Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Sci Rep ; 11(1): 117, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420188

ABSTRACT

We describe both phenotype and pathogenesis in two male siblings with typical retinitis pigmentosa (RP) and the potentially X-linked RP (XLRP) carrier phenotype in their mother. Two affected sons, two unaffected daughters, and their mother underwent detailed ophthalmological assessments including Goldmann perimetry, color vision testing, multimodal imaging and ISCEV-standard electroretinography. Genetic testing consisted of targeted next-generation sequencing (NGS) of known XLRP genes and whole exome sequencing (WES) of known inherited retinal disease genes (RetNet-WES). Variant validation and segregation analysis were performed by Sanger sequencing. The mutational load of the RHO variant in the mother was assessed in DNA from leucocytes, buccal cells and hair follicles using targeted NGS. Both affected sons showed signs of classical RP, while the mother displayed patches of hyperautofluorescence on blue light autofluorescence imaging and regional, intraretinal, spicular pigmentation, reminiscent of a carrier phenotype of XLRP. XLRP testing was negative. RetNet-WES testing revealed RHO variant c.404G > C p.(Arg135Pro) in a mosaic state (21% of the reads) in the mother and in a heterozygous state in both sons. Targeted NGQSS of the RHO variant in different maternal tissues showed a mutation load between 25.06% and 41.72%. We report for the first time that somatic mosaicism of RHO variant c.404G > C p.(Arg135Pro) mimics the phenotype of a female carrier of XLRP, in combination with heterozygosity for the variant in the two affected sons.


Subject(s)
Mosaicism , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Adult , Base Sequence , Female , Gene Dosage , Heterozygote , Humans , Male , Middle Aged , Phenotype , Point Mutation , Retinitis Pigmentosa/congenital , Retinitis Pigmentosa/metabolism , Rhodopsin/metabolism , Young Adult
2.
Hum Mutat ; 41(5): 998-1011, 2020 05.
Article in English | MEDLINE | ID: mdl-31999394

ABSTRACT

Inactivating variants in the centrosomal CEP78 gene have been found in cone-rod dystrophy with hearing loss (CRDHL), a particular phenotype distinct from Usher syndrome. Here, we identified and functionally characterized the first CEP78 missense variant c.449T>C, p.(Leu150Ser) in three CRDHL families. The variant was found in a biallelic state in two Belgian families and in a compound heterozygous state-in trans with c.1462-1G>T-in a third German family. Haplotype reconstruction showed a founder effect. Homology modeling revealed a detrimental effect of p.(Leu150Ser) on protein stability, which was corroborated in patients' fibroblasts. Elongated primary cilia without clear ultrastructural abnormalities in sperm or nasal brushes suggest impaired cilia assembly. Two affected males from different families displayed sperm abnormalities causing infertility. One of these is a heterozygous carrier of a complex allele in SPAG17, a ciliary gene previously associated with autosomal recessive male infertility. Taken together, our data indicate that a missense founder allele in CEP78 underlies the same sensorineural CRDHL phenotype previously associated with inactivating variants. Interestingly, the CEP78 phenotype has been possibly expanded with male infertility. Finally, CEP78 loss-of-function variants may have an underestimated role in misdiagnosed Usher syndrome, with or without sperm abnormalities.


Subject(s)
Alleles , Cell Cycle Proteins/genetics , Cone-Rod Dystrophies/genetics , Founder Effect , Hearing Loss/genetics , Infertility, Male/genetics , Mutation, Missense , Adolescent , Cell Cycle Proteins/chemistry , Cilia/metabolism , Cilia/ultrastructure , Cone-Rod Dystrophies/diagnosis , DNA Mutational Analysis , Female , Fibroblasts/metabolism , Genotype , Hearing Loss/diagnosis , Humans , Infertility, Male/diagnosis , Male , Middle Aged , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship , Syndrome , Exome Sequencing
3.
Clin Genet ; 97(3): 426-436, 2020 03.
Article in English | MEDLINE | ID: mdl-31721179

ABSTRACT

Biallelic MFSD8 variants are an established cause of severe late-infantile subtype of neuronal ceroid lipofuscinosis (v-LINCL), a severe lysosomal storage disorder, but have also been associated with nonsyndromic adult-onset maculopathy. Here, we functionally characterized two novel MFSD8 variants found in a child with juvenile isolated maculopathy, in order to establish a refined prognosis. ABCA4 locus resequencing was followed by the analysis of other inherited retinal disease genes by whole exome sequencing (WES). Minigene assays and cDNA sequencing were used to assess the effect of a novel MFSD8 splice variant. MFSD8 expression was quantified with qPCR and overexpression studies were analyzed by immunoblotting. Transmission electron microscopy (TEM) was performed on a skin biopsy and ophthalmological and neurological re-examinations were conducted. WES revealed two novel MFSD8 variants: c.[590del];[439+3A>C] p.[Gly197Valfs*2];[Ile67Glufs*3]. Characterization of the c.439+3A>C variant via splice assays showed exon-skipping (p.Ile67Glufs*3), while overexpression studies of the corresponding protein indicated expression of a truncated polypeptide. In addition, a significantly reduced MFSD8 RNA expression was noted in patient's lymphocytes. TEM of a skin biopsy revealed typical v-LINCL lipopigment inclusions while neurological imaging of the proband displayed subtle cerebellar atrophy. Functional characterization demonstrated the pathogenicity of two novel MFSD8 variants, found in a child with an initial diagnosis of juvenile isolated maculopathy but likely evolving to v-LINCL with a protracted disease course. Our study allowed a refined neurological prognosis in the proband and expands the natural history of MFSD8-associated disease.


Subject(s)
Macular Degeneration/genetics , Membrane Transport Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Child , Female , Genetic Variation , Homozygote , Humans , Macular Degeneration/diagnostic imaging , Macular Degeneration/physiopathology , Microscopy, Electron, Transmission , Mutation , Neuronal Ceroid-Lipofuscinoses/physiopathology , Retina/diagnostic imaging , Retina/physiopathology , Exome Sequencing
4.
Genet Med ; 21(8): 1761-1771, 2019 08.
Article in English | MEDLINE | ID: mdl-30670881

ABSTRACT

PURPOSE: ABCA4-associated disease, a recessive retinal dystrophy, is hallmarked by a large proportion of patients with only one pathogenic ABCA4 variant, suggestive for missing heritability. METHODS: By locus-specific analysis of ABCA4, combined with extensive functional studies, we aimed to unravel the missing alleles in a cohort of 67 patients (p), with one (p = 64) or no (p = 3) identified coding pathogenic variants of ABCA4. RESULTS: We identified eight pathogenic (deep-)intronic ABCA4 splice variants, of which five are novel and six structural variants, four of which are novel, including two duplications. Together, these variants account for the missing alleles in 40.3% of patients. Furthermore, two novel variants with a putative cis-regulatory effect were identified. The common hypomorphic variant c.5603A>T p.(Asn1868Ile) was found as a candidate second allele in 43.3% of patients. Overall, we have elucidated the missing heritability in 83.6% of our cohort. In addition, we successfully rescued three deep-intronic variants using antisense oligonucleotide (AON)-mediated treatment in HEK 293-T cells and in patient-derived fibroblast cells. CONCLUSION: Noncoding pathogenic variants, novel structural variants, and a common hypomorphic allele of the ABCA4 gene explain the majority of unsolved cases with ABCA4-associated disease, rendering this retinopathy a model for missing heritability in autosomal recessive disorders.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Genes, Recessive/genetics , Oligonucleotides, Antisense/genetics , Retinal Dystrophies/genetics , Adult , Alleles , Cohort Studies , Exons/genetics , Female , Gene Frequency , HEK293 Cells , Humans , Introns/genetics , Male , Middle Aged , Mutation/genetics , Oligonucleotides, Antisense/pharmacology , Pedigree , Phenotype , Retinal Dystrophies/pathology
5.
PLoS One ; 12(1): e0170038, 2017.
Article in English | MEDLINE | ID: mdl-28076437

ABSTRACT

PURPOSE: Autosomal dominant retinitis pigmentosa (adRP) is characterized by an extensive genetic heterogeneity, implicating 27 genes, which account for 50 to 70% of cases. Here 86 Belgian probands with possible adRP underwent genetic testing to unravel the molecular basis and to assess the contribution of the genes underlying their condition. METHODS: Mutation detection methods evolved over the past ten years, including mutation specific methods (APEX chip analysis), linkage analysis, gene panel analysis (Sanger sequencing, targeted next-generation sequencing or whole exome sequencing), high-resolution copy number screening (customized microarray-based comparative genomic hybridization). Identified variants were classified following American College of Medical Genetics and Genomics (ACMG) recommendations. RESULTS: Molecular genetic screening revealed mutations in 48/86 cases (56%). In total, 17 novel pathogenic mutations were identified: four missense mutations in RHO, five frameshift mutations in RP1, six mutations in genes encoding spliceosome components (SNRNP200, PRPF8, and PRPF31), one frameshift mutation in PRPH2, and one frameshift mutation in TOPORS. The proportion of RHO mutations in our cohort (14%) is higher than reported in a French adRP population (10.3%), but lower than reported elsewhere (16.5-30%). The prevalence of RP1 mutations (10.5%) is comparable to other populations (3.5%-10%). The mutation frequency in genes encoding splicing factors is unexpectedly high (altogether 19.8%), with PRPF31 the second most prevalent mutated gene (10.5%). PRPH2 mutations were found in 4.7% of the Belgian cohort. Two families (2.3%) have the recurrent NR2E3 mutation p.(Gly56Arg). The prevalence of the recurrent PROM1 mutation p.(Arg373Cys) was higher than anticipated (3.5%). CONCLUSIONS: Overall, we identified mutations in 48 of 86 Belgian adRP cases (56%), with the highest prevalence in RHO (14%), RP1 (10.5%) and PRPF31 (10.5%). Finally, we expanded the molecular spectrum of PRPH2, PRPF8, RHO, RP1, SNRNP200, and TOPORS-associated adRP by the identification of 17 novel mutations.


Subject(s)
Mutation , RNA Splicing Factors/genetics , Adult , Aged , Belgium , Cohort Studies , DNA Mutational Analysis , Eye Proteins/genetics , Family , Female , Gene Frequency , Genes, Dominant , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Retinitis Pigmentosa/genetics
6.
Genet Med ; 19(4): 457-466, 2017 04.
Article in English | MEDLINE | ID: mdl-27608171

ABSTRACT

PURPOSE: Our goal was to design a customized microarray, arrEYE, for high-resolution copy number variant (CNV) analysis of known and candidate genes for inherited retinal dystrophy (iRD) and retina-expressed noncoding RNAs (ncRNAs). METHODS: arrEYE contains probes for the full genomic region of 106 known iRD genes, including those implicated in retinitis pigmentosa (RP) (the most frequent iRD), cone-rod dystrophies, macular dystrophies, and an additional 60 candidate iRD genes and 196 ncRNAs. Eight CNVs in iRD genes identified by other techniques were used as positive controls. The test cohort consisted of 57 patients with autosomal dominant, X-linked, or simplex RP. RESULTS: In an RP patient, a novel heterozygous deletion of exons 7 and 8 of the HGSNAT gene was identified: c.634-408_820+338delinsAGAATATG, p.(Glu212Glyfs*2). A known variant was found on the second allele: c.1843G>A, p.(Ala615Thr). Furthermore, we expanded the allelic spectrum of USH2A and RCBTB1 with novel CNVs. CONCLUSION: The arrEYE platform revealed subtle single-exon to larger CNVs in iRD genes that could be characterized at the nucleotide level, facilitated by the high resolution of the platform. We report the first CNV in HGSNAT that, combined with another mutation, leads to RP, further supporting its recently identified role in nonsyndromic iRD.Genet Med 19 4, 457-466.


Subject(s)
Comparative Genomic Hybridization/methods , DNA Copy Number Variations , Oligonucleotide Array Sequence Analysis/methods , Retinal Dystrophies/genetics , Acetyltransferases/genetics , Extracellular Matrix Proteins/genetics , Female , Guanine Nucleotide Exchange Factors/genetics , Humans , Male , RNA, Untranslated/genetics , Sequence Deletion
7.
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
8.
Invest Ophthalmol Vis Sci ; 52(1): 324-33, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-20881294

ABSTRACT

PURPOSE: Anterior segment dysgenesis (ASD) comprises a heterogeneous group of developmental abnormalities that affect several structures of the anterior segment of the eye. The main purpose of this study was to assess the proportion of FOXC1 and PITX2 mutations and copy number changes in 80 probands with ASD. METHODS: The patients were examined for FOXC1 and PITX2 copy number changes and mutations using MLPA (multiplex ligation-dependent probe amplification) and direct sequencing. Subsequently, the identified copy number changes were fine-mapped using high-resolution microarrays. In the remaining mutation-negative patients, sequencing of the FOXC1 andPITX2 3' untranslated regions (UTRs) and three other candidate genes (P32, PDP2, and FOXC2) was performed. RESULTS: Thirteen FOXC1 and eight PITX2 mutations were identified, accounting for 26% (21/80) of the cases. In addition, six FOXC1 and five PITX2 deletions were found, explaining 14% (11/80) of the cases. The smallest FOXC1 and PITX2 deletions were 5.4 and 1.6 kb in size, respectively. Six patients carrying FOXC1 deletions presented with variable extraocular phenotypic features such as hearing defects (in 4/6) and mental retardation (in 2/6). No further genetic defects were found in the remaining mutation-negative patients. CONCLUSIONS: FOXC1 and PITX2 genetic defects explain 40% of our large ASD cohort. The current spectrum of intragenic FOXC1 and PITX2 mutations was extended considerably, the identified copy number changes were fine mapped, the smallest FOXC1 and PITX2 deletions reported so far were identified, and the need for dedicated copy number screening of the FOXC1 and PITX2 genomic landscape was emphasized. This study is unique in that sequence and copy number changes were screened simultaneously in both genes.


Subject(s)
Anterior Eye Segment/abnormalities , Eye Abnormalities/genetics , Forkhead Transcription Factors/genetics , Gene Dosage/genetics , Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , 3' Untranslated Regions , Adolescent , Adult , Carrier Proteins , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Mitochondrial Proteins/genetics , Nucleic Acid Amplification Techniques , Polymerase Chain Reaction , Homeobox Protein PITX2
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...