Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genet Med ; 22(3): 598-609, 2020 03.
Article in English | MEDLINE | ID: mdl-31700164

ABSTRACT

PURPOSE: Most classical aniridia is caused by PAX6 haploinsufficiency. PAX6 missense variants can be hypomorphic or mimic haploinsufficiency. We hypothesized that missense variants also cause previously undescribed disease by altering the affinity and/or specificity of PAX6 genomic interactions. METHODS: We screened PAX6 in 372 individuals with bilateral microphthalmia, anophthalmia, or coloboma (MAC) from the Medical Research Council Human Genetics Unit eye malformation cohort (HGUeye) and reviewed data from the Deciphering Developmental Disorders study. We performed cluster analysis on PAX6-associated ocular phenotypes by variant type and molecular modeling of the structural impact of 86 different PAX6 causative missense variants. RESULTS: Eight different PAX6 missense variants were identified in 17 individuals (15 families) with MAC, accounting for 4% (15/372) of our cohort. Seven altered the paired domain (p.[Arg26Gln]x1, p.[Gly36Val]x1, p.[Arg38Trp]x2, p.[Arg38Gln]x1, p.[Gly51Arg]x2, p.[Ser54Arg]x2, p.[Asn124Lys]x5) and one the homeodomain (p.[Asn260Tyr]x1). p.Ser54Arg and p.Asn124Lys were exclusively associated with severe bilateral microphthalmia. MAC-associated variants were predicted to alter but not ablate DNA interaction, consistent with the electrophoretic mobility shifts observed using mutant paired domains with well-characterized PAX6-binding sites. We found no strong evidence for novel PAX6-associated extraocular disease. CONCLUSION: Altering the affinity and specificity of PAX6-binding genome-wide provides a plausible mechanism for the worse-than-null effects of MAC-associated missense variants.


Subject(s)
Eye Abnormalities/genetics , Genetic Predisposition to Disease , Microphthalmos/genetics , PAX6 Transcription Factor/genetics , Adolescent , Adult , Binding Sites/genetics , Child , Child, Preschool , Cohort Studies , DNA-Binding Proteins/genetics , Eye Abnormalities/pathology , Female , Heterozygote , Humans , Infant , Male , Microphthalmos/pathology , Mutation, Missense/genetics , Pedigree , Young Adult
2.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27486781

ABSTRACT

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Subject(s)
Alleles , Guanine Nucleotide Exchange Factors/genetics , Mutation, Missense/genetics , Retinal Dystrophies/genetics , Ubiquitination/genetics , Adolescent , Adult , Age of Onset , Child , Consanguinity , Cullin Proteins/metabolism , Exome/genetics , Female , Founder Effect , Genes, Recessive , Haplotypes/genetics , Homozygote , Humans , Lymphocytes/metabolism , Male , NF-E2-Related Factor 2/metabolism , Pedigree , Phenotype , RNA, Messenger/genetics , Retina/metabolism , Syndrome , Turkey
3.
Am J Hum Genet ; 98(5): 981-992, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27108798

ABSTRACT

Gillespie syndrome (GS) is characterized by bilateral iris hypoplasia, congenital hypotonia, non-progressive ataxia, and progressive cerebellar atrophy. Trio-based exome sequencing identified de novo mutations in ITPR1 in three unrelated individuals with GS recruited to the Deciphering Developmental Disorders study. Whole-exome or targeted sequence analysis identified plausible disease-causing ITPR1 mutations in 10/10 additional GS-affected individuals. These ultra-rare protein-altering variants affected only three residues in ITPR1: Glu2094 missense (one de novo, one co-segregating), Gly2539 missense (five de novo, one inheritance uncertain), and Lys2596 in-frame deletion (four de novo). No clinical or radiological differences were evident between individuals with different mutations. ITPR1 encodes an inositol 1,4,5-triphosphate-responsive calcium channel. The homo-tetrameric structure has been solved by cryoelectron microscopy. Using estimations of the degree of structural change induced by known recessive- and dominant-negative mutations in other disease-associated multimeric channels, we developed a generalizable computational approach to indicate the likely mutational mechanism. This analysis supports a dominant-negative mechanism for GS variants in ITPR1. In GS-derived lymphoblastoid cell lines (LCLs), the proportion of ITPR1-positive cells using immunofluorescence was significantly higher in mutant than control LCLs, consistent with an abnormality of nuclear calcium signaling feedback control. Super-resolution imaging supports the existence of an ITPR1-lined nucleoplasmic reticulum. Mice with Itpr1 heterozygous null mutations showed no major iris defects. Purkinje cells of the cerebellum appear to be the most sensitive to impaired ITPR1 function in humans. Iris hypoplasia is likely to result from either complete loss of ITPR1 activity or structure-specific disruption of multimeric interactions.


Subject(s)
Aniridia/etiology , Aniridia/pathology , Cerebellar Ataxia/etiology , Cerebellar Ataxia/pathology , Genes, Dominant/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Intellectual Disability/etiology , Intellectual Disability/pathology , Mutation/genetics , Adolescent , Adult , Animals , Cells, Cultured , Child , Female , Humans , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Microscopy, Confocal , Middle Aged , Pedigree , Protein Conformation
4.
Hum Mutat ; 36(12): 1188-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26316326

ABSTRACT

Leber congenital amaurosis (LCA) is a severe autosomal-recessive retinal dystrophy leading to congenital blindness. A recently identified LCA gene is NMNAT1, located in the LCA9 locus. Although most mutations in blindness genes are coding variations, there is accumulating evidence for hidden noncoding defects or structural variations (SVs). The starting point of this study was an LCA9-associated consanguineous family in which no coding mutations were found in the LCA9 region. Exploring the untranslated regions of NMNAT1 revealed a novel homozygous 5'UTR variant, c.-70A>T. Moreover, an adjacent 5'UTR variant, c.-69C>T, was identified in a second consanguineous family displaying a similar phenotype. Both 5'UTR variants resulted in decreased NMNAT1 mRNA abundance in patients' lymphocytes, and caused decreased luciferase activity in human retinal pigment epithelial RPE-1 cells. Second, we unraveled pseudohomozygosity of a coding NMNAT1 mutation in two unrelated LCA patients by the identification of two distinct heterozygous partial NMNAT1 deletions. Molecular characterization of the breakpoint junctions revealed a complex Alu-rich genomic architecture. Our study uncovered hidden genetic variation in NMNAT1-associated LCA and emphasized a shift from coding to noncoding regulatory mutations and repeat-mediated SVs in the molecular pathogenesis of heterogeneous recessive disorders such as hereditary blindness.


Subject(s)
5' Untranslated Regions , DNA Copy Number Variations , Leber Congenital Amaurosis/genetics , Mutation , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Alleles , Alu Elements , Child , Chromosome Breakpoints , Chromosome Mapping , Computational Biology/methods , Consanguinity , Exons , Female , Gene Expression , Genetic Association Studies , Genotype , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leber Congenital Amaurosis/diagnosis , Male , Pedigree , Phenotype , RNA, Messenger/genetics , Sequence Analysis, DNA , Young Adult
5.
Hum Mutat ; 36(1): 39-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25346251

ABSTRACT

Autosomal-recessive Stargardt disease (STGD1) is hallmarked by a large proportion of patients with a single heterozygous causative variant in the disease gene ABCA4. Braun et al. () reported deep intronic variants of ABCA4 in STGD1 patients with one coding variant, prompting us to perform an augmented screen in 131 Belgian STGD1 patients with one or no ABCA4 variant to uncover deep intronic causal ABCA4 variants. This revealed a second variant in 28.6% of cases. Twenty-six percent of these carry the same causal variant c.4539+2001G>A (V4). Haplotyping in V4 carriers showed a common region of 63 kb, suggestive of a founder mutation. Genotype-phenotype correlations suggest a moderate-to-severe impact of V4 on the STGD1 phenotype. In conclusion, V4 occurs in a high fraction of Belgian STGD1 patients and represents the first deep intronic founder mutation in ABCA4. This emphasizes the importance of augmented molecular genetic testing of ABCA4 in Belgian STGD1.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Founder Effect , Macular Degeneration/congenital , Belgium , Genetic Association Studies , Haplotypes , Humans , Introns , Macular Degeneration/genetics , Mutation , Stargardt Disease
6.
Am J Hum Genet ; 91(3): 527-32, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22901948

ABSTRACT

Achromatopsia (ACHM) is an autosomal-recessive retinal dystrophy characterized by color blindness, photophobia, nystagmus, and severely reduced visual acuity. Its prevalence has been estimated to about 1 in 30,000 individuals. Four genes, GNAT2, PDE6C, CNGA3, and CNGB3, have been implicated in ACHM, and all encode functional components of the phototransduction cascade in cone photoreceptors. Applying a functional-candidate-gene approach that focused on screening additional genes involved in this process in a cohort of 611 index cases with ACHM or other cone photoreceptor disorders, we detected a homozygous single base change (c.35C>G) resulting in a nonsense mutation (p.Ser12(∗)) in PDE6H, encoding the inhibitory γ subunit of the cone photoreceptor cyclic guanosine monophosphate phosphodiesterase. The c.35C>G mutation was present in three individuals from two independent families with a clinical diagnosis of incomplete ACHM and preserved short-wavelength-sensitive cone function. Moreover, we show through immunohistochemical colocalization studies in mouse retina that Pde6h is evenly present in all retinal cone photoreceptors, a fact that had been under debate in the past. These findings add PDE6H to the set of genes involved in autosomal-recessive cone disorders and demonstrate the importance of the inhibitory γ subunit in cone phototransduction.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/genetics , Codon, Nonsense , Color Vision Defects/genetics , Adult , Base Sequence , Female , Genes, Recessive , Humans , Male , Middle Aged , Young Adult
7.
Am J Hum Genet ; 90(2): 321-30, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22325361

ABSTRACT

Congenital stationary night blindness (CSNB) is a heterogeneous retinal disorder characterized by visual impairment under low light conditions. This disorder is due to a signal transmission defect from rod photoreceptors to adjacent bipolar cells in the retina. Two forms can be distinguished clinically, complete CSNB (cCSNB) or incomplete CSNB; the two forms are distinguished on the basis of the affected signaling pathway. Mutations in NYX, GRM6, and TRPM1, expressed in the outer plexiform layer (OPL) lead to disruption of the ON-bipolar cell response and have been seen in patients with cCSNB. Whole-exome sequencing in cCSNB patients lacking mutations in the known genes led to the identification of a homozygous missense mutation (c.1807C>T [p.His603Tyr]) in one consanguineous autosomal-recessive cCSNB family and a homozygous frameshift mutation in GPR179 (c.278delC [p.Pro93Glnfs(∗)57]) in a simplex male cCSNB patient. Additional screening with Sanger sequencing of 40 patients identified three other cCSNB patients harboring additional allelic mutations in GPR179. Although, immunhistological studies revealed Gpr179 in the OPL in wild-type mouse retina, Gpr179 did not colocalize with specific ON-bipolar markers. Interestingly, Gpr179 was highly concentrated in horizontal cells and Müller cell endfeet. The involvement of these cells in cCSNB and the specific function of GPR179 remain to be elucidated.


Subject(s)
Exome , Mutation , Myopia/genetics , Night Blindness/genetics , Receptors, G-Protein-Coupled/genetics , Alleles , Animals , Electroretinography/methods , Eye Diseases, Hereditary , Female , Genetic Diseases, X-Linked , Genetic Heterogeneity , Genotyping Techniques/methods , Heterozygote , Homozygote , Humans , Male , Mice , Phenotype , Polymorphism, Single Nucleotide , Protein Structure, Tertiary , Proteoglycans/genetics , Receptors, Metabotropic Glutamate/genetics , Retina/abnormalities , TRPM Cation Channels/genetics
8.
Genet Med ; 17(4): 291-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25122145

ABSTRACT

PURPOSE: The aim of this study was to identify the genetic cause of early-onset autosomal recessive cerebellar ataxia associated with retinal dystrophy in a consanguineous family. METHODS: An affected 6-month-old child underwent neurological and ophthalmological examinations. Genetic analyses included homozygosity mapping, copy number analysis, conventional polymerase chain reaction, Sanger sequencing, quantitative polymerase chain reaction, and whole-exome sequencing. Expression analysis of GRID2 was performed by quantitative polymerase chain reaction and immunohistochemistry. RESULTS: A homozygous deletion of exon 2 of GRID2 (p.Gly30_Glu81del) was identified in the proband. GRID2 encodes an ionotropic glutamate receptor known to be selectively expressed in cerebellar Purkinje cells. Here, we demonstrated GRID2 expression in human adult retina and retinal pigment epithelium. In addition, Grid2 expression was demonstrated in different stages of murine retinal development. GRID2 immunostaining was shown in murine and human retina. Whole-exome sequencing in the proband did not provide arguments for other disease-causing mutations, supporting the idea that the phenotype observed represents a single clinical entity. CONCLUSION: We identified GRID2 as an underlying disease gene of early-onset autosomal recessive cerebellar ataxia with retinal dystrophy, expanding the clinical spectrum of GRID2 deletion mutants. We demonstrated for the first time GRID2 expression and localization in human and murine retina, providing evidence for a novel functional role of GRID2 in the retina.


Subject(s)
Receptors, Glutamate/genetics , Retinal Dystrophies/genetics , Spinocerebellar Degenerations/genetics , Animals , Child, Preschool , DNA Copy Number Variations/genetics , Exons/genetics , Female , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Infant , Mice , Pedigree , Receptors, Glutamate/biosynthesis , Retina/metabolism , Retina/pathology , Retinal Dystrophies/complications , Retinal Dystrophies/pathology , Sequence Deletion , Spinocerebellar Degenerations/complications , Spinocerebellar Degenerations/pathology
9.
Genet Med ; 16(9): 671-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24625443

ABSTRACT

PURPOSE: Autosomal recessive retinal dystrophies are clinically and genetically heterogeneous, which hampers molecular diagnosis. We evaluated identity-by-descent-guided Sanger sequencing or whole-exome sequencing in 26 families with nonsyndromic (19) or syndromic (7) autosomal recessive retinal dystrophies to identify disease-causing mutations. METHODS: Patients underwent genome-wide identity-by-descent mapping followed by Sanger sequencing (16) or whole-exome sequencing (10). Whole-exome sequencing data were filtered against identity-by-descent regions and known retinal dystrophy genes. The medical history was reviewed in mutation-positive families. RESULTS: We identified mutations in 14 known retinal dystrophy genes in 20/26 (77%) families: ABCA4, CERKL, CLN3, CNNM4, C2orf71, IQCB1, LRAT, MERTK, NMNAT1, PCDH15, PDE6B, RDH12, RPGRIP1, and USH2A. Whole-exome sequencing in single individuals revealed mutations in either the largest or smaller identity-by-descent regions, and a compound heterozygous genotype in NMNAT1. Moreover, a novel deletion was found in PCDH15. In addition, we identified mutations in CLN3, CNNM4, and IQCB1 in patients initially diagnosed with nonsyndromic retinal dystrophies. CONCLUSION: Our study emphasized that identity-by-descent-guided mutation analysis and/or whole-exome sequencing are powerful tools for the molecular diagnosis of retinal dystrophy. Our approach uncovered unusual molecular findings and unmasked syndromic retinal dystrophies, guiding future medical management. Finally, elucidating ABCA4, LRAT, and MERTK mutations offers potential gene-specific therapeutic perspectives.


Subject(s)
Consanguinity , DNA Mutational Analysis , Exome , Mutation , Retinal Dystrophies/diagnosis , Retinal Dystrophies/genetics , Adolescent , Cadherin Related Proteins , Cadherins/genetics , Child , Child, Preschool , Female , Genes, Recessive , Genome-Wide Association Study , Homozygote , Humans , Male , Mutation, Missense , Ophthalmoscopes , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Tooth/pathology
10.
J Neuroophthalmol ; 34(2): 137-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24621862

ABSTRACT

Congenital fixed dilated pupils (congenital mydriasis) is characterized by hypoplasia or aplasia of the iris muscles, with absence of iris between the collarette and pupillary border, creating a scalloped pupillary margin. This condition has been reported in a multisystemic smooth muscle cell dysfunction syndrome, combined with congenital patent ductus arteriosus, cerebrovascular disease (Moya-moya-like), coronary artery disease, thoracic aorta aneurysm, and dysfunction of smooth muscle cells in organs throughout the body. All affected individuals carry a p.R179H heterozygous mutation in the ACTA2 gene. We add to the ophthalmologic involvement with 3 more patients. Congenital fixed dilated pupils is a rare condition and should alert ophthalmologists to the possibility of the coexistence of systemic life-threatening disorders.


Subject(s)
Actins/genetics , Muscle, Smooth/pathology , Muscular Diseases/pathology , Pupil Disorders/genetics , Pupil Disorders/pathology , Adolescent , Female , Humans , Magnetic Resonance Imaging , Muscular Diseases/complications , Muscular Diseases/genetics , Pupil Disorders/complications , Young Adult
11.
Am J Hum Genet ; 85(5): 730-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19896109

ABSTRACT

Congenital stationary night blindness (CSNB) is a clinically and genetically heterogeneous group of retinal disorders characterized by nonprogressive impaired night vision and variable decreased visual acuity. We report here that six out of eight female probands with autosomal-recessive complete CSNB (cCSNB) had mutations in TRPM1, a retinal transient receptor potential (TRP) cation channel gene. These data suggest that TRMP1 mutations are a major cause of autosomal-recessive CSNB in individuals of European ancestry. We localized TRPM1 in human retina to the ON bipolar cell dendrites in the outer plexifom layer. Our results suggest that in humans, TRPM1 is the channel gated by the mGluR6 (GRM6) signaling cascade, which results in the light-evoked response of ON bipolar cells. Finally, we showed that detailed electroretinography is an effective way to discriminate among patients with mutations in either TRPM1 or GRM6, another autosomal-recessive cCSNB disease gene. These results add to the growing importance of the diverse group of TRP channels in human disease and also provide new insights into retinal circuitry.


Subject(s)
Mutation , Night Blindness/congenital , Night Blindness/genetics , TRPM Cation Channels/genetics , Amino Acid Sequence , Case-Control Studies , Chromosome Deletion , Cohort Studies , Electroretinography/standards , Exons , Female , Genes, Recessive , Heterozygote , Homozygote , Humans , Models, Biological , Molecular Sequence Data , Mutation, Missense , Night Blindness/physiopathology , Nuclear Family , Retinal Rod Photoreceptor Cells/physiology , Signal Transduction , White People/genetics
12.
Am J Hum Genet ; 84(4): 493-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19327736

ABSTRACT

Nonsyndromic autosomal-recessive optic neuropathies are rare conditions of unknown genetic and molecular origin. Using an approach of whole-genome homozygosity mapping and positional cloning, we have identified the first gene, to our knowledge, responsible for this condition, TMEM126A, in a large multiplex inbred Algerian family and subsequently in three other families originating from the Maghreb. TMEM126A is conserved in higher eukaryotes and encodes a transmembrane mitochondrial protein of unknown function, supporting the view that mitochondrial dysfunction may be a hallmark of inherited optic neuropathies including isolated autosomal-recessive forms.


Subject(s)
Mitochondrial Proteins/genetics , Mutation , Optic Atrophies, Hereditary/genetics , Algeria , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Codon, Nonsense , Female , Gene Expression , Genes, Recessive , Haplotypes , Humans , Male , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/metabolism , Sequence Homology, Amino Acid , Transfection
13.
Genet Med ; 14(6): 576-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22261762

ABSTRACT

PURPOSE: Leber congenital amaurosis (LCA) is a rare congenital retinal dystrophy associated with 16 genes. Recent breakthroughs in LCA gene therapy offer the first prospect of treating inherited blindness, which requires an unequivocal and early molecular diagnosis. While present genetic tests do not address this due to a tremendous genetic heterogeneity, massively parallel sequencing (MPS) strategies might bring a solution. Here, we developed a comprehensive molecular test for LCA based on targeted MPS of all exons of 16 known LCA genes. METHODS: We designed a unique and flexible workflow for targeted resequencing of all 236 exons from 16 LCA genes based on quantitative PCR (qPCR) amplicon ligation, shearing, and parallel sequencing of multiple patients on a single lane of a short-read sequencer. Twenty-two prescreened LCA patients were included, five of whom had a known molecular cause. RESULTS: Validation of 107 variations was performed as proof of concept. In addition, the causal genetic defect and a single heterozygous mutation were identified in 3 and 5, respectively, of 17 patients without previously identified mutations. CONCLUSION: We propose a novel targeted MPS-based approach that is suitable for accurate, fast, and cost-effective early molecular testing in LCA, and easily applicable in other genetic disorders.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Leber Congenital Amaurosis/diagnosis , Molecular Diagnostic Techniques/methods , Adaptor Proteins, Signal Transducing , Antigens, Neoplasm/genetics , Biomarkers/analysis , Blindness/congenital , Blindness/genetics , Carrier Proteins/genetics , Case-Control Studies , Cell Cycle Proteins , Child , Child, Preschool , Consanguinity , Cytoskeletal Proteins , Exons/genetics , Eye Proteins/genetics , Genetic Heterogeneity , Guanylate Cyclase/genetics , Heterozygote , Homeodomain Proteins/genetics , Humans , Leber Congenital Amaurosis/genetics , Membrane Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Real-Time Polymerase Chain Reaction/methods , Receptors, Cell Surface/genetics , Trans-Activators/genetics , Validation Studies as Topic , cis-trans-Isomerases/genetics
14.
Mol Vis ; 17: 2072-9, 2011.
Article in English | MEDLINE | ID: mdl-21850183

ABSTRACT

PURPOSE: Optic nerve aplasia (ONA, OMIM 165550) is a very rare unilateral or bilateral condition that leads to blindness in the affected eye, and is usually associated with other ocular abnormalities. Although bilateral ONA often occurs in association with severe congenital anomalies of the brain, nonsyndromic sporadic forms with bilateral ONA have been described. So far, no autosomal-dominant nonsyndromic ONA has been reported. The genetic basis of this condition remains largely unknown, as no developmental genes other than paired box gene 6 (PAX6) are known to be implicated in sporadic bilateral ONA. METHODS: The individuals reported underwent extensive ophthalmological, endocrinological, and neurologic evaluation, including neuroimaging of the visual pathways. In addition genomewide copy number screening was performed. RESULTS: Here we report an autosomal-dominant form of nonsyndromic ONA in a Belgian pedigree, with unilateral microphthalmia and ONA in the second generation (II:1), and bilateral ONA in two sibs of the third generation (III:1; III:2). No PAX6 mutation was found. Genome wide copy number screening revealed a microdeletion of maximal 363 kb of chromosome 10q23.33q23.33 in all affected individuals (II:1, III:1; III:2) and in unaffected I:1, containing three genes: exocyst complex component 6 (EXOC6), cytochrome p450, subfamily XXVIA, polypeptide 1 (CYP26A1), and cytochrome p450, subfamily XXVIC, polypeptide 1 (CYP26C1). The latter two encode retinoic acid-degrading enzymes. CONCLUSIONS: This is the first study reporting an autosomal-dominant form of nonsyndromic ONA. The diagnostic value of neuroimaging in uncovering ONA in microphthalmic patients is demonstrated. Although involvement of other genetic factors cannot be ruled out, our study might point to a role of CYP26A1 and CYP26C1 in the pathogenesis of nonsyndromic ONA.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Eye Proteins/genetics , Microphthalmos/genetics , Optic Nerve , Asymptomatic Diseases , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 10/chemistry , Chromosomes, Human, Pair 10/genetics , Cytochrome P450 Family 26 , DNA Mutational Analysis , Female , Gene Dosage , Genes, Dominant , Genetic Linkage , Genome-Wide Association Study , Humans , Male , Microphthalmos/physiopathology , Middle Aged , Mutation , Neuroimaging , Optic Nerve/abnormalities , Optic Nerve/metabolism , Pedigree , Phenotype , Retinoic Acid 4-Hydroxylase , Tretinoin/metabolism , Vision Tests
15.
Hum Mutat ; 31(10): E1709-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20683928

ABSTRACT

Leber Congenital Amaurosis (LCA), the most severe inherited retinal dystrophy, is genetically heterogeneous, with 14 genes accounting for 70% of patients. Here, 91 LCA probands underwent LCA chip analysis and subsequent sequencing of 6 genes (CEP290, CRB1, RPE65, GUCY2D, AIPL1and CRX), revealing mutations in 69% of the cohort, with major involvement of CEP290 (30%). In addition, 11 patients with early-onset retinal dystrophy (EORD) and 13 patients with Senior-Loken syndrome (SLS), LCA-Joubert syndrome (LCA-JS) or cerebello-oculo-renal syndrome (CORS) were included. Exhaustive re-inspection of the overall phenotypes in our LCA cohort revealed novel insights mainly regarding the CEP290-related phenotype. The AHI1 gene was screened as a candidate modifier gene in three patients with the same CEP290 genotype but different neurological involvement. Interestingly, a heterozygous novel AHI1 mutation, p.Asn811Lys, was found in the most severely affected patient. Moreover, AHI1 screening in five other patients with CEP290-related disease and neurological involvement revealed a second novel missense variant, p.His758Pro, in one LCA patient with mild mental retardation and autism. These two AHI1 mutations might thus represent neurological modifiers of CEP290-related disease.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alleles , Antigens, Neoplasm/genetics , Genetic Testing , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Neoplasm Proteins/genetics , Adaptor Proteins, Vesicular Transport , Adolescent , Adult , Belgium , Cell Cycle Proteins , Child , Child, Preschool , Cytoskeletal Proteins , DNA Mutational Analysis/methods , Gene Expression Profiling , Genotype , Humans , Infant , Leber Congenital Amaurosis/diagnosis , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Phenotype , Proteins/genetics , Retinal Degeneration/genetics , Retinal Dystrophies/genetics , Retinal Dystrophies/pathology , Young Adult
16.
Ophthalmology ; 116(1): 154-162.e1, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004499

ABSTRACT

OBJECTIVE: To report a novel phenotype of autosomal dominant atypical congenital cataract associated with variable expression of microcornea, microphthalmia, and iris coloboma linked to chromosome 2. Molecular analysis of this phenotype may improve our understanding of anterior segment development. DESIGN: Observational case study, genome linkage analysis, and gene mutation screening. PARTICIPANTS: Three families, 1 Egyptian and 2 Belgians, with a total of 31 affected were studied. METHODS: Twenty-one affected subjects and 9 first-degree relatives underwent complete ophthalmic examination. In the Egyptian family, exclusion of PAX6, CRYAA, and MAF genes was demonstrated by haplotype analysis using microsatellite markers on chromosomes 11, 16, and 21. Genome-wide linkage analysis was then performed using 385 microsatellite markers on this family. In the 2 Belgian families, the PAX6 gene was screened for mutations by direct sequencing of all exons. MAIN OUTCOME MEASURES: Phenotype description, genome-wide linkage of the phenotype, linkage to the PAX6, CRYAA, and MAF genes, and mutation detection in the PAX6 gene. RESULTS: Affected members of the 3 families had bilateral congenital cataracts inherited in an autosomal dominant pattern. A novel form of hexagonal nuclear cataract with cortical riders was expressed. Among affected subjects with available data, 95% had microcornea, 39% had microphthalmia, and 38% had iris coloboma. Seventy-five percent of the colobomata were atypical, showing a nasal superior location in 56%. A positive lod score of 4.86 was obtained at theta = 0 for D2S2309 on chromosome 2, a 4.9-Mb common haplotype flanked by D2S2309 and D2S2358 was obtained in the Egyptian family, and linkage to the PAX6, CRYAA, or MAF gene was excluded. In the 2 Belgian families, sequencing of the junctions and all coding exons of PAX6 did not reveal any molecular change. CONCLUSIONS: We describe a novel phenotype that includes the combination of a novel form of congenital hexagonal cataract, with variably expressed microcornea, microphthalmia, and atypical iris coloboma, not caused by PAX6 and mapping to chromosome 2. FINANCIAL DISCLOSURE(S): The authors have no proprietary or commercial interest in any materials discussed in this article.


Subject(s)
Cataract/congenital , Chromosomes, Human, Pair 2/genetics , Coloboma/genetics , Cornea/abnormalities , Iris/abnormalities , Microphthalmos/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosome Mapping , Crystallins/genetics , DNA Mutational Analysis , Eye Proteins/genetics , Female , Genetic Linkage , Haplotypes , Homeodomain Proteins/genetics , Humans , Lod Score , MafF Transcription Factor/genetics , Male , Microsatellite Repeats , Middle Aged , Nuclear Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Pedigree , Repressor Proteins/genetics
17.
Hum Mutat ; 29(11): E205-19, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18642388

ABSTRACT

Blepharophimosis syndrome (BPES) is caused by loss-of-function mutations in the single-exon forkhead transcription factor gene FOXL2 and by genomic rearrangements of the FOXL2 locus. Here, we focus on 92 new intragenic FOXL2 mutations, 34 of which are novel. Specifically, we found 10 nonsense mutations (11%), 13 missense mutations (14%), 40 deletions or insertions leading to a frameshift (43%), and 29 in-frame changes (32%), of which 28 (30%) lead to a polyalanine expansion. This study confirms the existence of two previously described mutational hotspots. Moreover, we gained novel insights in genotype-phenotype correlations, emphasizing the need to interpret genotype-phenotype correlations individually and always in the context of further clinical observations.


Subject(s)
Blepharophimosis/genetics , Forkhead Transcription Factors/genetics , Frameshift Mutation , Mutation, Missense , Adolescent , Adult , Amino Acid Sequence , Child , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Eyelids/abnormalities , Female , Forkhead Box Protein L2 , Genotype , Humans , Infant , Infant, Newborn , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phenotype , Primary Ovarian Insufficiency/genetics , Sequence Alignment , Young Adult
18.
Am J Med Genet A ; 146A(8): 1038-41, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18348267

ABSTRACT

Type 4 Waardenburg syndrome represents a well define entity caused by neural crest derivatives anomalies (melanocytes, intrinsic ganglion cells, central, autonomous and peripheral nervous systems) leading, with variable expressivity, to pigmentary anomalies, deafness, mental retardation, peripheral neuropathy, and Hirschsprung disease. Autosomal dominant mode of inheritance is prevalent when Sox10 gene mutation is identified. We report the natural history of a child who presented with synophrys, vivid blue eye, deafness, bilateral complete semicircular canals agenesis with mental retardation, subtle signs for peripheral neuropathy and lack of Hirschsprung disease. SOX10 gene sequencing identified "de novo" splice site mutation (c.698-2A > C). The present phenotype and the genotype findings underline the wide spectrum of SOX10 gene implication in unusual type 4 Waardenburg syndrome patient.


Subject(s)
DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Hirschsprung Disease/physiopathology , Mutation , Transcription Factors/genetics , Waardenburg Syndrome/genetics , Waardenburg Syndrome/physiopathology , Child, Preschool , Genotype , Humans , Male , Phenotype , SOXE Transcription Factors , Sequence Analysis, DNA , Severity of Illness Index , Waardenburg Syndrome/diagnosis
19.
J Med Genet ; 44(5): 322-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17220209

ABSTRACT

Harboyan syndrome, or corneal dystrophy and perceptive deafness (CDPD), consists of congenital corneal endothelial dystrophy and progressive perceptive deafness, and is transmitted as an autosomal recessive trait. CDPD and autosomal recessive, non-syndromic congenital hereditary endothelial corneal dystrophy (CHED2) both map at overlapping loci at 20p13, and mutations of SLC4A11 were reported recently in CHED2. A genotype study on six families with CDPD and on one family with either CHED or CDPD, from various ethnic backgrounds (in the seventh family, hearing loss could not be assessed because of the proband's young age), is reported here. Novel SLC4A11 mutations were found in all patients. Why some mutations cause hearing loss in addition to corneal dystrophy is presently unclear. These findings extend the implication of the SLC4A11 borate transporter beyond corneal dystrophy to perceptive deafness.


Subject(s)
Abnormalities, Multiple/genetics , Anion Transport Proteins/genetics , Antiporters/genetics , Borates/metabolism , Corneal Dystrophies, Hereditary/genetics , Endothelium/abnormalities , Hearing Loss, Sensorineural/genetics , Mutation/genetics , Adolescent , Adult , Amino Acid Sequence , Anion Transport Proteins/chemistry , Antiporters/chemistry , Base Sequence , Child , Child, Preschool , DNA Mutational Analysis , Humans , Male , Molecular Sequence Data , Pedigree , Syndrome
20.
Invest Ophthalmol Vis Sci ; 47(3): 1167-76, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16505055

ABSTRACT

PURPOSE: To test the efficiency of a microarray chip as a diagnostic tool in a cohort of northwestern European patients with Leber congenital amaurosis (LCA) and to perform a genotype-phenotype analysis in patients in whom pathologic mutations were identified. METHODS: DNAs from 58 patients with LCA were analyzed using a microarray chip containing previously identified disease-associated sequence variants in six LCA genes. Mutations identified by chip analysis were confirmed by sequence analysis. On identification of one mutation, all protein coding exons of the relevant genes were sequenced. In addition, sequence analysis of the RDH12 gene was performed in 22 patients. Patients with mutations were phenotyped. RESULTS: Pathogenic mutations were identified in 19 of the 58 patients with LCA (32.8%). Four novel sequence variants were identified. Mutations were most frequently found in CRB1 (15.5%), followed by GUCY2D (10.3%). The p.R768W mutation was found in 8 of 10 GUCY2D alleles, suggesting that it is a founder mutation in the northwest of Europe. In early childhood, patients with AIPL1 or GUCY2D mutations show normal fundi. Those with AIPL1-associated LCA progress to an RP-like fundus before the age of 8, whereas patients with GUCY2D-associated LCA still have relatively normal fundi in their mid-20s. Patients with CRB1 mutations present with distinct fundus abnormalities at birth and consistently show characteristics of RP12. Pathogenic GUCY2D mutations result in the most severe form of LCA. CONCLUSIONS: Microarray-based mutation detection allowed the identification of 32% of LCA sequence variants and represents an efficient first-pass screening tool. Mutations in CRB1, and to a lesser extent, in GUCY2D, underlie most LCA cases in this cohort. The present study establishes a genotype-phenotype correlation for AIPL1, CRB1, and GUCY2D.


Subject(s)
Blindness/congenital , Blindness/genetics , Carrier Proteins/genetics , Eye Proteins/genetics , Guanylate Cyclase/genetics , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Receptors, Cell Surface/genetics , Adaptor Proteins, Signal Transducing , Alcohol Oxidoreductases/genetics , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Testing/methods , Genotype , Humans , Infant , Male , Phenotype , Retinitis Pigmentosa/congenital , Retinitis Pigmentosa/genetics , cis-trans-Isomerases
SELECTION OF CITATIONS
SEARCH DETAIL