• No results found

The Xenopus homologue of Down syndrome critical region protein 6 drives dorsoanterior gene expression and embryonic axis formation by antagonising polycomb group proteins

N/A
N/A
Protected

Academic year: 2020

Share "The Xenopus homologue of Down syndrome critical region protein 6 drives dorsoanterior gene expression and embryonic axis formation by antagonising polycomb group proteins"

Copied!
11
0
0

Loading.... (view fulltext now)

Full text

(1)

ABSTRACT

Mesoderm and embryonic axis formation in vertebrates is mediated by maternal and zygotic factors that activate the expression of target genes. Transcriptional derepression plays an important role in the regulation of expression in different contexts; however, its involvement and possible mechanism in mesoderm and embryonic axis formation are largely unknown. Here we demonstrate that XDSCR6, a Xenopushomologue of human Down syndrome critical region protein 6 (DSCR6, or RIPPLY3), regulates mesoderm and embryonic axis formation through derepression of polycomb group (PcG) proteins. Xdscr6maternal mRNA is enriched in the endoderm of the early gastrula and potently triggers the formation of dorsal mesoderm and neural tissues in ectoderm explants; it also dorsalises ventral mesoderm during gastrulation and induces a secondary embryonic axis. A WRPW motif, which is present in all DSCR6 homologues, is necessary and sufficient for the dorsal mesoderm-and axis-inducing activity. Knockdown of Xdscr6 inhibits dorsal mesoderm gene expression and results in head deficiency. We further show that XDSCR6 physically interacts with PcG proteins through the WRPW motif, preventing the formation of PcG bodies and antagonising their repressor activity in embryonic axis formation. By chromatin immunoprecipitation, we demonstrate that XDSCR6 releases PcG proteins from chromatin and allows dorsal mesoderm gene transcription. Our studies suggest that XDSCR6 might function to sequester PcG proteins and identify a novel derepression mechanism implicated in embryonic induction and axis formation.

KEY WORDS: Transcriptional derepression, Embryonic axis formation, Mesoderm induction, Down syndrome, Polycomb group proteins, Xenopus

INTRODUCTION

During amphibian early development, mesoderm is formed through inductive signals emanating from endoderm cells of the vegetal hemisphere. The molecular basis of this inductive event has been extensively studied and now partly elucidated. Several conserved signalling pathways, including nodal, BMP, Wnt and FGF, are involved in mesoderm induction and patterning (Heasman, 2006; Kimelman, 2006). They act as morphogens to exert long-range effects on responding cells by activating the expression of various target genes (Smith, 2009). Subsequently, the dorsoventral

RESEARCH ARTICLE

1Laboratory of Developmental Biology, CNRS UMR 7622, University Pierre et Marie Curie, 9 quai Saint-Bernard, 75005 Paris, France. 2Department of Marine Biology, Ocean University of China, Qingdao 266003, China.

*Authors for correspondence (clemence.carron_homo@upmc.fr; de-li.shi@upmc.fr)

Received 26 April 2013; Accepted 18 September 2013

patterning of mesoderm is largely mediated by factors expressed in the Spemann’s organizer, which is a source of secreted antagonists for zygotic BMP and Wnt signals (De Robertis et al., 2000; De Robertis and Kuroda, 2004; Niehrs, 2004).

The polycomb group (PcG) proteins are conserved chromatin-interacting factors that are essential for development. They are transcriptional repressors required for maintaining the correct spatial and temporal expression of key developmental genes (Kerppola, 2009; Sawarkar and Paro, 2010). The PcG proteins form multiprotein repressive complexes (polycomb repressive complexes, PRCs), which repress transcription by a mechanism that involves the modification of chromatin (Bracken and Helin, 2009). It has been shown that PcG proteins are concentrated in nuclear foci called PcG bodies, which contribute to epigenetic silencing of Hox genes in Drosophila

(Bantignies et al., 2011; Pirrotta and Li, 2012). Removal of PcG proteins in different tissues of vertebrates leads to the transcriptional activation of genes that are normally repressed (Pietersen et al., 2008; Ezhkova et al., 2011; Juan et al., 2011). In the Xenopusembryo, misexpression of PcG proteins alters the expression of anteroposterior neural genes and produces anterior deficiency (Yoshitake et al., 1999; Barnett et al., 2001). Thus, the PcG system allows context-dependent gene expression and an appreciation of its control of the balance between repression and derepression is crucial in understanding developmental gene regulation; however, the mechanism of derepression by PcG proteins is hitherto unclear.

Down syndrome is one of the most frequent human birth defects, occurring in 1 out of 600 to 1000 births. Phenotypic and molecular analyses of patients have identified a region of chromosome 21, called the Down syndrome critical region (DSCR), which is responsible for many of the characteristic features of Down syndrome when present in three copies (Delabar et al., 1993). However, the function of most of the genes within this region in key early developmental events remains largely unexplored.

Here we report the characterisation of the Xenopus DSCR6 homologue (XDSCR6), which is member of the RIPPLY family proteins (DSCR6 is also known as RIPPLY3). We found that Xdscr6

is a maternal mRNA that is enriched in the endoderm of the early gastrula and functions as a novel and distinct mesoderm and axis inducer. It potently triggers the formation of dorsal mesoderm and neural tissues in ectoderm explants. XDSCR6 also possesses dorsalising activity and can induce a secondary axis. Knockdown of

Xdscr6suggests that it is required for anterior development. We also show that a WRPW motif, which is present in all RIPPLY family proteins, is necessary and sufficient for these activities. Furthermore, we find that XDSCR6 physically and functionally interacts with PcG proteins through the WRPW motif, preventing their accumulation in PcG bodies, releasing them from chromatin, and counteracting their repressive activity. Our results thus identify a novel derepression mechanism and demonstrate that XDSCR6 is an

The

Xenopus

homologue of Down syndrome critical region

protein 6 drives dorsoanterior gene expression and embryonic

axis formation by antagonising polycomb group proteins

Hong-Yan Li1,2, Raphaëlle Grifone1, Audrey Saquet1, Clémence Carron1,* and De-Li Shi1,*

(2)

endogenous inducer for dorsal mesoderm and the embryonic axis. These findings should also help in understanding the biological function and activity of the DSCR6gene in early development and in the pathogenesis of Down syndrome.

RESULTS

XDSCR6 triggers the formation of dorsal mesoderm and neural tissues

XDSCR6 is a member of the RIPPLY family of proteins known to be involved in somite segmentation (Wardle and Papaioannou, 2008); however, whether it plays an early role in development is not clear. RT-PCR analyses indicated that Xdscr6is a maternal mRNA that is expressed at a constant level from fertilisation until mid-gastrula stage (supplementary material Fig. S1A). In the early gastrula, Xdscr6 transcripts are enriched in the endoderm (supplementary material Fig. S1B,C), and its expression could be induced in the ectoderm by nodal signalling, but not by Wnt signalling (supplementary material Fig. S1D). This suggests that

Xdscr6 might play an early role during development. We thus performed functional analyses by injecting Xdscr6mRNA (0.2 ng) into the animal pole region and culturing ectoderm explants to different stages. At the early neurula stage equivalent, uninjected explants remained rounded (Fig. 1A), whereas Xdscr6-injected explants exhibited extensive elongation, which is characteristic of dorsal mesoderm formation (Fig. 1B). At the late neurula stage equivalent, in situ hybridisation analyses showed that Xdscr6 -injected explants expressed the muscle-specific myosin light chain

(MLC) gene, the cement gland marker XCG1and the neuroectoderm gene Sox3(Fig. 1C-H). This shows that overexpression of XDSCR6 triggered the formation of dorsal mesoderm and neural tissues.

We then injected different amounts (0.1, 0.4 and 1 ng) of Xdscr6

mRNA into the animal pole region and cultured ectoderm explants to the early gastrula and the late neurula stage equivalent to further analyse its inducing activity. At the early gastrula stage, Xdscr6at both low and high doses potently induced the expression of dorsoventral mesoderm genes, but not the endoderm gene Sox17α or the Wnt/β-catenin target gene Siamois(Fig. 1I). However, Sox17α expression was induced in ectoderm explants treated with activin (1 ng). Injection of Xdscr6also induced the expression of Xnr1, Xnr2

and Xnr6, but not Xnr4or Xnr5(supplementary material Fig. S2). Consistent with the induction of dorsal mesoderm genes at the early gastrula stage, ectoderm explants injected with high doses of Xdscr6

mRNA (0.4 or 1 ng) and cultured to the late neurula stage equivalent expressed somitic mesoderm genes including muscle actin and

MyoD, as well as various anteroposterior neuroectoderm genes (Fig. 1I). Real-time RT-PCR analyses on early gastrula ectoderm explants injected with low (0.2 ng) and high (1 ng) amounts of Xdscr6

mRNA indicated a dose-dependent induction of mesoderm, but not endoderm, gene expression. In comparison, ectoderm explants treated with low (0.2 ng/ml) and high (1 ng/ml) doses of activin exhibited a dose-dependent induction for both mesoderm and endoderm genes (Fig. 1J). These analyses suggest that XDSCR6 induces only mesoderm gene expression at the early gastrula stage. Since mesoderm induction and patterning depend on both maternal and zygotic factors (Kofron et al., 1999), we tested the activity of zygotic Xdscr6in mesoderm and neural induction by injecting Xdscr6DNA, which allows the transcription of Xdscr6

[image:2.612.57.547.414.659.2]

mRNA under the control of the CMV promoter to occur only after the mid-blastula transition (Rupp and Weintraub, 1991). As shown in Fig. 1I, injection of Xdscr6DNA (0.4 ng) also induced, although

Fig. 1. XDSCR6 triggers the formation of dorsal mesoderm and neural tissues in Xenopusectoderm explants.(A,B) Morphology of control (A) and

Xdscr6-injected (B) explants at the early neurula stage equivalent. (C-H) In situhybridisation analyses of the expression of MLC, XCG1and Sox3genes in control (C,E,G) and Xdscr6-injected (D,F,H) ectoderm explants at the late neurula stage equivalent. (I) RT-PCR analyses of gene expression in ectoderm explants cultured to stage 10.5 and 20 equivalent, following injection of the indicated amounts of Xdscr6mRNA, Xdscr6DNA or after activin treatment. ODC

was used as a loading control. Ms., muscle specific. (J) Real-time RT-PCR analyses of the expression level of the indicated genes in early gastrula ectoderm explants injected with low and high amounts of Xdscr6mRNA (top row) or treated with low and high doses of activin (bottom row). Similar results were obtained

(3)

weakly, the expression of dorsal mesoderm genes in ectoderm explants at the early gastrula stage and of neural genes at the late neurula stage. This result indicates that XDSCR6 is also able to induce dorsal mesoderm gene expression when expressed after zygotic transcription.

XDSCR6 induces a secondary embryonic axis

The activity of XDSCR6 in mesoderm and neural induction led us to examine whether it induces the formation of an embryonic axis. We first injected Xdscr6mRNA (0.4 ng) in the ventro-vegetal region of the 4-cell stage embryo (Fig. 2A); this dose was sufficient to induce the expression of most dorsal mesoderm genes (see Fig. 1I). When injected embryos developed to the early gastrula stage, the formation of a precocious ventral blastopore was apparent (Fig. 2B,C). At the late neurula stage, an ectopic axis became evident (Fig. 2D,E). At the late tail-bud stage, Xdscr6-injected embryos formed a secondary axis with essentially trunk and posterior regions (Fig. 2F,G), although anterior structure including cement gland and eyes was observed in 9% of the injected embryos (supplementary material Table S3). Injection of a high dose of Xdscr6mRNA (1 ng) did not further change the percentage or character of the secondary axis (not shown). Ventro-animal injection of Xdscr6mRNA also induced a secondary axis, although less frequently. In addition, ventro-vegetal injection of Xdscr6 DNA (0.4 ng) induced a secondary axis with high frequency (Fig. 2H; supplementary material Table S3). We then co-injected lacZmRNA (0.5 ng) into one ventral blastomere at the 4-cell stage to analyse how Xdscr6

induces a secondary axis from three independent experiments. When

lacZmRNA was injected alone, β-galactosidase-labelled cells were localised to the posterior and ventral region of the embryo at the late tail-bud stage (Fig. 2I; 100%, n=48). However, in embryos co-injected with Xdscr6and lacZmRNAs, β-galactosidase-labelled cells were mostly localised to the anterior region of the secondary axis (Fig. 2J; 97%, n=64) and formed a mass of muscle (Fig. 2K), indicating that Xdscr6had transformed ventral and posterior cells into dorsal cells. In situhybridisation analyses indicated that the secondary axis expressed the muscle marker MLC(Fig. 2L; 100%,

n=47), the forebrain marker Otx2(Fig. 2M; 67%, n=49) and the spinal cord marker HoxB9 (Fig. 2N; 100%, n=58) but not the notochord marker keratin 8(Fig. 2O; 98%, n=55). Taken together, these results show that XDSCR6 can induce a secondary axis with somites and neural tissues.

Although human RIPPLY proteins show relatively low overall identity (35-40%) with XDSCR6, they similarly induced mesoderm gene expression and a secondary axis (supplementary material Fig. S3). Furthermore, an amphioxus DSCR6 homologue (Li et al., 2006b) could also induce a secondary axis (data not shown). These observations suggest that RIPPLY family proteins from different species exhibit similar activity when overexpressed in the Xenopus

embryo.

XDSCR6 possesses dorsalising and neuralising activity

We further characterised the activity of XDSCR6 in mesoderm patterning. Ventral injection of Xdscr6mRNA (0.4 ng) at the 4-cell stage induced ectopic expression of Otx2and chordin(Fig. 3A-D) and inhibited the expression of Wnt8and Xvent1(Fig. 3E-H) at the early gastrula stage, indicating that it could dorsalise ventral mesoderm. To directly test its dorsalising activity, Xdscr6mRNA or DNA (0.4 ng) was injected into the two ventral blastomeres at the 4-cell stage and dissected ventral marginal zone (VMZ) was cultured to the late neurula stage equivalent in order to examine phenotypic and molecular changes (Fig. 3I). Dorsal marginal zone (DMZ) explants from uninjected embryos undergo elongation (Fig. 3J), which mimics gastrulation movements. However, VMZ explants from uninjected embryos remained rounded (Fig. 3K). By contrast, VMZ explants injected with Xdscr6 mRNA undergo extensive elongation as DMZ explants (Fig. 3L). VMZ explants injected with Xdscr6DNA also elongate, albeit to a lesser extent (Fig. 3M). RT-PCR analyses indicated that injection of Xdscr6

mRNA or DNA induced ventral mesoderm to express dorsal mesoderm genes including muscle actin, Myf5and MyoD, as well as the anterior neuroectoderm gene Otx2and cement gland marker

[image:3.612.48.373.481.742.2]

XCG1(Fig. 3N). Furthermore, radial injection of Xdscr6mRNA in the equatorial region at the 8-cell stage produced embryos with trunk

Fig. 2. XDSCR6 induces a secondary embryonic axis.(A) Xdscr6mRNA or DNA was injected into one ventral blastomere (arrow) at the 4-cell stage. (B,C) Vegetal view of an uninjected early gastrula embryo (B) and a Xdscr6mRNA-injected early gastrula embryo showing the precocious formation of the ventral blastopore (C, arrow). (D,E) Dorsal view, with anterior region at the top, of an uninjected late neurula embryo (D) and a Xdscr6mRNA-injected late neurula embryo with a secondary neural tube (E). (F-H) Phenotypes of control (F), Xdscr6mRNA-injected (G) and Xdscr6DNA-injected (H) embryos at stage 35. (I,J) β-galactosidase staining of stage 32 embryos injected with lacZmRNA alone (I) or co-injected with

Xdscr6mRNA (J). (K) Transverse section

corresponding to J showing β-galactosidase staining in XDSCR6-induced muscles. (L-O) In situhybridisation analyses of the expression of the indicated genes in

Xdscr6mRNA-injected embryos at stage 32. I and II designate the primary axis and the secondary axis, respectively.

