Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 597
Filter
1.
Lancet ; 404(10456): 962-970, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244273

ABSTRACT

BACKGROUND: Leber congenital amaurosis 1 (LCA1), caused by mutations in GUCY2D, is a rare inherited retinal disease that typically causes blindness in early childhood. The aim of this study was to evaluate the safety and preliminary efficacy of ascending doses of ATSN-101, a subretinal AAV5 gene therapy for LCA1. METHODS: 15 patients with genetically confirmed biallelic mutations in GUCY2D were included in this phase 1/2 study. All patients received unilateral subretinal injections of ATSN-101. In the dose-escalation phase, three adult cohorts (n=3 each) were treated with three ascending doses: 1·0 × 1010 vg/eye (low dose), 3·0 × 1010 vg/eye (middle dose), and 1·0 × 1011 vg/eye (high dose). In the dose-expansion phase, one adult cohort (n=3) and one paediatric cohort (n=3) were treated at the high dose. The primary endpoint was the incidence of treatment-emergent adverse events (TEAEs), and secondary endpoints included full-field stimulus test (FST) and best-corrected visual acuity (BCVA). A multi-luminance mobility test (MLMT) was also done. Data through the 12-month main study period are reported. FINDINGS: Patients were enrolled between Sept 12, 2019, and May 5, 2022. A total of 68 TEAEs were observed, 56 of which were related to the surgical procedure. No serious TEAE was related to the study drug. Ocular inflammation was mild and reversible with steroid treatment. For patients who received the high dose, mean change in dark-adapted FST was 20·3 decibels (dB; 95% CI 6·6 to 34·0) for treated eyes and 1·1 dB (-3·7 to 5·9) for untreated eyes at month 12 (white stimulus); improvements were first observed at day 28 and persisted over 12 months (p=0·012). Modest improvements in BCVA were also observed (p=0·10). Three of six patients who received the high dose and did the MLMT achieved the maximum score in the treated eye. INTERPRETATION: ATSN-101 is well tolerated 12 months after treatment, with no drug-related serious adverse events. Clinically significant improvements in retinal sensitivity were sustained in patients receiving the high dose. FUNDING: Atsena Therapeutics.


Subject(s)
Genetic Therapy , Guanylate Cyclase , Leber Congenital Amaurosis , Receptors, Cell Surface , Adolescent , Adult , Child , Humans , Genetic Therapy/methods , Guanylate Cyclase/genetics , Injections, Intraocular , Leber Congenital Amaurosis/genetics , Mutation , Receptors, Cell Surface/genetics , Treatment Outcome , Visual Acuity
2.
Nat Commun ; 15(1): 5943, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009597

ABSTRACT

Inherited retinopathies are devastating diseases that in most cases lack treatment options. Disease-modifying therapies that mitigate pathophysiology regardless of the underlying genetic lesion are desirable due to the diversity of mutations found in such diseases. We tested a systems pharmacology-based strategy that suppresses intracellular cAMP and Ca2+ activity via G protein-coupled receptor (GPCR) modulation using tamsulosin, metoprolol, and bromocriptine coadministration. The treatment improves cone photoreceptor function and slows degeneration in Pde6ßrd10 and RhoP23H/WT retinitis pigmentosa mice. Cone degeneration is modestly mitigated after a 7-month-long drug infusion in PDE6A-/- dogs. The treatment also improves rod pathway function in an Rpe65-/- mouse model of Leber congenital amaurosis but does not protect from cone degeneration. RNA-sequencing analyses indicate improved metabolic function in drug-treated Rpe65-/- and rd10 mice. Our data show that catecholaminergic GPCR drug combinations that modify second messenger levels via multiple receptor actions provide a potential disease-modifying therapy against retinal degeneration.


Subject(s)
Disease Models, Animal , Drug Repositioning , Retinitis Pigmentosa , Animals , Mice , Dogs , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Mutation , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Mice, Knockout , Leber Congenital Amaurosis/drug therapy , Leber Congenital Amaurosis/genetics , Bromocriptine/pharmacology , Bromocriptine/therapeutic use , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism , Humans , Drug Therapy, Combination , Mice, Inbred C57BL , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Female , Cyclic AMP/metabolism , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Male , Calcium/metabolism
3.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38892339

