Skip to main content
Advertisement
Browse Subject Areas
?

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

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Transcriptional Profiling of Wnt3a Mutants Identifies Sp Transcription Factors as Essential Effectors of the Wnt/β-catenin Pathway in Neuromesodermal Stem Cells

  • William C. Dunty Jr ,

    Contributed equally to this work with: William C. Dunty Jr, Mark W. L. Kennedy

    Affiliation Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America

  • Mark W. L. Kennedy ,

    Contributed equally to this work with: William C. Dunty Jr, Mark W. L. Kennedy

    Affiliation Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America

  • Ravindra B. Chalamalasetty,

    Affiliation Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America

  • Kenneth Campbell,

    Affiliation Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America

  • Terry P. Yamaguchi

    yamagute@mail.nih.gov

    Affiliation Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America

Correction

18 Apr 2014: The PLOS ONE Staff (2014) Correction: Transcriptional Profiling of Wnt3a Mutants Identifies Sp Transcription Factors as Essential Effectors of the Wnt/β-catenin Pathway in Neuromesodermal Stem Cells. PLOS ONE 9(4): e95947. https://doi.org/10.1371/journal.pone.0095947 View correction

Abstract

Neuromesodermal (NM) stem cells reside in the primitive streak (PS) of gastrulating vertebrate embryos and generate precursors of the spinal cord and musculoskeletal system. Although Wnt3a/β-catenin signaling is crucial for NM stem cell maintenance and differentiation, few key transcriptional effectors have been identified. Through a concerted transcriptional profiling and genetic approach we have determined that two Zn2+-finger transcription factors, Sp5 and Sp8, are regulated by Wnt3a in the PS, and are essential for neural and musculoskeletal patterning. These results identify Sp5 and Sp8 as pivotal downstream effectors of Wnt3a, and suggest that they are essential for the self-renewal and differentiation of NM stem cells.

Introduction

Germ layer specification is a fundamental aspect of embryogenesis that occurs in the PS region during gastrulation. Under the influence of signals emanating from the PS, pluripotent epiblast stem cells differentiate into the multipotent progenitors that will form the germ layers, ie. the ectoderm, mesoderm and endoderm. As development proceeds, the differentiation potential of descendants of these early embryonic stem cells becomes progressively restricted [1]. For example, the descendants of NM stem cells, which originate in the epiblast adjacent to the PS, become restricted to the multipotent neuroectoderm and paraxial mesoderm cell lineages upon differentiation [2][6]. The response of NM stem cells to signals secreted by the PS, such as Wnt3a, determines whether they self-renew or differentiate into these lineages, however the mechanisms that govern NM cell fate decisions are not well understood.

Wnt3a is a member of the ‘Wnt’ family of secreted glycoproteins that interact with Frizzled/Lrp receptor complexes to prevent the proteolytic degradation of the transcriptional co-activator β-catenin. In the presence of Wnt3a, stabilized β-catenin enters the nucleus where it interacts with members of the Tcf/Lef family of DNA binding transcription factors. Together, β-catenin and Tcf/Lef form the core of a multipartite transcription complex that controls target gene expression [7]. Embryos lacking Wnt3a die at mid-gestation with severe posterior axial truncations that reflect a loss of neuro-musculoskeletal progenitors [4], [8][12]. Cells expressing the stem cell marker and T-box transcription factor, T, including NM stem cells, are transiently established in Wnt3a−/− mutants but are not maintained [10]. Interestingly, Wnt3a−/− mutants form ectopic neural tubes that are speculated to arise from the transfating of paraxial mesoderm progenitors in the absence of a Wnt signal [10], [12]. Similar defects are also observed in Tcf1−/−;Lef1−/− embryos [13]. Together, these findings strongly suggest that Wnt3a/β-catenin signaling regulates the maintenance and differentiation of NM stem cells. A thorough understanding of the target genes and the gene regulatory network controlled by Wnt3a will provide crucial insights into how the fundamental neural and paraxial mesoderm lineages are established.

Here we report the comprehensive analysis of the Wnt3a-dependent transcriptome in PS and node stem cells. In silico analysis demonstrates that Wnt3a regulates the expression of many genes associated with the primitive streak, neuroectoderm and paraxial mesoderm. Our analysis identified 2 candidate target genes, Sp5 and Sp8, that when genetically disrupted recapitulate the Wnt3a null phenotype. Thus, Sp5 and Sp8 are redundant factors that function as key effectors of the Wnt3a/β-catenin pathway in NM stem cells.

Materials and Methods

Mice

Ctnnb1tm2Kem, Ctnnb1lox(ex3), Wnt3a, T-cre, Sp5lacZ/lacZ and Sp8Δ/+ mice were described previously [10], [14][18]. T-cre mice were used to conditionally delete Ctnnb1tm2Kem or Ctnnb1lox(ex3) in NM stem cells and their progeny. Sp5;Sp8 dko embryos were obtained by intercrossing Sp5lacZ/lacZ;Sp8Δ/+mice. This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Frederick National Lab Animal Care and Use Committee (Animal Study Proposal #12-408). All efforts were made to minimize suffering. Rodents were euthanized by CO2 inhalation in accordance with the most recent AVMA Guidelines on Euthanasia.

