Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Dev Dyn ; 251(11): 1798-1815, 2022 11.
Article in English | MEDLINE | ID: mdl-35710880

ABSTRACT

BACKGROUND: The vitamin A derivative all-trans retinoic acid (RA) regulates early stages of inner ear development. As the early disruption of the RA pathway results in profound mispatterning of the developing inner ear, this confounds analyses of specific roles in later stages. Therefore, we used the temporal-specific exposure of all-trans RA or diethylaminobenzaldehyde to evaluate RA functions in late otic development. RESULTS: Perturbing late RA signaling causes behavioral defects analogous to those expected in larvae suffering from vestibular dysfunction. These larvae also demonstrate malformations of the semi-circular canals, as visualized through (a) use of the transgenic strain nkhspdmc12a, a fluorescent reporter expressed in otic epithelium; and (b) injection of the fluorescent lipophilic dye DiI. We also noted the altered expression of genes encoding ECM proteins or modifying enzymes. Other malformations of the inner ear observed in our work include the loss or reduced size of the utricular and saccular otoliths, suggesting a role for RA in otolith maintenance. CONCLUSION: Our work has identified a previously undescribed late phase of RA activity in otic development, demonstrating that vestibular defects observed in human patients in relation to perturbed RA signaling are not solely due to its early disruption in otic development.


Subject(s)
Tretinoin , Zebrafish , Animals , Humans , Zebrafish/genetics , Otolithic Membrane , Embryonic Development , Semicircular Canals , Morphogenesis
2.
J Biol Chem ; 296: 100073, 2021.
Article in English | MEDLINE | ID: mdl-33187986

ABSTRACT

Tubby-like proteins (TULPs) are characterized by a conserved C-terminal domain that binds phosphoinositides. Collectively, mammalian TULP1-4 proteins play essential roles in intracellular transport, cell differentiation, signaling, and motility. Yet, little is known about how the function of these proteins is regulated in cells. Here, we present the protein-protein interaction network of TULP3, a protein that is responsible for the trafficking of G-protein-coupled receptors to cilia and whose aberrant expression is associated with severe developmental disorders and polycystic kidney disease. We identify several protein interaction nodes linked to TULP3 that include enzymes involved in acetylation and ubiquitination. We show that acetylation of two key lysine residues on TULP3 by p300 increases TULP3 protein abundance and that deacetylation of these sites by HDAC1 decreases protein levels. Furthermore, we show that one of these sites is ubiquitinated in the absence of acetylation and that acetylation inversely correlates with ubiquitination of TULP3. This mechanism is evidently conserved across species and is active in zebrafish during development. Finally, we identify this same regulatory module in TULP1, TULP2, and TULP4 and demonstrate that the stability of these proteins is similarly modulated by an acetylation switch. This study unveils a signaling pathway that links nuclear enzymes to ciliary membrane receptors via TULP3, describes a dynamic mechanism for the regulation of all tubby-like proteins, and explores how to exploit it pharmacologically using drugs.


Subject(s)
Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Eye Proteins/genetics , HEK293 Cells , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Stability , p300-CBP Transcription Factors/genetics
3.
Hum Genet ; 141(8): 1385-1407, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35089417

ABSTRACT

Coloboma, a congenital disorder characterized by gaps in ocular tissues, is caused when the choroid fissure fails to close during embryonic development. Several loci have been associated with coloboma, but these represent less than 40% of those that are involved with this disease. Here, we describe a novel coloboma-causing locus, BMP3. Whole exome sequencing and Sanger sequencing of patients with coloboma identified three variants in BMP3, two of which are predicted to be disease causing. Consistent with this, bmp3 mutant zebrafish have aberrant fissure closure. bmp3 is expressed in the ventral head mesenchyme and regulates phosphorylated Smad3 in a population of cells adjacent to the choroid fissure. Furthermore, mutations in bmp3 sensitize embryos to Smad3 inhibitor treatment resulting in open choroid fissures. Micro CT scans and Alcian blue staining of zebrafish demonstrate that mutations in bmp3 cause midface hypoplasia, suggesting that bmp3 regulates cranial neural crest cells. Consistent with this, we see active Smad3 in a population of periocular neural crest cells, and bmp3 mutant zebrafish have reduced neural crest cells in the choroid fissure. Taken together, these data suggest that Bmp3 controls Smad3 phosphorylation in neural crest cells to regulate early craniofacial and ocular development.


Subject(s)
Coloboma , Animals , Coloboma/genetics , Eye , Neural Crest , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
J Anat ; 239(4): 801-828, 2021 10.
Article in English | MEDLINE | ID: mdl-34047378

ABSTRACT

The vertebrate inner ear is a labyrinthine sensory organ responsible for perceiving sound and body motion. While a great deal of research has been invested in understanding the auditory system, a growing body of work has begun to delineate the complex developmental program behind the apparatuses of the inner ear involved with vestibular function. These animal studies have helped identify genes involved in inner ear development and model syndromes known to include vestibular dysfunction, paving the way for generating treatments for people suffering from these disorders. This review will provide an overview of known inner ear anatomy and function and summarize the exciting discoveries behind inner ear development and the evolution of its vestibular apparatuses.


Subject(s)
Vestibule, Labyrinth , Animals , Humans , Vertebrates
5.
PLoS Genet ; 14(3): e1007246, 2018 03.
Article in English | MEDLINE | ID: mdl-29522511

ABSTRACT

The eye primordium arises as a lateral outgrowth of the forebrain, with a transient fissure on the inferior side of the optic cup providing an entry point for developing blood vessels. Incomplete closure of the inferior ocular fissure results in coloboma, a disease characterized by gaps in the inferior eye and recognized as a significant cause of pediatric blindness. Here, we identify eight patients with defects in tissues of the superior eye, a congenital disorder that we term superior coloboma. The embryonic origin of superior coloboma could not be explained by conventional models of eye development, leading us to reanalyze morphogenesis of the dorsal eye. Our studies revealed the presence of the superior ocular sulcus (SOS), a transient division of the dorsal eye conserved across fish, chick, and mouse. Exome sequencing of superior coloboma patients identified rare variants in a Bone Morphogenetic Protein (Bmp) receptor (BMPR1A) and T-box transcription factor (TBX2). Consistent with this, we find sulcus closure defects in zebrafish lacking Bmp signaling or Tbx2b. In addition, loss of dorsal ocular Bmp is rescued by concomitant suppression of the ventral-specific Hedgehog pathway, arguing that sulcus closure is dependent on dorsal-ventral eye patterning cues. The superior ocular sulcus acts as a conduit for blood vessels, with altered sulcus closure resulting in inappropriate connections between the hyaloid and superficial vascular systems. Together, our findings explain the existence of superior coloboma, a congenital ocular anomaly resulting from aberrant morphogenesis of a developmental structure.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Coloboma/embryology , Coloboma/genetics , Cytochrome P-450 CYP1B1/genetics , Eye/embryology , Adult , Animals , Animals, Genetically Modified , Bone Morphogenetic Protein Receptors, Type I/metabolism , Chick Embryo , Embryo, Nonmammalian , Growth Differentiation Factor 6/genetics , Growth Differentiation Factor 6/metabolism , Humans , Infant , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Am J Med Genet C Semin Med Genet ; 184(3): 590-610, 2020 09.
Article in English | MEDLINE | ID: mdl-32852110

ABSTRACT

Ocular coloboma is a congenital disorder of the eye where a gap exists in the inferior retina, lens, iris, or optic nerve tissue. With a prevalence of 2-19 per 100,000 live births, coloboma, and microphthalmia, an associated ocular disorder, represent up to 10% of childhood blindness. It manifests due to the failure of choroid fissure closure during eye development, and it is a part of a spectrum of ocular disorders that include microphthalmia and anophthalmia. Use of genetic approaches from classical pedigree analyses to next generation sequencing has identified more than 40 loci that are associated with the causality of ocular coloboma. As we have expanded studies to include singleton cases, hereditability has been very challenging to prove. As such, researchers over the past 20 years, have unraveled the complex interrelationship amongst these 40 genes using vertebrate model organisms. Such research has greatly increased our understanding of eye development. These genes function to regulate initial specification of the eye field, migration of retinal precursors, patterning of the retina, neural crest cell biology, and activity of head mesoderm. This review will discuss the discovery of loci using patient data, their investigations in animal models, and the recent advances stemming from animal models that shed new light in patient diagnosis.


Subject(s)
Coloboma/genetics , Eye/growth & development , Microphthalmos/genetics , Animals , Child , Coloboma/pathology , Eye/metabolism , Humans , Microphthalmos/pathology , Neural Crest/growth & development , Neural Crest/metabolism , Organogenesis/genetics
7.
Dev Biol ; 388(2): 192-204, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24457098

ABSTRACT

Axial patterning of the developing eye is critically important for proper axonal pathfinding as well as for key morphogenetic events, such as closure of the optic fissure. The dorsal retina is initially specified by the actions of Bone Morphogenetic Protein (BMP) signaling, with such identity subsequently maintained by the Wnt-ß catenin pathway. Using zebrafish as a model system, we demonstrate that Secreted frizzled-related protein 1a (Sfrp1a) and Sfrp5 work cooperatively to pattern the retina along the dorso-ventral axis. Sfrp1a/5 depleted embryos display a reduction in dorsal marker gene expression that is consistent with defects in BMP- and Wnt-dependent dorsal retina identity. In accord with this finding, we observe a marked reduction in transgenic reporters of BMP and Wnt signaling within the dorsal retina of Sfrp1a/5 depleted embryos. In contrast to studies in which canonical Wnt signaling is blocked, we note an increase in BMP ligand expression in Sfrp1a/5 depleted embryos, a phenotype similar to that seen in embryos with inhibited BMP signaling. Overexpression of a low dose of sfrp5 mRNA causes an increase in dorsal retina marker gene expression. We propose a model in which Sfrp proteins function as facilitators of both BMP and Wnt signaling within the dorsal retina.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Retina/embryology , Signal Transduction , Wnt Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Base Sequence , DNA Primers , In Situ Hybridization , Morphogenesis , Real-Time Polymerase Chain Reaction , Zebrafish
8.
Hum Mol Genet ; 22(7): 1432-42, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23307924

ABSTRACT

Retinal dystrophies are predominantly caused by mutations affecting the visual phototransduction system and cilia, with few genes identified that function to maintain photoreceptor survival. We reasoned that growth factors involved with early embryonic retinal development would represent excellent candidates for such diseases. Here we show that mutations in the transforming growth factor-ß (TGF-ß) ligand Growth Differentiation Factor 6, which specifies the dorso-ventral retinal axis, contribute to Leber congenital amaurosis. Furthermore, deficiency of gdf6 results in photoreceptor degeneration, so demonstrating a connection between Gdf6 signaling and photoreceptor survival. In addition, in both murine and zebrafish mutant models, we observe retinal apoptosis, a characteristic feature of human retinal dystrophies. Treatment of gdf6-deficient zebrafish embryos with a novel aminopropyl carbazole, P7C3, rescued the retinal apoptosis without evidence of toxicity. These findings implicate for the first time perturbed TGF-ß signaling in the genesis of retinal dystrophies, support the study of related morphogenetic genes for comparable roles in retinal disease and may offer additional therapeutic opportunities for genetically heterogeneous disorders presently only treatable with gene therapy.


Subject(s)
Cell Survival , Growth Differentiation Factor 6/genetics , Leber Congenital Amaurosis/genetics , Retinitis Pigmentosa/genetics , Amino Acid Sequence , Animals , Apoptosis , DNA Mutational Analysis , Genetic Association Studies , Growth Differentiation Factor 6/physiology , Humans , Leber Congenital Amaurosis/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Pedigree , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/physiology , Retina/pathology , Retinitis Pigmentosa/pathology , Zebrafish
9.
Front Cell Dev Biol ; 12: 1338070, 2024.
Article in English | MEDLINE | ID: mdl-38385025

ABSTRACT

Humans and other jawed vertebrates rely heavily on their craniofacial skeleton for eating, breathing, and communicating. As such, it is vital that the elements of the craniofacial skeleton develop properly during embryogenesis to ensure a high quality of life and evolutionary fitness. Indeed, craniofacial abnormalities, including cleft palate and craniosynostosis, represent some of the most common congenital abnormalities in newborns. Like many other organ systems, the development of the craniofacial skeleton is complex, relying on specification and migration of the neural crest, patterning of the pharyngeal arches, and morphogenesis of each skeletal element into its final form. These processes must be carefully coordinated and integrated. One way this is achieved is through the spatial and temporal deployment of cell signaling pathways. Recent studies conducted using the zebrafish model underscore the importance of the Transforming Growth Factor Beta (TGF-ß) and Bone Morphogenetic Protein (BMP) pathways in craniofacial development. Although both pathways contain similar components, each pathway results in unique outcomes on a cellular level. In this review, we will cover studies conducted using zebrafish that show the necessity of these pathways in each stage of craniofacial development, starting with the induction of the neural crest, and ending with the morphogenesis of craniofacial elements. We will also cover human skeletal and craniofacial diseases and malformations caused by mutations in the components of these pathways (e.g., cleft palate, craniosynostosis, etc.) and the potential utility of zebrafish in studying the etiology of these diseases. We will also briefly cover the utility of the zebrafish model in joint development and biology and discuss the role of TGF-ß/BMP signaling in these processes and the diseases that result from aberrancies in these pathways, including osteoarthritis and multiple synostoses syndrome. Overall, this review will demonstrate the critical roles of TGF-ß/BMP signaling in craniofacial development and show the utility of the zebrafish model in development and disease.

10.
BMC Dev Biol ; 13: 31, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23937294

ABSTRACT

BACKGROUND: The reiterated architecture of cranial motor neurons aligns with the segmented structure of the embryonic vertebrate hindbrain. Anterior-posterior identity of cranial motor neurons depends, in part, on retinoic acid signaling levels. The early vertebrate embryo maintains a balance between retinoic acid synthetic and degradative zones on the basis of reciprocal expression domains of the retinoic acid synthesis gene aldhehyde dehydrogenase 1a2 (aldh1a2) posteriorly and the oxidative gene cytochrome p450 type 26a1 (cyp26a1) in the forebrain, midbrain, and anterior hindbrain. RESULTS: This manuscript investigates the role of zinc finger of the cerebellum (zic) transcription factors in regulating levels of retinoic acid and differentiation of cranial motor neurons. Depletion of zebrafish Zic2a and Zic2b results in a strong downregulation of aldh1a2 expression and a concomitant reduction in activity of a retinoid-dependent transgene. The vagal motor neuron phenotype caused by loss of Zic2a/2b mimics a depletion of Aldh1a2 and is rescued by exogenously supplied retinoic acid. CONCLUSION: Zic transcription factors function in patterning hindbrain motor neurons through their regulation of embryonic retinoic acid signaling.


Subject(s)
Rhombencephalon/metabolism , Signal Transduction , Transcription Factors/metabolism , Tretinoin/metabolism , Aldehyde Dehydrogenase 1 Family , Cytochrome P-450 Enzyme System/genetics , Humans , Neurons/metabolism , Retinal Dehydrogenase/genetics , Retinoic Acid 4-Hydroxylase , Vagus Nerve/cytology , Vagus Nerve/metabolism , Zebrafish Proteins
11.
Neurobiol Dis ; 55: 11-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23523635

ABSTRACT

The function of the cellular prion protein (PrP(C)) in healthy brains remains poorly understood, in part because Prnp knockout mice are viable. On the other hand, transient knockdown of Prnp homologs in zebrafish (including two paralogs, prp1 and prp2) has suggested that PrP(C) is required for CNS development, cell adhesion, and neuroprotection. It has been argued that zebrafish Prp2 is most similar to mammalian PrP(C), yet it has remained intransigent to the most thorough confirmations of reagent specificity during knockdown. Thus we investigated the role of prp2 using targeted gene disruption via zinc finger nucleases. Prp2(-/-) zebrafish were viable and did not display overt developmental phenotypes. Back-crossing female prp2(-/-) fish ruled out a role for maternal mRNA contributions. Prp2(-/-) larvae were found to have increased seizure-like behavior following exposure to the convulsant pentylenetetrazol (PTZ), as compared to wild type fish. In situ recordings from intact hindbrains demonstrated that prp2 regulates closing of N-Methyl-d-aspartate (NMDA) receptors, concomitant with neuroprotection during glutamate excitotoxicity. Overall, the knockout of Prp2 function in zebrafish independently confirmed hypothesized roles for PrP, identifying deeply conserved functions in post-developmental regulation of neuron excitability that are consequential to the etiology of prion and Alzheimer diseases.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Mutation/genetics , Neurons/metabolism , Prions/genetics , Age Factors , Animals , Animals, Genetically Modified , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/physiopathology , Gene Library , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Larva , Mice , Mutagenesis, Site-Directed , Pentylenetetrazole/toxicity , Phenotype , Receptors, N-Methyl-D-Aspartate/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zinc Fingers/genetics
12.
Dev Biol ; 355(1): 55-64, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21539831

ABSTRACT

Mutations in H6-homeobox (HMX) genes are linked to neural mispatterning and neural tube closure defects in humans. We demonstrate that zebrafish Hmx4 regulates the signaling of two morphogens critical for neural development, retinoic acid (RA) and Sonic hedgehog (Shh). Hmx4-depleted embryos have a strongly narrowed eye field and reduced forebrain Shh target gene expression. hmx4 morphants fail to properly transcribe the Shh signal transducer gli3, and have reduced ventral forebrain specification. Hmx4-depleted embryos also have neural tube patterning defects that phenocopy RA-deficiency. We show that Hmx4 is required for the initiation and maintenance of aldh1a2, the principal RA-synthesizing gene. Loss of RA is the primary defect in Hmx4-depleted embryos, as RA treatment rescues a number of the neural patterning defects. Surprisingly, RA treatment also rescues forebrain morphology, gli3 transcription, and Shh signaling. We propose that Hmx4 is a critical regulator of retinoic acid synthesis in a developing embryo, and that this regulation is essential for controlling Shh signaling and forebrain development.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Homeodomain Proteins/metabolism , Prosencephalon/embryology , Tretinoin/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Aldehyde Dehydrogenase/genetics , Animals , Body Patterning/genetics , Homeodomain Proteins/genetics , Prosencephalon/metabolism , Signal Transduction , Transcription Factors/genetics , Zebrafish/genetics , Zinc Finger Protein Gli3
13.
Biochim Biophys Acta ; 1812(3): 390-401, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20850526

ABSTRACT

In this review, we highlight recent literature concerning the signaling mechanisms underlying the development of two neural birth defects, holoprosencephaly and coloboma. Holoprosencephaly, the most common forebrain defect, occurs when the cerebral hemispheres fail to separate and is typically associated with mispatterning of embryonic midline tissue. Coloboma results when the choroid fissure in the eye fails to close. It is clear that Sonic hedgehog (Shh) signaling regulates both forebrain and eye development, with defects in Shh, or components of the Shh signaling cascade leading to the generation of both birth defects. In addition, other intercellular signaling pathways are known factors in the incidence of holoprosencephaly and coloboma. This review will outline recent advances in our understanding of forebrain and eye embryonic pattern formation, with a focus on zebrafish studies of Shh and retinoic acid pathways. Given the clear overlap in the mechanisms that generate both diseases, we propose that holoprosencephaly and coloboma can represent mild and severe aspects of single phenotypic spectrum resulting from aberrant forebrain development. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.


Subject(s)
Coloboma/etiology , Gene Expression Regulation, Developmental , Holoprosencephaly/etiology , Prosencephalon/abnormalities , Prosencephalon/metabolism , Signal Transduction , Animals , Coloboma/metabolism , Coloboma/pathology , Disease Models, Animal , Holoprosencephaly/metabolism , Holoprosencephaly/pathology , Humans , Prosencephalon/embryology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
14.
Hum Mol Genet ; 19(2): 287-98, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19864492

ABSTRACT

Ocular mal-development results in heterogeneous and frequently visually disabling phenotypes that include coloboma and microphthalmia. Due to the contribution of bone morphogenetic proteins to such processes, the function of the paralogue Growth Differentiation Factor 3 was investigated. Multiple mis-sense variants were identified in patients with ocular and/or skeletal (Klippel-Feil) anomalies including one individual with heterozygous alterations in GDF3 and GDF6. These variants were characterized, individually and in combination, through integrated biochemical and zebrafish model organism analyses, demonstrating appreciable effects with western blot analyses, luciferase based reporter assays and antisense morpholino inhibition. Notably, inhibition of the zebrafish co-orthologue of GDF3 accurately recapitulates patient phenotypes. By demonstrating the pleiotropic effects of GDF3 mutation, these results extend the contribution of perturbed BMP signaling to human disease and potentially implicate multi-allelic inheritance of BMP variants in developmental disorders.


Subject(s)
Eye Abnormalities/genetics , Growth Differentiation Factor 3/genetics , Muscle, Skeletal/abnormalities , Mutation , Amino Acid Sequence , Animals , Cell Line , Eye Abnormalities/metabolism , Female , Growth Differentiation Factor 3/chemistry , Growth Differentiation Factor 3/metabolism , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Pedigree , Sequence Alignment , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Genesis ; 49(9): 725-42, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21714061

ABSTRACT

In flies, the zinc-finger protein Teashirt promotes trunk segmental identities, in part, by repressing the expression and function of anterior hox paralog group (PG) 1-4 genes that specify head fates. Anterior-posterior patterning of the vertebrate hindbrain also requires Hox PG 1-4 function, but the role of vertebrate teashirt-related genes in this process has not been investigated. In this work, we use overexpression and structure-function analyses to show that zebrafish tshz3b antagonizes Hox-dependent hindbrain segmentation. Ectopic Tshz3b perturbs the specification of rhombomere identities and leads to the caudal expansion of r1, the only rhombomere whose identity is specified independently of Hox function. This overexpression phenotype does not require the homeodomain and C-terminal zinc fingers that are unique to vertebrate Teashirt-related proteins, but does require that Tshz3b function as a repressor. Together, these results argue that the negative regulation of Hox PG 1-4 function is a conserved characteristic of Teashirt-related proteins.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Rhombencephalon/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning/genetics , Down-Regulation/genetics , Gene Expression/genetics , Homeodomain Proteins/genetics , Mutation , Phenotype , RNA, Messenger/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rhombencephalon/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zinc Fingers/genetics , Zinc Fingers/physiology
16.
Hum Mol Genet ; 18(6): 1110-21, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19129173

ABSTRACT

Proteins of the bone morphogenetic protein (BMP) family are known to have a role in ocular and skeletal development; however, because of their widespread expression and functional redundancy, less progress has been made identifying the roles of individual BMPs in human disease. We identified seven heterozygous mutations in growth differentiation factor 6 (GDF6), a member of the BMP family, in patients with both ocular and vertebral anomalies, characterized their effects with a SOX9-reporter assay and western analysis, and demonstrated comparable phenotypes in model organisms with reduced Gdf6 function. We observed a spectrum of ocular and skeletal anomalies in morphant zebrafish, the latter encompassing defective tail formation and altered expression of somite markers noggin1 and noggin2. Gdf6(+/-) mice exhibited variable ocular phenotypes compatible with phenotypes observed in patients and zebrafish. Key differences evident between patients and animal models included pleiotropic effects, variable expressivity and incomplete penetrance. These data establish the important role of this determinant in ocular and vertebral development, demonstrate the complex genetic inheritance of these phenotypes, and further understanding of BMP function and its contributions to human disease.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Growth Differentiation Factor 6/genetics , Penetrance , Amino Acid Sequence , Animals , DNA Mutational Analysis , Genes, Reporter , Growth Differentiation Factor 6/chemistry , Humans , Mice , Models, Animal , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation/genetics , Oligonucleotides, Antisense/pharmacology , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
17.
Genome ; 54(12): 973-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22077099

ABSTRACT

Ladybird (Lbx) homeodomain transcription factors function in neural and muscle development--roles conserved from Drosophila to vertebrates. Lbx expression in mice specifies neural cell types, including dorsally located interneurons and association neurons, within the neural tube. Little, however, is known about the regulation of vertebrate lbx family genes. Here we describe the expression pattern of three zebrafish ladybird genes via mRNA in situ hybridization. Zebrafish lbx genes are expressed in distinct but overlapping regions within the developing neural tube, with strong expression within the hindbrain and spinal cord. The Hox family of transcription factors, in cooperation with cofactors such as Pbx and Meis, regulate hindbrain segmentation during embryogenesis. We have identified a novel regulatory interaction in which lbx1 genes are strongly downregulated in Pbx-depleted embryos. Further, we have produced a transgenic zebrafish line expressing dTomato and EGFP under the control of an lbx1b enhancer--a useful tool to acertain neuron location, migration, and morphology. Using this transgenic strain, we have identified a minimal neural lbx1b enhancer that contains key regulatory elements for expression of this transcription factor.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Repressor Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Biomarkers , Cloning, Molecular , DNA-Binding Proteins/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryonic Development , Enhancer Elements, Genetic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization/methods , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
18.
Genes (Basel) ; 12(2)2021 01 26.
Article in English | MEDLINE | ID: mdl-33530637

ABSTRACT

Precise spatiotemporal expression of the Nodal-Lefty-Pitx2 cascade in the lateral plate mesoderm establishes the left-right axis, which provides vital cues for correct organ formation and function. Mutations of one cascade constituent PITX2 and, separately, the Forkhead transcription factor FOXC1 independently cause a multi-system disorder known as Axenfeld-Rieger syndrome (ARS). Since cardiac involvement is an established ARS phenotype and because disrupted left-right patterning can cause congenital heart defects, we investigated in zebrafish whether foxc1 contributes to organ laterality or situs. We demonstrate that CRISPR/Cas9-generated foxc1a and foxc1b mutants exhibit abnormal cardiac looping and that the prevalence of cardiac situs defects is increased in foxc1a-/-; foxc1b-/- homozygotes. Similarly, double homozygotes exhibit isomerism of the liver and pancreas, which are key features of abnormal gut situs. Placement of the asymmetric visceral organs relative to the midline was also perturbed by mRNA overexpression of foxc1a and foxc1b. In addition, an analysis of the left-right patterning components, identified in the lateral plate mesoderm of foxc1 mutants, reduced or abolished the expression of the NODAL antagonist lefty2. Together, these data reveal a novel contribution from foxc1 to left-right patterning, demonstrating that this role is sensitive to foxc1 gene dosage, and provide a plausible mechanism for the incidence of congenital heart defects in Axenfeld-Rieger syndrome patients.


Subject(s)
Anterior Eye Segment/abnormalities , Eye Abnormalities/diagnosis , Eye Abnormalities/etiology , Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/etiology , Forkhead Transcription Factors/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Phenotype , Alleles , Animals , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Genetic Association Studies/methods , Genotype , Humans , Mesoderm/embryology , Mesoderm/metabolism , Mutation , Zebrafish
19.
Dev Biol ; 333(1): 37-47, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19545559

ABSTRACT

Dorsal-ventral patterning of the vertebrate retina is essential for accurate topographic mapping of retinal ganglion cell (RGC) axons to visual processing centers. Bone morphogenetic protein (Bmp) growth factors regulate dorsal retinal identity in vertebrate models, but the developmental timing of this signaling and the relative roles of individual Bmps remain unclear. In this study, we investigate the functions of two zebrafish Bmps, Gdf6a and Bmp4, during initiation of dorsal retinal identity, and subsequently during lens differentiation. Knockdown of zebrafish Gdf6a blocks initiation of retinal Smad phosphorylation and dorsal marker expression, while knockdown of Bmp4 produces no discernable retinal phenotype. These data, combined with analyses of embryos ectopically expressing Bmps, demonstrate that Gdf6a is necessary and sufficient for initiation of dorsal retinal identity. We note a profound expansion of ventral retinal identity in gdf6a morphants, demonstrating that dorsal BMP signaling antagonizes ventral marker expression. Finally, we demonstrate a role for Gdf6a in non-neural ocular tissues. Knockdown of Gdf6a leads to defects in lens-specific gene expression, and when combined with Bmp signaling inhibitors, disrupts lens fiber cell differentiation. Taken together, these data indicate that Gdf6a initiates dorsal retinal patterning independent of Bmp4, and regulates lens differentiation.


Subject(s)
Growth Differentiation Factor 6/physiology , Lens, Crystalline/embryology , Retina/embryology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/physiology , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Embryo, Nonmammalian/physiology , Gene Knockdown Techniques , Growth Differentiation Factor 6/biosynthesis , Growth Differentiation Factor 6/genetics , Lens, Crystalline/metabolism , Retina/metabolism , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Smad Proteins/physiology , T-Box Domain Proteins/genetics , Zebrafish , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
20.
Hum Mol Genet ; 17(4): 525-38, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17998248

ABSTRACT

Holoprosencephaly (HPE) is the most common human congenital forebrain defect, affecting specification of forebrain tissue and subsequent division of the cerebral hemispheres. The causes of HPE are multivariate and heterogeneous, and include exposure to teratogens, such as retinoic acid (RA), and mutations in forebrain patterning genes. Many of the defects in HPE patients resemble animal models with aberrant RA levels, which also show severe forebrain abnormalities. RA plays an important role in early neural patterning of the vertebrate embryo: expression of RA-synthesizing enzymes initiates high RA levels in the trunk, which are required for proper anterior-posterior patterning of the hindbrain and spinal cord. In the forebrain and midbrain, RA-degrading enzymes are expressed, protecting these regions from the effects of RA. However, the mechanisms that regulate RA-synthesizing and RA-degrading enzymes are poorly understood. Mutations in the gene TGIF are associated with incidence of HPE. We demonstrate in zebrafish that Tgif plays a key role in regulating RA signaling, and is essential to properly pattern the forebrain. Tgif is necessary for normal initiation of genes that control RA synthesis and degradation, resulting in defects in RA-dependent central nervous system patterning in Tgif-depleted embryos. The loss of the forebrain-specific RA-degrading enzyme cyp26a1 causes a forebrain phenotype that mimics tgif morphants. We propose a model in which Tgif controls forebrain patterning by regulating RA degradation. The consequences of abnormal RA levels for forebrain patterning are profound, and imply that in human patients with TGIF deficiencies, increased forebrain RA levels contribute to the development of HPE.


Subject(s)
Holoprosencephaly/embryology , Holoprosencephaly/metabolism , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Tretinoin/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Base Sequence , Body Patterning , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA, Complementary/genetics , Disease Models, Animal , Gene Expression Regulation, Developmental , Genes, Homeobox , Holoprosencephaly/genetics , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Oligodeoxyribonucleotides, Antisense/genetics , Phenotype , Prosencephalon/abnormalities , Prosencephalon/embryology , Prosencephalon/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Retinoic Acid 4-Hydroxylase , Rhombencephalon/abnormalities , Rhombencephalon/embryology , Rhombencephalon/metabolism , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL