
,
N.
,
Ayres
,
M.
,
Bensinger
,
A.
,
Bernard
,
A.
,
Boe
,
A. F.
,
Boguski
,
M. S.
,
Brockway
,
K. S.
,
Byrnes
,
E. J.
et al. 
(
2006
).
Genome-wide atlas of gene expression in the adult mouse brain
.
Nature
445
,
168
-
176
.
Lewis
,
A. E.
,
Vasudevan
,
H. N.
,
O'Neill
,
A. K.
,
Soriano
,
P.
and
Bush
,
J. O.
(
2013
).
The widely used Wnt1-Cre transgene causes developmental phenotypes by ectopic activation of Wnt signaling
.
Dev. Biol.
379
,
229
-
234
.
Li
,
C.
,
Hu
,
L.
,
Xiao
,
J.
,
Chen
,
H.
,
Li
,
J. T.
,
Bellusci
,
S.
,
Delanghe
,
S.
and
Minoo
,
P.
(
2005
).
Wnt5a regulates Shh and Fgf10 signaling during lung development
.
Dev. Biol.
287
,
86
-
97
.
Liddelow
,
S. A.
,
Dziegielewska
,
K. M.
,
VandeBerg
,
J. L.
and
Saunders
,
N. R.
(
2010
).
Development of the lateral ventricular choroid plexus in a marsupial, Monodelphis domestica
.
Cerebrospinal Fluid Res.
7
,
16
.
Lun
,
M. P.
,
Johnson
,
M. B.
,
Broadbelt
,
K. G.
,
Watanabe
,
M.
,
Kang
,
Y.-J.
,
Chau
,
K. F.
,
Springel
,
M. W.
,
Malesz
,
A.
,
Sousa
,
A. M. M.
,
Pletikos
,
M.
et al. 
(
2015
).
Spatially heterogeneous choroid plexus transcriptomes encode positional identity and contribute to regional CSF production
.
J. Neurosci.
35
,
4903
-
4916
.
Machon
,
O.
,
Masek
,
J.
,
Machonova
,
O.
,
Krauss
,
S.
and
Kozmik
,
Z.
(
2015
).
Meis2 is essential for cranial and cardiac neural crest development
.
BMC Dev. Biol.
15
,
40
.
Madisen
,
L.
,
Zwingman
,
T. A.
,
Sunkin
,
S. M.
,
Oh
,
S. W.
,
Zariwala
,
H. A.
,
Gu
,
H.
,
Ng
,
L. L.
,
Palmiter
,
R. D.
,
Hawrylycz
,
M. J.
,
Jones
,
A. R.
et al. 
(
2010
).
A robust and high-throughput Cre reporting and characterization system for the whole mouse brain
.
Nat. Neurosci.
13
,
133
-
140
.
Mentink
,
R. A.
,
Rella
,
L.
,
Radaszkiewicz
,
T. W.
,
Gybel
,
T.
,
Betist
,
M. C.
,
Bryja
,
V.
and
Korswagen
,
H. C.
(
2018
).
The planar cell polarity protein VANG-1/Vangl negatively regulates Wnt/β-catenin signaling through a Dvl dependent mechanism
.
PLoS Genet.
14
,
e1007840
.
Mercader
,
N.
,
Selleri
,
L.
,
Criado
,
L. M.
,
Pallares
,
P.
,
Parras
,
C.
,
Cleary
,
M. L.
and
Torres
,
M.
(
2009
).
Ectopic Meis1 expression in the mouse limb bud alters P-D patterning in a Pbx1-independent manner
.
Int. J. Dev. Biol.
53
,
1483
-
1494
.
Muthusamy
,
N.
,
Vijayakumar
,
A.
,
Cheng
,
G.
and
Ghashghaei
,
H. T.
(
2014
).
A Knock-in Foxj1CreERT2:: GFP mouse for recombination in epithelial cells with motile cilia
.
Genesis
52
,
350
-
358
.
Nakamura
,
E.
,
Nguyen
,
M.-T.
and
Mackem
,
S.
(
2006
).
Kinetics of tamoxifen-regulated Cre activity in mice using a cartilage-specific CreERT to assay temporal activity windows along the proximodistal limb skeleton
.
Dev. Dyn.
235
,
2603
-
2612
.
Niehrs
,
C.
(
2012
).
The complex world of WNT receptor signalling
.
Nat. Rev. Mol. Cell Biol.
13
,
767
-
779
.
Nielsen
,
C. M.
and
Dymecki
,
S. M.
(
2010
).
Sonic hedgehog is required for vascular outgrowth in the hindbrain choroid plexus
.
Dev. Biol.
340
,
430
-
437
.
Philipp
,
I.
,
Aufschnaiter
,
R.
,
Ozbek
,
S.
,
Pontasch
,
S.
,
Jenewein
,
M.
,
Watanabe
,
H.
,
Rentzsch
,
F.
,
Holstein
,
T. W.
and
Hobmayer
,
B.
(
2009
).
Wnt/beta-catenin and noncanonical Wnt signaling interact in tissue evagination in the simple eumetazoan Hydra
.
Proc. Natl. Acad. Sci. USA
106
,
4290
-
4295
.
Pietilä
,
I.
,
Prunskaite-Hyyryläinen
,
R.
,
Kaisto
,
S.
,
Tika
,
E.
,
van Eerde
,
A. M.
,
Salo
,
A. M.
,
Garma
,
L.
,
Miinalainen
,
I.
,
Feitz
,
W. F.
,
Bongers
,
E. M. H. F.
et al. 
(
2016
).
Wnt5a deficiency leads to anomalies in ureteric tree development, tubular epithelial cell organization and basement membrane integrity pointing to a role in kidney collecting duct patterning
.
PLoS ONE
11
,
e0147171
.
Pourreyron
,
C.
,
Reilly
,
L.
,
Proby
,
C.
,
Panteleyev
,
A.
,
Fleming
,
C.
,
McLean
,
K.
,
South
,
A. P.
and
Foerster
,
J.
(
2012
).
Wnt5a is strongly expressed at the leading edge in non-melanoma skin cancer, forming active gradients, while canonical Wnt signalling is repressed
.
PLoS ONE
7
,
e31827
.
Ramakrishnan
,
V. M.
,
Tien
,
K. T.
,
McKinley
,
T. R.
,
Bocard
,
B. R.
,
McCurry
,
T. M.
,
Williams
,
S. K.
,
Hoying
,
J. B.
and
Boyd
,
N. L.
(
2016
).
Wnt5a Regulates the Assembly of Human Adipose Derived Stromal Vascular Fraction-Derived Microvasculatures
.
PLoS ONE
11
,
e0151402
.
Roarty
,
K.
,
Baxley
,
S. E.
,
Crowley
,
M. R.
,
Frost
,
A. R.
and
Serra
,
R.
(
2009
).
Loss of TGF-β or Wnt5a results in an increase in Wnt/β-catenin activity and redirects mammary tumour phenotype
.
Breast Cancer Res.
11
,
R19
.
Ryu
,
Y. K.
,
Collins
,
S. E.
,
Ho
,
H.-Y. H.
,
Zhao
,
H.
and
Kuruvilla
,
R.
(
2013
).
An autocrine Wnt5a-Ror signaling loop mediates sympathetic target innervation
.
Dev. Biol.
377
,
79
-
89
.
Saito-Diaz
,
K.
,
Chen
,
T. W.
,
Wang
,
X.
,
Thorne
,
C. A.
,
Wallace
,
H. A.
,
Page-McCaw
,
A.
and
Lee
,
E.
(
2013
).
The way Wnt works: components and mechanism
.
Growth Factors
31
,
1
-
31
.
Sato
,
A.
,
Yamamoto
,
H.
,
Sakane
,
H.
,
Koyama
,
H.
and
Kikuchi
,
A.
(
2010
).
Wnt5a regulates distinct signalling pathways by binding to Frizzled2
.
EMBO J.
29
,
41
-
54
.
Silva-Vargas
,
V.
,
Maldonado-Soto
,
A. R.
,
Mizrak
,
D.
,
Codega
,
P.
and
Doetsch
,
F.
(
2016
).
Age-dependent Niche signals from the choroid plexus regulate adult neural stem cells
.
Cell Stem Cell
19
,
643
-
652
.
Sittaramane
,
V.
,
Pan
,
X.
,
Glasco
,
D. M.
,
Huang
,
P.
,
Gurung
,
S.
,
Bock
,
A.
,
Li
,
S.
,
Wang
,
H.
,
Kawakami
,
K.
,
Matise
,
M. P.
et al. 
(
2013
).
The PCP protein Vangl2 regulates migration of hindbrain motor neurons by acting in floor plate cells, and independently of cilia function
.
Dev. Biol.
382
,
400
-
412
.
Stephens
,
W. Z.
,
Senecal
,
M.
,
Nguyen
,
M.
and
Piotrowski
,
T.
(
2010
).
Loss of adenomatous polyposis coli (apc) results in an expanded ciliary marginal zone in the zebrafish eye
.
Dev. Dyn.
239
,
2066
-
2077
.
Sui
,
L.
and
Dahmann
,
C.
(
2020
).
Wingless counteracts epithelial folding by increasing mechanical tension at basal cell edges in Drosophila
.
Development
147
,
dev184713
.
Sun
,
M. Z.
,
Oh
,
M. C.
,
Ivan
,
M. E.
,
Kaur
,
G.
,
Safaee
,
M.
,
Kim
,
J. M.
,
Phillips
,
J. J.
,
Auguste
,
K. I.
and
Parsa
,
A. T.
(
2014
).
Current management of choroid plexus carcinomas
.
Neurosurg. Rev.
37
,
179
-
192
.
Tamai
,
K.
,
Semenov
,
M.
,
Kato
,
Y.
,
Spokony
,
R.
,
Liu
,
C.
,
Katsuyama
,
Y.
,
Hess
,
F.
,
Saint-Jeannet
,
J.-P.
and
He
,
X.
(
2000
).
LDL-receptor-related proteins in Wnt signal transduction
.
Nature
407
,
530
.
Topol
,
L.
,
Jiang
,
X.
,
Choi
,
H.
,
Garrett-Beal
,
L.
,
Carolan
,
P. J.
and
Yang
,
Y.
(
2003
).
Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent β-catenin degradation
.
J. Cell Biol.
162
,
899
-
908
.
van Amerongen
,
R.
,
Fuerer
,
C.
and
Mizutani
,
M.
(
2012
).
Wnt5a can both activate and repress Wnt/β-catenin signaling during mouse embryonic development
.
Dev. Biol.
369
,
101
-
114
.
Wilting
,
J.
and
Christ
,
B.
(
1989
).
An experimental and ultrastructural study on the development of the avian choroid plexus
.
Cell Tissue Res.
255
,
487
-
494
.
Xu
,
H.
,
Fame
,
R. M.
,
Sadegh
,
C.
,
Sutin
,
J.
,
Naranjo
,
C.
,
Syau
,
C. D.
,
Cui
,
J.
,
Shipley
,
F. B.
,
Vernon
,
A.
,
Gao
,
F.
et al. 
(
2021
).
Choroid plexus NKCC1 mediates cerebrospinal fluid clearance during mouse early postnatal development
.
Nat. Commun.
12
,
447
.
Yamaguchi
,
T. P.
,
Bradley
,
A.
,
McMahon
,
A. P.
and
Jones
,
S.
(
1999
).
A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo
.
Development
126
,
1211
-
1223
.
Yamamoto
,
H.
,
Yoo
,
S. K.
,
Nishita
,
M.
,
Kikuchi
,
A.
and
Minami
,
Y.
(
2007
).
Wnt5a modulates glycogen synthase kinase 3 to induce phosphorylation of receptor tyrosine kinase Ror2
.
Genes Cells
12
,
1215
-
1223
.
Yamamoto
,
H.
,
Awada
,
C.
,
Matsumoto
,
S.
,
Kaneiwa
,
T.
,
Sugimoto
,
T.
,
Takao
,
T.
and
Kikuchi
,
A.
(
2015
).
Basolateral secretion of Wnt5a in polarized epithelial cells is required for apical lumen formation
.
J. Cell Sci.
128
,
1051
-
1063
.
Zhang
,
Y.
,
Liu
,
T.
,
Meyer
,
C. A.
,
Eeckhoute
,
J.
,
Johnson
,
D. S.
,
Bernstein
,
B. E.
,
Nussbaum
,
C.
,
Myers
,
R. M.
,
Brown
,
M.
,
Li
,
W.
et al. 
(
2008
).
Model-based analysis of ChIP-Seq (MACS)
.
Genome Biol.
9
,
R137
.
Zhu
,
L. J.
,
Gazin
,
C.
,
Lawson
,
N. D.
,
Pagès
,
H.
,
Lin
,
S. M.
,
Lapointe
,
D. S.
and
Green
,
M. R.
(
2010
).
ChIPpeakAnno: a Bioconductor package to annotate ChIP-seq and ChIP-chip data
.
BMC Bioinformatics
11
,
237
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information