Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Am J Med Genet A ; 191(4): 1007-1012, 2023 04.
Article in English | MEDLINE | ID: mdl-36595661

ABSTRACT

The purpose of this article is to determine the cause of Leber congenital amaurosis (LCA) in Chuuk state, Federated States of Micronesia (FSM). In this prospective observational case series, five patients with early-onset vision loss were examined in Chuuk state, FSM, during an ocular genetics visit to study the elevated incidence of microphthalmia. Because of their low vision these patients were incorrectly assumed to have microphthalmia. A complete ophthalmological exam established a clinical diagnosis of LCA. Candidate gene exons were sequenced with a targeted retinal dystrophy panel. Five subjects in three related families were diagnosed with LCA. All five were from Tonoas Island, within the Chuuk Lagoon, with ages ranging from 6 months to 16 years. DNA sequencing of affected individuals revealed a homozygous CRB1 NM_201253.3:c.3134del pathogenic variant, which was heterozygous in their parents. CRB1 genotypes were confirmed by a PCR restriction assay. We report identification of a founder pathogenic variant in CRB1 responsible for autosomal recessive LCA in this isolated community. This discovery will lead to appropriate recurrence risk counseling.


Subject(s)
Leber Congenital Amaurosis , Microphthalmos , Humans , Leber Congenital Amaurosis/genetics , Mutation , Genotype , Eye , Pedigree , Eye Proteins/genetics , DNA Mutational Analysis , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics
2.
Hum Mol Genet ; 30(17): 1591-1606, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34046667

ABSTRACT

The forkhead transcription factor FOXE3 is critical for vertebrate eye development. Recessive and dominant variants cause human ocular disease but the full range of phenotypes and mechanisms of action for the two classes of variants are unknown. We identified FOXE3 variants in individuals with congenital eye malformations and carried out in vitro functional analysis on selected alleles. Sixteen new recessive and dominant families, including six novel variants, were identified. Analysis of new and previously reported genetic and clinical data demonstrated a broad phenotypic range with an overlap between recessive and dominant disease. Most families with recessive alleles, composed of truncating and forkhead-domain missense variants, had severe corneal opacity (90%; sclerocornea in 47%), aphakia (83%) and microphthalmia (80%), but some had milder features including isolated cataract. The phenotype was most variable for recessive missense variants, suggesting that the functional consequences may be highly dependent on the type of amino acid substitution and its position. When assessed, aniridia or iris hypoplasia were noted in 89% and optic nerve anomalies in 60% of recessive cases, indicating that these defects are also common and may be underrecognized. In dominant pedigrees, caused by extension variants, normal eye size (96%), cataracts (99%) and variable anterior segment anomalies were seen in most, but some individuals had microphthalmia, aphakia or sclerocornea, more typical of recessive disease. Functional studies identified variable effects on the protein stability, DNA binding, nuclear localization and transcriptional activity for recessive FOXE3 variants, whereas dominant alleles showed severe impairment in all areas and dominant-negative characteristics.


Subject(s)
Eye Abnormalities/genetics , Eye/embryology , Forkhead Transcription Factors/genetics , Adolescent , Alleles , Cataract/genetics , Child , Corneal Opacity/genetics , Developmental Disabilities/genetics , Eye/growth & development , Eye Abnormalities/enzymology , Female , Forkhead Transcription Factors/metabolism , Humans , Male , Mutation , Pedigree , Phenotype
3.
Sci Adv ; 7(11)2021 03.
Article in English | MEDLINE | ID: mdl-33712461

ABSTRACT

Retinal ganglion cells (RGCs) relay visual information from the eye to the brain. RGCs are the first cell type generated during retinal neurogenesis. Loss of function of the transcription factor Atoh7, expressed in multipotent early neurogenic retinal progenitors leads to a selective and essentially complete loss of RGCs. Therefore, Atoh7 is considered essential for conferring competence on progenitors to generate RGCs. Despite the importance of Atoh7 in RGC specification, we find that inhibiting apoptosis in Atoh7-deficient mice by loss of function of Bax only modestly reduces RGC numbers. Single-cell RNA sequencing of Atoh7;Bax-deficient retinas shows that RGC differentiation is delayed but that the gene expression profile of RGC precursors is grossly normal. Atoh7;Bax-deficient RGCs eventually mature, fire action potentials, and incorporate into retinal circuitry but exhibit severe axonal guidance defects. This study reveals an essential role for Atoh7 in RGC survival and demonstrates Atoh7-dependent and Atoh7-independent mechanisms for RGC specification.

4.
Dev Dyn ; 250(6): 807-821, 2021 06.
Article in English | MEDLINE | ID: mdl-32864847

ABSTRACT

BACKGROUND: Vertebrate eye formation requires coordinated inductive interactions between different embryonic tissue layers, first described in amphibians. A network of transcription factors and signaling molecules controls these steps, with mutations causing severe ocular, neuronal, and craniofacial defects. In eyeless mutant axolotls, eye morphogenesis arrests at the optic vesicle stage, before lens induction, and development of ventral forebrain structures is disrupted. RESULTS: We identified a 5-bp deletion in the rax (retina and anterior neural fold homeobox) gene, which was tightly linked to the recessive eyeless (e) axolotl locus in an F2 cross. This frameshift mutation, in exon 2, truncates RAX protein within the homeodomain (P154fs35X). Quantitative RNA analysis shows that mutant and wild-type rax transcripts are equally abundant in E/e embryos. Translation appears to initiate from dual start codons, via leaky ribosome scanning, a conserved feature among gnathostome RAX proteins. Previous data show rax is expressed in the optic vesicle and diencephalon, deeply conserved among metazoans, and required for eye formation in other species. CONCLUSION: The eyeless axolotl mutation is a null allele in the rax homeobox gene, with primary defects in neural ectoderm, including the retinal and hypothalamic primordia.


Subject(s)
Ambystoma mexicanum/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , Ambystoma mexicanum/metabolism , Animals , Embryonic Development/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 117(35): 21690-21700, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817515

ABSTRACT

The retinal ganglion cell (RGC) competence factor ATOH7 is dynamically expressed during retinal histogenesis. ATOH7 transcription is controlled by a promoter-adjacent primary enhancer and a remote shadow enhancer (SE). Deletion of the ATOH7 human SE causes nonsyndromic congenital retinal nonattachment (NCRNA) disease, characterized by optic nerve aplasia and total blindness. We used genome editing to model NCRNA in mice. Deletion of the murine SE reduces Atoh7 messenger RNA (mRNA) fivefold but does not recapitulate optic nerve loss; however, SEdel/knockout (KO) trans heterozygotes have thin optic nerves. By analyzing Atoh7 mRNA and protein levels, RGC development and survival, and chromatin landscape effects, we show that the SE ensures robust Atoh7 transcriptional output. Combining SE deletion and KO and wild-type alleles in a genotypic series, we determined the amount of Atoh7 needed to produce a normal complement of adult RGCs, and the secondary consequences of graded reductions in Atoh7 dosage. Together, these data reveal the workings of an evolutionary fail-safe, a duplicate enhancer mechanism that is hard-wired in the machinery of vertebrate retinal ganglion cell genesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Optic Nerve/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Retina/metabolism , Transcription Factors/metabolism
6.
Hum Genomics ; 13(1): 10, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30770771

ABSTRACT

BACKGROUND: Despite a number of different transgenes that can mediate DNA deletion in the developing lens, each has unique features that can make a given transgenic line more or less appropriate for particular studies. The purpose of this work encompasses both a review of transgenes that lead to the expression of Cre recombinase in the lens and a comparative analysis of currently available transgenic lines with a particular emphasis on the Le-Cre and P0-3.9GFPCre lines that can mediate DNA deletion in the lens placode. Although both of these transgenes are driven by elements of the Pax6 P0 promoter, the Le-Cre transgene consistently leads to ocular abnormalities in homozygous state and can lead to ocular defects on some genetic backgrounds when hemizygous. RESULT: Although both P0-3.9GFPCre and Le-Cre hemizygous transgenic mice undergo normal eye development on an FVB/N genetic background, Le-Cre homozygotes uniquely exhibit microphthalmia. Examination of the expression patterns of these two transgenes revealed similar expression in the developing eye and pancreas. However, lineage tracing revealed widespread non-ocular CRE reporter gene expression in the P0-3.9GFPCre transgenic mice that results from stochastic CRE expression in the P0-3.9GFPCre embryos prior to lens placode formation. Postnatal hemizygous Le-Cre transgenic lenses express higher levels of CRE transcript and protein than the hemizygous lenses of P0-3.9GFPCre mice. Transcriptome analysis revealed that Le-Cre hemizygous lenses deregulated the expression of 15 murine genes, several of which are associated with apoptosis. In contrast, P0-3.9GFPCre hemizygous lenses only deregulated two murine genes. No known PAX6-responsive genes or genes directly associated with lens differentiation were deregulated in the hemizygous Le-Cre lenses. CONCLUSIONS: Although P0-3.9GFPCre transgenic mice appear free from ocular abnormalities, extensive non-ocular CRE expression represents a potential problem for conditional gene deletion studies using this transgene. The higher level of CRE expression in Le-Cre lenses versus P0-3.9GFPCre lenses may explain abnormal lens development in homozygous Le-Cre mice. Given the lack of deregulation of PAX6-responsive transcripts, we suggest that abnormal eye development in Le-Cre transgenic mice stems from CRE toxicity. Our studies reinforce the requirement for appropriate CRE-only expressing controls when using CRE as a driver of conditional gene targeting strategies.


Subject(s)
Gene Deletion , Integrases/genetics , Lens, Crystalline/physiology , Mice, Transgenic , Animals , Female , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Lens, Crystalline/embryology , Lens, Crystalline/physiopathology , Mice, Inbred Strains
7.
Cell Rep ; 23(9): 2643-2652, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847795

ABSTRACT

Maternally skewed transmission of traits has been associated with genomic imprinting and oocyte-derived mRNA. We report canine congenital eye malformations, caused by an amino acid deletion (K12del) near the N terminus of retinol-binding protein (RBP4). The disease is only expressed when both dam and offspring are deletion homozygotes. RBP carries vitamin A (retinol) from hepatic stores to peripheral tissues, including the placenta and developing eye, where it is required to synthesize retinoic acid. Gestational vitamin A deficiency is a known risk factor for ocular birth defects. The K12del mutation disrupts RBP folding in vivo, decreasing its secretion from hepatocytes to serum. The maternal penetrance effect arises from an impairment in the sequential transfer of retinol across the placenta, via RBP encoded by maternal and fetal genomes. Our results demonstrate a mode of recessive maternal inheritance, with a physiological basis, and they extend previous observations on dominant-negative RBP4 alleles in humans.


Subject(s)
Dogs/genetics , Eye Diseases/congenital , Eye Diseases/veterinary , Genes, Recessive , Maternal Inheritance/genetics , Retinol-Binding Proteins, Plasma/genetics , Amino Acid Sequence , Animals , Base Pairing/genetics , Eye Diseases/blood , Eye Diseases/genetics , Female , Genetic Loci , Genotype , HeLa Cells , Humans , Male , Microphthalmos/blood , Microphthalmos/genetics , Pedigree , Phenotype , Prealbumin/metabolism , Protein Folding , Retinol-Binding Proteins, Plasma/chemistry , Sequence Deletion , Vitamin A/blood
8.
Gene Expr Patterns ; 27: 114-121, 2018 01.
Article in English | MEDLINE | ID: mdl-29225067

ABSTRACT

The Atoh7 transcription factor catalyzes the rate-limiting step in the specification of retinal ganglion cells (RGCs). As a tool to study vertebrate retinal development, we validate an antibody that recognizes human and mouse Atoh7 polypeptide, using informative knockout and transgenic mouse tissues and overexpression experiments. The transient features of Atoh7 protein expression during retinal neurogenesis match the expected pattern at the tissue and cellular level. Further, we compare endogenous Atoh7 to established RGC markers, reporter mouse lines and cell cycle markers, demonstrating the utility of the antibody to investigate molecular mechanisms of retinal histogenesis.


Subject(s)
Antibodies, Monoclonal/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/metabolism , Neurogenesis , Retina/metabolism , Retinal Ganglion Cells/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Cells, Cultured , Embryo, Mammalian/cytology , HEK293 Cells , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Retina/cytology , Retinal Ganglion Cells/cytology , Sequence Homology
9.
Glia ; 65(10): 1697-1716, 2017 10.
Article in English | MEDLINE | ID: mdl-28722174

ABSTRACT

Immature astrocytes and blood vessels enter the developing mammalian retina at the optic nerve head and migrate peripherally to colonize the entire retinal nerve fiber layer (RNFL). Retinal vascularization is arrested in retinopathy of prematurity (ROP), a major cause of bilateral blindness in children. Despite their importance in normal development and ROP, the factors that control vascularization of the retina remain poorly understood. Because astrocytes form a reticular network that appears to provide a substrate for migrating endothelial cells, they have long been proposed to guide angiogenesis. However, whether astrocytes do in fact impose a spatial pattern on developing vessels remains unclear, and how astrocytes themselves are guided is unknown. Here we explore the cellular mechanisms that ensure complete retinal coverage by astrocytes and blood vessels in mouse. We find that migrating astrocytes associate closely with the axons of retinal ganglion cells (RGCs), their neighbors in the RNFL. Analysis of Robo1; Robo2 mutants, in which RGC axon guidance is disrupted, and Math5 (Atoh7) mutants, which lack RGCs, reveals that RGCs provide directional information to migrating astrocytes that sets them on a centrifugal trajectory. Without this guidance, astrocytes exhibit polarization defects, fail to colonize the peripheral retina, and display abnormal fine-scale spatial patterning. Furthermore, using cell type-specific chemical-genetic tools to selectively ablate astrocytes, we show that the astrocyte template is required for angiogenesis and vessel patterning. Our results are consistent with a model whereby RGC axons guide formation of an astrocytic network that subsequently directs vessel development.


Subject(s)
Astrocytes/physiology , Axons/physiology , Neovascularization, Physiologic/physiology , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Diphtheria Toxin/pharmacology , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic , Mutation/genetics , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Retinal Ganglion Cells/cytology , Homeobox Protein SIX3
10.
Cell ; 161(3): 634-646, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25910211

ABSTRACT

Gestational vitamin A (retinol) deficiency poses a risk for ocular birth defects and blindness. We identified missense mutations in RBP4, encoding serum retinol binding protein, in three families with eye malformations of differing severity, including bilateral anophthalmia. The mutant phenotypes exhibit dominant inheritance, but incomplete penetrance. Maternal transmission significantly increases the probability of phenotypic expression. RBP normally delivers retinol from hepatic stores to peripheral tissues, including the placenta and fetal eye. The disease mutations greatly reduce retinol binding to RBP, yet paradoxically increase the affinity of RBP for its cell surface receptor, STRA6. By occupying STRA6 nonproductively, the dominant-negative proteins disrupt vitamin A delivery from wild-type proteins within the fetus, but also, in the case of maternal transmission, at the placenta. These findings establish a previously uncharacterized mode of maternal inheritance, distinct from imprinting and oocyte-derived mRNA, and define a group of hereditary disorders plausibly modulated by dietary vitamin A.


Subject(s)
Eye Diseases, Hereditary/genetics , Mutation, Missense , Retinol-Binding Proteins, Plasma/genetics , Amino Acid Sequence , Animals , DNA Mutational Analysis , Female , Genes, Dominant , Humans , Male , Maternal-Fetal Exchange , Molecular Sequence Data , Pedigree , Penetrance , Pregnancy , Retinol-Binding Proteins, Plasma/chemistry , Sequence Alignment , Vitamin A Deficiency/metabolism
11.
Mol Cell Neurosci ; 54: 108-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23481413

ABSTRACT

Retinal neurons and glia arise from a common progenitor pool in a temporal order, with retinal ganglion cells (RGCs) appearing first, and Müller glia last. The transcription factors Atoh7/Math5 and Ascl1/Mash1 represent divergent bHLH clades, and exhibit distinct spatial and temporal retinal expression patterns, with little overlap during early development. Here, we tested the ability of Ascl1 to change the fate of cells in the Atoh7 lineage when misexpressed from the Atoh7 locus, using an Ascl1-IRES-DsRed2 knock-in allele. In Atoh7(Ascl1KI/+) and Atoh7(Ascl1KI/Ascl1KI) embryos, ectopic Ascl1 delayed cell cycle exit and differentiation, even in cells coexpressing Atoh7. The heterozygous retinas recovered, and eventually produced a normal complement of RGCs, while homozygous substitution of Ascl1 for Atoh7 did not promote postnatal retinal fates precociously, nor rescue Atoh7 mutant phenotypes. However, our analyses revealed two unexpected findings. First, ectopic Ascl1 disrupted cell cycle progression within the marked Atoh7 lineage, but also nonautonomously in other retinal cells. Second, the size of the Atoh7 retinal lineage was unaffected, supporting the idea of a compensatory shift of the non-proliferative cohort to maintain lineage size. Overall, we conclude that Ascl1 acts dominantly to block cell cycle exit, but is incapable of redirecting the fates of early RPCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Lineage , Nerve Tissue Proteins/metabolism , Retina/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Division , Gene Expression Regulation, Developmental , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neuroglia/cytology , Neuroglia/metabolism , Phenotype , Retina/growth & development , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
12.
Hum Mol Genet ; 22(4): 696-703, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23161670

ABSTRACT

Focal facial dermal dysplasia (FFDD) Type IV is a rare syndrome characterized by facial lesions resembling aplasia cutis in a preauricular distribution along the line of fusion of the maxillary and mandibular prominences. To identify the causative gene(s), exome sequencing was performed in a family with two affected siblings. Assuming autosomal recessive inheritance, two novel sequence variants were identified in both siblings in CYP26C1-a duplication of seven base pairs, which was maternally inherited, c.844_851dupCCATGCA, predicting p.Glu284fsX128 and a missense mutation, c.1433G>A, predicting p.Arg478His, that was paternally inherited. The duplication predicted a frameshift mutation that led to a premature stop codon and premature chain termination, whereas the missense mutation was not functional based on its in vitro expression in mammalian cells. The FFDD skin lesions arise along the sites of fusion of the maxillary and mandibular prominences early in facial development, and Cyp26c1 was expressed exactly along the fusion line for these facial prominences in the first branchial arch in mice. Sequencing of four additional, unrelated Type IV FFDD patients and eight Type II or III TWIST2-negative FFDD patients revealed that three of the Type IV patients were homozygous for the duplication, whereas none of the Type II or III patients had CYP26C1 mutations. The seven base pairs duplication was present in 0.3% of healthy controls and 0.3% of patients with other birth defects. These findings suggest that the phenotypic manifestations of FFDD Type IV can be non-penetrant or underascertained. Thus, FFDD Type IV results from the loss of function mutations in CYP26C1.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Ectodermal Dysplasia/genetics , Mutation, Missense , Animals , COS Cells , Chlorocebus aethiops , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 26 , DNA Mutational Analysis , Ectodermal Dysplasia/enzymology , Focal Facial Dermal Dysplasias , Frameshift Mutation , Genetic Association Studies , Humans , Mice , Microsatellite Repeats
13.
Mol Vis ; 18: 1740-9, 2012.
Article in English | MEDLINE | ID: mdl-22815627

ABSTRACT

PURPOSE: The migratory neural crest cell population makes a significant contribution to the anterior segment structures of the eye. Consequently, several anterior segment dysgenesis phenotypes are associated with mutations in genes expressed during neural crest development. The forkhead box D3 (FOXD3) gene encodes a forkhead transcription factor that plays an important role in neural crest specification in vertebrates and therefore may be involved in human eye disease. METHODS: We screened 310 probands with developmental ocular conditions for variations in FOXD3. RESULTS: Six nonsynonymous FOXD3 variants were identified. Four of these changes, c.47C>T (p.Thr16Met), c.359C>T (p.Pro120Leu), c.517A>C (p.Asn173His), and c.818_829dup (p.Arg273_Gly276dup), affected conserved regions and were observed primarily in probands with aniridia or Peters anomaly; out of these four variants, one, p.Arg273_Gly276dup, was not detected in control populations and two, p.Pro120Leu and p.Asn173His, were statistically enriched in cases with aniridia or Peters anomaly. The p.Arg273_Gly276dup variant was seen in a proband with aniridia as well as two additional unrelated probands affected with anophthalmia or congenital cataracts. The p.Asn173His variant affects Helix 2 of the DNA-binding domain and was observed in two unrelated patients with Peters anomaly or aniridia; in both cases, one parent carried the same allele. CONCLUSIONS: FOXD3 variants increase the risk of anterior segment dysgenesis phenotypes in humans. The p.Asn173His mutation affects a residue in the forkhead domain that is 100% conserved among vertebrate orthologs and is predicted to participate in protein-protein interactions. Its phenotypic effects may be modulated by transcriptional cofactors which have yet to be identified.


Subject(s)
Aniridia/genetics , Anophthalmos/genetics , Cataract/genetics , Corneal Opacity/genetics , Eye Abnormalities/genetics , Forkhead Transcription Factors/genetics , Glaucoma/genetics , Alleles , Amino Acid Sequence , Animals , Anterior Eye Segment/abnormalities , Cataract/congenital , Conserved Sequence , DNA Mutational Analysis , Gene Frequency , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Molecular Sequence Data , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Risk Factors
14.
Neuroreport ; 23(10): 631-4, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22660169

ABSTRACT

The basic helix-loop-helix factor Math5 (Atoh7) is critical for the determination of retinal ganglion cell (RGC) fate in mice. Recently, genome-wide association studies have identified the ATOH7 locus as a major determinant of variation in the human optic disc area, which is directly correlated with the RGC number. These studies suggest that the level of Math5 expression may determine the ultimate number of RGCs. To test this hypothesis, we systematically compared optic nerve area and RGC axon number in C57BL/6J congenic Math5+/- and +/+ mice at young adult and neonatal ages by transmission electron microscopy. Optic disc area and RGC abundance were not significantly different in adults, but heterozygotes had thinner optic nerves and 25-30% fewer RGCs at birth than wild-type littermates (P<0.05). Our results suggest that Math5 dosage is important for the genesis, but not the ultimate number, of RGCs. Our findings highlight the importance of ganglion cell culling as a compensatory mechanism for retinal homeostasis, and support a quantitative role for Math5 in RGC specification.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Dosage/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Retinal Ganglion Cells/physiology , Animals , Mice , Mice, 129 Strain , Mice, Congenic , Mice, Inbred C57BL
15.
Hum Mol Genet ; 21(16): 3681-94, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22645276

ABSTRACT

The vertebrate basic helix-loop-helix (bHLH) transcription factor ATOH7 (Math5) is specifically expressed in the embryonic neural retina and is required for the genesis of retinal ganglion cells (RGCs) and optic nerves. In Atoh7 mutant mice, the absence of trophic factors secreted by RGCs prevents the development of the intrinsic retinal vasculature and the regression of fetal blood vessels, causing persistent hyperplasia of the primary vitreous (PHPV). We therefore screened patients with hereditary PHPV, as well as bilateral optic nerve aplasia (ONA) or hypoplasia (ONH), for mutations in ATOH7. We identified a homozygous ATOH7 mutation (N46H) in a large family with an autosomal recessive PHPV disease trait linked to 10q21, and a heterozygous variant (R65G, p.Arg65Gly) in one of five sporadic ONA patients. High-density single-nucleotide polymorphism analysis also revealed a CNTN4 duplication and an OTX2 deletion in the ONA cohort. Functional analysis of ATOH7 bHLH domain substitutions, by electrophoretic mobility shift and luciferase cotransfection assays, revealed that the N46H variant cannot bind DNA or activate transcription, consistent with structural modeling. The N46H variant also failed to rescue RGC development in mouse Atoh7-/- retinal explants. The R65G variant retains all of these activities, similar to wild-type human ATOH7. Our results strongly suggest that autosomal recessive persistent hyperplastic primary vitreous is caused by N46H and is etiologically related to nonsyndromic congenital retinal nonattachment. The R65G allele, however, cannot explain the ONA phenotype. Our study firmly establishes ATOH7 as a retinal disease gene and provides a functional basis to analyze new coding variants.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hyperplasia/genetics , Retinal Diseases/genetics , Vitreous Body/pathology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromosomes, Human, Pair 10 , Female , Genes, Recessive , Helix-Loop-Helix Motifs/genetics , Humans , Infant , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Optic Nerve/abnormalities , Optic Nerve/pathology , Organ Culture Techniques/methods , Pedigree , Retinal Diseases/pathology
16.
Dev Biol ; 368(2): 214-30, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22609278

ABSTRACT

The basic-helix-loop helix factor Math5 (Atoh7) is required for retinal ganglion cell (RGC) development. However, only 10% of Math5-expressing cells adopt the RGC fate, and most become photoreceptors. In principle, Math5 may actively bias progenitors towards RGC fate or passively confer competence to respond to instructive factors. To distinguish these mechanisms, we misexpressed Math5 in a wide population of precursors using a Crx BAC or 2.4 kb promoter, and followed cell fates with Cre recombinase. In mice, the Crx cone-rod homeobox gene and Math5 are expressed shortly after cell cycle exit, in temporally distinct, but overlapping populations of neurogenic cells that give rise to 85% and 3% of the adult retina, respectively. The Crx>Math5 transgenes did not stimulate RGC fate or alter the timing of RGC births. Likewise, retroviral Math5 overexpression in retinal explants did not bias progenitors towards the RGC fate or induce cell cycle exit. The Crx>Math5 transgene did reduce the abundance of early-born (E15.5) photoreceptors two-fold, suggesting a limited cell fate shift. Nonetheless, retinal histology was grossly normal, despite widespread persistent Math5 expression. In an RGC-deficient (Math5 knockout) environment, Crx>Math5 partially rescued RGC and optic nerve development, but the temporal envelope of RGC births was not extended. The number of early-born RGCs (before E13) remained very low, and this was correlated with axon pathfinding defects and cell death. Together, these results suggest that Math5 is not sufficient to stimulate RGC fate. Our findings highlight the robust homeostatic mechanisms, and role of pioneering neurons in RGC development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Mammalian/metabolism , Nerve Tissue Proteins/metabolism , Retinal Ganglion Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Retinal Ganglion Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Mol Cell Neurosci ; 50(2): 160-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22579728

ABSTRACT

The vertebrate neural retina contains seven major cell types, which arise from a common multipotent progenitor pool. During neurogenesis, these cells stop cycling, commit to a single fate, and differentiate. The mechanism and order of these steps remain unclear. The first-born type of retinal neurons, ganglion cells (RGCs), develop through the actions of Math5 (Atoh7), Brn3b (Pou4f2) and Islet1 (Isl1) factors, whereas inhibitory amacrine and horizontal precursors require Ptf1a for differentiation. We have examined the link between these markers, and the timing of their expression during the terminal cell cycle, by nucleoside pulse-chase analysis in the mouse retina. We show that G2 phase lasts 1-2 h at embryonic (E) 13.5 and E15.5 stages. Surprisingly, we found that cells expressing Brn3b and/or Isl1 were frequently co-labeled with EdU after a short chase (<1 h) in early embryos (E15), Brn3b and Isl1 were exclusively expressed in post-mitotic cells, even as new RGCs are still generated. In contrast, Ptf1a and amacrine marker AP2α were detected only after terminal mitosis, at all developmental stages. Using a retroviral tracer in embryonic retinal explants (E12-E13), we identified two-cell clones containing paired ganglion cells, consistent with RGC fate commitment prior to terminal mitosis. Thus, although cell cycle exit and fate determination are temporally correlated during retinal neurogenesis, the order of these events varies according to developmental stage and final cell type.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Retinal Neurons/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Biomarkers/metabolism , Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/biosynthesis , Mice , Nerve Tissue Proteins/biosynthesis , Retina/embryology , Retina/physiology , Retinal Neurons/cytology , Retinal Neurons/physiology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factor Brn-3B/biosynthesis , Transcription Factors/biosynthesis
18.
Dev Biol ; 365(2): 395-413, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22445509

ABSTRACT

The basic helix-loop-helix (bHLH) transcription factor Math5 (Atoh7) is transiently expressed during early retinal histogenesis and is necessary for retinal ganglion cell (RGC) development. Using nucleoside pulse-chase experiments and clonal analysis, we determined that progenitor cells activate Math5 during or after the terminal division, with progressively later onset as histogenesis proceeds. We have traced the lineage of Math5+ cells using mouse BAC transgenes that express Cre recombinase under strict regulatory control. Quantitative analysis showed that Math5+ progenitors express equivalent levels of Math5 and contribute to every major cell type in the adult retina, but are heavily skewed toward early fates. The Math5>Cre transgene labels 3% of cells in adult retina, including 55% of RGCs. Only 11% of Math5+ progenitors develop into RGCs; the majority become photoreceptors. The fate bias of the Math5 cohort, inferred from the ratio of cone and rod births, changes over time, in parallel with the remaining neurogenic population. Comparable results were obtained using Math5 mutant mice, except that ganglion cells were essentially absent, and late fates were overrepresented within the lineage. We identified Math5-independent RGC precursors in the earliest born (embryonic day 11) retinal cohort, but these precursors require Math5-expressing cells for differentiation. Math5 thus acts permissively to establish RGC competence within a subset of progenitors, but is not sufficient for fate specification. It does not autonomously promote or suppress the determination of non-RGC fates. These data are consistent with progressive and temporal restriction models for retinal neurogenesis, in which environmental factors influence the final histotypic choice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Retina/embryology , Retinal Ganglion Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Retinal Ganglion Cells/metabolism
19.
Nat Neurosci ; 14(5): 578-86, 2011 May.
Article in English | MEDLINE | ID: mdl-21441919

ABSTRACT

Individuals with nonsyndromic congenital retinal nonattachment (NCRNA) are totally blind from birth. The disease afflicts ∼1% of Kurdish people living in a group of neighboring villages in North Khorasan, Iran. We found that NCRNA is caused by a 6,523-bp deletion that spans a remote cis regulatory element 20 kb upstream from ATOH7 (Math5), a bHLH transcription factor gene that is required for retinal ganglion cell (RGC) and optic nerve development. In humans, the absence of RGCs stimulates massive neovascular growth of fetal blood vessels in the vitreous and early retinal detachment. The remote ATOH7 element appears to act as a secondary or 'shadow' transcriptional enhancer. It has minimal sequence similarity to the primary enhancer, which is close to the ATOH7 promoter, but drives transgene expression with an identical spatiotemporal pattern in the mouse retina. The human transgene also functions appropriately in zebrafish, reflecting deep evolutionary conservation. These dual enhancers may reinforce ATOH7 expression during early critical stages of eye development when retinal neurogenesis is initiated.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Enhancer Elements, Genetic/genetics , Neurogenesis/genetics , Retina/cytology , Retinal Detachment/genetics , Sequence Deletion/genetics , Adolescent , Animals , Animals, Genetically Modified , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 10 , Computational Biology/methods , Consanguinity , DNA Mutational Analysis , Embryo, Mammalian , Family Health , Female , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Humans , Iran/ethnology , Luminescent Proteins/genetics , Magnetic Resonance Imaging , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide/genetics , Retina/embryology , Retinal Detachment/congenital , Retinal Detachment/pathology , Superior Colliculi/pathology , Zebrafish/growth & development
20.
PLoS One ; 5(8): e12315, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20808762

ABSTRACT

The Math5 (Atoh7) gene is transiently expressed during retinogenesis by progenitors exiting mitosis, and is essential for ganglion cell (RGC) development. Math5 contains a single exon, and its 1.7 kb mRNA encodes a 149-aa polypeptide. Mouse Math5 mutants have essentially no RGCs or optic nerves. Given the importance of this gene in retinal development, we thoroughly investigated the possibility of Math5 mRNA splicing by Northern blot, 3'RACE, RNase protection assays, and RT-PCR, using RNAs extracted from embryonic eyes and adult cerebellum, or transcribed in vitro from cDNA clones. Because Math5 mRNA contains an elevated G+C content, we used graded concentrations of betaine, an isostabilizing agent that disrupts secondary structure. Although approximately 10% of cerebellar Math5 RNAs are spliced, truncating the polypeptide, our results show few, if any, spliced Math5 transcripts exist in the developing retina (<1%). Rare deleted cDNAs do arise via RT-mediated RNA template switching in vitro, and are selectively amplified during PCR. These data differ starkly from a recent study (Kanadia and Cepko 2010), which concluded that the vast majority of Math5 and other bHLH transcripts are spliced to generate noncoding RNAs. Our findings clarify the architecture of the Math5 gene and its mechanism of action. These results have implications for all members of the bHLH gene family, for any gene that is alternatively spliced, and for the interpretation of all RT-PCR experiments.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/genetics , RNA Splicing , Retina/growth & development , Retina/metabolism , Animals , Base Sequence , Blotting, Northern , Cerebellum/metabolism , Cloning, Molecular , DNA, Complementary/genetics , Mice , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...