Cardiac malformations due to aberrant development of the atrioventricular(AV) valves are among the most common forms of congenital heart disease. At localized swellings of extracellular matrix known as the endocardial cushions,the endothelial lining of the heart undergoes an epithelial to mesenchymal transition (EMT) to form the mesenchymal progenitors of the AV valves. Further growth and differentiation of these mesenchymal precursors results in the formation of portions of the atrial and ventricular septae, and the generation of thin, pliable valves. Gata4, which encodes a zinc finger transcription factor, is expressed in the endothelium and mesenchyme of the AV valves. Using a Tie2-Cre transgene, we selectively inactivated Gata4 within endothelial-derived cells. Mutant endothelium failed to undergo EMT, resulting in hypocellular cushions. Mutant cushions had decreased levels of Erbb3, an EGF-family receptor essential for EMT in the atrioventricular cushions. In Gata4 mutant embryos, Erbb3downregulation was associated with impaired activation of Erk, which is also required for EMT. Expression of a Gata4 mutant protein defective in interaction with Friend of Gata (FOG) cofactors rescued the EMT defect, but resulted in a decreased proliferation of mesenchyme and hypoplastic cushions that failed to septate the ventricular inlet. We demonstrate two novel functions of Gata4 in development of the AV valves. First, Gata4 functions as an upstream regulator of an Erbb3-Erk pathway necessary for EMT, and second, Gata4 acts to promote cushion mesenchyme growth and remodeling.

Cardiac malformations attributable to aberrant development of the atrioventricular (AV) valvuloseptal complex are among the most common forms of congenital heart disease (Pierpont et al.,2000). Development of the AV valvuloseptal complex can be considered to occur in several steps (reviewed by Armstrong and Bischoff, 2004). First, endocardial cells at the AV canal (AVC) undergo an epithelial to mesenchymal transition (EMT), forming mesenchymal cells that invade swellings of extracellular matrix to form the endocardial cushions (EC). Next, the cellularized superior and inferior EC grow and fuse, forming the AV valvuloseptal complex that divides the ventricular inflow into a right and a left AV valve. Further elongation and remodeling of the cushions results in formation of the mature valve leaflets.

Transformation of AV endocardium to cushion mesenchyme requires the input of multiple signaling molecules secreted from the adjacent myocardium,including Tgfβ and Egf family members(Camenisch et al., 2000; Nakajima et al., 2000a; Sugi et al., 2004; Rivera-Feliciano and Tabin,2006). Ablation of Erbb3, which encodes an Egf family receptor, results in failure of endocardial cells of the AV cushions to undergo EMT to form cushion mesenchyme(Camenisch et al., 2000). One mechanism by which Erbb3 promotes EMT is to activate Ras(Camenisch et al., 2000). Decreased Ras signaling results in defective EMT(Camenisch et al., 2000; Lakkis and Epstein, 1998),and, conversely, increased Ras signaling results in increased EMT and hypercellular, enlarged EC (Gitler et al.,2003; Lakkis and Epstein,1998).

The transcription factor Gata4 is essential for heart formation(Kuo et al., 1997; Molkentin et al., 1997; Watt et al., 2004; Zeisberg et al., 2005). In humans, GATA4 heterozygous mutations have been associated with defects in the muscular septum separating atria or ventricles, and variably associated with valvar pulmonary stenosis(Garg et al., 2003; Hirayama-Yamada et al., 2005; Okubo et al., 2004). In addition to its expression in the myocardium, Gata4 is robustly expressed in the endocardium and the EC(Charron and Nemer, 1999; Heikinheimo et al., 1994). This expression pattern, and the presence of EC defects in mouse embryos homozygous for two different hypomorphic Gata4 alleles(Crispino et al., 2001; Pu et al., 2004), suggested that Gata4 might be an important regulator of EC development.

To further investigate the role of Gata4 in EC development, we specifically inactivated Gata4 in endothelium and endothelium-derived cushion mesenchyme. We show that Gata4 expression in endothelium-derived cells is required at two stages of AV valve formation, illustrating novel cell-autonomous roles for Gata4 in the endocardium. First, Gata4 is required to promote EMT of endocardial cells to generate AV cushion mesenchyme. Second,Gata4 activity in endocardial-derived cells is required later during AV valve maturation for growth and remodeling of the AV cushions to septate the ventricular inlet.

Mice

Gata4H and Gata4flox alleles(Fig. 1A) have been described previously (Pu et al., 2004; Zeisberg et al., 2005). Gata4H expresses reduced amounts of protein compared with the wild-type allele (Pu et al.,2004). Gata4wt/Ki and R26RstoplacZmice (Crispino et al., 2001; Mao et al., 1999) were obtained from Stuart Orkin (Harvard Medical School). Transgenic mice expressing Cre from Tie2 (also known as Tek)regulatory elements (T2Cre) were obtained from M. Yanagisawa(Kisanuki et al., 2001). Wnt1-Cre mice (Danielian et al.,1998) were obtained from Jackson Laboratories. All mice were maintained in a mixed C57BL6/129 genetic background. Gata4wt/flox; T2Cre+ mice were crossed with Gata4flox/flox mice to yield Gata4flox/flox; T2Cre+(Gata4T2del) mice. All animal care and procedures were performed under protocols approved by the Institutional Animal Care and Use Committee.

Fig. 1.

Endocardial-restricted inactivation of Gata4. (A)Structure of wild-type (wt), hypomorphic (H), floxed, and deleted (Δex2) Gata4 alleles. (B) Fate mapping of T2Cre-recombined endothelial cells. (B1,B2) In T2Cre+; R26RstoplacZ embryos at E9.5,β-galactosidase reporter expression (blue) was seen in the endocardium(arrow) and EC mesenchyme (arrowhead) of the AVC, and in the OT endothelium.(B3) By E11.5, the β-galactosidase-positive cells populated the AV cushions (asterisks). Endothelial-derived OT cushion mesenchyme (yellow arrow)was largely confined to the most proximal portion of the OT cushion. Most of the outflow cushion was not recombined by T2Cre (green arrow).(C) Endocardial-restricted inactivation of Gata4 by T2Cre at E9.5. Control (C1,C2) and Gata4T2del(C3,C4) embryos, hybridized to a Gata4 exon 2-specific probe (red pseudocolor). Blue, DAPI counterstain. The Gata4 exon 2 in situ hybridization signal was present in the endocardium of control embryos but not of mutant embryos (white arrowheads). Gata4 expression in the myocardium (yellow arrowheads) and proepicardium (star) was unaffected.(D) Inactivation of Gata4 in endothelium and endothelium-derived cells at E11.5. In control embryos (D1), Gata4exon 2 in situ hybridization signal was present in the endocardium (white arrowhead), myocardium (yellow arrowhead), and AV and OT cushion mesenchyme. In mutant embryos (D2), Gata4 was not detected in endothelium (white arrowhead) or AVC mesenchyme. Strong signal was still present in mid and distal OT cushion mesenchyme (green arrow) and myocardium (yellow arrowhead). a, atria; v, ventricle; ot, OT. Scale bars in C and D: 100 μm.

Fig. 1.

Endocardial-restricted inactivation of Gata4. (A)Structure of wild-type (wt), hypomorphic (H), floxed, and deleted (Δex2) Gata4 alleles. (B) Fate mapping of T2Cre-recombined endothelial cells. (B1,B2) In T2Cre+; R26RstoplacZ embryos at E9.5,β-galactosidase reporter expression (blue) was seen in the endocardium(arrow) and EC mesenchyme (arrowhead) of the AVC, and in the OT endothelium.(B3) By E11.5, the β-galactosidase-positive cells populated the AV cushions (asterisks). Endothelial-derived OT cushion mesenchyme (yellow arrow)was largely confined to the most proximal portion of the OT cushion. Most of the outflow cushion was not recombined by T2Cre (green arrow).(C) Endocardial-restricted inactivation of Gata4 by T2Cre at E9.5. Control (C1,C2) and Gata4T2del(C3,C4) embryos, hybridized to a Gata4 exon 2-specific probe (red pseudocolor). Blue, DAPI counterstain. The Gata4 exon 2 in situ hybridization signal was present in the endocardium of control embryos but not of mutant embryos (white arrowheads). Gata4 expression in the myocardium (yellow arrowheads) and proepicardium (star) was unaffected.(D) Inactivation of Gata4 in endothelium and endothelium-derived cells at E11.5. In control embryos (D1), Gata4exon 2 in situ hybridization signal was present in the endocardium (white arrowhead), myocardium (yellow arrowhead), and AV and OT cushion mesenchyme. In mutant embryos (D2), Gata4 was not detected in endothelium (white arrowhead) or AVC mesenchyme. Strong signal was still present in mid and distal OT cushion mesenchyme (green arrow) and myocardium (yellow arrowhead). a, atria; v, ventricle; ot, OT. Scale bars in C and D: 100 μm.

Histology

Embryos were fixed in 4% paraformaldehyde overnight at 4°C, and then paraffin-wax embedded and sectioned at 10 μm. Alcian Blue staining was performed by using 0.15 mg/ml Alcian Blue in 5% acetic acid. BrdU labeling and TUNEL staining were performed as described previously(Zeisberg et al., 2005).

Tissue culture

For short-term embryo culture, E9.5 embryos were incubated in M199 supplemented with 1% FBS for 30 minutes at 37°C in 5% CO2. Heregulin (Sigma; 100 ng/ml) or U0126 (Calbiochem; 10 μM) were added as indicated.

AV explant culture was carried out as described(Rivera-Feliciano and Tabin,2006). Where indicated, U0126 (Calbiochem), vehicle (DMSO) or a growth factor cocktail [Tgfβ2 (Cell Biosciences), 50 ng/ml; Bmp2 (R&D Systems), 200 ng/ml; hyaluronic acid (Sigma), 500 ng/ml; heregulin (Sigma), 50 ng/ml] was added to the media at the start of the explant culture.

Human umbilical vein endothelial cells (HUVEC; passage <6; Cambrex) were cultured in complete endothelial growth media (Cambrex). BT20 human breast cancer cells (ATCC) were transfected using Fugene6 (Roche).

