Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 50(10): 4531-41, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19407022

ABSTRACT

PURPOSE: To determine the temporal and spatial expression of Pitx2, a bicoid-like homeobox transcription factor, during postnatal development of mouse extraocular muscle and to evaluate its role in the growth and phenotypic maintenance of postnatal extraocular muscle. METHODS: Mouse extraocular muscles of different ages were examined for the expression of Pitx2 by RT-PCR, q-PCR, and immunostaining. A conditional mutant mouse strain, in which Pitx2 function is inactivated at postnatal day (P)0, was generated with a Cre-loxP strategy. Histology, immunostaining, real-time PCR, in vitro muscle contractility, and in vivo ocular motility were used to study the effect of Pitx2 depletion on extraocular muscle. RESULTS: All three Pitx2 isoforms were expressed by extraocular muscle and at higher levels than in other striated muscles. Immunostaining demonstrated the presence of Pitx2 mainly in extraocular muscle myonuclei. However, no obvious expression patterns were observed in terms of anatomic region (orbital versus global layer), innervation zone, or muscle fiber types. The mutant extraocular muscle had no obvious pathology but had altered muscle fiber sizes. Expression levels of myosin isoforms Myh1, Myh6, Myh7, and Myh13 were reduced, whereas Myh2, Myh3, Myh4, and Myh8 were not affected by postnatal loss of Pitx2. In vitro, Pitx2 loss made the extraocular muscles stronger, faster, and more fatigable. Eye movement recordings found saccades to have a lower peak velocity. CONCLUSIONS: Pitx2 is important in maintaining the mature extraocular muscle phenotype and regulating the expression of critical contractile proteins. Modulation of Pitx2 expression can influence extraocular muscle function with long-term therapeutic implications.


Subject(s)
Gene Expression Regulation/physiology , Homeodomain Proteins/genetics , Oculomotor Muscles/metabolism , Transcription Factors/genetics , Animals , Eye Movements/physiology , Female , Fluorescent Antibody Technique, Indirect , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Myosins/genetics , Oculomotor Muscles/growth & development , Phenotype , Protein Isoforms/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Homeobox Protein PITX2
2.
Hum Mol Genet ; 17(23): 3643-54, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18723525

ABSTRACT

Axenfeld-Rieger syndrome (ARS) patients with PITX2 point mutations exhibit a wide range of clinical features including mild craniofacial dysmorphism and dental anomalies. Identifying new PITX2 targets and transcriptional mechanisms are important to understand the molecular basis of these anomalies. Chromatin immunoprecipitation assays demonstrate PITX2 binding to the FoxJ1 promoter and PITX2C transgenic mouse fibroblasts and PITX2-transfected cells have increased endogenous FoxJ1 expression. FoxJ1 is expressed at embryonic day 14.5 (E14.5) in early tooth germs, then down-regulated from E15.5-E17.5 and re-expressed in the inner enamel epithelium, oral epithelium, tongue epithelium, sub-mandibular salivary gland and hair follicles during E18.5 and neonate day 1. FoxJ1 and Pitx2 exhibit overlapping expression patterns in the dental and oral epithelium. PITX2 activates the FoxJ1 promoter and, Lef-1 and beta-catenin interact with PITX2 to synergistically regulate the FoxJ1 promoter. FoxJ1 physically interacts with the PITX2 homeodomain to synergistically regulate FoxJ1, providing a positive feedback mechanism for FoxJ1 expression. Furthermore, FoxJ1, PITX2, Lef-1 and beta-catenin act in concert to activate the FoxJ1 promoter. The PITX2 T68P ARS mutant protein physically interacts with FoxJ1; however, it cannot activate the FoxJ1 promoter. These data indicate a mechanism for the activity of the ARS mutant proteins in specific cell types and provides a basis for craniofacial/ tooth anomalies observed in these patients. These data reveal novel transcriptional mechanisms of FoxJ1 and demonstrate a new role of FoxJ1 in oro-facial morphogenesis.


