Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Gene Expr Patterns ; 50: 119344, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37844855

RESUMEN

Tissue fusion is a critical process that is repeated in multiple contexts during embryonic development and shares common attributes to processes such as wound healing and metastasis. Ocular coloboma is a developmental eye disorder that presents as a physical gap in the ventral eye, and is a major cause of childhood blindness. Coloboma results from fusion failure between opposing ventral retinal epithelia, but there are major knowledge gaps in our understanding of this process at the molecular and cell behavioural levels. Here we catalogue the expression of cell adhesion proteins: N-cadherin, E-cadherin, R-cadherin, ZO-1, and the EMT transcriptional activator and cadherin regulator SNAI2, in the developing chicken embryonic eye. We find that fusion pioneer cells at the edges of the fusing optic fissure have unique and dynamic expression profiles for N-cad, E-cad and ZO-1, and that these are temporally preceded by expression of SNAI2. This highlights the unique properties of these cells and indicates that regulation of cell adhesion factors may be a critical process in optic fissure closure.


Asunto(s)
Coloboma , Retina , Animales , Embrión de Pollo , Femenino , Embarazo , Humanos , Adhesión Celular , Retina/metabolismo , Coloboma/metabolismo , Coloboma/patología , Factores de Transcripción/metabolismo , Pollos/genética , Pollos/metabolismo , Ojo/metabolismo
2.
Biomolecules ; 13(2)2023 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-36830662

RESUMEN

Ocular coloboma (OC) is a failure of complete optic fissure closure during embryonic development and presents as a tissue defect along the proximal-distal axis of the ventral eye. It is classed as part of the clinical spectrum of structural eye malformations with microphthalmia and anophthalmia, collectively abbreviated to MAC. Despite deliberate attempts to identify causative variants in MAC, many patients remain without a genetic diagnosis. To reveal potential candidate genes, we utilised transcriptomes experimentally generated from embryonic eye tissues derived from humans, mice, zebrafish, and chicken at stages coincident with optic fissure closure. Our in-silico analyses found 10 genes with optic fissure-specific enriched expression: ALDH1A3, BMPR1B, EMX2, EPHB3, NID1, NTN1, PAX2, SMOC1, TENM3, and VAX1. In situ hybridization revealed that all 10 genes were broadly expressed ventrally in the developing eye but that only PAX2 and NTN1 were expressed in cells at the edges of the optic fissure margin. Of these conserved optic fissure genes, EMX2, NID1, and EPHB3 have not previously been associated with human MAC cases. Targeted genetic manipulation in zebrafish embryos using CRISPR/Cas9 caused the developmental MAC phenotype for emx2 and ephb3. We analysed available whole genome sequencing datasets from MAC patients and identified a range of variants with plausible causality. In combination, our data suggest that expression of genes involved in ventral eye development is conserved across a range of vertebrate species and that EMX2, NID1, and EPHB3 are candidate loci that warrant further functional analysis in the context of MAC and should be considered for sequencing in cohorts of patients with structural eye malformations.


Asunto(s)
Coloboma , Anomalías del Ojo , Neuropéptidos , Femenino , Embarazo , Humanos , Animales , Ratones , Coloboma/genética , Coloboma/metabolismo , Ojo/metabolismo , Pez Cebra/genética , Perfilación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Neuropéptidos/metabolismo , Proteínas de Homeodominio/metabolismo
3.
Dev Dyn ; 252(4): 495-509, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36576487

RESUMEN

BACKGROUND: RERE is a highly conserved transcriptional co-regulator that is associated with a human neurodevelopmental disorder with or without anomalies of the brain, eye, or heart (NEDBEH, OMIM: 616975). RESULTS: We show that the zebrafish rerea mutant (babyface) robustly recapitulates optic fissure closure defects resulting from loss of RERE function, as observed in humans. These defects result from expansion of proximal retinal optic stalk (OS) and reduced expression of some of the ventral retinal fate genes due to deregulated protein signaling. Using zebrafish and cell-based assays, we determined that NEDBEH-associated human RERE variants function as hypomorphs in their ability to repress shh signaling and some exhibit abnormal nuclear localization. Inhibiting shh signaling by the protein inhibitor HPI-1 rescues coloboma, confirming our observation that coloboma in rerea mutants is indeed due to deregulation of shh signaling. CONCLUSIONS: Zebrafish rerea mutants exhibit OS and optic fissure closure defects. The optic fissure closure defect was rescued by an shh signaling inhibitor, suggesting that this defect could arise due to deregulated shh signaling.