Gene Expression Profiling

Total RNA was isolated from dissected node and PS regions of E7.75-E8 wildtype and Wnt3a mutant embryos as previously described [15]. cDNA and cRNA synthesis, hybridization to GeneChip® Mouse Genome 430 2.0 Arrays, washing, staining, and array scanning were carried out according to manufacturer’s protocols (Affymetrix, Santa Clara, CA) as previously described [15], [19]. Briefly, 30–50 ng of total RNA from individual embryos was used as template for first cycle cDNA synthesis. For second cycle cDNA synthesis, equivalent amounts of cRNA from three stage-matched embryos per genotype were pooled (600 ng total) to generate labeled-target per array. Array hybridizations were performed in triplicate per genotype.

Statistical analysis was performed on probe-intensity level data (CEL files) using BRB ArrayTools version 4.3.0 or GeneSpringGX software, with similar results. Data presented herein was generated using BRB ArrayTools. Genes were considered statistically significant if their parametric p-value ≤0.05. For hierarchical clustering of genes in Fig. 1F, centered correlation and average linkage method was used for the maximally expressed probe set measured by average intensity across all arrays. Signal transduction and tissue pathway analysis was performed using Genomatix suite (GePS) software. The microarray data in this study was previously deposited in the GEO database under accession code GSE29995 [19].

thumbnail
Figure 1. Transcriptional profiling of wild type and Wnt3a−/− embryos.

A) WISH of a wt embryo at E7.75 illustrating Wnt3a expression in the PS and node (left panel). (right panel) The corresponding domain of Wnt/β-catenin activity as assessed by X-gal staining of the transgenic Wnt/β-catenin reporter, BAT-lacZ [15]. Dotted lines demarcate dissection boundaries. B) Volcano plot depicting all statistically significant DEGs (p<0.05, indicated by blue color). C–E) Gene ontology (GO) analysis of DEGs identified a highly significant association with the Wnt/β-catenin pathway (C, D) and with expression in embryonic mesoderm, somites and neural tissue (E). D) A selection of the shortest path network representation of Wnt pathway genes in (C) analyzed in Genomatix GePS. For complete network see Figure S2. F) Analysis of tissue-specific DEGs shows that posteriorly restricted genes expressed in the node/PS, PSM, and NP are down-regulated while anterior PSM, anterior NP, IM, and LPM genes were up-regulated. Green: down-regulated genes; Red: up-regulated genes.

https://doi.org/10.1371/journal.pone.0087018.g001

In situ Hybridization, Immunofluorescence and Tissue Sectioning

Brachyury, Wnt3a and Uncx4.1 probe synthesis and whole-mount in situ hybridizations (WISH) were performed as previously described [15]. The mouse Sp5 cDNA (cat# EMM1002-5885071) was obtained from Open Biosystems. The mouse Sp8 DNA template was obtained by TA-TOPO cloning (Invitrogen) the Sp8 coding region from ES cell cDNA. Paraffin and cryosectioning was performed at the Pathology and Histology Laboratory (PHL, NCI-Frederick). Cryosections were washed in 1×PBT (1×PBS +0.1% tween-20), incubated in 1×PBS +0.2% Triton-X for 20 minutes, washed in PBT. Sections were incubated in primary antibodies in 0.02% block (Roche Blocking reagent in PBT, filtered) overnight at 4°C. Sections were then washed 3 times in PBT. Secondary antibodies in 0.02% block were applied at room temperature for 2 hours. Sections were washed 3 times, 1 nM DAPI (4′,6-diamidino-2-phenylindole) was included in the second wash for 20 minutes. Embryos/sections were photographed on a Leica MZFLIII stereoscope equipped with a Zeiss Axiovision HRc digital camera, and Zeiss Axiovision imaging software (v4.5). Unless indicated otherwise, at least 3 mutant embryos were examined with each probe/antibody, and all yielded similar results.

ES Cell Culture and mRNA Expression Analysis

T-GFP ES cells were maintained on gelatin-coated plates and differentiated as embyoid bodies (EBs) in serum-free, feeder-free conditions, as previously described [20]. Day 2 EBs were dissociated with Trypsin-EDTA and 4×105 cells were cultured in ultra-low attachment 6-well dishes with 100 ng/ml recombinant Wnt3a (R&D Systems) or 1 µM CHIR99021 (ToCris) for 2 days. Total RNA was extracted using the RNeasy miniprep kit (Qiagen) and first stand cDNA synthesized using iScript cDNA synthesis kit (Quanta Biosciences). Relative gene expression was analyzed with iTaq universal SYBR Green (BioRad) using the CFX96 Real-Time PCR Detection system (BioRad). Relative fold changes were calculated by normalizing to Gapdh levels using the 2−ΔΔCt method. qPCR primers are listed in Table S4.