(4)

and posterior deficiency (supplementary material Fig. S4). These observations clearly indicate that XDSCR6 could dorsalise ventral mesoderm during gastrulation.

To examine whether XDSCR6 could initiate neural induction in the absence of dorsal mesoderm, we injected a low amount of Xdscr6

mRNA (0.1 ng) into the animal pole region along with VegT-EnR

mRNA (50 pg) or antivinmRNA (20 pg). Although RT-PCR analyses on dissected early gastrula ectoderm explants indicated that neither VegT-EnR nor antivin blocked the mesoderm-inducing activity of XDSCR6 (supplementary material Fig. S5), muscle was absent at the

late neurula stage. However, expression of the cement gland marker

XCG1, which correlates with neural induction, was detected (Fig. 3O; see also Fig. 1I). This suggests that XDSCR6 could trigger the formation of neural tissue in the absence of muscle, although this activity might be achieved by the early expression of chordin.

Xdscr6 knockdown affects anterior development

Since XDSCR6 possesses the activity to induce dorsal mesoderm and a secondary embryonic axis, we then examined whether it is required for these processes. We dorsally injected antisense MOs against Xdscr6 into 4-cell stage embryos and analysed the expression of mesoderm and neuroectoderm genes. We assayed three MOs (MO1, MO2 and MO3) targeting the 5′ untranslated region through the first 25 bases of coding sequence. Since MO1 was more efficient than MO2 and MO3 (data not shown), we used MO1, hereafter designated Xdscr6MO, in the following experiments. At the early gastrula stage, in situ hybridisation analyses revealed that injection of Xdscr6MO (40 ng) downregulated the expression of chordin, noggin and goosecoid

(Fig. 4A-F), indicating that Xdscr6 knockdown affects dorsal mesoderm development. At the end of neurulation, Xdscr6

morphants exhibited reduced expression of Otx2, XCG1and Sox3

(Fig. 4G-L). As development proceeds, injected embryos showed anterior deficiency characterised by the absence of head structure (Fig. 4M,N). These effects could be rescued by co-injection of myc-Xdscr6mRNA (0.1 ng), which is not targeted by Xdscr6MO (Fig. 4O,P). Western blot analyses of early gastrula embryos previously co-injected at the 4-cell stage with Xdscr6MO, Xdscr6-FlagmRNA (0.2 ng) and myc-Xdscr6mRNA (0.1 ng), showed that Xdscr6MO strongly inhibited protein synthesis from Xdscr6-Flag mRNA, which possesses the MO targeting sequence, whereas it had no effect on protein synthesis from myc-Xdscr6 mRNA (Fig. 4Q), indicating that the targeting effect of Xdscr6MO should be specific. We conclude that XDSCR6 is required for dorsal mesoderm formation and anterior development.

The WRPW motif confers XDSCR6 activity

RIPPLY family proteins have no clearly defined functional domain, except for a conserved WRPW tetrapeptide in the N-terminal region (supplementary material Fig. S3A). We overexpressed various Xdscr6mutants (Fig. 5A) by injecting the corresponding mRNA (0.4 ng) into one ventro-vegetal blastomere at the 4-cell stage to investigate the structure-function relationship of XDSCR6 protein in axis induction. The results unambiguously showed that, similar to the wild-type protein, the N-terminal half of XDSCR6 (XDSCR6N-Flag) induced the formation of a secondary axis, whereas the C-terminal half of XDSCR6 (XDSCR6C-Flag) and a point mutant (XDSCR6m-Flag), which transforms WRPW into WRSG, had no axis-inducing activity (Fig. 5B). RT-PCR analyses indicated that wild-type XDSCR6 and XDSCR6N induced Otx2, chordin and Xbra expression in ectoderm explants, whereas XDSCR6C and XDSCR6m had no effect (supplementary material Fig. S6). This indicates that the N-terminal half, in particular the WRPW tetrapeptide, is required for XDSCR6 function.

[image:4.612.46.299.58.482.2]

Since XDSCR6-related proteins were shown to function as a repressor during somitogenesis through physical interaction with Groucho-related proteins (Kawamura et al., 2005; Kondow et al., 2006), we ventrally injected Xdscr6-EnRor Xdscr6-VP16mRNA (0.2 ng) to examine whether the activity of XDSCR6 in mesoderm induction and axis formation is related to a repressor function. Phenotypic analyses showed that both XDSCR6-EnR and Fig. 3. Dorsalisation of ventral mesoderm by XDSCR6.(A-H) In situ

hybridisation analyses of Otx2, chordin, Wnt8and Xvent1expression in uninjected (A,C,E,G) and Xdscr6-injected (B,D,F,H) embryos at the early gastrula stage. Ectopic Otx2and chordinexpression sites are indicated by arrowheads. (I) Scheme of the dorsalisation experiment. Embryos were ventrally injected at the 4-cell stage (double arrows) and DMZ (D) or VMZ (V) explants were dissected at the early gastrula stage and cultured to late neurula stage equivalent. (J-M) Morphology of the uninjected and injected DMZ or VMZ explants. (N) RT-PCR analyses of gene expression in uninjected and injected DMZ or VMZ explants. (O) In situhybridisation of

MLCand XCG1expression in ectoderm explants injected with the mRNAs indicated at the top and cultured to late neurula stage equivalent. DMZ, dorsal marginal zone; VMZ, ventral marginal zone.

(5)

XDSCR6-VP16 induced the formation of a secondary axis (Fig. 5B). Furthermore, injection of myc-WRPWmRNA (0.5 ng) was also sufficient to induce the formation of a secondary axis (Fig. 5B). To exclude the possibility that myc tags might behave as an activation domain for transcription (Ferreiro et al., 1998), we injected GFP-WRPW mRNA (0.5 ng) and found that it similarly induced a secondary axis (Fig. 5B,C). However, injection of myc-WRSGor

GFP-WRSGmRNA (0.5 ng) had no effect (Fig. 5B,D). Western blot analyses indicated that wild-type and mutated proteins were expressed at a similar level (data not shown). In situhybridisation analyses showed that XDSCR6-EnR, XDSCR6-VP16 and myc-WRPW similarly induced ectopic chordin expression (Fig. 5E). These analyses highlight the importance of the WRPW motif in dorsal mesoderm and axis induction and suggest that the axis-inducing activity of XDSCR6 is independent of a repressor or activator function.

XDSCR6 physically interacts with PcG proteins

Although the WRPW motif is generally involved in interaction with Groucho-related proteins (Fisher et al., 1996), we found that the mesoderm- and axis-inducing activity of XDSCR6 was not influenced by co-expression of the Groucho homologue XGrg4

[Xenopusgroucho-related gene 4; also known as transducin-like enhancer of split 4 (Tle4)], or by inhibition of XGrg4 using an MO or the naturally truncated XGrg5, a dominant inhibitory Groucho protein (data not shown). Thus, interaction between XDSCR6 and Groucho-related proteins cannot be the mechanism underlying its mesoderm- and axis-inducing activity. We therefore performed a yeast two-hybrid screen in order to identify novel partners of XDSCR6 and attempt to provide a mechanism of XDSCR6 in mesoderm induction. By this approach, we identified enhancer of zeste homologue 2 (Ezh2). GST pull-down was used to confirm the physical interaction. Xenopus embryos at the 4-cell stage were injected with synthetic mRNA (0.5 ng) encoding XEzh2-myc, or XGrg4-myc as a positive control, and embryo extracts were prepared at the late gastrula stage. The results indicated that both XEzh2-myc and XGrg4-myc bound to GST-XDSCR6 (Fig. 6A). Co-immunoprecipitation further confirmed the interaction between XDSCR6 and XEzh2 (Fig. 6B).

Ezh2 is a component of PcG protein complex PRC2. We tested whether XDSCR6 also interacts with the core PRC1 component BMI1, which was not isolated in the yeast two-hybrid screen. Both GST pull-down and co-immunoprecipitation showed that XDSCR6, but not XDSCR6m, interacts with BMI1 (Fig. 6C,D). GST

pull-Fig. 4. XDSCR6 is required for anterior development. (A,B) Dorso-vegetal view of chordinexpression in a CoMO (A) and a Xdscr6MO-injected (B) embryo at stage 11. (C,D) Dorso-vegetal view of nogginexpression in a CoMO (C) and a

Xdscr6MO-injected (D) embryo at stage 10.5. (E,F) Dorso-vegetal view of goosecoidexpression in a CoMO (E) and a

Xdscr6MO-injected (F) embryo at stage 10. (G,H) Anterior view of Otx2expression in a CoMo (G) and a Xdscr6MO-injected (H) embryo at stage 20. (I,J) Ventral view, with anterior region at the top, of XCG1expression in a CoMO (I) and a Xdscr6 MO-injected (J) embryo at stage 20. (K,L) Dorsal view, with anterior region at the top, of Sox3expression in a CoMO (K) and a

Xdscr6MO-injected (L) embryo at stage 18. (M-O) Phenotypes of a CoMo-injected embryo (M), a Xdscr6morphant embryo (N) and a myc-Xdscr6rescued embryo (O) at stage 38.

(P) Summary of the rescue results. Numbers at the top indicate total embryos scored from four independent experiments. (Q) Western blot analysis shows the blockade of protein synthesis from Xdscr6-FlagmRNA, but not from myc-Xdscr6

mRNA, by Xdscr6MO. Tubulin is a loading control. CoMO, mismatch control MO.

[image:5.612.47.345.55.480.2]
(6)

down using in vitrotranslated proteins further demonstrated a direct interaction between XDSCR6, XEzh2 and BMI1 (Fig. 6E,F). Taken together, these results strongly suggest that Ezh2 and BMI1 are novel partners of XDSCR6 and that the WRPW motif is involved in the physical interaction.

XDSCR6 disrupts the formation of PcG bodies and releases PcG proteins from chromatin

PcG proteins accumulate at pericentric heterochromatin as discrete loci called PcG bodies, which contribute to epigenetic gene silencing (Hernández-Muñoz et al., 2005; Bantignies et al., 2011). To explore the functional significance of the biochemical interaction with Ezh2 and BMI1, we examined whether XDSCR6 can influence the subcellular localisation of endogenous PcG proteins. Nuclear localisation and fluorescence intensity of endogenous Ezh2 and BMI1 in HeLa cells transfected with Xdscr6-RFP (250 ng),

Xdscr6m-RFP(250 ng) or myc-WRPW(250 ng) were analysed by confocal microscopy. In untransfected cells (arrows in Fig. 7), Ezh2 and BMI1 exhibited a punctate nuclear distribution as previously described in this cell line (Saurin et al., 1998; Voncken et al., 1999). However, the nuclear fluorescence intensity for both Ezh2 and BMI1 was significantly reduced in Xdscr6-RFP- or myc-WRPW -transfected cells (arrowheads, Fig. 7A-C,I-K,M-O,U-W). By contrast, transfection of Xdscr6m-RFPdid not affect the nuclear staining of Ezh2 and BMI1 (arrowheads, Fig. 7E-G,Q-S), highlighting the importance of the WRPW motif in interaction with Ezh2 and BMI1. Quantification of nuclear fluorescence intensity confirmed these results (Fig. 7D,H,L,P,T,X). Because PcG bodies represent the site of accumulation of PcG complexes and hence the site of transcriptional repression (Bantignies et al., 2011; Hodgson and Brock, 2011), this observation suggests that XDSCR6 might prevent the targeting of PcG proteins to chromatin and thus avert their transcriptional repression activity.

We then performed ChIP experiments to test this issue directly. We chose to examine the goosecoidpromoter, which was activated by XDSCR6 in ectoderm explants. One-cell stage embryos were injected in the animal pole region with Xdscr6mRNA (0.5 ng) and ectoderm explants were cultured until stage 11. Chromatin from uninjected and injected ectoderm explants was immunoprecipitated and analysed by semi-quantitative and quantitative PCR. The results showed that, in the absence of XDSCR6, a −390/–167 (relative to transcription start site at +1) goosecoidpromoter region was amplified from chromatin precipitated by anti-Ezh2 and anti-BMI1 antibodies. However, in

Xdscr6-injected explants, this region was less efficiently amplified (Fig. 8A-C). As a specificity control, no goosecoidpromoter region was amplified from chromatin immunoprecipitated by an anti-GFP antibody, either in the absence or presence of XDSCR6 (Fig. 8A,D). This indicates that XDSCR6 specifically released Ezh2 and BMI1 from the goosecoidpromoter, a region that also exhibits an increased acetylated histone H4 mark, reflecting an opened state of chromatin in this location (Shogren-Knaak et al., 2006). As a result, the

goosecoidpromoter could be amplified from chromatin precipitated by the anti-acetylated histone H4 antibody (Fig. 8A,E). These observations strongly suggest that the activation of mesoderm gene expression by XDSCR6 results from a derepression mechanism, which is achieved through the dissociation of PcG proteins from chromatin.

XDSCR6 and PcG proteins mutually antagonise in mesoderm and axis formation

To understand the functional significance of this antagonism during development, we co-expressed BMI1 with XDSCR6 in

[image:6.612.49.266.53.515.2]

Xenopus embryos. Dorsal injection of Xdscr6 mRNA (0.2 ng) alone had no significant effect and most embryos (87%) developed normally (Fig. 9A,B; supplementary material Table S4). By contrast, dorsal injection of BMI1mRNA (0.5 ng) alone produced anterior deficiency in 70% of injected embryos (Fig. 9C; supplementary material Table S4). In BMI1and Xdscr6co-injected Fig. 5. The WRPW motif is necessary and sufficient to induce a

secondary axis.(A) Structure of the various Xdscr6constructs. (B) Summary of secondary axis formation in control (Ctl) and injected embryos (constructs numbered as in A). XDSCR6 (1), XDSCR6N (2), XDSCR6-EnR (5), XDSCR6-VP16 (6), myc-WRPW (7) and GFP-WRPW (9) similarly induce a secondary axis, whereas XDSCR6C (3), XDSCR6m (4), myc-WRSG (8) and GFP-WRSG (10) have no effect. The results are expressed as a percentage and numbers at the top indicate total embryos scored from three independent experiments. (C) A GFP-WRPW-injected stage 35 embryo with green fluorescence concentrated in the anterior region of the secondary axis (arrow). (D) A GFP-WRSG-injected embryo lacks a secondary axis, with green fluorescence distributed in the posterior region. (E) In situhybridisation analyses of chordinexpression at stage 10.5 following overexpression of XDSCR6-EnR, XDSCR6-VP16 or myc-WRPW in the ventro-vegetal region at the 4-cell stage. All embryos are in vegetal view.

(7)

embryos, 76% of embryos were morphologically normal (Fig. 9D; supplementary material Table S4), indicating that XDSCR6 counteracts the activity of BMI1 during anterior development. Conversely, ventral injection of Xdscr6mRNA (0.1 ng) induced a secondary axis in 66% of injected embryos (Fig. 9E,F; supplementary material Table S5), whereas ventral injection of

BMI1 mRNA (0.5 ng) had no effect (Fig. 9G; supplementary material Table S5). Co-injection of BMI1 mRNA with Xdscr6

mRNA reduced the prevalence of a secondary axis to 36% (Fig. 9H; supplementary material Table S5), which correlated with a blockade of ectopic chordin expression induced by XDSCR6 (supplementary material Table S6, Fig. S7). In accordance with these in vivoobservations, in vitroanalyses showed that XDSCR6 strongly activated a −1500/+3 goosecoid and a −907/–5 Xnr1

promoter-luciferase reporter gene, whereas these were repressed by co-expression of BMI1 (Fig. 9I,J). Altogether, these results suggest that XDSCR6 releases PcG proteins from chromatin (Fig. 9K) and thus contributes to transcriptional derepression.