Subject(s)
Craniofacial Abnormalities/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Maxillofacial Development , Morphogenesis , Transcription, Genetic , Animals , CHO Cells , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/metabolism , Cricetinae , Cricetulus , Face/abnormalities , Face/embryology , Female , Forkhead Transcription Factors/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Homeobox Protein PITX2
3.
BMC Mol Biol ; 9: 31, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18373856

ABSTRACT

BACKGROUND: Members of the Paired-like homeodomain transcription factor (PITX) gene family, particularly PITX1 and PITX2, play important roles in normal development and in differentiated cell functions. Three major isoforms of PITX2 were previously reported to be produced through both alternative mRNA splicing (PITX2A and PITX2B) and alternative promoter usage (PITX2C). The proteins derived from these mRNAs contain identical homeodomain and carboxyl termini. Differences in the amino-termini of the proteins may confer functional differences in some contexts. RESULTS: Here, we report the identification of two novel PITX2 isoforms. First, we demonstrate that the Pitx2c mRNA generates two protein products, PITX2Calpha and PITX2Cbeta, via alternative translation initiation. Second, we identified a novel mRNA splice variant, Pitx2b2, which uses the same 5' splice donor in intron 2 as Pitx2b (hereafter referred to as Pitx2b1), but employs an alternative 3' splice acceptor, leading to an in-frame deletion of 39 base pairs relative to Pitx2b1. Pitx2b2 mRNA is expressed in both murine and human pituitary. The data show that in a murine gonadotrope cell line and adult murine pituitary what was previously thought to be PITX2B1 is actually PITX2Cbeta, or perhaps PITX2B2. PITX2B1 is expressed at lower levels than previously thought. PITX2Cbeta and PITX2B2 activate gonadotrope-specific gene promoter-reporters similarly to known PITX2 isoforms. CONCLUSION: We have identified and characterized two novel isoforms of PITX2, generated by alternative translation initiation (PITX2Cbeta) and alternative mRNA splicing (PITX2B2). These proteins show similar DNA binding and trans-activation functions as other PITX2 isoforms in vitro, though their conservation across species suggests that they may play distinct, as yet unidentified, roles in vivo.


Subject(s)
Alternative Splicing , Homeodomain Proteins/genetics , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/physiology , Humans , Male , Mice , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Sequence Alignment , Transcription Factors/biosynthesis , Transcription Factors/physiology , Homeobox Protein PITX2
4.
Mol Endocrinol ; 22(7): 1633-46, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417734

ABSTRACT

Ad4BP/SF-1 [adrenal4 binding protein/steroidogenic factor-1 (NR5A1)] is a factor important for animal reproduction and endocrine regulation, and its expression is tightly regulated in the gonad, adrenal gland, ventromedial hypothalamic nucleus, and pituitary gonadotrope. Despite its functional significance in the pituitary, the mechanisms underlying pituitary-specific expression of the gene remain to be uncovered. In this study, we demonstrate by transgenic mouse assays that the pituitary gonadotrope-specific enhancer is localized within the sixth intron of the gene. Functionally, the enhancer recapitulates endogenous Ad4BP/SF-1 expression in the fetal Rathke's pouch to the adult pituitary gonadotrope. Structurally, the enhancer consists of several elements conserved among animal species. Mutational analyses confirmed the significance of these elements for the enhancer function. One of these elements was able to interact both in vitro and in vivo with Pitx2 (pituitary homeobox 2), demonstrating that pituitary homeobox 2 regulates Ad4BP/SF-1 gene transcription in the pituitary gonadotrope via interaction with the gonadotrope-specific enhancer.


Subject(s)
Gene Expression Regulation , Gonadotropins/metabolism , Homeodomain Proteins/metabolism , Pituitary Gland/metabolism , Steroidogenic Factor 1/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , Conserved Sequence , Enhancer Elements, Genetic , Introns , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Homeobox Protein PITX2
5.
BMC Dev Biol ; 8: 25, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18312615

ABSTRACT

BACKGROUND: Limb development and patterning originate from a complex interplay between the skeletal elements, tendons, and muscles of the limb. One of the genes involved in patterning of limb muscles is the homeobox transcription factor Pitx2 but its role in forelimb development is uncharacterized. Pitx2 is expressed in the majority of premature presumptive forelimb musculature at embryonic day 12.5 and then maintained throughout embryogenesis to adult skeletal muscle. RESULTS: To further study the role of Pitx2 in forelimb development we have generated transgenic mice that exhibit a pulse of PITX2 over-expression at embryonic day 13.5 and 14.5 in the developing forelimb mesenchyme. These mice exhibit a distal misplacement of the biceps brachii insertion during embryogenesis, which twists the forelimb musculature resulting in severe skeletal malformations. The skeletal malformations have some similarities to the forearm deformities present in Leri-Weill dyschondrosteosis. CONCLUSION: Taken together, the tendon, muscle, and bone anomalies further support a role of Pitx2 in forelimb development and may also shed light on the interaction between the skeletal elements and muscles of the limb during embryogenesis.


Subject(s)
Forelimb/abnormalities , Forelimb/embryology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Animals, Newborn , Apoptosis , Cyclin D2 , Cyclins/metabolism , Embryo, Mammalian/abnormalities , Forelimb/pathology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Humans , Humerus/abnormalities , Mice , Muscle Cells/metabolism , Proteoglycans/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radius/abnormalities , Tendons/abnormalities , Transcription Factors/metabolism , Ulna/abnormalities , Homeobox Protein PITX2
6.
Expert Rev Mol Med ; 7(25): 1-17, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16274491

ABSTRACT

Axenfeld-Rieger syndrome (ARS) is a rare autosomal dominant inherited disorder affecting the development of the eyes, teeth and abdomen. The syndrome is characterised by complete penetrance but variable expressivity. The ocular component of the ARS phenotype has acquired most clinical attention and has been dissected into a spectrum of developmental eye disorders, of which open-angle glaucoma represents the main challenge in terms of treatment. Mutations in several chromosomal loci have been implicated in ARS, including PITX2, FOXC1 and PAX6. Full-spectrum ARS is caused primarily by mutations in the PITX2 gene. The homeobox transcription factor PITX2 is produced as at least four different transcriptional and splicing isoforms, with different biological properties. Intriguingly, PITX2 is also involved in left-right polarity determination, although asymmetry defects are not a feature of ARS. In experimental animal models and in cell culture experiments using PITX2, abundant evidence indicates that a narrow window of expression level of this gene is vital for its correct function.


Subject(s)
Abdomen/abnormalities , Eye Abnormalities/genetics , Tooth Abnormalities/genetics , Animals , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Syndrome , Tooth Abnormalities/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Homeobox Protein PITX2
7.
Mol Endocrinol ; 19(7): 1893-903, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15761027

ABSTRACT

The PITX family of transcription factors regulate the development of many organs. Pitx1 mutants have a mild pituitary phenotype, but Pitx2 is necessary for the development of Rathke's pouch, expression of essential transcription factors in gonadotropes, and expansion of the Pit1 lineage. We report that lack of Pitx2 causes the pouch to undergo excessive cell death, resulting in severe pituitary hypoplasia. Transgenic overexpression of PITX2 in the pituitary can increase the gonadotrope population, suggesting that the absolute concentration of PITX2 is important for normal pituitary cell lineage expansion. We show that PITX1 and PITX2 proteins are present in similar expression patterns throughout pituitary development and in the mature pituitary. Both transcription factors are preferentially expressed in adult gonadotropes and thyrotropes, suggesting the possibility of overlap in maintenance of adult pituitary functions within these cell types. Double knockouts of Pitx1 and Pitx2 exhibit severe pituitary hypoplasia and fail to express the transcription factor LHX3. This indicates that these PITX genes are upstream of Lhx3 and have compensatory roles during development. Thus, the combined dosage of these PITX family members is vital for pituitary development, and their persistent coexpression in the adult pituitary suggests a continued role in maintenance of pituitary function.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Pituitary Gland/abnormalities , Transcription Factors/genetics , Animals , Cell Death/genetics , Cell Survival/genetics , Homeodomain Proteins/analysis , LIM-Homeodomain Proteins , Mice , Mice, Knockout , Mutation , Paired Box Transcription Factors , Pituitary Gland/cytology , Pituitary Gland/immunology , Transcription Factors/analysis , Transcription Factors/metabolism , Transcriptional Activation , Transgenes , Homeobox Protein PITX2
8.
J Biol ; 4(3): 11, 2005.
Article in English | MEDLINE | ID: mdl-16403239

ABSTRACT

BACKGROUND: Development of the eye depends partly on the periocular mesenchyme derived from the neural crest (NC), but the fate of NC cells in mammalian eye development and the signals coordinating the formation of ocular structures are poorly understood. RESULTS: Here we reveal distinct NC contributions to both anterior and posterior mesenchymal eye structures and show that TGFbeta signaling in these cells is crucial for normal eye development. In the anterior eye, TGFbeta2 released from the lens is required for the expression of transcription factors Pitx2 and Foxc1 in the NC-derived cornea and in the chamber-angle structures of the eye that control intraocular pressure. TGFbeta enhances Foxc1 and induces Pitx2 expression in cell cultures. As in patients carrying mutations in PITX2 and FOXC1, TGFbeta signal inactivation in NC cells leads to ocular defects characteristic of the human disorder Axenfeld-Rieger's anomaly. In the posterior eye, NC cell-specific inactivation of TGFbeta signaling results in a condition reminiscent of the human disorder persistent hyperplastic primary vitreous. As a secondary effect, retinal patterning is also disturbed in mutant mice. CONCLUSION: In the developing eye the lens acts as a TGFbeta signaling center that controls the development of eye structures derived from the NC. Defective TGFbeta signal transduction interferes with NC-cell differentiation and survival anterior to the lens and with normal tissue morphogenesis and patterning posterior to the lens. The similarity to developmental eye disorders in humans suggests that defective TGFbeta signal modulation in ocular NC derivatives contributes to the pathophysiology of these diseases.


Subject(s)
Eye Abnormalities/etiology , Neural Crest/physiology , Signal Transduction , Stem Cells/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Movement , Cells, Cultured , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lens, Crystalline/metabolism , Mice , Mice, Mutant Strains , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Rats , Stem Cells/cytology , Transcription Factors , Homeobox Protein PITX2
9.
Am J Pathol ; 165(5): 1633-41, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509533

ABSTRACT

The human autosomal-dominant disorder Axenfeld-Rieger syndrome presents with defects in development of the eyes, teeth, and umbilicus. The eye manifests with iris ruptures, irido-corneal adhesions, cloudy corneas, and glaucoma. Transcription factors such as PITX2 and FOXC1 have been found to carry point mutations, causing the disorder. However, for approximately 40% of the cases, the pathogenesis is unknown. It has been reported that some mutations in PITX2 increase transactivation, whereas most mutations cause defects in DNA binding or transactivation. It is not known whether up-regulation of PITX2 activity can cause the disorder as well. Here we test this hypothesis directly by overexpressing PITX2A as a transgene in mouse corneal mesenchyme and iris, using keratocan-flanking sequences. The mice presented with corneal opacification, corneal hypertrophy, irido-corneal adhesions, and severely degenerated retina, resembling glaucoma. The corneal hypertrophy also resembles the corneal hypertrophy of Pitx2-/- mice. Control transgenic mice carrying point mutations T68P or K88E in PITX2A were normal. These findings indicate a novel pathogenetic mechanism in which excess corneal and iridal PITX2A cause glaucoma and anterior defects that closely resemble Axenfeld-Rieger syndrome.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Alleles , Animals , Blotting, Southern , Chromosome Mapping , Collagen/chemistry , Cornea/pathology , DNA/metabolism , Disease Models, Animal , Eye Abnormalities/genetics , Eye Diseases/pathology , Genotype , Glaucoma/pathology , Hypertrophy , Immunohistochemistry , In Situ Nick-End Labeling , Iris/pathology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Point Mutation , Retina/metabolism , Retina/pathology , Reverse Transcriptase Polymerase Chain Reaction , Syndrome , Transcriptional Activation , Transgenes , Up-Regulation , Homeobox Protein PITX2
10.
J Biol Chem ; 278(25): 22437-45, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12692125

ABSTRACT

PITX2 and Nkx2.5 are two of the earliest known transcriptional markers of vertebrate heart development. Pitx2-/- mice present with severe cardiac malformations and embryonic lethality, demonstrating a role for PITX2 in heart development. However, little is known about the downstream targets of PITX2 in cardiogenesis. We report here that the atrial natriuretic factor (ANF) promoter is a target of PITX2. PITX2A, PITX2B, and PITX2C isoforms differentially activate the ANF promoter. However, only PITX2C can synergistically activate the ANF promoter in the presence of Nkx2.5. We further demonstrate that the procollagen lysyl hydroxylase (PLOD1) promoter is regulated by Nkx2.5. Mechanistically, PITX2C and Nkx2.5 synergistically regulate ANF and PLOD1 expression through binding to their respective DNA elements. Surprisingly, PITX2A activation of the ANF and PLOD1 promoters is repressed by co-transfection of Nkx2.5 in the C3H10T1/2 embryonic fibroblast cell line. Pitx2a and Pitx2c are endogenously expressed in C3H10T1/2 cells, and these cells express factors that differentially regulate PITX2 isoform activities. We provide a new mechanism for the regulation of heart development by PITX2 isoforms through the regulation of ANF and PLOD1 gene expression and Nkx2.5 transcriptional activity.


Subject(s)
Atrial Natriuretic Factor/genetics , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Xenopus Proteins , Animals , Base Sequence , CHO Cells , Cell Line , Cricetinae , Drug Synergism , Genes, Reporter , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transfection , Homeobox Protein PITX2
11.
J Biol Chem ; 277(28): 25001-10, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-11948188

ABSTRACT

Three major PITX2 isoforms are differentially expressed in human, mice, zebrafish, chick, and frog tissues. To demonstrate differential regulation of gene expression by these isoforms we used three different promoters and three cell lines. Transient transfection of Chinese hamster ovary, HeLa, and LS-8 cell lines revealed differences in PITX2A and PITX2C activation of the PLOD1 and Dlx2 promoters, however, PITX2B is inactive. In contrast, PITX2B actives the pituitary-specific Prolactin promoter at higher levels than either PITX2A or PITX2C. Interestingly, co-transfection of either PITX2A or PITX2C with PITX2B results in a synergistic activation of the PLOD1 and Dlx2 promoters. Furthermore, PITX2 isoforms have different transcriptional activity dependent upon the cells used for transfection analysis. We have isolated a fourth PITX2 isoform (PITX2D) expressed only in humans, which acts to suppress the transcriptional activity of the other PITX2 isoforms. Electrophoretic mobility shift assays and glutathione S-transferase pull-down experiments demonstrated that all isoforms interact with PITX2D and that PITX2B forms heterodimeric complexes with PITX2A and PITX2C. Our research provides a molecular basis for differential gene regulation through the expression of PITX2 isoforms. PITX2 isoform activities are both promoter- and cell-specific, and our data reveal new mechanisms for PITX2-regulated gene expression.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Nuclear Proteins , Protein Isoforms/physiology , Transcription Factors/physiology , Animals , Base Sequence , Cell Line , Cytoskeletal Proteins , DNA , DNA-Binding Proteins/genetics , Dimerization , Humans , Prolactin/genetics , Promoter Regions, Genetic , RNA-Binding Proteins , Transcription, Genetic/physiology , Transfection , Homeobox Protein PITX2
SELECTION OF CITATIONS
SEARCH DETAIL
...