Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 29(4): 591-604, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31814023

ABSTRACT

Mutations in the key transcription factor, SOX2, alone account for 20% of anophthalmia (no eye) and microphthalmia (small eye) birth defects in humans-yet its regulation is not well understood, especially on the post-transcription level. We report the unprecedented finding that the conserved RNA-binding motif protein, RBM24, positively controls Sox2 mRNA stability and is necessary for optimal SOX2 mRNA and protein levels in development, perturbation of which causes ocular defects, including microphthalmia and anophthalmia. RNA immunoprecipitation assay indicates that RBM24 protein interacts with Sox2 mRNA in mouse embryonic eye tissue. and electrophoretic mobility shift assay shows that RBM24 directly binds to the Sox2 mRNA 3'UTR, which is dependent on AU-rich elements (ARE) present in the Sox2 mRNA 3'UTR. Further, we demonstrate that Sox2 3'UTR AREs are necessary for RBM24-based elevation of Sox2 mRNA half-life. We find that this novel RBM24-Sox2 regulatory module is essential for early eye development in vertebrates. We show that Rbm24-targeted deletion using a constitutive CMV-driven Cre in mouse, and rbm24a-CRISPR/Cas9-targeted mutation or morpholino knockdown in zebrafish, results in Sox2 downregulation and causes the developmental defects anophthalmia or microphthalmia, similar to human SOX2-deficiency defects. We further show that Rbm24 deficiency leads to apoptotic defects in mouse ocular tissue and downregulation of eye development markers Lhx2, Pax6, Jag1, E-cadherin and gamma-crystallins. These data highlight the exquisite specificity that conserved RNA-binding proteins like RBM24 mediate in the post-transcriptional control of key transcription factors, namely, SOX2, associated with organogenesis and human developmental defects.


Subject(s)
Anophthalmos/pathology , Eye Abnormalities/pathology , Microphthalmos/pathology , Mutation , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/physiology , SOXB1 Transcription Factors/genetics , Animals , Anophthalmos/genetics , Anophthalmos/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/metabolism , Organogenesis , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish
2.
PLoS Genet ; 15(5): e1008130, 2019 05.
Article in English | MEDLINE | ID: mdl-31048900

ABSTRACT

Nanophthalmos is a rare, potentially devastating eye condition characterized by small eyes with relatively normal anatomy, a high hyperopic refractive error, and frequent association with angle closure glaucoma and vision loss. The condition constitutes the extreme of hyperopia or farsightedness, a common refractive error that is associated with strabismus and amblyopia in children. NNO1 was the first mapped nanophthalmos locus. We used combined pooled exome sequencing and strong linkage data in the large family used to map this locus to identify a canonical splice site alteration upstream of the last exon of the gene encoding myelin regulatory factor (MYRF c.3376-1G>A), a membrane bound transcription factor that undergoes autoproteolytic cleavage for nuclear localization. This variant produced a stable RNA transcript, leading to a frameshift mutation p.Gly1126Valfs*31 in the C-terminus of the protein. In addition, we identified an early truncating MYRF frameshift mutation, c.769dupC (p.S264QfsX74), in a patient with extreme axial hyperopia and syndromic features. Myrf conditional knockout mice (CKO) developed depigmentation of the retinal pigment epithelium (RPE) and retinal degeneration supporting a role of this gene in retinal and RPE development. Furthermore, we demonstrated the reduced expression of Tmem98, another known nanophthalmos gene, in Myrf CKO mice, and the physical interaction of MYRF with TMEM98. Our study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.


Subject(s)
Glaucoma, Angle-Closure/genetics , Hyperopia/genetics , Membrane Proteins/genetics , Microphthalmos/genetics , Retinal Degeneration/genetics , Transcription Factors/genetics , Adult , Animals , Child , Child, Preschool , Exons , Family , Female , Frameshift Mutation/genetics , Genetic Variation/genetics , Glaucoma, Angle-Closure/metabolism , Humans , Hyperopia/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/metabolism , Middle Aged , Pedigree , RNA Splice Sites/genetics , Refractive Errors/genetics , Transcription Factors/metabolism
3.
J Vasc Res ; 58(4): 252-266, 2021.
Article in English | MEDLINE | ID: mdl-33873190

ABSTRACT

PURPOSE: The retinal vasculature is heavily invested by pericytes. Small GTPase R-Ras is highly expressed in endothelial cells and pericytes, suggesting importance of this Ras homolog for the regulation of the blood vessel wall. We investigated the specific contribution of pericyte-expressed R-Ras to the development of the retinal vasculature. METHODS: The effect of R-Ras deficiency in pericytes was analyzed in pericyte-targeted conditional Rras knockout mice at birth and during the capillary plexus formation in the neonatal retina. RESULTS: The offspring of these mice frequently exhibited unilateral microphthalmia. Analyses of the developing retinal vasculature in the eyes without microphthalmia revealed excessive endothelial cell proliferation, sprouting, and branching of the capillary plexus in these animals. These vessels were structurally defective with diminished pericyte coverage and basement membrane formation. Furthermore, these vessels showed reduced VE-cadherin staining and significantly elevated plasma leakage indicating the breakdown of the blood-retinal barrier. This defect was associated with considerable macrophage infiltration in the retina. CONCLUSIONS: The normal retinal vascular development is dependent on R-Ras expression in pericytes, and the absence of it leads to unattenuated angiogenesis and significantly weakens the blood-retinal barrier. Our findings underscore the importance of R-Ras for pericyte function during the normal eye development.


