J Mol Cell Biol-2014-Xu-272-85.pdf

14
Article Maternal Eomesodermin regulates zygotic nodal gene expression for mesendoderm induction in zebrafish embryos Pengfei Xu , Gaoyang Zhu , Yixia Wang, Jiawei Sun, Xingfeng Liu, Ye-Guang Chen, and Anming Meng * State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua2Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China These authors contributed equally to this work. * Correspondence to: Anming Meng, E-mail: [email protected] Development of animal embryos before zygotic genome activation at the midblastula transition (MBT) is essentially supported by egg- derived maternal products. Nodal proteins are crucial signals for mesoderm and endoderm induction after the MBT. It remains unclear which maternal factors activate zygotic expression of nodal genes in the ventrolateral blastodermal margin of the zebrafish blastulas. In this study, we show that loss of maternal Eomesodermin a (Eomesa), a T-box transcription factor, impairs zygotic expression of the nodal genes ndr1 and ndr2 as well as mesodermal and endodermal markers, indicating an involvement in mesendoderm induction. Maternal Eomesa is also required for timely zygotic expression of the transcription factor gene mxtx2, a regulator of nodal gene expres- sion. Eomesa directly binds to the Eomes-binding sites in the promoter or enhancer of ndr1, ndr2, and mxtx2 to activate their transcrip- tion. Furthermore, human and mouse Nodal genes are also regulated by Eomes. Transfection of zebrafish eomesa into murine embryonic stem cells promotes mesendodermal differentiation with constant higher levels of endogenous Nodal expression, suggest- ing a conserved function of Eomes. Taken together, our findings reveal a conserved role of maternal T-box transcription factors in regu- lating nodal gene expression and mesendoderm induction in vertebrate embryos. Keywords: Eomesodermin, Nodal, transcription, mesoderm, endoderm, embryo, zebrafish Introduction Large amounts of maternal products, including RNAs and pro- teins, are stored in animal eggs and support early embryonic development upon fertilization before the zygotic genome starts to transcribe at the midblastula transition (MBT) (Tadros and Lipshitz, 2009). The germ layers are formed during gastrulation stages, but their progenitors are specified at and after MBT by coor- dinated action of multiple signals. It has been found that Nodal pro- teins, TGF-b superfamily members, play an essential role in mesoderm and endoderm induction in vertebrate embryos (Tian and Meng, 2006). For example, Nodal-deficient mouse embryos fail to form the primitive streak and mesoderm and endoderm pre- cursors (Conlon et al., 1991, 1994; Zhou et al., 1993); in zebrafish embryos, double mutants with simultaneous loss-of-function of two zygotic nodal genes ndr1/squint (sqt) and ndr2/cyclops (cyc) lack endodermal tissues and most of mesodermal tissues (Feldman et al., 1998), and maternal ndr1 transcripts may also be required for dorsal mesodermal specification independent of Ndr1 protein (Gore et al., 2005; Hong et al., 2011; Lim et al., 2012; Kumari et al., 2013); during Xenopus embryogenesis, Nodal signals are also essential for mesoderm induction and pat- terning (Jones et al., 1995; Osada and Wright, 1999; Agius et al., 2000). An interesting question is how maternal factors contribute to zygotic transcription of nodal genes during developmental course. In Xenopus and zebrafish embryos, nodal gene expression in the dorsal organizer is induced partially by maternally supplied b-catenin (Kelly et al., 2000; Xanthos et al., 2002; Bellipanni et al., 2006). Studies in Xenopus indicate that zygotic expression of nodal genes is initiated in the entire vegetal endoderm, which also requires other maternal factors such as the vegetally loca- lized T-box transcriptional factor VegT (Agius et al., 2000; Xanthos et al., 2002). However, fish or mammalian orthologs of VegT remain to be verified or identified. In zebrafish embryos, ndr1 and ndr2 are expressed during mid-to-late blastulation period in the yolk syncytial layer (YSL), an extraembryonic syncyt- ial layer of cytoplasm and nuclei formed by breakdown of Received January 14, 2014. Revised April 23, 2014. Accepted April 29, 2014. # The Author (2014). Published by Oxford University Press on behalf of Journal of Molecular Cell Biology, IBCB, SIBS, CAS. All rights reserved. 272 | Journal of Molecular Cell Biology (2014), 6(4), 272 285 doi:10.1093/jmcb/mju028 Published online June 12, 2014 by guest on October 14, 2014 http://jmcb.oxfordjournals.org/ Downloaded from

Transcript of J Mol Cell Biol-2014-Xu-272-85.pdf

Page 1: J Mol Cell Biol-2014-Xu-272-85.pdf

Article

Maternal Eomesodermin regulates zygotic nodalgene expression for mesendoderm induction inzebrafish embryosPengfei Xu†, Gaoyang Zhu†, Yixia Wang, Jiawei Sun, Xingfeng Liu, Ye-Guang Chen, and Anming Meng*

State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua2Peking Center for Life Sciences, School of Life Sciences, Tsinghua University,

Beijing 100084, China† These authors contributed equally to this work.

* Correspondence to: Anming Meng, E-mail: [email protected]

Development of animal embryos before zygotic genome activation at the midblastula transition (MBT) is essentially supported by egg-

derived maternal products. Nodal proteins are crucial signals for mesoderm and endoderm induction after the MBT. It remains unclear

which maternal factors activate zygotic expression of nodal genes in the ventrolateral blastodermal margin of the zebrafish blastulas.

In this study, we show that loss of maternal Eomesodermin a (Eomesa), a T-box transcription factor, impairs zygotic expression of the

nodal genes ndr1 and ndr2 as well as mesodermal and endodermal markers, indicating an involvement in mesendoderm induction.

Maternal Eomesa is also required for timely zygotic expression of the transcription factor gene mxtx2, a regulator of nodal gene expres-

sion. Eomesa directly binds to the Eomes-binding sites in the promoter or enhancer of ndr1, ndr2, and mxtx2 to activate their transcrip-

tion. Furthermore, human and mouse Nodal genes are also regulated by Eomes. Transfection of zebrafish eomesa into murine

embryonic stem cells promotes mesendodermal differentiation with constant higher levels of endogenous Nodal expression, suggest-

ing a conserved function of Eomes. Taken together, our findings reveal a conserved role of maternal T-box transcription factors in regu-

lating nodal gene expression and mesendoderm induction in vertebrate embryos.

Keywords: Eomesodermin, Nodal, transcription, mesoderm, endoderm, embryo, zebrafish

Introduction

Large amounts of maternal products, including RNAs and pro-

teins, are stored in animal eggs and support early embryonic

development upon fertilization before the zygotic genome starts

to transcribe at the midblastula transition (MBT) (Tadros and

Lipshitz, 2009). The germ layers are formed during gastrulation

stages, but their progenitors are specified at and after MBT by coor-

dinated action of multiple signals. It has been found that Nodal pro-

teins, TGF-b superfamily members, play an essential role in

mesoderm and endoderm induction in vertebrate embryos (Tian

and Meng, 2006). For example, Nodal-deficient mouse embryos

fail to form the primitive streak and mesoderm and endoderm pre-

cursors (Conlon et al., 1991, 1994; Zhou et al., 1993); in zebrafish

embryos, double mutants with simultaneous loss-of-function of

two zygotic nodal genes ndr1/squint (sqt) and ndr2/cyclops

(cyc) lack endodermal tissues and most of mesodermal tissues

(Feldman et al., 1998), and maternal ndr1 transcripts may also be

required for dorsal mesodermal specification independent of

Ndr1 protein (Gore et al., 2005; Hong et al., 2011; Lim et al.,

2012; Kumari et al., 2013); during Xenopus embryogenesis,

Nodal signals are also essential for mesoderm induction and pat-

terning (Jones et al., 1995; Osada and Wright, 1999; Agius et al.,

2000). An interesting question is how maternal factors contribute

to zygotic transcription of nodal genes during developmental

course.

In Xenopus and zebrafish embryos, nodal gene expression in

the dorsal organizer is induced partially by maternally supplied

b-catenin (Kelly et al., 2000; Xanthos et al., 2002; Bellipanni

et al., 2006). Studies in Xenopus indicate that zygotic expression

of nodal genes is initiated in the entire vegetal endoderm, which

also requires other maternal factors such as the vegetally loca-

lized T-box transcriptional factor VegT (Agius et al., 2000;

Xanthos et al., 2002). However, fish or mammalian orthologs of

VegT remain to be verified or identified. In zebrafish embryos,

ndr1 and ndr2 are expressed during mid-to-late blastulation

period in the yolk syncytial layer (YSL), an extraembryonic syncyt-

ial layer of cytoplasm and nuclei formed by breakdown of

Received January 14, 2014. Revised April 23, 2014. Accepted April 29, 2014.# The Author (2014). Published by Oxford University Press on behalf of Journal of

Molecular Cell Biology, IBCB, SIBS, CAS. All rights reserved.

272 | Journal of Molecular Cell Biology (2014), 6(4), 272–285 doi:10.1093/jmcb/mju028

Published online June 12, 2014

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 2: J Mol Cell Biol-2014-Xu-272-85.pdf

blastomeres lying against the yolk cell at the midblastula transi-

tion, as well as in overlying blastodermal marginal cells that

contain mesoderm and endoderm precursors reside (Erter et al.,

1998; Feldman et al., 1998; Rebagliati et al., 1998; Sampath

et al., 1998; Fan et al., 2007). The expression of ndr1 and ndr2

in the ventrolateral blastodermal margin requires YSL-derived

signals (Chen and Kimelman, 2000) and their own transcripts

that are present in the YSL (Fan et al., 2007). Knockdown of

YSL-specific expression of mxtx2, which encodes a member of

the Mix/Bix transcription factor family and is zygotically

expressed (Hirata et al., 2000), inhibits ndr1 and ndr2 expression

in the ventrolateral margin of blastulas (Hong et al., 2011), indi-

cating that Mxtx2 is an important regulator of nodal gene expres-

sion. It is unknown whether in the zebrafish embryo there exist

maternal transcription factors that, like VegT in the Xenopus

embryo, play a role in mesendoderm induction by promoting

ndr1 and ndr2 expression.

Eomesodermin (Eomes) is a T-box transcription factor. Several

pieces of evidence suggest a role of Eomes in mesendodermal

development: mouse embryos deficient for zygotic Eomes show

abnormal mesoderm and endoderm tissue formation (Russ

et al., 2000; Arnold et al., 2008); zebrafish embryos overexpres-

sing eomesa increase the expression of some endodermal and

dorsal mesodermal markers (Bruce et al., 2003; Bjornson et al.,

2005); and interfering with Eomes in MZmid embryos carrying a

foxh1 mutation causes a loss of endodermal and nonaxial meso-

dermal tissues in addition to axial mesodermal defects (Slagle

et al., 2011). It is believed that Eomes may act downstream and

mediate function of Nodal signaling in mesodermal and endo-

dermal cell lineage development (Ryan et al., 1996; Brennan

et al., 2001; Bjornson et al., 2005; Slagle et al., 2011). However,

mouse Eomes and zebrafish eomesodermin a (eomesa) gene

are maternally expressed (Bruce et al., 2003; Bjornson et al.,

2005; McConnell et al., 2005), implying that maternally provided

Eomes may act upstream of Nodal signaling. Recently, the eome-

safh105 mutant line has been generated in zebrafish, which carries

a C to A mutation at position 300 to create a premature stop codon

(Du et al., 2012). MZeomesa mutant embryos, which lack both

maternal and zygotic Eomesa, display delayed epiboly initiation

with defects in endodermal gene expression, indicating a role in

epibolic process and endoderm specification (Du et al., 2012).

On the other hand, it remains inconclusive whether eomesa

plays a role in regulating nodal gene expression and mesoderm

specification.

In this study, we further demonstrate that maternal product

of eomesa is implicated in promoting ndr1 and ndr2 expression

in the ventrolateral margin during mesendoderm specification

in the zebrafish embryo. Mechanistically, maternal Eomesa binds

to the promoter or enhancer and activates zygotic expression of

ndr1 and ndr2 in cooperation with Mxtx2 that is also activated by

maternal Eomesa. Furthermore, we show that ectopic expression

of zebrafish eomesa in mouse embryonic stem cells (mESCs) can

enhance the expression of endogenous Nodal gene and promote

their differentiation into mesendoderm precursors. Therefore,

Eomes may act on the top of the regulatory hierarchy of

mesendoderm induction during embryogenesis.

Results

Loss-of-function of maternal Eomesa impairs mesendoderm

specification in zebrafish embryos

Before the eomesa mutant line became available, we developed

a strategy to knock down maternal eomesa by injecting eomesa-

MO into immature oocytes followed by in vitro oocyte maturation

and fertilization (oKD) (Supplementary Figure S1A), which was

modified from previously reported protocols (Gore et al., 2005;

Bontems et al., 2009; Nair et al., 2013). We showed that oKD was

able to effectively inhibit function of maternal oep gene product

(Supplementary Figure S1B) and to block the translation of the re-

porter eomesa-gfp mRNA during oocyte maturation (Supplementary

Figure S1C). As revealed by in situ hybridization, knockdown of

eomesa by oKD led to missing of the pan-mesodermal marker

ntla and the endodermal marker sox32 in some ventrolateral blas-

todermal marginal areas but retaining of their expression in the

dorsal margin at shield and 30% epiboly stages, whereas knock-

down of eomesa in embryos after fertilization (zKD) did not cause

obvious changes of these markers (Figure 1A). A marked reduction

for the dorsal mesodermal marker gsc during midgastrulation and

the ventral mesodermal marker eve1 at the shield stage was also

observed in oKD eomesa morphants (Figure 1B). Real-time RT–

PCR analyses confirmed the reduction of these marker expression

levels (Supplementary Figure S2A). These results suggest that ma-

ternal Eomesa is implicated in mesodermal and endodermal fate

specification.

A recent study in MZeomesa mutants has demonstrated import-

ant functions of maternal Eomesa in epiboly initiation and endo-

derm specification, but failed to draw a reliable conclusion about

its role in mesoderm specification (Du et al., 2012). We set to re-

examine the expression patterns of endodermal and mesodermal

markers in MZeomesa mutants in detail. We found, by in situ hy-

bridization, that the expression of sox32 was missing in the ventro-

lateral domains of the blastodermal margin in MZeomesa mutants

at the 30% epiboly stage and was still undetectable in some ventro-

lateral domains at the shield stage in mutants while its expression

occurred in the whole margin in wild-type (WT) control embryos at

these two stages (Figure 1C), which were consistent with the previ-

ous finding that sox32 expression was absent in the ventrolateral

margin of MZeomesa mutants at the 40% epiboly stage (Du

et al., 2012). We confirmed, by real-time RT–PCR analysis, that

MZeomesa mutants had reduced amounts of sox32 transcripts at

30% epiboly and shield stages compared with WT embryos

(Supplementary Figure S2B). Therefore, maternal Eomesa plays a

role in endoderm specification.

Our in situ hybridization results disclosed that, in WT embryos,

ntla was expressed in the whole blastodermal margin at 30%

epiboly and shield stages; in contrast, the majority of MZeomesa

mutants exhibited an absence of ntla expression in the ventrolat-

eral domains of the blastodermal margin at the 30% epiboly

stage, and they recovered ntla expression at the shield stage in

some but not all ventrolateral domains (Figure 1D). The reduction

of ntla expression in MZeomesa at these stages was verified by

Maternal Eomes for zygotic nodal gene expression | 273

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 3: J Mol Cell Biol-2014-Xu-272-85.pdf

real-time RT–PCR results (Supplementary Figure S2C). Du et al.

(2012) also noted that ntla was expressed in fewer tiers of blasto-

dermal marginal cells in MZeomesa mutants at the 50% epiboly

stage. These data together indicate that deficiency of maternal

Eomesa is likely to impair mesoderm specification, at least in

ventrolateral domains of the blastodermal margin.

We found that, compared with WT embryos, about half of

MZeomesa mutants at the 30% epiboly stage expressed the

dorsal mesoderm marker gsc in a narrower dorsal margin; at

the shield stage when WT embryos expressed gsc exclusively in

the shield at high levels, 28/43 of mutants had gsc-positive cells

in the shield as well as in the surrounding areas including the

dorsal margin, which might result from abnormal convergence

and extension during early gastrulation, and the remaining

portion (15/43) of mutants showed gsc expression only in the

shield but at lower levels (Figure 1E). Real-time RT–PCR analysis

indicated that the relative mRNA expression level of gsc in

mutants was significantly reduced compared with WT embryos

Figure 1 Deficiency of maternal Eomesa results in a partial loss of the mesendoderm markers ntla and sox32. (A and B) Mesendodermal marker

expression revealed by in situ hybridization following eomesa knockdown. Embryos treated differently were collected at morphologically match-

able stages. Stages for in situ: ntla and eve1, shield stage; sox32, 30% epiboly; gsc, midgastrulation. Orientation: animal-pole views for ntla,

sox32, and eve1 with dorsal to the right; dorsal views for gsc with animal pole to the top. Ctr, naturally fertilized, uninjected WT embryos; zKD,

embryos injected with 5 ng eomesa-MO at the one-cell stage; oCtr, embryos derived from oocytes maturated and fertilized in vitro; oKD,

embryos derived from oocytes injected with 5 ng eomesa-MO. (C–E) sox32 (C), ntla (D), and gsc (E) expression revealed by in situ hybridization

at 30% epiboly and shield stages. All embryos were in animal-pole view with dorsal to the right. MZeomesa, mutants lacking maternal and zygotic

Eomesa. (F) boz expression revealed by in situ hybridization at indicated stages. All embryos were laterally viewed with dorsal to the right. Mutant

embryos in C–F were fixed at time points when the control embryos were collected at indicated stages. The ratio of embryos with the representative

expression pattern is indicated. Scale bar, 200 mm. Corresponding real-time RT–PCR results are shown in Supplementary Figure S2.

274 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 4: J Mol Cell Biol-2014-Xu-272-85.pdf

(Supplementary Figure S2D). Our results support a previous

finding that eomesa knockdown in WT embryos led to a slight re-

duction of gsc and flh expression at the shield stage (Bruce et al.,

2003). However, Du et al. (2012) claimed, based on in situ hybrid-

ization results, that the dorsal mesodermal markers gsc and flh

were normally expressed in MZeomesa mutants at the 50%

epiboly stage. The discrepancy may lie in different developmental

stages of the examined embryos and variations of the gsc expres-

sion pattern among individuals.

To test whether abnormal expression of the mesendodermal

markers in MZeomesa mutants was due to a possible develop-

mental delay, we examined the expression of bozozok (boz), a

direct target of canonical Wnt signaling (Leung et al., 2003). Both

in situ hybridization and real-time RT–PCR analyses failed to

detect alterations of boz expression in MZeomesa embryos at

various stages (Figure 1F, and Supplementary Figure S2E). Thus,

defects in mesendoderm specification are unlikely to be caused

by any epibolic defects in MZeomesa embryos.

Eomesa acts upstream of Nodal signaling

A previous report demonstrated that injection of eomesa mRNA

into WT embryos induced ectopic expression of the dorsal meso-

dermal markers at the shield stage (Bruce et al., 2003). Given

that Nodal signaling plays an essential role in mesendoderm induc-

tion (Feldman et al., 1998; Gritsman et al., 1999), we wondered

whether the induction activity of Eomesa was dependent on

Nodal signaling. To address this issue, we compared the effect of

eomesa overexpression between WT and MZoep mutant embryos

devoid of Nodal signaling (Gritsman et al., 1999). Results showed

that overexpression of myc-eomesa mRNA induced ectopic expres-

sion of gsc, flh and chd in WT embryos, but failed to do so in MZoep

mutants (Figure 2). Therefore, we speculate that eomesa functions

upstream or parallel of Nodal signaling.

Deficiency of maternal Eomesa impairs nodal gene expression

during mesendodermal specification

To further investigate the relationship between Eomesa and

Nodal signaling, we first examined ndr1 and ndr2 expression

in oKD eomesa morphants. As shown in Figure 3A and B, their ex-

pression was restricted to the dorsal blastoderm margin of oKD

embryos at the dome stage while in the control embryos ndr1

and ndr2 were expressed throughout the blastodermal margin;

the reduced expression was still obvious at the 30% epiboly

stage. These preliminary results raised the possibility that nodal

gene expression was regulated by Eomesa.

Du et al. (2012) recently reported that ndr1 expression was

normal in MZeomesa mutants at the sphere stage but both ndr1

and ndr2 expressions were reduced in a small portion of mutants

at the 40% epiboly stage, suggesting that regulation of nodal

gene expression by maternal Eomesa is stage- or/and individual-

dependent. Then, we analyzed the time-course expression of

ndr1 and ndr2 in MZeomesa and WT embryos (Figure 3C–F). As

shown in Figure 3C, like WT embryos, MZeomesa mutants at the

high stage expressed ndr1 in a restricted dorsal margin; at the

oblong stage, WT embryos showed an expansion of ndr1 expres-

sion domain in the dorsal margin, but MZeomesa mutants had a

narrower ndr1 expression domain; from dome to 30% epiboly

stages, ndr1 was expressed in the entire blastodermal margin in

WT embryos, but its expression in MZeomesa mutants was still

restricted to the dorsal margin though laterally expanded; at the

40% epiboly stage, MZeomesa embryos had ndr1 expression in

the ventrolateral margin, resembling WT embryos; at the shield

stage, ndr1 expression was very weak in WT embryos, while its ex-

pression was retained throughout the blastodermal margin at high

levels in MZeomesa mutants; at the 60% epiboly stage, its expres-

sion in WT embryos was undetectable, but still occurred in a few

domains of the blastodermal margin in MZeomesa mutants. The

dynamic alterations of ndr1 expression were similarly observed in

Meomesa mutants devoid of maternal Eomesa only, implying

that ndr1 expression is regulated by maternal Eomesa. To quantify

the changes in ndr1 mRNA levels, we performed real-time RT–PCR

analysis. Results showed that, compared with WT embryos,

MZeomesa mutants had significantly reduced amounts of ndr1

transcripts from oblong to 30% epiboly stages, increased

Figure 2 Dependence of eomesa function on Nodal signaling. The expression of the dorsal markers gsc, flh, and chd was examined by in situ hy-

bridization at the shield stage. In WT embryos, injection of 50 pg myc-eomesa mRNA caused ectopic expression of the markers in some areas of the

blastodermal margin (indicated by arrows) (B, D, and F). In MZoep mutant embryos deficient for Nodal signaling, myc-eomesa mRNA (50 pg per

embryo) overexpression failed to induce ectopic expression of the markers (H, J, and L). All embryos were orientated in animal-pole view with dorsal

to the right. The ratio of embryos with the representative expression pattern is indicated. Scale bar, 200 mm.

Maternal Eomes for zygotic nodal gene expression | 275

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 5: J Mol Cell Biol-2014-Xu-272-85.pdf

Figure 3 Deficiency of maternal Eomesa reduces ndr1 and ndr2 expression during germ layer specification. (A and B) In situ hybridization patterns

of ndr1 (A) and ndr2 (B) in embryos derived from oocytes maturated and fertilized in vitro. Injected and uninjected embryos were harvested at

matchable stages based on morphology. oCtr, uninjected control embryos; eomesa oKD, embryos derived from oocytes injected with 5 ng

eomesa-MO. (C and E) In situ hybridization detection of ndr1 (C) and ndr2 (E) expression in WT control (Ctr), MZeomesa and Meomesa mutant

embryos. Mutant embryos were fixed at time points when the control embryos were collected at indicated stages. All embryos were shown in

animal-pole view with dorsal to the right. The ratio of embryos with representative pattern is indicated in the right corner of each image. Scale

bar, 200 mm. Note that weak expression of ndr2 at the oblong stage could be detected after staining for a longer time. (D and F) Relative expression

levels of ndr1 (D) and ndr2 (F) mRNAs in WT (Ctr) and MZeomesa embryos detected by real-time RT–PCR at indicated stages.

276 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 6: J Mol Cell Biol-2014-Xu-272-85.pdf

amounts at the shield stage, and comparable amounts at high and

40% epiboly stages (Figure 3D), which all were consistent with in

situ hybridization results. Therefore, we conclude that maternal

Eomesa may not be required for initiation of ndr1 expression in

the dorsal margin, but required for its timely activation in the

ventrolateral blastodermal margin.

An interesting question is why ndr1 expression, upon activated,

lasts a longer time in MZeomesa or Meomesa mutants (Figure 3C).

We hypothesize that maternal Eomesa may also positively regulate

the expression of Nodal signaling repressors. Then, we examined

expression of the Nodal antagonist genes lefty1 and lefty2 (Tian

and Meng, 2006). As shown in Supplementary Figure S3A, lefty1 ex-

pression appeared normal in MZeomesa mutants at the dome

stage, was absent in the ventrolateral margin at the 30% epiboly

stage and occurred in the whole blastodermal margin at the 40%

epiboly stage, which resembled the change of ndr1 expression in

mutants. At the shield stage, the majority of mutants showed a

much weaker expression of lefty1 in the dorsal margin, contrasting

retained strong expression in WT embryos. lefty2 expression

appeared to be absent in the ventrolateral margin in MZeomesa

mutants at all examined stages ranging from dome to shield

stages, which strikingly differed from its expression in the whole

blastodermal margin in WT embryos (Supplementary Figure S3B).

Thus, the up-regulated expression of ndr1 after the onset of gastru-

lation in MZeomesa mutants may be related, at least in part, to the

abnormal expression of lefty2 and lefty1.

The expression of ndr2 was also reduced in MZeomesa and

Meomesa mutant embryos, more strikingly in the ventrolateral

margin, from oblong to 30% epiboly stages (Figure 3E and F).

Like ndr1, ndr2 expression was recovered in the ventrolateral

margin in mutants at the 40% epiboly stage. It appeared that

ndr2 expression in the dorsal margin of mutants at 40% epiboly

and shield stages was stronger than in WT embryos, which might

be also related to the abnormal expression of lefty1 and lefty2.

We further tested the relatedness of ndr1 and ndr2 reduction in

MZeomesa mutants to loss of Eomesa by overexpressing synthetic

eomesa mRNA in mutants. We found that the missing expression of

ndr1 and ndr2 in ventrolateral margin of MZeomesa embryos could

be effectively rescued by injection of myc-eomesa mRNA

(Supplementary Figure S4).

Eomesa binds to and activates transcription of ndr1 and ndr2 loci

We next asked whether Eomesa directly regulated nodal gene

expression in zebrafish embryos. Based on the consensus Eomes-

binding motif (T/C)(C/A)(A/G)CAC(C/T)(T/C) identified in Xenopus

embryos by Conlon et al. (2001), we identified two putative

Eomes-binding sites, EBS1ndr1 (from 2258 to 2251) and EBS2

ndr1

(from 299 to 292), in the proximal promoter of the ndr1 locus

(Figure 4A). Chromatin immunoprecipitation (ChIP) analysis dis-

closed that overexpressed Myc-Eomesa bound to the DNA region

containing these two sites in embryos at the 30% epiboly stage

(Figure 4B). As reported before (Fan et al., 2007), a distal enhancer

(a) plus proximal promoter (p) of ndr1 in the construct pndr1ap:gfp

could drive gfp expression in the blastodermal margin at 30%–40%

epiboly stages (Figure 4C and D). When the EBS1ndr1 and EBS2

ndr1 in

the proximal promoter were individually or simultaneously mutated,

the percentage of GFP-positive embryos and the GFP intensity were

reduced (Figure 4D). These results imply that both Eomes-binding

sites are required for ndr1 expression. Using luciferase as a reporter

for testing ndr1 promoter/enhancer activity, we also showed that

injection of eomesa mRNA or eomesa-enR mRNA coding for a dom-

inant negative form of Eomesa (Bjornson et al., 2005) could increase

or reduce the luciferase expression level in WTembryos, respectively

(Figure 4E), and that the luciferase expression was inhibited by

mutations of EBS1ndr1 and EBS2

ndr1 (Figure 4F). Takingthese datato-

gether, we conclude that Eomesa activates ndr1 transcription de-

pendent on binding to the promoter.

We found that there is one putative Eomes-binding site (EBSndr2

at positions from 3090 to 3097) in the first intron of the ndr2 locus

(Figure 5A). ChIP assay demonstrated an association of overex-

pressed Myc-Eomesa with this site in fish embryos (Figure 5B).

When the EBSndr2-containing fragment derived from the first

intron of ndr2 was placed into pndr1apm1+2:gfp in which both

EBS1ndr1 and EBS2

ndr1 were mutated, an increasing proportion

of injected embryos could express GFP, and the mutation of the

EBSndr2 led to a relatively smaller proportion of GFP-positive

embryos (Figure 5C). These results indicate that the EBSndr2 pos-

sesses an enhancer activity. We made luciferase reporter con-

structs pEBSndr2GL3 and pInndr2GL3 by inserting an EBSndr2-

containing fragment from intron 1 of ndr2 and a control fragment

without EBS from intron 2 of ndr2 into the vector pGL3, respectively

(Figure 5D). The construct pmEBSndr2GL3 was derived from

pEBSndr2GL3 but contained the mutated EBSndr2. These constructs

were injected into embryos and the relative luciferase activity was

measured. Compared with pEBSndr2GL3 injection, pmEBSndr2GL3

injection resulted in a significantly lower level of the luciferase ac-

tivity (Figure 5E). Co-injection of myc-eomesa or eomesa-enR

mRNA with pEBSndr2GL3 caused a marked increase or reduction

of the luciferase activity, respectively; however, myc-eomesa or

eomesa-enR mRNA injection had no effect on the luciferase activity

expressed by pIn2ndr2GL3 (Figure 5F). These results further con-

firmed the responsiveness of the EBSndr2 to Eomesa.

Maternal eomesa directly activates transcription of the Nodal

upstream activator mxtx2

The expression of mxtx2 occurs in blastodermal cells at and

before the oblong stage and then in both marginal blastoderm

and YSL at the sphere stage, but is restricted to the YSL at the

dome stage (Hirata et al., 2000). Mxtx2 is reported to regulate

ndr1 and ndr2 expression in the ventrolateral margin of the blasto-

derm during mesendoderm induction (Hong et al., 2011). We found

that knockdown of maternal Eomesa led to partial or complete loss

of mxtx2 expression at the dome stage (Figure 6A). We confirmed

the previous finding that mxtx2 expression was temporally inhibited

in MZeomesa mutants (Du et al., 2012), and disclosed the same

changes in Meomesa mutants (Figure 6A and B). Importantly, over-

expression of myc-eomesa mRNA in MZeomesa could efficiently

initiate mxtx2 expression as early as the dome stage (Figure 6C).

To test whether Eomesa directly regulates mxtx2 expression, we

isolated a 1-kb promoter region of mxtx2, which contains a putative

Maternal Eomes for zygotic nodal gene expression | 277

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 7: J Mol Cell Biol-2014-Xu-272-85.pdf

Eomes-binding site (EBSmxtx2, 2189AGGTGTGA2182) (Figure 6D),

and made a luciferase reporter by placing it upstream of the luc

coding sequence. The expression of this reporter in WT embryos

was enhanced by eomesa overexpression but inhibited by eomesa-

enR overexpression (Figure 6E, left panel). Furthermore, mutation

of the EBSmxtx2 in the reporter construct caused a reduction of the re-

porter expression (Figure 6E, right panel). ChIP assay demonstrated

an association of overexpressed Myc-Eomesa with endogenous

EBSmxtx2 element (Figure 6F). These data together support the idea

that maternal Eomesa activates mxtx2 transcription.

Eomesa and Mxtx2 cooperatively activate nodal gene expression

We hypothesized, based on the above findings and the previ-

ous report (Hong et al., 2011), that Eomesa regulated ndr1 and

ndr2 expression directly by binding to their promoters/enhan-

cers and indirectly through induction of Mxtx2. To further test

this hypothesis, we studied functional interaction between

Eomesa and Mxtx2 on nodal gene expression. Injection of

myc-eomesa mRNA alone into WT embryos could not cause

ectopic expression of ndr1 and ndr2, but its co-injection with

mxtx2 mRNA enhanced mxtx2-induced ectopic expression of

ndr1 and ndr2 in the animal-pole area (Figure 7A). The ectopic ex-

pression of the Nodal target genes ntla and sox32 in the animal-

pole area was also more efficiently induced by co-overexpression

of myc-eomesa and mxtx2 compared with mxtx2 overexpression

alone (Figure 7A). These results suggest that Eomesa and Mxtx2

cooperatively activate nodal gene expression during mesendo-

derm induction.

We next investigated epistatic interaction between Eomesa and

Mxtx2. As shown in Figure 7B, mxtx2 knockdown in WT or MZeomesa

embryos, using a specific antisense morpholino (mxtx2-MO) (Hong

et al., 2011), inhibited ndr1 and ndr2 expression in the ventrolateral

blastodermal margin at the 30% epiboly stage, which is consistent

with the previous finding by Hong et al. (2011). In contrast, mxtx2

knockdown in MZeomesa embryos did not cause a further reduction

of the ndr1 and ndr2 expression domains (Figure 7B), which could be

explained by the absence of mxtx2 transcripts in the ventrolateral

blastodermal margin of mutants (Figure 6B). Co-injection of

myc-eomesa mRNA with mxtx2-MO into WT or MZeomesa embryos

led to certain levels of recovery of ndr1 and ndr2 expression in the

ventrolateral margin compared with mxtx2-MO injection alone

(Figure 7B). These results indicate that Eomesa regulates ndr1 and

Figure 4 Eomesa directly regulates ndr1 transcription. (A) Illustration of the ndr1 locus with Eomes-binding sites (EBS) indicated. (B) ChIP-PCR

results show occupancy of EBS-containing region of ndr1 by Myc-Eomesa. Embryos injected with 50 pg of myc-eomesa mRNA were harvested

around the 30% epiboly stage for ChIP. The amplified EBS-containing region is indicated by arrows in A. (C) Illustration of GFP or luciferase reporter

constructs using promoter/enhancers of ndr1. (D2F) Expression of GFP (D) or luciferase (E and F) in embryos injected with different ndr1 con-

structs. GFP was observed or luciferase activity was assayed at 30%240% epiboly stage. WT, M1, M2, and M1+2 were different constructs har-

boring original EBSs, mutated EBS1ndr1, mutated EBS2

ndr1, and mutated EBS1ndr1 plus EBS2

ndr1, respectively. Injection doses: GFP reporter DNA,

80 pg; luciferase reporter DNA, 50 pg; myc-eomesa mRNA, 50 pg; eomesa-enR mRNA, 200 pg.

278 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 8: J Mol Cell Biol-2014-Xu-272-85.pdf

ndr2 expression in both Mxtx2-dependent and Mxtx2-independent

fashions.

Eomes promotes Nodal expression and mesendodermal

differentiation of murine embryonic stem cells

It has been reported that Eomes in human embryonic stem cells

(ESCs) is required for definitive endodermal differentiation induced

by extrinsic signals such as Activin, BMP4 and FGF2 (Teo et al.,

2011) and that Eomes overexpression in murine ESCs promotes

cardiac differentiation in the absence of Activin (van den Ameele

et al., 2012). However, the regulatory relationship between

Eomes and Nodal remains elusive during ESCs differentiation. To

investigate their relationship and to test whether the regulatory

mechanism of Nodal expression by Eomes is evolutionarily con-

served, we established a murine R1 ESCs line with stable expres-

sion of zebrafish eomesa fused to gfp, i.e. R1-eomesa. In ESC

culture medium with LIF, R1-eomesa cells expressed several stem-

ness and differentiation markers at levels comparable to R1-GFP

cells that were stably expressing GFP (Figure 8A), suggesting that

overexpression of eomesa is not sufficient to induce differentiation

of ESCs under self-renewal conditions. ESCs can form embryoid

body (EB) and autonomously differentiate into multiple lineages

in the absence of LIF (Fei et al., 2010). Using the EB differentiation

system, we found that, compared with the R1-GFP cells, the

Figure 5 Eomesa directly regulates ndr2 transcription. (A) Illustration of ndr2 locus with Eomes-binding sites (EBS) indicated. (B) ChIP-PCR results

show occupancy of EBSndr2-containing region by Myc-Eomesa. The amplified EBSndr2-containing region is indicated by arrows in A. See Figure 4B

for PCR result of the control, as the same batch of immunoprecipitated DNA pools was used. (C) A 994-bp EBSndr2-containing fragment shows the

enhancer activity. All three constructs were made based on pndr1ap:gfp (Figure 4C). After injection with 80 pg GFP reporter DNA at the one-cell

stage, GFP was observed at 30%240% epiboly stage. The ratios of embryos with different GFP intensities are shown in the bar graph. n, number of

observed embryos. (D) Illustration of luciferase reporter constructs. In1(EBSndr2) is a 994-bp EBSndr2-containing fragment derived from intron 1,

while In2 is a 961-bp fragment without any EBS derived from intron 2 of the ndr2 locus. In pmEBSndr2GL3, the EBSndr2 was mutated as shown in C.

(E and F) Relative luciferase activity expressed by different reporter constructs. WT embryos were injected at the one-cell stage with 80 pg reporter

DNA alone or together with 50 pg myc-eomesa or 200 pg eomesa-enR mRNA and harvested at 30%240% epiboly stage for luciferase assay.

Maternal Eomes for zygotic nodal gene expression | 279

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 9: J Mol Cell Biol-2014-Xu-272-85.pdf

R1-eomesa cells expressed significantly higher levels of

the mesendodermal markers Braychury (T ), GATA4 and Eomes,

but lower levels of the ectodermal marker Sox1 at Day 6 of differen-

tiation (Figure 8B), and the expression of the endodermal marker

Sox17 and the ectodermal marker FGF5 were also increased and

decreased, respectively, though not statistically significant.

These results indicate that eomesa overexpression causes prefer-

ential differentiation of ESCs toward the mesendodermal fates.

The R1-eomesa cells maintained higher levels of Nodal

expression than the R1-GFP cells during differentiation

(Figure 8C), implying that eomesa overexpression may directly

promote the expression of endogenous Nodal gene. We identified

a putative EBS (2168TCACACCT2161) in the promoter region of the

mouse Nodal locus. In the R1-eomesa cells, this EBS was bound

by overexpressed Eomesa-GFP (Figure 8D). The human NODAL

locus also contains a putative EBS in the promoter region

(2728TAACACCT2721). The EBS derived from either mouse or

human Nodal locus could act as an enhancer to promote GFP

Figure 6 Maternal Eomesa is involved in activation of mxtx2 expression. (A) eomesa knockdown in oocytes inhibited mxtx2 expression at the dome

stage. (B) mxtx2 expression was impaired in MZeomesa and Meomesa embryos. Mutant embryos were fixed at time points when the control

embryos were collected at indicated stages. (C) myc-eomesa overexpression rescued mxtx2 expression in MZeomesa mutants. MZeomesa

embryos were injected with 50 pg myc-eomesa mRNA at the one-cell stage and examined at indicated stages for mxtx2 expression by in situ hy-

bridization. Embryos were laterally viewed. The ratio of embryos with representative pattern is indicated. Scale bar, 200 mm. (D) Illustration of the

mxtx2 locus. (E) Luciferase reporter expression driven by the mxtx2 promoter harboring EBS. The EBS was mutated to AAGCGTGC in the mxtx2m-luc

construct. Embryos were injected with 50 pg mxtx2-luc plasmid DNA and 5 pg renila DNA, with 50 pg myc-eomesa or 200 pg eomesa-enR mRNA

co-injected when needed, and harvested at 30%240% epiboly stage for luciferase assay. (F) Occupancy of the EBS of mxtx2 by Myc-Eomesa in

embryos, as assayed by ChIP-PCR. The amplified EBSmxtx2-containing region indicated by arrows in D. See Figure 4B for ChIP-PCR result of the

control region, as the same batch of immunoprecipitated DNA pools was used.

280 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 10: J Mol Cell Biol-2014-Xu-272-85.pdf

expression when it was placed upstream of pndr1apm1+2:gfp

(Figure 8E). Therefore, the mouse or human Nodal gene may

require Eomes for activation during embryonic development.

Discussion

We have demonstrated in this study that maternal Eomesa is an

upstream regulator of zygotic ndr1 and ndr2 expression during

mesendoderm induction in the zebrafish embryo. We found that de-

ficiency of maternal Eomesa causes a failure of ndr1 and ndr2 ex-

pression in the ventral and lateral blastodermal margins from

mid- to late-blastula stages. This finding suggests that maternal

Eomesa is essential for timely activation of ndr1 and ndr2 in the

ventrolateral margin but other factors are required for activating

their expression in the dorsal margin. Given that ndr1 expression

in the dorsal margin is absent in ichabod/b-catenin 2 mutant

embryos (Kelly et al., 2000; Bellipanni et al., 2006), it is most

likely that the activation of ndr1 and ndr2 expression in the

dorsal margin is dependent on maternal Wnt/b-catenin signaling.

Based on our and others’ findings (Kelly et al., 2000; Dougan

et al., 2003; Bellipanni et al., 2006; Hong et al., 2011), we

propose a simplified model for transcription of nodal genes

during zebrafish mesendoderm induction (Supplementary Figure

S5). Briefly, maternal Eomesa activates zygotic mxtx2 expression;

in the ventrolateral margin, Eomesa and synthesized Mxtx2

cooperate to activate ndr1 and ndr2 expression; in the dorsal

margin, maternal b-catenin 2 activates ndr1 and ndr2 expression,

which may be enhanced by maternal Eomesa and zygotic Mxtx2;

Nodal proteins then transduce the signals intracellularly, ultimate-

ly inducing the expression of downstream mesendodermal genes.

It is worth noting that Mxtx2 has been reported to regulate ndr2 ex-

pression directly but may regulate ndr1 expression indirectly (Hong

et al., 2011). Nevertheless, maternal Eomesa contributes to mesen-

doderm induction in zebrafish embryos in a way similar to maternal

VegT in Xenopus embryos. The high-level expression of Eomes in

mouse eggs (McConnell et al., 2005) suggests its possible involve-

ment in activating zygotic Nodal gene expression during embryo-

genesis. It appears that the specification of mesendodermal

lineages during embryogenesis by the maternal T-box transcription

factors/Nodal signals is conserved across vertebrate species.

Our reporter and ChIP assays revealed that Eomesa may directly

regulate ndr1, ndr2 and mxtx2 expression. However, the missing

expression of these genes during mid- to late-blastulation can

be largely recovered at later stages (�40% epiboly stage)

(Figures 3 and 6), reflecting the complexity of underlying regula-

tory mechanisms. The possible explanations for the rescue of

ndr1 and ndr2 expression in the ventrolateral blastodermal

margin of MZeomesa mutants at late blastula stages may

include: (i) ventrolateral propagation of existing Nodal signals

Figure 7 Functional interaction of Eomesa with Mxtx2. (A) Eomesa and Mxtx2 cooperate to induce expression of nodal genes and their targets. WT

embryos at the one-cell stage were injected with 50 pg myc-eomesa or 30 pg mxtx2 mRNA alone or both and harvested at indicated stages for

examination of ndr1, ndr2, ntla, and sox32 expressions by in situ hybridization. The ratio of embryos with the representative pattern is indicated.

(B) Eomesa regulates nodal gene expression partially through Mxtx2. WT or MZeomesa mutant embryos at the one-cell stage were injected with

4 ng mxtx2-MO alone or together with 50 pg myc-eomesa mRNA and harvested at the 30% epiboly stage for examination of ndr1 and ndr2 expres-

sions by in situ hybridization. The recovered ndr1 and ndr2 expression domains in embryos co-injected with myc-eomesa mRNA and mxtx2-MO

were indicated by arrows (compared with embryos injected with mxtx2-MO alone). All embryos were orientated in animal-pole view with dorsal

to the right. The ratio of embryos with the representative pattern is indicated.

Maternal Eomes for zygotic nodal gene expression | 281

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 11: J Mol Cell Biol-2014-Xu-272-85.pdf

in the dorsal side, which involves positive autoregulation of Nodal

signaling; (ii) unknown activators of nodal genes, which may be

expressed or activated during late blastulation in the absence of

Eomesa; (iii) incomplete recovery of the expression of the Nodal

antagonists such as lefty1 and lefty2 (Supplementary Figure

S3). We noted that ndr2 expression in the dorsal margin of

MZeomesa and Meomesa mutants at 30% epiboly and shield

stages (Figure 3E) was stronger than in WT embryos, which con-

curred with stronger expression of mxtx2 expression in the

dorsal margin of mutants (Figure 6B). Since Mxtx2 is a direct regu-

lator of ndr2 (Hong et al., 2011), more abundant Mxtx2 in the

dorsal margin of mutants at late blastula stages may also

account for enhanced ndr2 expression.

The expression of mxtx2 was undetectable in mutants depleted

of maternal Eomesa before the 30% epiboly stage (Figure 6B), indi-

cating an essential role of maternal Eomesa in timely activation

of mxtx2 expression. At and after the 30% epiboly stage, mxtx2

started to be expressed in the dorsal YSL of mutants. There might

be unknown factors that are responsible for late activation of mxtx2

expression in the dorsal side in the absence of maternal Eomesa.

Previous studies have found that maternal Eomesa protein exist

throughout cleavage period in zebrafish embryos (Bruce et al.,

2003; Bjornson et al., 2005; Du et al., 2012). However, its target

genes mxtx2, ndr1 and ndr2 start to transcribe at midblastula

stages. Two mechanisms may account for this transcriptional

delay: (i) chromatin structures at those loci are not poised for

Figure 8 Overexpression of zebrafish eomesa in murine ESCs promotes Nodal expression and mesendodermal differentiation. (A–C) Gene expres-

sion levels detected by real-time RT–PCR analysis in murine R1 ESC line stably expressing Eomesa-GFP (R1-eomesa line) or GFP (R1-GFP line). (A)

Cells were cultured in the KSR-ESC medium without feeder cells but in the presence of LIF for 8 days. (B) EBs after 6-day culture in the absence of LIF.

Eom, endogenous mouse Eomes; G4, Gata4. (C) Expression of endogenous Nodal gene in EBs forming at different days in the absence of LIF. (D)

Occupancy of the EBS-containing region of the murine Nodal locus by Eomesa-GFP in R1-eomesa cells. Ctr was amplified from the 3′UTR of the

murine Nodal locus. (E) A human or mouse EBS-containing enhancer promoted GFP expression from an inactive, ndr1-based GFP reporter con-

struct. Two representative embryos with different GFP intensities are shown, with the ratios for each group in the bar graph.

282 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 12: J Mol Cell Biol-2014-Xu-272-85.pdf

transcription; (ii) cofactors required for their transcription are not

available at earlier stages. These mechanisms are worthy of investi-

gation through experiments in the future.

Materials and methods

Zebrafish lines

WT embryos were obtained from AB strain. The mutant line

eomesafh105 (Du et al., 2012) was obtained from the Zebrafish

International Resource Center and the line oeptz257 (Gritsman et al.,

1999) was a gift from Dr Alex Schier. Embryos derived from eome-

safh105/+ heterozygous intercrosses were raised to adulthood and

then homozygous female and male were identified by PCR genotyp-

ing. Homozygous eomesafh105/fh105 female were unable to naturally

produce eggs when mated to male. Therefore, eggs squeezed from

homozygous eomesafh105/fh105 female were in vitro fertilized by

sperms squeezed from eomesafh105/fh105 or WT male to produce

MZeomesa or Meomesa mutant embryos, respectively. Less than

10% of MZeomesa or Meomesa mutants could grow up to adulthood.

MZoep embryos were produced by crossing oeptz257/tz257 femalewith

oeptz257/tz257 male. Embryos were staged according to Kimmel et al.

(1995). Ethical approval was obtained from the Animal Care and Use

Committee of Tsinghua University.

In vitro oocyte maturation and fertilization

The procedures were modified from previous reports (Gore et al.,

2005; Seki et al., 2008; Bontems et al., 2009; Nair et al., 2013).

Briefly, ovaries taken out from sacrificed female were placed in

oocyte culture medium (OCM: 90% Leibovitz’s L-15 medium

(Gibco), pH9.0, 0.5 mg/ml BSA (Amresco)) at room temperature,

and stage III oocytes were chosen to culture in OCM with 1 mg/ml

17a-20b-dihydroxy-4 pregnen-3-one (DHP, Sigma-Aldrich) at 268C.

About 5 h later when proceeded to stage V, oocytes were defol-

licled manually and then fertilized in a fresh plate by adding

squeezed sperms. When needed, morpholino or mRNA was injected

into oocytes after first 2-h maturation when they proceeded close

to stage IV. Using this method, the fertilization rate was usually

10–15%, which might further drop if oocytes were injected.

Constructs, microinjection, and embryonic assays

mRNAs were synthesized in vitro using mMESSAGE mMACHINE

Kit (Ambion). Transgene constructs were generated by PCR-based

cloning. Sequences of the used cloning primers and morpholinos

(MO) were listed in Supplementary Tables S1 and S2. Unless other-

wise stated, DNA, MO, or mRNA was injected into one-cell stage

embryos. Whole-mount in situ hybridization was performed using

Digoxigenin-labeled RNA probes as usual. For real-time RT–PCR,

�15 embryos derived from oocytes matured and fertilized in vitro

or 50 embryos derived from natural fertilization were used to

extract total RNAs, and RT–PCR analysis was done as before

(Jia et al., 2009). Primers for RT–PCR were listed in Supplementary

Table S3. GFP in embryos were observed by fluorescence micros-

copy. For luciferase activity assays, 50 pg reporter DNA was coin-

jected with 5 pg renilla DNA (an internal control) and luciferase

activity was measured in embryos at 30%–40% epiboly stages as

before (Liu et al., 2013).

ChIP assay was performed as before (Liu et al., 2011; Jia et al.,

2012). In brief, �2000 embryos injected with myc-eomesa mRNA

were collected at the 30% epiboly stage and dechorionated. The

embryos were incubated for crosslinking in 1% formaldehyde for

15 min with occasional inversion at room temperature, followed

by adding 1/20 volume of 2.5 M glycine and incubating for 5 min

to stop crosslinking. After washed with pre-cooled PBS three

times, embryos were resuspended in 2 ml lysis buffer (10 mM

Tris–HCl pH8.0, 10 mM NaCl, 0.5% NP-40) for 15 min with gentle

rocking at 48C, and the lysate was precipitated after spinning

at 1000 rpm and resuspended in 1 ml of pre-cooled nuclei lysis

buffer (50 mM Tris–HCl pH8.0, 10 mM EDTA, 1% SDS). After incu-

bation for 10 min with gentle rocking at 48C, 6 ml pre-cooled IP di-

lution buffer (20 mM Tris–HCl pH 8.0, 150 mM NaCl, 2 mM EDTA,

0.01% SDS) was added and mixed well. The lysate was sonicated

using a sonifier to generate chromatin fragments of 200–1500 bp

in length with an enrichment of 200–500 bp fragments, and the

supernatant was collected after spinning at 14000 rpm for 10 min

at 48C. The chromatin solution was diluted by adding 3 ml IP dilution

buffer and 400 ml 10% Triton X-100. To 5 ml of the chromatin solu-

tion, 100 ml pre-equilibrated Protein A Sepharose beads was added

for pre-cleaning. Following incubation with rotation at 48C for

30 min and spin at 1000 rpm, the supernatant was collected. About

20 mg anti-Myc antibody or mouse IgG was added to the supernatant

and incubated overnight with rotation at 48C and the incubation was

extended for additional 2 h after addition of 100 ml pre-equilibrated

Protein A Sepharose beads. After several rounds of wash, DNA was

eluted from the beads and the elute was treated with proteinase K

and RNase A. Finally, the DNA was purified by phenol/chloroform

extractions. The purified DNA was used for PCR using specific

primers (Supplementary Table S4). The same batch of the immuno-

precipitated DNA pool was used for amplifying corresponding

regions of ndr1, ndr2 and mxtx2 loci as well as the control sequence

spanning the second intron and the third exon of ndr1 locus, which

does not contain Eomes-binding sites. The control region for ChIP

assay in murine ESCs is located in the 3′UTR of the Nodal locus.

Murine stem cell culture and differentiation

Zebrafish Eomesa with GFP tag was cloned into the lentiviral

vector p2K7 and the resulted construct was used to transfect

HEK293FT cells to make lentiviruses. The virus-containing super-

natant was used to infect murine R1 ES cells. The stable R1 cell

lines were established by selection with 250 mg/ml G418

(Invitrogen) for 5 days and maintained in normal ES culture

medium in the presence of feeder cells and 1000 units/ml LIF

(Millipore). For differentiation in embryoid bodies (EB), single

cells were plated and cultured in the feeder-free, KSR-containing

medium in the absence of LIF to allow generation of floating EBs

with cell differentiation (Fei et al., 2010). EB cells were collected

at different days for analyzing markers expression by RT–PCR.

Statistical analysis

Significance between the means was analyzed using Student’s

t-test. Significance levels were indicated by *(P , 0.05) and

**(P , 0.01).

Maternal Eomes for zygotic nodal gene expression | 283

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 13: J Mol Cell Biol-2014-Xu-272-85.pdf

Supplementary material

Supplementary material is available at Journal of Molecular Cell

Biology online.

Acknowledgements

We thank Drs Alex Schier and Susan Mango (Department of

Molecular and Cellular Biology, Harvard University, Cambridge,

MA, USA) for discussion and suggestions, Dr David Kimelman

(Department of Biochemistry, University of Washington, Seattle,

WA, USA) for myc-eomesa construct, and members of the Meng

lab for discussion and technical assistance.

Funding

This work was financially supported by grants from the Major

Science Research Programs of China (2011CB943800) and the

National Natural Science Foundation of China (31221064).

Conflict of interest: none declared.

ReferencesAgius, E., Oelgeschlager, M., Wessely, O., et al. (2000). Endodermal

Nodal-related signals and mesoderm induction in Xenopus. Development

127, 1173–1183.

Arnold, S.J., Hofmann, U.K., Bikoff, E.K., et al. (2008). Pivotal roles for

eomesodermin during axis formation, epithelium-to-mesenchyme tran-

sition and endoderm specification in the mouse. Development 135,

501 –511.

Bellipanni, G., Varga, M., Maegawa, S., et al. (2006). Essential and opposing

roles of zebrafish beta-catenins in the formation of dorsal axial structures

and neurectoderm. Development 133, 1299–1309.

Bjornson, C.R., Griffin, K.J., Farr, G.H., III, et al. (2005). Eomesodermin is a loca-

lized maternal determinant required for endoderm induction in zebrafish. Dev.

Cell 9, 523–533.

Bontems, F., Stein, A., Marlow, F., et al. (2009). Bucky ball organizes germ plasm

assembly in zebrafish. Curr. Biol. 19, 414–422.

Brennan, J., Lu, C.C., Norris, D.P., et al. (2001). Nodal signalling in the epiblast

patterns the early mouse embryo. Nature 411, 965–969.

Bruce, A.E., Howley, C., Zhou, Y., et al. (2003). The maternally expressed zebra-

fish T-box gene eomesodermin regulates organizer formation. Development

130, 5503–5517.

Chen, S., and Kimelman, D. (2000). The role of the yolk syncytial layer in germ

layer patterning in zebrafish. Development 127, 4681–4689.

Conlon, F.L., Barth, K.S., and Robertson, E.J. (1991). A novel retrovirally induced

embryonic lethal mutation in the mouse: assessment of the developmental

fate of embryonic stem cells homozygous for the 413.d proviral integration.

Development 111, 969–981.

Conlon, F.L., Lyons, K.M., Takaesu, N., et al. (1994). A primary requirement for

nodal in the formation and maintenance of the primitive streak in the

mouse. Development 120, 1919–1928.

Conlon, F.L., Fairclough, L., Price, B.M., et al. (2001). Determinants of T box

protein specificity. Development 128, 3749–3758.

Dougan, S.T., Warga, R.M., Kane, D.A., et al. (2003). The role of the zebrafish

nodal-related genes squint and cyclops in patterning of mesendoderm.

Development 130, 1837–1851.

Du, S., Draper, B.W., Mione, M., et al. (2012). Differential regulation of epiboly

initiation and progression by zebrafish Eomesodermin A. Dev. Biol. 362,

11–23.

Erter, C.E., Solnica-Krezel, L., and Wright, C.V. (1998). Zebrafish nodal-related 2

encodes an early mesendodermal inducer signaling from the extraembryonic

yolk syncytial layer. Dev. Biol. 204, 361–372.

Fan, X., Hagos, E.G., Xu, B., et al. (2007). Nodal signals mediate interactions

between the extra-embryonic and embryonic tissues in zebrafish. Dev. Biol.

310, 363–378.

Fei, T., Zhu, S., Xia, K., et al. (2010). Smad2 mediates Activin/Nodal signaling in

mesendoderm differentiation of mouse embryonic stem cells. Cell Res. 20,

1306–1318.

Feldman, B., Gates, M.A., Egan, E.S., et al. (1998). Zebrafish organizer develop-

ment and germ-layer formation require nodal-related signals. Nature 395,

181–185.

Gore, A.V., Maegawa, S., Cheong, A., et al. (2005). The zebrafish dorsal axis is

apparent at the four-cell stage. Nature 438, 1030–1035.

Gritsman, K., Zhang, J., Cheng, S., et al. (1999). The EGF-CFC protein one-eyed

pinhead is essential for nodal signaling. Cell 97, 121–132.

Hirata, T., Yamanaka, Y., Ryu, S.L., et al. (2000). Novel mix-family homeobox

genes in zebrafish and their differential regulation. Biochem. Biophys. Res.

Commun. 271, 603–609.

Hong, S.K., Jang, M.K., Brown, J.L., et al. (2011). Embryonic mesoderm and endo-

derm induction requires the actions of non-embryonic Nodal-related ligands

and Mxtx2. Development 138, 787–795.

Jia, S., Wu, D., Xing, C., et al. (2009). Smad2/3 activities are required for induc-

tion and patterning of the neuroectoderm in zebrafish. Dev. Biol. 333,

273–284.

Jia, S., Dai, F., Wu, D., et al. (2012). Protein phosphatase 4 cooperates with

Smads to promote BMP signaling in dorsoventral patterning of zebrafish

embryos. Dev. Cell 22, 1065–1078.

Jones, C.M., Kuehn, M.R., Hogan, B.L., et al. (1995). Nodal-related signals induce

axial mesoderm and dorsalize mesoderm during gastrulation. Development

121, 3651–3662.

Kelly, C., Chin, A.J., Leatherman, J.L., et al. (2000). Maternally controlled

(beta)-catenin-mediated signaling is required for organizer formation in the

zebrafish. Development 127, 3899–3911.

Kimmel, C.B., Ballard, W.W., Kimmel, S.R., et al. (1995). Stages of embryonic de-

velopment of the zebrafish. Dev. Dyn. 203, 253–310.

Kumari, P., Gilligan, P.C., Lim, S., et al. (2013). An essential role for maternal

control of Nodal signaling. Elife 2, e00683.

Leung, T., Soll, I., Arnold, S.J., et al. (2003). Direct binding of Lef1 to sites in the

boz promoter may mediate pre-midblastula-transition activation of boz ex-

pression. Dev. Dyn. 228, 424–432.

Lim, S., Kumari, P., Gilligan, P., et al. (2012). Dorsal activity of maternal squint is

mediated by a non-coding function of the RNA. Development 139, 2903–2915.

Liu, Z., Lin, X., Cai, Z., et al. (2011). Global identification of SMAD2 target genes

reveals a role for multiple co-regulatory factors in zebrafish early gastrulas.

J. Biol. Chem. 286, 28520–28532.

Liu, X., Xiong, C., Jia, S., et al. (2013). Araf kinase antagonizes Nodal-Smad2 ac-

tivity in mesendoderm development by directly phosphorylating the Smad2

linker region. Nat. Commun. 4, 1728.

McConnell, J., Petrie, L., Stennard, F., et al. (2005). Eomesodermin is expressed in

mouse oocytes and pre-implantation embryos. Mol. Reprod. Dev. 71, 399–404.

Nair, S., Lindeman, R.E., and Pelegri, F. (2013). In vitro oocyte culture-based ma-

nipulation of zebrafish maternal genes. Dev. Dyn. 242, 44–52.

Osada, S.I., and Wright, C.V. (1999). Xenopus nodal-related signaling is essential

for mesendodermal patterning during early embryogenesis. Development

126, 3229–3240.

Rebagliati, M.R., Toyama, R., Fricke, C., et al. (1998). Zebrafish nodal-related

genes are implicated in axial patterning and establishing left-right asymmetry.

Dev. Biol. 199, 261–272.

Russ, A.P., Wattler, S., Colledge, W.H., et al. (2000). Eomesodermin is required

for mouse trophoblast development and mesoderm formation. Nature 404,

95–99.

Ryan, K., Garrett, N., Mitchell, A., et al. (1996). Eomesodermin, a key early gene in

Xenopus mesoderm differentiation. Cell 87, 989–1000.

Sampath, K., Rubinstein, A.L., Cheng, A.M., et al. (1998). Induction of the zebra-

fish ventral brain and floorplate requires cyclops/nodal signalling. Nature

395, 185–189.

Seki, S., Kouya, T., Tsuchiya, R., et al. (2008). Development of a reliable in vitro

maturation system for zebrafish oocytes. Reproduction 135, 285–292.

Slagle, C.E., Aoki, T., and Burdine, R.D. (2011). Nodal-dependent mesendoderm

284 | Xu et al.

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from

Page 14: J Mol Cell Biol-2014-Xu-272-85.pdf

specification requires the combinatorial activities of FoxH1 and Eomesodermin.

PLoS Genet. 7, e1002072.

Tadros, W., and Lipshitz, H.D. (2009). The maternal-to-zygotic transition: a play

in two acts. Development 136, 3033–3042.

Teo, A.K., Arnold, S.J., Trotter, M.W., et al. (2011). Pluripotency factors regulate

definitive endoderm specification through eomesodermin. Genes Dev. 25,

238–250.

Tian, T., and Meng, A.M. (2006). Nodal signals pattern vertebrate embryos. Cell.

Mol. Life Sci. 63, 672–685.

van den Ameele, J., Tiberi, L., Bondue, A., et al. (2012). Eomesodermin induces

Mesp1 expression and cardiac differentiation from embryonic stem cells in

the absence of Activin. EMBO Rep. 13, 355–362.

Xanthos, J.B., Kofron, M., Tao, Q., et al. (2002). The roles of three signaling path-

ways in the formation and function of the Spemann Organizer. Development

129, 4027–4043.

Zhou, X., Sasaki, H., Lowe, L., et al. (1993). Nodal is a novel TGF-beta-like

gene expressed in the mouse node during gastrulation. Nature 361,

543–547.

Maternal Eomes for zygotic nodal gene expression | 285

by guest on October 14, 2014

http://jmcb.oxfordjournals.org/

Dow

nloaded from