Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Publication year range
1.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
2.
Hum Mol Genet ; 33(11): 945-957, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38453143

ABSTRACT

Inherited retinal diseases (IRDs) are a group of rare genetic eye conditions that cause blindness. Despite progress in identifying genes associated with IRDs, improvements are necessary for classifying rare autosomal dominant (AD) disorders. AD diseases are highly heterogenous, with causal variants being restricted to specific amino acid changes within certain protein domains, making AD conditions difficult to classify. Here, we aim to determine the top-performing in-silico tools for predicting the pathogenicity of AD IRD variants. We annotated variants from ClinVar and benchmarked 39 variant classifier tools on IRD genes, split by inheritance pattern. Using area-under-the-curve (AUC) analysis, we determined the top-performing tools and defined thresholds for variant pathogenicity. Top-performing tools were assessed using genome sequencing on a cohort of participants with IRDs of unknown etiology. MutScore achieved the highest accuracy within AD genes, yielding an AUC of 0.969. When filtering for AD gain-of-function and dominant negative variants, BayesDel had the highest accuracy with an AUC of 0.997. Five participants with variants in NR2E3, RHO, GUCA1A, and GUCY2D were confirmed to have dominantly inherited disease based on pedigree, phenotype, and segregation analysis. We identified two uncharacterized variants in GUCA1A (c.428T>A, p.Ile143Thr) and RHO (c.631C>G, p.His211Asp) in three participants. Our findings support using a multi-classifier approach comprised of new missense classifier tools to identify pathogenic variants in participants with AD IRDs. Our results provide a foundation for improved genetic diagnosis for people with IRDs.


Subject(s)
Computer Simulation , Pedigree , Retinal Diseases , Humans , Retinal Diseases/genetics , Female , Male , Mutation , Genes, Dominant , Genetic Predisposition to Disease , Computational Biology/methods , Phenotype , Adult
3.
Hum Mol Genet ; 32(12): 2005-2015, 2023 06 05.
Article in English | MEDLINE | ID: mdl-36811936

ABSTRACT

Despite increasing success in determining genetic diagnosis for patients with inherited retinal diseases (IRDs), mutations in about 30% of the IRD cases remain unclear or unsettled after targeted gene panel or whole exome sequencing. In this study, we aimed to investigate the contributions of structural variants (SVs) to settling the molecular diagnosis of IRD with whole-genome sequencing (WGS). A cohort of 755 IRD patients whose pathogenic mutations remain undefined were subjected to WGS. Four SV calling algorithms including include MANTA, DELLY, LUMPY and CNVnator were used to detect SVs throughout the genome. All SVs identified by any one of these four algorithms were included for further analysis. AnnotSV was used to annotate these SVs. SVs that overlap with known IRD-associated genes were examined with sequencing coverage, junction reads and discordant read pairs. Polymerase Chain Reaction (PCR) followed by Sanger sequencing was used to further confirm the SVs and identify the breakpoints. Segregation of the candidate pathogenic alleles with the disease was performed when possible. A total of 16 candidate pathogenic SVs were identified in 16 families, including deletions and inversions, representing 2.1% of patients with previously unsolved IRDs. Autosomal dominant, autosomal recessive and X-linked inheritance of disease-causing SVs were observed in 12 different genes. Among these, SVs in CLN3, EYS and PRPF31 were found in multiple families. Our study suggests that the contribution of SVs detected by short-read WGS is about 0.25% of our IRD patient cohort and is significantly lower than that of single nucleotide changes and small insertions and deletions.


Subject(s)
Retinal Diseases , Humans , Retinal Diseases/genetics , Mutation , Whole Genome Sequencing , Exome Sequencing , Alleles , Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Eye Proteins/genetics
4.
Genet Med ; 26(6): 101106, 2024 06.
Article in English | MEDLINE | ID: mdl-38420906

ABSTRACT

PURPOSE: Inherited retinal diseases (IRDs) are a group of monogenic conditions that can lead to progressive blindness. Their missing heritability is still considerable, due in part to the presence of disease genes that await molecular identification. The purpose of this work was to identify novel genetic associations with IRDs. METHODS: Patients underwent a comprehensive ophthalmological evaluation using standard-of-care tests, such as detailed retinal imaging (macular optical coherence tomography and short-wavelength fundus autofluorescence) and electrophysiological testing. Exome and genome sequencing, as well as computer-assisted data analysis were used for genotyping and detection of DNA variants. A minigene-driven splicing assay was performed to validate the deleterious effects of 1 of such variants. RESULTS: We identified 8 unrelated families from Hungary, the United States, Israel, and The Netherlands with members presenting with a form of autosomal recessive and nonsyndromic retinal degeneration, predominantly described as rod-cone dystrophy but also including cases of cone/cone-rod dystrophy. Age of disease onset was very variable, with some patients experiencing first symptoms during their fourth decade of life or later. Myopia greater than 5 diopters was present in 5 of 7 cases with available refractive data, and retinal detachment was reported in 2 cases. All ascertained patients carried biallelic loss-of-function variants in UBAP1L (HGNC: 40028), a gene with unknown function and with homologies to UBAP1, encoding a protein involved in ubiquitin metabolism. One of these pathogenic variants, the intronic NM_001163692.2:c.910-7G>A substitution, was identified in 5 unrelated families. Minigene-driven splicing assays in HEK293T cells confirmed that this DNA change is responsible for the creation of a new acceptor splice site, resulting in aberrant splicing. CONCLUSION: We identified UBAP1L as a novel IRD gene. Although its function is currently unknown, UBAP1L is almost exclusively expressed in photoreceptors and the retinal pigment epithelium, hence possibly explaining the link between pathogenic variants in this gene and an ocular phenotype.


Subject(s)
Pedigree , Retinal Degeneration , Humans , Male , Female , Adult , Retinal Degeneration/genetics , Middle Aged , Loss of Function Mutation , Genes, Recessive , Child , Adolescent , Cone-Rod Dystrophies/genetics , Hungary , Young Adult , Genetic Predisposition to Disease
5.
Am J Hum Genet ; 107(5): 802-814, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33022222

ABSTRACT

The cause of autosomal-dominant retinitis pigmentosa (adRP), which leads to loss of vision and blindness, was investigated in families lacking a molecular diagnosis. A refined locus for adRP on Chr17q22 (RP17) was delineated through genotyping and genome sequencing, leading to the identification of structural variants (SVs) that segregate with disease. Eight different complex SVs were characterized in 22 adRP-affected families with >300 affected individuals. All RP17 SVs had breakpoints within a genomic region spanning YPEL2 to LINC01476. To investigate the mechanism of disease, we reprogrammed fibroblasts from affected individuals and controls into induced pluripotent stem cells (iPSCs) and differentiated them into photoreceptor precursor cells (PPCs) or retinal organoids (ROs). Hi-C was performed on ROs, and differential expression of regional genes and a retinal enhancer RNA at this locus was assessed by qPCR. The epigenetic landscape of the region, and Hi-C RO data, showed that YPEL2 sits within its own topologically associating domain (TAD), rich in enhancers with binding sites for retinal transcription factors. The Hi-C map of RP17 ROs revealed creation of a neo-TAD with ectopic contacts between GDPD1 and retinal enhancers, and modeling of all RP17 SVs was consistent with neo-TADs leading to ectopic retinal-specific enhancer-GDPD1 accessibility. qPCR confirmed increased expression of GDPD1 and increased expression of the retinal enhancer that enters the neo-TAD. Altered TAD structure resulting in increased retinal expression of GDPD1 is the likely convergent mechanism of disease, consistent with a dominant gain of function. Our study highlights the importance of SVs as a genomic mechanism in unsolved Mendelian diseases.


Subject(s)
Chromosomes, Human, Pair 17/chemistry , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Transcription Factors/genetics , Adult , Amino Acid Sequence , Cell Differentiation , Cellular Reprogramming , Child , Chromosome Mapping , Cohort Studies , Enhancer Elements, Genetic , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Genes, Dominant , Genome, Human , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Nuclear Proteins/metabolism , Organoids/metabolism , Organoids/pathology , Phosphoric Diester Hydrolases/metabolism , Polymorphism, Genetic , Primary Cell Culture , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Transcription Factors/metabolism , Whole Genome Sequencing
6.
Ophthalmology ; 130(12): 1313-1326, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37541626

ABSTRACT

PURPOSE: Individuals with Zellweger spectrum disorder (ZSD) manifest a spectrum of clinical phenotypes but almost all have retinal degeneration leading to blindness. The onset, extent, and progression of retinal findings have not been well described. It is crucial to understand the natural history of vision loss in ZSD to define reliable endpoints for future interventional trials. Herein, we describe ophthalmic findings in the largest number of ZSD patients to date. DESIGN: Retrospective review of longitudinal data from medical charts and review of cross-sectional data from the literature. PARTICIPANTS: Sixty-six patients with ZSD in the retrospective cohort and 119 patients reported in the literature, divided into 4 disease phenotypes based on genotype or clinical severity. METHODS: We reviewed ophthalmology records collected from the retrospective cohort (Clinicaltrials.gov NCT01668186) and performed a scoping review of the literature for ophthalmic findings in patients with ZSD. We extracted available ophthalmic data and analyzed by age and disease severity. MAIN OUTCOME MEASURES: Visual acuity (VA), posterior and anterior segment descriptions, nystagmus, refraction, electroretinography findings, visual evoked potentials, and OCT results and images. RESULTS: Visual acuity was worse at younger ages in those with severe disease compared with older patients with intermediate to mild disease for all 78 participants analyzed, with a median VA of 0.93 logarithm of the minimum angle of resolution (Snellen 20/320). Longitudinal VA data revealed slow loss over time and legal blindness onset at an average age of 7.8 years. Funduscopy showed retinal pigmentation, macular abnormalities, small or pale optic discs, and attenuated vessels with higher prevalence in milder severity groups and did not change with age. Electroretinography waveforms were diminished in 91% of patients, 46% of which were extinguished and did not change with age. OCT in milder patients revealed schitic changes in 18 of 23 individuals (age range 1.8 to 30 years), with evolution or stable macular edema. CONCLUSIONS: In ZSD, VA slowly deteriorates and is associated with disease severity, serial electroretinography is not useful for documenting vision loss progression, and intraretinal schitic changes may be common. Multiple systematic measures are required to assess retinal dystrophy accurately in ZSD, including functional vision measures. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Subject(s)
Evoked Potentials, Visual , Zellweger Syndrome , Humans , Child , Infant , Child, Preschool , Adolescent , Young Adult , Adult , Cross-Sectional Studies , Retrospective Studies , Blindness , Retina
7.
Genet Med ; 24(7): 1523-1535, 2022 07.
Article in English | MEDLINE | ID: mdl-35486108

ABSTRACT

PURPOSE: This study aimed to investigate the clinical and genetic aspects of solute carrier (SLC) genes in inherited retinal diseases (IRDs). METHODS: Exome sequencing data were filtered to identify pathogenic variants in SLC genes. Analysis of transcript and protein expression was performed on fibroblast cell lines and retinal sections. RESULTS: Comprehensive analysis of 433 SLC genes in 913 exome sequencing IRD samples revealed homozygous pathogenic variants in 6 SLC genes, including 2 candidate novel genes, which were 2 variants in SLC66A1, causing autosomal recessive retinitis pigmentosa (ARRP), and a variant in SLC39A12, causing autosomal recessive mild widespread retinal degeneration with marked macular involvement. In addition, we present 4 families with ARRP and homozygous null variants in SLC37A3 that were previously suggested to cause retinitis pigmentosa, 2 of which cause exon skipping. The recently reported SLC4A7- c.2007dup variant was found in 2 patients with ARRP resulting in the absence of protein. Finally, variants in SLC24A1 were found in 4 individuals with either ARRP or congenital stationary night blindness. CONCLUSION: We report on SLC66A1 and SLC39A12 as candidate novel IRD genes, establish SLC37A3 pathogenicity, and provide further evidence of SLC4A7 as IRD genes. We extend the phenotypic spectrum of SLC24A1 and suggest that its ARRP phenotype may be more common than previously reported.


Subject(s)
Retinitis Pigmentosa , DNA Mutational Analysis/methods , Genes, Recessive , Genetic Association Studies , Humans , Mutation , Pedigree , Phenotype , Retinitis Pigmentosa/genetics
8.
Mol Vis ; 27: 95-106, 2021.
Article in English | MEDLINE | ID: mdl-33907365

ABSTRACT

Purpose: Despite the extensive use of next-generation sequencing (NGS) technology to identify disease-causing genomic variations, a major gap in our understanding of Mendelian diseases is the unidentified molecular lesion in a significant portion of patients. For inherited retinal degenerations (IRDs), although currently close to 300 disease-associated genes have been identified, the mutations in approximately one-third of patients remain unknown. With mounting evidence that noncoding mutations might contribute significantly to disease burden, we aimed to systematically investigate the contributions of noncoding regions in the genome to IRDs. Methods: In this study, we focused on RPGRIP1, which has been linked to various IRD phenotypes, including Leber congenital amaurosis (LCA), retinitis pigmentosa (RP), and macular dystrophy (MD). As several noncoding mutant alleles have been reported in RPGRIP1, and we observed that the mutation carrier frequency of RPGRIP1 is higher in patient cohorts with unsolved IRDs, we hypothesized that mutations in the noncoding regions of RPGRIP1 might be a significant contributor to pathogenicity. To test this hypothesis, we performed whole-genome sequencing (WGS) for 25 patients with unassigned IRD who carry a single mutation in RPGRIP1. Results: Three noncoding variants in RPGRIP1, including a 2,890 bp deletion and two deep-intronic variants (c.2710+233G>A and c.1468-263G>C), were identified as putative second hits of RPGRIP1 in three patients with LCA. The mutant alleles were validated with direct sequencing or in vitro assays. Conclusions: The results highlight the significance of the contribution of noncoding pathogenic variants to unsolved IRD cases.


Subject(s)
Cytoskeletal Proteins/genetics , Mutation/genetics , RNA, Untranslated/genetics , Retinal Degeneration/genetics , Adult , Alleles , Child, Preschool , Cloning, Molecular , Electroretinography , Female , Humans , Male , Phenotype , Real-Time Polymerase Chain Reaction , Retina/physiopathology , Retinal Degeneration/diagnosis , Retinal Degeneration/physiopathology , Tomography, Optical Coherence , Transfection , Visual Acuity/physiology , Whole Genome Sequencing
9.
Retina ; 41(5): 898-907, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33595255

ABSTRACT

PURPOSE: Leber congenital amaurosis due to CEP290 mutations (LCA10) is an inherited retinal disease that often results in severe visual impairment or blindness in early childhood. Currently, there are no approved treatments, highlighting the considerable unmet medical need associated with LCA10. We aimed to review the clinical characteristics of LCA10, its impact on patients and society, and the investigational treatment strategies currently in development. METHODS: Review of the current literature. RESULTS: LCA10 is an autosomal recessive ciliopathy, for which the CEP290 intronic variant c.2991+1655A>G (p.Cys998X) is the most common mutation. Usually diagnosed in early childhood, most patients with LCA10 have severe visual impairment during their first decade of life, which significantly affects the quality of life and development. LCA10 also has a significant societal burden (direct and indirect costs). RNA editing using antisense oligonucleotides or Staphylococcus aureus CRISPR-associated protein-9 nuclease is currently under investigation for treatment of p.Cys998X LCA10. Specifically, the antisense oligonucleotide therapy QR-110 (sepofarsen) has demonstrated encouraging safety and efficacy data in a first-in-human trial; a phase 3 clinical trial is ongoing. CONCLUSION: Interventions that can preserve or improve vision in patients with LCA10 have considerable potential to improve the patient quality of life and reduce burden of disease.


Subject(s)
Antigens, Neoplasm/genetics , Blindness/etiology , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , DNA/genetics , Disease Management , Health Services Needs and Demand/standards , Leber Congenital Amaurosis/genetics , Antigens, Neoplasm/metabolism , Blindness/diagnosis , Blindness/therapy , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , DNA Mutational Analysis , Humans , Leber Congenital Amaurosis/complications
10.
PLoS Pathog ; 13(1): e1006174, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28118396

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1005772.].

11.
Am J Med Genet A ; 179(11): 2233-2236, 2019 11.
Article in English | MEDLINE | ID: mdl-31390148

ABSTRACT

Aymé-Gripp syndrome is an intellectual disability syndrome characterized by autism spectrum disorder, cataracts, sensorineural hearing loss, skeletal involvement, seizures, cardiac anomalies, and distinctive facial features. The condition is caused by pathogenic variants in MAF. To date, less than 20 cases have been reported, the majority having de novo mutations. Here, we report a patient with classical features of Aymé-Gripp syndrome who inherited a MAF variant, c.206C>G (p.P69R), from a mother with normal intellectual function and normal hearing but with cataract and significant proteinuria. To the best of our knowledge, this is the first report of a patient who inherited a MAF causative variant from a parent with normal intellect. Although the syndrome typically has multiple malformations and intellectual disability, we suggest that a mild phenotype could exist. In addition, we suggest that the basal ganglia calcifications present in our proband could be a novel finding associated with MAF variants and offer further support for the relationship between these variants and late manifestations of renal disease.


Subject(s)
Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Variation , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Maternal Inheritance , Proto-Oncogene Proteins c-maf/genetics , Adolescent , Brain/abnormalities , Brain/diagnostic imaging , Genetic Association Studies , Humans , Male , Phenotype , Syndrome
12.
Mol Ther ; 26(6): 1581-1593, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29673930

ABSTRACT

Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.


Subject(s)
Blindness/metabolism , Blindness/therapy , Dependovirus/genetics , Genetic Therapy/methods , Animals , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/therapy , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
13.
Hum Mol Genet ; 25(8): 1479-88, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26908613

ABSTRACT

A growing number of human diseases have been linked to defects in protein glycosylation that affects a wide range of organs. Among them, O-mannosylation is an unusual type of protein glycosylation that is largely restricted to the muscular and nerve system. Consistently, mutations in genes involved in the O-mannosylation pathway result in infantile-onset, severe developmental defects involving skeleton muscle, brain and eye, such as the muscle-eye-brain disease (MIM no. 253280). However, the functional importance of O-mannosylation in these tissues at later stages remains largely unknown. In our study, we have identified recessive mutations in POMGNT1, which encodes an essential component in O-mannosylation pathway, in three unrelated families with autosomal recessive retinitis pigmentosa (RP), but without extraocular involvement. Enzymatic assay of these mutant alleles demonstrate that they greatly reduce the POMGNT1 enzymatic activity and are likely to be hypomorphic. Immunohistochemistry shows that POMGNT1 is specifically expressed in photoreceptor basal body. Taken together, our work identifies a novel disease-causing gene for RP and indicates that proper protein O-mannosylation is not only essential for early organ development, but also important for maintaining survival and function of the highly specialized retinal cells at later stages.


Subject(s)
Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Retinitis Pigmentosa/pathology , Sequence Analysis, DNA/methods , Adult , Aged , Animals , Cells, Cultured , Exome , Female , Genes, Recessive , Genetic Predisposition to Disease , Glycosylation , Humans , Male , Mice , Middle Aged , Pedigree , Photoreceptor Cells, Vertebrate/metabolism , Retinitis Pigmentosa/genetics
14.
PLoS Pathog ; 12(7): e1005772, 2016 07.
Article in English | MEDLINE | ID: mdl-27454487

ABSTRACT

Spliceosomal SNRNP200 is a Ski2-like RNA helicase that is associated with retinitis pigmentosa 33 (RP33). Here we found that SNRNP200 promotes viral RNA sensing and IRF3 activation through the ability of its amino-terminal Sec63 domain (Sec63-1) to bind RNA and to interact with TBK1. We show that SNRNP200 relocalizes into TBK1-containing cytoplasmic structures upon infection, in contrast to the RP33-associated S1087L mutant, which is also unable to rescue antiviral response of SNRNP200 knockdown cells. This functional rescue correlates with the Sec63-1-mediated binding of viral RNA. The hindered IFN-ß production of knockdown cells was further confirmed in peripheral blood cells of RP33 patients bearing missense mutation in SNRNP200 upon infection with Sendai virus (SeV). This work identifies a novel immunoregulatory role of the spliceosomal SNRNP200 helicase as an RNA sensor and TBK1 adaptor for the activation of IRF3-mediated antiviral innate response.


Subject(s)
Immunity, Innate/immunology , Interferon Regulatory Factor-3/immunology , RNA, Viral/immunology , Ribonucleoproteins, Small Nuclear/immunology , Virus Diseases/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , Spliceosomes/immunology
15.
Hum Mutat ; 38(11): 1521-1533, 2017 11.
Article in English | MEDLINE | ID: mdl-28714225

ABSTRACT

The genetic heterogeneity of Mendelian disorders results in a significant proportion of patients that are unable to be assigned a confident molecular diagnosis after conventional exon sequencing and variant interpretation. Here, we evaluated how many patients with an inherited retinal disease (IRD) have variants of uncertain significance (VUS) that are disrupting splicing in a known IRD gene by means other than affecting the canonical dinucleotide splice site. Three in silico splice-affecting variant predictors were leveraged to annotate and prioritize variants for splicing functional validation. An in vitro minigene system was used to assay each variant's effect on splicing. Starting with 745 IRD patients lacking a confident molecular diagnosis, we validated 23 VUS as splicing variants that likely explain disease in 26 patients. Using our results, we optimized in silico score cutoffs to guide future variant interpretation. Variants that alter base pairs other than the canonical GT-AG dinucleotide are often not considered for their potential effect on RNA splicing but in silico tools and a minigene system can be utilized for the prioritization and validation of such splice-disrupting variants. These variants can be overlooked causes of human disease but can be identified using conventional exon sequencing with proper interpretation guidelines.


Subject(s)
Exons , Gene Expression , Genes, Reporter , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Genetic Variation , RNA Splicing , Alleles , Chromosome Mapping , Computational Biology/methods , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genotype , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Annotation , Pedigree , Reproducibility of Results
16.
Pharmacogenet Genomics ; 26(1): 20-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26426212

ABSTRACT

OBJECTIVE: The aim of the study was to investigate the role of single-nucleotide polymorphisms (SNPs) located in the neuropilin-1 (NRP1) gene in treatment response to antivascular endothelial growth factor (VEGF) therapy for neovascular age-related macular degeneration (nvAMD). METHODS: Four SNPs in the NRP1 gene (rs2229935, rs2247383, rs2070296, and rs2804495) were genotyped in a study cohort of 377 nvAMD patients who received the loading dose of three monthly ranibizumab injections. Treatment response was assessed as the change in visual acuity after three monthly loading injections compared with baseline. RESULTS: SNP rs2070296 was associated with change in visual acuity after 3 months of treatment. Patients carrying the GA or AA genotypes performed significantly worse than individuals carrying the GG genotype (P=0.01). A cumulative effect of rs2070296 in the NRP1 gene and rs4576072 located in the VEGF receptor 2 (VEGFR2 or KDR) gene, previously associated with treatment response, was observed. Patients carrying two risk alleles performed significantly worse than patients carrying zero or one risk allele (P=0.03), and patients with more than two risk alleles responded even worse to the therapy (P=3×10). The combined effect of these two SNPs on the response was also seen after 6 and 12 months of treatment. CONCLUSION: This study suggests that genetic variation in NRP1, a key molecule in VEGFA-driven neovascularization, influences treatment response to ranibizumab in nvAMD patients. The results of this study may be used to generate prediction models for treatment response, which in the future may help tailor medical care to individual needs.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Neuropilin-1/genetics , Polymorphism, Single Nucleotide/drug effects , Ranibizumab/administration & dosage , Wet Macular Degeneration/drug therapy , Aged , Aged, 80 and over , Angiogenesis Inhibitors/pharmacology , Female , Humans , Male , Ranibizumab/pharmacology , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/genetics , Wet Macular Degeneration/genetics
17.
Am J Hum Genet ; 93(3): 571-8, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23993198

ABSTRACT

Vitelliform macular dystrophies (VMD) are inherited retinal dystrophies characterized by yellow, round deposits visible upon fundus examination and encountered in individuals with juvenile Best macular dystrophy (BMD) or adult-onset vitelliform macular dystrophy (AVMD). Although many BMD and some AVMD cases harbor mutations in BEST1 or PRPH2, the underlying genetic cause remains unknown for many affected individuals. In a large family with autosomal-dominant VMD, gene mapping and whole-exome sequencing led to the identification of a c.713T>G (p.Leu238Arg) IMPG1 mutation, which was subsequently found in two other families with autosomal-dominant VMD and the same phenotype. IMPG1 encodes the SPACR protein, a component of the rod and cone photoreceptor extracellular matrix domains. Structural modeling indicates that the p.Leu238Arg substitution destabilizes the conserved SEA1 domain of SPACR. Screening of 144 probands who had various forms of macular dystrophy revealed three other IMPG1 mutations. Two individuals from one family affected by autosomal-recessive VMD were homozygous for the splice-site mutation c.807+1G>T, and two from another family were compound heterozygous for the mutations c.461T>C (p.Leu154Pro) and c.1519C>T (p.Arg507(∗)). Most cases had a normal or moderately decreased electrooculogram Arden ratio. We conclude that IMPG1 mutations cause both autosomal-dominant and -recessive forms of VMD, thus indicating that impairment of the interphotoreceptor matrix might be a general cause of VMD.


Subject(s)
Extracellular Matrix Proteins/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Proteoglycans/genetics , Vitelliform Macular Dystrophy/genetics , Adult , Amino Acid Sequence , Base Sequence , Chromosomes, Human/genetics , Extracellular Matrix Proteins/chemistry , Eye Proteins/chemistry , Female , Fundus Oculi , Humans , Inheritance Patterns/genetics , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phenotype , Proteoglycans/chemistry , Young Adult
18.
Genet Med ; 18(10): 1044-51, 2016 10.
Article in English | MEDLINE | ID: mdl-26820066

ABSTRACT

PURPOSE: Leber congenital amaurosis (LCA) is an early-onset form of retinal degeneration. Six of the 22 known LCA genes encode photoreceptor ciliary proteins. Despite the identification of 22 LCA genes, the genetic basis of ~30% of LCA patients remains unknown. We sought to investigate the cause of disease in the remaining 30% by examining cilia-associated genes. METHODS: Whole-exome sequencing was performed on an LCA cohort of 212 unsolved probands previously screened for mutations in known retinal-disease genes. Immunohistochemistry using mouse retinas was used to confirm protein localization and zebrafish were used to perform rescue experiments. RESULTS: A homozygous nonsynonymous mutation was found in a single proband in CLUAP1, a gene required for ciliogenesis and cilia maintenance. Cluap1 knockout zebrafish exhibit photoreceptor cell death as early as 5 days after fertilization, and rescue experiments revealed that our proband's mutation is significantly hypomorphic. CONCLUSION: Consistent with the knowledge that CLUAP1 plays an important role in cilia function and that cilia are critical to photoreceptor function, our results indicate that hypomorphic mutations in CLUAP1 can result in dysfunctional photoreceptors without systemic abnormalities. This is the first report linking mutations in CLUAP1 to human disease and establishes CLUAP1 as a candidate LCA gene.Genet Med 18 10, 1044-1051.


Subject(s)
Antigens, Neoplasm/genetics , Cilia/genetics , Leber Congenital Amaurosis/genetics , Retinal Degeneration/genetics , Animals , Child, Preschool , Cilia/metabolism , Cilia/pathology , Exome/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Leber Congenital Amaurosis/pathology , Male , Mutation , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Zebrafish
19.
Doc Ophthalmol ; 132(3): 213-29, 2016 06.
Article in English | MEDLINE | ID: mdl-27041556

ABSTRACT

PURPOSE: A patient initially presented with constricted visual field, attenuated retinal vasculature, pigmentary clumping and reduced ERG in OS only, suggestive of unilateral retinitis pigmentosa (RP). This patient was subsequently seen on eight occasions (over three decades), and, with time, the initially normal eye (OD) gradually showed signs of RP-like degeneration. The purpose of this study was to evaluate which clinical modality (visual field, funduscopy or electroretinography) could have first predicted this fate. METHODS: At each time points, data obtained from our patient were compared to normative data using Z tests. RESULTS: At initial visit, all tests were significantly (p < 0.05) altered in OS and normal in OD. Visual field and retinal vessel diameter in OD reduced gradually to reach statistical significance at the 5th visit and 6th visit (21 and 22 years after the first examination, respectively). In OD, the amplitude of the scotopic and photopic ERGs reduced gradually and was significantly smaller than normal at the 2nd visit (after 11 years) and 3rd visit (after 18 years), respectively. When the photopic ERG was analyzed using the discrete wavelet transform (DWT), we were able to detect a significant change at the 2nd visit (after 11 years) instead of the 3rd visit (18 years). CONCLUSIONS: Our study allowed us to witness the earliest manifestation of an RP disease process. The ERG was the first test to detect significant RP changes. A significantly earlier detection of ERG anomalies was obtained when the DWT was used, demonstrating its advantage for early detection of ERG changes.


Subject(s)
Electroretinography/methods , Ophthalmoscopy/methods , Retinitis Pigmentosa/diagnosis , Visual Field Tests , Adult , Female , Follow-Up Studies , Humans , Vision Disorders/diagnosis , Visual Acuity , Visual Fields/physiology
20.
Adv Exp Med Biol ; 854: 193-200, 2016.
Article in English | MEDLINE | ID: mdl-26427411

ABSTRACT

Whole-genome linkage mapping identified a region on chromosome 10q21.3-q22.1 with a maximum LOD score of 3.0 at 0 % recombination in a six-generation family with autosomal dominant retinitis pigmentosa (adRP). All known adRP genes and X-linked RP genes were excluded in the family by a combination of methods. Whole-exome next-generation sequencing revealed a missense mutation in hexokinase 1, HK1 c.2539G > A, p.Glu847Lys, tracking with disease in all affected family members. One severely-affected male is homozygous for this region by linkage analysis and has two copies of the mutation. No other potential mutations were detected in the linkage region nor were any candidates identified elsewhere in the genome. Subsequent testing detected the same mutation in four additional, unrelated adRP families, for a total of five mutations in 404 probands tested (1.2 %). Of the five families, three are from the Acadian population in Louisiana, one is French Canadian and one is Sicilian. Haplotype analysis of the affected chromosome in each family and the homozygous individual revealed a rare, shared haplotype of 450 kb, suggesting an ancient founder mutation. HK1 is a widely-expressed gene, with multiple, abundant retinal transcripts, coding for hexokinase 1. Hexokinase catalyzes phosphorylation of glucose to glusose-6-phospate, the first step in glycolysis. The Glu847Lys mutation is in a highly-conserved site, outside of the active site or known functional sites.


Subject(s)
Chromosomes, Human, Pair 10/genetics , Genetic Predisposition to Disease/genetics , Hexokinase/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Base Sequence , DNA Mutational Analysis , Exome/genetics , Family Health , Female , Genes, Dominant , Genotype , Haplotypes , High-Throughput Nucleotide Sequencing , Humans , Male , Pedigree , Retinitis Pigmentosa/diagnosis , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL