Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Graefes Arch Clin Exp Ophthalmol ; 262(6): 1737-1744, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38206414

ABSTRACT

BACKGROUND: Kabuki Syndrome is a rare and genetically heterogenous condition with both ophthalmic and systemic complications and typical facial features. We detail the macular phenotype in two unrelated patients with Kabuki syndrome due to de novo nonsense variants in KMT2D, one novel. A follow-up of 10 years is reported. Pathogenicity of both de novo nonsense variants is analyzed. METHODS: Four eyes of two young patients were studied by full clinical examination, kinetic perimetry, short wavelength autofluorescence, full field (ff) ERGs, and spectral-domain optical coherence tomography (SD-OCT). One patient had adaptive optic (AO) imaging. Whole exome sequencing was performed in both patients. RESULTS: Both patients had de novo nonsense variants in KMTD2. One patient had c.14843C>G; p. (Ser4948ter) novel variant and the second c.11119C>T; p. (Arg3707ter). Both had a stable Snellen visual acuity of 0.2-0.3. The retinal multimodal imaging demonstrated abnormalities at the fovea in both eyes: hyperreflectivity to blue light and a well-delimited gap-disruption of ellipsoid and interdigitation layer on OCT. The dark area on AO imaging is presumed to be absent for, or with structural change to photoreceptors. The ff ERGs and kinetic visual fields were normal. The foveal findings remained stable over several years. CONCLUSION: Kabuki syndrome-related maculopathy is a distinct loss of photoreceptors at the fovea as shown by multimodal imaging including, for the first time, AO imaging. This report adds to the literature of only one case with maculopathy with two additional macular dystrophies in patients with Kabuki syndrome. Although underestimated, these cases further raise awareness of the potential impact of retinal manifestations of Kabuki syndrome not only among ophthalmologists but also other healthcare professionals involved in the care of patients with this multisystem disorder.


Subject(s)
Abnormalities, Multiple , Electroretinography , Face , Fluorescein Angiography , Hematologic Diseases , Multimodal Imaging , Neoplasm Proteins , Phenotype , Tomography, Optical Coherence , Vestibular Diseases , Visual Acuity , Humans , Vestibular Diseases/genetics , Vestibular Diseases/diagnosis , Vestibular Diseases/physiopathology , Face/abnormalities , Hematologic Diseases/genetics , Hematologic Diseases/diagnosis , Hematologic Diseases/physiopathology , Tomography, Optical Coherence/methods , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnosis , Follow-Up Studies , Male , Female , Neoplasm Proteins/genetics , Fluorescein Angiography/methods , DNA-Binding Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Neck , Fundus Oculi , DNA/genetics , Exome Sequencing , DNA Mutational Analysis , Macula Lutea/pathology , Time Factors , Adult , Adolescent
2.
Int J Mol Sci ; 23(8)2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35457110

ABSTRACT

Biallelic gene defects in MFSD8 are not only a cause of the late-infantile form of neuronal ceroid lipofuscinosis, but also of rare isolated retinal degeneration. We report clinical and genetic data of seven patients compound heterozygous or homozygous for variants in MFSD8, issued from a French cohort with inherited retinal degeneration, and two additional patients retrieved from a Swiss cohort. Next-generation sequencing of large panels combined with whole-genome sequencing allowed for the identification of twelve variants from which seven were novel. Among them were one deep intronic variant c.998+1669A>G, one large deletion encompassing exon 9 and 10, and a silent change c.750A>G. Transcript analysis performed on patients' lymphoblastoid cell lines revealed the creation of a donor splice site by c.998+1669A>G, resulting in a 140 bp pseudoexon insertion in intron 10. Variant c.750A>G produced exon 8 skipping. In silico and in cellulo studies of these variants allowed us to assign the pathogenic effect, and showed that the combination of at least one severe variant with a moderate one leads to isolated retinal dystrophy, whereas the combination in trans of two severe variants is responsible for early onset severe retinal dystrophy in the context of late-infantile neuronal ceroid lipofuscinosis.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Retinal Dystrophies , Exons/genetics , Homozygote , Humans , Membrane Transport Proteins/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Retinal Dystrophies/genetics
3.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948090

ABSTRACT

Usher syndrome is an autosomal recessive disorder characterized by congenital hearing loss combined with retinitis pigmentosa, and in some cases, vestibular areflexia. Three clinical subtypes are distinguished, and MYO7A and USH2A represent the two major causal genes involved in Usher type I, the most severe form, and type II, the most frequent form, respectively. Massively parallel sequencing was performed on a cohort of patients in the context of a molecular diagnosis to confirm clinical suspicion of Usher syndrome. We report here 231 pathogenic MYO7A and USH2A genotypes identified in 73 Usher type I and 158 Usher type II patients. Furthermore, we present the ACMG classification of the variants, which comprise all types. Among them, 68 have not been previously reported in the literature, including 12 missense and 16 splice variants. We also report a new deep intronic variant in USH2A. Despite the important number of molecular studies published on these two genes, we show that during the course of routine genetic diagnosis, undescribed variants continue to be identified at a high rate. This is particularly pertinent in the current era, where therapeutic strategies based on DNA or RNA technologies are being developed.


Subject(s)
Extracellular Matrix Proteins/genetics , Genotype , Mutation, Missense , Myosin VIIa/genetics , RNA Splice Sites , Usher Syndromes , Adult , Female , France , Humans , Male , Usher Syndromes/classification , Usher Syndromes/genetics
4.
Ophthalmic Genet ; 40(4): 365-368, 2019 08.
Article in English | MEDLINE | ID: mdl-31438752

ABSTRACT

Background: Posterior pole staphylomata (PSS) is an outward bulging of ocular wall, rarely reported in association with inherited retinal degenerations. Patients and methods: We report a large French family of Jewish ancestry with a peculiar form of dominant retinitis pigmentosa (RP) and posterior pole staphyloma (PPS). Eight members were clinically and genetically examined. Results: All affected members complained of night blindness from early childhood and their ERGs were extinguished in the first decade of life. Seven out of eight presented PPS on fundus examination and SD-OCT. The youngest patient did not present PPS at 11 months of age, but the signs of posterior pole bowing became evident at age 8 years. There was no association between the presence of PPS and refraction. Patients with PPS were either hyperopic or myopic, but all have a high with-the-rule astigmatism. A myopic shift was observed for all of them at follow-up. In this family, the disease segregated with the c.886A>G mutation in RHO gene. Conclusion: A PPS development was observed in initially non-myopic patients of a family with unusually severe dominant RP. The PPS concerned only the area with relatively preserved outer retinal layers (outer nuclear layer and ellipsoid zone). How the outer retina could guide choroid and scleral remodelling remains unclear.


Subject(s)
Mutation , Myopia/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Scleral Diseases/genetics , Severity of Illness Index , Adult , Child , Female , Follow-Up Studies , Humans , Infant , Male , Myopia/complications , Myopia/pathology , Pedigree , Prognosis , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/pathology , Retrospective Studies , Scleral Diseases/complications , Scleral Diseases/pathology
5.
Ophthalmic Genet ; 40(2): 161-164, 2019 04.
Article in English | MEDLINE | ID: mdl-30942644

ABSTRACT

BACKGROUND: Congenital nystagmus is one of the most common neuro-ophthalmological disorders. X chromosome-linked forms are associated with pathogenic variants of the GPR143 and FRMD7 genes. MATERIALS AND METHODS: Patients' DNA was analyzed using a next-generation sequencing (NGS) panel of genes involved in albinism and related pathologies (TYR, OCA2, TYRP1, SLC45A2, SLC24A5, C10ORF11, GPR143, SLC38A8, HPS 1 to 10, LYST, MITF, FRMD7) Results: We report a 4 generation family with 5 affected members initially referred for molecular diagnosis of ocular albinism. A missense variant of FRMD7 was found in 3 affected cases and one female carrier. We show that the disease in the affected girl is due to skewed inactivation of the X chromosome. CONCLUSIONS: By compiling all the published cases we discuss the variable penetrance among females due to different types of mutation and to X-inactivation.


Subject(s)
Albinism, Ocular/genetics , Cytoskeletal Proteins/genetics , Genetic Diseases, X-Linked/genetics , Membrane Proteins/genetics , Mutation, Missense , Nystagmus, Congenital/genetics , X Chromosome Inactivation/genetics , Albinism, Ocular/diagnosis , Child , Diagnosis, Differential , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Nystagmus, Congenital/diagnosis , Pedigree , Slit Lamp Microscopy , Tomography, Optical Coherence
6.
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
7.
Br J Ophthalmol ; 103(9): 1239-1247, 2019 09.
Article in English | MEDLINE | ID: mdl-30472657

ABSTRACT

AIM: Oculocutaneous albinism type 1 (OCA1) is due to TYR mutations. c.1205G>A/p.Arg402Gln (R402Q) is a thermosensitive variant of the TYR gene that has been reported to be responsible for mild forms of OCA1. The aim of our study was to define the phenotype associated with this variant. METHODS: In our retrospective series, among 268 patients diagnosed with OCA1, 122 (45.5%) harboured one pathogenic variant of TYR, and the R402Q variant ensured to be in trans by segregation analysis in 69 patients (25.7%), constituting the 'R402Q-OCA1' group. 146 patients harboured two pathogenic variants of the TYR gene other than R402Q. Clinical records were available for 119 of them, constituting the 'Classical-OCA1' group. RESULTS: Most R402Q-OCA1 patients presented with white or yellow-white hair at birth (71.43%), blond hair later (46.97%), a light phototype but with residual pigmentation (69.64%), and blue eyes (76.56%). Their pigmentation was significantly higher than in the classical-OCA1 group. All patients from the R402Q-OCA1 group presented with ocular features of albinism. However the prevalence of photophobia (78.13%) and iris transillumination (83.87%) and the severity scores of iris transillumination, retinal hypopigmentation and foveal hypoplasia were lower in the R402Q-OCA1 group. Visual acuity was higher in the R402Q-OCA1 group (0.38±0.21 logarithm of the minimum angle of resolution vs 0.76±0.24). Investigations concerning a possible additive effect of the c.575C>A/p.Ser192 (S192Y) variant of TYR in cis with R402Q, suggested by others, showed no significant impact on the phenotype. CONCLUSION: The R402Q variant leads to variable but generally mild forms of albinism whose less typical presentation may lead to underdiagnosis.


Subject(s)
Albinism, Oculocutaneous/genetics , Monophenol Monooxygenase/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Albinism, Oculocutaneous/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation , Pedigree , Phenotype , Retrospective Studies , Young Adult
9.
Sci Rep ; 8(1): 6840, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717154

ABSTRACT

EMAP (Extensive Macular Atrophy with Pseudodrusen) is a maculopathy we recently described that shares pseudodrusen and geographic atrophy with Age-related Macular Disease (AMD). EMAP differs from AMD by an earlier age of onset (50-55 years) and a characteristic natural history comprising a night blindness followed by a severe visual loss. In a prospective case-control study, ten referral centers included 115 EMAP (70 women, 45 men) patients and 345 matched controls to appraise dietary, environmental, and genetic risk factors. The incidence of EMAP (mean 2.95/1.106) was lower in Provence-Côte d'Azur with a Mediterranean diet (1.9/1.106), and higher in regions with intensive farming or industrialized activities (5 to 20/1.106). EMAP patients reported toxic exposure during professional activities (OR 2.29). The frequencies of common AMD complement factor risk alleles were comparable in EMAP. By contrast, only one EMAP patient had a rare AMD variant. This study suggests that EMAP could be a neurodegenerative disorder caused by lifelong toxic exposure and that it is associated with a chronic inflammation and abnormal complement pathway regulation. This leads to diffuse subretinal deposits with rod dysfunction and cone apoptosis around the age of 50 with characteristic extensive macular atrophy and paving stones in the far peripheral retina.


Subject(s)
Genetic Predisposition to Disease , Geographic Atrophy/epidemiology , Geographic Atrophy/genetics , Retinal Drusen/epidemiology , Retinal Drusen/genetics , Adult , Aged , Case-Control Studies , Diet, Mediterranean , Environmental Exposure/adverse effects , Feeding Behavior , Female , France/epidemiology , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies , Risk Factors
10.
Sci Rep ; 6: 32544, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27601084

ABSTRACT

To revisit the autosomal dominant Sorsby fundus dystrophy (SFD) as a syndromic condition including late-onset pulmonary disease. We report clinical and imaging data of ten affected individuals from 2 unrelated families with SFD and carrying heterozygous TIMP3 mutations (c.572A > G, p.Y191C, exon 5, in family 1 and c.113C > G, p.S38C, exon 1, in family 2). In family 1, all SFD patients older than 50 (two generations) had also a severe emphysema, despite no history of smoking or asthma. In the preceding generation, the mother died of pulmonary emphysema and she was blind after the age of 50. Her two great-grandsons (<20 years), had abnormal Bruch Membrane thickness, a sign of eye disease. In family 2, eye and lung diseases were also associated in two generations, both occurred later, and lung disease was moderate (bronchiectasis). This is the first report of a syndromic SFD in line with the mouse model uncovering the role of TIMP3 in human lung morphogenesis and functions. The TIMP3 gene should be screened in familial pulmonary diseases with bronchiectasis, associated with a medical history of visual loss. In addition, SFD patients should be advised to avoid tobacco consumption, to practice sports, and to undergo regular pulmonary examinations.


Subject(s)
Genetic Predisposition to Disease , Macular Degeneration/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Aged , Base Sequence , Family , Female , Fundus Oculi , Humans , Lung/pathology , Macular Degeneration/diagnostic imaging , Male , Middle Aged , Mutation , Pedigree , Protein Structure, Secondary , Tissue Inhibitor of Metalloproteinase-3/chemistry , Tomography, X-Ray Computed
11.
Ophthalmology ; 123(9): 1865-73, 2016 09.
Article in English | MEDLINE | ID: mdl-27320518

ABSTRACT

PURPOSE: To assess the association of clinical and biological factors with extensive macular atrophy with pseudodrusen (EMAP) characterized by bilateral macular atrophy occurring in patients aged 50 to 60 years and a rapid progression to legal blindness within 5 to 10 years. DESIGN: A national matched case-control study. PARTICIPANTS: Participants were recruited in 10 French Departments of Ophthalmology and their associated clinical investigation centers. All 115 patients with EMAP had symptoms before the age of 55 years due to bilateral extensive macular atrophy with a larger vertical axis and diffuse pseudodrusen. Three controls without age-related macular degeneration (AMD) or retinal disease at fundus examination were matched for each patient with EMAP by gender, age, and geographic area (in total 415). METHODS: Subjects and controls underwent an eye examination including color, red-free autofluorescent fundus photographs and spectral-domain optical coherence tomography with macular analysis. The interviews collected demographic, lifestyle, family and personal medical history, medications, and biological data. Associations of risk factors were estimated using conditional logistic regression. MAIN OUTCOME MEASURES: Extensive macular atrophy with pseudodrusen status (cases vs. controls). RESULTS: Extensive macular atrophy with pseudodrusen most frequently affected women (70 women, 45 men). After multivariate adjustment, family history of glaucoma or AMD was strongly associated with EMAP (odds ratio [OR], 2.3, P = 0.008 and OR, 1.5, P = 0.01, respectively). No association was found with cardiac diseases or their risk factors. Mild and moderate kidney disease and higher neutrophil rate were associated with a reduced risk of EMAP (OR, 0.58, P = 0.04; OR, 0.34, P = 0.01; and OR, 0.59, P = 0.003, respectively). On the contrary, eosinophilia (OR, 1.6; P = 0.0002), lymphocytosis (OR, 1.84; P = 0.0002), increased erythrocyte sedimentation rate (OR, 6.5; P = 0.0005), decreased CH50 (P = 0.001), and high plasma C3 level (P = 0.023) were significantly associated with a higher risk of EMAP. CONCLUSIONS: This study documents an association between EMAP and family history of AMD and glaucoma, a clear female predominance, and a systemic inflammatory profile. The reduced CH50 and increased C3 plasma values could reflect a more severe complement pathway dysfunction than in AMD, leading to early pseudodrusen and rapid development of geographic atrophy. There is no association of EMAP with AMD cardiac diseases or cardiac risks, including cigarette smoking.


Subject(s)
Geographic Atrophy/epidemiology , Macular Degeneration/epidemiology , Retinal Drusen/epidemiology , Adult , Aged , Aged, 80 and over , Blindness , Case-Control Studies , Choroidal Neovascularization/epidemiology , Diagnostic Techniques, Ophthalmological , Disease Progression , Female , France/epidemiology , Geographic Atrophy/etiology , Humans , Macular Degeneration/etiology , Male , Middle Aged , Odds Ratio , Photography , Retinal Drusen/etiology , Risk Factors , Sex Distribution , Tomography, Optical Coherence , Visual Acuity
12.
Doc Ophthalmol ; 131(2): 149-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138751

ABSTRACT

INTRODUCTION: Oligocone trichromacy (OT) is an uncommon cone dysfunction disorder, the mechanism of which remains poorly understood. OT has been thought to be non-progressive, but its long-term visual outcome has been seldom reported in the literature. Our aim was to present two OT patients followed at our institution over 18 years. MATERIALS AND METHODS: Complete ocular examination, color vision, visual fields, and full-field electroretinography (ERG) were performed at initial presentation and follow-up. Spectral-domain optical coherence tomography (OCT) was performed during follow-up when available at our institution. RESULTS: Initial ocular examination showed satisfactory visual acuities with normal fundus examination and near-to-normal color vision. However, computerized perimetry demonstrated a ring-shaped scotoma around fixation, and ERG showed a profound cone dysfunction. The discrepancy between preserved color vision and profound cone dysfunction leads to the diagnosis of OT. Subsequent follow-ups over 18 years showed subtle degradation of visual acuities along with progression of the myopia in both patients and slight worsening of color vision in one patient. Initial OCT revealed a focal interruption of the ellipsoid line along with decreased thickness of the perifoveal macula. Subsequent OCT imaging performed 2 years later did not show any macular changes. CONCLUSION: Although OT is known to be a non-progressive cone dysfunction, our results suggest that subtle degradation of the visual function might happen over time.


Subject(s)
Color Vision Defects/diagnosis , Retinal Cone Photoreceptor Cells/pathology , Adolescent , Child , Color Vision Defects/physiopathology , Electroretinography , Female , Fluorescein Angiography , Follow-Up Studies , Humans , Pedigree , Retrospective Studies , Tomography, Optical Coherence/methods , Visual Acuity/physiology , Visual Field Tests , Visual Fields/physiology
13.
Am J Ophthalmol ; 160(2): 364-372.e1, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25982971

ABSTRACT

PURPOSE: To describe a series of patients with Bardet-Biedl syndrome (BBS) and predominantly retinal cone dysfunction, a previously only rarely reported association. DESIGN: Retrospective observational case series. METHODS: Seven patients with clinically proven Bardet-Biedl syndrome had undergone detailed ocular phenotyping, which included fundus examination, Goldmann visual fields, fundus autofluorescence imaging (FAF), optical coherence tomography (OCT), and electroretinography (ERG). Mutational screening in the BBS genes was performed either by direct Sanger sequencing or targeted next-generation sequencing. RESULTS: All 7 patients had proven BBS mutations; 1 had a cone dystrophy phenotype on ERG and 6 had a cone-rod pattern of dysfunction. Macular atrophy was present in all patients, usually with central hypofluorescence surrounded by a continuous hyperfluorescent ring on fundus autofluorescence imaging. OCT confirmed loss of outer retinal structure within the atrophic areas. No clear genotype-phenotype relationship was evident. CONCLUSIONS: Patients with Bardet-Biedl syndrome usually develop early-onset retinitis pigmentosa. In contrast, the patients described herein, with molecularly confirmed Bardet-Biedl syndrome, developed early cone dysfunction, including the first reported case of a cone dystrophy phenotype associated with the disorder. The findings significantly expand the phenotype associated with Bardet-Biedl syndrome.


Subject(s)
Bardet-Biedl Syndrome/complications , DNA/genetics , Eye Proteins/genetics , Mutation , Retinal Cone Photoreceptor Cells/physiology , Retinal Degeneration/etiology , Adult , Bardet-Biedl Syndrome/genetics , DNA Mutational Analysis , Electroretinography , Female , Fluorescein Angiography , Fundus Oculi , Humans , Male , Middle Aged , Phenotype , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Retrospective Studies , Tomography, Optical Coherence , Visual Acuity , Visual Fields
14.
Am J Med Genet A ; 164A(6): 1537-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24668847

ABSTRACT

The 15q13.3 heterozygous microdeletion is a fairly common microdeletion syndrome with marked clinical variability and incomplete penetrance. The average size of the deletion, which comprises six genes including CHRNA7, is 1.5 Mb. CHRNA7 has been identified as the gene responsible for the neurological phenotype in this microdeletion syndrome. Only seven patients with a homozygous microdeletion that includes at least CHRNA7, and is inherited from both parents have been described in the literature. The aim of this study was to further describe the distinctive eye manifestations from the analysis in the three French patients diagnosed with the classical 1.5 Mb homozygous microdeletion. Patients' ages ranged from 30 months to 9 years, and included one sib pair. They all displayed a remarkably severe identifiable clinical phenotype that included congenital blindness and convulsive encephalopathy with inconstant abnormal movements. The ophthalmological examination revealed a lack of eye tracking, optic nerve pallor, an immature response with increased latencies with no response to the checkerboard stimulations at the visual evoked potential examination, and a distinctive retina dystrophy with a negative electroretinogram in which the "b" wave was smaller than the "a" wave after a dark adapted pupil and bright flash in all patients. Clear genotype-phenotype correlations emerged, showing that this eye phenotype was secondary to homozygous deletion of TRPM1, the gene responsible for autosomal recessive congenital stationary night blindness. The main differential diagnosis is ceroid lipofuscinosis.


Subject(s)
Blindness/genetics , Chromosome Disorders/genetics , Intellectual Disability/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Seizures/genetics , TRPM Cation Channels/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/pathology , Chromosomes, Human, Pair 15/genetics , Electroretinography , Eye/pathology , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Female , Genetic Association Studies , Genetic Diseases, X-Linked/genetics , Humans , Intellectual Disability/pathology , Male , Myopia/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Night Blindness/genetics , Optic Nerve/abnormalities , Retinal Dystrophies/genetics , Seizures/pathology
15.
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
16.
Am J Hum Genet ; 85(5): 720-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19896113

ABSTRACT

Night vision requires signaling from rod photoreceptors to adjacent bipolar cells in the retina. Mutations in the genes NYX and GRM6, expressed in ON bipolar cells, lead to a disruption of the ON bipolar cell response. This dysfunction is present in patients with complete X-linked and autosomal-recessive congenital stationary night blindness (CSNB) and can be assessed by standard full-field electroretinography (ERG), showing severely reduced rod b-wave amplitude and slightly altered cone responses. Although many cases of complete CSNB (cCSNB) are caused by mutations in NYX and GRM6, in approximately 60% of the patients the gene defect remains unknown. Animal models of human diseases are a good source for candidate genes, and we noted that a cCSNB phenotype present in homozygous Appaloosa horses is associated with downregulation of TRPM1. TRPM1, belonging to the family of transient receptor potential channels, is expressed in ON bipolar cells and therefore qualifies as an excellent candidate. Indeed, mutation analysis of 38 patients with CSNB identified ten unrelated cCSNB patients with 14 different mutations in this gene. The mutation spectrum comprises missense, splice-site, deletion, and nonsense mutations. We propose that the cCSNB phenotype in these patients is due to the absence of functional TRPM1 in retinal ON bipolar cells.


Subject(s)
Genes, Recessive , Mutation , Night Blindness/congenital , Night Blindness/genetics , TRPM Cation Channels/genetics , Electroretinography , Female , Heterozygote , Homozygote , Humans , Male , Models, Genetic , Night Blindness/physiopathology , Nuclear Family , Pedigree
17.
Am J Ophthalmol ; 133(5): 723-5, 2002 May.
Article in English | MEDLINE | ID: mdl-11992882

ABSTRACT

PURPOSE: To report a case of acute comitant esotropia successfully treated with suboccipital decompression in a 9-year-old male patient with Chiari I malformation. DESIGN: Interventional case report. METHODS: A 9-year-old male with Chiari I malformation had acute onset of diplopia, headache, and comitant esotropia. RESULTS: About 9 months after suboccipital decompression, diplopia resolved and there was near orthophoria on examination 15 months after surgery. CONCLUSION: In view of our case and after a review of literature, we advocate primary suboccipital decompression to treat acute comitant esotropia in patients with Chiari I malformation. A follow-up period of at least 1 year rather than 6 months seems necessary to assess surgery effects.


Subject(s)
Arnold-Chiari Malformation/surgery , Esotropia/surgery , Acute Disease , Arnold-Chiari Malformation/complications , Arnold-Chiari Malformation/diagnosis , Child , Decompression, Surgical , Diplopia/etiology , Diplopia/physiopathology , Esotropia/etiology , Headache/etiology , Headache/physiopathology , Humans , Magnetic Resonance Imaging , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...