Pitx2 is a homeodomain transcription factor that is mutated in Rieger syndrome, a haploinsufficiency disorder affecting eyes and teeth. Pitx2 also has a postulated role in left-right axis determination. We assessed the requirements for Pitx2 directly by generating hypomorphic and null alleles. Heterozygotes for either allele have eye abnormalities consistent with Rieger syndrome. The ventral body wall fails to close in embryos homozygous for the null allele, leaving the heart and abdominal organs externalized and the body axis contorted. In homozygotes for either allele, the heart tube undergoes normal, rightward looping and the stomach is positioned normally. In contrast, homozygotes for both alleles exhibit right isomerization of the lungs. Thus, Pitx2 is required for left-right asymmetry of the lungs but not other organs. Homozygotes for either allele exhibit septal and valve defects, and null homozygotes have a single atrium proving that a threshold level of Pitx2 is required for normal heart development. Null homozygotes exhibit arrest of pituitary gland development at the committed Rathke pouch stage and eye defects including optic nerve coloboma and absence of ocular muscles. This allelic series establishes that Pitx2 is required for the development of mulitple organs in a dosage-sensitive manner.

The bicoid-like homeobox gene Pitx2 is a member of a multigene family with overlapping and distinctive expression patterns. Multiple members of this gene family have been identified in vertebrates including amphibians, fish, birds and mammals (Gage et al., 1999). Identification of Pitx in Drosophila implied an important evolutionary role for the Pitx gene family, yet Drosophila mutants have no obvious anomalies (Vorbruggen et al., 1997). Recent experiments in chick and frog have indicated a role for Pitx2 downstream of sonic hedgehog and nodal in a genetic pathway regulating laterality of heart, gut and other asymmetric organs (Logan et al., 1998; Piedra et al., 1998; Ryan et al., 1998; Yoshioka et al., 1998). Pitx2 expression is altered in mouse mutants with laterality defects, including iv, inv and a targeted mutant in Lefty-2, suggesting that Pitx2 participates in left-right determination in mammals as well (Meno et al., 1998; Piedra et al., 1998; Ryan et al., 1998).

The importance of each member of the Pitx gene family is emerging through analysis of human patients and targeted mutations in mice. PITX2 mutations cause Rieger syndrome, type I (RGS1, MIM 180500) (Semina et al., 1996), an autosomal dominant haploinsufficiency syndrome with defects in the anterior segment of the eye (cornea, trabecular meshwork and iris stroma), and other features of varying severity including cataracts, glaucoma, missing or misplaced teeth (hypodontia), umbilical abnormalities and cardiac defects (Jorgenson et al., 1978; Semina et al., 1996). PITX3 mutations result in autosomal dominant cataracts and anterior segment mesenchymal dysgenesis (MIM 107250) (Semina et al., 1998). In contrast to PITX2 and PITX3, Pitx1 mutations in mice are recessive, affecting development of the palate, mandible and limbs (Lanctot et al., 1999; Szeto et al., 1999).

Pitx2 is expressed in many tissues during development, including the left lateral plate mesoderm, derivatives of the first branchial arch, the eye, brain, pituitary gland, mandible, heart and limbs (Arakawa et al., 1998; Gage and Camper, 1997; Kitamura et al., 1997; Muccielli et al., 1996; Semina et al., 1996). The phenotypes of RGS patients with null alleles establish an essential role for this gene in eye, tooth and umbilical development, and glaucoma (Amendt et al., 1998). The occasional occurrence of isolated growth insufficiency and aortic arch defects in RGS patients suggests that PITX2 may also play a role in pituitary gland and heart development (Mammi et al., 1998; Sadeghi-Nedjad and Senior, 1974). However, the importance of PITX2 is still enigmatic because of the one remaining functional allele in these heterozygous patients.

We used gene targeting in embryonic stem (ES) cells, combined with cre-loxP technology to generate an allelic series of Pitx2 mutations of varying severity. Heterozygotes for hypomorphic (neo) or null (−) alleles for Pitx2 mimic some aspects of RGS, including eye and tooth defects. The phenotypes of homozygotes for both alleles establish that Pitx2 is essential for positioning and development of the heart and for lung asymmetry. While pituitary and eye development are grossly normal in neo/neo fetuses, −/− mice eye exhibit an early arrest in pituitary development and multiple eye defects. The differing sensitivity of various organs to Pitx2 deficiency demonstrates a tissue-specific dosage-dependence on Pitx2 for both laterality and organogenesis.

Targeting of Pitx2 gene

A P1 clone containing Pitx2 was obtained from a 129/OLA genomic library by screening with Pitx2 exon 5 PCR primers (Sternberg, 1992; Genomic Systems, Inc.; St. Louis, MO). Since herpes simplex virus thymidine kinase (HSV-TK) interferes with spermatogenesis (Al-Shawi et al., 1991), pFLOXΔRI was generated by deleting the HSV-TK sequences from pFLOX (Orban et al., 1992) by EcoRI digestion and religation. Three contiguous Pitx2 genomic fragments were inserted sequentially into pFLOXΔRI: a 631-bp fragment spanning the homeodomain-encoding exon 4 was placed between loxP2 and loxP3, a 1.7 kb fragment was placed upstream of loxP1 and the phosphoglycerate kinase-neoR cassette, and a 3.2 kb fragment was placed downstream of loxP3 (Fig. 1).

Fig. 1.

Pitx2 allelic series. (A) The structure of the targeting vector is shown enlarged in relationship to Pitx2+. The complete structure of Pitx2neo and abbreviated structures for Pitx2null and Pitx2flox are also shown at the same scale as Pitx2+. Exon (boxes) and coding sequence (black boxes). (B) Southern analysis illustrating homologous recombination 5′ of the neoR cassette in two cell lines heterozygous for Pitx2neo. A 5′ probe located outside of the targeting vector detected the expected 24 and 11 kb SpeI fragments (S) from the wild-type and targeted Pitx2 alleles, respectively. (C) Genotyping wild-type(+/+) embryos, and embryos heterozygous and heterozygous for Pitx2neo (neo/+ and neo/neo) or Pitx2null (−/+ and −/−) by multiplex PCR. Pitx2 genomic primers (° in A) located 5′ of loxP1 and 3′ of loxP3 amplify 952 bp and 482 bp products from Pitx2+ and Pitx2null, respectively. The 5′ Pitx2 primer and an antisense neoR primer (in B) amplifies a 631 bp product. The Pitx2 genomic primer pair does not readily amplify across Pitx2neo.

Fig. 1.

Pitx2 allelic series. (A) The structure of the targeting vector is shown enlarged in relationship to Pitx2+. The complete structure of Pitx2neo and abbreviated structures for Pitx2null and Pitx2flox are also shown at the same scale as Pitx2+. Exon (boxes) and coding sequence (black boxes). (B) Southern analysis illustrating homologous recombination 5′ of the neoR cassette in two cell lines heterozygous for Pitx2neo. A 5′ probe located outside of the targeting vector detected the expected 24 and 11 kb SpeI fragments (S) from the wild-type and targeted Pitx2 alleles, respectively. (C) Genotyping wild-type(+/+) embryos, and embryos heterozygous and heterozygous for Pitx2neo (neo/+ and neo/neo) or Pitx2null (−/+ and −/−) by multiplex PCR. Pitx2 genomic primers (° in A) located 5′ of loxP1 and 3′ of loxP3 amplify 952 bp and 482 bp products from Pitx2+ and Pitx2null, respectively. The 5′ Pitx2 primer and an antisense neoR primer (in B) amplifies a 631 bp product. The Pitx2 genomic primer pair does not readily amplify across Pitx2neo.