Asunto(s)
Coloboma , Proteínas de Pez Cebra , Pez Cebra , Animales , Humanos , Proteínas Portadoras/metabolismo , Coloboma/genética , Coloboma/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Retina/metabolismo , Transducción de Señal/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Invest Ophthalmol Vis Sci ; 63(12): 5, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36326727

RESUMEN

Purpose: Uveal coloboma is a congenital eye malformation caused by failure of the optic fissure to close in early human development. Despite significant progress in identifying genes whose regulation is important for executing this closure, mutations are detected in a minority of cases using known gene panels, implying additional genetic complexity. We have previously shown knockdown of znf503 (the ortholog of mouse Zfp503) in zebrafish causes coloboma. Here we characterize Zfp503 knockout (KO) mice and evaluate transcriptomic profiling of mutant versus wild-type (WT) retinal pigment epithelium (RPE)/choroid. Methods: Zfp503 KO mice were generated by gene targeting using homologous recombination. Embryos were characterized grossly and histologically. Patterns and level of developmentally relevant proteins/genes were examined with immunostaining/in situ hybridization. The transcriptomic profile of E11.5 KO RPE/choroid was compared to that of WT. Results: Zfp503 is dynamically expressed in developing mouse eyes, and loss of its expression results in uveal coloboma. KO embryos exhibit altered mRNA levels and expression patterns of several key transcription factors involved in eye development, including Otx2, Mitf, Pax6, Pax2, Vax1, and Vax2, resulting in a failure to maintain the presumptive RPE, as evidenced by reduced melanin pigmentation and its differentiation into a neural retina-like lineage. Comparison of RNA sequencing data from WT and KO E11.5 embryos demonstrated reduced expression of melanin-related genes and significant overlap with genes known to be dynamically regulated at the optic fissure. Conclusions: These results demonstrate a critical role of Zfp503 in maintaining RPE fate and optic fissure closure.


Asunto(s)
Coloboma , Neuropéptidos , Animales , Humanos , Ratones , Coloboma/genética , Coloboma/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Melaninas/metabolismo , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuropéptidos/genética , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pez Cebra/genética
5.
Sci Rep ; 11(1): 3111, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542446

RESUMEN

Cat eye syndrome (CES), a human genetic disorder caused by the inverted duplication of a region on chromosome 22, has been known since the late 1890s. Despite the significant impact this disorder has on affected individuals, models for CES have not been produced due to the difficulty of effectively duplicating the corresponding chromosome region in an animal model. However, the study of phenotypes associated with individual genes in this region such as CECR2 may shed light on the etiology of CES. In this study we have shown that deleterious loss of function mutations in mouse Cecr2 effectively demonstrate many of the abnormal features present in human patients with CES, including coloboma and specific skeletal, kidney and heart defects. Beyond phenotypic analyses we have demonstrated the importance of utilizing multiple genetic backgrounds to study disease models, as we see major differences in penetrance of Cecr2-related abnormal phenotype between mouse strains, reminiscent of the variability in the human syndrome. These findings suggest that Cecr2 is involved in the abnormal features of CES and that Cecr2 mice can be used as a model system to study the wide range of phenotypes present in CES.


Asunto(s)
Trastornos de los Cromosomas/genética , Coloboma/genética , Modelos Animales de Enfermedad , Anomalías del Ojo/genética , Cardiopatías/genética , Mutación con Pérdida de Función , Factores de Transcripción/genética , Aneuploidia , Animales , Huesos/metabolismo , Huesos/patología , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/patología , Duplicación Cromosómica , Cromosomas Humanos Par 22/química , Cromosomas Humanos Par 22/genética , Cromosomas Humanos Par 22/metabolismo , Coloboma/metabolismo , Coloboma/patología , Embrión de Mamíferos , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Femenino , Expresión Génica , Cardiopatías/metabolismo , Cardiopatías/patología , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Penetrancia , Especificidad de la Especie , Factores de Transcripción/deficiencia
6.
Hum Mol Genet ; 29(20): 3373-3387, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33075808

RESUMEN

Uveal coloboma represents one of the most common congenital ocular malformations accounting for up to 10% of childhood blindness (~1 in 5000 live birth). Coloboma originates from defective fusion of the optic fissure (OF), a transient gap that forms during eye morphogenesis by asymmetric, ventral invagination. Genetic heterogeneity combined with the activity of developmentally regulated genes suggests multiple mechanisms regulating OF closure. The tumor suppressor and FERM domain protein Neurofibromin 2 (NF2) controls diverse processes in cancer, development and regeneration, via Hippo pathway and cytoskeleton regulation. In humans, NF2 mutations can cause ocular abnormalities, including coloboma, however, its actual role in OF closure is unknown. Using conditional inactivation in the embryonic mouse eye, our data indicate that loss of Nf2 function results in a novel underlying cause for coloboma. In particular, mutant eyes show substantially increased retinal pigmented epithelium (RPE) proliferation in the fissure region with concomitant acquisition of RPE cell fate. Cells lining the OF margin can maintain RPE fate ectopically and fail to transition from neuroepithelial to cuboidal shape. In the dorsal RPE of the optic cup, Nf2 inactivation leads to a robust increase in cell number, with local disorganization of the cytoskeleton components F-actin and pMLC2. We propose that RPE hyperproliferation is the primary cause for the observed defects causing insufficient alignment of the OF margins in Nf2 mutants and failure to fuse properly, resulting in persistent coloboma. Our findings indicate that limiting proliferation particularly in the RPE layer is a critical mechanism during OF closure.


Asunto(s)
Proliferación Celular , Coloboma/patología , Ojo/patología , Regulación del Desarrollo de la Expresión Génica , Neurofibromina 2/fisiología , Organogénesis , Epitelio Pigmentado de la Retina/patología , Animales , Coloboma/etiología , Coloboma/metabolismo , Ojo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epitelio Pigmentado de la Retina/metabolismo
7.
Int J Mol Sci ; 21(8)2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316164

RESUMEN

Colobomata, persistent optic fissures, frequently cause congenital blindness. Here, we focused on optic fissure fusion using in vivo time-lapse imaging in zebrafish. We identified the fusion initiating cells, which we termed "pioneer cells." Based on morphology, localization, and downregulation of the neuroretinal (NR) precursor marker rx2, these cells could be considered as retinal pigment epithelial (RPE) progenitors. Notably, pioneer cells regain rx2 expression and integrate into the NR after fusion, indicating that they do not belong to the pool of RPE progenitors, supported by the lack of RPE marker expression in pioneer cells. They establish the first cellular contact between the margins in the proximal fissure region and separate the hyaloid artery and vein. After initiation, the fusion site is progressing distally, increasing the distance between the hyaloid artery and vein. A timed BMP (Bone Morphogenetic Protein) induction, resulting in coloboma, did not alter the morphology of the fissure margins, but it did affect the expression of NR and RPE markers within the margins. In addition, it resulted in a persisting basal lamina and persisting remnants of periocular mesenchyme and hyaloid vasculature within the fissure, supporting the necessity of BMP antagonism within the fissure margins. The hampered fissure fusion had severe effects on the vasculature of the eye.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Membrana Basal/metabolismo , Vasos Sanguíneos/anatomía & histología , Proteínas Morfogenéticas Óseas/genética , Coloboma/metabolismo , Coloboma/patología , Disco Óptico/anomalías , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Imagen de Lapso de Tiempo , Proteínas de Pez Cebra/genética
8.
Clin Genet ; 96(4): 371-375, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31343737

RESUMEN

Retinoid acid receptors (RAR) are transcription factors that bind retinoic acid (RA), a metabolite of vitamin A. RARs are composed of three subunits encoded by RARA, RARB and RARG. In humans, RARB defects cause syndromic microphthalmia. So far, no germline pathogenic variants have been identified in RARA or RARG. We describe a girl with a de novo mutation NM_000964 c.826C > T (p.Arg276Trp) in RARA with symptoms overlapping those described in RARB patients (coloboma, muscular hypotonia, dilated pulmonary artery, ectopic kidney). RARA Arg276 residue is functionally important, as it was previously shown that its substitution for Ala or Gln causes a 50- or 21-fold impairment of RA binding, respectively. Moreover, in leukemic cells, the p.Arg611Trp mutation in a chimeric PML/RARA gene (corresponding to the RARA p.Arg276Trp detected in our patient) conferred resistance to therapy by decreasing binding of all-trans RA. The functional effect of RARA p.Arg276Trp was further confirmed by in silico modeling which showed that binding of RA by the Trp276 variant was similarly defective as in the deleterious model Ala276 mutant. We propose that RARA p.Arg276Trp causes the disease by affecting RA interaction with the RARA receptor.


Asunto(s)
Aminoácidos/metabolismo , Coloboma/genética , Coloboma/metabolismo , Heterocigoto , Mutación , Receptor alfa de Ácido Retinoico/genética , Tretinoina/metabolismo , Niño , Coloboma/diagnóstico , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Modelos Moleculares , Linaje , Fenotipo , Receptor alfa de Ácido Retinoico/química , Relación Estructura-Actividad
9.
Open Biol ; 9(2): 180179, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30958096

RESUMEN

Optic cup morphogenesis is an intricate process. Especially, the formation of the optic fissure is not well understood. Persisting optic fissures, termed coloboma, are frequent causes for congenital blindness. Even though the defective fusion of the fissure margins is the most acknowledged reason for coloboma, highly variable morphologies of coloboma phenotypes argue for a diverse set of underlying pathomechanisms. Here, we investigate optic fissure morphogenesis in zebrafish to identify potential morphogenetic defects resulting in coloboma. We show that the formation of the optic fissure depends on tissue flow movements, integrated into the bilateral distal epithelial flow forming the optic cup. On the temporal side, the distal flow translates into a ventral perpendicular flow, shaping the temporal fissure margin. On the nasal side, however, the distal flow is complemented by tissue derived from the optic stalk, shaping the nasal fissure margin. Notably, a distinct population of TGFß-signalling positive cells is translocated from the optic stalk into both fissure margins. Furthermore, we show that induced BMP signalling as well as Wnt-signalling inhibition result in morphogenetic defects of the optic fissure. Our data also indicate that morphogenesis is crucial for a proper positioning of pre-specified dorsal-ventral optic cup domains.


Asunto(s)
Morfogénesis , Disco Óptico/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Coloboma/embriología , Coloboma/genética , Coloboma/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Hibridación in Situ/métodos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Disco Óptico/embriología , Imagen de Lapso de Tiempo/métodos , Proteínas Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
10.
Sci Rep ; 9(1): 1541, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733552

RESUMEN

Incomplete fusion of the optic fissure leads to ocular coloboma, a congenital eye defect that affects up to 7.5 per 10,000 births and accounts for up to 10 percent of childhood blindness. The molecular and cellular mechanisms that facilitate optic fissure fusion remain elusive. We have profiled global gene expression during optic fissure morphogenesis by transcriptome analysis of tissue dissected from the margins of the zebrafish optic fissure and the opposing dorsal retina before (32 hours post fertilisation, hpf), during (48 hpf) and after (56 hpf) optic fissure fusion. Differential expression analysis between optic fissure and dorsal retinal tissue resulted in the detection of several known and novel developmental genes. The expression of selected genes was validated by qRT-PCR analysis and localisation investigated using in situ hybridisation. We discuss significantly overrepresented functional ontology categories in the context of optic fissure morphogenesis and highlight interesting transcripts from hierarchical clustering for subsequent analysis. We have identified netrin1a (ntn1a) as highly differentially expressed across optic fissure fusion, with a resultant ocular coloboma phenotype following morpholino antisense translation-blocking knockdown and downstream disruption of atoh7 expression. To support the identification of candidate genes in human studies, we have generated an online open-access resource for fast and simple quantitative querying of the gene expression data. Our study represents the first comprehensive analysis of the zebrafish optic fissure transcriptome and provides a valuable resource to facilitate our understanding of the complex aetiology of ocular coloboma.


Asunto(s)
Retina/metabolismo , Transcriptoma , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Análisis por Conglomerados , Coloboma/genética , Coloboma/metabolismo , Coloboma/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Perfilación de la Expresión Génica/métodos , Hibridación Fluorescente in Situ , Morfolinos/metabolismo , Netrina-1/genética , Netrina-1/metabolismo , Análisis de Componente Principal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo
11.
Hum Mol Genet ; 28(6): 912-927, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445451

RESUMEN

Polyglutamine (polyQ) expansion in Ataxin-7 (ATXN7) results in spinocerebellar ataxia type 7 (SCA7) and causes visual impairment. SCA7 photoreceptors progressively lose their outer segments (OSs), a structure essential for their visual function. ATXN7 is a subunit of the transcriptional coactivator Spt-Ada-Gcn5 Acetyltransferase complex, implicated in the development of the visual system in flies. To determine the function of ATXN7 in the vertebrate eye, we have inactivated ATXN7 in zebrafish. While ATXN7 depletion in flies led to gross retinal degeneration, in zebrafish, it primarily results in ocular coloboma, a structural malformation responsible for pediatric visual impairment in humans. ATXN7 inactivation leads to elevated Hedgehog signaling in the forebrain, causing an alteration of proximo-distal patterning of the optic vesicle during early eye development and coloboma. At later developmental stages, malformations of photoreceptors due to incomplete formation of their OSs are observed and correlate with altered expression of crx, a key transcription factor involved in the formation of photoreceptor OS. Therefore, we propose that a primary toxic effect of polyQ expansion is the alteration of ATXN7 function in the daily renewal of OS in SCA7. Together, our data indicate that ATXN7 plays an essential role in vertebrate eye morphogenesis and photoreceptor differentiation, and its loss of function may contribute to the development of human coloboma.


Asunto(s)
Ataxina-7/deficiencia , Coloboma/etiología , Coloboma/metabolismo , Predisposición Genética a la Enfermedad , Células Fotorreceptoras/metabolismo , Subunidades de Proteína/deficiencia , Transactivadores/genética , Animales , Animales Modificados Genéticamente , Biomarcadores , Tipificación del Cuerpo/genética , Diferenciación Celular , Coloboma/patología , Modelos Animales de Enfermedad , Edición Génica , Regulación de la Expresión Génica , Histonas/metabolismo , Inmunohistoquímica , Modelos Biológicos , Nervio Óptico/embriología , Nervio Óptico/metabolismo , Organogénesis/genética , Fenotipo , Células Fotorreceptoras/patología , Procesamiento Proteico-Postraduccional , Transactivadores/química , Transactivadores/metabolismo , Pez Cebra
12.
Biochem Biophys Res Commun ; 506(3): 478-484, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30352686

RESUMEN

Microphthalmia is a malformation that reduces the size of the ocular globe. The etiologies of this anomaly are various, but the genetic background appears to have a predominant influence on its development through mutations of genes controlling ocular developmental processes. LRP4 is a type I single transmembrane protein that is essential for the formation of neuromuscular junctions. We created and experimented on homozygous Lrp4-deficient mice and found the microphthalmia phenotype in their eyes. The loss of Lrp4 resulted in an elevated incidence of microphthalmia and affected the mRNA expression of the members of bone morphogenetic protein, fibroblast growth factor, Sonic hedgehog, and WNT signaling pathways and of several pathogenic genes for microphthalmia. Moreover, the loss of Lrp4 enhanced the incidence of aberrant retinal folds, which appeared pleated and corrugated in the eyeball.


Asunto(s)
Eliminación de Gen , Microftalmía/genética , Receptores de LDL/deficiencia , Animales , Coloboma/metabolismo , Feto/anomalías , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Incidencia , Proteínas Relacionadas con Receptor de LDL , Ratones Endogámicos C57BL , Microftalmía/patología , Fenotipo , Receptores de LDL/metabolismo , Retina/anomalías
13.
Invest Ophthalmol Vis Sci ; 59(3): 1167-1177, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29625437

RESUMEN

Purpose: Coloboma is a sight-threatening congenital eye disease caused by a failure in optic fissure (OF) closure. The aim of this study was to investigate the role of Adamts16, a metalloproteinase, in OF closure. Methods: RNA in situ hybridization was used to examine the expression of Adamts16 in developing mouse and zebrafish eyes. Morpholino knockdowns were performed to study adamts16 function during zebrafish eye development. Additionally, immunofluorescent staining, RNA in situ hybridization, bromodeoxyuridine (BrdU) labeling, TUNEL assays, and high-throughput sequencing were used to examine altered cellular and molecular events in adamts16-morphant optic cups (OCs). Results: Adamts16 is expressed at the edges of the closing OF in both mice and zebrafish eyes. Zebrafish adamts16 knockdown resulted in coloboma formation. In adamts16-morphant eyes, the basement membrane failed to disassemble at the closing OF edges, OC cells exhibited decreased proliferation and increased apoptosis, and fibroblast growth factor 8 (fgf8) was ectopically upregulated in the OC. Conclusions: adamts16 is required for proper OF closure in zebrafish eyes. adamts16 controls OF closure possibly through the combined functions of degrading the basement membrane at the closing OF edges, promoting cell proliferation and survival, and restricting fgf8 expression. Our study linked a metalloproteinase to OF closure, which may facilitate future etiologic studies on human coloboma cases.


Asunto(s)
Proteínas ADAMTS/fisiología , Coloboma/embriología , Disco Óptico/anomalías , Disco Óptico/embriología , Proteínas ADAMTS/metabolismo , Animales , Membrana Basal/patología , Coloboma/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Disco Óptico/metabolismo , Pez Cebra
14.
Sci Rep ; 7(1): 10222, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860541

RESUMEN

COACH syndrome is an autosomal recessive developmental disorder, a subtype of Joubert syndrome and related disorders, characterized by cerebellar vermis hypoplasia, oligophrenia, ataxia, coloboma, and hepatic fibrosis. Although mutations in TMEM67 (transmembrane protein 67)/MKS3 (Meckel-Gruber syndrome, type 3) were reported to cause COACH syndrome, this causality has not verified by functional studies. In a 20-year-old Korean man, we found cerebellar ataxia, isolated elevation in serum γ-glutamyl transpeptidase (γ-GTP) activity, oligophrenia, the molar tooth sign (MTS) in the brain MR images and congenital hepatic fibrosis (CHF). Two novel compound heterozygous mutations were found in TMEM67 in the patient: i) missense mutation (c.395 G > C and p.Gly132Ala) in exon 3, and ii) deletion in exon 26 (c.2758delT and p.Tyr920ThrfsX40). Western blotting showed that the p.Tyr920ThrfsX40 mutation accelerates turnover of the TMEM67 protein. Although wild-type human TMEM67 RNA rescued phenotypes of zebrafish embryos injected with anti-sense oligonucleotide morpholinos against tmem67, the two human TMEM67 RNAs individually harboring the two mutations did not. Finally, Wnt signaling, but not Hedgehog signaling, was suppressed in tmem67 morphants. To the best of our knowledge, this is the first report verifying the causality between COACH syndrome and TMEM67, which will further our understanding of molecular pathogenesis of the syndrome.


Asunto(s)
Anomalías Múltiples/genética , Ataxia/genética , Encéfalo/anomalías , Colestasis/genética , Coloboma/genética , Hepatopatías/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Anomalías Múltiples/tratamiento farmacológico , Anomalías Múltiples/metabolismo , Animales , Ataxia/tratamiento farmacológico , Ataxia/metabolismo , Encéfalo/metabolismo , Colestasis/tratamiento farmacológico , Colestasis/metabolismo , Coloboma/tratamiento farmacológico , Coloboma/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hepatopatías/tratamiento farmacológico , Hepatopatías/metabolismo , Masculino , Morfolinos/administración & dosificación , Morfolinos/farmacología , Mutación Missense , Eliminación de Secuencia , Vía de Señalización Wnt , Adulto Joven , Pez Cebra
15.
Ophthalmology ; 124(7): 992-1003, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28412069

RESUMEN

PURPOSE: To identify the genetic cause of and describe the phenotype in 4 families with autosomal recessive retinitis pigmentosa (arRP) that can be associated with pseudocoloboma. DESIGN: Case series. PARTICIPANTS: Seven patients from 4 unrelated families with arRP, among whom 3 patients had bilateral early-onset macular pseudocoloboma. METHODS: We performed homozygosity mapping and whole-exome sequencing in 5 probands and 2 unaffected family members from 4 unrelated families. Subsequently, Sanger sequencing and segregation analysis were performed in additional family members. We reviewed the medical history of individuals carrying IDH3A variants and performed additional ophthalmic examinations, including full-field electroretinography, fundus photography, fundus autofluorescence imaging, and optical coherence tomography. MAIN OUTCOME MEASURES: IDH3A variants, age at diagnosis, visual acuity, fundus appearance, visual field, and full-field electroretinography, fundus autofluorescence, and optical coherence tomography findings. RESULTS: We identified 7 different variants in IDH3A in 4 unrelated families, that is, 5 missense, 1 nonsense, and 1 frameshift variant. All participants showed symptoms early in life, ranging from night blindness to decreased visual acuity, and were diagnosed between the ages of 1 and 11 years. Four participants with biallelic IDH3A variants displayed a typical arRP phenotype and 3 participants were diagnosed with arRP and pseudocoloboma of the macula. CONCLUSIONS: IDH3A variants were identified as a novel cause of typical arRP in some individuals associated with macular pseudocoloboma. We observed both phenotypes in 2 siblings carrying the same compound heterozygous variants, which could be explained by variable disease expression and warrants caution when making assertions about genotype-phenotype correlations.


Asunto(s)
Coloboma/genética , ADN/genética , Proteínas del Ojo/genética , Estudios de Asociación Genética , Mácula Lútea/patología , Mutación , Retinitis Pigmentosa/genética , Adolescente , Adulto , Niño , Preescolar , Coloboma/diagnóstico , Coloboma/metabolismo , Análisis Mutacional de ADN , Electrorretinografía , Exoma , Proteínas del Ojo/metabolismo , Femenino , Genes Recesivos , Homocigoto , Humanos , Masculino , Linaje , Fenotipo , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/metabolismo , Tomografía de Coherencia Óptica , Agudeza Visual , Campos Visuales , Adulto Joven
17.
Dev Biol ; 407(2): 256-64, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26407529

RESUMEN

Previous results have shown that Bone Morphogenetic Protein (BMP) signaling is essential for lens specification and differentiation. How BMP signals are regulated in the prospective lens ectoderm is not well defined. To address this issue we have modulated BMP activity in a chicken embryo pre-lens ectoderm explant assay, and also studied transgenic mice, in which the type I BMP receptors, Bmpr1a and Acvr1, are deleted from the prospective lens ectoderm. Our results show that chicken embryo pre-lens ectoderm cells express BMPs and require BMP signaling for lens specification in vitro, and that in vivo inhibition of BMP signals in the mouse prospective lens ectoderm interrupts lens placode formation and prevents lens invagination. Furthermore, our results provide evidence that BMP expression is negatively auto-regulated in the lens-forming ectoderm, decreasing when the tissue is exposed to exogenous BMPs and increasing when BMP signaling is prevented. In addition, eyes lacking BMP receptors in the prospective lens placode develop coloboma in the adjacent wild type optic cup. In these eyes, Bmp7 expression increases in the ventral optic cup and the normal dorsal-ventral gradient of BMP signaling in the optic cup is disrupted. Pax2 becomes undetectable and expression of Sfrp2 increases in the ventral optic cup, suggesting that increased BMP signaling alter their expression, resulting in failure to close the optic fissure. In summary, our results suggest that negative and positive auto-regulation of BMP expression is important to regulate early eye development.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Ojo/embriología , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Células CHO , Embrión de Pollo , Coloboma/embriología , Coloboma/metabolismo , Coloboma/patología , Cricetinae , Cricetulus , Ectodermo/embriología , Ectodermo/metabolismo , Inmunohistoquímica , Hibridación Fluorescente in Situ , Cristalino/embriología , Cristalino/metabolismo , Ratones Transgénicos
18.
Mech Dev ; 133: 218-29, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25151399

RESUMEN

During vertebrate eye development retinal progenitor cells (RPCs) differentiate into all neural cell types of the retina. Retinal ganglion cells (RGCs) represent the first cell type to be generated. For their development, Atoh7, a basic Helix Loop Helix (bHLH) transcription factor is crucial. Atoh7 loss of function results in a massive reduction or even a total loss of RGCs. However, inconsistent results have been obtained in atoh7 gain of function experiments with respect to ganglion cell genesis, implying that the effect of Atoh7 is likely to be dependent on the competence state of the RPC. In this study we addressed the differential susceptibilities of early RPCs to Atoh7 in vivo, using medaka. Unexpectedly, we observed a largely normal development of the dorsal retina, although atoh7 was precociously expressed. However, the development of the retina close to the optic nerve head (part of the ventral retina) was disturbed severely. Photoreceptors were largely absent and the Müller glia cell number was reduced significantly. The majority of cells in this domain were ganglion cells and the abnormal development of this area affected the closure of the optic fissure resulting in coloboma.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Peces/genética , Oryzias/embriología , Oryzias/genética , Retina/embriología , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Recuento de Células , Diferenciación Celular , Coloboma/embriología , Coloboma/genética , Coloboma/metabolismo , Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica , Oryzias/metabolismo , Retina/citología , Retina/metabolismo
19.
PLoS Genet ; 10(7): e1004491, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010521

RESUMEN

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.


Asunto(s)
Coloboma/genética , Desarrollo Embrionario/genética , Proteínas Hedgehog/biosíntesis , Proteínas Hedgehog/genética , Factores de Transcripción SOXC/genética , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/genética , Animales , Enfermedades de la Coroides/genética , Enfermedades de la Coroides/metabolismo , Enfermedades de la Coroides/patología , Coloboma/metabolismo , Coloboma/patología , Embrión no Mamífero , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Humanos , Morfogénesis/genética , ARN Mensajero/biosíntesis , Factores de Transcripción SOXC/biosíntesis , Transducción de Señal/genética , Pez Cebra/genética
20.
J AAPOS ; 18(1): 90-2, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24568996

RESUMEN

Colobomata are etiologically heterogeneous and may occur as an isolated defect or as a feature of a variety of single-gene disorders, chromosomal syndromes, or malformation syndromes. Although not classically associated with Marfan syndrome, colobomata have been described in several reports of Marfan syndrome, typically involving the lens and rarely involving other ocular structures. While colobomata of the lens have been described in Marfan syndrome, there are very few reports of coloboma involving other ocular structures. We report a newborn boy presenting with coloboma of the iris, lens, retina, and optic disk who was subsequently diagnosed with Marfan syndrome. Marfan syndrome is a disorder of increased TGFß signaling, and recent work in the mouse model suggests a role for TGFß signaling in eye development and coloboma formation, suggesting a causal association between Marfan syndrome and coloboma.


Asunto(s)
Coloboma/etiología , Iris/anomalías , Cristalino/anomalías , Síndrome de Marfan/complicaciones , Disco Óptico/anomalías , Retina/anomalías , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Aracnodactilia/diagnóstico , Coloboma/diagnóstico , Coloboma/metabolismo , Análisis Mutacional de ADN , Femenino , Fibrilinas , Edad Gestacional , Humanos , Recién Nacido , Masculino , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Proteínas de Microfilamentos/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA