The C. elegans MS blastomere, born at the 7-cell stage of embryogenesis, generates primarily mesodermal cell types, including pharynx cells, body muscles and coelomocytes. A presumptive null mutation in the T-box factor gene tbx-35, a target of the MED-1 and MED-2 divergent GATA factors, was previously found to result in a profound decrease in the production of MS-derived tissues, although the tbx-35(-) embryonic arrest phenotype was variable. We report here that the NK-2 class homeobox gene ceh-51 is a direct target of TBX-35 and at least one other factor, and that CEH-51 and TBX-35 share functions. Embryos homozygous for a ceh-51 null mutation arrest as larvae with pharynx and muscle defects, although these tissues appear to be specified correctly. Loss of tbx-35 and ceh-51 together results in a synergistic phenotype resembling loss of med-1 and med-2. Overexpression of ceh-51 causes embryonic arrest and generation of ectopic body muscle and coelomocytes. Our data show that TBX-35 and CEH-51 have overlapping function in MS lineage development. As T-box regulators and NK-2 homeodomain factors are both important for heart development in Drosophila and vertebrates, our results suggest that these regulators function in a similar manner in C. elegans to specify a major precursor of mesoderm.

During metazoan development, embryonic cells must select from among multiple possible fates, and, ultimately, their descendants will produce gene products typical of a differentiated tissue. In the nematode C. elegans, early embryonic cells acquire transient, distinct identities after the zygote undergoes a series of asymmetrical cleavages. These form the six so-called `founder cells', each of which undergoes a stereotyped pattern of cell divisions to give rise to a nearly invariant set of descendants(Fig. 1A)(Sulston et al., 1983). The emergent paradigm of blastomere/lineage specification is that maternal factors first specify blastomere identity by zygotic activation of blastomere-specific factors, which ultimately leads to activation of tissue-specific gene networks(Labouesse and Mango, 1999; Lei et al., 2009; Maduro, 2009). Blastomere-specific factors are transiently expressed and act for a short time in development, whereas tissue-specific factors tend to maintain their expression throughout the lifespan. An understanding of how lineage-specific activation of tissue factors is achieved will close the gap between studies of blastomere fate and studies of tissue identity, generating a comprehensive gene network that describes development.

The 7-cell stage MS blastomere generates many mesodermal cell types,including cells of the pharynx and body musculature(Fig. 1A,C). The gene cascade that specifies MS has been studied for almost two decades(Fig. 1B). Initial specification of MS requires maternal activity of the bZIP/homeodomain factor SKN-1 (Bowerman et al., 1993; Bowerman et al., 1992). Loss of skn-1 leads to a lack of MS-derived tissues and a somewhat less penetrant loss of endoderm from E, the sister cell of MS(Bowerman et al., 1992). skn-1 mutants also lack the AB-derived portion of the pharynx owing to failure of a Notch/GLP-1-mediated induction from MS to the AB lineage(Priess et al., 1987; Shelton and Bowerman, 1996). In skn-1 mutants, mis-specified MS and E cells adopt the fate of the mesectodermal precursor C (Bowerman et al.,1992). In EMS (the mother of MS and E), SKN-1 activates the zygotic med-1 med-2 (med-1,2) divergent GATA factor gene pair (Coroian et al., 2005; Maduro et al., 2001). Loss of med-1,2 has a similar effect on MS specification as loss of skn-1, but a much weaker effect on E specification owing to parallel contributions to endoderm from SKN-1 and other factors(Goszczynski and McGhee, 2005; Maduro et al., 2005a; Maduro et al., 2001). In MS,MED-1,2 activate the T-box factor gene tbx-35(Broitman-Maduro et al., 2006). Loss of tbx-35 has variable effects on MS lineage development and morphogenesis, although the most severely affected mutants resemble skn-1 or med-1,2 embryos and lack most tissues made by MS(Broitman-Maduro et al.,2006).

The regulatory cascade initiated by SKN-1 works combinatorially with other factors that restrict MS fate to the appropriate blastomere. Within the EMS lineage, SKN-1 and its target genes collaborate with the Wnt/β-catenin asymmetry pathway to distinguish MS and E identity(Maduro et al., 2002; Rocheleau et al., 1997; Shetty et al., 2005; Thorpe et al., 1997). EMS receives an induction from its posterior sister P2 that ultimately results in differential nucleocytoplasmic localization of the nuclear effector TCF/POP-1 within MS and E, referred to as POP-1 asymmetry(Goldstein, 1992; Lin et al., 1998; Lo et al., 2004; Maduro et al., 2005a; Rocheleau et al., 1999). Within the E cell, reduced nuclear POP-1 permits POP-1 to function as an endoderm activator through association with the divergent β-catenin SYS-1(Huang et al., 2007; Phillips et al., 2007). Blockage of the induction, or of the components that act upstream of POP-1,results in EMS dividing to produce two MS-like cells(Goldstein, 1992; Rocheleau et al., 1997; Rocheleau et al., 1999; Thorpe et al., 1997). Outside of the EMS lineage, multiple factors block inappropriate expression of SKN-1 or prevent its timely degradation, either of which can otherwise lead to ectopic mis-specification of MS or E fates(Lin, 2003; Mello et al., 1992; Page et al., 2007; Shirayama et al., 2006).

Fig. 1.

Developmental context of the MS lineage and its gene regulatory network. (A) Partial cell lineage showing the production of major tissue types (number of cells in brackets) from early blastomeres(Sulston et al., 1983). The MS lineage is expanded to show the origin of pharynx, muscle and coelomocytes.(B) Gene regulatory network for MS specification [modified with permission from Maduro (Maduro,2009)]. (C) Embryo stages. Blastomeres are indicated on the 8-cell stage embryo. In the 1.5-fold embryo, all pharynx nuclei, and body muscle nuclei of the left half of the embryo, are shown. Darker-shaded nuclei are those derived from MS. The left-side embryonic coelomocytes (cc) are shown as circles with an X. For the L1 larva, tissues are indicated along with their blastomere of origin. A C. elegans embryo is ∼50 μm long. Here and in subsequent figures, anterior is to the left and dorsal is up.

Fig. 1.

Developmental context of the MS lineage and its gene regulatory network. (A) Partial cell lineage showing the production of major tissue types (number of cells in brackets) from early blastomeres(Sulston et al., 1983). The MS lineage is expanded to show the origin of pharynx, muscle and coelomocytes.(B) Gene regulatory network for MS specification [modified with permission from Maduro (Maduro,2009)]. (C) Embryo stages. Blastomeres are indicated on the 8-cell stage embryo. In the 1.5-fold embryo, all pharynx nuclei, and body muscle nuclei of the left half of the embryo, are shown. Darker-shaded nuclei are those derived from MS. The left-side embryonic coelomocytes (cc) are shown as circles with an X. For the L1 larva, tissues are indicated along with their blastomere of origin. A C. elegans embryo is ∼50 μm long. Here and in subsequent figures, anterior is to the left and dorsal is up.

The organ-identity factors that specify the two major tissues made by MS,pharynx and muscle, have been well characterized. Pharynx is specified by FoxA/PHA-4 (Horner et al.,1998; Kalb et al.,1998), which is at the top of a network of at least several hundred genes (Gaudet and Mango,2002) that includes the pharynx muscle-specific gene ceh-22 (Okkema and Fire,1994). Body muscle is specified by the activity of three regulators, MyoD/HLH-1, HAND/HND-1 and SRF/UNC-120(Fukushige et al., 2006). All three genes have overlapping function, as each can specify muscle fate when overexpressed and muscle specification is blocked only when the activity of all three has been compromised (Fukushige et al., 2006). Approximately 1300 genes are known to be enriched for expression in muscle (Fox et al.,2007), suggesting that HLH-1, HND-1 and UNC-120 are at the top of a complex tissue-specific muscle gene network.

In the present study we identify the NK-2 homeobox gene ceh-51 as a direct target of TBX-35, and present evidence that CEH-51 and TBX-35 have distinct and shared functions. Whereas loss of ceh-51 function causes subtle muscle and pharynx defects and larval lethality, simultaneous loss of ceh-51 and tbx-35 results in a highly penetrant loss of MS-derived tissues and an embryonic arrest phenotype that is strikingly similar to that of med-1,2(-) embryos, thus explaining the weaker phenotype of single tbx-35 mutants. Our results add an important regulator, CEH-51, to the MS gene regulatory network, and suggest that combinatorial control of mesoderm through T-box and NK-2 factors has been evolutionarily conserved.

Strains used

C. elegans animals were cultured on E. coli OP50 using standard methods (Sulston and Hodgkin,1988). The wild-type strain was N2. Mutations: LG X: hnd-1(q740), med-1(ok804). LG I: unc-120(st364). LG II: tbx-35(tm1789), hlh-1(cc561ts). LG III: unc-119(ed4),med-2(cx9744). LG IV: skn-1(zu67). LG V: ceh-51(tm2123). Rearrangement: nT1 [unc-?(n754)let-?](IV;V). Transgenes: gvIs401 V [unc-120::GFP], gvIs402 I [unc-120::GFP], cuIs1 V[ceh-22::GFP], ccIs7963 V [hlh-1::GFP], qIs55 [hnd-1::GFP], irIs57 III[hs-ceh-51], irIs70 [hs-ceh-51], cdIs41 II[cup-4::GFP], cdIs42 I [cup-4::GFP], ruIs37 III [myo-2::GFP], pxIs[pha-4::GFP] IV, irIs39 III [ceh-51::GFP], irIs41 [ceh-51::GFP], irIs42 X [hs-tbx-35], irIs58 [hs-ceh-51], irIs89 [ceh-51(+)], qtIs9 [nhr-25::YFP]. We have previously observed a lack of strict additivity and variability in the number of cells expressing tissue-specific reporters(Lin et al., 2009). We attribute this primarily to expression mosaicism between animals and the difficulty of resolving adjacent cells.

Identification of ceh-51

Y80D3A.3 (previously dlx-1) was named ceh-51 in consultation with Thomas Burglin and Jonathan Hodgkin (Karolinska Institute,Stockholm, Sweden and University of Oxford, Oxford, UK). ceh-51resides within intron 12 of Y80D3A.2/emb-4 (WormBase, WS200 release). Four ESTs support a single transcript with one intron for ceh-51 that does not overlap emb-4 exonic sequences(Kohara, 2001). As RNAi targeted to introns does not affect mature transcripts(Fire et al., 1998), it is unlikely that RNAi targeted to ceh-51 would affect transcripts of emb-4. Indeed, RNAi of emb-4 results in embryonic lethality(Katic and Greenwald, 2006),not larval arrest (see text).