DISCUSSION

This study has uncovered a novel derepression mechanism underlying dorsal mesoderm and embryonic axis formation during

Xenopus early development. We show that XDSCR6, through interaction with PcG proteins, removes them from chromatin and triggers the expression of dorsoanterior genes, which then repress the expression of ventral genes.

XDSCR6 is a novel dorsal mesoderm and axis inducer

The activity of XDSCR6 in mesoderm and secondary axis induction presents several characteristic features. First, both maternal and zygotic XDSCR6 potently induces the expression of dorsal mesoderm genes, including several Xnr members. However, the activity of XDSCR6 to initiate the formation of mesoderm and neural tissue is unaffected in the presence of antivin, implying that it might function independently of nodal signalling. Second, XDSCR6 does not have endoderm-inducing activity even at high concentration, whereas other mesoderm-inducing factors, including activin and XNR proteins, can induce both endoderm and mesoderm (Green et al., 1992; Osada and Wright, 1999) (this study). Third, in the absence of muscle, XDSCR6 potently triggers the formation of

both anterior and posterior neural tissues. Finally, XDSCR6-expressing cells participate in the formation of the secondary axis, which lacks a notochord.

The ability of XDSCR6 to induce a secondary axis may be attributed to its dorsal mesoderm-inducing and/or dorsalising activity during gastrulation. Although ventral gene expression could be detected in Xdscr6-injected ectoderm explants, whether this is direct or a consequence of mesoderm formation remains to be determined. In situ hybridisation analyses clearly showed that ventral overexpression of XDSCR6 dorsalises ventral mesoderm through ectopic induction of dorsoanterior genes and downregulation of ventral genes including Wnt8and Xvent1. Thus, the induction of dorsal genes and the repression of ventral genes trigger the formation of a secondary axis. Our findings suggest that XDSCR6 represents a novel mesoderm and embryonic axis inducer with both distinct and shared activity with respect to previously identified mesoderm-inducing factors.

A WRPW motif confers the mesoderm- and axis-inducing activity of XDSCR6

XDSCR6 belongs to the RIPPLY family, which have no conserved functional domain except for a WRPW motif in the N-terminal half. This motif was shown to interact with Groucho-related proteins during somitogenesis (Kawamura et al., 2005; Kondow et al., 2006); however, we find that neither overexpression nor knockdown of XGrg4 affects the activity of XDSCR6 in mesoderm induction and axis formation. This indicates that XDSCR6 and XGrg4 have distinct activity in mesoderm induction. Most strikingly, we found that the WRPW motif is strictly required and sufficient for the dorsal mesoderm- and axis-inducing activity of XDSCR6, whereas the C-terminal half is totally dispensable. By contrast, the C-C-terminal region is required for interaction with Tbx24 during somitogenesis (Kawamura et al., 2008). Furthermore, we find that the axis-inducing activity of XDSCR6 is independent of an activator or repressor function. Together, these differences imply that RIPPLY family proteins regulate mesoderm induction and somitogenesis through distinct mechanisms.

[image:7.612.50.529.58.223.2]

The observation that a WRPW motif alone could function as an axis inducer raises the question of whether other proteins with this motif have similar activity. It is likely that such proteins, to some Fig. 6. XDSCR6 physically interacts with PcG proteins.(A) GST pull-down of embryonically expressed XEzh2-myc or XGrg4-myc by GST-XDSCR6. (B) Co-immunoprecipitation of XEzh2-myc by XDSCR6-Flag. (C) GST pull-down of embryonically expressed XDSCR6-myc, but not the mutant XDSCR6m-myc, by GST-BMI1. (D) Co-immunoprecipitation of XDSCR6-myc by BMI1-Flag. (E) GST pull-down of in vitrotranslated XDSCR6-myc, but not the mutant XDSCR6m-myc, by GST-XEzh2 or GST-BMI1. (F) GST pull-down of in vitrotranslated BMI1-GFP or XEzh2-myc by GST-XDSCR6. IP, immunoprecipitation; WB, western blot.

(8)

extent, exhibit similar function to XDSCR6. For example, Hairy-related proteins may function as dual transcriptional regulators (activator and repressor) in embryonic axis formation (Murato et al., 2006). In the ascidian embryo, Posterior end mark (PEM), which has a WRPW motif and lacks a DNA-binding domain, plays a role in anteroposterior patterning (Yoshida et al., 1996). Thus, WRPW-containing proteins might regulate embryonic patterning with both shared and distinct specificity.

XDSCR6 counteracts the repressor activity of PcG proteins

[image:8.612.48.568.61.357.2]

We also demonstrated that the WRPW motif is indispensable for physical and functional interactions with protein components within both PRC1 and PRC2, which are chromatin-associated multiprotein complexes involved in the maintenance of stable gene repression, especially during early development and stem cell fate decision (Schwartz and Pirrotta, 2008; Kerppola, 2009; Sawarkar and Paro, 2010; Bantignies et al., 2011; Ezhkova et al., 2011). In Fig. 7. XDSCR6 interferes with the nuclear localisation of endogenous Ezh2 and BMI1.Confocal microscopy analyses of Ezh2- and BMI1-associated PcG bodies in HeLa cells. In cells transfected with Xdscr6-RFPor myc-WRPW(arrowheads in A-C,M-O,I-K,U-W), the intensity of endogenous Ezh2 and BMI1 staining is significantly decreased compared with neighbouring untransfected cells in the same culture dish (arrows). In Xdscr6m-RFP-transfected cells (arrowheads in E-G,Q-S), the intensity of fluorescence is comparable to that of neighbouring untransfected cells (arrows). Scale bar: 10 μm. (D,H,L,P,T,X) Quantification of nuclear fluorescence intensity. Red lines represent the median for each condition. Data are presented as mean ± s.e.m. P-values (Student’s

t-test) and number of measures (n) are indicated.

Fig. 8. XDSCR6 releases endogenous Ezh2 and BMI1 from the goosecoidpromoter.ChIP experiments using uninjected (uninj.) or Xdscr6-injected

Xenopusectoderm explants. (A) A representative semi-quantitative PCR result. (B-E) Quantitative PCR analyses of the goosecoidpromoter using chromatin immunoprecipitated from uninjected or Xdscr6-injected ectoderm explants by the indicated antibodies. Data represent the mean of triplicate experiments (error

[image:8.612.50.555.553.694.2]
(9)

functional assays, XDSCR6 prevents the proper nuclear localisation of both Ezh2 and BMI1 in PcG bodies, which contribute to epigenetic silencing. Thus, XDSCR6 may regulate embryonic induction and patterning through interference with endogenous PcG protein function and derepression of dorsal mesoderm genes.

At present, it is unclear how PcG protein-mediated transcriptional silencing established during development is reversed to allow gene expression. In Drosophila, it has been demonstrated that testis-specific TATA box-binding protein (TBP)-associated factors bind to target promoters, reduce binding of PcG proteins and promote gene expression (Chen et al., 2005). XDSCR6 might counteract PcG proteins through both a similar and a distinct mechanism. Based on the interference of XDSCR6 in the formation of PcG bodies and on the binding of PcG proteins to chromatin, we propose that a physical interaction between XDSCR6 and PcG proteins may remove the latter from target promoters, open the chromatin and thus promote gene expression. In support of this hypothesis, it has been shown that several key genes involved in mesoderm induction, including

nodaland brachyury, are targets of PcG proteins. In the absence of PcG function, these genes are expressed independently of the inductive signals (Dahle et al., 2010). Furthermore, there are several lines of evidence showing that conditional knockout of

Ezh2in different mouse tissues is accompanied by transcriptional activation of genes that are normally repressed in these cells

(Chen et al., 2009; Juan et al., 2011). BMI1 has been shown to bind to a specific locus in pancreatic β-cells and decreased binding leads to transcriptional derepression (Dhawan et al., 2009). Thus, it is likely that XDSCR6 triggers mesoderm induction by releasing PcG proteins from chromatin to allow transcriptional activation of mesoderm genes.

In conclusion, the DSCR on chromosome 21 is associated with specific features of Down syndrome that are likely to be caused by an increased gene dosage (Pritchard et al., 2008). Functional analyses of these genes should help to understand how they regulate key aspects of embryogenesis and how their duplication leads to the pathogenesis of Down syndrome. The finding that XDSCR6 antagonises the repressor activity of PcG proteins in embryonic axis formation suggests a novel mechanism of derepression during development.

MATERIALS AND METHODS Embryo manipulations

Xenopuseggs were obtained from females injected with 500 IU of human chorionic gonadotropin (Sigma) and artificially fertilised. For activin treatment, ectoderm explants at the blastula stage were incubated in 0.2 to 1 ng/ml activin solution for 1 hour and then cultured to appropriate stages.

Plasmid constructs

[image:9.612.53.507.362.645.2]

The full-length Xdscr6(AB073615) and human RIPPLY2(NM_001009994) sequences were PCR amplified and cloned into the pCS2 vector (Rupp and Weintraub, 1991). Xdscr6-Flag, Xdscr6N-Flagand Xdscr6C-Flagwere cloned in the pCS2 vector, upstream of the sequence coding for the Flag epitope.

Fig. 9. XDSCR6 and BMI1 are mutually antagonistic during embryonic axis formation.(A-D) XDSCR6 rescues anterior deficiency produced by BMI1 overexpression. Embryos at the 4-cell stage were either left uninjected (A) or dorsally injected with Xdscr6mRNA (B), BMI1mRNA (C) or co-injected with BMI1

and Xdscr6mRNAs (D), and cultured to the stage 32. (E-H) BMI1 inhibits the axis-inducing activity of XDSCR6. Embryos at the 4-cell stage were either left uninjected (E) or ventrally injected with Xdscr6mRNA (F), BMI1mRNA (G) or co-injected with BMI1and Xdscr6mRNAs (H), and cultured to the stage 35. (I,J) BMI1 represses the goosecoidpromoter (gsc-luc) and Xnr1promoter (Xnr1-luc) activity that had been stimulated by overexpression of XDSCR6 in ectoderm explants. Values were expressed relative to the value obtained from uninjected control explants. Mean ± s.e.m.; P<0.05 (Student’s t-test). RLU, relative luciferase units. (K) Model of XDSCR6 function in transcriptional derepression. (Top) In the absence of XDSCR6, PcG proteins bind to chromatin and repress the transcription of mesoderm genes. (Bottom) The interaction between XDSCR6 and PcG proteins removes PRC1 and PRC2 from chromatin and

(10)

Xdscr6m-Flag, bearing mutations in the WRPW coding sequence, was created by site-directed mutagenesis according to the manufacturer’s recommendations (Stratagene). A myc-tagged version of Xdscr6(myc-Xdscr6) was made by cloning the coding sequence, without the initiation codon, after the six myc epitopes in the pCS2 vector. Xdscr6-EnRand Xdscr6-VP16were constructed by cloning the Xdscr6coding sequence in frame with the engrailed repressor domain or the VP16 activator domain, respectively. The myc-tagged or GFP-tagged WRPW and WRSG constructs were obtained by cloning the WRPW or WRSG coding sequence after the six myc epitopes or the GFP in the pCS2 vector. BMI1-Flag, BMI1-GFP, Xdscr6-myc, Xdscr6m-myc, Xdscr6-RFPand

Xdscr6m-RFPwere also obtained by PCR. All constructs were sequenced before use. XEzh2-myc, VegT-EnRand antivinconstructs were described previously (Thisse and Thisse, 1999; Barnett et al., 2001; Li et al., 2006a). Capped mRNAs were synthesized by in vitrotranscription using appropriate RNA polymerases as previously described (Djiane et al., 2000).

Morpholinos (MOs)

The MOs for Xdscr6(MO1, 5′-AACTCCGCTGTTGATTCGGCTCCAT-3′; MO2, 5′-CCGCAACTCCGCTGTTGATTCGGCT-3′; MO3, 5′AGA -ACAGTTCGGCCGTGTCTCTGAC-3′) and the control MO (CoMO) incorporating five mismatches (indicated in lowercase) in the MO1 sequence (5′-AAgTCCcCTGTTcATTCcGCTgCAT-3′) were from Gene Tools, and suspended in sterile water.

In situ hybridisation and cell lineage tracing

Whole-mount in situhybridisation was performed according to a standard protocol (Harland, 1991). Probes were labelled using digoxigenin-11-UTP and appropriate RNA polymerases. Cell lineage tracing was performed by injection of lacZmRNA followed by β-galactosidase staining using X-Gal as substrate.

RT-PCR and real-time RT-PCR

For RT-PCR, total RNA extraction and reverse transcription were as described (Li et al., 2010). PCR primers are listed in supplementary material Table S1 or were described previously (Shi et al., 2002). PCR products amplified in a reaction mixture containing 1 μCi [α-32P]dCTP were resolved

on a 5% non-denaturing polyacrylamide gel and visualised using a phosphorimager (Bio-Rad).

For real-time RT-PCR, total RNA from 15 ectoderm explants was extracted using the RNeasy Mini Kit (Qiagen) and reverse transcribed using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) according to the manufacturer’s instructions. Quantitative PCR was performed using SsoAdvanced SYBR Green Supermix (Bio-Rad) according to the manufacturer’s instructions. PCR primers are listed in supplementary material Table S2. The data were analysed using both ornithine decarboxylase(ODC) and EF1αas reference.

Yeast two-hybrid screening

The Xdscr6coding sequence was cloned in the pGBKT7 vector (Clontech) in frame with the Gal4 DNA-binding domain (Gal4-Xdscr6). The AH109yeast strain was transformed with this construct and expression of the fusion protein was detected by western blot using anti-Gal4 or anti-myc antibody. The

AH109-Gal4-Xdscr6 strain was mated with an 11-day mouse embryo Matchmaker cDNA library pre-transformed into yeast strain Y187(Clontech) and ~2.4×106independent clones were screened at high stringency.

GST pull-down and co-immunoprecipitation

GST-Xdscr6 and GST-BMI1 constructs were obtained by cloning their coding sequences in pGEX4T1, and fusion proteins produced in E. colicells were bound to glutathione-agarose (Sigma). Synthetic mRNAs encoding epitope-tagged proteins were injected into Xenopusembryos and lysates were incubated with glutathione-agarose-bound GST fusion proteins overnight at 4°C. Bound proteins were subjected to western blot analyses. The experiment was also performed with in vitrotranslated proteins using the TNT Coupled Reticulocyte Lysate System (Promega). Co-immunoprecipitation was performed as previously described (Carron et al., 2003).

Cell transfection and confocal immunofluorescence microscopy

Xdscr6-RFP, Xdscr6m-RFPor myc-WRPWplasmids were transfected into HeLa cells using Fugene 6 reagent (Roche). Transfected cells were cultured for 24 hours and then fixed in PBS-buffered 4% paraformaldehyde. After permeabilisation in PBS containing 0.1% Tween 20 and blocking in PBS containing 5% BSA, immunodetection was performed with rabbit anti-Ezh2 antibody (1:800; Abcam, 1 mg/ml stock, code ab3748), mouse anti-BMI1 antibody (1:100; Abcam, 1 mg/ml stock, code ab14389) or 9E10 antibody (1:2000; Santa Cruz Biotechnology, 200 µg/ml, code sc-40), followed by Alexa Fluor 488- or Alexa Fluor 594-conjugated secondary antibody. The cells were then stained with 4⬘,6-diamidino-2-phenylindole dihydrochloride (DAPI). Images were acquired with a Leica SP5 confocal microscope and quantification of nuclear fluorescence intensity was by fluorescent particle counting in an area of 20 μm2using ImageJ software (NIH). Box plots were

generated using R software. Statistical significance was assessed by Student’s t-test in triplicate.

Chromatin immunoprecipitation (ChIP)

ChIP was based on a published method (Havis et al., 2006). One-cell stage embryos were injected in the animal pole region with Xdscr6mRNA (0.5 ng) and ectoderm explants were dissected at stage 11. Sonicated chromatin from 100 ectoderm explants was incubated with 10 μg anti-Ezh2 antibody (Abcam), 5 μg anti-BMI1 antibody (Abcam), 8 µl anti-acetylated histone H4 antibody (Millipore, unpurified serum, code 06-866) or 10 μg anti-GFP antibody (Roche, 400 mg/ml stock, code 11814460001). The goosecoid

promoter regions (−390/–167 and −390/–290) were analysed by semi-quantitative PCR or by semi-quantitative PCR in triplicate using the primers listed in supplementary material Tables S1 and S2.

Luciferase assay

The −1500/+3 goosecoidand −907/–5 Xnr1promoter-luciferase reporter plasmid DNAs (Watabe et al., 1995; Cao et al., 2007; Cao et al., 2008) were injected at 100 pg with synthetic mRNAs in the animal pole region at the 2-cell stage, and the luciferase assay was performed using ten early gastrula ectoderm explants as described (Cao et al., 2012). The experiments were carried out in triplicate using different batches of embryos and the data were analysed using Student’s t-test.

Acknowledgements

We thank E. Jones, H. Clevers, A. Ciechanover, Y. Cao, B. Thisse and M. van Lohuizen for reagents; the imaging facility at IFR83 (University Pierre et Marie Curie) for help with confocal image acquisition; and C. Antoniewski, R. Gautier, D. Cladière, E. Mouchel-Viehl, R. Le Bouffant and E. Havis for helpful discussion and technical advice.

Competing interests

The authors declare no competing financial interests.

Author contributions

H.-Y.L. performed the experiments with assistance from R.G. and A.S.; C.C. and D.-L.S. designed and performed the experiments, analysed results and wrote the paper.

Funding

This work was supported by grants from Association Française contre les Myopathies (AFM), Ligue Nationale Contre le Cancer (LNCC) and Agence Nationale de la Recherche (ANR) [ANR-09-BLAN-0262-03].

Supplementary material

Supplementary material available online at

http://dev.biologists.org/lookup/suppl/doi:10.1242/dev.098319/-/DC1

References

Bantignies, F., Roure, V., Comet, I., Leblanc, B., Schuettengruber, B., Bonnet, J., Tixier, V., Mas, A. and Cavalli, G.(2011). Polycomb-dependent regulatory contacts between distant Hox loci in Drosophila. Cell144, 214-226.

Barnett, M. W., Seville, R. A., Nijjar, S., Old, R. W. and Jones, E. A.(2001). Xenopus Enhancer of Zeste (XEZ); an anteriorly restricted polycomb gene with a role in neural patterning. Mech. Dev.102, 157-167.

(11)

Cao, Y., Siegel, D., Donow, C., Knöchel, S., Yuan, L. and Knöchel, W.(2007). POU-V factors antagonize maternal POU-VegT activity and beta-Catenin signaling in Xenopus embryos. EMBO J.26, 2942-2954.

Cao, Y., Siegel, D., Oswald, F. and Knöchel, W.(2008). Oct25 represses transcription of nodal/activin target genes by interaction with signal transducers during Xenopus gastrulation. J. Biol. Chem.283, 34168-34177.

Cao, J. M., Li, S. Q., Zhang, H. W. and Shi, D. L.(2012). High mobility group B proteins regulate mesoderm formation and dorsoventral patterning during zebrafish and Xenopus early development. Mech. Dev.129, 263-274.

Carron, C., Pascal, A., Djiane, A., Boucaut, J. C., Shi, D. L. and Umbhauer, M. (2003). Frizzled receptor dimerization is sufficient to activate the Wnt/beta-catenin pathway. J. Cell Sci.116, 2541-2550.

Chen, X., Hiller, M., Sancak, Y. and Fuller, M. T. (2005). Tissue-specific TAFs counteract Polycomb to turn on terminal differentiation. Science310, 869-872. Chen, H., Gu, X., Su, I. H., Bottino, R., Contreras, J. L., Tarakhovsky, A. and Kim,

S. K. (2009). Polycomb protein Ezh2 regulates pancreatic beta-cell Ink4a/Arf expression and regeneration in diabetes mellitus. Genes Dev.23, 975-985. Dahle, Ø., Kumar, A. and Kuehn, M. R.(2010). Nodal signaling recruits the histone

demethylase Jmjd3 to counteract polycomb-mediated repression at target genes. Sci. Signal.3, ra48.

De Robertis, E. M. and Kuroda, H.(2004). Dorsal-ventral patterning and neural induction in Xenopus embryos. Annu. Rev. Cell Dev. Biol.20, 285-308.

De Robertis, E. M., Larraín, J., Oelgeschläger, M. and Wessely, O.(2000). The establishment of Spemann’s organizer and patterning of the vertebrate embryo. Nat. Rev. Genet.1, 171-181.

Delabar, J. M., Theophile, D., Rahmani, Z., Chettouh, Z., Blouin, J. L., Prieur, M., Noel, B. and Sinet, P. M.(1993). Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur. J. Hum. Genet.1, 114-124.

Dhawan, S., Tschen, S. I. and Bhushan, A. (2009). Bmi-1 regulates the Ink4a/Arf locus to control pancreatic beta-cell proliferation. Genes Dev.23, 906-911. Djiane, A., Riou, J. F., Umbhauer, M., Boucaut, J. C. and Shi, D. L.(2000). Role of

frizzled 7 in the regulation of convergent extension movements during gastrulation in Xenopus laevis. Development127, 3091-3100.

Ezhkova, E., Lien, W. H., Stokes, N., Pasolli, H. A., Silva, J. M. and Fuchs, E. (2011). EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essential for hair follicle homeostasis and wound repair. Genes Dev.25, 485-498.

Ferreiro, B., Artinger, M., Cho, K. and Niehrs, C.(1998). Antimorphic goosecoids. Development125, 1347-1359.

Fisher, A. L., Ohsako, S. and Caudy, M.(1996). The WRPW motif of the hairy-related basic helix-loop-helix repressor proteins acts as a 4-amino-acid transcription repression and protein-protein interaction domain. Mol. Cell. Biol.16, 2670-2677. Green, J. B., New, H. V. and Smith, J. C.(1992). Responses of embryonic Xenopus

cells to activin and FGF are separated by multiple dose thresholds and correspond to distinct axes of the mesoderm. Cell71, 731-739.

Harland, R. M.(1991). In situ hybridization: an improved whole mount method for Xenopusembryos. Methods Cell Biol. 36, 685-695.

Havis, E., Le Mevel, S., Morvan Dubois, G., Shi, D. L., Scanlan, T. S., Demeneix, B. A. and Sachs, L. M.(2006). Unliganded thyroid hormone receptor is essential for Xenopus laevis eye development. EMBO J.25, 4943-4951.

Heasman, J.(2006). Maternal determinants of embryonic cell fate. Semin. Cell Dev. Biol.17, 93-98.

Hernández-Muñoz, I., Taghavi, P., Kuijl, C., Neefjes, J. and van Lohuizen, M. (2005). Association of BMI1 with polycomb bodies is dynamic and requires PRC2/EZH2 and the maintenance DNA methyltransferase DNMT1. Mol. Cell. Biol. 25, 11047-11058.

Hodgson, J. W. and Brock, H. W.(2011). Are polycomb group bodies gene silencing factories? Cell144, 170-171.

Juan, A. H., Derfoul, A., Feng, X., Ryall, J. G., Dell’Orso, S., Pasut, A., Zare, H., Simone, J. M., Rudnicki, M. A. and Sartorelli, V.(2011). Polycomb EZH2 controls self-renewal and safeguards the transcriptional identity of skeletal muscle stem cells. Genes Dev.25, 789-794.

Kawamura, A., Koshida, S., Hijikata, H., Ohbayashi, A., Kondoh, H. and Takada, S.(2005). Groucho-associated transcriptional repressor ripply1 is required for proper transition from the presomitic mesoderm to somites. Dev. Cell9, 735-744. Kawamura, A., Koshida, S. and Takada, S.(2008). Activator-to-repressor conversion

of T-box transcription factors by the Ripply family of Groucho/TLE-associated mediators. Mol. Cell. Biol.28, 3236-3244.

Kerppola, T. K. (2009). Polycomb group complexes – many combinations, many functions. Trends Cell Biol.19, 692-704.

Kimelman, D.(2006). Mesoderm induction: from caps to chips. Nat. Rev. Genet.7, 360-372.

Kofron, M., Demel, T., Xanthos, J., Lohr, J., Sun, B., Sive, H., Osada, S., Wright, C., Wylie, C. and Heasman, J.(1999). Mesoderm induction in Xenopus is a zygotic event regulated by maternal VegT via TGFbeta growth factors. Development126, 5759-5770.

Kondow, A., Hitachi, K., Ikegame, T. and Asashima, M.(2006). Bowline, a novel protein localized to the presomitic mesoderm, interacts with Groucho/TLE in Xenopus. Int. J. Dev. Biol.50, 473-479.

Li, H. Y., Bourdelas, A., Carron, C., Gomez, C., Boucaut, J. C. and Shi, D. L. (2006a). FGF8, Wnt8 and Myf5 are target genes of Tbx6 during anteroposterior specification in Xenopus embryo. Dev. Biol.290, 470-481.

Li, X. Y., Zhang, W., Chen, D. Y., Lin, Y. S., Huang, X. W., Shi, D. L. and Zhang, H. W.(2006b). Expression of a novel somite-formation-related gene, AmphiSom, during amphioxus development. Dev. Genes Evol.216, 52-55.

Li, H. Y., Bourdelas, A., Carron, C. and Shi, D. L.(2010). The RNA-binding protein Seb4/RBM24 is a direct target of MyoD and is required for myogenesis during Xenopus early development. Mech. Dev.127, 281-291.

Murato, Y., Yamaguti, M., Katamura, M., Cho, K. W. and Hashimoto, C.(2006). Two modes of action by which Xenopus hairy2b establishes tissue demarcation in the Spemann-Mangold organizer. Int. J. Dev. Biol.50, 463-471.

Niehrs, C.(2004). Regionally specific induction by the Spemann-Mangold organizer. Nat. Rev. Genet.5, 425-434.

Osada, S. I. and Wright, C. V.(1999). Xenopus nodal-related signaling is essential for mesendodermal patterning during early embryogenesis. Development126, 3229-3240.

Pietersen, A. M., Evers, B., Prasad, A. A., Tanger, E., Cornelissen-Steijger, P., Jonkers, J. and van Lohuizen, M. (2008). Bmi1 regulates stem cells and proliferation and differentiation of committed cells in mammary epithelium. Curr. Biol. 18, 1094-1099.

Pirrotta, V. and Li, H. B.(2012). A view of nuclear Polycomb bodies. Curr. Opin. Genet. Dev.22, 101-109.

Pritchard, M., Reeves, R. H., Dierssen, M., Patterson, D. and Gardiner, K. J. (2008). Down syndrome and the genes of human chromosome 21: current knowledge and future potentials. Cytogenet. Genome Res.121, 67-77.

Rupp, R. A. and Weintraub, H. (1991). Ubiquitous MyoD transcription at the midblastula transition precedes induction-dependent MyoD expression in presumptive mesoderm of X. laevis. Cell65, 927-937.

Saurin, A. J., Shiels, C., Williamson, J., Satijn, D. P., Otte, A. P., Sheer, D. and Freemont, P. S. (1998). The human polycomb group complex associates with pericentromeric heterochromatin to form a novel nuclear domain. J. Cell Biol.142, 887-898.

Sawarkar, R. and Paro, R.(2010). Interpretation of developmental signaling at chromatin: the Polycomb perspective. Dev. Cell19, 651-661.

Schwartz, Y. B. and Pirrotta, V.(2008). Polycomb complexes and epigenetic states. Curr. Opin. Cell Biol.20, 266-273.

Shi, D. L., Bourdelas, A., Umbhauer, M. and Boucaut, J. C. (2002). Zygotic Wnt/beta-catenin signaling preferentially regulates the expression of Myf5 gene in the mesoderm of Xenopus. Dev. Biol.245, 124-135.

Shogren-Knaak, M., Ishii, H., Sun, J. M., Pazin, M. J., Davie, J. R. and Peterson, C. L. (2006). Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science311, 844-847.

Smith, J. C.(2009). Forming and interpreting gradients in the early Xenopus embryo. Cold Spring Harb. Perspect. Biol.1, a002477.

Thisse, C. and Thisse, B.(1999). Antivin, a novel and divergent member of the TGFbeta superfamily, negatively regulates mesoderm induction. Development126, 229-240.

Voncken, J. W., Schweizer, D., Aagaard, L., Sattler, L., Jantsch, M. F. and van Lohuizen, M.(1999). Chromatin-association of the Polycomb group protein BMI1 is cell cycle-regulated and correlates with its phosphorylation status. J. Cell Sci.112, 4627-4639.

Wardle, F. C. and Papaioannou, V. E.(2008). Teasing out T-box targets in early mesoderm. Curr. Opin. Genet. Dev.18, 418-425.

Watabe, T., Kim, S., Candia, A., Rothbächer, U., Hashimoto, C., Inoue, K. and Cho, K. W.(1995). Molecular mechanisms of Spemann’s organizer formation: conserved growth factor synergy between Xenopus and mouse. Genes Dev.9, 3038-3050. Yoshida, S., Marikawa, Y. and Satoh, N.(1996). Posterior end mark, a novel maternal

gene encoding a localized factor in the ascidian embryo. Development122, 2005-2012.

Yoshitake, Y., Howard, T. L., Christian, J. L. and Hollenberg, S. M. (1999). Misexpression of Polycomb-group proteins in Xenopus alters anterior neural development and represses neural target genes. Dev. Biol.215, 375-387.

Figure

Fig. 1. XDSCR6 triggers the formation of dorsal mesoderm and neural tissues in Xenopus ectoderm explants
Fig. 2. XDSCR6 induces a secondary embryonicaxis.neurula embryo with a secondary neural tube (E)
Fig. 3. Dorsalisation of ventral mesoderm by XDSCR6. (A-H) indicated at the top and cultured to late neurula stage equivalent
Fig. 4. XDSCR6 is required for anterior development.
+5

References

Related documents

In this study, there was no significant correlation between outcome of meningitis with age, sex, or the estimated interval between hospital admission and the

In this study, we identified several strains with mutations in the ribosomal proteins putatively associated with linezolid resistance; two linezolid- resistant isolates harbored only

ABSTRACT Mycobacterium haemophilum is an emerging pathogen associated with a variety of clinical syndromes, most com- monly skin infections in immunocompromised

Gene expression of Pseudomonas aeruginosa in a mucin-containing syn- thetic growth medium mimicking cystic fibrosis lung sputum. A survey of TonB-dependent

14 He also provided for the right of appeal, and for trial by a jury of six men.' 5 This jury had no legal standing under Mexican law, but it represented fair trial to

In an effort to provide more certainty to foreign limited partnerships in light of the potentially harsh consequences resulting from failure to qualify, TRLPA

This unique quality of HRV/HRC analyses has led to interest in its application in pre-hospital triage and emergency clinical decision making; interest which has been bol- stered by

Our study successfully implemented a referral program for maternal contraceptive services within pediatric care by having pediatric residents assess postpartum women ’ s needs