ABSTRACT

Leber congenital amaurosis (LCA)/early-onset severe retinal dystrophy (EOSRD) stand as primary causes of incurable childhood blindness. This study investigates the clinical and molecular architecture of syndromic and non-syndromic LCA/EOSRD within a Chilean cohort (67 patients/60 families). Leveraging panel sequencing, 95.5% detection was achieved, revealing 17 genes and 126 variants (32 unique). CRB1, LCA5, and RDH12 dominated (71.9%), with CRB1 being the most prevalent (43.8%). Notably, four unique variants (LCA5 p.Glu415*, CRB1 p.Ser1049Aspfs*40 and p.Cys948Tyr, RDH12 p.Leu99Ile) constituted 62.7% of all disease alleles, indicating their importance for targeted analysis in Chilean patients. This study underscores a high degree of inbreeding in Chilean families affected by pediatric retinal blindness, resulting in a limited mutation repertoire. Furthermore, it complements and reinforces earlier reports, indicating the involvement of ADAM9 and RP1 as uncommon causes of LCA/EOSRD. These data hold significant value for patient and family counseling, pharmaceutical industry endeavors in personalized medicine, and future enrolment in gene therapy-based treatments, particularly with ongoing trials (LCA5) or advancing preclinical developments (CRB1 and RDH12).


Subject(s)
Mutation , Retinal Dystrophies , Humans , Retinal Dystrophies/genetics , Retinal Dystrophies/therapy , Retinal Dystrophies/diagnosis , Chile/epidemiology , Male , Female , Child , Child, Preschool , Alcohol Oxidoreductases/genetics , Membrane Proteins/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Leber Congenital Amaurosis/diagnosis , Pedigree , Nerve Tissue Proteins/genetics , Adolescent , Alleles , Genetic Variation , Eye Diseases, Hereditary
4.
Commun Biol ; 7(1): 676, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830954

ABSTRACT

TRiC/CCT is a chaperonin complex required for the folding of cytoplasmic proteins. Although mutations in each subunit of TRiC/CCT are associated with various human neurodegenerative diseases, their impact in mammalian models has not yet been examined. A compound heterozygous mutation in CCT2 (p.[Thr400Pro]; p.[Arg516His]) is causal for Leber congenital amaurosis. Here, we generate mice carrying each mutation and show that Arg516His (R516H) homozygosity causes photoreceptor degeneration accompanied by a significant depletion of TRiC/CCT substrate proteins in the retina. In contrast, Thr400Pro (T400P) homozygosity results in embryonic lethality, and the compound heterozygous mutant (T400P/R516H) mouse showed aberrant cone cell lamination and died 2 weeks after birth. Finally, CCDC181 is identified as a interacting protein for CCTß protein, and its localization to photoreceptor connecting cilia is compromised in the mutant mouse. Our results demonstrate the distinct impact of each mutation in vivo and suggest a requirement for CCTß in ciliary maintenance.


Subject(s)
Chaperonin Containing TCP-1 , Disease Models, Animal , Leber Congenital Amaurosis , Animals , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Mice , Chaperonin Containing TCP-1/genetics , Chaperonin Containing TCP-1/metabolism , Mutation , Heterozygote , Mice, Inbred C57BL
5.
Nat Commun ; 15(1): 5471, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942756

ABSTRACT

The clinical success of CRISPR therapies hinges on the safety and efficacy of Cas proteins. The Cas9 from Francisella novicida (FnCas9) is highly precise, with a negligible affinity for mismatched substrates, but its low cellular targeting efficiency limits therapeutic use. Here, we rationally engineer the protein to develop enhanced FnCas9 (enFnCas9) variants and broaden their accessibility across human genomic sites by ~3.5-fold. The enFnCas9 proteins with single mismatch specificity expanded the target range of FnCas9-based CRISPR diagnostics to detect the pathogenic DNA signatures. They outperform Streptococcus pyogenes Cas9 (SpCas9) and its engineered derivatives in on-target editing efficiency, knock-in rates, and off-target specificity. enFnCas9 can be combined with extended gRNAs for robust base editing at sites which are inaccessible to PAM-constrained canonical base editors. Finally, we demonstrate an RPE65 mutation correction in a Leber congenital amaurosis 2 (LCA2) patient-specific iPSC line using enFnCas9 adenine base editor, highlighting its therapeutic utility.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Francisella , Gene Editing , Humans , Gene Editing/methods , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Francisella/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Leber Congenital Amaurosis/genetics , Streptococcus pyogenes/genetics , HEK293 Cells , Mutation , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Protein Engineering/methods , Genome, Human
6.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38811164