Gene expression

Whole-mount staining for detection of β-galactosidase activity was performed as described (Lobe et al.,1999). Immunostaining was performed using the following antibodies: Nfatc1 (1:200, Santa Cruz), Desmin (1:4, Biomeda), Chd5 (1:20,Santa Cruz), phospho-Erk1/2 (1:200, Cell Signaling),α-SMA-Cy3-conjugated (Clone 1A4, Sigma 1:200) and biotin-conjugated CD31(Pecam1, 1:100) monoclonal antibody (Clone MEC 13.3, BD Pharmingen). Erbb3 western blotting was performed with antibody C-17 (Santa Cruz, 1:200) and normalized to Gapdh (Research Diagnostics, 1:10,000).

For RNA analysis, four mutant and four control RNA samples were prepared(Pico-Pure RNA Isolation Kit, Arcturus), each consisting of 10 microdissected AVCs. Probe was prepared from 50 ng total RNA using an isothermal amplification protocol (NuGen), and hybridized to Affymetrix Mouse 430 2.0 microarrays. Two control samples were excluded because of excessive noise. We excluded probe sets that may cross-hybridize to unrelated targets (probe name ending with `_x_at') or that received `Absent' calls across all samples. The 27,082 remaining probe sets were ranked by the `relative difference' d-score(Tusher et al., 2001), using the Significance Analysis of Microarray (SAM) software package(http://www-stat.stanford.edu/~tibs/SAM/).

For quantitative RT-PCR, RNA samples were converted to cDNA and amplified by Ovation isothermal amplification. The cDNA was then used for quantitative PCR on an ABI7300 thermal cycler, with Sybr Green or Taqman detection. Primer sequences are provided in Table 1.

Table 1.

Primer and probe sequences

GeneForwardReverseNote
Real-time PCR assays    
Ankrd1 CAGTTGTAGAGAAATTCCTGTCAGACA GTGGAGTGCCGTCCGTTTAT Sybr Green 
Erbb3 TAGACGTCAATGGTGCAGATCTG TAGACGTCAATGGTGCAGATCTG Sybr Green, mouse 
Erbb3 GTGGCACTCAGGGAGCATTTA TCTGGGACTGGGGAAAAGG Sybr Green, human 
Gapdh Proprietary taqman assay from Applied Biosystems  Taqman 
Gata2 GGCAGTGGCCCTGTGATC CGAAGCTTGGGACACTTTTTATG Sybr Green 
Igf1 ACAGGCTATGGCTCCAGCAT GCTCCGGAAGCAACACTCAT Sybr Green 
Nrp2 ACGTGGCATTTGGGCATT GAGCATCCCCAGCATGTTG Sybr Green 
Pdga GGTCCACCACCGCAGTGT CCTGGACCTCTTTCAATTTTGG Sybr Green 
Plxc1 AGATGACCACTGCCACTTGATTC CTTTGAACTTGTGTTTCCCTCGAT Sybr Green 
Sgk3 TGCCTGGGCGCTGTTC AGCAACATCTCGGCAGTAAAAAG Sybr Green 
Thbs1 CCTCAAGAAAATGCAGTTTTCAAA AAACCCAAGGAACTCTTCATGGT Sybr Green 
Tnc TCCCCAAGAGAATTTACAGCTACAG GAGGTATCCAGTGACCGATGCT Sybr Green 
Vcam1 CGTCGCGAGGTTGTTTAGAGTA CAGTCCAAGCAACACTCTCTGATT Sybr Green 
In situ hybridization probes    
Bmp2 AACTAGAAGCCGTGGAGGAACTTCCA TTGTGGAGGGCTGCGGGTGTCGTTAG PCR template 
Cdh5 ACAACCATGACAACACCGCCAACA ATCCAGGTTGCAATGAGGTTGGGT PCR template 
Erbb3 BE947142  EST template 
Gata4 exon 2 195-981 of accession AB075549  PCR template 
HRT1 A1316788  EST template 
Nfatc1 TGGTGGTTGAGATACCACCTTTCC TAGAAGCGTCACCATGAGGACAAC PCR template 
Nkx2-5   (Tanaka et al., 1999
Notch1 AW047868  EST template 
Nppa   (Tanaka et al., 1999
Pecam CCAACAGAGCCAGCAGTATGAGGACCAG CACCCCCCGAAACACAAGGAAGATAGG PCR template 
Plxnc1 BE981975  EST template 
Snail CAAACCCACTCGGATGTGAAGAGA GTGAAACAGGTGTCACCAGGACAA PCR template 
Tbx2   (Bruneau et al., 2001
Tgfb2   (Pelton et al., 1989
Thbs1 AW122997  EST template 
Primers for amplifying Erbb3 promoter and intronic enhancer homology arms    
Prom 5′ arm GCGGTACCGAGAAAGCCTGTTGACTTGAGTTCCATC GGCTCGAGACTAGTCACCATATCCGGCTCTCCTGCTAC  
Prom 3′ arm GGACTAGTCCTAATCGCTGGCTGTTCAGGTGG GACCATGGTACCTGCCTGAGAGTTGCCCATC  
Enh 5′ arm GCGGTACCGCTCCCTTCACCCTCCACTGTAATC GGCTCGAGACTAGTCAGAACCCAAACTCCTTGCCCAGG  
Enh 3′ arm GGACTAGTCCTGGATAGAATTGCTTCATTGTG GGCTCGAGAACATTTACCCTCTCCTCCTCAACGG  
Site-directed mutagenesis    
Prom top ATCTCCTCTTAACCGTCTCCGGGGGAAAT   
Prom bot ATTTCCCCCGGAGACGGTTAAGAGGAGAT   
Enh m1 top CTTAGCTCCTCCCCTAGATGAGGAGAAGGGTTG   
Enh m1 bot CAACCCTTCTCCTCATCTAGGGGAGGAGCTAAG   
Enh m2 top GGAGGCCACCAGTTCCTTAGAATCCAGCTTTAAAGACAAAG   
Enh m2 bot CTTTGTCTTTAAAGCTGGATTCTAAGGAACTGGTGGCCTCC   
GeneForwardReverseNote
Real-time PCR assays    
Ankrd1 CAGTTGTAGAGAAATTCCTGTCAGACA GTGGAGTGCCGTCCGTTTAT Sybr Green 
Erbb3 TAGACGTCAATGGTGCAGATCTG TAGACGTCAATGGTGCAGATCTG Sybr Green, mouse 
Erbb3 GTGGCACTCAGGGAGCATTTA TCTGGGACTGGGGAAAAGG Sybr Green, human 
Gapdh Proprietary taqman assay from Applied Biosystems  Taqman 
Gata2 GGCAGTGGCCCTGTGATC CGAAGCTTGGGACACTTTTTATG Sybr Green 
Igf1 ACAGGCTATGGCTCCAGCAT GCTCCGGAAGCAACACTCAT Sybr Green 
Nrp2 ACGTGGCATTTGGGCATT GAGCATCCCCAGCATGTTG Sybr Green 
Pdga GGTCCACCACCGCAGTGT CCTGGACCTCTTTCAATTTTGG Sybr Green 
Plxc1 AGATGACCACTGCCACTTGATTC CTTTGAACTTGTGTTTCCCTCGAT Sybr Green 
Sgk3 TGCCTGGGCGCTGTTC AGCAACATCTCGGCAGTAAAAAG Sybr Green 
Thbs1 CCTCAAGAAAATGCAGTTTTCAAA AAACCCAAGGAACTCTTCATGGT Sybr Green 
Tnc TCCCCAAGAGAATTTACAGCTACAG GAGGTATCCAGTGACCGATGCT Sybr Green 
Vcam1 CGTCGCGAGGTTGTTTAGAGTA CAGTCCAAGCAACACTCTCTGATT Sybr Green 
In situ hybridization probes    
Bmp2 AACTAGAAGCCGTGGAGGAACTTCCA TTGTGGAGGGCTGCGGGTGTCGTTAG PCR template 
Cdh5 ACAACCATGACAACACCGCCAACA ATCCAGGTTGCAATGAGGTTGGGT PCR template 
Erbb3 BE947142  EST template 
Gata4 exon 2 195-981 of accession AB075549  PCR template 
HRT1 A1316788  EST template 
Nfatc1 TGGTGGTTGAGATACCACCTTTCC TAGAAGCGTCACCATGAGGACAAC PCR template 
Nkx2-5   (Tanaka et al., 1999
Notch1 AW047868  EST template 
Nppa   (Tanaka et al., 1999
Pecam CCAACAGAGCCAGCAGTATGAGGACCAG CACCCCCCGAAACACAAGGAAGATAGG PCR template 
Plxnc1 BE981975  EST template 
Snail CAAACCCACTCGGATGTGAAGAGA GTGAAACAGGTGTCACCAGGACAA PCR template 
Tbx2   (Bruneau et al., 2001
Tgfb2   (Pelton et al., 1989
Thbs1 AW122997  EST template 
Primers for amplifying Erbb3 promoter and intronic enhancer homology arms    
Prom 5′ arm GCGGTACCGAGAAAGCCTGTTGACTTGAGTTCCATC GGCTCGAGACTAGTCACCATATCCGGCTCTCCTGCTAC  
Prom 3′ arm GGACTAGTCCTAATCGCTGGCTGTTCAGGTGG GACCATGGTACCTGCCTGAGAGTTGCCCATC  
Enh 5′ arm GCGGTACCGCTCCCTTCACCCTCCACTGTAATC GGCTCGAGACTAGTCAGAACCCAAACTCCTTGCCCAGG  
Enh 3′ arm GGACTAGTCCTGGATAGAATTGCTTCATTGTG GGCTCGAGAACATTTACCCTCTCCTCCTCAACGG  
Site-directed mutagenesis    
Prom top ATCTCCTCTTAACCGTCTCCGGGGGAAAT   
Prom bot ATTTCCCCCGGAGACGGTTAAGAGGAGAT   
Enh m1 top CTTAGCTCCTCCCCTAGATGAGGAGAAGGGTTG   
Enh m1 bot CAACCCTTCTCCTCATCTAGGGGAGGAGCTAAG   
Enh m2 top GGAGGCCACCAGTTCCTTAGAATCCAGCTTTAAAGACAAAG   
Enh m2 bot CTTTGTCTTTAAAGCTGGATTCTAAGGAACTGGTGGCCTCC   

In situ hybridization was performed on 10 μm paraffin sections using digoxigenin-labeled or S35-labeled RNA probes as described(Brent et al., 2003; Tanaka et al., 1999). Signal from S35-labeled probes was detected in dark field and pseudocolored red using PhotoShop. Sections were counterstained with DAPI. For in situ hybridization probes refer to Table 1. Results shown are representative of at least two embryos.