The targeting vector was linearized with HindIII and electroporated into R1 ES cells (Nagy et al., 1993). G418-resistant colonies were screened by PCR to identify homologous recombinants. The 29 positive clones (3.3%) were rescreened by Southern blot to confirm homologous recombination 5′ and 3′ of the neoR cassette, and by PCR to demonstrate the incorporation of loxP3. Two cell lines heterozygous for Pitx2neo (6A.C10 and 8A.A9) were injected into C57BL/6J blastocysts to generate chimeric mice, which were mated to C57BL/6J females to transmit the neo allele.

The two neo/+ cell lines were also transiently transfected with a cre expression vector (pMC-Cre, (Gu et al., 1993)) to derive the null (−) and floxed alleles by cre-mediated site-specific recombination. The +/− and +/flox cell lines were identified by PCR (Fig 1). Two +/− cell lines were injected into C57BL/6J blastocysts; resulting chimeric founder males were bred to C57BL/6J females to transmit the null allele. Heterozygotes of the N1-N3 generations were mated to generate timed pregnancies. The morning after mating was designated e0.5.

Experiments with mice and recombinant DNA were approved by university committees.

RT-PCR

Total RNA was isolated from embryos or cell line pellets using TRIzol reagent (Gibco; Gaithersburg, MD) (Gage and Camper, 1997). PCR was performed on first-strand synthesis product using Pitx2-specific primers located in exons 1C and 4.

Histology, immunohistochemistry and in situ hybridization

Embryos were flash frozen or fixed overnight in 4% phosphate-buffered saline (pH 7.2). Fixed embryos were embedded in ParaPlast (Oxford Labware; St Louis, MO). Sections were stained in Hematoxylin (Fisher; Fair Lawn, NJ) and Eosin (Sigma; St. Louis, MO). In situ histochemistry was previously described (Schaeren-Weimers and Gerfin-Moser, 1993). Plasmid templates for Lhx3 (from H. Westphal) and Rpx (officially designated as Hesx1, from K. Mahon) were used as described (Hermesz et al., 1996; Zhadanov et al., 1995).

Construction of Pitx2 allelic series

Analysis of Pitx2 mRNA transcripts reveals the potential for three different protein isoforms of wild-type Pitx2 that arise through the use of alternative promoters and splicing (Fig. 1A) (Gage et al., 1999). Pitx2a and Pitx2b use an upstream promoter and differ from each other by alternative splicing of exon 3. Pitx2c utilizes a downstream promoter located in intron 3. Each Pitx2 isoform would contain the homeodomain, encoded primarily in exon 4, and have identical C termini but different N termini (Gage et al., 1999).

We used gene targeting in ES cells and cre-lox technology (Gu et al., 1994) to construct two Pitx2 alleles that differ in severity. Gene targeting was performed with a vector engineered to contain loxP sites flanking the critical exon 4 and a neomycin resistance selectable marker cassette (neoR) flanked by loxP sites and inserted into intron 4 (Fig. 1A). The Pitx2neo allele was generated by homologous recombination after electroporation of the targeting vector into ES cells. Transfection of these clones with a cre expression vector produced the Pitx2null and Pitx2flox alleles. Clones heterozygous for Pix2null (+/−) and Pitx2flox (flox/+) were identified by PCR (Fig. 1C). Deletion of both the neoRcassette and exon 4 via recombination at sites P1 and P3 produced a presumptive null allele due to loss of the essential homeodomain and a frameshift leading to a series of missense codons and a premature stop codon. Deletion of only the neoR cassette, by recombination between lox P1 and P2, produced Pitx2flox, an allele that should be fully functional and conditionally converted to Pitx2null in the presence of tissue-specific or temporally regulated cre transgenes.

Two neo/+ clones and two +/− clones were injected into C57BL/6J blastocysts to generate chimeric mice. Chimeric males were bred to C57BL/6J females to transmit the modified Pitx2 alleles, and independent mouse colonies derived from each injected cell line were established. Progeny were genotyped by PCR (Fig 1C). Heterozygotes for both alleles exhibited normal fertility. The phenotypes associated with each allele were replicated in two independent lines.

Pitx2neo is hypomorphic

Insertion of a neoR cassette into an intron by gene targeting can alter or disrupt gene expression by virtue of the cryptic splice sites and polyadenylation signals in the cassette, leading to hypomorphic or null alleles (Meyers et al., 1998). neo/+ mice were intercrossed to test for viable progeny. No neo/neo mice were identified in 334 two-week old progeny (Table 1), demonstrating that the neo allele is not functioning normally. Genetic analysis of timed pregnant litters from e9.5 to p1 revealed that individual homozygotes die throughout gestation beginning at e10.5. The loss of viable embryos is significant at e18.5 when only 2.4% (1/41) of the fetuses were neo homozygotes instead of the expected 25% (P<0.01). Death this late in gestation is usually attributable to structural cardiovascular defects or hematopoietic problems (Copp, 1995).

Table 1.

Transmission of Pitx2neo

Transmission of Pitx2neo
Transmission of Pitx2neo

Normal Pitx2 transcripts, produced by splicing from exon 1A to exon 4, were detected in mRNA from neo/neo embryos by RT-PCR (Fig. 2A). This observation and the more severe effects of the null allele (see below) demonstrate that the neo allele is a hypomorph, rather than a null (Table 2).

Table 2.

Pitx2 allelic series

Pitx2 allelic series
Pitx2 allelic series
Fig. 2.

RT-PCR detection of Pitx2 mRNA. (A) Detection of Pitx2c in total RNA from neo/neo, neo/+ and +/+ embryos at e12.5. Each reaction was carried out in the presence (+) and absence (−) of reverse transcriptase. (B) Detection of Pitx2c in total RNA representing all 5 anterior pituitary cell lineages. Cell lines: AtT20, corticotrope; GHFT-1, Pit1-dependent lineages; GH3, somatomammotrope; G7, AtT20 subclone; aT3, gonadotrope; aTSH, thyrotrope.

Fig. 2.

RT-PCR detection of Pitx2 mRNA. (A) Detection of Pitx2c in total RNA from neo/neo, neo/+ and +/+ embryos at e12.5. Each reaction was carried out in the presence (+) and absence (−) of reverse transcriptase. (B) Detection of Pitx2c in total RNA representing all 5 anterior pituitary cell lineages. Cell lines: AtT20, corticotrope; GHFT-1, Pit1-dependent lineages; GH3, somatomammotrope; G7, AtT20 subclone; aT3, gonadotrope; aTSH, thyrotrope.

Pitx2 is required for both anterior and posterior eye development

The RGS phenotype can be highly variable, even within families. However, all patients exhibit congenital defects of the anterior segment of the eye. Abnormalities consistent with RGS1 were evident. These included corectopia (asymmetrically placed pupils), anisocoria (uneven pupil size), multiple pupillary openings and clouded lenses (Fig. 3). Like RGS in humans, these features were not present in all heterozygotes for the neo or null (−) allele (∼10%). The majority of the +/− mice (n>30) had grossly normal teeth and body size, however two +/− males were approximately 1/3 normal body size, had mal-occluded incisors and obviously small eyes.

Fig. 3.

Eyes and teeth of Pitx2 heterozygotes. (A,B) Pitx2neo/+ eye pair showing corectopia (A) and anisocoria (B). (C) neo/+ eye with multiple iris openings (bracket). (D) Magnification of eye in C. (E) Mal-occluded teeth in −/+ mouse.

Fig. 3.

Eyes and teeth of Pitx2 heterozygotes. (A,B) Pitx2neo/+ eye pair showing corectopia (A) and anisocoria (B). (C) neo/+ eye with multiple iris openings (bracket). (D) Magnification of eye in C. (E) Mal-occluded teeth in −/+ mouse.

Examination of −/− embryos revealed a broad requirement for Pitx2 during eye development (Fig. 4A,B,D,G). Pitx2 is expressed in the perioptic mesenchyme by e9.5, but it is not expressed in the optic stalk or eminence. At e12.5, +/+ and neo/neo eyes are located immediately beneath the surface with the rim of the optic cup in contact with the developing cornea. In contrast, −/− eyes are smaller than normal and lie below the surface epithelium (Fig. 4C,E,H). They are rotated toward the nose so that the lens does not directly face the surface ectoderm. The developing corneal epithelium is thicker in −/− mice, and the optic cup is separated from the surface ectoderm by an abnormally thick mesenchymal layer (Fig. 4E,H). We confirmed the increased thickness of corneal epithelium by staining with a Pax6 antibody (data not shown). Vestigial lens pits are present in some −/− eyes at e12.5 (Fig. 4H), while lens pit closure is complete in +/+ eyes by e11.

Fig. 4.

Lack of Pitx2 effects eye development. (A-C) Embryo eyes at e12.5 with choroidal fissure oriented toward lower right (arrowhead). (D,E,G,H) Transverse sections at e12.5 illustrate the lens (L), neural retina (N), cornea (C), extrinsic ocular muscles (M), and optic nerve (*) in +/+ (D,G) and −/− eyes (E,H). Mutant eyes (E,H) are inappropriately positioned, corneal epithelia and mesenchyme are hypercellular, closure of the optic fissure and stalk are defective or severely delayed, extrinsic ocular muscles are absent and a vestigial lens pit is present. (F,I) Sagittal sections behind +/+ (F) and −/− (I) eyes. Note the unclosed optic stalk and absent ocular muscles in the mutant eyes.

Fig. 4.

Lack of Pitx2 effects eye development. (A-C) Embryo eyes at e12.5 with choroidal fissure oriented toward lower right (arrowhead). (D,E,G,H) Transverse sections at e12.5 illustrate the lens (L), neural retina (N), cornea (C), extrinsic ocular muscles (M), and optic nerve (*) in +/+ (D,G) and −/− eyes (E,H). Mutant eyes (E,H) are inappropriately positioned, corneal epithelia and mesenchyme are hypercellular, closure of the optic fissure and stalk are defective or severely delayed, extrinsic ocular muscles are absent and a vestigial lens pit is present. (F,I) Sagittal sections behind +/+ (F) and −/− (I) eyes. Note the unclosed optic stalk and absent ocular muscles in the mutant eyes.

Mesenchymal condensations leading to extrinsic eye muscle formation are clearly evident in wild-type eyes but absent in −/− embryos at e12.5 (Fig. 4D-I). Muscles form in neo/neo eyes but they are reduced in size (data not shown). Closure of the optic fissure is also defective or significantly delayed in the −/− embryos. No other gross defects were noted in the development of the neo/neo eyes or in the neural retina and retinal pigmented epithelium of −/− eyes.

Pitx2 regulates pituitary gland development

The anterior and intermediate lobes of the pituitary gland derive from Rathke’s pouch, an invagination of oral ectoderm within the roof of the mouth that begins at e8.5. Sequentially, the early steps are formation of the pouch rudiment, development of the committed pouch and pouch expansion (Sheng et al., 1996). Pitx2 is broadly expressed early in Rathke’s pouch, including in the rudiment (Gage and Camper, 1997; Muccielli et al., 1996). The Pitx2a and Pitx2b mRNA isoforms are expressed in the anterior pituitary cells of the thyrotrope, somatotrope, lactotrope and gonadotrope lineages, but not corticotropes (Gage and Camper, 1997). The Pitx2c isoform is expressed in all five anterior pituitary cell lineages, including corticotropes (Fig. 2B). Examination of −/− embryos revealed a committed pouch at e10.5, indicating that Pitx2 is not required for the initial stages of pituitary development (Fig. 5A,B). However, the mesenchymal layer surrounding the pouch was clearly hypercellular at this time and comparison of serial sagittal sections spanning the midline from +/+ and −/− littermates at e10.5 demonstrated that the pouch was smaller. Arrest of growth and differentiation is clearly evident in −/− pouches by e12.5. The wild-type pouch wall is thicker and cell proliferation on the ventral side of the pouch has formed the nascent anterior lobe. In contrast, the ventral and dorsal aspects of the −/− pouch wall remain relatively thin (Fig. 5D). This indicates that Pitx2 is required for pituitary gland development shortly after formation of the committed pouch. No changes were noted in the nascent posterior pituitary gland. Pituitary glands from neo/neo embryos were morphologically normal at e16.5 and e18.5.

Fig. 5.

Lack of Pitx2 affects pituitary gland development. (A,B) Midline sagittal comparison of +/+ and −/− Rathke’s pouch (*) at e10.5. Note the hypercellularity of the mesenchymal cells surrounding the mutant pouch. (C-F) Midline sagittal (C,D) and transverse (E,F) comparison of +/+ and −/− Rathke’s pouch at e12.5. In the mutant, dorsal and ventral walls are threadlike and there is little evidence of cell proliferation at the ventral side. Lack of lateral development is clear in transverse sections (F). (G-L) In situ hybridization of midline sagittal sections with probes for Hesx1 (G,H), Lhx3 (I,J), and immunohistochemical staining with Pomc antibodies (K,L) in normal (G,I,K) and −/− pituitaries (H,J,L).

Fig. 5.

Lack of Pitx2 affects pituitary gland development. (A,B) Midline sagittal comparison of +/+ and −/− Rathke’s pouch (*) at e10.5. Note the hypercellularity of the mesenchymal cells surrounding the mutant pouch. (C-F) Midline sagittal (C,D) and transverse (E,F) comparison of +/+ and −/− Rathke’s pouch at e12.5. In the mutant, dorsal and ventral walls are threadlike and there is little evidence of cell proliferation at the ventral side. Lack of lateral development is clear in transverse sections (F). (G-L) In situ hybridization of midline sagittal sections with probes for Hesx1 (G,H), Lhx3 (I,J), and immunohistochemical staining with Pomc antibodies (K,L) in normal (G,I,K) and −/− pituitaries (H,J,L).

Hesx1 (Rpx) and Lhx3 are homeobox genes that are required early during early pituitary development (Hermesz et al., 1996; Sheng et al., 1996). Pitx2 is expressed before Hesx1 and Lhx3 during pouch development, making them candidates for downstream targets of Pitx2. Hesx1 expression was detectable throughout the pouch in +/+ embryos at e12.5, but no transcripts were evident in −/− littermates (Fig. 5G,H). This indicated that Pitx2 is a regulator of Hesx1 expression during pituitary development. In contrast, similar levels of Lhx3 expression were detected in both +/+ and −/− pouches, demonstrating that Lhx3 expression is not dependent on Pitx2 (Fig. 5I,J). Essential inductive interactions occur between Rathke’s pouch and the base of the diencephalon early during pituitary development, including induction of pro-opiomelanocortin (Pomc) in the diencephalon (Diakoku et al., 1982; Sheng et al., 1996). We demonstrated that Pomc expression in the diencephalon of +/+ versus −/− embryos is indistinguishable, indicating that some inductive interactions are intact in −/− mice (Fig. 5K,L).

Null homozygotes have gross morphological defects

Neo/neo embryos are indistinguishable externally from wild type (Fig. 6A,B). The −/− embryos are moderately undersized relative to their heterozygous or wild-type littermates, although there were no signs of edema (Fig. 6C,D). All the mutant embryos bend severely rightward rostral to the developing hindlimb. In some embryos (∼5%), the crown of the head is open over the midbrain. Other brain defects were also varied but included excess mesenchyme and abnormalities of the expansion of the pontine and midbrain flexures. Rarely, embryos lacked the left forelimb (∼5%, Fig. 6J), mandibular arch, tongue and/or aspects of the caudal region.

Fig. 6.

Gross phenotypes of Pitx2neo and Pitx2null homozygotes. (A-D) Left lateral aspects of +/+ (A), neo/neo (B) and −/− (C,D) embryos at e12.5. Note reduced body size and externalization of ventral organs in −/− embryos. (E-H) Magnified left lateral flanks from embryos in A-D. Heart of Pitx2null/null embryos are external (ectopia cordis), as are the liver and other developing gut organs (gastroschisis). Note the distended, single atrium in both mutants (asterisk). Chest wall, and left forelimb and shoulder of +/+ and neo/neo embryos were dissected away for visualization. (I,J) Dorsal aspects of embryos in A and D. A-P axis of −/− mice bend severely right rostral to the hindlimbs. Note the extreme lateral placement of the heart and liver, and the absence of a left forelimb. (K,L,P) Low and high magnification ventral views. (L,P) Edge of ventral wall (arrowhead) was contiguous with the amnion prior to resection in this view of the right flank from the dorsal side. (M,N) Heart and liver of e13.5 −/− embryo with intact amnion. (O) Caudal view of e13.5 embryo.

Fig. 6.

Gross phenotypes of Pitx2neo and Pitx2null homozygotes. (A-D) Left lateral aspects of +/+ (A), neo/neo (B) and −/− (C,D) embryos at e12.5. Note reduced body size and externalization of ventral organs in −/− embryos. (E-H) Magnified left lateral flanks from embryos in A-D. Heart of Pitx2null/null embryos are external (ectopia cordis), as are the liver and other developing gut organs (gastroschisis). Note the distended, single atrium in both mutants (asterisk). Chest wall, and left forelimb and shoulder of +/+ and neo/neo embryos were dissected away for visualization. (I,J) Dorsal aspects of embryos in A and D. A-P axis of −/− mice bend severely right rostral to the hindlimbs. Note the extreme lateral placement of the heart and liver, and the absence of a left forelimb. (K,L,P) Low and high magnification ventral views. (L,P) Edge of ventral wall (arrowhead) was contiguous with the amnion prior to resection in this view of the right flank from the dorsal side. (M,N) Heart and liver of e13.5 −/− embryo with intact amnion. (O) Caudal view of e13.5 embryo.

Individuals with RGS1 have phenotypes consistent with variable defects in development of the ventral body wall. Failure of umbilical cord involution ranges in severity from a protruding navel to omphalocele (umbilical hernia) requiring surgical repair (Jorgenson et al., 1978). In wild-type mice, the thoracic and abdominal organs are normally internalized by e12.5 (Fig. 6A). Remarkably, the heart, liver and other abdominal organs are almost universally external and displaced profoundly leftward and outward in −/− embryos at e12.5 due to a large fissure in the ventral body wall (Fig. 6C,D). This fissure results from incomplete ventral closure and is generally displaced towards the left of the midline. The open ventral walls thicken as they extend outwardly and dorsally (Fig. 6L,P). They remain contiguous with the amnion, similar to their relationship during and shortly after turning of the embryo. The heart and liver are usually within thin membranes, presumably the remnants of the pericardial sac and liver capsule, respectively (Fig. 6K). Thus, the phenotype of −/− mice appears to be a more severe expression of the omphalocele characteristic of some RGS patients.

Hearts of −/− embryos are dramatically enlarged by e12.5 and continue to increase in size (Fig. 6 G,H). A septal ridge is clearly visible extending across the ventricle, indicating that septation has at least initiated. However, even as late as e12.5-13.5 only a single, hugely distended atrium can be identified. Contraction of this single atrium injects blood into both ventricles. Blood flows from the atrium into the ventricles between contractions and atrial contraction can inject blood backward into the vena cava.

Requirement of Pitx2 for asymmetry of lung

Several organs of the body exhibit left-right asymmetry, including the lungs, heart and stomach. The heart begins as a symmetrical, straight tube and initates asymmetry by looping to the right in an ‘S-shaped’ structure. The atria and inflow tracts at the caudal end of the tube move dorsally and anteriorly behind the ventricular regions. Right looping is initated properly in both neo/neo and −/− embryos and the asymmetry appears grossly normal at later stages (Fig. 7A-H). In rodents, the left lung is a single lobe but the right lung consists of four lobes. Both neo/neo and −/− embryos exhibit right-isomerization of the lungs (Fig. 7I-L). Both the right and left lungs were mirror images with multiple lobes normally characteristic of the right lung. The stomach was positioned on the left, as expected, in both neo/neo and −/− mutants (data not shown). Positioning of the tail and umbilicus over the right flank indicated that mutant embryos turn in the normal direction (data not shown). However, the orientation of the hindlimbs is sometimes in the opposite direction to the forelimbs, suggesting that the process of turning is not always complete.

Fig. 7.

Pitx2 deficiency causes right isomerization of the lungs.(A-D) Heart tubes loop rightward in neo/neo (B) and −/− (D) embryos at e9.5, similar to normal littermates (A, C). (E,F) Hearts dissected from neo/neo embryos at e16.5 (F) have asymmetric morphology similar to normal littermates (E). (G,H) Hearts from −/− embryos at e14.5 (H) are severely dysmorphic . (I-L) The lungs of neo/neo and −/− embryos are right-isomerized. Note the separation between the median and posteror lobes of the lungs (arrowhead). Lungs of normal e16.5 (I) and e14.5 (K) embryos and neo/neo (J) and −/− embryos (L) at the same time points.

Fig. 7.

Pitx2 deficiency causes right isomerization of the lungs.(A-D) Heart tubes loop rightward in neo/neo (B) and −/− (D) embryos at e9.5, similar to normal littermates (A, C). (E,F) Hearts dissected from neo/neo embryos at e16.5 (F) have asymmetric morphology similar to normal littermates (E). (G,H) Hearts from −/− embryos at e14.5 (H) are severely dysmorphic . (I-L) The lungs of neo/neo and −/− embryos are right-isomerized. Note the separation between the median and posteror lobes of the lungs (arrowhead). Lungs of normal e16.5 (I) and e14.5 (K) embryos and neo/neo (J) and −/− embryos (L) at the same time points.

Partial and complete loss of Pitx2 differentially affect heart development

The extremely abnormal heart morphology indicated that −/− embryos probably died of heart defects. A massive proliferation of mesoderm cells has displaced the mutant heart ventrally and leftward at e12.5 which, combined with the deficit in ventral body wall closure, results in its grossly external position (Fig. 8A,D). There is a single, distended atrium located to the left of the ventricles (Fig. 8E). Ventricular septation in −/− hearts is dysmorphic and there is little evidence of atrial septation. In contrast, +/+ hearts have distinct left and right atria developing on either side of the ventricles and ventricular septation is complete (Fig. 8B). Both the leaflets of the vena cava and atrial-ventricular (A-V) valve formation are defective in −/− embryos (Fig. 8E,F). In the case of the A-V valve, this appears to result from a reduction in the membranous component of the valve. Although the hearts of neo/neo animals were located within the thoracic cavity (Fig. 8I), they exhibited ventricular septation defects possibly resulting from a reduction in the membranous component (Fig. 8J). The structural defects of the heart are likely to account for the death of both the neo/neo and −/− embyros.

Fig. 8.

Heart defects in Pitx2 mutant embryos. (A-F) Hearts of e12.5 +/+ and −/− littermates. Displacement of −/− heart by mesenchymal proliferation is evident (A,D). Mutant A-V valve (*) formation is significantly reduced relative to +/+ (B,E). Leaflets (arrow) leading from the vena cava into the atrium are reduced in −/− embryos (F) relative to +/+ (C). (G-J) Hearts of +/+ and neo/neo littermates at e16.5. Mutant hearts are within the thoracic cavity (I). Septa are reversed and incomplete (arrow) in neo/neo hearts (J). A, atrium; V, ventricle; VC, vena cava. All panels are oriented with ventral at the top and left at the left.

Fig. 8.

Heart defects in Pitx2 mutant embryos. (A-F) Hearts of e12.5 +/+ and −/− littermates. Displacement of −/− heart by mesenchymal proliferation is evident (A,D). Mutant A-V valve (*) formation is significantly reduced relative to +/+ (B,E). Leaflets (arrow) leading from the vena cava into the atrium are reduced in −/− embryos (F) relative to +/+ (C). (G-J) Hearts of +/+ and neo/neo littermates at e16.5. Mutant hearts are within the thoracic cavity (I). Septa are reversed and incomplete (arrow) in neo/neo hearts (J). A, atrium; V, ventricle; VC, vena cava. All panels are oriented with ventral at the top and left at the left.

Malpositioning of the heart is evident in homozygotes for both alleles. The position of the hearts in neo/neo embryos was biased toward the right and the ventricular septum appeared reversed relative to wild-type hearts (compare Fig. 8I,J with 8G,H). The isomerization of the lungs results in a larger than normal left lung. This may contribute to the apparent shift in position of the heart, as heart looping initiated properly. While the heart was placed on the left in −/− embryos, its orientation was off by approximately 90°. The size of the left lung and the mass of mesoderm cells on the left could contribute to this rotation.

An allelic series ranging from partial to complete loss of function can be especially valuable in assessing gene function (Greco et al., 1996). The embryonic lethality frequently resulting from complete loss-of-function mutations in homeobox genes makes the availability of milder alleles highly desirable and relevant as models for human birth defects. We have generated an allelic series for the murine Pitx2 gene (Table 2). Two alleles differ in severity and the floxed allele has the potential for tissue-specific and temporally induced disruption of Pitx2 function. The constellation of phenotypes produced in heterozygotes indicate that these mice are a model for Rieger syndrome, type I. Pitx2 is essential for many aspects of normal development, with striking roles in eye, pituitary gland, heart and lung ontogeny, including developing functional heart valves and establishing the asymmetry of the lungs.

The congenital eye defects of RGS patients indicate an essential role for PITX2 in development of the anterior segment of the eye. In −/− embryos, the anterior segment is abnormally thick, due to an increase in both the epithelial and mesenchymal components of the cornea. It is striking that both layers are affected since Pitx2 is not expressed in the corneal epithelium (Semina et al., 1996). RGS patients exhibit corneal opacity. Therefore, the changes observed in −/− mice are consistent with the defects seen in RGS patients. The two posterior eye defects, lack of ocular muscles and optic nerve coloboma, were unexpected because RGS involves anterior segment abnormalities. However, the majority of RGS patients develop early onset glaucoma, a progressive loss of retinal ganglion cells and degeneration of the optic nerve head (Chang et al., 1999). The optic fissure defects in −/− embryos suggest that subtle optic nerve defects in heterozygotes might result in glaucoma as the animals age, providing another parallel with RGS patients.

Only a small fraction of adult +/neo or +/− mice exhibited anterior chamber defects of the eye, consistent with the remarkable variability in RGS and septo-optic dysplasia (Hesx1 MIM #182230) (Dattani et al., 1998). In contrast to the variability in heterozygotes, profound disturbances in eye development were observed consistently in the absence of Pitx2. The dosage sensitivity of Pitx2 may contribute to the variability of the phenotype. Inbred strains of mice and strains with sensitizing mutations in Pax6, Chx10 and Trp1 have been used to demonstrate the effects of genetic background on eye development and glaucoma (Burmeister et al., 1996; Chang et al., 1999; Glaser et al., 1999). Modifiers of PITX2 may be responsible for unlinked cases of RGS in addition to other

eye abnormalities and diseases such as cataracts and glaucoma. Thus, the Pitx2 mutants we have generated could be valuable for identifying other genes important for eye development and disease.

A genetic hierarchy of homeobox genes, including Hesx1, Lhx3, Prop1 and Pit1, is essential for pituitary gland development. (Dattani et al., 1998; Gage et al., 1996; Sheng et al., 1996; Sornson et al., 1996). Loss of Hesx1, Lhx3 and Prop1 results in dysmorphology of the pituitary primordium. Pit1 is required later during pituitary development for specification of three anterior pituitary cell types (Camper et al., 1990; Li et al., 1990). Molecular genetic analyses suggest that some of the genes act sequentially in pituitary development: Hesx1Prop1Pit1 (Anderson et al., 1995; Gage et al., 1996; Sornson et al., 1996). Our demonstration of altered Hesx1 expression in −/− mice places Pitx2 early in this pathway. Lhx3 and Pitx2 affect pouch development similarly and both genes are required for Hesx1 expression. Lhx3 expression does not require Pitx2. Thus, Pitx2 is an early regulator of pituitary ontogeny that acts with Hesx1 and Lhx3, prior to the action of Prop1 and Pit1 to advance development of the committed Rathke pouch.

The expression patterns of Pitx1 and Pitx2 overlap in the developing pituitary and they have nearly identical homeodomains, suggesting functional redundancy (Lamonerie et al., 1996; Lanctot et al., 1997; Muccielli et al., 1996; Szeto et al., 1996; Tremblay et al., 1998). Pitx1−/− fetuses have normal pituitary morphology and only modest quantitative changes in three of five pituitary cell types (Szeto et al., 1999). In contrast, development is arrested at the committed pouch stage in Pitx2−/− fetuses. The relative roles of Pitx1 and Pitx2 may be similar to the relative roles of Lhx4 and Lhx3. Loss of Lhx4 alone results in a relatively mild pituitary phenotype, similar to Pitx1 null mice (Sheng et al., 1997), but loss of Lhx3 has an early and dramatic effect on pouch development, like Pitx2. Loss of both Lhx3 and Lhx4 affected pouch development more profoundly, indicating a partial ability of these two genes to compensate for each other (Sheng et al., 1997). Analysis of Pitx1, Pitx2 double mutants will be necessary to reveal any functional overlap between these two genes.

Prior reports of asymmetric expression of Pitx2 in early mouse development, before the onset of visceral organogenesis, suggested it might have a role in development of left-right asymmetry (Logan et al., 1998; Piedra et al., 1998; Ryan et al., 1998; Yoshioka et al., 1998). Its expression is perturbed in mouse mutants with reversals of the heart and visceral organs (Meno et al., 1998; Piedra et al., 1998; Ryan et al., 1998). Manipulation of Pitx2 expression was sufficient to reprogram the left/right axis in chick and frog, providing further circumstantial evidence for an important role. In these experiments, Pitx2 was expressed ectopically on the right side and an early player in the pathway was blocked (sonic hedgehog, Shh), preventing activation of Pitx2 on the left. Isomerized or reversed development of the heart and gastrointestinal tract resulted. The defects in the ventral body wall that caused externalization of the viscera in −/− embryos revealed the importance of Pitx2 at the time the embryo turns. However, the right-ward looping of the heart initiated properly and the stomach was located on the left in both neo/neo and −/− fetuses, indicating that Pitx2 is not required to initiate the left-right asymmetry of the heart or the gastrointestinal tract. However, the lungs were right-isomerized, suggesting that Pitx2 is required for establishing the lobular pattern characteristic of the left lung. Thus, there is a dramatic difference between ectopic expression of Pitx2, and absent or low levels of Pitx2 in establishing the left-right asymmetry of the body axis. Ectopic expression of Pitx2 is sufficient to reprogram the left-right body axis, but it is only required for developing the asymmetry of the lungs. This suggests that there are other genes that partially compensate for the lack of Pitx2.

The defects that we observed in homozygous mutant mice clarify some questions about the Rieger syndrome phenotype. Retrospective studies have demonstrated that defects of the heart and umbilicus are more common than previously appreciated (Jorgenson et al., 1978; Mammi et al., 1998). However, the possibility existed that these features resulted from a contiguous gene syndrome. This is unlikely because reduced Pitx2 levels cause heart defects similar to those found in Rieger patients and failure of the ventral body wall to close that is consistent with omphalocele requiring surgical repair.

The concentration of Pitx2 is important for normal development. This is evident by comparing the mild eye phenotypes of adult +/− and +/neo mice with the severe eye defects in −/− embryos and by comparing the heart development of neo/neo and −/− fetuses. The sensitivity of each organ to Pitx2 levels is variable. The eye and heart appear to be more sensitive than the pituitary, for example. Pitx1 is expressed in the pituitary but not the eye or heart, thus functional redundancy may account for some of the differential sensitivity of each organ. In conjunction with tissue-specific, developmentally regulated, and/or inducible cre transgenes, the Pitx2flox allele will allow spatial and temporal requirements of Pitx2 function to be dissected.

We thank the following: S. Lindert, P. Gillespie, T. Saunders and the University of Michigan Transgenic Animal Core for assistance with generation and propogation of the mice; T. Glaser, N. Brown and J. Lauderdale for discussions of eye development and Pax6 antibodies, G. Dressler for Pax2, A. Seasholtz for pituitary cell line RNA, and D. M. Martin and S. O’Shea for helpful discussions. This work was supported by The Glaucoma Research Foundation (PJG) and NICHD (34283 and 30428, SAC).

Al-Shawi
,
R.
,
Burke
,
J.
,
Wallace
,
H.
,
Jones
,
C.
,
Harrison
,
S.
,
Buxton
,
D.
,
Maley
,
S.
,
Chandley
,
A.
and
Bishop
,
J. O.
(
1991
).
The herpes simplex virus type 1 thymidine kinase is expressed in the testes of transgenic mice under the control of a cryptic promoter
.
Mol. Cell Biol
.
11
,
4207
4216
.
Amendt
,
B.
,
Sutherland
,
L.
,
Semina
,
E.
and
Russo
,
A.
(
1998
).
The molecular basis of Rieger syndrome: Analysis of Pitx2 homeodomain protein activities
.
J. Biol. Chem
.
273
,
20066
20072
.
Anderson
,
B.
,
Pearse II
,
R. V.
,
Jenne
,
K.
,
Sornson
,
M.
,
Lin
,
S.-C.
,
Bartke
,
A.
and
Rosenfeld
,
M. G.
(
1995
).
The Ames dwarf gene is required for Pit-1 gene activation
.
Dev. Biol
.
172
,
495
503
.
Arakawa
,
H.
,
Nakamura
,
T.
,
Zhadanov
,
A. B.
,
Fidanza
,
V.
,
Yano
,
T.
,
Bullrich
,
F.
,
Shimizu
,
M.
,
Blechman
,
J.
,
Mazo
,
A.
,
Canaani
,
E.
et al.  (
1998
).
Identification and characterization of the ARP1 gene, a target for the human acute leukemia ALL1 gene
.
Proc. Natl Acad. Sci. USA
95
,
4573
4578
.
Burmeister
,
M.
,
Novak
,
J.
,
Liang
,
M.
,
Basu
,
S.
,
Ploder
,
L.
,
Hawes
,
N.
,
Vidgen
,
D.
,
Hoover
,
F.
,
Goldman
,
D.
,
Kalnins
,
V.
et al.  (
1996
).
Ocular retardation mouse caused Chx10 homeobox null allele: inpaired retinal progenitor proliferation and bipolar cell differentiation
.
Nat. Genet
.
12
,
376
384
.
Camper
,
S. A.
,
Saunders
,
T. L.
,
Katz
,
R. W.
and
Reeves
,
R. H.
(
1990
).
The Pit-1 transcription factor gene is a candidate for the Snell dwarf mutation
.
Genomics
8
,
586
590
.
Chang
,
B.
,
Smith
,
R.
,
Hawes
,
N.
,
Anderson
,
M.
,
Zabaleta
,
A.
,
Savinova
,
O.
,
Roderick
,
T.
,
Heckenlively
,
J.
,
Davisson
,
M.
and
John
,
S.
(
1999
).
Interacting loci cause severe iris atrophy and glaucoma in DBA/2J mice
.
Nat. Genet
.
21
,
405
409
.
Copp
,
A.
(
1995
).
Death before birth: clues from gene knockouts and mutations
.
Trends Genet
.
11
,
87
93
.
Dattani
,
M. T.
,
Martinez-Barbera
,
J.-P.
,
Thomas
,
P. Q.
,
Brickman
,
J. M.
,
Gupta
,
R.
,
Mårtensson
,
I.-L.
,
Toresson
,
H.
,
Fox
,
M.
,
Wales
,
J. K. H.
,
Hindmarsh
,
P. C.
et al.  (
1998
).
Mutations in the homeobox gene HESX1/Hesx1 associated with septo-optic dysplasia in human and mouse
.
Nat. Genet
.
19
,
125
133
.
Diakoku
,
S.
,
Chikamori
,
M.
,
Adachi
,
T.
and
Maki
,
Y.
(
1982
).
Effect of basal diencephalon on the development of Rathke’s pouch in rats: a study in combined organ cultures
.
Dev. Biol
.
90
,
198
202
.
Gage
,
P. J.
,
Brinkmeier
,
M. L.
,
Scarlett
,
L. M.
,
Knapp
,
L. T.
,
Camper
,
S. A.
and
Mahon
,
K. A.
(
1996
).
The Ames dwarf gene, df, is required early in pituitary ontogeny for the extinction of Rpx transcription and initiation of lineage specific cell proliferation
.
Mol. Endocrinol
.
10
,
1570
1581
.
Gage
,
P. J.
and
Camper
,
S. A.
(
1997
).
Pituitary homeobox 2, a novel member of the bicoid-related family of homeobox genes, is a potential regulator of anterior structure formation
.
Hum. Mol. Genet
.
6
,
457
464
.
Gage
,
P. J.
,
Suh
,
H.
and
Camper
,
S. A.
(
1999
).
Review: The bicoid-related Pitx gene family in development
.
Mammalian Genome
10
,
197
200
.
Glaser
,
T.
,
Tucker
,
P.
,
Munson
,
A.
,
Kanekar
,
S.
,
Vetter
,
M.
and
Laemle
,
L.
(
1999
).
Analysis of an eyeless strain of mice: Identification of a Rx/rax homeobox gene mutation
.
Invest. Ophthalmol. Vis. Sci
.
40
,
S205
.
Greco
,
T. L.
,
Takada
,
S.
,
Newhouse
,
M. M.
,
McMahon
,
J. A.
,
McMahon
,
A. P.
and
Camper
,
S. A.
(
1996
).
Analysis of the vestigial tail mutation demonstrates that Wnt-3a gene dosage regulates mouse axial development
.
Genes Dev
.
10
,
313
324
.
Gu
,
H.
,
Marth
,
J. D.
,
Orban
,
P. C.
,
Mossmann
,
H.
and
Rajewsky
,
K.
(
1994
).
Deletion of a DNA polymerase β gene segment in T cells using cell type-specific gene targeting
.
Science
265
,
103
106
.
Gu
,
H.
,
Zou
,
Y. R.
and
Rajevsky
,
K.
(
1993
).
Independent control of immunoglobulin switch recombination at individual switch regions evidenced through Cre-loxP-mediate gene targeting
.
Cell
73
,
1155
1164
.
Hermesz
,
E.
,
Mackem
,
S.
and
Mahon
,
K. A.
(
1996
).
Rpx: A novel anterior-restricted homeobox gene progressively activated in the prechordal plate, anterior neural plate, and Rathke’s pouch of the mouse embryo
.
Development
122
,
41
52
.
Jorgenson
,
R.
,
Levin
,
L.
,
Cross
,
H.
,
Yoder
,
F.
and
Kelly
,
T.
(
1978
).
The Rieger Syndrome
.
Am. J. Med. Genet
.
2
,
307
318
.
Kitamura
,
K.
,
Miura
,
H.
,
Yanazawa
,
M.
,
Miyashita
,
T.
and
Kato
,
K.
(
1997
).
Expression patterns of Brx1 (Rieg gene), Sonic hedgehog, Nkx2.2, Dlx1 and Arx during zona limitans intrathalamica and embryonic ventral lateral geniculate nuclear formation
.
Mech. Dev
.
67
,
83
96
.
Lamonerie
,
T.
,
Tremblay
,
J. J.
,
Lanctot
,
C.
,
Therrien
,
M.
,
Gauthier
,
Y.
and
Drouin
,
J.
(
1996
).
Ptx1, a bicoid-related homeo box transcription factor involved in transcription of the pro-opiomelanocortin gene
.
Genes Dev
.
10
,
1284
1295
.
Lanctot
,
C.
,
Lamolet
,
B.
and
Drouin
,
J.
(
1997
).
The bicoid-related homeoprotein Ptx1 defines the most anterior domain of the embryo and differentiates posterior from anterior lateral mesoderm
.
Development
124
,
2807
2817
.
Lanctot
,
C.
,
Moreau
,
A.
,
Chamberland
,
M.
,
Tremblay
,
M. L.
and
Drouin
,
J.
(
1999
).
Hindlimb patterning and mandible development require the Ptx1 gene
.
Development
126
,
1805
1810
.
Li
,
S.
, Crenshaw 3
rd
,
E. B.
,
Rawson
,
E. J.
,
Simmons
,
D. M.
,
Swanson
,
L. W.
and
Rosenfeld
,
M. G.
(
1990
).
Dwarf locus mutants lacking three pituitary cell types result from mutations in the POU-domain gene Pit-1
.
Nature
347
,
528
533
.
Logan
,
M.
,
Pagan-Westphal
,
S. M.
,
Smith
,
D. M.
,
Paganessi
,
L.
and
Tabin
,
C. J.
(
1998
).
The transcription factor Pitx2 mediates situs-specific morphogenesis in response to left-right asymmetric signals
.
Cell
94
,
307
317
.
Mammi
,
I.
,
De Giorgio
,
P.
,
Clementi
,
M.
and
Renconi
,
R.
(
1998
).
Cardiovascular anomaly in Rieger Syndrome: Heterogeneity or contiguity?
Acta Ophthalmologica Scandinavica
76
,
509
512
.
Meno
,
C.
,
Shimono
,
A.
,
Saijoh
,
Y.
,
Yashiro
,
K.
,
Mochida
,
K.
,
Ohishi
,
S.
,
Noji
,
S.
,
Kondoh
,
H.
and
Hamada
,
H.
(
1998
).
lefty-1 is required for left-right determination as a regulator of lefty-2 and nodal
.
Cell
94
,
287
297
.
Meyers
,
E.
,
Lewandoski
,
M.
and
Martin
,
G.
(
1998
).
An Fgf8 mutant allelic series generated by Cre-and FLP-mediated recombination
.
Nat. Genet
.
18
,
136
141
.
Muccielli
,
M. L.
,
Martinez
,
S.
,
Pattyn
,
A.
,
Goridis
,
C.
and
Brunet
,
J. F.
(
1996
).
Otlx2, an Otx-related homeobox gene expressed in the pituitary gland and in a restricted pattern in the forebrain
.
Molec. Cell. Neurosci
.
8
,
258
271
.
Nagy
,
A.
,
Rossant
,
J.
,
Nagy
,
R.
,
Abramow-Newerly
,
W.
and
Roder
,
J. C.
(
1993
).
Derivation of completely cell culture-derived mice from early-passage embryonic stem cells
.
Proc. Natl. Acad. Sci. USA
90
,
8424
8428
.
Orban
,
P. C.
,
Chui
,
D.
and
Marth
,
J. D.
(
1992
).
Tissue-and site-specific DNA recombination in transgenic mice
.
Proc. Natl. Acad. Sci. USA
89
,
6861
6865
.
Piedra
,
M. E.
,
Icardo
,
J. M.
,
Albajar
,
M.
,
Rodriquez-Rey
,
J. C.
and
Ros
,
M. A.
(
1998
).
Pitx2 participates in the late phase of the pathway conrolling left-right asymmetry
.
Cell
94
,
319
324
.
Ryan
,
A. K.
,
tBlumberg
,
B.
,
Rodriguez-Estaban
,
C.
,
Yonei-Tamura
,
S.
,
Tamura
,
K.
,
Tsukui
,
T.
, de al
Peña
,
J.
,
Sabbagh
,
W.
,
Greenwald
,
J.
,
Choe
,
S.
et al.  (
1998
).
Pitx2 determines left-right asymmetry of internal organs in vertebrates
.
Nature
394
, 1998.
Sadeghi-Nedjad
,
A.
and
Senior
,
B.
(
1974
).
Autosomal dominant transmission of isolated growth hormone deficiency in iris-dental dysplasia (Rieger’s Syndrome)
.
J. Pediatr
.
85
,
644
648
.
Schaeren-Weimers
,
N.
and
Gerfin-Moser
,
A.
(
1993
).
A single protocol to detect transcripts of various types and expression levels in neural and cultured cells: in situ hybridization using digoxigenin-labeled cRNA probes
.
Histochemistry
100
,
431
440
.
Semina
,
E. V.
,
Ferrell
,
R. E.
,
Mintz-Hittner
,
H. A.
,
Bitoun
,
P.
,
Alward
,
W. L.
,
Reiter
,
R. S.
,
Funkhauser
,
C.
,
Daack-Hirsch
,
S.
and
Murray
,
J. C.
(
1998
).
A novel homeobox gene PITX3 is mutated in families with autosomal-dominant cataracts and ASMD
.
Nat. Genet
.
19
,
167
170
.
Semina
,
E. V.
,
Reiter
,
R.
,
Leysens
,
N. J.
,
Alward
,
W. L.
,
Small
,
K. W.
,
Datson
,
N. A.
,
Siegel-Bartelt
,
J.
,
Bierke-Nelson
,
D.
,
Bitoun
,
P.
,
Zabel
,
B. U.
et al.  (
1996
).
Cloning and characterization of a novel bicoid-related homeobox transcription factor gene, RIEG, involved in Rieger syndrome
.
Nat. Genet
.
14
,
392
399
.
Sheng
,
H.
,
Moriyama
,
K.
,
Yamashita
,
T.
,
Li
,
H.
,
Potter
,
S.
,
Mahon
,
K.
and
Westphal
,
H.
(
1997
).
Multistep control of pituitary organogenesis
.
Science
278
,
1809
1812
.
Sheng
,
H. Z.
,
Zhadanov
,
A. B.
,
Mosinger
,
B. J.
,
Fujii
,
T.
,
Bertuzzi
,
S.
,
Grinberg
,
A.
,
Lee
,
E. J.
,
Huang
,
S. P.
,
Mahon
,
K. A.
and
Westphal
,
H.
(
1996
).
Specification of pituitary cell lineages by the LIM homeobox gene Lhx3
.
Science
272
,
1004
1007
.
Sornson
,
M. W.
,
Wu
,
W.
,
Dasen
,
J. S.
,
Flynn
,
S. E.
,
Norman
,
D. J.
,
O’Connell
,
S. M.
,
Gukovsky
,
I.
,
Carrière
,
C.
,
Ryan
,
A. K.
,
Miller
,
A. P.
et al.  (
1996
).
Pituitary lineage determination by the Prophet of Pit-1 homeodomain factor defective in Ames dwarfism
.
Nature
384
,
327
333
.
Sternberg
,
N. L.
(
1992
).
Cloning high molecular weight DNA fragments by the bacteriophage P1 system
.
Trends Genet
.
8
,
11
16
.
Szeto
,
D.
,
Ryan
,
A.
,
O’Connell
,
S.
and
Rosenfeld
,
M.
(
1996
).
P-OTX: A PIT-1-interacting homeodomain factor expressed during anterior pituitary gland development
.
Proc. Natl. Acad. Sci. USA
93
,
7706
7710
.
Szeto
,
D. P.
,
Rodriquez-Estaban
,
C.
,
Ryan
,
A. K.
,
O’Connell
,
S. M.
,
Liu
,
F.
, C., K.,
Gleiberman
,
A. S.
,
Izpisua-Belmonte
,
J. C.
and
Rosenfeld
,
M. G.
(
1999
).
Role of Bicoid-related homeodomain factor Pitx1 in specifying hindlimb morphogenesis and pituitary development
.
Genes Dev
.
13
,
484
494
.
Tremblay
,
J. J.
,
Lanctôt
,
C.
and
Drouin
,
J.
(
1998
).
The pan-Pituitary Activator of Transcription, Ptx1 (Pituitary Homeobox 1), Acts in Synergy with SF-1 and Pit1 and Is an Upstream Regulator of the Lim-Homeodomain gene Lim3/Lhx3
.
Mol. Endocrinol
.
12
,
428
441
.
Vorbruggen
,
G.
,
Constien
,
R.
,
Zilian
,
O.
,
Wimmer
,
E. A.
,
Dowe
,
G.
,
Taubert
,
H.
,
Noll
,
M.
and
Jackle
,
H.
(
1997
).
Embryonic expression and characterization of a Ptx1 homolog in Drosophila
.
Mech. Dev
.
68
,
139
147
.
Yoshioka
,
H.
,
Meno
,
C.
,
Koshiba
,
K.
,
Sugihara
,
M.
,
Itoh
,
H.
,
Ishimaru
,
Y.
,
Inoue
,
T.
,
Ohuchi
,
H.
,
Semina
,
E. V.
,
Murray
,
J. C.
et al.  (
1998
).
Pitx2, a bicoid-type homeobox gene, is involved in a lefty signaling pathway in determination of left-right asymmetry
.
Cell
94
,
299
305
.
Zhadanov
,
A.
,
Bertuzzi
,
S.
,
Taira
,
M.
,
Dawid
,
I.
and
Westphal
,
H.
(
1995
).
Expression pattern of the murine LIM class homeobox gene Lhx3 in subsets of neural and neuroendocrine tissues
.
Dev. Dyn
.
202
,
354
364
.