ABSTRACT

The canonical visual cycle employing RPE65 as the retinoid isomerase regenerates 11-cis-retinal to support both rod- and cone-mediated vision. Mutations of RPE65 are associated with Leber congenital amaurosis that results in rod and cone photoreceptor degeneration and vision loss of affected patients at an early age. Dark-reared Rpe65-/- mouse has been known to form isorhodopsin that employs 9-cis-retinal as the photosensitive chromophore. The mechanism regulating 9-cis-retinal synthesis and the role of the endogenous 9-cis-retinal in cone survival and function remain largely unknown. In this study, we found that ablation of fatty acid transport protein-4 (FATP4), a negative regulator of 11-cis-retinol synthesis catalyzed by RPE65, increased the formation of 9-cis-retinal, but not 11-cis-retinal, in a light-independent mechanism in both sexes of RPE65-null rd12 mice. Both rd12 and rd12;Fatp4-/- mice contained a massive amount of all-trans-retinyl esters in the eyes, exhibiting comparable scotopic vision and rod degeneration. However, expression levels of M- and S-opsins as well as numbers of M- and S-cones surviving in the superior retinas of rd12;Fatp4-/ - mice were at least twofold greater than those in age-matched rd12 mice. Moreover, FATP4 deficiency significantly shortened photopic b-wave implicit time, improved M-cone visual function, and substantially deaccelerated the progression of cone degeneration in rd12 mice, whereas FATP4 deficiency in mice with wild-type Rpe65 alleles neither induced 9-cis-retinal formation nor influenced cone survival and function. These results identify FATP4 as a new regulator of synthesis of 9-cis-retinal, which is a "cone-tropic" chromophore supporting cone survival and function in the retinas with defective RPE65.


Subject(s)
Fatty Acid Transport Proteins , Leber Congenital Amaurosis , Retinal Cone Photoreceptor Cells , Animals , Retinal Cone Photoreceptor Cells/metabolism , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Mice , Fatty Acid Transport Proteins/metabolism , Fatty Acid Transport Proteins/genetics , Male , Female , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism , cis-trans-Isomerases/deficiency , Cell Survival , Mice, Knockout , Diterpenes , Vision, Ocular/physiology , Disease Models, Animal , Mice, Inbred C57BL , Retinaldehyde
7.
Am J Ophthalmol ; 265: 257-274, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38777102

ABSTRACT

PURPOSE: To investigate autism spectrum disorder (ASD) indicators in children with Leber congenital amaurosis (LCA). STUDY DESIGN: Cross-sectional, prospective, and correlational study. METHODS: Setting: It was conducted at the Institute of Ocular Genetics, the Department of Ophthalmology at Federal University of São Paulo (UNIFESP), and the Autism Spectrum Disorder Laboratory, in São Paulo, Brazil. PATIENT POPULATION: Participants included patients aged 2 to 16 years with LCA confirmed by genetic testing. There were 20 individuals with ciliopathies (LCA cilio) and 26 with other gene mutations (LCA other). As intervention, the instrument used for ASD screening was the Autism Behavior Checklist (ABC). Marginal descriptive analyses, non-parametric tests, and a linear regression model were conducted. The main outcomes were the scores on the tests correlated with clinical variables. RESULTS: Of the 46 participants, 6 had ASD scores. There was no statistically significant correlation between the different groups (LCA cilio and LCA other) (p = 0.438). There was no statistically significant correlation between age and ASD (p = 0.308). However, there was a statistically significant correlation between visual acuity and ASD (p = 0.008) and between male gender and ASD (p = 0.025). CONCLUSIONS: This study suggests that there is no correlation between LCA cilio, LCA other and ASD. These findings bring new insights to the existing literature, which previously lacked robust data on the relationship between LCA and ASD. These data demonstrate that visual acuity plays a crucial role in the development of children with visual impairment as poorer visual acuity is associated with a higher incidence of ASD. Based on this study, early interventions can be designed, especially for individuals without light perception, with the aim of maximizing their developmental outcomes. Furthermore, such data indicates that any improvement in visual acuity outcomes in treatment clinical trials become relevant for child development. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.


Subject(s)
Autism Spectrum Disorder , Leber Congenital Amaurosis , Humans , Child , Male , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/physiopathology , Female , Adolescent , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/diagnosis , Leber Congenital Amaurosis/physiopathology , Child, Preschool , Prospective Studies , Cross-Sectional Studies , Visual Acuity/physiology , Mutation , Eye Proteins/genetics
8.
Life Sci Alliance ; 7(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38570189

ABSTRACT

Crumbs homolog 1 (CRB1) is one of the key genes linked to retinitis pigmentosa and Leber congenital amaurosis, which are characterized by a high clinical heterogeneity. The Crumbs family member CRB2 has a similar protein structure to CRB1, and in zebrafish, Crb2 has been shown to interact through the extracellular domain. Here, we show that CRB1 and CRB2 co-localize in the human retina and human iPSC-derived retinal organoids. In retina-specific pull-downs, CRB1 was enriched in CRB2 samples, supporting a CRB1-CRB2 interaction. Furthermore, novel interactors of the crumbs complex were identified, representing a retina-derived protein interaction network. Using co-immunoprecipitation, we further demonstrate that human canonical CRB1 interacts with CRB1 and CRB2, but not with CRB3, which lacks an extracellular domain. Next, we explored how missense mutations in the extracellular domain affect CRB1-CRB2 interactions. We observed no or a mild loss of CRB1-CRB2 interaction, when interrogating various CRB1 or CRB2 missense mutants in vitro. Taken together, our results show a stable interaction of human canonical CRB2 and CRB1 in the retina.


Subject(s)
Leber Congenital Amaurosis , Retinitis Pigmentosa , Animals , Humans , Zebrafish/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Retina/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Carrier Proteins/metabolism
9.
Stem Cell Res ; 77: 103413, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631180

ABSTRACT

Leber Congenital Amaurosis 2 is an early onset retinal dystrophy that occurs due to mutation in RPE65 gene. Here, we report the generation of two patient specific induced pluripotent stem cell lines harboring nonsense mutations in exon 7 (c.646A > T) and exon 9 (c.992G > A) of RPE65 gene, respectively, which leads to premature translational termination and formation of defective protein. These lines were generated by the reprogramming of human dermal fibroblast cells using integration-free, episomal constructs expressing stemness genes. The stable lines maintained a normal karyotype, expressed the key stemness factors, underwent trilineage differentiation, and maintained their genetic identity and genomic integrity.


Subject(s)
Induced Pluripotent Stem Cells , Leber Congenital Amaurosis , cis-trans-Isomerases , Humans , Induced Pluripotent Stem Cells/metabolism , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism , Mutation , Cell Line , Cell Differentiation , Male , Fibroblasts/metabolism , Female
10.
Graefes Arch Clin Exp Ophthalmol ; 262(9): 3029-3038, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38662103

ABSTRACT

PURPOSE: Leber congenital amaurosis (LCA) is a group of early-onset retinal degenerative disorders, resulting in blindness in children. This study aimed to describe the clinical and genetic characteristics of a cohort of patients with LCA and to investigate the retinal vascular characteristics in LCA patients. METHODS: Fifty-two children with LCA were included in the study. All patients underwent detailed ocular examinations. Electroretinography (ERG) was used to evaluate the retinal function. Optical coherence tomography (OCT) was used to assess the structure change of the retina for those patients who were able to cooperate very well. Panel-based next-generation sequencing was performed to identify pathogenic variants in genes associated with LCA. Diameters of the retinal vessels were measured using the EVision AI screening system with an artificial intelligence (AI) technique. An ultrasound Doppler was used to evaluate hemodynamic parameters, including peak systolic velocity (PSV), resistive index (RI), and pulsatility index (PI), in the ophthalmic, central retinal, posterior ciliary, carotid, and internal carotid as well as external carotid arteries in 12 patients aged from 3 to 14 years. RESULTS: We detected 75 pathogenic variants from ten genes of RPGRIP1, CEP290, GUCY2D, LCA5, AIPL1, CRB1, RPE65, CRX, RDH12, and TULP1, including 29 novel and 36 previously reported variants in 52 affected children with LCA, with the highest detective rate in RPGRIP1 (26.9%). Fundus appearance is diverse in patients with LCA, ranging from normal to severe peripheral or central retinopathy. Retinal vasculature was evaluated in 12 patients with different gene variants, showing narrowed arteries with an average diameter of 43.6 ± 3.8 µm compared to that of 51.7 ± 2.6 µm in the normal controls (P < 0.001, n = 12). Meanwhile, their hemodynamic parameters were changed as well in the ophthalmic artery (OA), with a decreased PSV (P = 0.0132, n = 12) and slightly increased PI (P = 0.0488, n = 12) compared to the normal controls. However, the hemodynamic parameters did not change significantly in the other vessels. CONCLUSIONS: Blood supply to the eyeball is predicted to be reduced in patients with LCA, presumably due to photoreceptor cell degeneration. The novel identified variants will expand the spectrum of variants in LCA-related genes and be useful for studying the molecular mechanisms of LCA.


Subject(s)
Electroretinography , Leber Congenital Amaurosis , Mutation , Tomography, Optical Coherence , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Leber Congenital Amaurosis/diagnosis , Male , Child , Female , Tomography, Optical Coherence/methods , Adolescent , Child, Preschool , Retinal Vessels , Eye Proteins/genetics , Visual Acuity , DNA/genetics , DNA Mutational Analysis , Retina/physiopathology
11.
Eye (Lond) ; 38(13): 2504-2515, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38627549

ABSTRACT

Biallelic mutations in the RPE65 gene affect nearly 8% of Leber Congenital Amaurosis and 2% of Retinitis Pigmentosa cases. Voretigene neparvovec (VN) is the first gene therapy approach approved for their treatment. To date, real life experience has demonstrated functional improvements following VN treatment, which are consistent with the clinical trials outcomes. However, there is currently no consensus on the characteristics for eligibility for VN treatment. We reviewed relevant literature to explore whether recommendations on patient eligibility can be extrapolated following VN marketing. We screened 166 papers through six research questions, following scoping reviews methodology, to investigate: (1) the clinical and genetic features considered in VN treatment eligibility; (2) the psychophysical tests and imaging modalities used in the pre-treatment and follow-up; (3) the potential correlations between visual function and retinal structure that can be used to define treatment impact on disease progression; (4) retinal degeneration; (5) the most advanced testing modalities; and (6) the impact of surgical procedure on treatment outcomes. Current gaps concerning patients' eligibility in clinical settings, such as pre-treatment characteristics and outcomes are not consistently reported across the studies. No upper limit of retinal degeneration can be defined as the univocal factor in patient eligibility, although evidence suggested that the potential for function rescue is related to the preservation of photoreceptors before treatment. In general, paediatric patients retain more viable cells, present a less severe disease stage and show the highest potential for improvements, making them the most suitable candidates for treatment.


Subject(s)
Genetic Therapy , Mutation , Retinal Dystrophies , cis-trans-Isomerases , Humans , cis-trans-Isomerases/genetics , Genetic Therapy/methods , Retinal Dystrophies/genetics , Retinal Dystrophies/therapy , Clinical Trials as Topic , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Leber Congenital Amaurosis/physiopathology , Dependovirus/genetics , Visual Acuity/physiology , Genetic Vectors
12.
Invest Ophthalmol Vis Sci ; 65(3): 11, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38466290

ABSTRACT

Purpose: The purpose of this study was to investigate the genotypic and phenotypic characteristics of CRB1-associated early onset retinal dystrophy (CRB1-eoRD) and retinal architecture by swept-source optical coherence tomography (SS-OCT). Methods: Eleven probands with CRB1-eoRD were recruited. Clinical information, genetic analysis, and comprehensive ophthalmic examinations including SS-OCT and SS-OCT angiography (SS-OCTA) were conducted. Results: A total of 81.8% (9/11) of CRB1-eoRD presented as Leber congenital amaurosis (LCA). Common clinical manifestations included coin-like yellow-white retinal spots (20/22, 90.9%) and para-arteriolar retinal pigment epithelial retention (12/22, 54.5%). Nineteen different CRB1 variants were detected in our case series, including 12 missense, 3 frameshifts, 3 nonsense, and 1 splicing. Of them, 12 variants had been reported, and 7 were novel. SS-OCT showed thinner central macula (the LCA group, P < 0.0001), thicker total retina (P < 0.0001), thinner outer retina (P < 0.05), and thicker inner retina (P < 0.0001) compared with the healthy control. The inner/outer (I/O) retina thickness ratio of CRB1-eoRD was 3.0, higher than the healthy control of 1.2 and other inherited retinal diseases (IRDs) of 2.2 (P < 0.0001 and P = 0.0027, respectively). SS-OCTA revealed an increased vascular density and perfusion area of the superficial vascular complex and deep vascular complex in CRB1-eoRD. Conclusions: LCA emerges as a frequently occurring phenotype in CRB1-eoRD. The unique features of SS-OCT and SS-OCTA are illustrated, and the novel biomarker, I/O ratio, may facilitate early diagnosis. The insights gained from this study hold significant value in determining the treatment window for potential forthcoming CRB1 gene therapy.


Subject(s)
Leber Congenital Amaurosis , Retinal Dystrophies , Humans , Retina/diagnostic imaging , Retinal Dystrophies/diagnosis , Retinal Dystrophies/genetics , Leber Congenital Amaurosis/diagnosis , Leber Congenital Amaurosis/genetics , Genotype , Phenotype , Eye Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics
13.
Am J Ophthalmol ; 263: 168-178, 2024 07.
Article in English | MEDLINE | ID: mdl-38461945

ABSTRACT

PURPOSE: To evaluate ocular and retinal features of CRB1-associated early onset severe retinal dystrophy/Leber congenital amaurosis (EOSRD/LCA) for age-related changes. DESIGN: Retrospective cohort study. METHODS: Sixteen pediatric patients with biallelic CRB1 EOSRD/LCA who had been followed for up to 18 years were reviewed. Results of comprehensive ophthalmic examinations-including visual acuity, refractive error, dark-adapted visual threshold, Goldmann perimetry, and macular optical coherence tomography (OCT)-were analyzed for significant age-related changes using mixed-effects models. RESULTS: Visual acuity dark-adapted visual sensitivity, and area of seeing visual field (all subnormal from the earliest ages recorded) declined with increasing age. Hyperopia was stable through childhood and adolescence. In CRB1 EOSRD/LCA, OCT extrafoveal inner and outer laminar thicknesses exceeded those in controls but varied little with age, and foveal metrics (depth, breadth, thickness at rim) differed significantly from those in controls, but variations in foveal metrics were not associated with declines in acuity. CONCLUSIONS: From the youngest ages, retinal and visual function is significantly subnormal and becomes progressively compromized. A goal of future therapies should be intervention at young ages, when there is more function to be rescued.


Subject(s)
Eye Proteins , Leber Congenital Amaurosis , Membrane Proteins , Nerve Tissue Proteins , Tomography, Optical Coherence , Visual Acuity , Visual Fields , Humans , Child , Retrospective Studies , Visual Acuity/physiology , Male , Adolescent , Female , Child, Preschool , Eye Proteins/genetics , Nerve Tissue Proteins/genetics , Membrane Proteins/genetics , Visual Fields/physiology , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/physiopathology , Visual Field Tests , Retinal Dystrophies/genetics , Retinal Dystrophies/physiopathology , Retinal Dystrophies/diagnosis , Dark Adaptation/physiology , Infant , Aging/physiology , Follow-Up Studies , Retina/physiopathology , Young Adult
14.
J Med Genet ; 61(7): 613-620, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38499336

ABSTRACT

BACKGROUND: As gene-specific therapy for inherited retinal dystrophy (IRD) advances, unified variant interpretation across institutes is becoming increasingly important. This study aims to update the genetic findings of 86 retinitis pigmentosa (RP)-related genes in a large number of Japanese patients with RP by applying the standardised variant interpretation guidelines for Japanese patients with IRD (J-IRD-VI guidelines) built upon the American College of Medical Genetics and Genomics and the Association for Molecular Pathology rules, and assess the contribution of these genes in RP-allied diseases. METHODS: We assessed 2325 probands with RP (n=2155, including n=1204 sequenced previously with the same sequencing panel) and allied diseases (n=170, newly analysed), including Usher syndrome, Leber congenital amaurosis and cone-rod dystrophy (CRD). Target sequencing using a panel of 86 genes was performed. The variants were interpreted according to the J-IRD-VI guidelines. RESULTS: A total of 3564 variants were detected, of which 524 variants were interpreted as pathogenic or likely pathogenic. Among these 524 variants, 280 (53.4%) had been either undetected or interpreted as variants of unknown significance or benign variants in our earlier study of 1204 patients with RP. This led to a genetic diagnostic rate in 38.6% of patients with RP, with EYS accounting for 46.7% of the genetically solved patients, showing a 9% increase in diagnostic rate from our earlier study. The genetic diagnostic rate for patients with CRD was 28.2%, with RP-related genes significantly contributing over other allied diseases. CONCLUSION: A large-scale genetic analysis using the J-IRD-VI guidelines highlighted the population-specific genetic findings for Japanese patients with IRD; these findings serve as a foundation for the clinical application of gene-specific therapies.


Subject(s)
Retinitis Pigmentosa , Female , Humans , Male , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/pathology , East Asian People/genetics , Genetic Predisposition to Disease , Genetic Variation , Japan , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Mutation , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Usher Syndromes/genetics
15.
Stem Cell Res ; 77: 103380, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38479331

ABSTRACT

Leber congenital amaurosis (LCA) is a congenital, early onset, autosomal recessive inherited retinal disease (IRD). This report describes an LCA12 patient-specific iPSC line (LVPEIi006-A), generated by the reprogramming of dermal fibroblasts using integration-free episomal plasmids.This disease-specific iPSC model carries a homozygous point mutation in RD3, within the donor splice site at the end of exon 2 (c.296 + 1G > A). The stable line at passage 15 has displayed a normal colony morphology, expressed multiple stemness and pluripotency markers, lost all transgenes, differentiated into cell types of all three germ layers, and maintained a normal karyotype.


Subject(s)
Homozygote , Induced Pluripotent Stem Cells , Leber Congenital Amaurosis , Induced Pluripotent Stem Cells/metabolism , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/pathology , Humans , Cell Line , Mutation , Cell Differentiation
16.
Clin Genet ; 106(2): 127-139, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38468396

ABSTRACT

Leber congenital amaurosis (LCA) and early-onset retinal degeneration (EORD) are inherited retinal diseases (IRD) characterized by early-onset vision impairment. Herein, we studied 15 Saudi families by whole exome sequencing (WES) and run-of-homozygosity (ROH) detection via AutoMap in 12/15 consanguineous families. This revealed (likely) pathogenic variants in 11/15 families (73%). A potential founder variant was found in RPGRIP1. Homozygous pathogenic variants were identified in known IRD genes (ATF6, CRB1, CABP4, RDH12, RIMS2, RPGRIP1, SPATA7). We established genotype-driven clinical reclassifications for ATF6, CABP4, and RIMS2. Specifically, we observed isolated IRD in the individual with the novel RIMS2 variant, and we found a retina-enriched RIMS2 isoform conserved but not annotated in mouse. The latter illustrates potential different phenotypic consequences of pathogenic variants depending on the particular tissue/cell-type specific isoforms they affect. Lastly, a compound heterozygous genotype in GUCY2D in one non-consanguineous family was demonstrated, and homozygous variants in novel candidate genes ATG2B and RUFY3 were found in the two remaining consanguineous families. Reporting these genes will allow to validate them in other IRD cohorts. Finally, the missing heritability of the two unsolved IRD cases may be attributed to variants in non-coding regions or structural variants that remained undetected, warranting future WGS studies.


Subject(s)
Consanguinity , Exome Sequencing , Pedigree , Phenotype , Humans , Female , Male , Retina/pathology , Homozygote , Retinal Diseases/genetics , Protein Isoforms/genetics , Exome/genetics , Mutation , Child , Genetic Predisposition to Disease , Leber Congenital Amaurosis/genetics , Cohort Studies , Genotype , Genetic Association Studies/methods
17.
Brain ; 147(9): 3234-3246, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38538211

ABSTRACT

Genetic diseases affecting the retina can result in partial or complete loss of visual function. Leber's congenital amaurosis (LCA) is a rare blinding disease, usually inherited in an autosomally recessive manner, with no cure. Retinal gene therapy has been shown to improve vision in LCA patients caused by mutations in the RPE65 gene (LCA2). However, little is known about how activity in central visual pathways is affected by the disease or by subsequent gene therapy. Functional MRI (fMRI) was used to assess retinal signal transmission in cortical and subcortical visual structures before and 1 year after retinal intervention. The fMRI paradigm consisted of 15-s blocks of flickering (8 Hz) black and white checkerboards interleaved with 15 s of blank (black) screen. Visual activation in the brain was assessed using the general linear model, with multiple comparisons corrected using the false discovery rate method. Response to visual stimulation through untreated eyes of LCA2 patients showed heightened fMRI responses in the superior colliculus and diminished activities in the lateral geniculate nucleus (LGN) compared to controls, indicating a shift in the patients' visual processing towards the retinotectal pathway. Following gene therapy, stimuli presented to the treated eye elicited significantly stronger fMRI responses in the LGN and primary visual cortex, indicating some re-engagement of the geniculostriate pathway (GS) pathway. Across patients, the post-treatment LGN fMRI responses correlated significantly with performance on a clinical test measuring light sensitivity. Our results demonstrate that the low vision observed in LCA2 patients involves a shift in visual processing toward the retinotectal pathway, and that gene therapy partially reinstates visual transmission through the GS pathway. This selective boosting of retinal output through the GS pathway and its correlation to improved visual performance, following several years of degenerative retinal disease, is striking. However, while retinal gene therapy and other ocular interventions have given hope to RPE65 patients, it may take years before development of therapies tailored to treat the diseases in other low vision patients are available. Our demonstration of a shift toward the retinotectal pathway in these patients may spur the development of new tools and rehabilitation strategies to help maximize the use of residual visual abilities and augment experience-dependent plasticity.


Subject(s)
Genetic Therapy , Leber Congenital Amaurosis , Magnetic Resonance Imaging , Visual Pathways , Humans , Genetic Therapy/methods , Visual Pathways/physiopathology , Visual Pathways/diagnostic imaging , Adult , Male , Female , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Leber Congenital Amaurosis/physiopathology , Young Adult , Photic Stimulation/methods , Middle Aged , cis-trans-Isomerases/genetics
18.
Cell ; 187(6): 1387-1401.e13, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38412859

ABSTRACT

The Crumbs homolog 1 (CRB1) gene is associated with retinal degeneration, most commonly Leber congenital amaurosis (LCA) and retinitis pigmentosa (RP). Here, we demonstrate that murine retinas bearing the Rd8 mutation of Crb1 are characterized by the presence of intralesional bacteria. While normal CRB1 expression was enriched in the apical junctional complexes of retinal pigment epithelium and colonic enterocytes, Crb1 mutations dampened its expression at both sites. Consequent impairment of the outer blood retinal barrier and colonic intestinal epithelial barrier in Rd8 mice led to the translocation of intestinal bacteria from the lower gastrointestinal (GI) tract to the retina, resulting in secondary retinal degeneration. Either the depletion of bacteria systemically or the reintroduction of normal Crb1 expression colonically rescued Rd8-mutation-associated retinal degeneration without reversing the retinal barrier breach. Our data elucidate the pathogenesis of Crb1-mutation-associated retinal degenerations and suggest that antimicrobial agents have the potential to treat this devastating blinding disease.


Subject(s)
Nerve Tissue Proteins , Retinal Degeneration , Animals , Mice , Bacterial Translocation , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
19.
Ophthalmic Genet ; 45(3): 303-312, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38323530

ABSTRACT

INTRODUCTION: Mutations in the retinal pigment epithelial 65 kilodalton protein (RPE65) gene are associated with various inherited retinal diseases (IRDs), including Leber congenital amaurosis (LCA), early-onset severe retinal dystrophy (EOSRD), and retinitis pigmentosa (RP). We screened for mutations in RPE65 in a series of Indian patients with these IRDs to determine the frequency/types of mutations and to describe the associated phenotypes. MATERIALS AND METHODS: Diagnosis of LCA, EOSRD, and RP was made by standard and pre-defined criteria. Patients were evaluated by clinical, retinal imaging, and electrophysiological parameters. Genomic DNA from patients and available family members were used for identifying mutations by direct Sanger sequencing of the RPE65 gene or targeted NGS gene panel for IRDs covering 260+ genes. Variations detected were tested in healthy control populations and for co-segregation with the disease in available family members. RESULTS: Mutations were found in eight patients, out of 220 total cases screened, all homozygous for the respective mutant alleles. Seven patients had mutations leading to premature termination codons and one patient had a missense change. The onset of visual loss ranged from birth to <2 years of life. At presentation, RPE mottling in the background retina was present in all cases with macular involvement in five cases with or without vascular attenuation and optic disc pallor. CONCLUSION: RPE65 mutations in this series were found in 3.6% of cases associated with severe, early-onset disease, with consistent RPE mottling and variable manifestations with regard to the extent of disc pallor, arteriolar attenuation, and appearance of the macula.


Subject(s)
Leber Congenital Amaurosis , Mutation , Retinal Dystrophies , Retinitis Pigmentosa , cis-trans-Isomerases , Humans , cis-trans-Isomerases/genetics , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/diagnosis , Male , India/epidemiology , Female , Retinal Dystrophies/genetics , Retinal Dystrophies/diagnosis , Adult , Child , Tertiary Care Centers , Child, Preschool , Pedigree , Adolescent , Electroretinography , Young Adult , Visual Acuity/physiology , DNA Mutational Analysis , Infant , Eye Diseases, Hereditary
20.
Int J Mol Sci ; 25(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38279252

ABSTRACT

This retrospective study examines the clinical characteristics and underlying genetic variants that exist in a Leber congenital amaurosis (LCA) patient cohort evaluated at the inherited retinal disease (IRD) clinic at the University of Minnesota (UMN)/M Health System. Our LCA cohort consisted of 33 non-syndromic patients and one patient with Joubert syndrome. We report their relevant history, clinical findings, and genetic testing results. We monitored disease presentation utilizing ocular coherence tomography (OCT) and fundus autofluorescence (FAF). Electroretinogram testing (ERG) was performed in patients when clinically indicated. Next-generation sequencing (NGS) and genetic counseling was offered to all evaluated patients. Advanced photoreceptor loss was noted in 85.7% of the subjects. All patients who underwent FAF had findings of either a ring of macular hypo/hyper AF or peripheral hypo-AF. All patients had abnormal ERG findings. A diagnostic genetic test result was identified in 74.2% of the patients via NGS single-gene testing or panel testing. Two patients in our cohort qualified for Luxturna® and both received treatment at the time of this study. These data will help IRD specialists to understand the genetic variants and clinical presentations that characterize our patient population in the Midwest region of the United States.


Subject(s)
Leber Congenital Amaurosis , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Retrospective Studies , Mutation , Retina/pathology , Genetic Therapy , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL