Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Exp Eye Res ; 234: 109599, 2023 09.
Article in English | MEDLINE | ID: mdl-37488009

ABSTRACT

Limbal epithelial stem cells are not only critical for corneal epithelial homeostasis but also have the capacity to change from a relatively quiescent mitotic phenotype to a rapidly proliferating cell in response to population depletion following corneal epithelial wounding. Pax6+/- mice display many abnormalities including corneal vascularization and these aberrations are consistent with a limbal stem cell deficiency (LSCD) phenotype. FoxC1 has an inhibitory effect on corneal avascularity and a positive role in stem cell maintenance in many tissues. However, the role of FoxC1 in limbal epithelial stem cells remains unknown. To unravel FoxC1's role(s) in limbal epithelial stem cell homeostasis, we utilized an adeno-associated virus (AAV) vector to topically deliver human FOXC1 proteins into Pax6 +/- mouse limbal epithelium. Under unperturbed conditions, overexpression of FOXC1 in the limbal epithelium had little significant change in differentiation (PAI-2, Krt12) and proliferation (BrdU, Ki67). Conversely, such overexpression resulted in a marked increase in the expression of putative limbal epithelial stem cell markers, N-cadherin and Lrig1. After corneal injuries in Pax6 +/- mice, FOXC1 overexpression enhanced the behavior of limbal epithelial stem cells from quiescence to a highly proliferative status. Overall, the treatment of AAV8-FOXC1 may be beneficial to the function of limbal epithelial stem cells in the context of a deficiency of Pax6 function.


Subject(s)
Corneal Diseases , Epithelium, Corneal , Limbus Corneae , Animals , Humans , Mice , Cornea , Corneal Diseases/metabolism , Debridement , Epithelial Cells , Epithelium, Corneal/metabolism , Limbus Corneae/metabolism , Stem Cells
2.
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
3.
EBioMedicine ; 67: 103360, 2021 May.
Article in English | MEDLINE | ID: mdl-33975254

ABSTRACT

Ocular morphogenesis in vertebrates is a highly organized process, orchestrated largely by intrinsic genetic programs that exhibit stringent spatiotemporal control. Alternations in these genetic instructions can lead to hereditary or nonhereditary congenital disorders, a major cause of childhood visual impairment, and contribute to common late-onset blinding diseases. Currently, limited treatment options exist for clinical phenotypes involving eye development. This review summarizes recent advances in our understanding of early-onset ocular disorders and highlights genetic complexities in development and diseases, specifically focusing on coloboma, congenital glaucoma and Leber congenital amaurosis. We also discuss innovative paradigms for potential therapeutic modalities.


Subject(s)
Eye Diseases, Hereditary/genetics , Child , Eye/embryology , Eye/metabolism , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/therapy , Genetic Therapy/methods , Humans , Molecular Targeted Therapy/methods , Stem Cell Transplantation/methods
4.
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
5.
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
6.
Dev Biol ; 453(1): 34-47, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31199900

ABSTRACT

Vascular smooth muscle of the head derives from neural crest, but developmental mechanisms and early transcriptional drivers of the vSMC lineage are not well characterized. We find that in early development, the transcription factor foxc1b is expressed in mesenchymal cells that associate with the vascular endothelium. Using timelapse imaging, we observe that foxc1b expressing mesenchymal cells differentiate into acta2 expressing vascular mural cells. We show that in zebrafish, while foxc1b is co-expressed in acta2 positive smooth muscle cells that associate with large diameter vessels, it is not co-expressed in capillaries where pdgfrß positive pericytes are located. In addition to being an early marker of the lineage, foxc1 is essential for vSMC differentiation; we find that foxc1 loss of function mutants have defective vSMC differentiation and that early genetic ablation of foxc1b or acta2 expressing populations blocks vSMC differentiation. Furthermore, foxc1 is expressed upstream of acta2 and is required for acta2 expression in vSMCs. Using RNA-Seq we determine an enriched intersectional gene expression profile using dual expression of foxc1b and acta2 to identify novel vSMC markers. Taken together, our data suggests that foxc1 is a marker of vSMCs and plays a critical functional role in promoting their differentiation.


Subject(s)
Cell Differentiation , Embryo, Nonmammalian/cytology , Forkhead Transcription Factors/metabolism , Head/blood supply , Head/embryology , Muscle, Smooth, Vascular/cytology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Brain/embryology , Brain/metabolism , Cell Differentiation/genetics , Embryo, Nonmammalian/metabolism , Endothelium/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , Transcriptome/genetics , Up-Regulation , Zebrafish/genetics
7.
Hum Mol Genet ; 28(8): 1298-1311, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30561643

ABSTRACT

Pigmentary glaucoma (PG) is a common glaucoma subtype that results from release of pigment from the iris, called pigment dispersion syndrome (PDS), and its deposition throughout the anterior chamber of the eye. Although PG has a substantial heritable component, no causative genes have yet been identified. We used whole exome sequencing of two independent pedigrees to identify two premelanosome protein (PMEL) variants associated with heritable PDS/PG. PMEL encodes a key component of the melanosome, the organelle essential for melanin synthesis, storage and transport. Targeted screening of PMEL in three independent cohorts (n = 394) identified seven additional PDS/PG-associated non-synonymous variants. Five of the nine variants exhibited defective processing of the PMEL protein. In addition, analysis of PDS/PG-associated PMEL variants expressed in HeLa cells revealed structural changes to pseudomelanosomes indicating altered amyloid fibril formation in five of the nine variants. Introduction of 11-base pair deletions to the homologous pmela in zebrafish by the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 method caused profound pigmentation defects and enlarged anterior segments in the eye, further supporting PMEL's role in ocular pigmentation and function. Taken together, these data support a model in which missense PMEL variants represent dominant negative mutations that impair the ability of PMEL to form functional amyloid fibrils. While PMEL mutations have previously been shown to cause pigmentation and ocular defects in animals, this research is the first report of mutations in PMEL causing human disease.


Subject(s)
Glaucoma, Open-Angle/genetics , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/physiology , Adult , Amyloid/metabolism , Animals , Female , HeLa Cells , Humans , Iris/metabolism , Male , Melanosomes/genetics , Middle Aged , Mutation, Missense/genetics , Pedigree , Pigmentation/genetics , Exome Sequencing/methods , Young Adult , Zebrafish
8.
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
9.
Proc Natl Acad Sci U S A ; 114(34): E7131-E7139, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28778995

ABSTRACT

EGR1 is an early growth response zinc finger transcription factor with broad actions, including in differentiation, mitogenesis, tumor suppression, and neuronal plasticity. Here we demonstrate that Egr1-/- mice on the C57BL/6 background have normal eyelid development, but back-crossing to BALB/c background for four or five generations resulted in defective eyelid development by day E15.5, at which time EGR1 was expressed in eyelids of WT mice. Defective eyelid formation correlated with profound ocular anomalies evident by postnatal days 1-4, including severe cryptophthalmos, microphthalmia or anophthalmia, retinal dysplasia, keratitis, corneal neovascularization, cataracts, and calcification. The BALB/c albino phenotype-associated Tyrc tyrosinase mutation appeared to contribute to the phenotype, because crossing the independent Tyrc-2J allele to Egr1-/- C57BL/6 mice also produced ocular abnormalities, albeit less severe than those in Egr1-/- BALB/c mice. Thus EGR1, in a genetic background-dependent manner, plays a critical role in mammalian eyelid development and closure, with subsequent impact on ocular integrity.


Subject(s)
Eyelids/growth & development , Mice/genetics , Mice/metabolism , Animals , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Eye/growth & development , Eye/metabolism , Eyelids/metabolism , Gene Expression Regulation, Developmental , Mice/growth & development , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
10.
Mol Vis ; 23: 952-962, 2017.
Article in English | MEDLINE | ID: mdl-29296075

ABSTRACT

Purpose: To evaluate the ability of a targeted genome-wide association study (GWAS) to identify genes associated with central corneal thickness (CCT). Methods: A targeted GWAS was used to investigate whether ten candidate genes with known roles in corneal development were associated with CCT in two Singaporean populations. The single nucleotide polymorphisms (SNPs) within a 500 kb interval encompassing each candidate were analyzed, and in light of the resulting data, members of the Wnt pathway were subsequently screened using similar methodology. Results: Variants within the 500 kb interval encompassing three candidate genes, DKK1 (rs1896368, p=1.32×10-3), DKK2 (rs17510449, p=7.34×10-4), and FOXO1 (rs7326616, p=1.56×10-4 and rs4943785, p=1.19×10-3), were statistically significantly associated with CCT in the Singapore Indian population. DKK2 was statistically significantly associated with CCT in a separate Singapore Malaysian population (rs10015200, p=2.26×10-3). Analysis of Wnt signaling pathway genes in each population demonstrated that TCF7L2 (rs3814573, p=1.18×10-3), RYK (rs6763231, p=1.12×10-3 and rs4854785, p=1.11×10-3), and FZD8 (rs640827, p=5.17×10-4) were statistically significantly associated with CCT. Conclusions: The targeted GWAS identified four genes (DKK1, DKK2, RYK, and FZD8) with novel associations with CCT and confirmed known associations with two genes, FOXO1 and TCF7L2. All six participate in the Wnt pathway, supporting a broader role for Wnt signaling in regulating the thickness of the cornea. In parallel, this study demonstrated that a hypothesis-driven candidate gene approach can identify associations in existing GWAS data sets.


Subject(s)
Cornea/anatomy & histology , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Quantitative Trait, Heritable , Wnt Signaling Pathway/genetics , Animals , Asian People/genetics , Corneal Pachymetry , Female , Forkhead Box Protein O1/genetics , Humans , India/ethnology , Intercellular Signaling Peptides and Proteins/genetics , Malaysia/ethnology , Male , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Singapore/epidemiology , Transcription Factor 7-Like 2 Protein/genetics
11.
CMAJ ; 188(6): 449-450, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27044787
12.
Hum Mol Genet ; 25(7): 1382-91, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26908622

ABSTRACT

Ocular coloboma is a common eye malformation resulting from incomplete fusion of the optic fissure during development. Coloboma is often associated with microphthalmia and/or contralateral anophthalmia. Coloboma shows extensive locus heterogeneity associated with causative mutations identified in genes encoding developmental transcription factors or components of signaling pathways. We report an ultra-rare, heterozygous frameshift mutation in FZD5 (p.Ala219Glufs*49) that was identified independently in two branches of a large family with autosomal dominant non-syndromic coloboma. FZD5 has a single-coding exon and consequently a transcript with this frameshift variant is not a canonical substrate for nonsense-mediated decay. FZD5 encodes a transmembrane receptor with a conserved extracellular cysteine rich domain for ligand binding. The frameshift mutation results in the production of a truncated protein, which retains the Wingless-type MMTV integration site family member-ligand-binding domain, but lacks the transmembrane domain. The truncated protein was secreted from cells, and behaved as a dominant-negative FZD5 receptor, antagonizing both canonical and non-canonical WNT signaling. Expression of the resultant mutant protein caused coloboma and microphthalmia in zebrafish, and disruption of the apical junction of the retinal neural epithelium in mouse, mimicking the phenotype of Fz5/Fz8 compound conditional knockout mutants. Our studies have revealed a conserved role of Wnt-Frizzled (FZD) signaling in ocular development and directly implicate WNT-FZD signaling both in normal closure of the human optic fissure and pathogenesis of coloboma.


Subject(s)
Frameshift Mutation , Frizzled Receptors/genetics , Wnt Signaling Pathway , Animals , DNA Mutational Analysis , Female , Humans , Male , Mice , Microphthalmos/genetics , Microphthalmos/metabolism , Pedigree , Zebrafish/genetics , Zebrafish/metabolism
13.
J Clin Invest ; 124(11): 4877-81, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25250569

ABSTRACT

Patients with cerebral small-vessel disease (CSVD) exhibit perturbed end-artery function and have an increased risk for stroke and age-related cognitive decline. Here, we used targeted genome-wide association (GWA) analysis and defined a CSVD locus adjacent to the forkhead transcription factor FOXC1. Moreover, we determined that the linked SNPs influence FOXC1 transcript levels and demonstrated that patients as young as 1 year of age with altered FOXC1 function exhibit CSVD. MRI analysis of patients with missense and nonsense mutations as well as FOXC1-encompassing segmental duplication and deletion revealed white matter hyperintensities, dilated perivascular spaces, and lacunar infarction. In a zebrafish model, overexpression or morpholino-induced suppression of foxc1 induced cerebral hemorrhage. Inhibition of foxc1 perturbed platelet-derived growth factor (Pdgf) signaling, impairing neural crest migration and the recruitment of mural cells, which are essential for vascular stability. GWA analysis also linked the FOXC1-interacting transcription factor PITX2 to CSVD, and both patients with PITX2 mutations and murine Pitx2-/- mutants displayed brain vascular phenotypes. Together, these results extend the genetic etiology of stroke and demonstrate an increasing developmental basis for human cerebrovascular disease.


Subject(s)
Cerebral Small Vessel Diseases/genetics , Forkhead Transcription Factors/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Cerebral Hemorrhage/genetics , Codon, Nonsense , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Leukoencephalopathies/genetics , Linkage Disequilibrium , Mutation, Missense , Platelet-Derived Growth Factor/physiology , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Signal Transduction , Zebrafish , Homeobox Protein PITX2
14.
PLoS Genet ; 10(7): e1004491, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25010521

ABSTRACT

Ocular coloboma is a sight-threatening malformation caused by failure of the choroid fissure to close during morphogenesis of the eye, and is frequently associated with additional anomalies, including microphthalmia and cataracts. Although Hedgehog signaling is known to play a critical role in choroid fissure closure, genetic regulation of this pathway remains poorly understood. Here, we show that the transcription factor Sox11 is required to maintain specific levels of Hedgehog signaling during ocular development. Sox11-deficient zebrafish embryos displayed delayed and abnormal lens formation, coloboma, and a specific reduction in rod photoreceptors, all of which could be rescued by treatment with the Hedgehog pathway inhibitor cyclopamine. We further demonstrate that the elevated Hedgehog signaling in Sox11-deficient zebrafish was caused by a large increase in shha transcription; indeed, suppressing Shha expression rescued the ocular phenotypes of sox11 morphants. Conversely, over-expression of sox11 induced cyclopia, a phenotype consistent with reduced levels of Sonic hedgehog. We screened DNA samples from 79 patients with microphthalmia, anophthalmia, or coloboma (MAC) and identified two novel heterozygous SOX11 variants in individuals with coloboma. In contrast to wild type human SOX11 mRNA, mRNA containing either variant failed to rescue the lens and coloboma phenotypes of Sox11-deficient zebrafish, and both exhibited significantly reduced transactivation ability in a luciferase reporter assay. Moreover, decreased gene dosage from a segmental deletion encompassing the SOX11 locus resulted in microphthalmia and related ocular phenotypes. Therefore, our study reveals a novel role for Sox11 in controlling Hedgehog signaling, and suggests that SOX11 variants contribute to pediatric eye disorders.


Subject(s)
Coloboma/genetics , Embryonic Development/genetics , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , SOXC Transcription Factors/genetics , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics , Animals , Choroid Diseases/genetics , Choroid Diseases/metabolism , Choroid Diseases/pathology , Coloboma/metabolism , Coloboma/pathology , Embryo, Nonmammalian , Eye/growth & development , Eye/metabolism , Humans , Morphogenesis/genetics , RNA, Messenger/biosynthesis , SOXC Transcription Factors/biosynthesis , Signal Transduction/genetics , Zebrafish/genetics
15.
Hum Mol Genet ; 23(10): 2511-26, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24412933

ABSTRACT

Ocular coloboma is a congenital defect resulting from failure of normal closure of the optic fissure during embryonic eye development. This birth defect causes childhood blindness worldwide, yet the genetic etiology is poorly understood. Here, we identified a novel homozygous mutation in the SALL2 gene in members of a consanguineous family affected with non-syndromic ocular coloboma variably affecting the iris and retina. This mutation, c.85G>T, introduces a premature termination codon (p.Glu29*) predicted to truncate the SALL2 protein so that it lacks three clusters of zinc-finger motifs that are essential for DNA-binding activity. This discovery identifies SALL2 as the third member of the Drosophila homeotic Spalt-like family of developmental transcription factor genes implicated in human disease. SALL2 is expressed in the developing human retina at the time of, and subsequent to, optic fissure closure. Analysis of Sall2-deficient mouse embryos revealed delayed apposition of the optic fissure margins and the persistence of an anterior retinal coloboma phenotype after birth. Sall2-deficient embryos displayed correct posterior closure toward the optic nerve head, and upon contact of the fissure margins, dissolution of the basal lamina occurred and PAX2, known to be critical for this process, was expressed normally. Anterior closure was disrupted with the fissure margins failing to meet, or in some cases misaligning leading to a retinal lesion. These observations demonstrate, for the first time, a role for SALL2 in eye morphogenesis and that loss of function of the gene causes ocular coloboma in humans and mice.


Subject(s)
Codon, Nonsense , Coloboma/genetics , Transcription Factors/genetics , Adolescent , Animals , Child , Consanguinity , DNA Mutational Analysis , DNA-Binding Proteins , Eye/embryology , Eye/pathology , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homozygote , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pedigree , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism
16.
Am J Hum Genet ; 93(1): 158-66, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23810382

ABSTRACT

SHORT syndrome is a rare, multisystem disease characterized by short stature, anterior-chamber eye anomalies, characteristic facial features, lipodystrophy, hernias, hyperextensibility, and delayed dentition. As part of the FORGE (Finding of Rare Disease Genes) Canada Consortium, we studied individuals with clinical features of SHORT syndrome to identify the genetic etiology of this rare disease. Whole-exome sequencing in a family trio of an affected child and unaffected parents identified a de novo frameshift insertion, c.1906_1907insC (p.Asn636Thrfs*18), in exon 14 of PIK3R1. Heterozygous mutations in exon 14 of PIK3R1 were subsequently identified by Sanger sequencing in three additional affected individuals and two affected family members. One of these mutations, c.1945C>T (p.Arg649Trp), was confirmed to be a de novo mutation in one affected individual and was also identified and shown to segregate with the phenotype in an unrelated family. The other mutation, a de novo truncating mutation (c.1971T>G [p.Tyr657*]), was identified in another affected individual. PIK3R1 is involved in the phosphatidylinositol 3 kinase (PI3K) signaling cascade and, as such, plays an important role in cell growth, proliferation, and survival. Functional studies on lymphoblastoid cells with the PIK3R1 c.1906_1907insC mutation showed decreased phosphorylation of the downstream S6 target of the PI3K-AKT-mTOR pathway. Our findings show that PIK3R1 mutations are the major cause of SHORT syndrome and suggest that the molecular mechanism of disease might involve downregulation of the PI3K-AKT-mTOR pathway.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Frameshift Mutation , Growth Disorders/genetics , Hypercalcemia/genetics , Metabolic Diseases/genetics , Nephrocalcinosis/genetics , Adolescent , Child , Child, Preschool , DNA Mutational Analysis/methods , Exome , Exons , Female , Genetic Carrier Screening , Heterozygote , Humans , Infant, Newborn , Male , Pedigree , Phenotype , Phosphorylation , Signal Transduction
17.
Invest Ophthalmol Vis Sci ; 54(8): 5871-9, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23847306

ABSTRACT

PURPOSE: To characterize the molecular mechanisms underlying retinal apoptosis induced by loss of Gdf6, a TGFß ligand. METHODS: The role of Gdf6 in regulating apoptosis was studied using a zebrafish gdf6a(-/-) mutant, which encodes a truncated, nonfunctional protein. To investigate whether intrinsic or extrinsic apoptotic mechanisms were involved, morpholino antisense oligonucleotides targeting baxa, baxb, and p53 were employed. Caspase-3 immunohistochemistry (IHC) was performed to assay apoptosis. Pharmacologic inhibition (using SB203580) and IHC were used to investigate the role of p38 mitogen activated protein (MAP) kinase activation in gdf6a(-/-)-induced apoptosis. To assess the role of Gdf6a in transcriptional regulation of TGFß signal transducers, in situ hybridization (ISH) was performed using probes to smad1, 5, 7, and 8. RESULTS: Results indicate maximal ocular apoptosis occurs 28 hours post fertilization (hpf) in gdf6a(-/-) mutants that is mediated independently of p53 by intrinsic mechanisms involving Bax proteins. Also, gdf6a(-/-) mutants exhibit markedly increased p38 MAP kinase activation that can be inhibited to significantly reduce retinal apoptosis. A reduction in retinal smad1 expression was also noted in gdf6a(-/-) mutants. CONCLUSIONS: gdf6a(-/-)-induced apoptosis is characterized by the involvement of intrinsic apoptotic pathways, p38 MAP kinases, and dysregulated smad expression. Modulation of key mediators can inhibit retinal apoptosis offering potential avenues of therapy. However, the efficacy of pharmacomodulation in improvement of visual function needs to be further examined.


Subject(s)
Apoptosis/physiology , Growth Differentiation Factor 6/physiology , Retina/physiology , Zebrafish Proteins/physiology , Animals , Caspase 3/metabolism , Growth Differentiation Factor 6/genetics , Immunohistochemistry , Retina/metabolism , Signal Transduction/physiology , Smad1 Protein/metabolism , Zebrafish , Zebrafish Proteins/genetics , p38 Mitogen-Activated Protein Kinases/physiology
18.
Mol Vis ; 19: 1082-95, 2013.
Article in English | MEDLINE | ID: mdl-23734077

ABSTRACT

PURPOSE: Longitudinal observation of retinal degeneration and regeneration in animal models is time-consuming and expensive. To address this challenge, we used a custom fundus lens and zebrafish transgenic lines with cell-specific fluorescent reporters to document the state of individual retinal neurons in vivo. METHODS: We empirically tested several versions of a custom fundus lens and assessed its capabilities under a stereomicroscope to image retinal neurons in transgenic zebrafish lines expressing fluorescent reporters. Vascular branch points provided spatial references enabling determination of whether changes induced by ablating photoreceptors were repaired over the course of several days. RESULTS: Individual ultraviolet- and blue-sensitive cone photoreceptors were readily visualized in vivo, and green fluorescent protein-labeled blood vessels were used as landmarks to facilitate orientation. Sequential imaging of the same retinal areas over several weeks permitted documentation of photoreceptor reappearance in individual animals. Photoreceptor regeneration in these regions was evidenced by the reappearance of individual fluorescent cells. CONCLUSIONS: This technique permits real-time in vivo serial examination of individual fish, permitting temporal analysis of changes to the retinal mosaic. The key benefits this technique offers include that the same retinal locations can be recovered and viewed at multiple time points, that in vivo observations are comparable to those made ex vivo, and that fewer animals need to be euthanized over the course of an experiment. Our results promise the ability to detect individual cells, including reappearing cone photoreceptors, and to monitor disease progression during screening of therapies in an adult animal model of late onset disease.


Subject(s)
Fundus Oculi , Lens, Crystalline/pathology , Lens, Crystalline/physiopathology , Optical Imaging , Regeneration , Retinal Degeneration/physiopathology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Fluorescence , Genes, Reporter , Green Fluorescent Proteins/metabolism , Mosaicism , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Ultraviolet Rays , Zebrafish/genetics
19.
Invest Ophthalmol Vis Sci ; 54(7): 4636-47, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23737474

ABSTRACT

PURPOSE: Vision is critically dependent on ocular size, which is regulated by environmental and genetic factors. Mutation of human Growth and Differentiation Factor 6 (GDF6) or zebrafish gdf6a results in a spectrum of small eye phenotypes (microphthalmia, anophthalmia, and coloboma). However, current models do not explain their etiology fully. As such, analyses of apoptosis and cell cycle regulation were undertaken in a zebrafish gdf6a mutant. METHODS: Microarray analysis was performed at 2 days after fertilization to uncover novel gdf6a-dependent cell cycle regulators. Altered expression of Gdf6a targets was confirmed by in situ hybridization, and resulting changes in cell proliferation were assessed by phosphohistone H3 immunohistochemistry. Analysis of apoptosis was evaluated through activated Caspase 3 immunohistochemistry and chemical inhibitors of cell death. RESULTS: Reduced numbers of retinal progenitor cells are observed at 24 hours post fertilization (hpf), resulting in microphthalmic eyes in gdf6a(-/-) embryos. At 28 hpf, a wave of apoptosis occurs; however, apoptosis inhibition does not rescue eye size, indicating a limited contribution. Mutants display altered proliferation and expression levels of cell cycle regulators, including members of the forkhead box i (foxi) transcription factor family expressed in the ciliary marginal zone. Notably, inhibition of foxi2 in gdf6a(-/-) embryos further reduces eye size. CONCLUSIONS: These data support a model whereby the gdf6a(-/-)-induced microphthalmia is based on early regulation of retinal progenitor cell number, and later by regulation of proliferation in the ciliary marginal zone. Foxi genes represent downstream effectors of Gdf6a function in the CMZ required for eye size determination.


Subject(s)
Apoptosis/physiology , Bone Morphogenetic Proteins/physiology , Cell Proliferation , Microphthalmos/pathology , Animals , Cell Cycle/genetics , Disease Models, Animal , Embryo, Nonmammalian , Eye/embryology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Growth Differentiation Factor 6/metabolism , Immunohistochemistry , Microarray Analysis , Microphthalmos/genetics , Microphthalmos/physiopathology , Stem Cells/pathology , Zebrafish/embryology , Zebrafish Proteins/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...