Plasmids

The Gata4 expression construct has been described previously(Lee et al., 1998). The Gata4Δex2 expression construct was generated by RT-PCR amplification of the Cre-recombined Gata4transcript. Gata4DBD-engrailed was constructed by PCR cloning the Gata4 DNA-binding domain upstream of the engrailed repressor domain. The murine Erbb3 promoter and intron 1 enhancer was cloned from a bacterial artificial chromosome by Red/ET recombineering (GeneBridges) into pGL3-BASIC (Promega) or pGL3-promoter (Promega), respectively. Luciferase assays were normalized for transfection efficiency using pRL-null(Promega).

Endothelial-restricted Gata4 inactivation

To determine the function of Gata4 within the endocardium and its derivatives, we inactivated a floxed Gata4 allele(Gata4flox; Fig. 1A) (Pu et al.,2004; Zeisberg et al.,2005) by expressing Cre recombinase from a Tie2promoter (T2Cre) (Kisanuki et al., 2001). Cre-mediated recombination of Gata4flox resulted in excision of a portion of exon 2,including the start codon and 46% of the Gata4 coding region. The recombined allele (Gata4Δex2)expresses a truncated protein containing both zinc fingers and the C-terminal activation domain, but lacking the N-terminal transactivation domains (see Fig. S1 in the supplementary material). The truncated protein failed to activate multiple cardiac and intestinal Gata4-dependent promoters in vitro (Fig. S1 in the supplementary material; T. Bosse and S. Krasinski,personal communication), consistent with previous results(Morrisey et al., 1997). These data, along with the observation that embryos homozygous for this mutation in their germline(Gata4Δex2/Δex2)resemble previously reported mice carrying Gata4 null alleles(Kuo et al., 1997; Molkentin et al., 1997) (data not shown), suggest that the Gata4Δex2 allele behaves as a loss-of-function mutation. However, we cannot exclude the possibility that Gata4Δex2 retains partial function.

In control experiments, we characterized the spatiotemporal pattern of recombination catalyzed by T2Cre using the reporter R26RstoplacZ, which expresses lacZ only after activation by Cre (Mao et al.,1999). In T2Cre+; R26RstoplacZ embryos, T2Cre activated reporter expression in the majority of endocardial cells by E9.5 (Fig. 1B1,B2). lacZ expression was not observed in epicardial or myocardial cells. Between E9.5 and E11.5, endocardial cells at the AVC transform into mesenchymal cells and populate the AV cushions. Consistent with this lineage history, mesenchymal cells of the AV cushions were recombined by Cre recombinase, and consequently expressed the lacZ reporter at E9.5 and E11.5 (Fig. 1B1, arrow; 1B3,asterisks).

The endocardium and EC mesenchyme of the developing heart express high levels of Gata4 (Fig. 1C,D) (Heikinheimo et al.,1994). To specifically inactivate Gata4 in endocardial-derived cells, we generated embryos with the genotype Gata4flox/flox; T2Cre+(Gata4T2del). We examined expression of Gata4 in these embryos by in situ hybridization using an exon 2-specific probe. In control embryos, Gata4 exon 2 transcripts were present in endocardial, epicardial and myocardial cells(Fig. 1C1,C2). In Gata4T2del embryos, expression from the Gata4exon 2 probe was unchanged in the epicardium and myocardium (star and yellow arrowheads, Fig. 1C3,C4), but expression in the endocardium was absent (white arrowheads, Fig. 1C3,C4). At E11.5, robust expression was detected in AVC mesenchyme in control embryos, but this tissue was largely deficient in Gata4T2del embryos (asterisks, Fig. 1D; see below). The lack of detectable Gata4 transcripts in mutant endocardium was not due to non-specific transcript degradation, or a general failure of endothelium to subspecialize into endocardium, as mutant endocardium continued to express normal levels of Pecam, an endothelial marker, and Nfatc1, a marker of endothelium subspecialized to line the heart (data not shown).

EC also contribute to formation of the outflow (OT) tract and the OT valves. However, only the most proximal portion of OT cushion mesenchyme derives from endothelial progenitors that were recombined by the T2Cre transgene (yellow arrow, Fig. 1B). The bulk of OT cushion mesenchyme is derived from neural crest, as demonstrated by fate mapping using the neural crest restricted Wnt1Cre transgene(Gitler et al., 2003; Verzi et al., 2005) (see Fig. S2 in the supplementary material). Gata4 is expressed in OT endothelium and both endocardial- and neural crest-derived OT mesenchyme(green arrow, Fig. 1D). Consistent with the lack of T2Cre-mediated recombination in the mid and distal OT mesenchyme (green arrow, Fig. 1B3), Gata4 expression in these regions was not affected in Gata4T2del embryos (green arrow, Fig. 1D).

Phenotypic characterization of Gata4T2delembryos

Out of 34 litters genotyped, no Gata4T2del mice survived to weaning. By E12.5, the prevalence of Gata4T2del embryos was 80% less than expected based on Mendelian ratios (Fig. 2A). At E12.5, surviving Gata4T2del embryos had pericardial effusion and peripheral hemorrhage, which are hallmarks of embryos with heart failure (Fig. 2B,C). The liver was hypoplastic (Fig. 2B; see also Fig. S3 in the supplementary material). Histological examination of the heart showed that these mutant embryos displayed a paucity of mesenchymal cells within the AV cushions (Fig. 2D-G). Additionally, the mutant AV endocardium was multiple cell layers thick at certain foci (arrow, Fig. 2G), whereas the AV endocardium of controls remained as a single cell layer epithelial sheet (Fig. 2E). Although the myocardium appeared normal in the majority of mutant embryos, in 30% of embryos (six out of 17 examined) the compact myocardium was abnormally thin (Fig. S4 in the supplementary material).

Fig. 2.

Phenotype of embryos with endothelial-restricted inactivation of Gata4 (Gata4T2del). (A) Gata4T2del embryos die by E12.5-E13.5. The incidence of Gata4T2del embryos is plotted against gestational age;expected incidence was 25%. Numbers indicate total number of embryos genotyped. (B) Gross appearance of E12.5 control and Gata4T2del littermates. Mutants showed liver hypoplasia(L) and variable growth retardation, peripheral hemorrhage and pericardial effusion (arrowhead). (C) E12.5 Gata4T2del embryo with severe pericardial effusion (arrowhead). Scale bars: 50 μm.(D-G) Formation of AV cushion mesenchyme requires Gata4activity in endothelial-derived cells. By E10.5, the AV cushions of control embryos were populated by numerous mesenchymal cells (asterisk, D), whereas in Gata4T2del embryos the AV cushions were severely hypocellular (asterisk, F). The endocardium overlying the AV cushions became several cell layers thick in mutant embryos (arrow, G), whereas it was one cell layer thick in control embryos (E).

Fig. 2.

Phenotype of embryos with endothelial-restricted inactivation of Gata4 (Gata4T2del). (A) Gata4T2del embryos die by E12.5-E13.5. The incidence of Gata4T2del embryos is plotted against gestational age;expected incidence was 25%. Numbers indicate total number of embryos genotyped. (B) Gross appearance of E12.5 control and Gata4T2del littermates. Mutants showed liver hypoplasia(L) and variable growth retardation, peripheral hemorrhage and pericardial effusion (arrowhead). (C) E12.5 Gata4T2del embryo with severe pericardial effusion (arrowhead). Scale bars: 50 μm.(D-G) Formation of AV cushion mesenchyme requires Gata4activity in endothelial-derived cells. By E10.5, the AV cushions of control embryos were populated by numerous mesenchymal cells (asterisk, D), whereas in Gata4T2del embryos the AV cushions were severely hypocellular (asterisk, F). The endocardium overlying the AV cushions became several cell layers thick in mutant embryos (arrow, G), whereas it was one cell layer thick in control embryos (E).

The hypocellular AV cushion phenotype was 100% penetrant and was not due to increased apoptosis, as measured by TUNEL staining (data not shown). A small and variable number of mesenchymal cells were observed in mutant AV cushions. To determine the origin of these cells, we fate mapped T2Cre-expressing cells using the R26RstoplacZ reporter in control and Gata4T2del mutant embryos(Fig. 3). We found that these cells were lacZ positive (arrowhead, Fig. 3B), indicating that they are derived from endothelium and not from an alternative tissue compartment,such as epicardium. In situ hybridization demonstrated that the residual mesenchymal cells did not express Gata4 (data not shown). T2Cre-recombined mesenchyme at the proximal tip of the OT cushions(yellow arrows, Fig. 3A,C) was missing in Gata4T2del mutants (yellow arrows, Fig. 3B,D).

Fig. 3.

Fate map of T2Cre-recombined cells in Gata4T2del hearts. (A-D) T2Cre-mediated activation of the R26RstoplacZ reporter(blue) was mapped in E11.5 control (A,C) or Gata4T2del(B,D) embryo hearts. Whole-mount embryos were X-gal stained, then sectioned in the sagittal plane. Sections through the AV (A,B) and OT (C,D) cushions are shown. In control hearts, T2Cre-recombined cells of endothelial origin gave rise to the AV cushion mesenchyme (asterisk), as well as the proximal tip of the OT cushion (yellow arrow). The mid- and distal OT cushions were not recombined by T2Cre (red arrows). In Gata4T2del embryos, the corresponding regions were markedly hypocellular. Residual cells in the AV cushions were descended from T2Cre-recombined endothelium (arrowhead). Scale bar: 100 μm.

Fig. 3.

Fate map of T2Cre-recombined cells in Gata4T2del hearts. (A-D) T2Cre-mediated activation of the R26RstoplacZ reporter(blue) was mapped in E11.5 control (A,C) or Gata4T2del(B,D) embryo hearts. Whole-mount embryos were X-gal stained, then sectioned in the sagittal plane. Sections through the AV (A,B) and OT (C,D) cushions are shown. In control hearts, T2Cre-recombined cells of endothelial origin gave rise to the AV cushion mesenchyme (asterisk), as well as the proximal tip of the OT cushion (yellow arrow). The mid- and distal OT cushions were not recombined by T2Cre (red arrows). In Gata4T2del embryos, the corresponding regions were markedly hypocellular. Residual cells in the AV cushions were descended from T2Cre-recombined endothelium (arrowhead). Scale bar: 100 μm.

Gata4 is necessary for AV cushion EMT

The process of EMT can be recapitulated in vitro by culturing explants of the AVC in a three-dimensional collagen gel(Runyan and Markwald, 1983). During EMT, endothelial cells first undergo an activation step during which they lose their cell-cell contacts. The cells then adopt an elongated morphology, upregulate mesenchymal markers such as SMA, and downregulate endothelial markers such as Pecam. These activated cells subsequently invade and migrate through the extracellular matrix to complete the transformation process.

When cultured in a collagen gel, control AVC explants produced a halo of invasive, migrating cells with mesenchymal morphology(Fig. 4A). By contrast, mutant explants failed to generate mesenchymal cells(Fig. 4B,C). Immunostaining with antibodies for SMA and Pecam delineated three classes of endothelial cells at different stages of activation in all explants examined: (1) rounded cells expressing Pecam but not SMA (arrow, Fig. 4D); (2) rare, round transitional cells expressing SMA (white arrowhead, Fig. 4D); and (3) elongated,SMA-expressing cells (yellow arrowhead, Fig. 4D). By contrast, explants from Gata4T2del embryos failed to produce mesenchymal,SMA-expressing cells (Fig. 4E). Mutant endothelium appeared multi-layered in a manner reminiscent of the morphology of sectioned Gata4T2del hearts (star, Fig. 4E; arrow, Fig. 2G). None of the aforementioned signs of endothelial cell activation were detected in explants examined by immunostaining for Pecam and SMA. These data suggest that endothelial recombination of Gata4 impairs the activation of endothelial cells and blocks their transformation to invasive mesenchyme.

Fig. 4.

Endocardial Gata4 activity is required for mesenchyme formation in AV cushion explants. (A,B) Gata4T2delAVC explants produced markedly fewer mesenchymal cells than control explants.(C) Quantitation of the number of cells with mesenchymal morphology from control and Gata4T2del explants (n=3).(D,E) Failure of endocardial activation in Gata4T2del AV explant cultures. In control embryos,Pecam-expressing endothelial cells lost cell-cell contacts and adopted a rounded shape (arrow). Some rounded cells expressed SMA (white arrowhead) and acquired a mesenchymal morphology (yellow arrowhead). In mutant explants,endothelial cells remained in a sheet (star) and did not adopt a rounded shape. No mesenchymal cells or cells positive for both Pecam and SMA were detected (n=4). Myocardium (M) of control and mutant explants stained positive for SMA.

Fig. 4.

Endocardial Gata4 activity is required for mesenchyme formation in AV cushion explants. (A,B) Gata4T2delAVC explants produced markedly fewer mesenchymal cells than control explants.(C) Quantitation of the number of cells with mesenchymal morphology from control and Gata4T2del explants (n=3).(D,E) Failure of endocardial activation in Gata4T2del AV explant cultures. In control embryos,Pecam-expressing endothelial cells lost cell-cell contacts and adopted a rounded shape (arrow). Some rounded cells expressed SMA (white arrowhead) and acquired a mesenchymal morphology (yellow arrowhead). In mutant explants,endothelial cells remained in a sheet (star) and did not adopt a rounded shape. No mesenchymal cells or cells positive for both Pecam and SMA were detected (n=4). Myocardium (M) of control and mutant explants stained positive for SMA.

Having established an essential role for Gata4 in the transformation of AVC endocardial cells to mesenchymal cells, we sought to elucidate the mechanism by which Gata4 acts in this process. Because adjacent myocardium is known to influence endocardial EMT, we investigated whether the endocardial inactivation of Gata4 blocked EMT in a non-cell-autonomous manner. Myocardial differentiation and specification of atrioventricular canal myocardium remained intact, as indicated by the patterns of expression of sarcomeric myosin, the myocardial specific transcription factor Nkx2-5, the chamber myocardium marker Nppa, and the AVC myocardial marker Tbx2 (Fig. 5; data not shown). Myocardial expression of Bmp2 and Tgfβ2, known paracrine activators of EMT(Nakajima et al., 2000b; Rivera-Feliciano and Tabin,2006; Sugi et al.,2004), was unchanged in Gata4T2del hearts(data not shown). Furthermore, culture of control AVC explants directly next to Gata4T2del explants did not rescue the EMT defect in mutant explants. Similarly, culture of explants in the presence of a mixture of growth factors known to promote EMT (Tgfβ2, Bmp2, heregulin and hyaluoronic acid) did not rescue the EMT defect in mutant explants, but did robustly stimulate the formation of mesenchymal cells in control explants. These data suggest that the phenotype of Gata4T2delexplants was not due to an absence of diffusible factor(s) produced by the myocardium that promote EMT.

We also considered the possibility that Gata4 could regulate EMT through modulation of the extracellular matrix, which is necessary for cushion mesenchyme formation (Camenisch et al.,2000). However, the extracellular matrix of the AV cushions was still present in Gata4T2del hearts, as assessed by binding of the stain Alcian Blue to acidic glycosaminoglycans present in the cushion extracellular matrix (Fig. 5D). Moreover, addition hyaluronic acid directly to explant cultures did not rescue the EMT defect of mutant explants (data not shown).

Gata4 regulates expression of Erbb3

We next turned our attention to cell-autonomous mechanisms that could account for the loss of EMT in Gata4T2del embryos. Downregulation of Snail as a result of Notch signaling leads to downregulation of VE-cadherin (Cdh5), and is necessary for endocardial EMT (Timmerman et al.,2004). In Gata4T2del hearts, we did not find altered expression of components of this pathway (Notch1, HRT1 and Snail) by in situ hybridization(Fig. 5C; data not shown) or of Cdh5 by immunohistochemistry (Fig. 5E).

To identify genes whose altered expression might contribute to the Gata4T2del phenotype, we performed genome-wide expression analysis using microarrays. We isolated RNA from the AV region of mutant and control E9.5 hearts (between 30 and 33 somites), and used the RNA to probe Affymetrix microarrays. We used the SAM algorithm(Tusher et al., 2001) to identify genes with significantly altered expression(Table 2). For a subset of these, we used qRT-PCR to validate differences in gene expression between mutant and control tissue (Fig. 6A). Out of 11 genes tested, qRT-PCR confirmed differential gene expression in four: Erbb3, thrombospondin 1 (Thbs1), plexin C1 (Plxnc1), and tenascin C (Tnc; Fig. 6A). We also confirmed differential expression of Erbb3, Thbs1 and Plxnc1 by in situ hybridization (Fig. 6B).

Table 2.

Genes differentially expressed between control and Gata4T2del AVCs

RankProbe set IDGene symbolGene nameUniGene IDFold change
Downregulated genes      
1418788_at Tek Endothelial-specific receptor tyrosine kinase Mm.14313 −1.6 
1423213_at Plxnc1a,c Plexin C1 Mm.256712 −1.8 
1434606_at Erbb3a,c v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (avian) Mm.29023 −2.1 
1421811_at Thbs1a,c Thrombospondin 1 Mm.4159 −2.2 
1426294_at Hapln1 Hyaluronan and proteoglycan link protein 1 Mm.266790 −1.8 
1456144_at Nav3 Neuron navigator 3 Mm.225050 −1.8 
1416342_at Tnca Tenascin C Mm.980 −2.0 
1444403_at Cbfa2t2h Core-binding factor, runt domain, alpha subunit 2, translocated to, 2 homolog(human) (Cbfa2t2h), mRNA Mm.29914 −1.5 
1449303_at Sesn3 Sestrin 3 Mm.227443 −1.7 
10 1422286_a_at Tgif TG interacting factor Mm.101034 −2.2 
11 1421908_a_at Tcf12 Transcription factor 12 Mm.171615 −1.6 
12 1448293_at Ebf1 Early B-cell factor 1 Mm.215971 −1.5 
13 1426295_at Hapln 1 Hyaluronan and proteoglycan link protein 1 Mm.266790 −1.9 
14 1442019_at 16 days neonate cerebellum cDNA, RIKEN full-length enriched library,clone:9630033M11 product: unclassifiable, full insert sequence Mm.152121 −2.7 
15 1435727_s_at D15Ertd366e DNA segment, Chr 15, ERATO Doi 366, expressed Mm.33207 −1.5 
16 1452366_at 4732435N03Rik RIKEN cDNA 4732435N03 gene Mm.334569 −2.6 
17 1432029_a_at Smap 1 Stromal membrane-associated protein 1 Mm.329963 −1.9 
18 1434957_at Cdon Cell adhesion molecule-related/downregulated by oncogenes Mm.80509 −1.8 
19 1434510_at Papss2 3′-phosphoadenosine 5′-phosphosulfate synthase 2 Mm.203916 −3.9 
20 1437401_at Igf1b Insulin-like growth factor 1 Mm.268521 −2.0 
21 1434413_at Transcribed locus Mm.380595 −2.1 
22 1451740_at Paip 1 Polyadenylate binding protein-interacting protein 1 Mm.132584 −2.0 
23 1439641_at LOC553090 Hypothetical LOC553090 −2.4 
24 1418120_at Rbm8a RNA binding motif protein 8a Mm.261972 −1.5 
25 1426862_at 9130023F12Rik RIKEN cDNA 9130023F12 gene Mm.356580 −1.7 
26 1417625_s_at Cmkor1 Chemokine orphan receptor 1 Mm.6522 −2.3 
27 1421917_at Pdgfra Platelet derived growth factor receptor, alpha polypeptide Mm.221403 −1.7 
Upregulated genes      
1449187_at Pdgfaa Platelet derived growth factor, alpha Mm.2675 1.6 
1439713_at Itga1 Integrin alpha 1 1.5 
1423100_at Fos FBJ osteosarcoma oncogene Mm.246513 1.9 
1426114_at Hnrpab Heterogeneous nuclear ribonucleoprotein A/B Mm.280842 1.7 
1421126_at Ryr2 Ryanodine receptor 2, cardiac Mm.239871 1.7 
1433657_at A130092J06Rik RIKEN cDNA A130092J06 gene Mm.250441 1.5 
1415949_at Cpe Carboxypeptidase E Mm.31395 1.5 
1419647_a_at ler3 Immediate early response 3 Mm.25613 1.6 
1456768_a_at 1.8 
10 1430781_at Ak7 Adenylate kinase 7 Mm.36006 1.6 
11 1425196_a_at Hint2 Histidine triad nucleotide binding protein 2 Mm.25285 1.5 
12 1420991_at Ankrd1b Ankyrin repeat domain 1 (cardiac muscle) Mm.10279 1.8 
13 1458917_at 1.9 
14 1439518_at 1.8 
15 1449897_a_at Mtcp1 Mature T-cell proliferation 1 Mm.16366 1.5 
16 1460519_a_at Mettl5 Methyltransferase like 5 Mm.268657 1.7 
17 1447845_s_at Vnn1 Vanin 1 Mm.27154 1.7 
18 1445938_at 5930427L02Rik RIKEN cDNA 5930427L02 gene Mm.379523 1.7 
19 1418486_at Vnn1 Vanin 1 Mm.27154 1.5 
20 1422529_s_at Casq2 Calsequestrin 2 Mm.15343 1.6 
21 1429317_at Qrsl1 Glutaminyl-tRNA synthase (glutamine-hydrolyzing)-like 1 Mm.5904 1.6 
22 1417313_at Lsm7 LSM7 homolog, U6 small nuclear RNA associated (S. cerevisiae) Mm.379101 3.8 
23 1451370_at Adult male kidney cDNA, RIKEN full-length enriched library, clone:0610042I08 product:RNA component of mitochondrial RNAase P, 1, full insert sequence Mm.289109 1.7 
24 1430787_at 2310050B05Rik RIKEN cDNA 2310050B05 gene 1.6 
25 1455251_at Itga1 Integrin alpha 1 1.6 
26 1431248_at 5031426D15Rik RIKEN cDNA 5031426D15 gene 1.6 
27 1426902_at Coq6 Coenzyme Q6 homolog (yeast) Mm.280062 1.6 
28 1437904_at MGI:2387367 Developmentally regulated RNA binding protein 1 Mm.33310 2.1 
29 1428960_at 4933434I06Rik RIKEN cDNA 4933434I06 gene Mm.27658 1.5 
30 1417150_at Slc6a4 Solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mm.300318 2.2 
31 1455227_at Aadacl1 Arylacetamide deacetylase-like 1 Mm.24576 1.6 
32 1434865_a_at Exoc7 Exocyst complex component 7 Mm.22530 1.7 
33 1435436_at Transcribed locus Mm.234875 2.2 
34 1417713_at Eif2s2 Eukaryotic translation initiation factor 2, subunit 2 (beta) Mm.383218 1.7 
35 1452840_at 1500009L16Rik RIKEN cDNA 1500009L16 gene Mm.271188 2.4 
36 1440397_at 2.0 
37 1430023_at 5133400G04Rik RIKEN cDNA 5133400G04 gene Mm.151498 1.6 
38 1427263_at Xist Inactive X specific transcripts 1.9 
39 1449146_at Notch4 Notch gene homolog 4 (Drosophila) Mm.173813 2.8 
40 1436723_at Fshprh1 FSH primary response 1 Mm.34903 1.6 
RankProbe set IDGene symbolGene nameUniGene IDFold change
Downregulated genes      
1418788_at Tek Endothelial-specific receptor tyrosine kinase Mm.14313 −1.6 
1423213_at Plxnc1a,c Plexin C1 Mm.256712 −1.8 
1434606_at Erbb3a,c v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (avian) Mm.29023 −2.1 
1421811_at Thbs1a,c Thrombospondin 1 Mm.4159 −2.2 
1426294_at Hapln1 Hyaluronan and proteoglycan link protein 1 Mm.266790 −1.8 
1456144_at Nav3 Neuron navigator 3 Mm.225050 −1.8 
1416342_at Tnca Tenascin C Mm.980 −2.0 
1444403_at Cbfa2t2h Core-binding factor, runt domain, alpha subunit 2, translocated to, 2 homolog(human) (Cbfa2t2h), mRNA Mm.29914 −1.5 
1449303_at Sesn3 Sestrin 3 Mm.227443 −1.7 
10 1422286_a_at Tgif TG interacting factor Mm.101034 −2.2 
11 1421908_a_at Tcf12 Transcription factor 12 Mm.171615 −1.6 
12 1448293_at Ebf1 Early B-cell factor 1 Mm.215971 −1.5 
13 1426295_at Hapln 1 Hyaluronan and proteoglycan link protein 1 Mm.266790 −1.9 
14 1442019_at 16 days neonate cerebellum cDNA, RIKEN full-length enriched library,clone:9630033M11 product: unclassifiable, full insert sequence Mm.152121 −2.7 
15 1435727_s_at D15Ertd366e DNA segment, Chr 15, ERATO Doi 366, expressed Mm.33207 −1.5 
16 1452366_at 4732435N03Rik RIKEN cDNA 4732435N03 gene Mm.334569 −2.6 
17 1432029_a_at Smap 1 Stromal membrane-associated protein 1 Mm.329963 −1.9 
18 1434957_at Cdon Cell adhesion molecule-related/downregulated by oncogenes Mm.80509 −1.8 
19 1434510_at Papss2 3′-phosphoadenosine 5′-phosphosulfate synthase 2 Mm.203916 −3.9 
20 1437401_at Igf1b Insulin-like growth factor 1 Mm.268521 −2.0 
21 1434413_at Transcribed locus Mm.380595 −2.1 
22 1451740_at Paip 1 Polyadenylate binding protein-interacting protein 1 Mm.132584 −2.0 
23 1439641_at LOC553090 Hypothetical LOC553090 −2.4 
24 1418120_at Rbm8a RNA binding motif protein 8a Mm.261972 −1.5 
25 1426862_at 9130023F12Rik RIKEN cDNA 9130023F12 gene Mm.356580 −1.7 
26 1417625_s_at Cmkor1 Chemokine orphan receptor 1 Mm.6522 −2.3 
27 1421917_at Pdgfra Platelet derived growth factor receptor, alpha polypeptide Mm.221403 −1.7 
Upregulated genes      
1449187_at Pdgfaa Platelet derived growth factor, alpha Mm.2675 1.6 
1439713_at Itga1 Integrin alpha 1 1.5 
1423100_at Fos FBJ osteosarcoma oncogene Mm.246513 1.9 
1426114_at Hnrpab Heterogeneous nuclear ribonucleoprotein A/B Mm.280842 1.7 
1421126_at Ryr2 Ryanodine receptor 2, cardiac Mm.239871 1.7 
1433657_at A130092J06Rik RIKEN cDNA A130092J06 gene Mm.250441 1.5 
1415949_at Cpe Carboxypeptidase E Mm.31395 1.5 
1419647_a_at ler3 Immediate early response 3 Mm.25613 1.6 
1456768_a_at 1.8 
10 1430781_at Ak7 Adenylate kinase 7 Mm.36006 1.6 
11 1425196_a_at Hint2 Histidine triad nucleotide binding protein 2 Mm.25285 1.5 
12 1420991_at Ankrd1b Ankyrin repeat domain 1 (cardiac muscle) Mm.10279 1.8 
13 1458917_at 1.9 
14 1439518_at 1.8 
15 1449897_a_at Mtcp1 Mature T-cell proliferation 1 Mm.16366 1.5 
16 1460519_a_at Mettl5 Methyltransferase like 5 Mm.268657 1.7 
17 1447845_s_at Vnn1 Vanin 1 Mm.27154 1.7 
18 1445938_at 5930427L02Rik RIKEN cDNA 5930427L02 gene Mm.379523 1.7 
19 1418486_at Vnn1 Vanin 1 Mm.27154 1.5 
20 1422529_s_at Casq2 Calsequestrin 2 Mm.15343 1.6 
21 1429317_at Qrsl1 Glutaminyl-tRNA synthase (glutamine-hydrolyzing)-like 1 Mm.5904 1.6 
22 1417313_at Lsm7 LSM7 homolog, U6 small nuclear RNA associated (S. cerevisiae) Mm.379101 3.8 
23 1451370_at Adult male kidney cDNA, RIKEN full-length enriched library, clone:0610042I08 product:RNA component of mitochondrial RNAase P, 1, full insert sequence Mm.289109 1.7 
24 1430787_at 2310050B05Rik RIKEN cDNA 2310050B05 gene 1.6 
25 1455251_at Itga1 Integrin alpha 1 1.6 
26 1431248_at 5031426D15Rik RIKEN cDNA 5031426D15 gene 1.6 
27 1426902_at Coq6 Coenzyme Q6 homolog (yeast) Mm.280062 1.6 
28 1437904_at MGI:2387367 Developmentally regulated RNA binding protein 1 Mm.33310 2.1 
29 1428960_at 4933434I06Rik RIKEN cDNA 4933434I06 gene Mm.27658 1.5 
30 1417150_at Slc6a4 Solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mm.300318 2.2 
31 1455227_at Aadacl1 Arylacetamide deacetylase-like 1 Mm.24576 1.6 
32 1434865_a_at Exoc7 Exocyst complex component 7 Mm.22530 1.7 
33 1435436_at Transcribed locus Mm.234875 2.2 
34 1417713_at Eif2s2 Eukaryotic translation initiation factor 2, subunit 2 (beta) Mm.383218 1.7 
35 1452840_at 1500009L16Rik RIKEN cDNA 1500009L16 gene Mm.271188 2.4 
36 1440397_at 2.0 
37 1430023_at 5133400G04Rik RIKEN cDNA 5133400G04 gene Mm.151498 1.6 
38 1427263_at Xist Inactive X specific transcripts 1.9 
39 1449146_at Notch4 Notch gene homolog 4 (Drosophila) Mm.173813 2.8 
40 1436723_at Fshprh1 FSH primary response 1 Mm.34903 1.6 

Genes differentially expressed between between control (n=2) and mutant (n=4) were identified by SAM (Δ>0.4 and fold-change>1.5) and ranked by the relative difference score.

a

Differential expression validated by qRT-PCR (P<0.05).

b

Differential expression failed validation by qRT-PCR(P>0.05).

c

Differential expression validated by in situ hybridization.

The downregulation of Erbb3 in Gata4T2del AV tissue was of particular interest. Egf family ligands signal through Erbb3 to activate Ras and promote EMT, and in the absence of Erbb3 AV endothelium fails to form cushion mesenchyme(Camenisch et al., 2002). We confirmed downregulation of Erbb3 transcripts by qRT-PCR(Fig. 6A) and in situ hybridization (Fig. 6B), and downregulation of Erbb3 protein by western blotting(Fig. 6C).

To obtain further evidence that Gata4 regulates Erbb3expression, we investigated whether ectopic expression of Gata4 in endothelial cells that normally lack this transcription factor is sufficient to activate Erbb3 expression. Adenoviral expression of Gata4in human umbilical vein endothelial cells (HUVEC) caused a 5-fold upregulation of Erbb3 expression compared with cells treated with a GFP-expressing adenovirus (Fig. 6D). The degree of upregulation is likely to be even higher in Gata4-expressing cells, as adenovirus transduced only 15-20% of cells. Conversely, we investigated whether Gata factors are necessary for Erbb3 expression in at least some cellular contexts. For this experiment, we used BT20 breast carcinoma cells, which natively express both Gata4 and Erbb3 proteins (Bouchard et al.,2005) (data not shown). Expression of a dominant-negative Gata4DBD-engrailed fusion protein strongly decreased Erbb3 transcript levels(Fig. 6E).

To determine whether Gata4 regulates Erbb3 expression at the level of transcription, we used rVista 2.0(Loots and Ovcharenko, 2004)to find Erbb3 non-coding sequences conserved between mouse and human. Conserved non-coding sequences upstream of the putative transcriptional start site and within the first intron were used to drive expression of luciferase reporters. We found that co-transfected Gata4 stimulated transcription from both the promoter and the intronic enhancer(Fig. 6F). By contrast, the truncated protein produced by the Gata4Δex2 allele failed to stimulate transcription from these Erbb3 regulatory elements(Fig. 6F). These regulatory elements contain three predicted GATA motifs conserved between mouse and human. In mobility shift experiments, the two consensus sites in the enhancer strongly bound Gata4, whereas the site in the promoter (nonconsensus GATG site) did not. Mutation of these sites did not significantly alter transcriptional stimulation by Gata4 (see Fig. S5 in the supplementary material).

Gata4 regulates an Erbb3-Erk pathway required for EMT

Next, we wished to determine whether Gata4T2del AV endothelium was functionally deficient in the transduction of Egf signals. We treated E9.5 Gata4T2del and control embryos with the Erbb3 ligand heregulin, and measured the phosphorylation of Erk,which is activated downstream of Ras. In heregulin-treated control embryos, activated Erk was readily observed in AV cushion endothelium(arrowheads, Fig. 7A, middle panel). By contrast, in heregulin-treated Gata4T2delembryos, Erk activation was strongly reduced in AV cushion endothelium(arrowheads, Fig. 7A, bottom panel). Erk activation in myocardium did not differ between genotypes (arrows, Fig. 7A), indicating that the downregulation in cushion endothelium was specific and unlikely to be due to technical factors.

Fig. 5.

Expression of myocardial and endocardial markers in Gata4T2del mutant hearts. (A,B) In situ hybridization for Nppa (A) and Tbx2 (B) in E9.5 embryos,showing intact expression in Gata4T2del hearts. Asterisk indicates the EC. (C) In situ hybridization for Notch1 in E10.5 embryos, showing unperturbed expression in AV endocardium (arrowheads).(D) Alcian Blue staining of acidic glycosaminoglycans, including hyaluronic acid, in E10.5 control and mutant cushions. (E,F)Cdh5 immunostaining of E9.5 embryo sections. Note the bunched up phenotype of the mutant AV endocardium compared with control AV endocardium (arrowheads). Panels in F are higher magnifications of the sections shown in E.

Fig. 5.

Expression of myocardial and endocardial markers in Gata4T2del mutant hearts. (A,B) In situ hybridization for Nppa (A) and Tbx2 (B) in E9.5 embryos,showing intact expression in Gata4T2del hearts. Asterisk indicates the EC. (C) In situ hybridization for Notch1 in E10.5 embryos, showing unperturbed expression in AV endocardium (arrowheads).(D) Alcian Blue staining of acidic glycosaminoglycans, including hyaluronic acid, in E10.5 control and mutant cushions. (E,F)Cdh5 immunostaining of E9.5 embryo sections. Note the bunched up phenotype of the mutant AV endocardium compared with control AV endocardium (arrowheads). Panels in F are higher magnifications of the sections shown in E.

We then investigated whether defective Erk activation downstream of Erbb3 might account for the marked reduction in EMT seen in the Gata4T2del AV cushions. We treated control and mutant explants with U0126, a selective inhibitor of Erk activation. We found that U0126 strongly reduced the extent of EMT in wild-type explants(Fig. 7B). We reasoned that if Gata4 and Erk are mutually required for EMT, then partial antagonism of each would inhibit EMT. To test this hypothesis, we generated AVC explants homozygous for Gata4H, a Gata4allele that expresses reduced levels of Gata4 protein(Pu et al., 2004). We treated Gata4H/H and littermate control explants with a 50%inhibitory concentration of U0126 (400 nM). In the presence of vehicle, Gata4H/H and control explants generated comparable numbers of mesenchymal cells. However, the number of mesenchymal cells generated by Gata4H/H explants treated with 400 nM U0126 was significantly reduced when compared with the number generated by similarly treated control explants (Fig. 7C). This result is consistent with a model in which Gata4 and Erk act in the same genetic pathway to promote EMT.

Gata4 is required for growth and remodeling of the AV cushions

Constitutive, partial loss of Gata4 function, as a result of either decreased protein expression (Gata4H/H) or a point mutation abrogating Fog1 and Fog2 (Zfpm1 and Zfpm2, respectively-Mouse Genome Informatics) interaction(Gata4Ki/Ki), resulted in embryonic lethality between E12.5-E16.5 (Crispino et al.,2001; Pu et al.,2004). These embryos had common atrioventricular canal defects,indicating a severe abnormality of EC maturation such that the superior and inferior cushions failed to fuse and divide the ventricular inflow tract into separate inlets for the right and left ventricle. This abnormality could, in principle, have been caused by impaired Gata4 function in the endocardium, or by defective Gata4 function in the myocardium with secondary abnormalities in the EC due to abnormal paracrine signaling. To determine whether there is a cell-autonomous requirement for Gata4 in later atrioventricular valve maturation, we generated embryos with the genotype Gata4flox/Ki; T2Cre+ (abbreviated Gata4T2del/Ki). In these embryos, Gata4flox complements Gata4Ki except in the endothelium and its derivatives, where T2Cre-mediated recombination inactivates it.

Gata4Ki/flox; T2Cre- control mice were present at weaning at the expected Mendelian frequency and had no obvious heart defects. By contrast, Gata4T2del/Ki embryos were present in the expected Mendelian ratio at E16.5, but were not present at weaning (out of eight litters genotyped), indicating lethality in late gestation or in the perinatal period. Mutant embryos examined at E16.5 had severe peripheral hemorrhage and edema, consistent with heart failure (data not shown). At E12.5, the heart (Fig. 8A-D) and the liver appeared normal(Fig. 8A,B). However, in later stage embryos, the AV valve leaflets were hypoplastic, and failure of fusion of the superior and inferior AV cushions resulted in a common atrioventricular canal defect (asterisk, Fig. 8E,F). Cell death was not increased in mutant cushions, as measured by TUNEL staining at E12.5 and E15.5 (data not shown). However, cell proliferation, as measured by BrdU uptake, was decreased in mutant cushion mesenchyme at E12.5 and E13.5 (Fig. 8G, data not shown). These data suggest that Gata4-Foginteraction within endocardium-derived cells is required for normal proliferation of the AV cushions.

Fig. 6.

Gata4 regulates Erbb3 expression. (A)qRT-PCR of control and mutant AVC samples. Gene expression was normalized to Gapdh. *P<0.05, n=3 per group.(B) In situ hybridization demonstrating downregulation of Thbs1,Plxnc1 and Erbb3 in Gata4T2del EC. Arrowheads indicate AV cushions. (C) Downregulation of Erbb3 protein in AVC whole cell lysates by western blotting. Each sample contained AVCs pooled from 10 hearts. Expression was quantitated by densitometry and normalized to Gapdh. (D) Adenoviral expression of Gata4 upregulated Erbb3 in HUVEC cells, compared with GFP expression from a control virus. Erbb3 expression was measured by qRT-PCR and normalized to Gapdh. The graph shows the average of three independent experiments.(E) BT20 cells were co-transfected with an EGFP expression vector and either Gata4DBD-engrailed or an empty expression vector. After sorting for transfected (GFP-expressing) cells, Erbb3 expression was measured by qRT-PCR and normalized to Gapdh. Results are representative of two independent experiments. (F) Constructs containing Erbb3promoter (-1356 to +214) and intron 1 enhancer (+104 to +3076) sequences driving luciferase were cotransfected with Gata4 or Gata4Δex2expression constructs into BT20 cells. Luciferase activity was normalized to pRL-null. Results are the average of five (enhancer) or eight (promoter)independent experiments (*P<0.05).

Fig. 6.

Gata4 regulates Erbb3 expression. (A)qRT-PCR of control and mutant AVC samples. Gene expression was normalized to Gapdh. *P<0.05, n=3 per group.(B) In situ hybridization demonstrating downregulation of Thbs1,Plxnc1 and Erbb3 in Gata4T2del EC. Arrowheads indicate AV cushions. (C) Downregulation of Erbb3 protein in AVC whole cell lysates by western blotting. Each sample contained AVCs pooled from 10 hearts. Expression was quantitated by densitometry and normalized to Gapdh. (D) Adenoviral expression of Gata4 upregulated Erbb3 in HUVEC cells, compared with GFP expression from a control virus. Erbb3 expression was measured by qRT-PCR and normalized to Gapdh. The graph shows the average of three independent experiments.(E) BT20 cells were co-transfected with an EGFP expression vector and either Gata4DBD-engrailed or an empty expression vector. After sorting for transfected (GFP-expressing) cells, Erbb3 expression was measured by qRT-PCR and normalized to Gapdh. Results are representative of two independent experiments. (F) Constructs containing Erbb3promoter (-1356 to +214) and intron 1 enhancer (+104 to +3076) sequences driving luciferase were cotransfected with Gata4 or Gata4Δex2expression constructs into BT20 cells. Luciferase activity was normalized to pRL-null. Results are the average of five (enhancer) or eight (promoter)independent experiments (*P<0.05).

We used a T2Cre transgene to recombine a conditional Gata4 allele (Gata4flox) within endothelial-derived cells and demonstrated that Gata4 is required at two stages of AVC morphogenesis. First, Gata4 is necessary for endothelial EMT to form AV cushion mesenchyme. Second, Gata4 is necessary for the growth and remodeling of the AV cushions, after they have been populated by mesenchymal cells. Gata4 interaction with Fog cofactors within endothelial-derived cells is dispensable for the former, but required for the latter.

The AV cushions are largely formed by mesenchyme derived from endothelial cells (see Fig. 1B, Fig. 3). Inactivation of Gata4 within endothelial-derived cells results in a marked decrease in the number of mesenchymal cells in the AV cushions(Fig. 2). By contrast, the OT cushions are formed by cells derived from at least two sources. OT endothelium undergoes EMT to form the mesenchyme of the proximal OT cushions, whereas neural crest contributes to the mid- and distal OT cushions(Fig. 1B; Fig. 3; see also Fig. S2 in the supplementary material). Gata4 is expressed in both the neural crest-derived and endothelial-derived portions of the OT cushions. However,its expression in the neural crest-derived portion is not required for normal OT development, as Gata4 inactivation by Wnt1Cre was compatible with normal survival and normal OT morphogenesis (W.T.P.,unpublished). Gata4 expression was required to form the proximal,endothelial-derived portion of the OT cushion(Fig. 3). Because Gata4T2del embryos do not survive to a stage at which mature OT valves are evident, we were unable to determine what effect the loss of this portion of OT cushion has on OT valve development. Congenital abnormalities of the pulmonary valve are associated with human GATA4mutation (Garg et al., 2003; Hirayama-Yamada et al., 2005; Okubo et al., 2004),suggesting that Gata4 activity within the proximal, endothelial-derived portion of the OT cushions may be important for development of the OT valves.

Gata4 function in formation of AV mesenchyme

Inactivation of Gata4 within endothelial-derived cells blocked endocardial EMT, resulting in a paucity of mesenchymal cells within the AV cushions. This was associated with strong downregulation of Erbb3(Fig. 6). Using heterologous expression systems, we show that Gata4 modulates Erbb3 transcript levels and transcriptional activity of Erbb3 regulatory elements(Fig. 6F). Although these regulatory elements contain evolutionarily conserved GATA binding sites, these binding sites were not required for transcriptional stimulation by Gata4 in vitro (see Fig. S5 in the supplementary material). These data suggest that Gata4 may regulate Erbb3 indirectly. Alternatively, these findings might represent limitations of the in vitro assay system. Additional in vivo studies of Erbb3 regulatory elements will be necessary to elucidate the mechanism by which Gata4 regulates Erbb3 in the endocardial cushions.

Fig. 7.

Gata4 regulates an Erbb3-Erk pathway required for EMT. (A)E9.5 control or Gata4T2del embryos were cultured in M199 containing 100 ng/ml heregulin. A phosphospecific Erk antibody (pErk; green staining) showed defective Erk ativation in the endocardium of mutant embryos(arrowheads, bottom panel) compared with control embryos (arrowheads, middle panel). Myocardial pErk levels were unchanged between genotypes. To demonstrate antibody specificity, a control embryo was incubated with U0126, a selective inhibitor of Erk activation (top panel). (B) U0126 (10 μM)blocked the formation of mesenchymal cells in wild-type AV explants.(C) Synthetic phenotype resulting from the combination of reduced Gata4 expression and partial Erk inhibition. Explants from embryos homozygous for a hypomorphic Gata4 allele (H/H) underwent EMT at the same rate as controls in the presence of vehicle (NS, no significant difference). However, in the presence of U0126 at a 50% inhibitory dose, EMT was strongly inhibited in hypomorphic explants (*P<0.05 compared with control-treated H/H or U0126-treated wt/wt; n=4). The extent of EMT was measured by the number of cells with mesenchymal morphology that invaded the collagen gel.

Fig. 7.

Gata4 regulates an Erbb3-Erk pathway required for EMT. (A)E9.5 control or Gata4T2del embryos were cultured in M199 containing 100 ng/ml heregulin. A phosphospecific Erk antibody (pErk; green staining) showed defective Erk ativation in the endocardium of mutant embryos(arrowheads, bottom panel) compared with control embryos (arrowheads, middle panel). Myocardial pErk levels were unchanged between genotypes. To demonstrate antibody specificity, a control embryo was incubated with U0126, a selective inhibitor of Erk activation (top panel). (B) U0126 (10 μM)blocked the formation of mesenchymal cells in wild-type AV explants.(C) Synthetic phenotype resulting from the combination of reduced Gata4 expression and partial Erk inhibition. Explants from embryos homozygous for a hypomorphic Gata4 allele (H/H) underwent EMT at the same rate as controls in the presence of vehicle (NS, no significant difference). However, in the presence of U0126 at a 50% inhibitory dose, EMT was strongly inhibited in hypomorphic explants (*P<0.05 compared with control-treated H/H or U0126-treated wt/wt; n=4). The extent of EMT was measured by the number of cells with mesenchymal morphology that invaded the collagen gel.

Fig. 8.

AV valve remodeling requires Gata4 expression in endocardium and its derivatives.Gata4T2del/Ki(Gata4Ki/flox; Tie2Cre+) embryos are deficient for Gata4-Fog interaction in the endocardium and its derivatives. (A-D)AV cushions were normally populated by mesenchymal cells at E12.5 (asterisks,C,D), indicating that the Gata4-Fog interaction is not essential for EMT. The liver (L) was normal in size and morphology. Boxed regions of A,B are enlarged in C,D. (E,F) A common atrioventricular canal (asterisk) was present in all Gata4T2del/Ki embryos at E14.5, whereas in control embryos the AVC was septated by the fusion of the superior and inferior cushions. (G) Decreased proliferation of cushion mesenchyme in Gata4T2del/Ki embryos. Proliferation was measured as the percentage of nuclei that stained positive for BrdU (red). Nuclei were stained with DAPI. Myocyctes were labeled with desmin (green). Three sections from three different E12.5 embryos were examined per group by a blinded observer(*P<0.05).

Fig. 8.

AV valve remodeling requires Gata4 expression in endocardium and its derivatives.Gata4T2del/Ki(Gata4Ki/flox; Tie2Cre+) embryos are deficient for Gata4-Fog interaction in the endocardium and its derivatives. (A-D)AV cushions were normally populated by mesenchymal cells at E12.5 (asterisks,C,D), indicating that the Gata4-Fog interaction is not essential for EMT. The liver (L) was normal in size and morphology. Boxed regions of A,B are enlarged in C,D. (E,F) A common atrioventricular canal (asterisk) was present in all Gata4T2del/Ki embryos at E14.5, whereas in control embryos the AVC was septated by the fusion of the superior and inferior cushions. (G) Decreased proliferation of cushion mesenchyme in Gata4T2del/Ki embryos. Proliferation was measured as the percentage of nuclei that stained positive for BrdU (red). Nuclei were stained with DAPI. Myocyctes were labeled with desmin (green). Three sections from three different E12.5 embryos were examined per group by a blinded observer(*P<0.05).

Downregulation of Erbb3 in Gata4T2del AVCs was associated with impaired Erk activation in response to the Erbb3 ligand heregulin (Fig. 7A). Erk activation was necessary for endocardial EMT(Fig. 7B). Partial inhibition of both Gata4 and Erk impaired the formation of cushion mesenchyme(Fig. 7C). This synthetic phenotype suggests a strong interaction between Gata4- and Erk-dependent pathways. Collectively, these data are consistent with a model in which Gata4 functions upstream of an Erbb3-Ras-Erk pathway that is necessary for the formation of cushion mesenchyme. Intriguingly, Gata4 itself is activated by Erk phosphorylation (Liang et al.,2001), suggesting the possibility of a positive-feedback loop that promotes endocardial EMT.

Although Gata4 is required for AV EMT, Gata4 interaction with Fog is dispensable for this step of AV valve development. Embryos deficient in Fog1 or Fog2, or in Gata4-Fog interactions(Gata4Ki/Ki), did not show any defect in the generation of AV valve mesenchyme by EMT (Crispino et al., 2001; Katz et al.,2003; Tevosian et al.,2000), indicating that Gata4 regulation of AV cushion EMT does not require Fog interaction. Consistent with this conclusion, Gata4T2del/Ki EC were normally populated with mesenchymal cells (Fig. 8A-D).

Gata4 function in AV valve growth and remodeling

After AV cushion mesenchyme is formed by EMT, it rapidly proliferates to fill the expanding AV cushions with mesenchymal cells. The growing superior and inferior AV cushions meet and fuse, dividing the ventricular inlet into a right and left channel. Gata4 is necessary for this process, as a decrease in Gata4 levels or germline abrogation of the Gata4-Fog interaction resulted in an unseptated ventricular inlet(Crispino et al., 2001; Pu et al., 2004). Here, we show that Gata4-Fog interaction within endothelial-derived cells is required for septation of the ventricular inlet(Fig. 8).

This study demonstrates an essential role for expression of Gata4in the endothelium and its derivatives. During the initial steps of AV valve formation, Gata4 is necessary for the expression of Erbb3,which acts through a Ras-Erk pathway to promote EMT. During subsequent steps of AV valve maturation, Gata4, in cooperation with a Fog cofactor, promotes the growth and fusion of the AV cushions, resulting in the division of the ventricular inlet into two separate channels guarded by two AV valves. These data suggest that Gata4 mutations might contribute to EC defects in humans; indeed, a Gata4 mutation has been reported to occur in association with AV septal defects(Garg et al., 2003).

The authors thank J. L. Galloway for comments on the manuscript, and members of the Tabin, Cepko, Dymecki, Izumo and Pu labs for their advice and support. This work was supported by grants from the NIH (C.J.T., R01 HD0045499; W.T.P., K08 HL004387-04 and 1 PO1 HL074734) and by a charitable donation from Edward Marran and Karen Carpenter. J.R.-F. was supported by a predoctoral National Research Service Award.

Armstrong, E. J. and Bischoff, J. (
2004
). Heart valve development: endothelial cell signaling and differentiation.
Circ. Res.
95
,
459
-470.
Bouchard, M. F., Taniguchi, H. and Viger, R. S.(
2005
). Protein kinase A-dependent synergism between GATA factors and the nuclear receptor, liver receptor homolog-1, regulates human aromatase(CYP19) PII promoter activity in breast cancer cells.
Endocrinology
146
,
4905
-4916.
Brent, A. E., Schweitzer, R. and Tabin, C. J.(
2003
). A somitic compartment of tendon progenitors.
Cell
113
,
235
-248.
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.
Camenisch, T. D., Spicer, A. P., Brehm-Gibson, T., Biesterfeldt,J., Augustine, M. L., Calabro, A., Jr, Kubalak, S., Klewer, S. E. and McDonald, J. A. (
2000
). Disruption of hyaluronan synthase-2 abrogates normal cardiac morphogenesis and hyaluronan-mediated transformation of epithelium to mesenchyme.
J. Clin. Invest.
106
,
349
-360.
Camenisch, T. D., Schroeder, J. A., Bradley, J., Klewer, S. E. and McDonald, J. A. (
2002
). Heart-valve mesenchyme formation is dependent on hyaluronan-augmented activation of ErbB2-ErbB3 receptors.
Nat. Med.
8
,
850
-855.
Charron, F. and Nemer, M. (
1999
). GATA transcription factors and cardiac development.
Semin. Cell Dev. Biol.
10
,
85
-91.
Crispino, J. D., Lodish, M. B., Thurberg, B. L., Litovsky, S. H., Collins, T., Molkentin, J. D. and Orkin, S. H. (
2001
). Proper coronary vascular development and heart morphogenesis depend on interaction of GATA-4 with FOG cofactors.
Genes Dev.
15
,
839
-844.
Danielian, P. S., Muccino, D., Rowitch, D. H., Michael, S. K. and McMahon, A. P. (
1998
). Modification of gene activity in mouse embryos in utero by a tamoxifen-inducible form of Cre recombinase.
Curr. Biol.
8
,
1323
-1326.
Garg, V., Kathiriya, I. S., Barnes, R., Schluterman, M. K.,King, I. N., Butler, C. A., Rothrock, C. R., Eapen, R. S., Hirayama-Yamada,K., Joo, K. et al. (
2003
). GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5.
Nature
424
,
443
-447.
Gitler, A. D., Zhu, Y., Ismat, F. A., Lu, M. M., Yamauchi, Y.,Parada, L. F. and Epstein, J. A. (
2003
). Nf1 has an essential role in endothelial cells.
Nat. Genet.
33
,
75
-79.
Heikinheimo, M., Scandrett, J. M. and Wilson, D. B.(
1994
). Localization of transcription factor GATA-4 to regions of the mouse embryo involved in cardiac development.
Dev. Biol.
164
,
361
-373.
Hirayama-Yamada, K., Kamisago, M., Akimoto, K., Aotsuka, H.,Nakamura, Y., Tomita, H., Furutani, M., Imamura, S., Takao, A., Nakazawa, M. et al. (
2005
). Phenotypes with GATA4 or NKX2.5 mutations in familial atrial septal defect.
Am. J. Med. Genet. A
135
,
47
-52.
Katz, S. G., Williams, A., Yang, J., Fujiwara, Y., Tsang, A. P.,Epstein, J. A. and Orkin, S. H. (
2003
). Endothelial lineage-mediated loss of the GATA cofactor Friend of GATA 1 impairs cardiac development.
Proc. Natl. Acad. Sci. USA
100
,
14030
-14035.
Kisanuki, Y. Y., Hammer, R. E., Miyazaki, J., Williams, S. C.,Richardson, J. A. and Yanagisawa, M. (
2001
). Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo.
Dev. Biol.
230
,
230
-242.
Kuo, C. T., Morrisey, E. E., Anandappa, R., Sigrist, K., Lu, M. M., Parmacek, M. S., Soudais, C. and Leiden, J. M. (
1997
). GATA4 transcription factor is required for ventral morphogenesis and heart tube formation.
Genes Dev.
11
,
1048
-1060.
Lakkis, M. M. and Epstein, J. A. (
1998
). Neurofibromin modulation of ras activity is required for normal endocardial-mesenchymal transformation in the developing heart.
Development
125
,
4359
-4367.
Lee, K. H., Evans, S., Ruan, T. Y. and Lassar, A. B.(
2004
). SMAD-mediated modulation of YY1 activity regulates the BMP response and cardiac-specific expression of a GATA4/5/6-dependent chick Nkx2.5 enhancer.
Development
131
,
4709
-4723.
Lee, Y., Shioi, T., Kasahara, H., Jobe, S. M., Wiese, R. J.,Markham, B. E. and Izumo, S. (
1998
). The cardiac tissue-restricted homeobox protein Csx/Nkx2.5 physically associates with the zinc finger protein GATA4 and cooperatively activates atrial natriuretic factor gene expression.
Mol. Cell. Biol.
18
,
3120
-3129.
Liang, Q., Wiese, R. J., Bueno, O. F., Dai, Y. S., Markham, B. E. and Molkentin, J. D. (
2001
). The transcription factor gata4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes.
Mol. Cell. Biol.
21
,
7460
-7469.
Lobe, C. G., Koop, K. E., Kreppner, W., Lomeli, H.,Gertsenstein, M. and Nagy, A. (
1999
). Z/AP, a double reporter for cre-mediated recombination.
Dev. Biol.
208
,
281
-292.
Loots, G. G. and Ovcharenko, I. (
2004
). rVISTA 2.0: evolutionary analysis of transcription factor binding sites.
Nucleic Acids Res.
32
,
W217
-W221.
Mao, X., Fujiwara, Y. and Orkin, S. H. (
1999
). Improved reporter strain for monitoring Cre recombinase-mediated DNA excisions in mice.
Proc. Natl. Acad. Sci. USA
96
,
5037
-5042.
Molkentin, J. D., Lin, Q., Duncan, S. A. and Olson, E. N.(
1997
). Requirement of the transcription factor GATA4 for heart tube formation and ventral morphogenesis.
Genes Dev.
11
,
1061
-1072.
Morrisey, E. E., Ip, H. S., Tang, Z. and Parmacek, M. S.(
1997
). GATA-4 activates transcription via two novel domains that are conserved within the GATA-4/5/6 subfamily.
J. Biol. Chem.
272
,
8515
-8524.
Nakajima, H., Nakajima, H. O., Salcher, O., Dittie, A. S.,Dembowsky, K., Jing, S. and Field, L. J. (
2000a
). Atrial but not ventricular fibrosis in mice expressing a mutant transforming growth factor-beta(1) transgene in the heart.
Circ. Res.
86
,
571
-579.
Nakajima, Y., Yamagishi, T., Hokari, S. and Nakamura, H.(
2000b
). Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: roles of transforming growth factor(TGF)-beta and bone morphogenetic protein (BMP).
Anat. Rec.
258
,
119
-127.
Okubo, A., Miyoshi, O., Baba, K., Takagi, M., Tsukamoto, K.,Kinoshita, A., Yoshiura, K., Kishino, T., Ohta, T., Niikawa, N. et al.(
2004
). A novel GATA4 mutation completely segregated with atrial septal defect in a large Japanese family.
J. Med. Genet.
41
,
e97
.
Pelton, R. W., Nomura, S., Moses, H. L. and Hogan, B. L.(
1989
). Expression of transforming growth factor beta 2 RNA during murine embryogenesis.
Development
106
,
759
-767.
Pierpont, M. E., Markwald, R. R. and Lin, A. E.(
2000
). Genetic aspects of atrioventricular septal defects.
Am. J. Med. Genet.
97
,
289
-296.
Pu, W. T., Ishiwata, T., Juraszek, A. L., Ma, Q. and Izumo,S. (
2004
). GATA4 is a dosage-sensitive regulator of cardiac morphogenesis.
Dev. Biol.
275
,
235
-244.
Rivera-Feliciano, J. and Tabin, C. J. (
2006
). Bmp2 instructs cardiac progenitors to form the heart-valve-inducing field.
Dev. Biol.
295
,
580
-588.
Runyan, R. B. and Markwald, R. R. (
1983
). Invasion of mesenchyme into threedimensional collagen gels: a regional and temporal analysis of interaction in embryonic heart tissue.
Dev. Biol.
95
,
108
-114.
Sugi, Y., Yamamura, H., Okagawa, H. and Markwald, R. R.(
2004
). Bone morphogenetic protein-2 can mediate myocardial regulation of atrioventricular cushion mesenchymal cell formation in mice.
Dev. Biol.
269
,
505
-518.
Tanaka, M., Chen, Z., Bartunkova, S., Yamasaki, N. and Izumo,S. (
1999
). The cardiac homeobox gene Csx/Nkx2.5 lies genetically upstream of multiple genes essential for heart development.
Development
126
,
1269
-1280.
Tevosian, S. G., Deconinck, A. E., Tanaka, M., Schinke, M.,Litovsky, S. H., Izumo, S., Fujiwara, Y. and Orkin, S. H.(
2000
). FOG-2, a cofactor for GATA transcription factors, is essential for heart morphogenesis and development of coronary vessels from epicardium.
Cell
101
,
729
-739.
Timmerman, L. A., Grego-Bessa, J., Raya, A., Bertran, E.,Perez-Pomares, J. M., Diez, J., Aranda, S., Palomo, S., McCormick, F.,Izpisua-Belmonte, J. C. et al. (
2004
). Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation.
Genes Dev.
18
,
99
-115.
Tusher, V. G., Tibshirani, R. and Chu, G.(
2001
). Significance analysis of microarrays applied to the ionizing radiation response.
Proc. Natl. Acad. Sci. USA
98
,
5116
-5121.
Verzi, M. P., McCulley, D. J., De Val, S., Dodou, E. and Black,B. L. (
2005
). The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field.
Dev. Biol.
287
,
134
-145.
Watt, A. J., Battle, M. A., Li, J. and Duncan, S. A.(
2004
). GATA4 is essential for formation of the proepicardium and regulates cardiogenesis.
Proc. Natl. Acad. Sci. USA
101
,
12573
-12578.
Zeisberg, E. M., Ma, Q., Juraszek, A. L., Moses, K., Schwartz,R. J., Izumo, S. and Pu, W. T. (
2005
). Morphogenesis of the right ventricle requires myocardial expression of Gata4.
J. Clin. Invest.
115
,
1522
-1531.

Supplementary information