Construction of ceh-51(tm2123) strains

We injected a heterozygous ceh-51(tm2123) strain (a gift from Shohei Mitani, National Bioresource Project, Japan) with overlapping genomic PCR products spanning the ceh-51 locus (but lacking any exonic emb-4 sequences; primer sites are shown in Fig. 2) and an unc-119::CFP reporter (pMM809) to produce MS1206, a line that segregated arrested larvae and unc-119::CFP(+) viables. We confirmed the correct splicing of emb-4 in the tm2123 strain by RT-PCR. After backcrossing, the array was replaced with another carrying ceh-51(+), unc-119::mCherry (pMM824) and myo-2::mCherry(pCFJ90) for the muscle phenotype synergy experiments. PCR confirmed homozygosity of the tm2123 deletion in this strain. A spontaneous integrant of a ceh-51(+) array, irIs89, showed that 96%(n=253) of ceh-51(tm2123); irIs89 embryos were rescued to full viability. A tbx-35(tm1789); ceh-51(tm2123) double mutant strain was made by crossing tbx-35; Ex[tbx-35(+), unc-119::YFP]males to ceh-51; Ex[ceh-51(+), unc-119::CFP] hermaphrodites,and identifying YFP/CFP-expressing F2 animals that gave arrested embryos/larvae and in which all viable animals expressed both YFP and CFP. The two arrays in MS1275 were replaced by a single array marked with unc-119::mCherry (pMM824) or sur-5::dsRed (pAS152).

Cloning and transgenics

To construct ceh-51::GFP (pGB196), a PCR product containing 788 bp upstream of the ceh-51 start codon and 204 bp of the coding region was cloned into the SphI-BamHI sites in pPD95.67. A smaller reporter, with 187 bp of upstream DNA and 5 bp of coding region, was cloned similarly (pWH270). TBX-35 sites were mutated into restriction sites by PCR in pWH270. A translational fusion was constructed by combining 358 bp of ceh-51 promoter, a GFP coding region from pPD95.67 and the genomic region of ceh-51 containing the exons, intron and 3′UTR. A heat-shock ceh-51 construct was created by cloning the coding region,intron and 468 bp of the 3′UTR into pPD49.78. Further PCR and cloning details are available on request. Transgenics and integrants were made as described (Maduro et al.,2001).

RNAi experiments

For feeding-induced RNAi, L4 animals were fed for 36 hours on E. coli HT115 from the OpenBioSystems RNAi Library or transformed with clones made in pPD129.36. Adults were transferred to fresh plates for egg laying for 4-6 hours at 25°C. Embryos were allowed to develop for 12-24 hours prior to scoring. For dsRNA synthesis, PCR products carrying the T7 RNA polymerase recognition sequence at each end were amplified from N2 DNA, cDNA clones or the Ahringer Lab RNAi Library(Kamath and Ahringer, 2003). dsRNA was synthesized using the Ambion MEGAscript T7 Kit and microinjected into late L4 worms or young adults as described(Ahringer, 2006). Injected animals were allowed to recover for 3-24 hours and transferred to fresh plates for egg laying.

In situ hybridization

Embryos were stained as described(Coroian et al., 2005). For pal-1 staining of med-1,2 and ceh-51; tbx-35embryos, a mixture of rescued and non-rescued embryos were stained, and the number of mutants was estimated from the array transmission frequency.

Phalloidin staining

Embryos or larvae were freeze-cracked on dry ice or frozen in liquid nitrogen, fixed in 4% formaldehyde and stained with Alexa Fluor 488-conjugated phalloidin (Molecular Probes, Eugene, OR, USA) as described(Shaham, 2006).

Laser ablation, microscopy and imaging

Laser ablations were performed as described(Lin et al., 2009). Animals were imaged on a Zeiss Axioplan using a Hamamatsu ORCA II digital camera, or on an Olympus BX-61 with a Canon 350D camera. For phalloidin-stained larvae, a Zeiss LSM510 confocal microscope was used (Microscopy and Imaging Core Facility, UC Riverside). Adobe Photoshop 7 and ImageJ v1.37 were used to adjust image brightness and generate overlays.

Heat-shock experiments

Embryos were heat shocked as a group for 30-45 minutes at 33°C while they were contained within hermaphrodite mothers, representing a developmental time interval of 0-3 hours. After heat shock, hermaphrodites were allowed to lay eggs for 3-4 hours. Embryos were allowed to develop for a further 6-12 hours before scoring. For in situ hybridizations after heat shock, mothers were left overnight at 15°C on plates without food.

Expression and purification of recombinant TBX-35

A cDNA fragment encoding amino acids 120-325, corresponding to the predicted TBX-35 DNA-binding domain, was cloned into the GST vector pGEX-4T-1(GE Healthcare) to generate pWH173. This was transformed into E. coliRosetta2 cells (Novagen), grown at 37°C to an OD of 0.3, and protein production was induced with 0.3 mM IPTG overnight at 25°C. Cells were resuspended in BugBuster HT (Novagen) with one tablet of Complete, Mini,EDTA-free Protease Inhibitor (Roche). Glutathione beads, swelled in phosphate-buffered saline (PBS), were added to the lysate for 1 hour. After three washes with PBS, the protein was eluted in 50 mM Tris-HCl (pH 7.5), 5 mM reduced glutathione, 80 mM NaCl, 0.03% Triton X-100, and desalted using a P6 column (BioRad). The protein was stored at -20°C in 50% glycerol with 1 mM DTT and 10 mM Tris (pH 7.5).

Gel shift and DNase I footprinting

EMSA probes were gel-purified PCR products generated with a 32P end-labeled primer and an unlabeled primer. The probes contained DNA corresponding to -187 bp to +5 bp relative to the ceh-51 ATG. Probes carrying mutated sites were amplified from the corresponding GFP reporters. Gel shift and DNase I footprinting were performed as previously described for MED-1 (Broitman-Maduro et al.,2005), except that 10 μM GST and 10, 25 and 50 μM GST::TBX-35(DBD) were used, 6% acrylamide gels were run, and complexes were treated with 0.5 units of DNase I (Epicentre) for 40 seconds prior to organic extraction. For competition arrays, complementary oligonucleotides were annealed at 95°C for 5 minutes, cooled for 15 minutes and added to reactions at a 50-fold excess.

Identification of CEH-51, a putative NK-2 class homeodomain transcription factor

Loss of med-1,2 leads to a highly expressive loss of MS-derived tissues, whereas loss of tbx-35 has a less expressive MS phenotype,especially at lower temperatures(Broitman-Maduro et al., 2006; Maduro et al., 2001) (this work), suggesting that an additional factor contributes to MS specification downstream of MED-1,2 (Broitman-Maduro et al., 2006). From embryonic transcriptome analyses(Baugh et al., 2005; Baugh et al., 2003), we identified Y80D3A.3 as a candidate early MS lineage gene. Transcripts were reported to accumulate when the MS lineage is undergoing its first divisions,and were reduced in mex-3(zu155); skn-1(RNAi) embryos, which do not correctly specify MS. In parallel, we identified Y80D3A.3 in an RNAi screen for enhancement of hlh-1(cc561ts) muscle defects (S.K. and P.W.S.,unpublished results).

The Y80D3A.3 gene encodes a putative homeodomain transcription factor,CEH-51 (Fig. 2). Of the 89 homeodomain proteins encoded by the C. elegans genome(Okkema and Krause, 2005),CEH-51 is most closely related to CEH-7(Kagoshima et al., 1999),CEH-24 (Harfe and Fire, 1998)and TAB-1 (CEH-29) [L. Carnell and M. Chalfie, unpublished data cited in Syntichaki and Tavernarakis (Syntichaki and Tavernarakis, 2004)], sharing 41-48% identity (57-58%similarity) within the homeodomain (Fig. 2B,C). The CEH-51 homeodomain is most closely related to those of NK-2 subfamily proteins, with which it shares 39-43% identity (59-62%similarity), although CEH-51 lacks the conserved tyrosine at position 54 of the homeodomain (asterisk in Fig. 2C) that is typical of NK-2 proteins(Harvey, 1996). The C. elegans pharynx muscle NK-2 factor CEH-22 is more closely related to other NK-2 family members, as it contains the conserved tyrosine and shares 85% identity (90% similarity) with Drosophila Vnd/NK-2 across the homeodomain. CEH-51 contains multiple serine residues in its N-terminus (16/50 residues), a feature noted for the N-termini of CEH-24(Harfe and Fire, 1998) and the endoderm-specifying END-1,3 GATA factors(Maduro et al., 2005b).

ceh-51 is expressed in the early MS lineage downstream of TBX-35

We confirmed that ceh-51 transcripts accumulate in the MS daughters and persist into the MS granddaughters, as observed in 91%(n=70) of embryos at the MS2 to MS4 stage(Fig. 3A,B). Similar expression was seen with a ceh-51::GFP transcriptional reporter carrying 788 bp of genomic DNA upstream of the predicted ATG(Fig. 3E), a GFP::CEH-51 translational fusion with 358 bp of upstream region(Fig. 3F), and from expression reported by others (Hunt-Newbury et al.,2007; Kohara,2001; Reece-Hoyes et al.,2007). As anticipated by the mis-specification of MS in skn-1 and med-1,2 mutant embryos(Bowerman et al., 1992; Maduro et al., 2001),expression of ceh-51::GFP was not observed in these backgrounds(Fig. 3G; data not shown). Conversely, ectopic ceh-51::GFP was observed in mex-1 and pie-1 RNAi backgrounds (Fig. 3I,J), in which additional MS-like cells are made from the AB and C lineages, respectively (Mello et al.,1992). We have previously found that tbx-35 is still expressed in MS in a pop-1(RNAi) background(Broitman-Maduro et al., 2006),even though in this background MS adopts an E-like fate(Lin et al., 1995). Unexpectedly, most pop-1(RNAi) embryos expressed ceh-51 in both the MS and E lineages (Fig. 3D,H).

Fig. 2.

Structure of ceh-51 and its gene product. (A)Location of ceh-51 and emb-4 exons on LG V. The locations of the mutant alleles tm2123 and tm2187 and the primer pairs A1/A2 and B1/B2 (used to generate overlapping PCR products for rescue of tm2123) are shown. A 3′UTR of ∼260 bases is predicted by EST yk51g7. Polyadenylation motifs of AATAAA and AATGAA(Hajarnavis et al., 2004) are found 40 bp and 260 bp, respectively, downstream of the stop codon. tm2123 is a 1610 bp deletion that includes the coding portion of exon 1 and part of exon 2, including the first six amino acids of the predicted homeodomain, and carries an additional 14 bp insertion. The remainder of the ceh-51 coding region in tm2123 lacks any in-frame ATG codons, suggesting that tm2123 is null. tm2187 is an intronic 540 bp deletion and was not studied. (B) Comparison of CEH-51 and other NK-2 proteins. Like all C. elegans NK-2 factors, CEH-51 lacks the Tinman (TN) and NK-2-specific (NK) domains that are found in many other NK-2 factors (Harvey,1996). Regions where at least 7/10 contiguous residues are serine are indicated by S. HD, homeodomain. (C) Homeodomain alignments. Identities with C. elegans CEH-51 are indicated by black boxes and similarities by gray boxes. A tyrosine residue found in NK-2 family members is indicated with an asterisk (Harvey,1996). Accession numbers: C. elegans (Ce) CEH-51,CAB60440; CEH-7, AAC36745; CEH-24, AAB81844; TAB-1 (CEH-29), AAA98021; CEH-22,NP_001076744; C. briggsae CBG20317, CAP37360; Oncorhynchus mykiss (Om) Nkx2.1b, BAD93686; Mus musculus (Mm) Nkx2.1,NP_033411; Nkx2.5, NP_032726; Drosophila melanogaster (Dm) Vnd,P22808.

Fig. 2.

Structure of ceh-51 and its gene product. (A)Location of ceh-51 and emb-4 exons on LG V. The locations of the mutant alleles tm2123 and tm2187 and the primer pairs A1/A2 and B1/B2 (used to generate overlapping PCR products for rescue of tm2123) are shown. A 3′UTR of ∼260 bases is predicted by EST yk51g7. Polyadenylation motifs of AATAAA and AATGAA(Hajarnavis et al., 2004) are found 40 bp and 260 bp, respectively, downstream of the stop codon. tm2123 is a 1610 bp deletion that includes the coding portion of exon 1 and part of exon 2, including the first six amino acids of the predicted homeodomain, and carries an additional 14 bp insertion. The remainder of the ceh-51 coding region in tm2123 lacks any in-frame ATG codons, suggesting that tm2123 is null. tm2187 is an intronic 540 bp deletion and was not studied. (B) Comparison of CEH-51 and other NK-2 proteins. Like all C. elegans NK-2 factors, CEH-51 lacks the Tinman (TN) and NK-2-specific (NK) domains that are found in many other NK-2 factors (Harvey,1996). Regions where at least 7/10 contiguous residues are serine are indicated by S. HD, homeodomain. (C) Homeodomain alignments. Identities with C. elegans CEH-51 are indicated by black boxes and similarities by gray boxes. A tyrosine residue found in NK-2 family members is indicated with an asterisk (Harvey,1996). Accession numbers: C. elegans (Ce) CEH-51,CAB60440; CEH-7, AAC36745; CEH-24, AAB81844; TAB-1 (CEH-29), AAA98021; CEH-22,NP_001076744; C. briggsae CBG20317, CAP37360; Oncorhynchus mykiss (Om) Nkx2.1b, BAD93686; Mus musculus (Mm) Nkx2.1,NP_033411; Nkx2.5, NP_032726; Drosophila melanogaster (Dm) Vnd,P22808.

The expression pattern of ceh-51 suggests that it is a direct target of TBX-35. Overexpression of TBX-35 was sufficient to cause ectopic ceh-51 activation (Fig. 3C), whereas weaker expression still occurred in approximately half of tbx-35(tm1789) mutants(Fig. 3K), demonstrating that TBX-35 is sufficient but not necessary for ceh-51 activation. In a tbx-35(tm1789); pop-1(RNAi) background, expression of ceh-51::GFP became undetectable(Fig. 3L), suggesting that activation of ceh-51 in a tbx-35 mutant background is POP-1-dependent.

To test for direct interaction of TBX-35 with ceh-51, we purified recombinant GST::TBX-35 DNA-binding domain (DBD) expressed in E. coli, and found that a 187 bp fragment of ceh-51 could be gel shifted (Fig. 4A, lanes 6-8). We identified four putative TBX-35 binding sites based on similarity to the consensus sequence for the founding T-box factor Brachyury(Kispert and Herrmann, 1993),and confirmed that they were protected in a DNase I footprinting assay(Fig. 4B). These regions define a consensus of RTSKCACCYNNYY(Fig. 4C), which matches 7/8 sites of the Brachyury half-site TCACACCT (matches underlined) (Kispert and Herrmann,1993). Hence, it is likely that TBX-35 binds DNA as a monomer,similar to mouse Tbx20 and Tbx5 (Ghosh et al., 2001; Macindoe et al.,2009; Stennard et al.,2003). A competitor oligonucleotide containing two of the candidate sites competed the shifts, whereas a competitor with both sites mutated did not (Fig. 4A, lanes 9-11), and all four sites appear to be important for TBX-35 binding(Fig. 4D). In vivo, a minimal ceh-51::GFP reporter carrying the four sites was expressed in the early MS lineage, its expression was abolished in a tbx-35(tm1789)background, and mutation of the sites resulted in a loss of expression(Fig. 4E). We conclude that TBX-35 directly activates ceh-51.

Fig. 3.

Expression of ceh-51. (A,B) ceh-51transcripts occur in (A) the MS daughter cells (MS2) and (B) in the MS granddaughters (MS4), as detected by in situ hybridization. The E daughters are outlined. Ninety-one percent (n=101) of embryos at this stage showed expression in MS2 or MS4 (nine embryos did not stain). (C) Ectopic expression of ceh-51 following heat shock of hs-tbx-35 embryos. (D) Eighty-six percent(n=44) of pop-1(RNAi) embryos showed ceh-51 mRNA in both the MS and E daughters. Two embryos showed normal expression and four embryos did not stain. (E) Embryos transgenic for a ceh-51::GFP transcriptional reporter with 788 bp of upstream sequence show expression at MS4 that persists in later MS descendants.(F) A translational ceh-51::GFP::CEH-51 fusion shows strong nuclear accumulation at MS8. (G) ceh-51::GFP is undetectable in med-1(ok804); med-2(cx9744) embryos (n=84).(H) Sixty-six percent (n=41) of pop-1(RNAi) embryos showed ceh-51::GFP in both the MS and E lineages (the remainder were similar to wild type). (I,J) mex-1(RNAi) (I) and pie-1(RNAi) (J) embryos displayed ectopic ceh-51::GFP in AB and C descendants. (K) In tbx-35(tm1789) embryos, the onset of ceh-51::GFP expression was undetectable (52%, n=89) or delayed until past the MS8 stage (48%) and at lower levels. The exposure in this image was 10-fold longer than that shown in E. (L) ceh-51::GFP was not detected in tbx-35(tm1789); pop-1(RNAi)embryos (n=49).

Fig. 3.

Expression of ceh-51. (A,B) ceh-51transcripts occur in (A) the MS daughter cells (MS2) and (B) in the MS granddaughters (MS4), as detected by in situ hybridization. The E daughters are outlined. Ninety-one percent (n=101) of embryos at this stage showed expression in MS2 or MS4 (nine embryos did not stain). (C) Ectopic expression of ceh-51 following heat shock of hs-tbx-35 embryos. (D) Eighty-six percent(n=44) of pop-1(RNAi) embryos showed ceh-51 mRNA in both the MS and E daughters. Two embryos showed normal expression and four embryos did not stain. (E) Embryos transgenic for a ceh-51::GFP transcriptional reporter with 788 bp of upstream sequence show expression at MS4 that persists in later MS descendants.(F) A translational ceh-51::GFP::CEH-51 fusion shows strong nuclear accumulation at MS8. (G) ceh-51::GFP is undetectable in med-1(ok804); med-2(cx9744) embryos (n=84).(H) Sixty-six percent (n=41) of pop-1(RNAi) embryos showed ceh-51::GFP in both the MS and E lineages (the remainder were similar to wild type). (I,J) mex-1(RNAi) (I) and pie-1(RNAi) (J) embryos displayed ectopic ceh-51::GFP in AB and C descendants. (K) In tbx-35(tm1789) embryos, the onset of ceh-51::GFP expression was undetectable (52%, n=89) or delayed until past the MS8 stage (48%) and at lower levels. The exposure in this image was 10-fold longer than that shown in E. (L) ceh-51::GFP was not detected in tbx-35(tm1789); pop-1(RNAi)embryos (n=49).

Overexpressed CEH-51 is sufficient to promote aspects of MS specification

We next assessed the ability of CEH-51 to specify the development of MS-derived cell types using a heat-shock (hs) ceh-51 transgene. Ninety-one percent (n=245) of heat shocked pregastrulation hs-ceh-51 embryos underwent arrest, whereas heat shock of wild types resulted in only 22% (n=243) embryonic arrest. We examined pharynx muscles with ceh-22::GFP (Okkema and Fire, 1994), using a skn-1(RNAi) background to eliminate MS-derived tissues and AB-derived pharynx(Bowerman et al., 1992). Among skn-1(RNAi); hs-ceh-51 embryos, we observed only a small number of ceh-22::GFP-positive cells following heat shock(Fig. 5F), and were unable to detect significant expression of the pharynx identity gene pha-4(Horner et al., 1998) or the pharyngeal myosin gene myo-2(Miller et al., 1986)(Fig. 5G; data not shown),suggesting that CEH-51 by itself has, at most, a weak ability to specify pharynx.

Next, we examined production of muscle in a skn-1(RNAi);pal-1(RNAi) background, which blocks specification of nearly all body muscles (Hunter and Kenyon,1996). In such embryos, hs-ceh-51 was sufficient to promote widespread muscle specification as scored by unc-120::GFP(Fukushige et al., 2006) and expression of the body muscle gene myo-3(Miller et al., 1986)(Fig. 5H,I). Hence, CEH-51 is sufficient to specify muscle cell fate.

We then examined production of the four embryonically derived coelomocytes,which arise fairly late in the MS lineage(Sulston et al., 1983), using cup-4::GFP (Patton et al.,2005). hs-ceh-51 was sufficient to cause specification of coelomocytes in a skn-1(RNAi) background, which by itself eliminates them (Table 1; Fig. 5E,F). We conclude that CEH-51 is sufficient to specify muscle and coelomocyte precursors. No attempt was made to optimize the time interval for CEH-51 responsiveness, although under the same conditions, overexpressed tbx-35 was able to cause specification of pharynx, muscle and coelomocytes(Broitman-Maduro et al., 2006)(data not shown).

Table 1.

MS-dependent tissues produced in wild-type and mutant embryos

GenotypePharynx cells(pha-4::GFP)Pharynx muscles(ceh-22::GFP)Muscle cells (hlh-1::GFP)Coelomocytes (cup-4::GFP)
Wild type 50.0±0.9 (21) 12.8±0.1 (37) 44.7±1.1 (20) 3.7±0.2 (105) 
skn-1 (RNAi) 4.8±0.4 (20) 0.0±0.0 (165) nd 0.15±0.04 (124) 
pal-1 (RNAi) 49.5±0.8 (10) 11.7±0.3 (12) 21.6±0.9 (13) 3.7±0.1 (103) 
pop-1 (RNAi) nd nd nd 0.0±0.0 (50) 
glp-1 (RNAi) 23.1±0.6 (15) 5.7±0.2 (38) nd nd 
tbx-35(tm1789) 15°C 40.6±1.2 (17) 5.9±0.3 (24) 37.3±1.6 (10) 3.8±0.2 (28) 
tbx-35(tm1789) 23°C 35.7±0.8 (16)** 5.2±0.2 (46)* 34.8±2.4 (10) 3.3±0.4 (20) 
tbx-35(tm1789); glp-1 (RNAi) 15°C 6.6±0.5 (23) 2.0±0.4 (26) nd nd 
tbx-35(tm1789); glp-1 (RNAi) 23°C 1.1±0.3 (32)** 1.0±0.2 (39)* nd nd 
tbx-35(tm1789); pal-1 (RNAi) 15°C 38.8±0.7 (15) 5.1±0.3 (14) 8.4±1.0 (17) 2.2±0.2 (47) 
tbx-35(tm1789); pal-1 (RNAi) 23°C 35.6±1.0 (14)* 4.7±0.3 (17) 5.7±0.5 (40)* 0.6±0.1 (49)** 
ceh-51 (tm2123) 47.8±0.9 (17) 9.2±0.2 (10) 42.4±1.4 (10) 2.1±0.1 (53) 
ceh-51 (tm2123); pal-1 (RNAi) nd nd 19.3±0.5 (11) 2.5±0.1 (84) 
med-1 (ok804); med-2(cx9744) 31.3±0.6 (26) 4.1±0.2 (32) 31.0±2.7 (10) 0.07±0.03 (34) 
ceh-51 (tm2123); tbx-35(tm1789) 30.2±0.5 (44) 4.4±0.2 (18) 30.1±1.0 (14) 0.19±0.04 (124)* 
med-1 (ok804); med-2(cx9744); glp-1 (RNAi) 1.4±0.4 (14) 0.3±0.1 (31) nd nd 
ceh-51 (tm2123); tbx-35(tm1789); glp-1 (RNAi) 1.9±0.5 (26) 0.5±0.1 (52) nd nd 
med-1 (ok804); med-2(cx9744); pal-1 (RNAi) nd nd 3.8±0.5 (13) nd 
ceh-51 (tm2123); tbx-35(tm1789); pal-1 (RNAi) nd nd 3.9±0.4 (15) nd 
GenotypePharynx cells(pha-4::GFP)Pharynx muscles(ceh-22::GFP)Muscle cells (hlh-1::GFP)Coelomocytes (cup-4::GFP)
Wild type 50.0±0.9 (21) 12.8±0.1 (37) 44.7±1.1 (20) 3.7±0.2 (105) 
skn-1 (RNAi) 4.8±0.4 (20) 0.0±0.0 (165) nd 0.15±0.04 (124) 
pal-1 (RNAi) 49.5±0.8 (10) 11.7±0.3 (12) 21.6±0.9 (13) 3.7±0.1 (103) 
pop-1 (RNAi) nd nd nd 0.0±0.0 (50) 
glp-1 (RNAi) 23.1±0.6 (15) 5.7±0.2 (38) nd nd 
tbx-35(tm1789) 15°C 40.6±1.2 (17) 5.9±0.3 (24) 37.3±1.6 (10) 3.8±0.2 (28) 
tbx-35(tm1789) 23°C 35.7±0.8 (16)** 5.2±0.2 (46)* 34.8±2.4 (10) 3.3±0.4 (20) 
tbx-35(tm1789); glp-1 (RNAi) 15°C 6.6±0.5 (23) 2.0±0.4 (26) nd nd 
tbx-35(tm1789); glp-1 (RNAi) 23°C 1.1±0.3 (32)** 1.0±0.2 (39)* nd nd 
tbx-35(tm1789); pal-1 (RNAi) 15°C 38.8±0.7 (15) 5.1±0.3 (14) 8.4±1.0 (17) 2.2±0.2 (47) 
tbx-35(tm1789); pal-1 (RNAi) 23°C 35.6±1.0 (14)* 4.7±0.3 (17) 5.7±0.5 (40)* 0.6±0.1 (49)** 
ceh-51 (tm2123) 47.8±0.9 (17) 9.2±0.2 (10) 42.4±1.4 (10) 2.1±0.1 (53) 
ceh-51 (tm2123); pal-1 (RNAi) nd nd 19.3±0.5 (11) 2.5±0.1 (84) 
med-1 (ok804); med-2(cx9744) 31.3±0.6 (26) 4.1±0.2 (32) 31.0±2.7 (10) 0.07±0.03 (34) 
ceh-51 (tm2123); tbx-35(tm1789) 30.2±0.5 (44) 4.4±0.2 (18) 30.1±1.0 (14) 0.19±0.04 (124)* 
med-1 (ok804); med-2(cx9744); glp-1 (RNAi) 1.4±0.4 (14) 0.3±0.1 (31) nd nd 
ceh-51 (tm2123); tbx-35(tm1789); glp-1 (RNAi) 1.9±0.5 (26) 0.5±0.1 (52) nd nd 
med-1 (ok804); med-2(cx9744); pal-1 (RNAi) nd nd 3.8±0.5 (13) nd 
ceh-51 (tm2123); tbx-35(tm1789); pal-1 (RNAi) nd nd 3.9±0.4 (15) nd 

Strains were grown at 20-23°C unless otherwise indicated. Data are shown as the mean ± s.e.m. *0.01<P<0.05, **P<0.01, Student's t-test, by comparison with the experiment immediately above. nd, not done.

Only pharynx expression of pha-4::GFP, anterior to the gut (when present), was scored.

The anatomy of the pharynx was considered in assigning expression to particular muscle cells.

Loss of ceh-51 function results in defects in MS-derived tissues

To evaluate the requirement for ceh-51 in MS specification, we examined ceh-51(RNAi) and ceh-51(tm2123) animals. Gonadal injection of ceh-51 dsRNA resulted in 47% (n=70) of progeny arresting as uncoordinated L1 larvae, whereas the remainder appeared normal(50%) or arrested as early embryos (3%). The putative null mutant, tm2123 (Fig. 2A),resulted in a fully penetrant recessive zygotic L1 arrest. This lethality could be rescued by a ceh-51(+) transgene (see Materials and methods).

We examined ceh-51 mutants for pharynx defects. ceh-51(tm2123) mutants had a poorly defined metacorpus and an incompletely developed grinder (Fig. 6A,D), and expression of the pharynx muscle reporter ceh-22::GFP (Okkema and Fire,1994) was observed both inside and outside of the pharynx basement membrane, suggesting defective pharynx integrity(Fig. 6B,E). We also observed detachment of the pharynx from the buccal cavity in 64% (n=56) of animals. Similar defects were apparent in ceh-51(RNAi) arrested larvae (data not shown). ceh-51(tm2123) mutants also had defects in the organization of actin filaments as detected by phalloidin staining(Fig. 6C,F). We scored production of all pharynx cells in ceh-51 mutants using a pha-4::GFP reporter (Horner et al., 1998), and found that the number of cells in ceh-51mutants (47.8±0.9, n=17) was similar to that in wild type(50.0±0.9, n=21, P=0.1). We compared the pharynx defects in ceh-51 mutants with those of tbx-35(tm1789)larvae produced at 15°C. Although such larvae also displayed grinder abnormalities and a defective terminal bulb, they showed a well-defined metacorpus and less disorganized muscle actin, although expression of ceh-22::GFP was often mosaic in posterior pharynx muscle cells(Fig. 6G-I). These results suggest that ceh-51 primarily affects pharynx development and not specification.

Fig. 4.

TBX-35 binds ceh-51. (A) GST::TBX-35(DBD) shifts a ceh-51 promoter fragment. Competitor oligonucleotides containing two binding sites competed the shift, whereas a mutant competitor did not.(B) DNase I footprinting of the ceh-51 promoter by GST::TBX-35(DBD) at four regions (boxed). The thin horizontal line is the join between two autoradiographs. (C) A provisional recognition sequence for TBX-35 (rendered by WebLogo, http://weblogo.berkeley.edu)resembles that of the Brachyury half-site(Kispert and Herrmann, 1993).(D) TBX-35-dependent shifting of a 187 bp fragment of the ceh-51 promoter as demonstrated with a subset of possible binding site mutants. (E) Expression of a minimal tbx-35-dependent ceh-51::GFP reporter in a wild-type background, in tbx-35(tm1789), and in a wild-type background in which the reporter has been mutated at two or more binding sites. At least two transgenic lines and 50 embryos were tested for each promoter.

Fig. 4.

TBX-35 binds ceh-51. (A) GST::TBX-35(DBD) shifts a ceh-51 promoter fragment. Competitor oligonucleotides containing two binding sites competed the shift, whereas a mutant competitor did not.(B) DNase I footprinting of the ceh-51 promoter by GST::TBX-35(DBD) at four regions (boxed). The thin horizontal line is the join between two autoradiographs. (C) A provisional recognition sequence for TBX-35 (rendered by WebLogo, http://weblogo.berkeley.edu)resembles that of the Brachyury half-site(Kispert and Herrmann, 1993).(D) TBX-35-dependent shifting of a 187 bp fragment of the ceh-51 promoter as demonstrated with a subset of possible binding site mutants. (E) Expression of a minimal tbx-35-dependent ceh-51::GFP reporter in a wild-type background, in tbx-35(tm1789), and in a wild-type background in which the reporter has been mutated at two or more binding sites. At least two transgenic lines and 50 embryos were tested for each promoter.

Next, we examined the production of body muscles using unc-120::GFP (Fukushige et al.,2006). Seventy-five percent (n=20) of ceh-51embryos at the 1.5-fold stage lacked proper expression in the anterior region of the embryo where MS-derived muscles are normally found(Fig. 7A,D)(Sulston et al., 1983). Mutants frequently displayed additional expression of unc-120::GFP displaced slightly to the posterior, suggesting that muscle cells might have migration defects (Fig. 7D). As Caudal/PAL-1 is required for nearly all non-MS body muscles(Hunter and Kenyon, 1996), we scored muscle cells produced in a pal-1(RNAi) background in wild type and ceh-51 mutants using hlh-1::GFP(Krause et al., 1994)(Table 1). ceh-51(tm2123);pal-1(RNAi) embryos made 19.3±0.5 (n=11) hlh-1::GFP cells, slightly less than pal-1(RNAi) alone(21.6±0.9, n=13, P=0.02). In the case of tbx-35(tm1789); pal-1(RNAi) at 20°C, the number of muscle cells was much lower (5.7±0.5, n=40, P=10-4).

We looked for further evidence of defects in muscle development by combining ceh-51(tm2123) or ceh-51(RNAi) with reduction in function of one of three factors, HND-1, HLH-1 and UNC-120, that together define the muscle fate in C. elegans(Fukushige et al., 2006). Individual loss of function results in mild impairments in muscle function,but their loss in combination causes a synergistic failure of muscle specification, resulting in a paralyzed, arrested 2-fold (Pat) phenotype(Fukushige et al., 2006; Williams and Waterston, 1994). Loss of ceh-51 synergistically enhanced the phenotypes of loss of hlh-1, hnd-1 or unc-120(Fig. 7G). Individually, RNAi for these factors produced less than 1% Pat embryos, but in a ceh-51(tm2123) background, 47% Pat resulted from hlh-1(RNAi)and ∼10% Pat from hnd-1(RNAi) or unc-120(RNAi) (e.g. Fig. 7E); a similar result was observed with ceh-51(RNAi) (Fig. 7F).

Finally, we observed a decrease in expression of the coelomocyte marker cup-4::GFP (Patton et al.,2005), from an average of 3.7±0.2 cells (n=105) in wild type to 2.1±0.1 (n=53) in ceh-51(tm2123)(Table 1), further suggesting that the development of MS descendants is compromised in mutants. Taken together, these results show that CEH-51 is required for the normal development of multiple MS tissue types.

Fig. 5.

Overexpression of CEH-51 promotes specification of MS-derived cell types. (A,F) A small number of cells expressing ceh-22::GFP are restored to skn-1(RNAi) embryos by hs-ceh-51. (B,G) Expression of the pharynx muscle gene myo-2 is largely absent in both skn-1(RNAi) and skn-1(RNAi); hs-ceh-51 embryos. The inset in B shows wild-type expression of myo-2. (C,H) Many hs-ceh-51 embryos display unc-120::GFP-expressing cells in a skn-1(RNAi); pal-1(RNAi) background, which depletes embryos of nearly all body muscles (Hunter and Kenyon,1996). (D,I) In a skn-1(zu67); pal-1(RNAi)background, heat shock of ceh-51 causes the generation of many cells expressing the muscle myosin gene myo-3. One hundred percent(n=79) of heat shocked non-transgenic embryos resembled those shown in D, whereas 53% (n=53) of heat shocked transgenics resembled those shown in I. (E,J) hs-ceh-51 embryos accumulate ectopic coelomocytes. (K-M) Bar charts summarizing the hs-ceh-51data.

Fig. 5.

Overexpression of CEH-51 promotes specification of MS-derived cell types. (A,F) A small number of cells expressing ceh-22::GFP are restored to skn-1(RNAi) embryos by hs-ceh-51. (B,G) Expression of the pharynx muscle gene myo-2 is largely absent in both skn-1(RNAi) and skn-1(RNAi); hs-ceh-51 embryos. The inset in B shows wild-type expression of myo-2. (C,H) Many hs-ceh-51 embryos display unc-120::GFP-expressing cells in a skn-1(RNAi); pal-1(RNAi) background, which depletes embryos of nearly all body muscles (Hunter and Kenyon,1996). (D,I) In a skn-1(zu67); pal-1(RNAi)background, heat shock of ceh-51 causes the generation of many cells expressing the muscle myosin gene myo-3. One hundred percent(n=79) of heat shocked non-transgenic embryos resembled those shown in D, whereas 53% (n=53) of heat shocked transgenics resembled those shown in I. (E,J) hs-ceh-51 embryos accumulate ectopic coelomocytes. (K-M) Bar charts summarizing the hs-ceh-51data.

TBX-35 and CEH-51 have overlapping function

Loss of med-1,2 results in an embryonic lethal phenotype in which arrested embryos elongate to between one and two times the length of the eggshell (Maduro et al., 2007; Maduro et al., 2001). By contrast, tbx-35 null mutants arrest with varying degrees of elongation, ranging from 1-fold to complete elongation and hatching(Broitman-Maduro et al., 2006). These results suggest that TBX-35 works with another factor. Two further observations support this notion. First, whereas med-1(ok804);med-2(cx9744) double mutants and skn-1(RNAi) embryos made less than 0.2 coelomocytes per embryo (Table 1; Fig. 8L), tbx-35(tm1789) embryos raised at 15°C made as many coelomocytes(3.8±0.2, n=28) as wild types (3.7±0.2, n=105, P>0.9). Second, tbx-35(tm1789) embryos achieved further elongation overall when raised at 15°C(Fig. 8P). This increased elongation correlated with an increase in production of MS-derived pharynx cells as scored by pha-4::GFP in a glp-1(RNAi) background,which eliminates AB-derived pharynx(Priess et al., 1987)(Table 1): tbx-35;glp-1(RNAi) embryos at 15°C made 6.6±0.5 pharynx cells(n=23), whereas at 23°C only 1.1±0.3 cells were made(n=32, P<10-11).

We hypothesized that tbx-35 and ceh-51 double mutants might show a stronger phenotype than either single mutant, given that ceh-51 is still activated in tbx-35(tm1789)(Fig. 3K). As shown in Fig. 8 and Table 1, ceh-51(tm2123);tbx-35(tm1789) double mutants displayed phenotypes that are indistinguishable from med-1(ok804); med-2(cx9744) (henceforth abbreviated as ceh-51; tbx-35 and med-1,2). First, ceh-51; tbx-35 double mutants displayed a strong embryonic arrest that is not temperature sensitive (P=0.48 for 15°C versus 20°C) (Fig. 8P) and which is comparable to that of med-1,2 double mutants at both temperatures(P=0.36 and P=0.43 for 15°C and 20°C, respectively). Second, development of MS-derived pharynx was eliminated in ceh-51;tbx-35 (Table 1; Fig. 8E,F), even at 15°C,at which single ceh-51 and tbx-35 mutants each displayed a partial grinder (Fig. 6D,G). Using glp-1(RNAi) to eliminate AB-derived pharynx, both med-1,2;glp-1(RNAi) and ceh-51; tbx-35; glp-1(RNAi) embryos made similarly low numbers of pharynx cells (less than two) as scored with pha-4::GFP or ceh-22::GFP (P=0.15 and P=0.3) (Table 1). Production of pal-1-independent body muscle cells was reduced in ceh-51; tbx-35; pal-1(RNAi) embryos to levels comparable to med-1,2; pal-1(RNAi) (P=0.9)(Table 1; Fig. 8H,I). Lastly, whereas single ceh-51 and tbx-35 mutants made reduced numbers of cup-4::GFP(+) cells, the double mutants displayed a synergistic reduction similar to that of a med-1,2 background (P=0.04)(Table 1; Fig. 8K,L).

MS adopts a C-like fate in med-1,2(RNAi) and skn-1 mutant embryos (Bowerman et al., 1992; Maduro et al., 2001), but this transformation is weaker in tbx-35 mutants as zygotic activation of pal-1 in the MS lineage, a marker of transformation of MS to C(Baugh et al., 2005), was detected in only ∼30% of embryos(Broitman-Maduro et al., 2006). We found that 75% (n=20) of med-1,2(-) and 60%(n=35) of ceh-51; tbx-35 embryos showed ectopic zygotic pal-1 mRNA in the early MS lineage (P>0.3)(Fig. 8N,O). We examined the fate of MS descendants in tbx-35; ceh-51 double mutants carrying a reporter fusion for nhr-25, a C-lineage gene that is expressed in hypodermal precursors and their descendants(Baugh et al., 2005), using a laser to ablate all other cells. Partial embryos resulting from isolated wild-type MS blastomeres failed to show significant nhr-25::YFP(n=3), whereas 9/9 MS blastomeres from tbx-35; ceh-51 double mutants, and 5/5 isolated C blastomeres from wild types, generated nhr-25::YFP descendants. Hence, ceh-51; tbx-35 embryos show a strong transformation of MS to C, suggesting that CEH-51 and TBX-35 together account for the majority of normal MS lineage development downstream of MED-1,2.

Fig. 6.

ceh-51 mutants and tbx-35 mutants raised at 15°C arrest as larvae with pharynx structural defects. Pharynxes were visualized by DIC optics (A,D,G), ceh-22::GFP expression (B,E,H)(Okkema and Fire, 1994) or phalloidin staining (C,F,I) (Franks et al., 2006). In the DIC panels, the lumen (arrows), grinder (gr),metacorpus (mc) and terminal bulb (tb) are indicated and the pharynx is outlined (dashed line). (A-C) Wild-type pharynx. (D-F) ceh-51(tm2123) pharynxes show lumen irregularities and an indistinct metacorpus (D). Protrusions accumulate GFP outside the pharynx, suggesting a defect in pharynx integrity (E). In F, phalloidin staining shows actin filament accumulations (large arrowheads), lumen abnormalities (small arrowheads) and an abnormal terminal bulb (arrow). (G-I) tbx-35(tm1789) raised at 15°C has a normal lumen but abnormal grinder (G). ceh-22::GFP expression (H) shows absence of expression of ceh-22::GFP in part of the posterior pharynx (arrowhead);contralateral expression in this region is likely to be in an MS-derived m7 muscle (Okkema and Fire, 1994; Sulston et al., 1983). In I,phalloidin staining shows some actin accumulations (arrowhead) and an abnormal terminal bulb (arrow).

Fig. 6.

ceh-51 mutants and tbx-35 mutants raised at 15°C arrest as larvae with pharynx structural defects. Pharynxes were visualized by DIC optics (A,D,G), ceh-22::GFP expression (B,E,H)(Okkema and Fire, 1994) or phalloidin staining (C,F,I) (Franks et al., 2006). In the DIC panels, the lumen (arrows), grinder (gr),metacorpus (mc) and terminal bulb (tb) are indicated and the pharynx is outlined (dashed line). (A-C) Wild-type pharynx. (D-F) ceh-51(tm2123) pharynxes show lumen irregularities and an indistinct metacorpus (D). Protrusions accumulate GFP outside the pharynx, suggesting a defect in pharynx integrity (E). In F, phalloidin staining shows actin filament accumulations (large arrowheads), lumen abnormalities (small arrowheads) and an abnormal terminal bulb (arrow). (G-I) tbx-35(tm1789) raised at 15°C has a normal lumen but abnormal grinder (G). ceh-22::GFP expression (H) shows absence of expression of ceh-22::GFP in part of the posterior pharynx (arrowhead);contralateral expression in this region is likely to be in an MS-derived m7 muscle (Okkema and Fire, 1994; Sulston et al., 1983). In I,phalloidin staining shows some actin accumulations (arrowhead) and an abnormal terminal bulb (arrow).

New regulatory interactions in the MS gene network

We have identified a new regulator, CEH-51, in MS specification. Our results suggest that TBX-35 and CEH-51 could participate in a `feed-forward'regulatory cascade (Lee et al.,2002), in which TBX-35 activates ceh-51, and both TBX-35 and CEH-51 activate common target genes in MS development. There is likely to be at least one other MS lineage activator of ceh-51 because a ceh-51::GFP reporter was still weakly expressed in a tbx-35null background (Fig. 3K). Whereas pal-1(RNAi) reduced coelomocyte production in tbx-35(tm1789) mutants (Table 1), there was no effect on ceh-51::GFP expression (data not shown). Instead, this activator appears to be downstream of POP-1 because simultaneous loss of pop-1 and tbx-35 resulted in loss of ceh-51::GFP expression (Fig. 3L). We also observed ectopic expression of ceh-51 in the early E lineage in pop-1(RNAi) embryos(Fig. 3D,H), suggesting that POP-1 might contribute to repression of ceh-51 in the E lineage. The observation that a tbx-35; pop-1 background abolishes all ceh-51::GFP expression suggests that ectopic TBX-35 is responsible for E lineage expression of ceh-51 in pop-1(RNAi). Although we failed to detect activation of tbx-35 in E in pop-1(-)embryos (Broitman-Maduro et al.,2006), such ectopic expression of tbx-35::GFP has been observed by others (P. Shetty and R. Lin, personal communication). We have recently shown that in the related nematode C. briggsae, POP-1 contributes positively to MS specification in parallel with SKN-1, and there is an apparent function for POP-1 in repression of the MS fate in E(Lin et al., 2009). Hence,these additional roles for POP-1 might be evolutionarily conserved.

Fig. 7.

Muscle defects in ceh-51(tm2123). (A,D) Loss of MS-derived unc-120::GFP expression (arrowheads) in ceh-51(D) as compared with wild type (A). Additional expression is indicated by small arrows. (B,C,E,F) Loss of ceh-51synergizes with partial muscle specification mutants to produce paralyzed,arrested 2-fold (Pat) embryos. Whereas more than 95% of hlh-1(cc561ts) mutants grown at 15°C (B), and more than 99% of ceh-51(tm2123) embryos (C), elongated to greater than 3-fold, between 47 and 49% of embryos produced by a combination of mutation of ceh-51with RNAi of hlh-1 (E), or vice versa (F), produced a synthetic Pat phenotype. (G) Summary of synthetic Pat phenotypes. Data are shown as the percentage of progeny arresting as embryos (%Emb)/percentage of progeny arresting as paralyzed, 2-fold (Pat) embryos (%Pat) (included in the Emb totals). Backgrounds have been shaded to indicate higher %Pat.

Fig. 7.

Muscle defects in ceh-51(tm2123). (A,D) Loss of MS-derived unc-120::GFP expression (arrowheads) in ceh-51(D) as compared with wild type (A). Additional expression is indicated by small arrows. (B,C,E,F) Loss of ceh-51synergizes with partial muscle specification mutants to produce paralyzed,arrested 2-fold (Pat) embryos. Whereas more than 95% of hlh-1(cc561ts) mutants grown at 15°C (B), and more than 99% of ceh-51(tm2123) embryos (C), elongated to greater than 3-fold, between 47 and 49% of embryos produced by a combination of mutation of ceh-51with RNAi of hlh-1 (E), or vice versa (F), produced a synthetic Pat phenotype. (G) Summary of synthetic Pat phenotypes. Data are shown as the percentage of progeny arresting as embryos (%Emb)/percentage of progeny arresting as paralyzed, 2-fold (Pat) embryos (%Pat) (included in the Emb totals). Backgrounds have been shaded to indicate higher %Pat.

Shared and distinct functions for CEH-51 and TBX-35

Although ceh-51(tm2123); tbx-35(tm1789) embryos have a synergistic phenotype compared with the single mutants, each gene has unique essential functions, as evidenced by their distinct phenotypes (Figs 6 and 7). Overexpressed CEH-51 was sufficient to promote specification of muscle and coelomocytes, but was apparently not as effective at promoting pharynx development(Fig. 5), whereas overexpressed TBX-35 could specify all three tissues efficiently(Broitman-Maduro et al., 2006)(data not shown). Conversely, ceh-51(tm2123) mutants had only mild defects in pharynx, muscle and coelomocytes, whereas tbx-35(tm1789)mutants had strong defects in pharynx and muscle at 20°C(Broitman-Maduro et al., 2006)(Table 1). At 15°C, ceh-51 is able to partially rescue these defects, resulting in a higher proportion of elongated animals and more normal specification of MS-derived tissues (Table 1). Hence, CEH-51 adds robustness to MS specification primarily at lower temperatures. In the future, identification of TBX-35 and CEH-51 target genes might explain the basis for their different activities, perhaps accounting for why CEH-51 does not rescue aspects of MS specification in tbx-35mutants at higher temperatures. We have identified putative TBX-35 binding sites in the promoters of hlh-1 and pha-4 (W.W.K.H. and M.F.M., unpublished), although we have not yet identified common targets for both TBX-35 and CEH-51.

Collaboration of T-box and NK-2 factors in mesoderm development

The apparent collaboration of TBX-35 and CEH-51 in C. elegansmesoderm development, downstream of MED-1,2, is highly reminiscent of the roles of related factors involved in cardiac development in other systems. In C. elegans, the pharynx is the structure that most closely resembles the heart, as it is a contractile pumping organ that expresses unique sets of myosins (Mango, 2007; Okkema et al., 1993). Expression of vertebrate Nkx2.5 is able to compensate for loss of ceh-22 in the C. elegans pharynx, suggesting a common evolutionary origin of heart and pharynx(Haun et al., 1998). Here, we have shown that TBX-35 and CEH-51 have both distinct and shared roles in pharynx progenitor specification and development. The Drosophila Nkx2.5 ortholog tinman is important for defining early domains that are restricted to forming heart, visceral muscle and some body muscles,as mutants have impairments in the development of these tissues(Azpiazu and Frasch, 1993; Bodmer, 1993). Activation of tinman in cardioblasts requires the T-box genes midline and H15 (Reim et al.,2005). In Xenopus, the T-box factor Tbx5 is expressed in heart precursors and is known to be essential for heart development(Horb and Thomsen, 1999). Similarly, Nkx2.5 is expressed in early cardioblasts(Lints et al., 1993) and plays an important role in heart patterning, as Nkx2.5 knockout mice show heart defects (Lyons et al.,1995). Finally, mouse Tbx5 and Nkx2.5 physically interact and collaborate with Gata4/5 in synergistic activation of cardiac genes(Bruneau et al., 2001; Hiroi et al., 2001; Stennard et al., 2003). Hence,the collaboration between TBX-35 and CEH-51 in C. elegans might be evolutionarily conserved. Future work aimed at elucidating the gene network downstream of TBX-35 and CEH-51 might uncover further conserved aspects of cardiac and mesoderm development.

Fig. 8.

Mutation of ceh-51 and tbx-35 together synergizes to a med-1,2(-) arrest phenotype. (A-C) Pharynx muscles marked by ceh-22::GFP (Okkema and Fire,1994) overlaid on DIC images. (A) Arrested 1.5-fold tbx-35(tm1789) embryo raised at 20°C showing AB-derived and MS-derived pharynx muscles. (B) ceh-51(tm2123); tbx-35(tm1789) double mutant arrested at ∼1.5-fold elongation. (C) med-1(ok804);med-2(cx9744) double mutant. (D-F) Polarized light images to show gut granules overlaid with pha-4::GFP(Horner et al., 1998). (D) At 15°C, tbx-35(tm1789); glp-1(RNAi) embryos display 6.6±0.5(n=23) pharynx cells. Some additional GFP-positive cells are seen(arrows); similar `stray' GFP expression is also seen in a skn-1(RNAi) background (see Table 1). Gut/rectum expression of pha-4::GFP coincides with birefringence of gut granules, which mark the intestine. (E) ceh-51(tm2123); tbx-35(tm1789); glp-1(RNAi) embryo showing a small number of pharynx cells (arrow). (F) med-1(ok804); med-2(cx9744);glp-1(RNAi) embryo. (G-I) Body muscle cells marked by hlh-1::GFP (Krause et al.,1990). (G) Wild-type embryo just before hatching. (H) ceh-51(tm2123); pal-1(RNAi) embryo. (I) ceh-51(tm2123);tbx-35(tm1789); pal-1(RNAi) embryo. (J-L) Coelomocytes marked by cup-4::GFP (Patton et al.,2005). (J) Wild-type embryo with four coelomocytes. (K,L) Double ceh-51(tm2123); tbx-35(tm1789) or med-1(ok804);med-2(cx9744) mutants produce little or no coelomocytes. (M) In situ hybridization showing expression of pal-1 in the early C and D lineages (Baugh et al., 2005).(N) Ectopic expression of pal-1 in ceh-51(tm2123);tbx-35(tm1789) double mutant. (O) Ectopic pal-1 in a med-1(ok804); med-2(cx9744) embryo. (P) Histogram summarizing elongation of wild-type and mutant embryos. *, P=0.05(χ2 test), for some dataset pairs (comparisons among other pairs are not shown). The total number (n) of embryos scored per experiment is shown above each bar.

Fig. 8.

Mutation of ceh-51 and tbx-35 together synergizes to a med-1,2(-) arrest phenotype. (A-C) Pharynx muscles marked by ceh-22::GFP (Okkema and Fire,1994) overlaid on DIC images. (A) Arrested 1.5-fold tbx-35(tm1789) embryo raised at 20°C showing AB-derived and MS-derived pharynx muscles. (B) ceh-51(tm2123); tbx-35(tm1789) double mutant arrested at ∼1.5-fold elongation. (C) med-1(ok804);med-2(cx9744) double mutant. (D-F) Polarized light images to show gut granules overlaid with pha-4::GFP(Horner et al., 1998). (D) At 15°C, tbx-35(tm1789); glp-1(RNAi) embryos display 6.6±0.5(n=23) pharynx cells. Some additional GFP-positive cells are seen(arrows); similar `stray' GFP expression is also seen in a skn-1(RNAi) background (see Table 1). Gut/rectum expression of pha-4::GFP coincides with birefringence of gut granules, which mark the intestine. (E) ceh-51(tm2123); tbx-35(tm1789); glp-1(RNAi) embryo showing a small number of pharynx cells (arrow). (F) med-1(ok804); med-2(cx9744);glp-1(RNAi) embryo. (G-I) Body muscle cells marked by hlh-1::GFP (Krause et al.,1990). (G) Wild-type embryo just before hatching. (H) ceh-51(tm2123); pal-1(RNAi) embryo. (I) ceh-51(tm2123);tbx-35(tm1789); pal-1(RNAi) embryo. (J-L) Coelomocytes marked by cup-4::GFP (Patton et al.,2005). (J) Wild-type embryo with four coelomocytes. (K,L) Double ceh-51(tm2123); tbx-35(tm1789) or med-1(ok804);med-2(cx9744) mutants produce little or no coelomocytes. (M) In situ hybridization showing expression of pal-1 in the early C and D lineages (Baugh et al., 2005).(N) Ectopic expression of pal-1 in ceh-51(tm2123);tbx-35(tm1789) double mutant. (O) Ectopic pal-1 in a med-1(ok804); med-2(cx9744) embryo. (P) Histogram summarizing elongation of wild-type and mutant embryos. *, P=0.05(χ2 test), for some dataset pairs (comparisons among other pairs are not shown). The total number (n) of embryos scored per experiment is shown above each bar.

We are grateful to Shohei Mitani for tm2123; L. Ryan Baugh and Craig Hunter for sharing embryo transcriptome data prior to publication; Ian Hope, Michael Krause, Craig Hunter, Johnny Fares, Jeb Gaudet, Peter Okkema,Jenny Hsieh and Andy Fire for sending GFP reporter strains; Yuji Kohara for ceh-51 ESTs; Christian Frøkjær-Jensen, Erik Jorgensen,Attila Stetak, Andy Fire and David Miller for plasmids; Serena Cervantes for preliminary characterization of the tbx-35 elongation defects; and two anonymous reviewers for helpful comments. Some nematode strains used in this work were provided by the Caenorhabditis Genetics Center, which is funded by the NIH National Center for Research Resources(NCRR). This work was funded by grants from the NSF(IBN#0416922 and IOS#0643325)and NIH(1R03HD054589-01) to M.F.M. P.W.S. is an Investigator with the Howard Hughes Medical Institute, which supported this work. Deposited in PMC for release after 6 months.

Ahringer, J. (2006). Reverse genetics. In WormBook(ed. The C. elegans Research Community), doi:10.1895/wormbook.1.47.1, http://www.wormbook.org.
Azpiazu, N. and Frasch, M. (
1993
). tinman and bagpipe: two homeo box genes that determine cell fates in the dorsal mesoderm of Drosophila.
Genes Dev.
7
,
1325
-1340.
Baugh, L. R., Hill, A. A., Slonim, D. K., Brown, E. L. and Hunter, C. P. (
2003
). Composition and dynamics of the Caenorhabditis elegans early embryonic transcriptome.
Development
130
,
889
-900.
Baugh, L. R., Hill, A. A., Claggett, J. M., Hill-Harfe, K., Wen,J. C., Slonim, D. K., Brown, E. L. and Hunter, C. P. (
2005
). The homeodomain protein PAL-1 specifies a lineage-specific regulatory network in the C. elegans embryo.
Development
132
,
1843
-1854.
Bodmer, R. (
1993
). The gene tinman is required for specification of the heart and visceral muscles in Drosophila.
Development
118
,
719
-729.
Bowerman, B., Eaton, B. A. and Priess, J. R.(
1992
). skn-1, a maternally expressed gene required to specify the fate of ventral blastomeres in the early C. elegans embryo.
Cell
68
,
1061
-1075.
Bowerman, B., Draper, B. W., Mello, C. C. and Priess, J. R.(
1993
). The maternal gene skn-1 encodes a protein that is distributed unequally in early C. elegans embryos.
Cell
74
,
443
-452.
Broitman-Maduro, G., Maduro, M. F. and Rothman, J. H.(
2005
). The noncanonical binding site of the MED-1 GATA factor defines differentially regulated target genes in the C. elegans mesendoderm.
Dev. Cell
8
,
427
-433.
Broitman-Maduro, G., Lin, K. T. H., Hung, W. and Maduro, M.(
2006
). Specification of the C. elegans MS blastomere by the T-box factor TBX-35.
Development
133
,
3097
-3106.
Bruneau, B. G., Nemer, G., Schmitt, J. P., Charron, F.,Robitaille, L., Caron, S., Conner, D. A., Gessler, M., Nemer, M., Seidman, C. E. et al. (
2001
). A murine model of Holt-Oram syndrome defines roles of the T-box transcription factor Tbx5 in cardiogenesis and disease.
Cell
106
,
709
-721.
Coroian, C., Broitman-Maduro, G. and Maduro, M. F.(
2005
). Med-type GATA factors and the evolution of mesendoderm specification in nematodes.
Dev. Biol.
289
,
444
-455.
Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E. and Mello, C. C. (
1998
). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans.
Nature
391
,
806
-811.
Fox, R. M., Watson, J. D., Von Stetina, S. E., McDermott, J.,Brodigan, T. M., Fukushige, T., Krause, M. and Miller, D. M., 3rd(
2007
). The embryonic muscle transcriptome of Caenorhabditis elegans.
Genome Biol.
8
,
R188
.
Franks, D. M., Izumikawa, T., Kitagawa, H., Sugahara, K. and Okkema, P. G. (
2006
). C. elegans pharyngeal morphogenesis requires both de novo synthesis of pyrimidines and synthesis of heparan sulfate proteoglycans.
Dev. Biol.
296
,
409
-420.
Fukushige, T., Brodigan, T. M., Schriefer, L. A., Waterston, R. H. and Krause, M. (
2006
). Defining the transcriptional redundancy of early bodywall muscle development in C. elegans: evidence for a unified theory of animal muscle development.
Genes Dev.
20
,
3395
-3406.
Gaudet, J. and Mango, S. E. (
2002
). Regulation of organogenesis by the Caenorhabditis elegans FoxA protein PHA-4.
Science
295
,
821
-825.
Ghosh, T. K., Packham, E. A., Bonser, A. J., Robinson, T. E.,Cross, S. J. and Brook, J. D. (
2001
). Characterization of the TBX5 binding site and analysis of mutations that cause Holt-Oram syndrome.
Hum. Mol. Genet.
10
,
1983
-1994.
Goldstein, B. (
1992
). Induction of gut in Caenorhabditis elegans embryos.
Nature
357
,
255
-257.
Goszczynski, B. and McGhee, J. D. (
2005
). Re-evaluation of the role of the med-1 and med-2 genes in specifying the C. elegans endoderm.
Genetics
171
,
545
-555.
Hajarnavis, A., Korf, I. and Durbin, R. (
2004
). A probabilistic model of 3′ end formation in Caenorhabditis elegans.
Nucleic Acids Res.
32
,
3392
-3399.
Harfe, B. D. and Fire, A. (
1998
). Muscle and nerve-specific regulation of a novel NK-2 class homeodomain factor in Caenorhabditis elegans.
Development
125
,
421
-429.
Harvey, R. P. (
1996
). NK-2 homeobox genes and heart development.
Dev. Biol.
178
,
203
-216.
Haun, C., Alexander, J., Stainier, D. Y. and Okkema, P. G.(
1998
). Rescue of Caenorhabditis elegans pharyngeal development by a vertebrate heart specification gene.
Proc. Natl. Acad. Sci. USA
95
,
5072
-5075.
Hiroi, Y., Kudoh, S., Monzen, K., Ikeda, Y., Yazaki, Y., Nagai,R. and Komuro, I. (
2001
). Tbx5 associates with Nkx2-5 and synergistically promotes cardiomyocyte differentiation.
Nat. Genet.
28
,
276
-280.
Horb, M. E. and Thomsen, G. H. (
1999
). Tbx5 is essential for heart development.
Development
126
,
1739
-1751.
Horner, M. A., Quintin, S., Domeier, M. E., Kimble, J.,Labouesse, M. and Mango, S. E. (
1998
). pha-4, an HNF-3 homolog, specifies pharyngeal organ identity in Caenorhabditis elegans.
Genes Dev.
12
,
1947
-1952.
Huang, S., Shetty, P., Robertson, S. M. and Lin, R.(
2007
). Binary cell fate specification during C. elegans embryogenesis driven by reiterated reciprocal asymmetry of TCF POP-1 and its coactivator beta-catenin SYS-1.
Development
134
,
2685
-2695.
Hunt-Newbury, R., Viveiros, R., Johnsen, R., Mah, A., Anastas,D., Fang, L., Halfnight, E., Lee, D., Lin, J., Lorch, A. et al.(
2007
). High-throughput in vivo analysis of gene expression in Caenorhabditis elegans.
PLoS Biol.
5
,
e237
.
Hunter, C. P. and Kenyon, C. (
1996
). Spatial and temporal controls target pal-1 blastomere-specification activity to a single blastomere lineage in C. elegans embryos.
Cell
87
,
217
-226.
Kagoshima, H., Cassata, G. and Burglin, T. R.(
1999
). A Caenorhabditis elegans homeobox gene expressed in the male tail, a link between pattern formation and sexual dimorphism?
Dev. Genes Evol.
209
,
59
-62.
Kalb, J. M., Lau, K. K., Goszczynski, B., Fukushige, T., Moons,D., Okkema, P. G. and McGhee, J. D. (
1998
). pha-4 is Ce-fkh-1, a fork head/HNF-3alpha,beta,gamma homolog that functions in organogenesis of the C. elegans pharynx.
Development
125
,
2171
-2180.
Kamath, R. S. and Ahringer, J. (
2003
). Genome-wide RNAi screening in Caenorhabditis elegans.
Methods
30
,
313
-321.
Katic, I. and Greenwald, I. (
2006
). EMB-4: a predicted ATPase that facilitates lin-12 activity in Caenorhabditis elegans.
Genetics
174
,
1907
-1915.
Kispert, A. and Herrmann, B. G. (
1993
). The Brachyury gene encodes a novel DNA binding protein.
EMBO J.
12
,
4898
-4899.
Kohara, Y. (
2001
). Systematic analysis of gene expression of the C. elegans genome.
Tanpakushitsu Kakusan Koso
46
,
2425
-2431.
Krause, M., Fire, A., Harrison, S. W., Priess, J. and Weintraub,H. (
1990
). CeMyoD accumulation defines the body wall muscle cell fate during C. elegans embryogenesis.
Cell
63
,
907
-919.
Krause, M., Harrison, S. W., Xu, S. Q., Chen, L. and Fire,A. (
1994
). Elements regulating cell- and stage-specific expression of the C. elegans MyoD family homolog hlh-1.
Dev. Biol.
166
,
133
-148.
Labouesse, M. and Mango, S. E. (
1999
). Patterning the C. elegans embryo: moving beyond the cell lineage.
Trends Genet.
15
,
307
-313.
Lee, T. I., Rinaldi, N. J., Robert, F., Odom, D. T., Bar-Joseph,Z., Gerber, G. K., Hannett, N. M., Harbison, C. T., Thompson, C. M., Simon, I. et al. (
2002
). Transcriptional regulatory networks in Saccharomyces cerevisiae.
Science
298
,
799
-804.
Lei, H., Liu, J., Fukushige, T., Fire, A. and Krause, M.(
2009
). Caudal-like PAL-1 directly activates the bodywall muscle module regulator hlh-1 in C. elegans to initiate the embryonic muscle gene regulatory network.
Development
136
,
1241
-1249.
Lin, K. T., Broitman-Maduro, G., Hung, W. W., Cervantes, S. and Maduro, M. F. (
2009
). Knockdown of SKN-1 and the Wnt effector TCF/POP-1 reveals differences in endomesoderm specification in C. briggsae as compared with C. elegans.
Dev. Biol.
325
,
296
-306.
Lin, R. (
2003
). A gain-of-function mutation in oma-1, a C. elegans gene required for oocyte maturation, results in delayed degradation of maternal proteins and embryonic lethality.
Dev. Biol.
258
,
226
-239.
Lin, R., Thompson, S. and Priess, J. R. (
1995
). pop-1 encodes an HMG box protein required for the specification of a mesoderm precursor in early C. elegans embryos.
Cell
83
,
599
-609.
Lin, R., Hill, R. J. and Priess, J. R. (
1998
). POP-1 and anterior-posterior fate decisions in C. elegans embryos.
Cell
92
,
229
-239.
Lints, T. J., Parsons, L. M., Hartley, L., Lyons, I. and Harvey,R. P. (
1993
). Nkx-2.5: a novel murine homeobox gene expressed in early heart progenitor cells and their myogenic descendants.
Development
119
,
419
-431.
Lo, M. C., Gay, F., Odom, R., Shi, Y. and Lin, R.(
2004
). Phosphorylation by the beta-catenin/MAPK complex promotes 14-3-3-mediated nuclear export of TCF/POP-1 in signal-responsive cells in C. elegans.
Cell
117
,
95
-106.
Lyons, I., Parsons, L. M., Hartley, L., Li, R., Andrews, J. E.,Robb, L. and Harvey, R. P. (
1995
). Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2-5.
Genes Dev.
9
,
1654
-1666.
Macindoe, I., Glockner, L., Vukasin, P., Stennard, F. A., Costa,M. W., Harvey, R. P., Mackay, J. P. and Sunde, M. (
2009
). Conformational stability and DNA binding specificity of the cardiac T-Box transcription factor Tbx20.
J. Mol. Biol.
389
,
606
-618.
Maduro, M. F. (
2009
). Structure and evolution of the C. elegans embryonic endomesoderm network.
Biochim Biophys Acta
1789
,
250
-260.
Maduro, M. F., Meneghini, M. D., Bowerman, B., Broitman-Maduro,G. and Rothman, J. H. (
2001
). Restriction of mesendoderm to a single blastomere by the combined action of SKN-1 and a GSK-3beta homolog is mediated by MED-1 and -2 in C. elegans.
Mol. Cell
7
,
475
-485.
Maduro, M. F., Lin, R. and Rothman, J. H.(
2002
). Dynamics of a developmental switch: recursive intracellular and intranuclear redistribution of Caenorhabditis elegans POP-1 parallels Wnt-inhibited transcriptional repression.
Dev. Biol.
248
,
128
-142.
Maduro, M. F., Kasmir, J. J., Zhu, J. and Rothman, J. H.(
2005a
). The Wnt effector POP-1 and the PAL-1/Caudal homeoprotein collaborate with SKN-1 to activate C. elegans endoderm development.
Dev. Biol.
285
,
510
-523.
Maduro, M., Hill, R. J., Heid, P. J., Newman-Smith, E. D., Zhu,J., Priess, J. and Rothman, J. (
2005b
). Genetic redundancy in endoderm specification within the genus Caenorhabditis.
Dev. Biol.
284
,
509
-522.
Maduro, M. F., Broitman-Maduro, G., Mengarelli, I. and Rothman,J. H. (
2007
). Maternal deployment of the embryonic SKN-1→MED-1,2 cell specification pathway in C. elegans.
Dev. Biol.
301
,
590
-601.
Mango, S. E. (2007). The C. elegans pharynx: a model for organogenesis. In WormBook (ed. The C. elegans Research Community), doi:10.1895/wormbook.1.129.1, http://www.wormbook.org.
Mello, C. C., Draper, B. W., Krause, M., Weintraub, H. and Priess, J. R. (
1992
). The pie-1 and mex-1 genes and maternal control of blastomere identity in early C. elegans embryos.
Cell
70
,
163
-176.
Miller, D. M., Stockdale, F. E. and Karn, J.(
1986
). Immunological identification of the genes encoding the four myosin heavy chain isoforms of Caenorhabditis elegans.
Proc. Natl. Acad. Sci. USA
83
,
2305
-2309.
Okkema, P. G. and Fire, A. (
1994
). The Caenorhabditis elegans NK-2 class homeoprotein CEH-22 is involved in combinatorial activation of gene expression in pharyngeal muscle.
Development
120
,
2175
-2186.
Okkema, P. G. and Krause, M. (2005). Transcriptional regulation. In WormBook (ed. The C. elegans Research Community), doi:10.1895/wormbook.1.45.1, http://www.wormbook.org.
Okkema, P. G., Harrison, S. W., Plunger, V., Aryana, A. and Fire, A. (
1993
). Sequence requirements for myosin gene expression and regulation in Caenorhabditis elegans.
Genetics
135
,
385
-404.
Page, B. D., Diede, S. J., Tenlen, J. R. and Ferguson, E. L.(
2007
). EEL-1, a Hect E3 ubiquitin ligase, controls asymmetry and persistence of the SKN-1 transcription factor in the early C. elegans embryo.
Development
134
,
2303
-2314.
Patton, A., Knuth, S., Schaheen, B., Dang, H., Greenwald, I. and Fares, H. (
2005
). Endocytosis function of a ligand-gated ion channel homolog in Caenorhabditis elegans.
Curr. Biol.
15
,
1045
-1050.
Phillips, B. T., Kidd, A. R., 3rd, King, R., Hardin, J. and Kimble, J. (
2007
). Reciprocal asymmetry of SYS-1/beta-catenin and POP-1/TCF controls asymmetric divisions in Caenorhabditis elegans.
Proc. Natl. Acad. Sci. USA
104
,
3231
-3236.
Priess, J. R., Schnabel, H. and Schnabel, R.(
1987
). The glp-1 locus and cellular interactions in early C. elegans embryos.
Cell
51
,
601
-611.
Reece-Hoyes, J. S., Shingles, J., Dupuy, D., Grove, C. A.,Walhout, A. J., Vidal, M. and Hope, I. A. (
2007
). Insight into transcription factor gene duplication from Caenorhabditis elegans Promoterome-driven expression patterns.
BMC Genomics
8
,
27
.
Reim, I., Mohler, J. P. and Frasch, M. (
2005
). Tbx20-related genes, mid and H15, are required for tinman expression, proper patterning, and normal differentiation of cardioblasts in Drosophila.
Mech. Dev.
122
,
1056
-1069.
Rocheleau, C. E., Downs, W. D., Lin, R., Wittmann, C., Bei, Y.,Cha, Y. H., Ali, M., Priess, J. R. and Mello, C. C. (
1997
). Wnt signaling and an APC-related gene specify endoderm in early C. elegans embryos.
Cell
90
,
707
-716.
Rocheleau, C. E., Yasuda, J., Shin, T. H., Lin, R., Sawa, H.,Okano, H., Priess, J. R., Davis, R. J. and Mello, C. C.(
1999
). WRM-1 activates the LIT-1 protein kinase to transduce anterior/posterior polarity signals in C. elegans.
Cell
97
,
717
-726.
Shaham, S. (2006). Methods in cell biology. In WormBook (ed. The C. elegans Research Community),doi:10.1895/wormbook.1.49.1, http://www.wormbook.org.
Shelton, C. A. and Bowerman, B. (
1996
). Time-dependent responses to glp-1-mediated inductions in early C. elegans embryos.
Development
122
,
2043
-2050.
Shetty, P., Lo, M. C., Robertson, S. M. and Lin, R.(
2005
). C. elegans TCF protein, POP-1, converts from repressor to activator as a result of Wnt-induced lowering of nuclear levels.
Dev. Biol.
285
,
584
-592.
Shirayama, M., Soto, M. C., Ishidate, T., Kim, S., Nakamura, K.,Bei, Y., van den Heuvel, S. and Mello, C. C. (
2006
). The conserved kinases CDK-1, GSK-3, KIN-19, and MBK-2 promote OMA-1 destruction to regulate the oocyte-to-embryo transition in C. elegans.
Curr. Biol.
16
,
47
-55.
Stennard, F. A., Costa, M. W., Elliott, D. A., Rankin, S.,Haast, S. J., Lai, D., McDonald, L. P., Niederreither, K., Dolle, P., Bruneau,B. G. et al. (
2003
). Cardiac T-box factor Tbx20 directly interacts with Nkx2-5, GATA4, and GATA5 in regulation of gene expression in the developing heart.
Dev. Biol.
262
,
206
-224.
Sulston, J. E. and Hodgkin, J. (
1988
). Methods. In
The Nematode Caenorhabditis elegans
(ed. W. B. Wood), pp.
587
-606. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press.
Sulston, J. E., Schierenberg, E., White, J. G. and Thomson, J. N. (
1983
). The embryonic cell lineage of the nematode Caenorhabditis elegans.
Dev. Biol.
100
,
64
-119.
Syntichaki, P. and Tavernarakis, N. (
2004
). Genetic models of mechanotransduction: the nematode Caenorhabditis elegans.
Physiol. Rev.
84
,
1097
-1153.
Thorpe, C. J., Schlesinger, A., Carter, J. C. and Bowerman,B. (
1997
). Wnt signaling polarizes an early C. elegans blastomere to distinguish endoderm from mesoderm.
Cell
90
,
695
-705.
Williams, B. D. and Waterston, R. H. (
1994
). Genes critical for muscle development and function in Caenorhabditis elegans identified through lethal mutations.
J. Cell Biol.
124
,
475
-490.