Subject(s)
Blood-Retinal Barrier/metabolism , Microphthalmos/metabolism , Neovascularization, Pathologic , Pericytes/metabolism , Retinal Vessels/metabolism , ras Proteins/deficiency , Animals , Animals, Newborn , Antigens, CD/metabolism , Blood-Retinal Barrier/pathology , Cadherins/metabolism , Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Genetic Predisposition to Disease , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/pathology , Pericytes/pathology , Phenotype , Receptor, Platelet-Derived Growth Factor beta/deficiency , Receptor, Platelet-Derived Growth Factor beta/genetics , Retinal Vessels/pathology , ras Proteins/genetics
4.
Nucleic Acids Res ; 47(6): 2822-2839, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30698748

ABSTRACT

The DNA methylation epigenetic signature is a key determinant during development. Rules governing its establishment and maintenance remain elusive especially at repetitive sequences, which account for the majority of methylated CGs. DNA methylation is altered in a number of diseases including those linked to mutations in factors that modify chromatin. Among them, SMCHD1 (Structural Maintenance of Chromosomes Hinge Domain Containing 1) has been of major interest following identification of germline mutations in Facio-Scapulo-Humeral Dystrophy (FSHD) and in an unrelated developmental disorder, Bosma Arhinia Microphthalmia Syndrome (BAMS). By investigating why germline SMCHD1 mutations lead to these two different diseases, we uncovered a role for this factor in de novo methylation at the pluripotent stage. SMCHD1 is required for the dynamic methylation of the D4Z4 macrosatellite upon reprogramming but seems dispensable for methylation maintenance. We find that FSHD and BAMS patient's cells carrying SMCHD1 mutations are both permissive for DUX4 expression, a transcription factor whose regulation has been proposed as the main trigger for FSHD. These findings open new questions as to what is the true aetiology for FSHD, the epigenetic events associated with the disease thus calling the current model into question and opening new perspectives for understanding repetitive DNA sequences regulation.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , DNA Methylation , Homeodomain Proteins/genetics , Microsatellite Repeats/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Choanal Atresia/genetics , Choanal Atresia/metabolism , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Male , Microphthalmos/genetics , Microphthalmos/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Nose/abnormalities
5.
Mol Vis ; 25: 129-142, 2019.
Article in English | MEDLINE | ID: mdl-30820148

ABSTRACT

Purpose: To identify the effects of a single copy deletion of Yap1 (Yap1 +/-) in the mouse eye, the ocular phenotypic consequences of Yap1 +/- were determined in detail. Methods: Complete ophthalmic examinations, as well as corneal esthesiometry, the phenol red thread test, intraocular pressure, and Fourier-domain optical coherence tomography were performed on Yap1 +/- and age-matched wild-type (WT) mice between eyelid opening (2 weeks after birth) and adulthood (2 months and 1 year after birth). Following euthanasia, enucleated eyes were characterized histologically. Results: Microphthalmia with small palpebral fissures, corneal fibrosis, and reduced corneal sensation were common findings in the Yap1 +/- mice. Generalized corneal fibrosis precluded clinical examination of the posterior structures. Histologically, thinning and keratinization of the corneal epithelium were observed in the Yap1 +/- mice in comparison with the WT mice. Distorted collagen fiber arrangement and hypercellularity of keratocytes were observed in the stroma. Descemet's membrane was extremely thin and lacked an endothelial layer in the Yap1 +/- mice. The iris was adherent to the posterior cornea along most of its surface creating a distorted contour. Most of the Yap1 +/- eyes were microphakic with swollen fibers and bladder cells. The retinas of the Yap1 +/- mice were normal at 2 weeks and 2 months of age, but the presence of retinal abnormalities, including retinoschisis and detachment, was markedly increased in the Yap1 +/- mice at 1 year of age. Conclusions: The results show that the heterozygous deletion of the Yap1 gene in mice leads to complex ocular abnormalities, including microphthalmia, corneal fibrosis, anterior segment dysgenesis, and cataract.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cataract/genetics , Eye Abnormalities/genetics , Microphthalmos/genetics , Phenotype , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Animals , Cataract/diagnostic imaging , Cataract/metabolism , Cataract/pathology , Cell Cycle Proteins , Corneal Stroma/diagnostic imaging , Corneal Stroma/metabolism , Corneal Stroma/pathology , Descemet Membrane/diagnostic imaging , Descemet Membrane/metabolism , Descemet Membrane/pathology , Epithelium, Corneal/diagnostic imaging , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Eye Abnormalities/diagnostic imaging , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Female , Fibrosis , Gene Expression , Heterozygote , Intraocular Pressure/physiology , Iris/diagnostic imaging , Iris/metabolism , Iris/pathology , Male , Mice , Mice, Knockout , Microphthalmos/diagnostic imaging , Microphthalmos/metabolism , Microphthalmos/pathology , Phosphoproteins/deficiency , Retina/diagnostic imaging , Retina/metabolism , Retina/pathology , Tomography, Optical Coherence , Tonometry, Ocular , YAP-Signaling Proteins
6.
Exp Eye Res ; 188: 107632, 2019 11.
Article in English | MEDLINE | ID: mdl-30991053