Results

Transcriptional Profiling of PS and Node Stem Cells

Wnt3a is highly expressed in the node and primitive streak during gastrulation, where it signals via β-catenin and Tcf1/Lef1 (Fig. 1A; [15]) to regulate genes essential for neuromesodermal development. To identify the effectors of this pathway, we used microarrays to generate transcriptional profiles of wild type and Wnt3a−/− primitive streak and node tissue. To enrich for early molecular regulators, tissues were excised between E7.75–8.0 when Wnt3a−/− mutants are morphologically indistinguishable from wild type controls. Comparisons of wild type and mutant transcriptional profiles identified 729 differentially expressed genes (DEGs, p<0.05) of which 379 were down-regulated and 350 were up-regulated (Fig. 1B, Table S1 and Figure S1). Included amongst the down-regulated genes are several well-known targets that encode components of the Wnt/β-catenin signaling pathway itself, ex. Axin2, Nkd1, and Lef1 (Fig. 1B, Table S1), thereby providing an initial validation of the screen [21][23]. An assessment of DEGs for their association with signaling pathways revealed that the Wnt/β-catenin pathway was most highly represented followed closely by the Notch, Hedgehog, and Fgf pathways (Fig. 1C, Table S2). The DEG data was mined further for associations with literature-based gene regulatory networks using Genomatix Pathway Systems software. This approach identified an interacting network of Wnt/β-catenin pathway genes that function, in part, to control the expression of Axin2 (Fig. 1D, Fig. S2). Included in this network of Wnt target genes are the transcription factors Msgn1 and Tbx6 that play critical roles in paraxial mesoderm and neural development [19], [24], [25]. Interestingly, two members of the large and well-characterized Sp1 family of DNA-binding Zn2+-finger transcription factors, Sp5 and Sp8, were also identified in this network.

We then asked if these DEGs are expressed in the appropriate embryonic tissues to be targets of Wnt3a. Detailed analysis of the DEGs using their associated gene ontology (GO) terms revealed that, as expected, their expression is most commonly associated with stem cells, the PS, neural plate, and mesoderm (Fig. 1E, Table S3). Analysis of genes clustered by their tissue-specific expression patterns revealed that primitive streak and posterior PSM genes were down regulated in Wnt3a−/−embryos, while anterior PSM genes were increased (Fig. 1F). These results are consistent with Wnt3a maintaining immature posterior PSM progenitors, while suppressing segmentation genes in the anterior PSM [19], [26]. Interestingly, intermediate and lateral plate mesoderm genes showed increased expression levels in the mutants suggesting Wnt3a has a mesodermal patterning role in the PS. It is also notable that anterior neural plate markers increased, but posterior neural markers decreased in the Wnt3a mutants, likely reflecting the established neural posteriorizing role for Wnt signaling [27], [28].

Validation of Candidate Wnt3a Target Genes

We focused on the validation of putative Wnt3a target genes that encode transcription factors, as they frequently function as master regulators of stem cell fate. Microarray analysis showed that the Sp5, Sp8, Klf5, Mixl1, and Lmo2 transcription factors were all down-regulated in Wnt3a mutants, and were thus selected for further expression analysis in vivo, along with the secreted Wnt feedback inhibitor Notum. We reasoned that candidate Wnt3a target genes should be spatially and temporally co-expressed with Wnt3a and be responsive to changes in Wnt3a/β-catenin signaling. WISH analysis demonstrated that Sp5, Sp8, Notum, Klf5, and Mixl1 were co-expressed with Wnt3a (Fig. 1a, left) in the wild type node/primitive steak region at E7.5–8.5 (Fig. 2A, B, F, G, K, L, P, Q, U, V). Lmo2 was undetectable in the PS at E7.5, but was easily detected at E8.5 (Fig. 2Z, AA). The lack of Lmo2 expression at E7.5, when Wnt3a is easily detected in the PS, suggests that Lmo2 requires additional signals for activation. Reduced Wnt3a/β-catenin activity led to significantly diminished expression of all 6 genes in the PS and posterior mesoderm domains of Wnt3a−/− embryos (Fig. 2C, H, M, R, W, BB), and in embryos conditionally lacking Ctnnb1 (β-catenin) activity in mesoderm progenitors (Fig. 2D, I, N, S, X, CC). Correspondingly, these genes were all highly up-regulated in the PS and PSM of embryos in which β-catenin was constitutively activated in mesoderm progenitors (Fig. 2E, J, O, T, Y, DD). Thus, all 6 genes lie downstream of Wnt3a and β-catenin in vivo, thereby validating the microarray results and furthering their candidacy as effectors of the Wnt3a/β-catenin signaling pathway.

thumbnail
Figure 2. Validation of candidate Wnt3a target gene expression in Wnt3a and Ctnnb1 mutants by WISH.

A comparison of Sp5 (A–E), Sp8 (F–J), Notum (K–O), Klf5 (P–T), MixL1 (U–Y), and Lmo2 (Z-DD) expression in wild type (A, B, F, G, K, L, P, Q, U, V, Z and AA), Wnt3a−/− (C, H, M, R, W and BB), Tcre;Ctnnb1flLOF (D, I, N, S, X and CC) and Tcre;Ctnnb1flGOF (E, J, O, T, Y and DD) embryos at E7.5 and 8.5 (LOF: loss-of-function; GOF: gain-of-function). Arrowhead in B–E points to the PS.

https://doi.org/10.1371/journal.pone.0087018.g002

Sp5 and Sp8 are Wnt3a-responsive and Expressed in the PS

Sp5 and Sp8 were identified by microarray as two of the more prominently down regulated genes in Wnt3a mutants. The lack of a phenotype in Sp5 null mice, taken together with the observation that Sp1, Sp5, and Sp8 all bind to the same DNA sequence in vitro [17], [29], [30], suggests that Sp factors could function redundantly during development. To determine whether additional members of the Sp family are expressed in the PS and are Wnt3a-responsive, we turned to an in vitro mouse embryonic stem cell (ESC) model of PS formation [20]. ESCs were cultured in defined media to allow for the stimulation of the Wnt/βcatenin signaling pathway with purified, recombinant Wnt3a protein in feeder-free and serum-free conditions. ESCs were primed for germ layer differentiation by withdrawing LIF and forming embryoid bodies (EBs) for 2 days. EBs were then treated with Wnt3a or the GSK-3β inhibitor CHIR99021 for an additional 2 days to promote PS and mesoderm gene expression [20], [31], [32]. Quantitative PCR of EBs revealed that of the 9 Sp genes (Sp1-9), only Sp5 and Sp8 were expressed during the in vitro differentiation process (Fig. 3A, D4 EB untreated). These expression levels were dramatically augmented by treatment with Wnt3a protein or CHIR99021 (Fig. 3A) demonstrating that Sp5 and Sp8 were induced during PS formation and were highly Wnt3a-responsive.

thumbnail
Figure 3. Analysis of Sp1-like family members for co-expression and Wnt3a responsiveness.

A) qPCR analysis of the Sp1-like family mRNA levels in ESCs stimulated with recombinant Wnt3a protein or the GSK3β inhibitor CHIR99021 on day 2 (D2) and assayed on day 4 (D4). mRNA levels were normalized to Gapdh expression. Values represent the mean, calculated from 3 independent qPCR reactions. Error bars, one standard deviation; *, statistical significance (p<0.05, 2-tailed students t-test); n.s., not significant. WISH of Sp5 (B) and Sp8 (C) at E8.5 illustrates the overlapping expression in the midbrain and the posterior of the embryo. (D, E) Sections generated at the positions indicated by lines in (B) and (C) show Sp5 and Sp8 co-expression across all three germ layers. Ectoderm (e), Mesoderm (m), and Hindgut (HG).

https://doi.org/10.1371/journal.pone.0087018.g003

To examine whether Sp5 and Sp8 are expressed in the PS in vivo, a detailed comparative analysis was performed on serial sections of E8.5 embryos processed for WISH (Fig. 3B, C). Section analysis shows definitively that Sp5 and Sp8 are both expressed in the PS and across all three germ layers. In addition to the expression in the PS ectoderm and mesoderm, Sp5 is highly expressed in the dorsal hindgut (Fig. 3D). Sp8 is weakly expressed in the dorsal hindgut but is highly expressed in the PS epiblast/caudal neural plate (Fig. 3E). Together, these results demonstrate that Sp5 and Sp8 respond to Wnt3a/β-catenin activity, and are co-expressed in the PS, including the NM stem cells in the caudal lateral epiblast.

Sp5 and Sp8 Function in the Wnt3a Genetic Pathway

Our bioinformatics analysis and ESC experiments suggest that Sp5 and Sp8 could participate in Wnt signaling. To address this hypothesis we first asked if Sp5 genetically interacts with Wnt3a. Although Sp5LacZ/LacZ null mice are viable and fertile, both homozygotes and heterozygotes develop “kinked” tails at a frequency of ∼2–3% (Fig. 4E–F and Fig. S3). This phenotype is observed at a similar frequency in Wnt3a heterozygotes (7.7%, Fig. 4F) and is indicative of defective somite formation leading to fused vertebrae [33]. The frequency of this tail defect rises to 17.5% in Sp5LacZ/+;Wnt3a+/− mice and 41.5% in Sp5LacZ/LacZ;Wnt3a+/− animals demonstrating an Sp5 gene dosage-dependent genetic interaction with Wnt3a during tail development (Fig. 4E–F). Similar genetic interactions have been reported for Sp5 and the Wnt3a target gene T [11], [17]. Furthermore, Sp5LacZ/LacZ;Wnt3a−/− double mutants display a loss of caudal somites and tailbud that is slightly more severe than Wnt3a−/− mutants alone (Fig. 4A–D). These results support a role for Sp5 in segmentation and the maintenance of tail progenitors and suggest that Sp5 functions in a genetic network with Wnt3a and T.

thumbnail
Figure 4. Genetic interactions between Sp5 and Wnt3a during tail development.

(A–D) Two-color WISH illustrating Uncx4.1 (purple) and T (orange) in Sp5+/+;Wnt3a+/− (A), Sp5lacz/lacz;Wnt3a+/− (B), Sp5+/+;Wnt3a−/− (C), and Sp5lacz/lacz;Wnt3a−/− double mutants (D) at E9.5. Double mutants (n = 8) display fewer and less-defined Uncx4.1+ anterior somites as compared to stage-matched Wnt3a null embryos. (E) At E18.5, Sp5lacz/+;Wnt3a+/− animals (bottom) display abnormal, fused tail vertebrae (arrow) compared to Sp5lacz/+;Wnt3a+/− mice (top). (F) Frequency of tail kinks (expressed as %) observed in Wnt3a+/− heterozygotes at 6 weeks of age increased dramatically as the Sp5 gene dosage was reduced. The number of animals examined (n) is indicated below each genotype. *, statistical significance (p<0.0001, Chi-squared t-test). Scale bar = 500 um (E).

https://doi.org/10.1371/journal.pone.0087018.g004

Sp8 null embryos are tailless and display spina bifida, reflecting a failure to form caudal somites and maintain a tailbud, and an eversion of the posterior neural folds, respectively [14], [34]. Interestingly, the Sp8Δ/Δ axis truncation phenotype bears a striking resemblance to the tail and spina bifida phenotypes observed in the Wnt3a hypomorph, vestigial tail (vt) [33]. The similarities between the hypomorphic Wnt3a and Sp8 mutant phenotypes, together with the demonstration that Wnt3a and Sp5 function in the same genetic pathway and that Sp5 and Sp8 are co-expressed in the PS, strongly suggests that Sp5 and Sp8 could function redundantly in the Wnt3a pathway to regulate NM stem cells. To test this hypothesis, we generated Sp5LacZ/LacZ;Sp8Δ/Δ double knockout (hereafter referred to as Sp5;Sp8 dko) embryos. At E9.5, Sp5LacZ/LacZ and Sp8Δ/Δ single mutants appear similar to wild type controls (Fig. 5A–C). Although Sp5LacZ/LacZ;Sp8Δ/+ mice are viable and fertile, 33.3% of adults develop “kinked” and/or shortened tails (Fig. S3). In contrast, tail defects were observed in only ∼2.2% of Sp8Δ/+ mice, suggesting a requirement for both Sp5 and Sp8 in tail segmentation and extension. Analysis of Sp5;Sp8 dko revealed embryonic lethality by ∼E13.5–14.5. Remarkably, Sp5;Sp8 dko embryos displayed a severe posterior mesoderm deficit at E9.5 (Fig. 5D) that closely resembled the phenotype observed in Wnt3a−/− mutants (Fig. 5E). Although Sp5;Sp8 dko formed ∼12 somites, compared to the 7–10 somites that typically develop in Wnt3a−/− mutants, the T+ NM stem cell population is completely depleted in Sp5;Sp8 dko by E9.5, just like in Wnt3a−/− mutants (Fig. 5F–H compared to Fig. 4C). It is notable that the open anterior neuropore that was present in all Sp8 nulls (n = 28, Fig. 5C arrowhead) was exacerbated in 17/31 double mutants (arrow, Fig. 5D). These results show that Sp5 and Sp8 are essential for NM cell development and that they may have additional redundant roles in anterior neural development.

thumbnail
Figure 5. Sp5;Sp8 double mutants phenocopy single Wnt3a−/− mutants.

Brightfield images depicting wild type (A), Sp5LacZ/LacZ (B) Sp8Δ/Δ, (C), Sp5LacZ/LacZ;Sp8Δ/Δ (D), and Wnt3a−/− (E) mutant phenotypes at E9.5. Arrows in (C) and (D) indicates the open anterior neuropore. Arrowheads in (D) and (E) indicate position of last formed somite. (F–H) Two-color WISH analysis of somite polarity (Uncx4.1) and tailbud progenitor (T) markers in wt and mutant embryos at E9.5. F) Normal T expression in the PS and posterior PSM in control embryos. T was severely down regulated in Sp8Δ/Δ embryos (G), and completely absent from Sp5LacZ/LacZ;Sp8Δ/Δ mutants (H). (I–T) Immunofluorescence analysis of Sox2 and Fibronectin protein expression in wild type (I–L), Wnt3a−/− (M–P), and Sp5LacZ/LacZ;Sp8Δ/Δ (Q–T) mutants. Note the absence of somites and the duplicated neural tubes (arrows) in the Wnt3a−/− and Sp5LacZ/LacZ;Sp8Δ/Δ mutants. Nuclei were labeled with DAPI. Approximate positions of sections are indicated by lines in (A), (D) and (E). Somites (s).

https://doi.org/10.1371/journal.pone.0087018.g005

In addition to being required for the maintenance of NM stem cells, Wnt3a plays a critical role in directing NM stem cells towards a paraxial mesoderm cell fate. In the absence of Wnt3a activity, presumptive paraxial mesoderm cells are thought to adopt neural fates and form ectopic neural tubes posterior to the last formed somite [10], [12]. To determine if Sp5;Sp8 dko mutants also display ectopic neural tissue, we analyzed cross sections of mutant embryos for the expression of the neural marker Sox2. Indeed, a supernumerary Sox2+ neural tube lying ventral to the primary neural tube was observed in Sp5;Sp8 dko similar to Wnt3a−/−mutants (Fig. 5M–P, Q–T). Fibronectin staining was used to highlight embryonic structure and clearly shows that embryos that display ectopic neural tubes also lack somites (arrowheads, Fig. 5K, O, S). Thus the severe posterior abnormalities resulting from changes in neural and mesodermal fates are nearly identical in both Sp5;Sp8 and Wnt3a mutants. These data show that like Wnt3a, Sp5 and Sp8 play a major role in determining NM stem cell fate.

