Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Hum Mutat ; 43(7): 832-858, 2022 07.
Article in English | MEDLINE | ID: mdl-35332618

ABSTRACT

Achromatopsia (ACHM) is a congenital cone photoreceptor disorder characterized by impaired color discrimination, low visual acuity, photosensitivity, and nystagmus. To date, six genes have been associated with ACHM (CNGA3, CNGB3, GNAT2, PDE6C, PDE6H, and ATF6), the majority of these being implicated in the cone phototransduction cascade. CNGA3 encodes the CNGA3 subunit of the cyclic nucleotide-gated ion channel in cone photoreceptors and is one of the major disease-associated genes for ACHM. Herein, we provide a comprehensive overview of the CNGA3 variant spectrum in a cohort of 1060 genetically confirmed ACHM patients, 385 (36.3%) of these carrying "likely disease-causing" variants in CNGA3. Compiling our own genetic data with those reported in the literature and in public databases, we further extend the CNGA3 variant spectrum to a total of 316 variants, 244 of which we interpreted as "likely disease-causing" according to ACMG/AMP criteria. We report 48 novel "likely disease-causing" variants, 24 of which are missense substitutions underlining the predominant role of this mutation class in the CNGA3 variant spectrum. In addition, we provide extensive in silico analyses and summarize reported functional data of previously analyzed missense, nonsense and splicing variants to further advance the pathogenicity assessment of the identified variants.


Subject(s)
Color Vision Defects , Cyclic Nucleotide-Gated Cation Channels , Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Humans , Mutation , Retinal Cone Photoreceptor Cells
2.
Int J Mol Sci ; 21(22)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182541

ABSTRACT

X-chromosomal retinitis pigmentosa (RP) frequently is caused by mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. We evaluated the potential of PTC124 (Ataluren, TranslamaTM) treatment to promote ribosomal read-through of premature termination codons (PTC) in RPGR. Expression constructs in HEK293T cells showed that the efficacy of read-through reagents is higher for UGA than UAA PTCs. We identified the novel hemizygous nonsense mutation c.1154T > A, p.Leu385* (NM_000328.3) causing a UAA PTC in RPGR and generated patient-derived fibroblasts. Immunocytochemistry of serum-starved control fibroblasts showed the RPGR protein in a dot-like expression pattern along the primary cilium. In contrast, RPGR was no longer detectable at the primary cilium in patient-derived cells. Applying PTC124 restored RPGR at the cilium in approximately 8% of patient-derived cells. RT-PCR and Western blot assays verified the pathogenic mechanisms underlying the nonsense variant. Immunofluorescence stainings confirmed the successful PTC124 treatment. Our results showed for the first time that PTC124 induces read-through of PTCs in RPGR and restores the localization of the RPGR protein at the primary cilium in patient-derived cells. These results may provide a promising new treatment option for patients suffering from nonsense mutations in RPGR or other genetic diseases.


Subject(s)
Codon, Nonsense/drug effects , Eye Proteins/genetics , Genetic Diseases, X-Linked/drug therapy , Genetic Diseases, X-Linked/genetics , Mutant Proteins/genetics , Oxadiazoles/therapeutic use , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Case-Control Studies , Cells, Cultured , Cilia/metabolism , Eye Proteins/biosynthesis , Genetic Diseases, X-Linked/metabolism , HEK293 Cells , Hemizygote , Humans , Mutant Proteins/biosynthesis , Proof of Concept Study , Protein Biosynthesis/drug effects , RNA Stability , Retinitis Pigmentosa/metabolism
3.
Int J Mol Sci ; 21(5)2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32111077

ABSTRACT

Best vitelliform macular dystrophy (BD), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and the autosomal recessive bestrophinopathy (ARB), together known as the bestrophinopathies, are caused by mutations in the bestrophin-1 (BEST1) gene affecting anion transport through the plasma membrane of the retinal pigment epithelium (RPE). To date, while no treatment exists a better understanding of BEST1-related pathogenesis may help to define therapeutic targets. Here, we systematically characterize functional consequences of mutant BEST1 in thirteen RPE patient cell lines differentiated from human induced pluripotent stem cells (hiPSCs). Both BD and ARB hiPSC-RPEs display a strong reduction of BEST1-mediated anion transport function compared to control, while ADVIRC mutations trigger an increased anion permeability suggesting a stabilized open state condition of channel gating. Furthermore, BD and ARB hiPSC-RPEs differ by the degree of mutant protein turnover and by the site of subcellular protein quality control with adverse effects on lysosomal pH only in the BD-related cell lines. The latter finding is consistent with an altered processing of catalytic enzymes in the lysosomes. The present study provides a deeper insight into distinct molecular mechanisms of the three bestrophinopathies facilitating functional categorization of the more than 300 known BEST1 mutations that result into the distinct retinal phenotypes.


Subject(s)
Bestrophins/genetics , Bestrophins/metabolism , Eye Diseases, Hereditary/genetics , Mutation , Phenotype , Retinal Diseases/genetics , Cell Line , Choroid Diseases/genetics , Choroid Diseases/metabolism , Choroid Diseases/pathology , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Genes, Recessive , Genetic Predisposition to Disease/genetics , Homeostasis , Humans , Hydrogen-Ion Concentration , Induced Pluripotent Stem Cells , Retina/metabolism , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Pigment Epithelium/metabolism , Vitelliform Macular Dystrophy
4.
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
5.
Hum Mutat ; 40(6): 765-787, 2019 06.
Article in English | MEDLINE | ID: mdl-30825406

ABSTRACT

Inherited retinal disorders (IRD) represent clinically and genetically heterogeneous diseases. To date, pathogenic variants have been identified in ~260 genes. Albeit that many genes are implicated in IRD, for 30-50% of the cases, the gene defect is unknown. These cases may be explained by novel gene defects, by overlooked structural variants, by variants in intronic, promoter or more distant regulatory regions, and represent synonymous variants of known genes contributing to the dysfunction of the respective proteins. Patients with one subgroup of IRD, namely incomplete congenital stationary night blindness (icCSNB), show a very specific phenotype. The major cause of this condition is the presence of a hemizygous pathogenic variant in CACNA1F. A comprehensive study applying direct Sanger sequencing of the gene-coding regions, exome and genome sequencing applied to a large cohort of patients with a clinical diagnosis of icCSNB revealed indeed that seven of the 189 CACNA1F-related cases have intronic and synonymous disease-causing variants leading to missplicing as validated by minigene approaches. These findings highlight that gene-locus sequencing may be a very efficient method in detecting disease-causing variants in clinically well-characterized patients with a diagnosis of IRD, like icCSNB.


Subject(s)
Calcium Channels, L-Type/genetics , Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Mutation , Myopia/genetics , Night Blindness/genetics , Sequence Analysis, DNA/methods , Genetic Predisposition to Disease , Hemizygote , Humans , Introns , Male , Pedigree , RNA Splicing , Silent Mutation
6.
Hum Mol Genet ; 26(1): 133-144, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28025326

ABSTRACT

Macular dystrophy leads to progressive loss of central vision and shows symptoms similar to age-related macular degeneration. Genetic screening of patients diagnosed with macular dystrophy disclosed a novel mutation in the GUCA1A gene, namely a c.526C > T substitution leading to the amino acid substitution p.L176F in the guanylate cyclase-activating protein 1 (GCAP1). The same variant was found in three families showing an autosomal dominant mode of inheritance. For a full functional characterization of the L176F mutant we expressed and purified the mutant protein and measured key parameters of its activating properties, its Ca2+/Mg2+-binding, and its Ca2+-induced conformational changes in comparison to the wildtype protein. The mutant was less sensitive to changes in free Ca2+, resulting in a constitutively active form under physiological Ca2+-concentration, showed significantly higher activation rates than the wildtype (90-fold versus 20-fold) and interacted with an higher apparent affinity with its target guanylate cyclase. However, direct Ca2+-binding of the mutant was nearly similar to the wildtype; binding of Mg2+ occurred with higher affinity. We performed molecular dynamics simulations for comparing the Ca2+-saturated inhibiting state of GCAP1 with the Mg2+-bound activating states. The L176F mutant exhibited significantly lower flexibility, when three Ca2+ or two Mg2+ were bound forming probably the structural basis for the modified GCAP1 function.


Subject(s)
Calcium/metabolism , Cyclic GMP/metabolism , Guanylate Cyclase-Activating Proteins/genetics , Macular Degeneration/genetics , Mutation/genetics , Retinal Cone Photoreceptor Cells/metabolism , Adolescent , Adult , Female , Guanylate Cyclase-Activating Proteins/chemistry , Guanylate Cyclase-Activating Proteins/metabolism , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Middle Aged , Molecular Dynamics Simulation , Pedigree , Protein Conformation , Retinal Cone Photoreceptor Cells/pathology , Young Adult
7.
Sci Rep ; 6: 36208, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27812022

ABSTRACT

The diagnoses of retinitis pigmentosa (RP) and stationary night blindness (CSNB) are two distinct clinical entities belonging to a group of clinically and genetically heterogeneous retinal diseases. The current study focused on the identification of causative mutations in the RP-affected index patient and in several members of the same family that reported a phenotype resembling CSNB. Ophthalmological examinations of the index patient confirmed a typical form of RP. In contrast, clinical characterizations and ERGs of another affected family member showed the Riggs-type CSNB lacking signs of RP. Applying whole exome sequencing we detected the non-synonymous substitution c.337G > A, p.E113 K in the rhodopsin (RHO) gene. The mutation co-segregated with the diseases. The identification of the pathogenic variant p.E113 K is the first description of a naturally-occurring mutation in the Schiff base counterion of RHO in human patients. The heterozygous mutation c.337G > A in exon 1 was confirmed in the index patient as well as in five CSNB-affected relatives. This pathogenic sequence change was excluded in a healthy family member and in 199 ethnically matched controls. Our findings suggest that a mutation in the biochemically well-characterized counterion p.E113 in RHO can be associated with RP or Riggs-type CSNB, even within the same family.


Subject(s)
Mutation, Missense , Night Blindness/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Adult , Aged, 80 and over , Amino Acid Sequence , Amino Acid Substitution , Case-Control Studies , DNA Mutational Analysis , Female , Heterozygote , Humans , Male , Middle Aged , Night Blindness/diagnostic imaging , Pedigree , Phenotype , Retinitis Pigmentosa/diagnostic imaging , Rhodopsin/chemistry , Schiff Bases , Sequence Analysis, DNA
8.
PLoS One ; 11(1): e0145951, 2016.
Article in English | MEDLINE | ID: mdl-26766544

ABSTRACT

Retinal dystrophies (RD) constitute a group of blinding diseases that are characterized by clinical variability and pronounced genetic heterogeneity. The different nonsyndromic and syndromic forms of RD can be attributed to mutations in more than 200 genes. Consequently, next generation sequencing (NGS) technologies are among the most promising approaches to identify mutations in RD. We screened a large cohort of patients comprising 89 independent cases and families with various subforms of RD applying different NGS platforms. While mutation screening in 50 cases was performed using a RD gene capture panel, 47 cases were analyzed using whole exome sequencing. One family was analyzed using whole genome sequencing. A detection rate of 61% was achieved including mutations in 34 known and two novel RD genes. A total of 69 distinct mutations were identified, including 39 novel mutations. Notably, genetic findings in several families were not consistent with the initial clinical diagnosis. Clinical reassessment resulted in refinement of the clinical diagnosis in some of these families and confirmed the broad clinical spectrum associated with mutations in RD genes.


Subject(s)
Mutation , Retinal Dystrophies/genetics , DNA Copy Number Variations , Exome , Eye Proteins/genetics , Female , Genetic Association Studies , Genetic Heterogeneity , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation Rate , Pedigree , Phenotype , Retinal Dystrophies/diagnosis
9.
J Neurol Sci ; 358(1-2): 522-4, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26474792

ABSTRACT

UNLABELLED: Optic glioma (OPG) accounts for 4-8% of all brain tumors in children. En-block removal of intraorbital tumor is recommended in cases with disfiguring exophthalmos and impaired vision. Surgical resection of intraorbital optic nerve (ON) poses the risks of permanent ptosis and globe atrophy. We present here the case of a 4-year-old boy with exophthalmos and near blindness due to an intraorbital OPG. Despite chemotherapy he showed progressive exophthalmos and vision loss. Bony orbital decompression with ON transection temporally reduced his exophthalmos. OPG resection was required later for recurrence of his exophthalmos secondary to tumor progression. Post operatively, he had preserved oculomotor nerve functions but developed globe ischemia. Unusually, his ischemic globe caused him to have pain and severe photophobia, which later lead to enucleation. Photophobia has been reported in blind patients. Animal models and MRI functional imaging showed activation of trigeminal pathway during photophobia in completely transected ON. However, the exact neuro-ophthalmology pathway requires further study. CONCLUSION: This is the first described case of photophobia after excision of OPG with ON denervation. Photophobia can be a serious side effect that significantly lowers the patient's quality of life.


Subject(s)
Blindness/etiology , Exophthalmos/etiology , Optic Nerve Glioma/complications , Optic Nerve/surgery , Photophobia/physiopathology , Child, Preschool , Denervation , Humans , Male , Optic Nerve Glioma/surgery , Orbit/surgery , Trigeminal Nerve/physiopathology
10.
Transl Vis Sci Technol ; 4(5): 3, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26396929

ABSTRACT

PURPOSE: Spontaneous venous pulsation correlates negatively with elevated intracranial pressure and papilledema, and it relates to glaucoma. Yet, its etiology remains unclear. A key element to elucidate its underlying mechanism is the time at which collapse occurs with respect to the heart cycle, but previous reports are contradictory. We assessed this question in healthy subjects using quantitative measurements of both vein diameters and artery lateral displacements; the latter being used as the marker of the ocular systole time. METHODS: We recorded 5-second fundus sequences with a near-infrared scanning laser ophthalmoscope in 12 young healthy subjects. The image sequences were coregistered, cleaned from microsaccades, and filtered via a principal component analysis to remove nonpulsatile dynamic features. Time courses of arterial lateral displacement and of diameter at sites of spontaneous venous pulsation or proximal to the disk were retrieved from those image sequences and compared. RESULTS: Four subjects displayed both arterial and venous pulsatile waveforms. On those, we observed venous diameter waveforms differing markedly among the subjects, ranging from a waveform matching the typical intraocular pressure waveform to a close replica of the arterial waveform. CONCLUSIONS: The heterogeneity in waveforms and arteriovenous phases suggests that the mechanism governing the venous outflow resistance differs among healthy subjects. TRANSLATIONAL RELEVANCE: Further characterizations are necessary to understand the heterogeneous mechanisms governing the venous outflow resistance as this resistance is altered in glaucoma and is instrumental when monitoring intracranial hypertension based on fundus observations.

11.
Proc Natl Acad Sci U S A ; 112(20): E2630-9, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25941382

ABSTRACT

In response to cell swelling, volume-regulated anion channels (VRACs) participate in a process known as regulatory volume decrease (RVD). Only recently, first insight into the molecular identity of mammalian VRACs was obtained by the discovery of the leucine-rich repeats containing 8A (LRRC8A) gene. Here, we show that bestrophin 1 (BEST1) but not LRRC8A is crucial for volume regulation in human retinal pigment epithelium (RPE) cells. Whole-cell patch-clamp recordings in RPE derived from human-induced pluripotent stem cells (hiPSC) exhibit an outwardly rectifying chloride current with characteristic functional properties of VRACs. This current is severely reduced in hiPSC-RPE cells derived from macular dystrophy patients with pathologic BEST1 mutations. Disruption of the orthologous mouse gene (Best1(-/-)) does not result in obvious retinal pathology but leads to a severe subfertility phenotype in agreement with minor endogenous expression of Best1 in murine RPE but highly abundant expression in mouse testis. Sperm from Best1(-/-) mice showed reduced motility and abnormal sperm morphology, indicating an inability in RVD. Together, our data suggest that the molecular identity of VRACs is more complex--that is, instead of a single ubiquitous channel, VRACs could be formed by cell type- or tissue-specific subunit composition. Our findings provide the basis to further examine VRAC diversity in normal and diseased cell physiology, which is key to exploring novel therapeutic approaches in VRAC-associated pathologies.


Subject(s)
Cell Size , Chloride Channels/metabolism , Eye Proteins/metabolism , Models, Biological , Retinal Pigment Epithelium/cytology , Amino Acid Sequence , Animals , Bestrophins , Eye Proteins/genetics , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Ion Channels/deficiency , Ion Channels/genetics , Male , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Oocytes/metabolism , Patch-Clamp Techniques , Spermatozoa/cytology , Statistics, Nonparametric , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...