ABSTRACT

During an ENU (N-ethyl-N-nitrosourea) mutagenesis screen, we observed a dominant small-eye mutant mouse with viable homozygotes. A corresponding mutant line was established and referred to as Aey69 (abnormality of the eye #69). Comprehensive phenotyping of the homozygous Aey69 mutants in the German Mouse Clinic revealed only a subset of statistically significant alterations between wild types and homozygous mutants. The mutation causes microphthalmia without a lens but with retinal hyperproliferation. Linkage was demonstrated to mouse chromosome 3 between the markers D3Mit188 and D3Mit11. Sequencing revealed a 358 A-> C mutation (Ile120Leu) in the Hist2h3c1 gene and a 71 T-> C (Val24Ala) mutation in the Gja8 gene. Detailed analysis of eye development in the homozygous mutant mice documented a perturbed lens development starting from the lens vesicle stage including decreasing expression of crystallins as well as of lens-specific transcription factors like PITX3 and FOXE3. In contrast, we observed an early expression of retinal progenitor cells characterized by several markers including BRN3 (retinal ganglion cells) and OTX2 (cone photoreceptors). The changes in the retina at the early embryonic stages of E11.5-E15.5 happen in parallel with apoptotic processes in the lens at the respective stages. The excessive retinal hyperproliferation is characterized by an increased level of Ki67. The hyperproliferation, however, does not disrupt the differentiation and appearance of the principal retinal cell types at postnatal stages, even if the overgrowing retina covers finally the entire bulbus of the eye. Morpholino-mediated knock-down of the hist2h3ca1 gene in zebrafish leads to a specific perturbation of lens development. When injected into zebrafish zygotes, only the mutant mouse mRNA leads to severe malformations, ranging from cyclopia to severe microphthalmia. The wild-type Hist2h3c1 mRNA can rescue the morpholino-induced defects corroborating its specific function in lens development. Based upon these data, it is concluded that the ocular function of the Hist2h3c1 gene (encoding a canonical H3.2 variant) is conserved throughout evolution. Moreover, the data highlight also the importance of Hist2h3c1 in the coordinated formation of lens and retina during eye development.


Subject(s)
Gene Knockdown Techniques , Histones/genetics , Lens Diseases/genetics , Microphthalmos/genetics , Mutation , Animals , Crystallins/metabolism , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Ki-67 Antigen/metabolism , Lens Diseases/embryology , Lens Diseases/metabolism , Lens Diseases/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microphthalmos/embryology , Microphthalmos/metabolism , Microphthalmos/pathology , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Transcription Factors/metabolism , Zebrafish
7.
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
8.
J Pathol ; 243(1): 9-15, 2017 09.
Article in English | MEDLINE | ID: mdl-28608987

ABSTRACT

SMARCA4 chromatin remodelling factor is mutated in 11% of Coffin-Siris syndrome (CSS) patients and in almost all small-cell carcinoma of the ovary hypercalcaemic type (SCCOHT) tumours. Missense mutations with gain-of-function or dominant-negative effects are associated with CSS, whereas inactivating mutations, leading to loss of SMARCA4 expression, have been exclusively found in SCCOHT. We applied whole-exome sequencing to study a 15-year-old patient with mild CSS who concomitantly developed SCCOHT at age 13 years. Interestingly, our patient also showed congenital microphthalmia, which has never previously been reported in CSS patients. We detected a de novo germline heterozygous nonsense mutation in exon 19 of SMARCA4 (c.2935C > T;p.Arg979*), and a somatic frameshift mutation in exon 6 (c.1236_1236delC;p.Gln413Argfs*88), causing complete loss of SMARCA4 immunostaining in the tumour. The immunohistochemical findings are supported by the observation that the c.2935C > T mutant transcript was detected by reverse transcription polymerase chain reaction at a much lower level than the wild-type allele in whole blood and the lymphoblastoid cell line of the proband, confirming nonsense-mediated mRNA decay. Accordingly, immunoblotting demonstrated that there was approximately half the amount of SMARCA4 protein in the proband's cells as in controls. This study suggests that SMARCA4 constitutional mutations associated with CSS are not necessarily non-truncating, and that haploinsufficiency may explain milder CSS phenotypes, as previously reported for haploinsufficient ARID1B. In addition, our case supports the dual role of chromatin remodellers in developmental disorders and cancer, as well as the involvement of SMARCA4 in microphthalmia, confirming previous findings in mouse models and the DECIPHER database. Finally, we speculate that mild CSS might be under-recognized in a proportion of SCCOHT patients harbouring SMARCA4 mutations. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Abnormalities, Multiple/genetics , Biomarkers, Tumor/genetics , Carcinoma, Small Cell/genetics , Codon, Nonsense , DNA Helicases/genetics , Face/abnormalities , Frameshift Mutation , Hand Deformities, Congenital/genetics , Hypercalcemia/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Microphthalmos/genetics , Neck/abnormalities , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/metabolism , Adolescent , Biomarkers, Tumor/analysis , Blotting, Western , Carcinoma, Small Cell/chemistry , Carcinoma, Small Cell/diagnosis , DNA Helicases/analysis , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/metabolism , Heterozygote , Humans , Hypercalcemia/diagnosis , Hypercalcemia/metabolism , Immunohistochemistry , Intellectual Disability/diagnosis , Intellectual Disability/metabolism , Male , Micrognathism/diagnosis , Micrognathism/metabolism , Microphthalmos/diagnosis , Microphthalmos/metabolism , Middle Aged , Nuclear Proteins/analysis , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/diagnosis , Pedigree , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis
9.
Stem Cells ; 34(11): 2625-2634, 2016 11.
Article in English | MEDLINE | ID: mdl-27301076

ABSTRACT

Few gene targets of Visual System Homeobox 2 (VSX2) have been identified despite its broad and critical role in the maintenance of neural retina (NR) fate during early retinogenesis. We performed VSX2 ChIP-seq and ChIP-PCR assays on early stage optic vesicle-like structures (OVs) derived from human iPS cells (hiPSCs), which highlighted WNT pathway genes as direct regulatory targets of VSX2. Examination of early NR patterning in hiPSC-OVs from a patient with a functional null mutation in VSX2 revealed mis-expression and upregulation of WNT pathway components and retinal pigmented epithelium (RPE) markers in comparison to control hiPSC-OVs. Furthermore, pharmacological inhibition of WNT signaling rescued the early mutant phenotype, whereas augmentation of WNT signaling in control hiPSC-OVs phenocopied the mutant. These findings reveal an important role for VSX2 as a regulator of WNT signaling and suggest that VSX2 may act to maintain NR identity at the expense of RPE in part by direct repression of WNT pathway constituents. Stem Cells 2016;34:2625-2634.


Subject(s)
Body Patterning/genetics , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/metabolism , Microphthalmos/genetics , Retinal Pigment Epithelium/metabolism , Transcription Factors/genetics , Wnt1 Protein/genetics , Amino Acid Substitution , Benzothiazoles/pharmacology , Biomarkers/metabolism , Cell Differentiation , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Gene Expression Profiling , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmos/metabolism , Microphthalmos/pathology , Mutation , Phenotype , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , Wnt1 Protein/agonists , Wnt1 Protein/antagonists & inhibitors , Wnt1 Protein/metabolism
10.
Subcell Biochem ; 81: 77-93, 2016.
Article in English | MEDLINE | ID: mdl-27830501

ABSTRACT

Vitamin A, retinol, circulates in blood bound to retinol binding protein (RBP). In some tissues, the retinol-RBP complex (holo-RBP) is recognized by a membrane receptor, termed STRA6, which mediates uptake of retinol into cells. Recent studies have revealed that, in addition to serving as a retinol transporter, STRA6 is a ligand-activated cell surface signaling receptor that, upon binding of holo-RBP activates JAK/STAT signaling, culminating in the induction of STAT target genes. It has further been shown that retinol transport and cell signaling by STRA6 are critically interdependent and that both are coupled to intracellular vitamin A metabolism. The molecular mechanism of action of STRA6 and its associated machinery is beginning to be revealed, but further work is needed to identify and characterize the complete range of genes and associated signaling cascades that are regulated by STRA6 in different tissues. An understanding of STRA6 is clinically relevant, as for example, it has been shown to be hyper- activated in obese animals, leading to insulin resistance. A potential role for STRA6 in other pathologies, including cancer, awaits further investigation.


Subject(s)
Membrane Proteins/physiology , Vitamin A/metabolism , Animals , Anophthalmos/genetics , Anophthalmos/metabolism , Biological Transport , Gene Expression Regulation , Humans , Insulin Resistance , Janus Kinases/physiology , Lung Diseases/genetics , Lung Diseases/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/metabolism , Obesity/metabolism , Organ Specificity , Prealbumin/physiology , Retinol-Binding Proteins, Plasma/metabolism , STAT Transcription Factors/physiology , Signal Transduction/physiology , Transcription, Genetic
11.
Am J Hum Genet ; 93(4): 765-72, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24075189

ABSTRACT

Anophthalmia and/or microphthalmia, pulmonary hypoplasia, diaphragmatic hernia, and cardiac defects are the main features of PDAC syndrome. Recessive mutations in STRA6, encoding a membrane receptor for the retinol-binding protein, have been identified in some cases with PDAC syndrome, although many cases have remained unexplained. Using whole-exome sequencing, we found that two PDAC-syndrome-affected siblings, but not their unaffected sibling, were compound heterozygous for nonsense (c.355C>T [p.Arg119(∗)]) and frameshift (c.1201_1202insCT [p.Ile403Serfs(∗)15]) mutations in retinoic acid receptor beta (RARB). Transfection studies showed that p.Arg119(∗) and p.Ile403Serfs(∗)15 altered RARB had no transcriptional activity in response to ligands, confirming that the mutations induced a loss of function. We then sequenced RARB in 15 subjects with anophthalmia and/or microphthalmia and at least one other feature of PDAC syndrome. Surprisingly, three unrelated subjects with microphthalmia and diaphragmatic hernia showed de novo missense mutations affecting the same codon; two of the subjects had the c.1159C>T (Arg387Cys) mutation, whereas the other one carried the c.1159C>A (p.Arg387Ser) mutation. We found that compared to the wild-type receptor, p.Arg387Ser and p.Arg387Cys altered RARB induced a 2- to 3-fold increase in transcriptional activity in response to retinoic acid ligands, suggesting a gain-of-function mechanism. Our study thus suggests that both recessive and dominant mutations in RARB cause anophthalmia and/or microphthalmia and diaphragmatic hernia, providing further evidence of the crucial role of the retinoic acid pathway during eye development and organogenesis.


Subject(s)
Hernia, Diaphragmatic/genetics , Microphthalmos/genetics , Mutation , Receptors, Retinoic Acid/genetics , Adolescent , Anophthalmos/genetics , Anophthalmos/metabolism , Exome , Female , Hernia, Diaphragmatic/metabolism , Humans , Infant, Newborn , Male , Microphthalmos/metabolism , Receptors, Retinoic Acid/metabolism , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/metabolism , Tretinoin/metabolism
12.
Exp Eye Res ; 149: 26-39, 2016 08.
Article in English | MEDLINE | ID: mdl-27235794

ABSTRACT

We investigated the corneal morphology of adult Mp/+ mice, which are heterozygous for the micropinna microphthalmia mutation, and identified several abnormalities, which implied that corneal epithelial maintenance was abnormal. The Mp/+ corneal epithelium was thin, loosely packed and contained goblet cells in older mice. Evidence also suggested that the barrier function was compromised. However, there was no major effect on corneal epithelial cell turnover and mosaic patterns of radial stripes indicated that radial cell movement was normal. Limbal blood vessels formed an abnormally wide limbal vasculature ring, K19-positive cells were distributed more widely than normal and K12 was weakly expressed in the peripheral cornea. This raises the possibilities that the limbal-corneal boundary was poorly defined or the limbus was wider than normal. BrdU label-retaining cell numbers and quantitative clonal analysis suggested that limbal epithelial stem cell numbers were not depleted and might be higher than normal. However, as corneal epithelial homeostasis was abnormal, it is possible that Mp/+ stem cell function was impaired. It has been shown recently that the Mp mutation involves a chromosome 18 inversion that disrupts the Fbn2 and Isoc1 genes and produces an abnormal, truncated fibrillin-2(MP) protein. This abnormal protein accumulates in the endoplasmic reticulum (ER) of cells that normally express Fbn2 and causes ER stress. It was also shown that Fbn2 is expressed in the corneal stroma but not the corneal epithelium, suggesting that the presence of truncated fibrillin-2(MP) protein in the corneal stroma disrupts corneal epithelial homeostasis in Mp/+ mice.


Subject(s)
Epithelium, Corneal/abnormalities , Microphthalmos/genetics , Mutation , Animals , Animals, Newborn , Cell Count , Cell Movement , Epithelium, Corneal/pathology , Female , Heterozygote , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microphthalmos/metabolism , Microphthalmos/pathology , Microscopy, Confocal
13.
Hum Mol Genet ; 22(16): 3250-8, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23591992

ABSTRACT

The major active retinoid, all-trans retinoic acid, has long been recognized as critical for the development of several organs, including the eye. Mutations in STRA6, the gene encoding the cellular receptor for vitamin A, in patients with Matthew-Wood syndrome and anophthalmia/microphthalmia (A/M), have previously demonstrated the importance of retinol metabolism in human eye disease. We used homozygosity mapping combined with next-generation sequencing to interrogate patients with anophthalmia and microphthalmia for new causative genes. We used whole-exome and whole-genome sequencing to study a family with two affected brothers with bilateral A/M and a simplex case with bilateral anophthalmia and hypoplasia of the optic nerve and optic chiasm. Analysis of novel sequence variants revealed homozygosity for two nonsense mutations in ALDH1A3, c.568A>G, predicting p.Lys190*, in the familial cases, and c.1165A>T, predicting p.Lys389*, in the simplex case. Both mutations predict nonsense-mediated decay and complete loss of function. We performed antisense morpholino (MO) studies in Danio rerio to characterize the developmental effects of loss of Aldh1a3 function. MO-injected larvae showed a significant reduction in eye size, and aberrant axonal projections to the tectum were noted. We conclude that ALDH1A3 loss of function causes anophthalmia and aberrant eye development in humans and in animal model systems.


Subject(s)
Aldehyde Oxidoreductases/genetics , Anophthalmos/genetics , Codon, Nonsense/genetics , Microphthalmos/genetics , Optic Chiasm/abnormalities , Optic Nerve/abnormalities , Aldehyde Oxidoreductases/metabolism , Animals , Anophthalmos/metabolism , Child , Child, Preschool , Exome , Eye/growth & development , Eye/pathology , Female , Genome , Homozygote , Humans , Infant , Larva/genetics , Larva/growth & development , Larva/metabolism , Male , Microphthalmos/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Zebrafish/embryology , Zebrafish/genetics
14.
Am J Hum Genet ; 91(5): 942-9, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23122588

ABSTRACT

Microphthalmia with linear skin lesions (MLS) is an X-linked dominant male-lethal disorder associated with mutations in holocytochrome c-type synthase (HCCS), which encodes a crucial player of the mitochondrial respiratory chain (MRC). Unlike other mitochondrial diseases, MLS is characterized by a well-recognizable neurodevelopmental phenotype. Interestingly, not all clinically diagnosed MLS cases have mutations in HCCS, thus suggesting genetic heterogeneity for this disorder. Among the possible candidates, we analyzed the X-linked COX7B and found deleterious de novo mutations in two simplex cases and a nonsense mutation, which segregates with the disease, in a familial case. COX7B encodes a poorly characterized structural subunit of cytochrome c oxidase (COX), the MRC complex IV. We demonstrated that COX7B is indispensable for COX assembly, COX activity, and mitochondrial respiration. Downregulation of the COX7B ortholog (cox7B) in medaka (Oryzias latipes) resulted in microcephaly and microphthalmia that recapitulated the MLS phenotype and demonstrated an essential function of complex IV activity in vertebrate CNS development. Our results indicate an evolutionary conserved role of the MRC complexes III and IV for the proper development of the CNS in vertebrates and uncover a group of mitochondrial diseases hallmarked by a developmental phenotype.


Subject(s)
Electron Transport Complex IV/genetics , Microphthalmos/genetics , Mitochondrial Diseases/genetics , Mutation , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Cell Line , Female , Gene Expression Regulation , Genes, X-Linked , Genotype , Humans , Lyases/genetics , Microphthalmos/metabolism , Microphthalmos/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Molecular Sequence Data , Oryzias/genetics , Oryzias/metabolism , Pedigree , Phenotype , Skin/pathology
15.
Clin Genet ; 88(5): 468-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25457163

ABSTRACT

Anophthalmia/microphthalmia (A/M) is a genetically heterogeneous birth defect for which the etiology is unknown in more than 50% of patients. We used exome sequencing with the ACE Exome(TM) (Personalis, Inc; 18 cases) and UCSF Genomics Core (21 cases) to sequence 28 patients with A/M and four patients with varied developmental eye defects. In the 28 patients with A/M, we identified de novo mutations in three patients (OTX2, p.(Gln91His), RARB, p.Arg387Cys and GDF6, p.Ala249Glu) and inherited mutations in STRA6 in two patients. In patients with developmental eye defects, a female with cataracts and cardiomyopathy had a de novo COL4A1 mutation, p.(Gly773Arg), expanding the phenotype associated with COL4A1 to include cardiomyopathy. A male with a chorioretinal defect, microcephaly, seizures and sensorineural deafness had two PNPT1 mutations, p.(Ala507Ser) and c.401-1G>A, and we describe eye defects associated with this gene for the first time. Exome sequencing was efficient for identifying mutations in pathogenic genes for which there is no clinical testing available and for identifying cases that expand phenotypic spectra, such as the PNPT1 and COL4A1-associated disorders described here.


Subject(s)
Anophthalmos/genetics , Eye Abnormalities/genetics , Microphthalmos/genetics , Mutation , Anophthalmos/metabolism , Collagen Type IV/genetics , DNA Mutational Analysis , Exome , Exoribonucleases/genetics , Female , Humans , Infant , Male , Membrane Proteins/genetics , Microphthalmos/metabolism , Otx Transcription Factors/genetics , Receptors, Retinoic Acid/genetics
16.
J Biol Chem ; 288(15): 10275-85, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23426374

ABSTRACT

Smad7 is an intracellular inhibitory protein that antagonizes the signaling of TGF-ß family members. Deletion of Smad7 in the mouse leads to an abnormality in heart development. However, whether Smad7 has a functional role in the development of other organs has been elusive. Here we present evidence that Smad7 imparts a role to eye development in the mouse. Smad7 is expressed in both the lens and retina in the developing embryonic eye. Depletion of Smad7 caused various degrees of coloboma and microphthalmia with alterations in cell apoptosis and proliferation in eyes. Smad7 was implicated in lens differentiation but was not required for the induction of the lens placode. The development of the periocular mesenchyme was retarded with the down-regulation of Bmp7 and Pitx2 in mutant mice. Retinal spatial patterning was affected by Smad7 deletion and was accompanied by altered bone morphogenetic protein (BMP) signaling. At late gestation stages, TGF-ß signaling was up-regulated in the differentiating retina. Smad7 mutant mice displayed an expanded optic disc with increasing of sonic hedgehog (SHH) signaling. Furthermore, loss of Smad7 led to a temporal change in retinal neurogenesis. In conclusion, our study suggests that Smad7 is essential for eye development. In addition, our data indicate that alterations in the signaling of BMP, TGF-ß, and SHH likely underlie the defects in eye development caused by Smad7 deletion.


Subject(s)
Cell Differentiation/physiology , Lens, Crystalline/embryology , Neurogenesis/physiology , Retina/embryology , Signal Transduction/physiology , Smad7 Protein/biosynthesis , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Coloboma/genetics , Coloboma/metabolism , Coloboma/pathology , Down-Regulation/physiology , Gene Deletion , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lens, Crystalline/cytology , Mice , Mice, Mutant Strains , Microphthalmos/genetics , Microphthalmos/metabolism , Microphthalmos/pathology , Retina/cytology , Smad7 Protein/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Homeobox Protein PITX2
17.
Hum Mol Genet ; 21(8): 1848-60, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22228100

ABSTRACT

Nineteen Wnt ligands and 10 Frizzled (Fz) receptors mediate multiple distinct cellular events during neuronal development. However, their precise roles in cell-type specification and organogenesis are poorly delineated because of overlapping functions and expression profiles. Here, we have explored the role of two closely related Frizzled receptors, Fz5 and Fz8, in mouse retinal development. We previously showed that Fz5(-/-) mice exhibit mild coloboma and microphthalmia at ~50% penetrance. Fz8 expression overlaps with Fz5 in the neural retina and optic fissure/disc. Mice lacking Fz8 show minimal eye and retinal defects. The embryos lacking both Fz5 and Fz8 die early in development, but a majority of triallelic Fz5(-/-);Fz8(+/-) mutants survive until birth. The triallelic mutant develops severe retinal coloboma and microphthalmia with full penetrance. At the cellular level, impaired neurogenesis is indicated by increased early-born retinal neurons that result from accelerated cell cycle exit of progenitors. Deficiency of apical retinal neuroepithelium is indicated by altered localization of apical junction markers, such as atypical protein kinase C, RhoA and ß-catenin. Hes1 expression, which is critical for retinal progenitor expansion, is down-regulated in the triallelic mutant mouse. Furthermore, blocking Frizzled receptors in cultured retinal explants led to basally shifted divisions of retinal progenitors. Together, our studies suggest a dose-dependent regulation of signaling by Fz5 and Fz8 in optic fissure/disc formation and progenitor expansion.


Subject(s)
Frizzled Receptors/genetics , Frizzled Receptors/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Retina/embryology , Retinal Neurons/physiology , Animals , Axons/physiology , Coloboma/genetics , Coloboma/metabolism , Down-Regulation , Gene Expression Regulation, Developmental , Gliosis/genetics , Gliosis/metabolism , Mice , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/metabolism , Mitosis , Mutation , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Optic Disk/embryology , Optic Disk/metabolism , Retina/metabolism , Retinal Neurons/cytology , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
18.
J Neuroinflammation ; 11: 92, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24884664

ABSTRACT

BACKGROUND: Peri-spinal subarachnoid (intrathecal; i.t.) injection of non-viral naked plasmid DNA encoding the anti-inflammatory cytokine, IL-10 (pDNA-IL-10) suppresses chronic neuropathic pain in animal models. However, two sequential i.t. pDNA injections are required within a discrete 5 to 72-hour period for prolonged efficacy. Previous reports identified phagocytic immune cells present in the peri-spinal milieu surrounding the i.t injection site that may play a role in transgene uptake resulting in subsequent IL-10 transgene expression. METHODS: In the present study, we aimed to examine whether factors known to induce pro-phagocytic anti-inflammatory properties of immune cells improve i.t. IL-10 transgene uptake using reduced naked pDNA-IL-10 doses previously determined ineffective. Both the synthetic glucocorticoid, dexamethasone, and the hexose sugar, D-mannose, were factors examined that could optimize i.t. pDNA-IL-10 uptake leading to enduring suppression of neuropathic pain as assessed by light touch sensitivity of the rat hindpaw (allodynia). RESULTS: Compared to dexamethasone, i.t. mannose pretreatment significantly and dose-dependently prolonged pDNA-IL-10 pain suppressive effects, reduced spinal IL-1ß and enhanced spinal and dorsal root ganglia IL-10 immunoreactivity. Macrophages exposed to D-mannose revealed reduced proinflammatory TNF-α, IL-1ß, and nitric oxide, and increased IL-10 protein release, while IL-4 revealed no improvement in transgene uptake. Separately, D-mannose dramatically increased pDNA-derived IL-10 protein release in culture supernatants. Lastly, a single i.t. co-injection of mannose with a 25-fold lower pDNA-IL-10 dose produced prolonged pain suppression in neuropathic rats. CONCLUSIONS: Peri-spinal treatment with D-mannose may optimize naked pDNA-IL-10 transgene uptake for suppression of allodynia, and is a novel approach to tune spinal immune cells toward pro-phagocytic phenotype for improved non-viral gene therapy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Genetic Therapy , Interleukin-10/therapeutic use , Mannose/therapeutic use , Neuralgia/therapy , Pain Threshold/physiology , Animals , Cells, Cultured , Chronic Disease , Constriction, Pathologic/complications , Cytokines/genetics , Cytokines/metabolism , Dexamethasone/therapeutic use , Disease Models, Animal , Interleukin-10/biosynthesis , Male , Mice , Microphthalmos/drug therapy , Microphthalmos/metabolism , Neuralgia/etiology , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/physiology
19.
Mol Syst Biol ; 9: 692, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24084807

ABSTRACT

Genes with common profiles of the presence and absence in disparate genomes tend to function in the same pathway. By mapping all human genes into about 1000 clusters of genes with similar patterns of conservation across eukaryotic phylogeny, we determined that sets of genes associated with particular diseases have similar phylogenetic profiles. By focusing on those human phylogenetic gene clusters that significantly overlap some of the thousands of human gene sets defined by their coexpression or annotation to pathways or other molecular attributes, we reveal the evolutionary map that connects molecular pathways and human diseases. The other genes in the phylogenetic clusters enriched for particular known disease genes or molecular pathways identify candidate genes for roles in those same disorders and pathways. Focusing on proteins coevolved with the microphthalmia-associated transcription factor (MITF), we identified the Notch pathway suppressor of hairless (RBP-Jk/SuH) transcription factor, and showed that RBP-Jk functions as an MITF cofactor.


Subject(s)
Evolution, Molecular , Genome , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmos/genetics , Phylogeny , Algorithms , Amino Acid Sequence , Animals , Bacteria/genetics , Bacteria/metabolism , Cell Line, Tumor , Chromosome Mapping , Databases, Genetic , Fungi/genetics , Fungi/metabolism , Gene Regulatory Networks , Genetic Loci , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/classification , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Metabolic Networks and Pathways , Microphthalmia-Associated Transcription Factor/classification , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmos/metabolism , Microphthalmos/pathology , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid
20.
J Hum Genet ; 59(6): 314-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24694763

ABSTRACT

Oculofaciocardiodental (OFCD) syndrome is a rare X-linked dominant condition. Mutations in BCOR have been described as causal in OFCD syndrome. Almost all BCOR mutations result in premature termination codons (PTCs); therefore, nonsense-mediated mRNA decay (NMD) might have an important role in pathogenesis. The purpose of this study was to identify BCOR mutations in two OFCD patients, if it present, and to clarify the pathogenesis of radiculomegaly using one OFCD patient's pulp and periodontal ligament (PDL) cells. In our genetic analysis, two novel BCOR mutations were found. We also examined the transcript levels and the effects of NMD using cultured pulp and PDL cells from one affected patient. BCOR expression was normal in pulp but reduced in PDL cells, which is consistent with the higher rates of NMD in PDL cells. The mutant PDL cells had unstable mutant transcripts and proliferated faster than did wild-type cells, but mutant pulp cells appeared normal by these measures. In summary, the nonsense and frameshift mutations, which introduce PTCs, were found to contribute to OFCD syndrome in our two patients. Furthermore, in PDL cells, the mutation resulting in a PTC corresponded to greater NMD, unstable mutant transcripts and increased cell proliferation, which may contribute to hyperactive root formation.


Subject(s)
Cataract/congenital , Codon, Nonsense , Dental Pulp/metabolism , Heart Septal Defects/genetics , Microphthalmos/genetics , Periodontal Ligament/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Adolescent , Adult , Cataract/genetics , Cataract/metabolism , Cataract/pathology , Cell Proliferation , Cells, Cultured , Dental Pulp/pathology , Female , Heart Septal Defects/metabolism , Heart Septal Defects/pathology , Humans , Microphthalmos/metabolism , Microphthalmos/pathology , Nonsense Mediated mRNA Decay , Periodontal Ligament/pathology , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL