29.04.2013 Views

2007-DevelopmentalBiology.pdf

2007-DevelopmentalBiology.pdf

2007-DevelopmentalBiology.pdf

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Abstract<br />

Genomes & Developmental Control<br />

Wnt/β-catenin signaling controls Mespo expression to regulate<br />

segmentation during Xenopus somitogenesis<br />

Jinhu Wang 1 , Shangwei Li 1 , Yuelei Chen, Xiaoyan Ding ⁎<br />

Laboratory of Molecular Cell Biology, Key Laboratory of Stem Cell Biology, Institute of Biochemistry and Cell Biology,<br />

Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China<br />

Graduate School of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China<br />

Received for publication 12 May 2006; revised 17 November 2006; accepted 14 December 2006<br />

Available online 21 December 2006<br />

The vertebral column is derived from somites, which are transient segments of the paraxial mesoderm that are present in developing vertebrates.<br />

The strict spatial and temporal regulation of somitogenesis is of crucial developmental importance. Signals such as Wnt and FGF play roles in<br />

somitogenesis, but details regarding how Wnt signaling functions in this process remain unclear. In this study, we report that Wnt/β-catenin<br />

signaling regulates the expression of Mespo, a basic–helix–loop–helix (bHLH) gene critical for segmental patterning in Xenopus somitogenesis.<br />

Transgenic analysis of the Mespo promoter identifies Mespo as a direct downstream target of Wnt/β-catenin signaling pathway. We also<br />

demonstrate that activity of Wnt/β-catenin signaling in somitogenesis can be enhanced by the PI3-K/AKT pathway. Our results illustrate that Wnt/<br />

β-catenin signaling in conjunction with PI3-K/AKT pathway plays a key role in controlling development of the paraxial mesoderm.<br />

© <strong>2007</strong> Published by Elsevier Inc.<br />

Keywords: AKT; Paraxial mesoderm; PI3-K; Wnt; Xenopus laevis<br />

Introduction<br />

During vertebrate embryogenesis, the paraxial mesoderm<br />

separates into somites, a metameric series of homologous<br />

subunits, which bud off sequentially from the rostral end of the<br />

presomitic mesoderm (PSM) at regular intervals along the<br />

anterior–posterior (AP) axis. The process in which the<br />

segmental or metameric pattern is established accompanying<br />

the constant posterior elongation of the embryo body axis is<br />

termed somitogenesis (Weisblat et al., 1994). Somitogenesis<br />

occurs in a strict spatial and temporal progression (Dubrulle<br />

et al., 2001; Pourquie, 2003).<br />

The molecular nature of the signals responsible for<br />

controlling somitogenesis remains elusive. One potential<br />

candidate for regulating this process is Wnt/β-catenin signaling<br />

(Aulehla et al., 2003; Yamaguchi et al., 1999). Previous studies<br />

have shown that Wnt/β-catenin signaling is implicated in<br />

⁎ Corresponding author. Fax: +86 21 34230165.<br />

E-mail address: xyding@sunm.shcnc.ac.cn (X. Ding).<br />

1 These authors contributed equally to this work.<br />

0012-1606/$ - see front matter © <strong>2007</strong> Published by Elsevier Inc.<br />

doi:10.1016/j.ydbio.2006.12.034<br />

Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

www.elsevier.com/locate/ydbio<br />

controlling the development of paraxial mesoderm (Aulehla<br />

et al., 2003; Galceran et al., 2004; Hofmann et al., 2004;<br />

Pourquie, 2001). In mouse embryos, Wnt3a is expressed in the<br />

primitive streak during gastrulation and in the tailbud during the<br />

later stages of mouse development. Animals homozygous for<br />

null alleles of Wnt3a lack somites posterior to the forelimbs<br />

(Ikeya and Takada, 2001; Takada et al., 1994; Wilson et al.,<br />

1993). The double mutant harboring mutations in lef1 and tcf1,<br />

identified as nuclear mediators of Wnt/β-catenin signaling that<br />

activate Wnt-responsive genes by associating with β-catenin,<br />

results in lack of paraxial mesoderm resembling the Wnt3a null<br />

mutation (Galceran et al., 1999). In chick embryos, it has been<br />

shown that altering Wnt/β-catenin signaling activity in the PSM<br />

causes defects in somite formation (Aulehla et al., 2003).<br />

However, the exact molecular nature of Wnt/β-catenin signaling<br />

in somitogenesis is not fully understood, and whether Wnt/<br />

β-catenin signaling is involved in somitogenesis of the lower<br />

vertebrates such as Xenopus is still questionable.<br />

The FGF signaling pathway also participates in somitogenesis<br />

(Delfini et al., 2005; Dubrulle et al., 2001; Moreno and Kintner,<br />

2004). Fgf8 mRNA forms a gradient along the PSM in mouse


embryos (Delfini et al., 2005; Dubrulle et al., 2001). Altering the<br />

activity of FGF signaling in the PSM resulted in segmental defects<br />

(Dubrulle et al., 2001). These data demonstrate that FGF signaling<br />

is required for somitogenesis. It has been proposed that FGF<br />

signaling may act through the PI3-K/AKT pathway in the PSM to<br />

participate in somitogenesis (Dubrulle and Pourquie, 2004). Since<br />

PI3-K/AKT signaling can enhance accumulation of β-catenin by<br />

inhibiting GSK-3β during cardiomyocyte growth (Haq et al.,<br />

2003), it may regulate Wnt/β-catenin activity in the segmentation<br />

process by a similar mechanism.<br />

Somite formation and patterning are thought to be controlled<br />

by Mespo, a member of the basic–helix–loop–helix (bHLH)<br />

transcription factor family (Moreno and Kintner, 2004; Yoon<br />

et al., 2000; Yoon and Wold, 2000). Mespo expression<br />

encompasses the entire caudal PSM of Xenopus embryos, and<br />

its expression is reduced when the PSM cells enter the<br />

segmentation process (Joseph and Cassetta, 1999; Kim et al.,<br />

2000; Moreno and Kintner, 2004). Gain of function experiments<br />

showed that Mespo can induce expression of paraxial mesoderm<br />

marker genes (Yoon et al., 2000). Mespo null mouse embryos<br />

exhibited no identifiable somites or segmental patterning in the<br />

trunk posterior to forelimbs, and severe disruption of gene<br />

expressions in posterior paraxial mesoderm (Yoon and Wold,<br />

2000). These results suggest that Mespo plays essential roles in<br />

somitogenesis. The phenotypic resemblance between Mespo<br />

and Wnt3a mutant mouse embryos provides a strong hint that<br />

Wnt/β-catenin signaling regulates the expression of Mespo<br />

during paraxial mesoderm development. However, whether<br />

Mespo expression is regulated by Wnt/β-catenin signaling in the<br />

PSM remains unclear.<br />

Here, we report that Wnt/β-catenin signaling directly<br />

regulates the expression of Mespo, and this regulation is<br />

enhanced by PI3-K/AKT signaling. These results demonstrate<br />

that Wnt/β-catenin signaling plays a key role in controlling<br />

somitogenesis by regulating Mespo gene expression, and that<br />

PI3-K/AKT signaling is also involved in this process.<br />

Materials and methods<br />

Embryo manipulations, drug treatment<br />

Preparation and injection of Xenopus embryos were carried out as previously<br />

described (Ding et al., 1998). Embryos were staged according to Nieuwkoop<br />

and Faber (1967). LY294002 (Calbiochem) was used at 100 μM. Chemicals<br />

were dissolved in DMSO and then diluted into 0.1× MMR. In each case, about<br />

20 embryos were examined per condition per probe. Each experiment was<br />

repeated three times independently.<br />

Morpholino oligomer and in vitro translation<br />

The 25-bp morpholino antisense oligomer for Xenopus Mespo was obtained<br />

from Gene Tools and consisted of the following sequence:<br />

MespoMO: GGGATGGTGCAGAGTCTCCATCAGT<br />

Morpholino was dissolved in water to a concentration of 1 mM, which was<br />

then further diluted to a give a working solution of 0.3 mM. For the rescue assay,<br />

we generated a mutated Mespo cDNA (mMespo) differed in seven bases<br />

(underlined) from Mespo: 5′-CTACTATGGAGACTCTGCACCATCCCCT.<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

The in vitro transcription/translation of Mespo was performed according to<br />

the protocol of TNT coupled reticulocyte lysate system (Promega, L4610).<br />

In situ hybridization, immunocytochemistry<br />

Whole-mount in situ hybridization was performed as previously described<br />

(Harland, 1991) using DIG-labeled probes. For bleaching of wild-type embryos,<br />

hybridized embryos were treated with bleaching solution (0.5× SSC with 1%<br />

hydrogen peroxide and 5% formamide) under a fluorescent light. The following<br />

plasmid templates were linearized, and DIG-labeled antisense RNA probes were<br />

made using either T7 or SP6 RNA polymerase: Thylacine1 (Sparrow et al.,<br />

1998); Mespo (Joseph and Cassetta, 1999); GFP (Geng et al., 2003); Xwnt8<br />

(Kazanskaya et al., 2004) and Dkk1 (Glinka et al., 1998); Paraxis (a PCRamplified<br />

full-length cDNA of Paraxis was cloned into pCS2+); AKT (a PCRamplified<br />

coding region of Xenopus AKT was cloned into pCS2+); PAPC (Kim<br />

et al., 2000); XBra (Smith et al., 1991).<br />

To visualize somite size, embryos were fixed in 2% paraformaldehyde, 2%<br />

glutaraldehyde in PBS, and stained with the muscle-specific monoclonal antibody<br />

12/101 (Kintner and Brockes, 1985). Then embryos were post-fixed in MEMFA<br />

(0.1 M MOPS pH 7.4, 2 mM EGTA, 1 mM MgSO4, 3.7% formaldehyde),<br />

embedded in paraffin, sectioned at 10 μm, and stained with DAPI.<br />

Mutagenesis and transgenesis<br />

The Exsite PCR-based site-directed mutagenesis system (Stratagene, cat.<br />

200502-5) was used to generate the mutants of site L, p-4317mLuc and p-511m.<br />

The primers are shown below (nucleotides altered shown in bold): 5′-<br />

AGGAGACAGCAAGGTGTTAACATG-3′ (reverse) and 5′-CTAGACTCC-<br />

TCCATTAAAACGCCCACT-3′ (forward).<br />

Transgenic Xenopus embryos were generated as described previously (Kroll<br />

and Amaya, 1996). Plasmids used for transgenesis assays were linearized by<br />

NotI digestion. Three independent rounds of transgenesis were performed for<br />

each plasmid.<br />

Western blot<br />

For measuring phospho-Akt levels, the caudal region of Xenopus embryos at<br />

stage 23 were divided into three equal parts corresponding to the caudal domain,<br />

the rostral domain of the PSM and a region including the last formed somites. In<br />

these experiments, 100 embryos were used. The samples were lysed in EDTAfree<br />

RIPA (0.1% NP40, 20 mM Tris–HCl pH 8, 10% glycerol) with cocktail<br />

protease inhibitors (Bio Basics) and Na 3VO 4.<br />

For measuring accumulation of β-catenin in the nucleus and cytosol, the<br />

caudal domain of the PSM was dissected and cultured in 1× MMR containing<br />

LY294002 at 100 μM or DMSO for 2 h. Then β-catenin in either nucleus or<br />

cytosol was extracted according to PIERCE protocol (NE-PER nuclear and<br />

cytoplasmic extraction reagents, 78833).<br />

All the samples were quantitated by Bradford assay and adjusted to equal<br />

concentration before loading. Western blot was performed as described (He<br />

et al., 2005). Polyclonal antibody P14L was used to visualize β-catenin (dilution<br />

1:2000), and anti-phospho-Akt polyclonal antibodies (dilution 1:2000; Cell<br />

Signaling Technology) were used to detect endogenous Akt and phospho-Akt.<br />

For a loading control, equal volume of proteins were separated by SDS–PAGE<br />

and visualized with Coomassie Blue.<br />

Chromatin immunoprecipitation (ChIP)<br />

Chromatin immunoprecipitation (ChIP) assay was performed, with modifications,<br />

according to the method described previously (Sachs and Shi, 2000).<br />

100 Xenopus embryos were selected at stage 13 from embryos injected with<br />

appropriate plasmids, and crosslinked with 1% formaldehyde at room<br />

temperature for 20 min. Following treatment with 0.125 M glycine, embryos<br />

were washed and resuspended in 200 μl ChIP lysis buffer. The lysates were<br />

sonicated on ice to obtain DNA fragments on an average around 500 bp and<br />

immunoprecipitated with anti-HA monoclonal antibody (dilution 1:3000;<br />

Sigma). DNA fragments were washed, eluted and recovered from the immunoprecipitated<br />

complex. PCR was performed under an optimized condition: 94 °C<br />

837


838 J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

for 30 s, 58 °C for 30 s, and 72 °C for 30 s, for 30–35 cycles. Primer sequences are<br />

as follows: Mespo promoter upstream 5′-TGAAAGCCAATTTAGCTGAAGG-<br />

3′, downstream 5′-GCTGGCAAAGTTCCATGTGAG-3′; ODC upstream 5′-<br />

GATCATGCACATGTCAAGCC-3′, downstream 5′-CAGGGAGAATGC-<br />

CATGTTCT-3′; pGL3-Basic upstream 5′-CAGGGGATAACGCAGGAAA-3′,<br />

downstream 5′-AAGGGAGAAAGGCGGACA G-3′.<br />

Results<br />

MO-based Mespo depletion caused defects of somite formation<br />

Previous analysis of the morphological defects in Mespo<br />

knockout mouse suggests that Mespo plays a role in somitogenesis<br />

(Yoon and Wold, 2000). In Xenopus, Mespo<br />

expression is firstly detected in ventrolateral mesoderm but is<br />

absent from dorsal mesoderm in gastrulae, and is localized to<br />

the most caudal (unsegmented) paraxial mesoderm in neurulae<br />

and tailbud stage (Joseph and Cassetta, 1999; Yoon et al.,<br />

2000). To test whether Mespo contributes to Xenopus<br />

somitogenesis, we used antisense morpholino oligonucleotides<br />

(MO) to deplete Mespo. We designed a Mespo MO that spans<br />

the ATG initiation codon (Fig. 1A). This MO inhibits<br />

translation of recombinant Mespo RNA in an in vitro<br />

transcription/translation reaction (Fig. 1B). To disrupt endogenous<br />

Mespo expression we injected Mespo MO into the<br />

ventral marginal zone (VMZ) of one blastomere at four-cell<br />

stage, with the uninjected side serving as a control. The somite<br />

morphology of the injected embryos was then examined at<br />

tadpole stage by whole-mount immunostaining with 12/101 (a<br />

monoclonal antibody specifically recognizes differentiated<br />

muscle cells). While control MO-injected embryos exhibit no<br />

morphological defects in somite formation (Fig. 1C), embryos<br />

injected with Mespo MO show remarkable malformation of<br />

Fig. 1. Antisense MO against Mespo depletes Mespo protein and causes somite formation defects. (A) Sequence of Mespo MO aligned with the 5′-untranslated region<br />

(UTR) of Mespo RNA. (B) In vitro transcription/translation of Mespo is inhibited by Mespo MO. Arrow indicates the band corresponding to Mespo protein. (C, D)<br />

Whole mount immunostaining with 12/101, a muscle-specific antibody, shows that the segmental pattern of somites in Mespo MO-injected embryos (D) is disrupted<br />

and the somite size is irregular (compared with control MO-injected embryo in panel C). Bracket in panel D marks the malformed somites. (E–H) Embryos injected<br />

with Mespo MO (F, H) or control MO (E, G) stained with 12/101, then sectioned longitudinally and labeled with DAPI. Panels E, G and Panels F, H are the same view<br />

of one embryo, respectively. Embryos injected with Mespo MO panel F, H show the disorganization of segmental pattern, based on myotomal morphology (panel F,<br />

compared with panel E) and the arrangement of somatic nuclei panel H, compared with G). Bracket in panel H marks the malformed somites.


somites. The defects include disrupted somite boundaries and<br />

irregular somite size (79%, N=100, Fig. 1D). The malformation<br />

of somites in Mespo MO-injected embryos is shown more<br />

clearly on frontal sections stained with 12/101 or with DAPI<br />

staining of nuclei. Compared with the uninjected side, somites<br />

in Mespo MO-injected side are poorly defined, with no<br />

metameric shape typical for this tissue. Also, somite nuclei<br />

are abnormally aligned, indicating that the patterning of somite<br />

is severely disrupted (67%, N=30, Figs. 1F, H). As a control,<br />

the somite morphology in control MO-injected embryos<br />

remains identical to that in the uninjected side, with regularly<br />

arranged allied cells (Figs. 1E, G).<br />

Aiming to verify that the phenotype described above is due to<br />

specific depletion of Mespo, we carried out rescue experiments.<br />

We prepared a mutated Mespo (mMespo) cDNA, which codes<br />

for the same polypeptide as the Mespo protein but differs from<br />

Mespo cDNA by seven bases. Injection of the resulting pCS–<br />

mMespo plasmid effectively rescues the phenotypic defects<br />

caused by Mespo MO in 85% (N=100) of the injected embryos<br />

(Supplemental Fig. 1). We also injected Mespo MO into DMZ<br />

of four-cell-stage embryos, where Mespo is not expressed, and<br />

found no effect on embryo development (data not shown).<br />

These results indicate that the effect of Mespo MO is specific.<br />

Taken together, our results indicate that depletion of Mespo<br />

caused the morphological defects of somite patterning.<br />

Depleting Mespo disrupts expression of genes in the PSM<br />

The results described above indicate that lacking of Mespo in<br />

embryos causes segmentation defects. In Xenopus, several<br />

molecules have been demonstrated to play roles in segmentation<br />

process, including PAPC (Kim et al., 2000), Thylacine1 (Thy)<br />

(Sparrow et al., 1998) and XDelta-1 (Pourquie, 2003). To<br />

evaluate whether depleting Mespo interferes the expression of<br />

segmental marker genes, we examined the expression of genes<br />

such as PAPC, Thy and XDelta-1 in embryos injected with<br />

Mespo MO. Compared with the uninjected side, the expression<br />

of PAPC is lost in Mespo MO-injected side (85%, N=78, Fig.<br />

2B). The loss of PAPC expression in embryos injected with<br />

Mespo MO is specific since it could be rescued by coinjecting<br />

with pCS–mMespo (70%, N=60, Fig. 2C). The segmental<br />

expression of Thy in somitomeres is severely disrupted (87%,<br />

N=65, Fig. 2E), and it was observed that the somitomeric<br />

expression pattern of XDelta-1 is also perturbed (80%, N=81,<br />

Fig. 2G). As a control, injection of a mixture of pCS–LacZ and<br />

control MO resulted in no difference in the expression level and<br />

pattern of PAPC, Thy or XDelta-1 in both injected and<br />

uninjected sides (Figs. 2A, D, F). These results indicate that<br />

depletion of Mespo profoundly changes the expression of<br />

PAPC, Thy and XDelta-1 in the PSM, suggesting that Mespo<br />

acts in the PSM to control segmental patterning. Since XDelta-1<br />

is a component of the Notch/Delta pathway, these results<br />

suggest that Mespo controls segmental patterning by regulating<br />

Notch/Delta signaling (Pourquie, 2003).<br />

To gain further insight into the cause of somite defects in<br />

embryos lacking Mespo function, we examined the expression<br />

of XBra that plays major roles in development of paraxial<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

mesoderm (Conlon and Smith, 1999). We found that in embryos<br />

depleted for Mespo, the expression of XBra is increased in the<br />

Mespo MO-injected side (67%, N=90, Fig. 2I), as compared<br />

to the control side, indicating that XBra expression is negatively<br />

regulated by Mespo, a phenomenon also observed in Mespo<br />

null mutant mouse (Yoon and Wold, 2000). XBra expression is<br />

not changed in control MO-injected embryos (Fig. 2H).<br />

Interestingly, in embryos injected with Mespo MO, Mespo<br />

mRNA is expressed at levels higher than the control side (78%,<br />

N=78, Fig. 2K). Expression of Mespo is not changed in the<br />

control MO-injected embryos (Fig. 2J). This result implies that<br />

Mespo has a negative regulatory effect on its own mRNA<br />

expression in the PSM.<br />

Taken together, we provide evidence here that Mespo is<br />

responsible not only for segmental specific gene expression, but<br />

is also involved in regulating Xbra and Mespo expression. Thus<br />

we conclude that Mespo is essential for somitogenesis.<br />

Wnt/β-catenin signaling in the PSM<br />

Observations from studies on mouse embryos have demonstrated<br />

that Wnt/β-catenin signaling is involved in somitogenesis<br />

(Aulehla et al., 2003). Regional similarity between<br />

expression patterns of Wnts and Mespo in mouse embryos, as<br />

well as the phenotypic similarities of Wnt and Mespo null<br />

mutant mouse (Takada et al., 1994; Yoon and Wold, 2000),<br />

raises a possibility that Wnt signaling regulates Mespo<br />

expression in somitogenesis.<br />

However, the existence of Wnt signaling in Xenopus PSM<br />

has not been documented. We performed whole-mount in situ<br />

hybridization to analyze Wnt gene expression in the PSM. Like<br />

in other vertebrates, Xenopus PSM is subdivided into two<br />

distinct regions featured by expression pattern of the bHLH<br />

family genes (Fig. 3K) (Kim et al., 2000; Pourquie, 2003). In<br />

the caudal domain of the PSM, Mespo is expressed in the entire<br />

region referred as the tailbud domain (TBD) (Figs. 3I, J). Thy<br />

transcripts are restricted in the rostral region of the PSM and<br />

localized to the anterior half of somitomeres (Figs. 3G, H, I, J).<br />

At the neurula stage, XWnt8 is expressed at high level in the<br />

TBD (Figs. 3A, B, E, F, G, H), whereas Wnt antagonist Dkk1 is<br />

uniformly expressed in somitomeres (Figs. 3C, D, E, F). The<br />

expression patterns of these genes indicate that Wnt signals<br />

exist in the TBD and support that Wnt/β-catenin signaling is<br />

involved in Xenopus somitogenesis.<br />

Altering Wnt/β-catenin signaling activity affects expression of<br />

Mespo<br />

To test whether Wnt/β-catenin signaling regulates Mespo<br />

expression, pCS–Lef1–VP16 (a dominant active form of Lef1)<br />

DNA was injected into VMZ of one cell of four-cell-stage<br />

embryos and Mespo expression was examined at stage 19.<br />

Mespo expression in the injected side is increased compared<br />

with the control side (75%, N=70, Fig. 4B). When embryos<br />

were injected with pCS–β-catenin DNA, Mespo expression is<br />

also increased (Supplemental Fig. 2B). In contrast, injection of<br />

pCS–Lef1–EnR (a dominant negative form of Lef1) DNA<br />

839


840 J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Fig. 2. Expression patterns of genes responsible for segmental patterning are altered in embryos lacking Mespo function. For all embryos, anterior is to the left. (A–K)<br />

One VMZ cell of four-cell-stage embryos injected with control MO (A, D, F, H and J), or Mespo MO (B, E, G, I and K) or Mespo MO plus 35 pg pCS–mMespo (C)<br />

and the embryos were fixed at neurula stage for in situ hybridization to detect the PSM marker gene expression: (A–C) PAPC, (D, E) Thy, (F,G)XDelta-1, (H, I)<br />

XBra, (J, K) Mespo. Bracket in panels B and E indicates the disruption of segmental expression of PAPC and Thy, respectively. Bracket in panel C indicates the<br />

rescue of PAPC expression by coinjecting Mespo MO with pCS–mMespo. Arrowhead in panel G indicates the disruption of segmental expression of XDelta-1.<br />

Arrowhead in panel I indicates the increase of XBra expression. Arrowhead in panel K indicates the increase of Mespo expression.<br />

results in the opposite effect where Mespo expression is<br />

markedly reduced (56%, N=65, Fig. 4C). We also observed<br />

that Mespo expression is decreased at stage 10 when injecting<br />

embryos with lef1–EnR mRNA at four-cell stage (Supplemental<br />

Fig. 2D). These changes are tightly associated with the<br />

changing of Wnt/β-catenin signaling activity, because the


Fig. 3. Wnt/β-catenin signaling in the PSM. (A–J) The neurula stage embryos<br />

were stained by whole-mount in situ hybridization for Xwnt8 (A, B, arrowhead),<br />

Dkk1 (C, D, arrow), Xwnt8 and Dkk1 (E, F, arrowhead indicates XWnt8 and<br />

arrow indicates DKK1), Xwnt8 and Thy (G, H, arrowhead indicates XWn8 and<br />

arrow indicates Thy), or Mespo and Thy (I, J, arrowhead indicates Mespo and<br />

arrow indicates Thy). Embryos in panels A, C, E, G and I are oriented with<br />

anterior to the left and shown in lateral views. Embryos in panels B, D, F, H and<br />

J show posterior–dorsal view. (K) Schematic diagram (adapted from Kim et al.,<br />

2000) illustrates gene expression patterns related to somite formation in the PSM<br />

of Xenopus embryos.<br />

expression patterns of Mespo or Paraxis are un-changed in<br />

control embryos or control side (Figs. 4A, D, E, F).<br />

These results indicate that the activity of Wnt/β-catenin<br />

signaling is responsible for Mespo expression in the PSM.<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Mespo is a direct downstream target of Wnt/β-catenin<br />

signaling cascade<br />

To experimentally determine how Wnt/β-catenin signaling<br />

regulates Mespo expression, we identified a 4.3 kb Mespo<br />

promoter that is able to recapitulate the endogenous Mespo<br />

expression from gastrulae to tailbud stage (Wang and Ding,<br />

2006) and used it for further analysis. We first tested whether<br />

pCS–Lef1–VP16 could induce the expression of a luciferase<br />

reporter controlled by the 4.3 kb Mespo promoter (p-4317Luc)<br />

in animal caps. A 10-fold induction of the reporter activity is<br />

observed (Fig. 5B).<br />

To investigate whether Mespo is a direct target of Wnt/<br />

β-catenin signaling, we examined the Mespo promoter for<br />

LEF/TCF binding sites. A perfectly conserved LEF/TCF<br />

binding site (site L) was found (van de Wetering et al.,<br />

1991), at position −73 to −67 relative to the transcription start.<br />

In a search of GenBank database for the 5′ regulatory regions<br />

of homologous Mespo promoters in zebrafish and chick, we<br />

found that this LEF/TCF binding site is conserved in zMespo<br />

(the zebrafish Mespo homolog) and cMespo-1 (the chick Mespo<br />

homolog) (Fig. 5A). The conservation within the<br />

regulatory regions among these vertebrates suggests that this<br />

LEF/TCF binding site plays an important role in the regulation<br />

of Mespo expression. We then generated a mutation in site L<br />

and examined the activity of the mutant promoter in animal<br />

cap assay. The results show that the mutant promoter construct<br />

(p-4317mLuc) has similar activity as the p-4317Luc (Fig. 5C).<br />

However, when coinjected with pCS–Lef1–VP16, the luciferase<br />

activity of the p-4317mLuc plasmid is significantly<br />

reduced as compared to that of the p-4317Luc plasmid (Fig.<br />

5D). Based on the results obtained from animal cap assays, we<br />

injected reporter plasmids into the VMZ of both cells of<br />

embryos at four-cell stage to examine the function of the LEF/<br />

TCF binding site (site L) in vivo. The results show that the<br />

luciferase activity of the p-4317mLuc plasmid is reduced to<br />

half of the p-4317Luc plasmid (Fig. 5E). The functional<br />

importance of site L was further confirmed by generating<br />

transgenic embryos with the 4.3 kb GFP construct carrying the<br />

site L mutation. When site L was mutated, GFP expression in<br />

the PSM is abolished in approximately 90% transgenic<br />

embryos (N=200, Fig. 5H) while the expression of GFP<br />

mRNA in approximately 51% p-4317GFP transgenic embryos<br />

(N=180, Fig. 5G) is detected in the PSM (Fig. 5I), strongly<br />

suggesting that the site L is indeed essential for Mespo<br />

expression in the PSM.<br />

LEF1 binds site L in vivo<br />

To examine whether Lef1 binds to site L, chromatin<br />

immunoprecipitation (ChIP) assay was performed as previously<br />

described (Sachs and Shi, 2000). HA-tagged Lef1 was coinjected<br />

with a truncated form of Mespo promoter, p-511, which<br />

comprised a proximal 511-bp region of the promoter, followed<br />

by precipitation of plasmid DNA bound to Lef1 with an anti-<br />

HA antibody. PCR amplification using specific primers<br />

flanking site L revealed that binding of Lef1 to wild type site<br />

841


842 J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Fig. 4. Wnt/β-catenin signaling regulates Mespo expression. For all embryos, anterior is to the left. Four-cell-stage embryos were injected in VMZ of one blastomere<br />

with lineage tracer pCS–LacZ as control (A, D), or together with pCS–Lef1–VP16 (B, E) or together with pCS–Lef1–EnR (C, F). At stage 19, whole mount in situ<br />

hybridization was performed for Mespo (A–C) or Paraxis (D–F), as indicated. Arrowhead in panel B shows the increase of Mespo expression. Arrowhead in panel C<br />

shows the decrease of Mespo expression.<br />

L is significantly stronger than that to the mutated one (Fig. 6A).<br />

We also examined whether the binding of Lef1 to the Mespo<br />

regulatory region can be detected in vivo. Indeed, when<br />

injecting HA-tagged Lef1 into both VMZ cells at four-cell<br />

stage, the binding of Lef1 to site L is detected (Fig. 6B). As a<br />

control, no binding of Lef1 is detected in the exon region of<br />

Mespo (Fig. 6B).<br />

These results indicate that Mespo is a direct downstream<br />

target of Wnt/β-catenin signaling in the PSM.<br />

PI3-K/AKT signaling regulates Mespo expression<br />

Recent work indicates that FGF signaling is involved in<br />

controlling somitogenesis through MKK1/ERK signaling pathway<br />

by regulating Mespo expression (Delfini et al., 2005;<br />

Moreno and Kintner, 2004). However, FGF signaling has other<br />

downstream effectors like PI3-K/AKT in the PSM (Dubrulle<br />

and Pourquie, 2004). This raises a question of whether PI3-K/<br />

AKT-mediated FGF signaling contributes to the regulation of<br />

Mespo expression.<br />

In order to answer this question, we checked whether Akt is<br />

present in the PSM as the phosphorylated form. Results from<br />

Western blot show that both Akt and its phosphorylated form<br />

are detected in the caudal PSM (Fig. 7B). To evaluate whether<br />

PI3-K/AKT signaling contributes to the expression of Mespo,<br />

we treated embryos with LY294002 which is a specific PI3<br />

kinase inhibitor (Knight et al., 2006; Walker et al., 2000) and<br />

then dissected caudal PSM for analysis. Treating embryos with<br />

LY294002 causes a reduction of Akt phosphorylation (Fig.<br />

7C), and leads to a decreased Mespo expression (67%, N=55,<br />

Fig. 7E) as compared with the control (Fig. 7D). LY294002 or<br />

DMSO treatment causes no change in Paraxis expression<br />

(Figs. 7F, G). The decrease of Mespo expression was further<br />

confirmed by RT–PCR. As shown in Fig. 7H, Mespo mRNA<br />

is dramatically downregulated when the caudal PSM cells<br />

were treated with LY294002 (Fig. 7H). These results indicate<br />

that PI3-K/AKT signaling contributes to Mespo expression.<br />

Interaction of PI3-K/AKT and Wnt signaling on Mespo<br />

expression<br />

Since Wnt/β-catenin signaling directly regulates Mespo<br />

expression and blocking PI3-K/AKT pathway decreases<br />

Mespo expression, we reasoned that Wnt/β-catenin and<br />

PI3-K/AKT pathway might cooperate to regulate Mespo<br />

expression. To test this possibility, we examined whether the<br />

activity of Wnt/β-catenin signaling was affected when PI3-K/<br />

AKT signaling was blocked. A sharp decrease of β-catenin<br />

protein in the nuclei of the caudal PSM cells was detected<br />

after a 2-h treatment with LY294002, and a decrease in<br />

cytosolic β-catenin was also observed (Fig. 8A). To test what<br />

led to the loss of β-catenin, we took advantage of a specific<br />

antibody, which only recognizes the phosphor-form of βcatenin<br />

that is phosphorylated by GSK-3β. Western blot using<br />

this antibody shows that when treating the PSM cells with<br />

LY294002 the phosphorylated form of β-catenin is significantly<br />

increased within 1.5 h, and decreased after 2.5 h (Fig.<br />

8B). This result indicates that the blocking of PI3-K/AKT<br />

pathway causes the phosphorylation and degradation of βcatenin<br />

via an Axin/GSK-3β-dependent mechanism (Aberle<br />

et al., 1997; Polakis, 2001). Therefore, PI3-K/AKT pathway<br />

affects the stability of β-catenin. On the other hand, when<br />

injecting embryos with pCS–Lef1–VP16 DNA to activate<br />

Wnt/β-catenin signaling, the injected side shows increased<br />

Mespo expression (79%, N =70, Fig. 8D). However,<br />

LY294002 treatment could not cause significant reduction of


the increased Mespo expression induced by injecting pCS–<br />

Lef1–VP16, while Mespo expression is clearly reduced in the<br />

uninjected side (61%, N=90, Fig. 8E). Thus injecting pCS–<br />

Lef1–VP16 expressing the constitutively active Lef1–VP16<br />

minimizes the effect of blocking the PI3-K/AKT pathway.<br />

Taken together, these results indicate that the PI3-K/AKT<br />

pathway participates in regulating Wnt/β-catenin signaling<br />

mediated Mespo expression, through the regulation of βcatenin<br />

stability.<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Fig. 5. The LEF/TCF binding site is responsible for the expression of Mespo. (A) Schematic representation of Mespo promoters from various species with a conserved<br />

LEF/TCF binding site (red). The position of potential LEF/TCF binding site (box) in Xenopus Mespo 5′-upstream sequence is indicated. (B–D) Fold induction of<br />

pCS–Lef1–VP16 in pGL3basic and p-4317Luc reporter plasmids (B), luciferase activities of p-4317Luc and p-4317mLuc reporter plasmids (C) or fold induction of<br />

pCS–Lef1–VP16 in p-4317Luc and p-4317mLuc reporter plasmids (D) in animal cap assays. Plasmids were injected into the animal pole at the four-cell stage. Animal<br />

caps were dissected at stage 8.5 and luciferase activities were measured at stage 12.5. RLU, relative light units. (E) Luciferase activities of the indicated plasmids<br />

injected into both VMZ cells at four-cell stage and measured at stage 19. RLU, relative light units. (F) Expression of endogenous Mespo mRNA at the tailbud stage.<br />

(G–I) Expression of GFP mRNA in p-4317GFP transgenic embryos (G) or in p-4317mGFP transgenic embryos (H) at the tailbud stage. The percentages of transgenic<br />

embryos that show the PSM expression (purple), non-specific expression (brown), and no detected signal (white) are shown in panel I.<br />

Discussion<br />

Mespo is involved in segmental patterning during Xenopus<br />

somitogenesis<br />

Mespo knock out mouse shows severe defects in somitogenesis,<br />

including perturbation of segmental patterning. The mutant<br />

mice have no identifiable somites, and the expression of genes<br />

associated with somitogenesis like Mesp2 (the ortholog of<br />

843


844 J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Fig. 6. Lef1 binds to site L in the Mespo promoter in vivo. (A) ChIP assay of<br />

embryos injected with pCS–Lef1–HA and p-511, or pCS–Lef1–HA and p-<br />

511m in both VMZ cells at four-cell stage to detect the binding of Lef1 to site L.<br />

(B) ChIP assay detecting the binding of Lef1 to endogenous Mespo regulatory<br />

sequence. pCS–Lef1–HA was injected into both VMZ cells of embryos at fourcell<br />

stage.<br />

Xenopus Thy), PAPC and Dll-3 (the ortholog of XDelta-1) is<br />

abolished in absence of Mespo (Yoon and Wold, 2000). This<br />

phenotype implies that Mespo is involved in establishing<br />

segmental patterning in somitogenesis. Here, we report that in<br />

Xenopus embryos depleting Mespo by Mespo Mo causes<br />

disorder of somite cell arrangement. During Xenopus somitogenesis,<br />

the rostral PSM cells compact to form cell aggregates<br />

named somitomeres, which later separate from the PSM to form<br />

somites. The nuclei of somite cells line up in the middle of each<br />

somite, representing the well-organized segmental pattern.<br />

There is no such distinct morphological feature in Mespodepleting<br />

Xenopus embryos. Instead, nuclei are randomly<br />

distributed, indicating the disorganization of somite cells (Fig.<br />

1H). The expression patterns of genes associated with<br />

segmental patterning are also perturbed (Fig. 2). Thus, our<br />

data indicate that Mespo functions in establishing the segmental<br />

pattern by means of regulating the genes involved in Xenopus<br />

somitogenesis.<br />

Wnt/β-catenin signaling controls gene expressions in<br />

somitogenesis<br />

Previous studies have demonstrated that Wnt3a is involved<br />

in somitogenesis (Aulehla et al., 2003; Galceran et al., 2004;<br />

Hofmann et al., 2004; Ikeya and Takada, 2001). In chick<br />

embryos, increasing Wnt/β-catenin signaling activity by<br />

implanting beads covered with Wnt3a expressing cells induced<br />

smaller somites (Aulehla et al., 2003). In mouse embryos, it<br />

has been proposed that Wnt3a is involved in somitogenesis via<br />

Axin2 (Aulehla et al., 2003) and Wnt/β-catenin signaling<br />

controlled segmentation by directly regulating Dll1 expression<br />

(Galceran et al., 2004; Hofmann et al., 2004). However, the<br />

effect of Wnt/β-catenin signaling in somitogenesis has not<br />

been fully understood. Our study provides new information on<br />

the role of Wnt/β-catenin signaling in somitogenesis. We<br />

checked XWnt3a and XWnt8 expression in Xenopus embryos<br />

and found that XWnt3a expression is restricted in neural crest<br />

cells, while XWnt8 expression is detected in the caudal PSM<br />

(Figs. 3A, B, E–H). Furthermore, the Wnt antagonist Dkk1 is<br />

expressed in somitomeres (Figs. 3C–F). These observations<br />

suggested that the activity of Wnt/β-catenin signaling exists in<br />

Fig. 7. PI3-K/AKT signaling regulates Mespo expression. (A) Hatched lines<br />

indicate the three caudal regions (C, R, and S) used for western blot of Akt and<br />

phosphorylated Akt. (B) Western blot of Akt and phosphorylated Akt in<br />

different regions indicated in panel A. (C) Western blot of phosphorylated Akt in<br />

the caudal PSM equivalent to region C (illustrated in panel A) treated with either<br />

LY294002 or DMSO. (D–G) The neurula stage embryos were treated with<br />

either DMSO (D, F), or LY294002 (E, G) for 2 h, and then stained for Mespo<br />

and Paraxis as indicated. (H) RT–PCR of Mespo mRNA in the caudal PSM<br />

equivalent to region C (illustrated in panel A) treated with either DMSO or<br />

LY294002. The caudal PSM were dissected at stage 23 and treated with drugs<br />

for 2 h, then harvested for RT–PCR.


Xenopus PSM and is restricted in the caudal PSM. This Wnt/<br />

β-catenin signaling activity is essential for Mespo expression.<br />

Increasing Wnt/β-catenin signaling activity by injecting pCS–<br />

Lef1–VP16 enhances Mespo expression in the PSM, and<br />

decreasing Wnt/β-catenin signaling activity by injecting pCS–<br />

Lef1–EnR downregulates Mespo expression (Fig. 4). We also<br />

found that Wnt/β-catenin signaling, acting upstream of Mespo,<br />

directly controls Mespo expression, based on the<br />

recruitment of Lef1 to the site in the Mespo 5′-regulatory<br />

region (Fig. 6B). Since Mespo expression is essential for<br />

segmental patterning, we concluded that Wnt/β-catenin<br />

signaling controls segmentation process by regulating Mespo<br />

expression.<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Fig. 8. Interaction between PI3-K/AKT and Wnt/β-catenin signaling in PSM. (A) Western blot of β-catenin in the nucleus and cytosol of the caudal PSM cells from<br />

embryos treated with either DMSO or LY294002 for 2 h. (B) Western blot of β-catenin, phosphorylated β-catenin, Akt and phosphorylated Akt in the caudal PSM<br />

cells treated with DMSO for 2.5 h, or LY294002 for 1.5 h or 2.5 h. (C–E) Four-cell-stage embryos were injected in VMZ of one cell with pCS–LacZ as control (C) or<br />

together with pCS–Lef1–VP16 (D, E). At the neurula stage, injected embryos were treated with DMSO (C, D) or LY294002 (E) for 2 h, then fixed, stained with X-gal<br />

to reveal the tracer (light blue) and analyzed by whole-mount in situ hybridization for Mespo. Arrow in panel D indicates the increase of Mespo expression in the<br />

injected side and arrowhead indicates the expression of Mespo in the uninjected side. Arrow in panel E indicates the increased Mespo expression in the injected side<br />

and arrowhead indicates the decrease of Mespo expression in the uninjected side.<br />

Regulation of Mespo expression in the PSM<br />

The bHLH family member Mespo is a key regulator of genes<br />

associated with proper segmentation and successful somitogenesis<br />

(Yoon and Wold, 2000). FGF and RA signaling were proposed to<br />

act upstream of Mespo. It has been shown that blocking FGF<br />

signaling by SU5402 or increasing RA signaling by treating with<br />

RA caused a decrease of Mespo expression (Moreno and Kintner,<br />

2004). However, the molecular nature of how Mespo expression is<br />

regulated remains unclear.<br />

We show here that Wnt/β-catenin signaling is required for<br />

Mespo expression. Increasing Wnt/β-catenin signaling activity<br />

by overexpressing β-catenin or Lef1–VP16 increases the<br />

845


846 J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

expression of Mespo (Fig. 4B and Supplemental Fig. 2B), and<br />

interfering Wnt/β-catenin signaling by overexpressing Lef1–<br />

EnR resulted in the opposite effect (Fig. 4C and Supplemental<br />

Fig. 2D). Our results also show that the PI3-K/AKT pathway<br />

participates in regulating Mespo expression (Fig. 7E). Since it<br />

has been proposed that MKK1/ERK signaling is involved in<br />

regulating Mespo expression (Moreno and Kintner, 2004), it<br />

seems that FGF signaling contributes to the expression of<br />

Mespo through PI3-K/AKT and MKK1/ERK pathway together.<br />

Thus, multiple signaling pathways regulate expression of<br />

Mespo that is a key regulator of somitogenesis.<br />

Interaction of signals in somitogenesis<br />

Previous studies have shown that FGF, Wnt and RA<br />

signaling are all involved in somitogenesis (Aulehla et al.,<br />

2003; Dubrulle et al., 2001; Galceran et al., 2004; Hofmann et<br />

al., 2004; Vermot and Pourquie, 2005). However, whether or<br />

how these signals interact to precisely control somite formation<br />

is poorly understood. In this report, we have provided evidence<br />

that Wnt/β-catenin signaling is involved in controlling<br />

somitogenesis by interacting with FGF signaling. FGF<br />

signaling downstream effector PI3-K/AKT regulated the<br />

stability of β-catenin in the nucleus of the PSM cells (Fig.<br />

8A). Several lines have also shown that RA and FGF signaling<br />

interact to regulate somitogenesis. RA signaling may inhibit<br />

the MAPK/ERK pathway by activating the expression of<br />

MKP3, and FGF signaling seems to regulate RA signaling<br />

activity by regulating the expression of enzymes for RA<br />

production and degradation (Moreno and Kintner, 2004).<br />

Further experiments are needed to explore whether Wnt and<br />

RA signaling interact in somitogenesis.<br />

Acknowledgments<br />

The authors are grateful to Drs. D. Sparrow, E. Joseph,<br />

C. Niehrs and J.C. Smith for sharing plasmids, Dr. Schneider<br />

(Max-Planck-Institut fur Entwicklungsbiologie) for β-catenin<br />

antibody. We thank Drs. W. Wu and Q. Tao for discussion of the<br />

project. This work was supported by the National Natural<br />

Science Foundation of China (90408005, 30270650) and the<br />

National Key Project for Basic Science Research of China<br />

(2001CB509901, 2005CB522704) to X.D. We thank Dr.<br />

Dangsheng Li for his professional polish of the MS.<br />

Appendix A. Supplementary data<br />

Supplementary data associated with this article can be found,<br />

in the online version, at doi:10.1016/j.ydbio.2006.12.034.<br />

References<br />

Aberle, H., Bauer, A., Stappert, J., Kispert, A., Kemler, R., 1997. beta-Catenin is<br />

a target for the ubiquitin–proteasome pathway. EMBO J. 16, 3797–3804.<br />

Aulehla, A., Wehrle, C., Brand-Saberi, B., Kemler, R., Gossler, A., Kanzler, B.,<br />

Herrmann, B.G., 2003. Wnt3a plays a major role in the segmentation clock<br />

controlling somitogenesis. Dev. Cell 4, 395–406.<br />

Conlon, F.L., Smith, J.C., 1999. Interference with brachyury function inhibits<br />

convergent extension, causes apoptosis, and reveals separate requirements in<br />

the FGF and activin signalling pathways. Dev. Biol. 213, 85–100.<br />

Delfini, M.C., Dubrulle, J., Malapert, P., Chal, J., Pourquie, O., 2005. Control of<br />

the segmentation process by graded MAPK/ERK activation in the chick<br />

embryo. Proc. Natl. Acad. Sci. U. S. A. 102, 11343–11348.<br />

Ding, X., Hausen, P., Steinbeisser, H., 1998. Pre-MBT patterning of early gene<br />

regulation in Xenopus: the role of the cortical rotation and mesoderm<br />

induction. Mech. Dev. 70, 15–24.<br />

Dubrulle, J., Pourquie, O., 2004. fgf8 mRNA decay establishes a gradient that<br />

couples axial elongation to patterning in the vertebrate embryo. Nature 427,<br />

419–422.<br />

Dubrulle, J., McGrew, M.J., Pourquie, O., 2001. FGF signaling controls somite<br />

boundary position and regulates segmentation clock control of spatiotemporal<br />

Hox gene activation. Cell 106, 219–232.<br />

Galceran, J., Farinas, I., Depew, M.J., Clevers, H., Grosschedl, R., 1999.<br />

Wnt3a−/−-like phenotype and limb deficiency in Lef1(−/−)Tcf1(−/−)<br />

mice. Genes Dev. 13, 709–717.<br />

Galceran, J., Sustmann, C., Hsu, S.C., Folberth, S., Grosschedl, R., 2004. LEF1mediated<br />

regulation of Delta-like1 links Wnt and Notch signaling in<br />

somitogenesis. Genes Dev. 18, 2718–2723.<br />

Geng, X., Xiao, L., Lin, G.F., Hu, R., Wang, J.H., Rupp, R.A., Ding, X., 2003.<br />

Lef/Tcf-dependent Wnt/beta-catenin signaling during Xenopus axis specification.<br />

FEBS Lett. 547, 1–6.<br />

Glinka, A., Wu, W., Delius, H., Monaghan, A.P., Blumenstock, C., Niehrs, C.,<br />

1998. Dickkopf-1 is a member of a new family of secreted proteins and<br />

functions in head induction. Nature 391, 357–362.<br />

Haq, S., Michael, A., Andreucci, M., Bhattacharya, K., Dotto, P., Walters, B.,<br />

Woodgett, J., Kilter, H., Force, T., 2003. Stabilization of beta-catenin by a<br />

Wnt-independent mechanism regulates cardiomyocyte growth. Proc. Natl.<br />

Acad. Sci. U. S. A. 100, 4610–4615.<br />

Harland, R.M., 1991. In situ hybridization: an improved whole-mount method<br />

for Xenopus embryos. Methods Cell Biol. 36, 685–695.<br />

He, Z., Li, J., Zhen, C., Feng, L., Ding, X., 2005. Knockdown of p53 by RNAi in<br />

ES cells facilitates RA-induced differentiation into muscle cells. Biochem.<br />

Biophys. Res. Commun. 335, 676–683.<br />

Hofmann, M., Schuster-Gossler, K., Watabe-Rudolph, M., Aulehla, A.,<br />

Herrmann, B.G., Gossler, A., 2004. WNT signaling, in synergy with T/<br />

TBX6, controls Notch signaling by regulating Dll1 expression in the<br />

presomitic mesoderm of mouse embryos. Genes Dev. 18, 2712–2717.<br />

Ikeya, M., Takada, S., 2001. Wnt-3a is required for somite specification along<br />

the anteroposterior axis of the mouse embryo and for regulation of cdx-1<br />

expression. Mech. Dev. 103, 27–33.<br />

Joseph, E.M., Cassetta, L.A., 1999. Mespo: a novel basic helix–loop–helix gene<br />

expressed in the presomitic mesoderm and posterior tailbud of Xenopus<br />

embryos. Mech. Dev. 82, 191–194.<br />

Kazanskaya, O., Glinka, A., del Barco Barrantes, I., Stannek, P., Niehrs, C., Wu,<br />

W., 2004. R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling<br />

and is required for Xenopus myogenesis. Dev. Cell 7, 525–534.<br />

Kim, S.H., Jen, W.C., De Robertis, E.M., Kintner, C., 2000. The protocadherin<br />

PAPC establishes segmental boundaries during somitogenesis in Xenopus<br />

embryos. Curr. Biol. 10, 821–830.<br />

Kintner, C.R., Brockes, J.P., 1985. Monoclonal antibodies to the cells of a<br />

regenerating limb. J. Embryol. Exp. Morphol. 89, 37–55.<br />

Knight, Z.A., Gonzalez, B., Feldman, M.E., Zunder, E.R., Goldenberg, D.D.,<br />

Williams, O., Loewith, R., Stokoe, D., Balla, A., Toth, B., Balla, T., Weiss, W.<br />

A., Williams, R.L., Shokat, K.M., 2006. A pharmacological map of the PI3-K<br />

family defines a role for p110alpha in insulin signaling. Cell 125, 733–747.<br />

Kroll, K.L., Amaya, E., 1996. Transgenic Xenopus embryos from sperm nuclear<br />

transplantations reveal FGF signaling requirements during gastrulation.<br />

Development 122, 3173–3183.<br />

Moreno, T.A., Kintner, C., 2004. Regulation of segmental patterning by retinoic<br />

acid signaling during Xenopus somitogenesis. Dev. Cell 6, 205–218.<br />

Nieuwkoop, P.D., Faber, J., 1967. Normal Table of Xenopus laevis. North<br />

Holland, Amsterdam.<br />

Polakis, P., 2001. More than one way to skin a catenin. Cell 105, 563–566.<br />

Pourquie, O., 2001. Vertebrate somitogenesis. Annu. Rev. Cell Dev. Biol. 17,<br />

311–350.


Pourquie, O., 2003. The segmentation clock: converting embryonic time into<br />

spatial pattern. Science 301, 328–330.<br />

Sachs, L.M., Shi, Y.B., 2000. Targeted chromatin binding and histone<br />

acetylation in vivo by thyroid hormone receptor during amphibian<br />

development. Proc. Natl. Acad. Sci. U. S. A. 97, 13138–13143.<br />

Smith, J.C., Price, B.M., Green, J.B., Weigel, D., Herrmann, B.G., 1991.<br />

Expression of a Xenopus homolog of brachyury (T) is an immediate-early<br />

response to mesoderm induction. Cell 67, 79–87.<br />

Sparrow, D.B., Jen, W.C., Kotecha, S., Towers, N., Kintner, C., Mohun, T.J.,<br />

1998. Thylacine 1 is expressed segmentally within the paraxial mesoderm of<br />

the Xenopus embryo and interacts with the Notch pathway. Development<br />

125, 2041–2051.<br />

Takada, S., Stark, K.L., Shea, M.J., Vassileva, G., McMahon, J.A., McMahon,<br />

A.P., 1994. Wnt-3a regulates somite and tailbud formation in the mouse<br />

embryo. Genes Dev. 8, 174–189.<br />

van de Wetering, M., Oosterwegel, M., Dooijes, D., Clevers, H., 1991.<br />

Identification and cloning of TCF-1, a T lymphocyte-specific transcription<br />

factor containing a sequence-specific HMG box. EMBO J. 10, 123–132.<br />

Vermot, J., Pourquie, O., 2005. Retinoic acid coordinates somitogenesis and<br />

left–right patterning in vertebrate embryos. Nature 435, 215–220.<br />

J. Wang et al. / Developmental Biology 304 (<strong>2007</strong>) 836–847<br />

Walker, E.H., Pacold, M.E., Perisic, O., Stephens, L., Hawkins, P.T., Wymann,<br />

M.P., Williams, R.L., 2000. Structural determinants of phosphoinositide<br />

3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and<br />

staurosporine. Mol. Cell 6, 909–919.<br />

Wang, J., Ding, X., 2006. Cloning and analyzing of Xenopus Mespo promoter in<br />

retinoic acid regulated Mespo expression. Acta Biochim. Biophys. Sin. 38<br />

(11), 759–764 (Shanghai).<br />

Weisblat, D.A., Wedeen, C.J., Kostriken, R.G., 1994. Evolution of developmental<br />

mechanisms: spatial and temporal modes of rostrocaudal patterning.<br />

Curr. Top. Dev. Biol. 29, 101–134.<br />

Wilson, V., Rashbass, P., Beddington, R.S., 1993. Chimeric analysis of T<br />

(Brachyury) gene function. Development 117, 1321–1331.<br />

Yamaguchi, T.P., Takada, S., Yoshikawa, Y., Wu, N., McMahon, A.P., 1999. T<br />

(Brachyury) is a direct target of Wnt3a during paraxial mesoderm<br />

specification. Genes Dev. 13, 3185–3190.<br />

Yoon, J.K., Wold, B., 2000. The bHLH regulator pMesogenin1 is required for<br />

maturation and segmentation of paraxial mesoderm. Genes Dev. 14,<br />

3204–3214.<br />

Yoon, J.K., Moon, R.T., Wold, B., 2000. The bHLH class protein pMesogenin1<br />

can specify paraxial mesoderm phenotypes. Dev. Biol. 222, 376–391.<br />

847

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!