Discussion

We have characterized the Wnt3a transcriptome in the node and primitive streak (PS) at a developmental stage when the mammalian embryo is undergoing gastrulation, segmentation and axis extension. Our genomic data strongly supports a role for Wnt3a as a major regulator of PS stem cells, including NM stem cells. Many PS-specific genes identified in the microarray were down-regulated implicating Wnt3a as an important regulator of PS fate. The most significant sets of differentially expressed genes were associated with neural or paraxial mesoderm cell identity lending strong support to our hypothesis that Wnt3a directly regulates NM progenitors. Interestingly, neural-specific gene expression was marked by increased anterior and decreased posterior neural gene expression. In parallel, posterior paraxial PSM markers were also down-regulated while the anterior PSM markers, Ripply2 and Mesp2 increased. These findings suggest that Wnt3a suppresses anterior, and activates posterior, gene expression in both the neural plate and the PSM, consistent with a proposed role for Wnt/β-catenin signaling as the major evolutionarily conserved posteriorizing signal during the establishment and maintenance of axial AP polarity [35].

Wnt/β-catenin signaling plays a well-established role in the regulation of embryonic and adult stem cell homeostasis [36]. Somewhat paradoxically, Wnt/β-catenin signaling can promote both the maintenance of pluripotent ESCs as well as their differentiation into germ layer progenitors [32], [36]. Although our studies did not focus on pluripotency, Klf5, which encodes a transcription factor redundantly required for ESC pluripotency [37], was down-regulated in Wnt3a mutants suggesting that Klf5 might regulate PS stem cell potency. This screen also led to the identification of the bHLH transcription factor, Msgn1, as a direct target gene of Wnt3a [19]. Msgn1 is a major activator of the oscillating segmentation clock - an activity that is a defining feature of PSM cells - suggesting that Msgn1 may be the key regulator of PSM differentiation.

Sp5 and Sp8 are expressed in numerous tissues throughout mouse development, [17], [38] but are restricted to important signaling centers where Wnts are active and where stem cells reside, such as the primitive streak. The orthologous genes Sp5l and Sp5 have been implicated as important regulators of posterior development in zebrafish [39], [40], but a role for mammalian Sp5 has not been clearly established. Although Sp5 is not required for mammalian embryonic development, an Sp5 null allele enhanced the short or kinked tail phenotypes in T [17] and Wnt3a heterozygotes suggesting it may indeed function in NM progenitors. The case for a role for Sp8 in NM progenitors is stronger given that genetic ablation of Sp8 resulted in the loss of all caudal (tail) somites and led to the development of neural anomalies [10], [14]. We have now shown, through the generation of Sp5;Sp8 double mutants, that Sp5 and Sp8 have essential, overlapping roles in trunk and tail development. With the exception of the anterior exencephaly defects, the Sp5;Sp8 dko phenotype is nearly identical to the Wnt3a−/− phenotype. Double mutants lack posterior paraxial mesoderm and display ectopic neural tubes, strongly suggesting that Sp5 and Sp8 function to regulate the fate of NM stem cells.

We have shown that Wnt3a/β-catenin signaling is both necessary and sufficient for Sp5 and Sp8 expression in the PS, PSM, and in ESCs. The high degree of similarity between Wnt3a, conditional β-catenin, Tcf1;Lef1, and Sp5;Sp8 mutants suggests that Sp5/8 play equally important roles as β-catenin and Tcf1/Lef1 to transduce Wnt3a signals. This raises the intriguing question as to whether Sp5/8 function as terminal effectors of a Wnt3a-initiated transcriptional cascade, or as novel, integral components of the β-catenin/Lef1 transcription complex. Alternatively, Sp5/Sp8 could function in a positive feedback loop to maintain Wnt3a expression. Future studies will address the precise molecular mechanisms of Sp5 and Sp8 activity in stem cells of the early embryo.

Supporting Information

Figure S1.

Heat Map of all 729 DEGs between wt and Wnt3a−/− mutants (p<0.05). Hierarchical clustering of all DEGs illustrates the consistency between replicates.

https://doi.org/10.1371/journal.pone.0087018.s001

(TIF)

Figure S2.

Wnt/wg gene regulatory network. Signal transduction pathway analysis (Genomatix, GePS software) identified 65 of the DEGs as members of the Wnt/wg gene regulatory network. Levels of differential gene expression are reflected by color and intensity: Green: down-regulated genes; Red: up-regulated genes Dashed lines highlight connections between network genes; diamonds indicate characterized and predicted promoter binding sites for connected factors; solid lines indicate interaction between proteins.

https://doi.org/10.1371/journal.pone.0087018.s002

(TIF)

Figure S3.

Genetic interaction between Sp5 and Sp8.

https://doi.org/10.1371/journal.pone.0087018.s003

(TIF)

Table S1.

List of differentially expressed genes (DEG) determined by comparing transcriptional profiles of Wnt3a−/− and wildtype PS/node.

https://doi.org/10.1371/journal.pone.0087018.s004

(XLSX)

Table S2.

Signal transduction pathway analysis of DEG.

https://doi.org/10.1371/journal.pone.0087018.s005

(XLS)

Table S3.

Tissue-specific expression patterns associated with DEG.

https://doi.org/10.1371/journal.pone.0087018.s006

(XLS)

Table S4.

qPCR primers used to analyze Sp1-like family expression.

https://doi.org/10.1371/journal.pone.0087018.s007

(DOCX)

Acknowledgments

We are particularly indebted to Jaime Greear and Ruth Wolfe (SAIC-Frederick) for excellent animal husbandry, Kristin Biris for technical assistance, and Rob Garriock for critiquing the manuscript. We thank David Wilkinson and Sally Dunwoodie for kindly providing us with the Sp5lacz/LacZ mouse line, and Gordon Keller (OCI, Canada) for the gift of T-GFP ESCs [20]. We also thank Jennifer Matta and Bobbi Smith (PHL, SAIC-Frederick), and the Laboratory of Molecular Technology (LMT, FNL CR) for technical assistance.

Author Contributions

Conceived and designed the experiments: WCD MWLK TPY. Performed the experiments: WCD MWLK. Analyzed the data: WCD MWLK RBC TPY. Contributed reagents/materials/analysis tools: KC. Wrote the paper: MWLK TPY.

References

  1. 1. Slack JM (2008) Origin of stem cells in organogenesis. Science 322: 1498–1501.
  2. 2. Cambray N, Wilson V (2002) Axial progenitors with extensive potency are localised to the mouse chordoneural hinge. Development 129: 4855–4866.
  3. 3. Cambray N, Wilson V (2007) Two distinct sources for a population of maturing axial progenitors. Development 134: 2829–2840.
  4. 4. Martin BL, Kimelman D (2012) Canonical Wnt signaling dynamically controls multiple stem cell fate decisions during vertebrate body formation. Dev Cell 22: 223–232.
  5. 5. McGrew MJ, Sherman A, Lillico SG, Ellard FM, Radcliffe PA, et al. (2008) Localised axial progenitor cell populations in the avian tail bud are not committed to a posterior Hox identity. Development 135: 2289–2299.
  6. 6. Tzouanacou E, Wegener A, Wymeersch FJ, Wilson V, Nicolas JF (2009) Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis. Dev Cell 17: 365–376.
  7. 7. Saito-Diaz K, Chen TW, Wang X, Thorne CA, Wallace HA, et al. (2013) The way Wnt works: components and mechanism. Growth Factors 31: 1–31.
  8. 8. Martin BL, Kimelman D (2008) Regulation of canonical Wnt signaling by Brachyury is essential for posterior mesoderm formation. Dev Cell 15: 121–133.
  9. 9. Martin BL, Kimelman D (2010) Brachyury establishes the embryonic mesodermal progenitor niche. Genes Dev 24: 2778–2783.
  10. 10. Takada S, Stark KL, Shea MJ, Vassileva G, McMahon JA, et al. (1994) Wnt-3a regulates somite and tailbud formation in the mouse embryo. Genes Dev 8: 174–189.
  11. 11. Yamaguchi TP, Takada S, Yoshikawa Y, Wu N, McMahon AP (1999) T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification. Genes Dev 13: 3185–3190.
  12. 12. Yoshikawa Y, Fujimori T, McMahon AP, Takada S (1997) Evidence that absence of Wnt-3a signaling promotes neuralization instead of paraxial mesoderm development in the mouse. Dev Biol 183: 234–242.
  13. 13. Galceran J, Farinas I, Depew MJ, Clevers H, Grosschedl R (1999) Wnt3a−/−like phenotype and limb deficiency in Lef1(−/−)Tcf1(−/−) mice. Genes Dev 13: 709–717.
  14. 14. Bell SM, Schreiner CM, Waclaw RR, Campbell K, Potter SS, et al. (2003) Sp8 is crucial for limb outgrowth and neuropore closure. Proc Natl Acad Sci U S A 100: 12195–12200.
  15. 15. Dunty WC Jr, Biris KK, Chalamalasetty RB, Taketo MM, Lewandoski M, et al. (2008) Wnt3a/beta-catenin signaling controls posterior body development by coordinating mesoderm formation and segmentation. Development 135: 85–94.
  16. 16. Harada N, Tamai Y, Ishikawa T, Sauer B, Takaku K, et al. (1999) Intestinal polyposis in mice with a dominant stable mutation of the beta-catenin gene. EMBO J 18: 5931–5942.
  17. 17. Harrison SM, Houzelstein D, Dunwoodie SL, Beddington RS (2000) Sp5, a new member of the Sp1 family, is dynamically expressed during development and genetically interacts with Brachyury. Dev Biol 227: 358–372.
  18. 18. Perantoni AO, Timofeeva O, Naillat F, Richman C, Pajni-Underwood S, et al. (2005) Inactivation of FGF8 in early mesoderm reveals an essential role in kidney development. Development 132: 3859–3871.
  19. 19. Chalamalasetty RB, Dunty WC Jr, Biris KK, Ajima R, Iacovino M, et al. (2011) The Wnt3a/beta-catenin target gene Mesogenin1 controls the segmentation clock by activating a Notch signalling program. Nat Commun 2: 390.
  20. 20. Gadue P, Huber TL, Paddison PJ, Keller GM (2006) Wnt and TGF-beta signaling are required for the induction of an in vitro model of primitive streak formation using embryonic stem cells. Proc Natl Acad Sci U S A 103: 16806–16811.
  21. 21. Hovanes K, Li TW, Munguia JE, Truong T, Milovanovic T, et al. (2001) Beta-catenin-sensitive isoforms of lymphoid enhancer factor-1 are selectively expressed in colon cancer. Nat Genet 28: 53–57.
  22. 22. Jho EH, Zhang T, Domon C, Joo CK, Freund JN, et al. (2002) Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol 22: 1172–1183.
  23. 23. Wharton KA Jr, Zimmermann G, Rousset R, Scott MP (2001) Vertebrate proteins related to Drosophila Naked Cuticle bind Dishevelled and antagonize Wnt signaling. Dev Biol 234: 93–106.
  24. 24. Chapman DL, Papaioannou VE (1998) Three neural tubes in mouse embryos with mutations in the T-box gene Tbx6. Nature 391: 695–697.
  25. 25. Yoon JK, Wold B (2000) The bHLH regulator pMesogenin1 is required for maturation and segmentation of paraxial mesoderm. Genes Dev 14: 3204–3214.
  26. 26. Biris KK, Dunty WC Jr, Yamaguchi TP (2007) Mouse Ripply2 is downstream of Wnt3a and is dynamically expressed during somitogenesis. Dev Dyn 236: 3167–3172.
  27. 27. McGrew LL, Lai CJ, Moon RT (1995) Specification of the anteroposterior neural axis through synergistic interaction of the Wnt signaling cascade with noggin and follistatin. Dev Biol 172: 337–342.
  28. 28. Satoh K, Kasai M, Ishidao T, Tago K, Ohwada S, et al. (2004) Anteriorization of neural fate by inhibitor of beta-catenin and T cell factor (ICAT), a negative regulator of Wnt signaling. Proc Natl Acad Sci U S A 101: 8017–8021.
  29. 29. Sahara S, Kawakami Y, Izpisua Belmonte JC, O’Leary DD (2007) Sp8 exhibits reciprocal induction with Fgf8 but has an opposing effect on anterior-posterior cortical area patterning. Neural Dev 2: 10.
  30. 30. Fujimura N, Vacik T, Machon O, Vlcek C, Scalabrin S, et al. (2007) Wnt-mediated down-regulation of Sp1 target genes by a transcriptional repressor Sp5. J Biol Chem 282: 1225–1237.
  31. 31. ten Berge D, Koole W, Fuerer C, Fish M, Eroglu E, et al. (2008) Wnt signaling mediates self-organization and axis formation in embryoid bodies. Cell Stem Cell 3: 508–518.
  32. 32. ten Berge D, Kurek D, Blauwkamp T, Koole W, Maas A, et al. (2011) Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat Cell Biol 13: 1070–1075.
  33. 33. Greco TL, Takada S, Newhouse MM, McMahon JA, McMahon AP, et al. (1996) Analysis of the vestigial tail mutation demonstrates that Wnt-3a gene dosage regulates mouse axial development. Genes Dev 10: 313–324.
  34. 34. Treichel D, Schock F, Jackle H, Gruss P, Mansouri A (2003) mBtd is required to maintain signaling during murine limb development. Genes Dev 17: 2630–2635.
  35. 35. Petersen CP, Reddien PW (2009) Wnt signaling and the polarity of the primary body axis. Cell 139: 1056–1068.
  36. 36. Merrill BJ (2012) Wnt pathway regulation of embryonic stem cell self-renewal. Cold Spring Harb Perspect Biol 4: a007971.
  37. 37. Ema M, Mori D, Niwa H, Hasegawa Y, Yamanaka Y, et al. (2008) Kruppel-like factor 5 is essential for blastocyst development and the normal self-renewal of mouse ESCs. Cell Stem Cell 3: 555–567.
  38. 38. Treichel D, Becker MB, Gruss P (2001) The novel transcription factor gene Sp5 exhibits a dynamic and highly restricted expression pattern during mouse embryogenesis. Mech Dev 101: 175–179.
  39. 39. Thorpe CJ, Weidinger G, Moon RT (2005) Wnt/beta-catenin regulation of the Sp1-related transcription factor sp5l promotes tail development in zebrafish. Development 132: 1763–1772.
  40. 40. Weidinger G, Thorpe CJ, Wuennenberg-Stapleton K, Ngai J, Moon RT (2005) The Sp1-related transcription factors sp5 and sp5-like act downstream of Wnt/beta-catenin signaling in mesoderm and neuroectoderm patterning. Curr Biol 15: 489–500.