Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 600
Filter
1.
Retina ; 44(10): 1836-1844, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39287548

ABSTRACT

PURPOSE: To ascertain the characteristics of achromatopsia (ACHM) in Japan by analyzing the genetic and phenotypic features of patients with ACHM. METHODS: The medical records of 52 patients from 47 Japanese families who were clinically diagnosed with ACHM were reviewed in this retrospective observational study. RESULTS: Thirty-six causative variants of ACHM were identified in 26 families via whole-exome sequencing: PDE6C (12 families), CNGA3 (10 families), CNGB3 (two families), and GNAT2 (two families). However, none of the 6 causative variants that are known to cause ACHM, or the 275 other genes listed in RetNet, were observed in 19 families. A significant trend toward older age and worsening of ellipsoid zone disruption on optical coherence tomography images was observed (P < 0.01). Progressive ellipsoid zone disruptions were observed in 13 eyes of seven patients during the follow-up visits. These patients harbored one or more variants in PDE6C. CONCLUSION: The ACHM phenotype observed in this study was similar to those observed in previous reports; however, the causative gene variants differed from those in Europe. The low identification ratio of causative genes in whole-exome sequencing suggests the presence of unique hotspots in Japanese patients with ACHM that were not detectable via ordinal whole-exome sequencing.


Subject(s)
Color Vision Defects , Exome Sequencing , Tomography, Optical Coherence , Humans , Color Vision Defects/genetics , Color Vision Defects/diagnosis , Male , Female , Retrospective Studies , Japan/epidemiology , Adult , Middle Aged , Child , Adolescent , Young Adult , Mutation , Pedigree , Visual Acuity , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Phenotype , Child, Preschool , Eye Proteins/genetics , Aged , Electroretinography , Cyclic Nucleotide-Gated Cation Channels/genetics , DNA Mutational Analysis
2.
Sci Rep ; 14(1): 22090, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333705

ABSTRACT

Retinitis pigmentosa (RP) is a progressive and degenerative retinal disease resulting in severe vision loss. RP have been extensively studied for pathogenetic mechanisms and treatments. Yet there is little information about alterations of RP associated proteins in phosphodiesterase 6 beta (Pde6b) mutated model. To explore the roles of RP causing proteins, we performed a label free quantitative mass spectrometry based proteomic analysis in rd10 mouse retinas. 3737 proteins were identified at the degenerative time points in rd10 mice. 222 and 289 differentially expressed proteins (DEPs) (fold change, FC > 2, p < 0.05) were detected at 5 and 8 weeks. Based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, visual perception and phototransduction were severely affected. The downregulated DEPs were significantly enriched in cilium assembly and protein localization. 25 decreased DEPs causing autosomal recessive/dominant retinitis pigmentosa were visualized by heatmaps. Protein-protein interaction network represented 13 DEPs interacted directly with Pde6b protein. 25 DEPs causing RP were involved in phototransduction, visual perception, response to stimulus, protein localization and cilium assembly pathways. The significantly reduced expressions of DEPs were further validated by quantitative reverse transcription polymerase chain reaction (qPCR), Western blots (WB) and immunohistochemistry (IHC). This study revealed the molecular mechanisms underlying early and late stage of RP, as well as changes of RP-causing proteins.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Disease Models, Animal , Mutation , Proteomics , Retinitis Pigmentosa , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Proteomics/methods , Mice , Eye Proteins/metabolism , Eye Proteins/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retina/metabolism , Retina/pathology , Protein Interaction Maps , Proteome/metabolism
3.
BMC Ophthalmol ; 24(1): 353, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160471

ABSTRACT

BACKGROUND: Mutations in PDE6A and PDE6B are known to cause autosomal recessive RP in humans, On the other hand, mutations in PDE6G are rare but can lead to severe early-onset RP. CASE PRESENTATION: An 8-year-old Chinese boy was referred to our hospital for poor vision issues. Refraction with cycloplegia showed high hyperopia with astigmatism both eyes. Funduscopic examination revealed typical bone spicule-type pigment deposits in the periphery and midperiphery. The patient was given glasses and a whole exome sequencing containing mitochondrial genes was performed. The results of genetic testing showed that there was a heterozygous frameshift mutation and a segment deletion in the proband's PDE6G gene. Analysis of the parental genes showed that frameshift mutation was inherited from the proband's mother and segment deletion from his father. CONCLUSIONS: In this paper, we give a firsthand report that the complex heterozygous mutations of PDE6G gene can causes autosomal recessiveRP (arRP), which expands the understanding of the pathogenic genes of RP.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Retinitis Pigmentosa , Humans , Male , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Child , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/diagnosis , Pedigree , DNA Mutational Analysis , Mutation , Frameshift Mutation , Tomography, Optical Coherence , Visual Acuity , Exome Sequencing
4.
BMC Med Genomics ; 17(1): 173, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38956522

ABSTRACT

BACKGROUND: Autosomal recessive non-syndromic hearing loss (NSHL) and cone dystrophies (CODs) are highly genetically and phenotypically heterogeneous disorders. In this study, we applied the whole exome sequencing (WES) to find the cause of HL and COD in an Iranian consanguineous family with three affected individuals. METHODS: Three members from an Iranian consanguineous family who were suffering from NSHL and visual impairment were ascertained in this study. Comprehensive clinical evaluations and genetic analysis followed by bioinformatic and co-segregation studies were performed to diagnose the cause of these phenotypes. Data were collected from 2020 to 2022. RESULTS: All cases showed congenital bilateral NSHL, decreased visual acuity, poor color discrimination, photophobia and macular atrophy. Moreover, cornea, iris and anterior vitreous were within normal limit in both eyes, decreased foveal sensitivity, central scotoma and generalized depression of visual field were seen in three cases. WES results showed two variants, a novel null variant (p.Trp548Ter) in the PDE6C gene causing COD type 4 (Achromatopsia) and a previously reported variant (p.Ile84Thr) in the PDZD7 gene causing NSHL. Both variants were found in the cis configuration on chromosome 10 with a genetic distance of about 8.3 cM, leading to their co-inheritance. However, two diseases could appear independently in subsequent generations due to crossover during meiosis. CONCLUSIONS: Here, we could successfully determine the etiology of a seemingly complex phenotype in two adjacent genes. We identified a novel variant in the PDE6C gene, related to achromatopsia. Interestingly, this variant could cooperatively cause visual disorders: cone dystrophy and cone-rod dystrophy.


Subject(s)
Color Vision Defects , Cyclic Nucleotide Phosphodiesterases, Type 6 , Pedigree , Adult , Child , Female , Humans , Male , Color Vision Defects/genetics , Consanguinity , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Exome Sequencing , Eye Proteins , Hearing Loss/genetics , Iran , Mutation , Phenotype
5.
Mol Metab ; 88: 101994, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39032643

ABSTRACT

OBJECTIVE: Retinitis pigmentosa (RP) is a hereditary retinal disease characterized by progressive photoreceptor degeneration, leading to vision loss. The best hope for a cure for RP lies in gene therapy. However, given that RP patients are most often diagnosed in the midst of ongoing photoreceptor degeneration, it is unknown how the retinal proteome changes as RP disease progresses, and which changes can be prevented, halted, or reversed by gene therapy. METHODS: Here, we used a Pde6b-deficient RP gene therapy mouse model and performed untargeted proteomic analysis to identify changes in protein expression during degeneration and after treatment. RESULTS: We demonstrated that Pde6b gene restoration led to a novel form of homeostatic plasticity in rod phototransduction which functionally compensates for the decreased number of rods. By profiling protein levels of metabolic genes and measuring metabolites, we observed an upregulation of proteins associated with oxidative phosphorylation in mutant and treated photoreceptors. CONCLUSION: In conclusion, the metabolic demands of the retina differ in our Pde6b-deficient RP mouse model and are not rescued by gene therapy treatment. These findings provide novel insights into features of both RP disease progression and long-term rescue with gene therapy.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Disease Models, Animal , Genetic Therapy , Retinitis Pigmentosa , Animals , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Mice , Genetic Therapy/methods , Retina/metabolism , Mice, Inbred C57BL , Retinal Rod Photoreceptor Cells/metabolism , Proteomics
6.
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
7.
J Med Chem ; 67(11): 8569-8584, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38758695

ABSTRACT

The trafficking chaperone PDE6D (or PDEδ) was proposed as a surrogate target for K-Ras, leading to the development of a series of inhibitors that block its prenyl binding pocket. These inhibitors suffered from low solubility and suspected off-target effects, preventing their clinical development. Here, we developed a highly soluble, low nanomolar PDE6D inhibitor (PDE6Di), Deltaflexin3, which has the lowest off-target activity as compared to three prominent reference compounds. Deltaflexin3 reduces Ras signaling and selectively decreases the growth of KRAS mutant and PDE6D-dependent cancer cells. We further show that PKG2-mediated phosphorylation of Ser181 lowers K-Ras binding to PDE6D. Thus, Deltaflexin3 combines with the approved PKG2 activator Sildenafil to more potently inhibit PDE6D/K-Ras binding, cancer cell proliferation, and microtumor growth. As observed previously, inhibition of Ras trafficking, signaling, and cancer cell proliferation remained overall modest. Our results suggest reevaluating PDE6D as a K-Ras surrogate target in cancer.


Subject(s)
Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 6 , Proto-Oncogene Proteins p21(ras) , Sildenafil Citrate , Humans , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Sildenafil Citrate/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mutation , Animals , Structure-Activity Relationship , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/chemical synthesis
8.
Gene ; 922: 148562, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38754567

ABSTRACT

BACKGROUND: Previously, we discovered a strain of Kunming mice, referred to as the KMush/ush strain, that exhibited notably abnormal electroretinogram (ERG) readings and elevated thresholds for auditory brainstem responses (ABRs), which resembled the characteristics of Usher Syndrome (USH). We successfully identified the pathogenic genes, Pde6b and Adgrv1, after KMush/ush crossbred with CBA/CaJ mice, referred to as CBA-1ush/ush, CBA-2ush/ush or CBA-2ush/ush. In this investigation, we crossbred KMush/ush and CBA/J mice to establish novel recombinant inbred lines and analysed their phenotypic and genotypic characteristics. METHODS: ERG readings, ABR testing, fundus morphology, histological examination of the retina and inner ear, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis, western blotting, DNA sequence analysis and behavioural experiments were performed to assess the phenotypes and genotypes of the progeny lines. RESULTS: No obvious waveforms in the ERG were detected in F1 hybrid mice while normal ABR results were recorded. The F2 hybrids, which were called J1ush/ush or J2ush/ush, exhibited segregated hearing-loss phenotypes. J1ush/ush mice had a retinitis pigmentosa (RP) phenotype with elevated ABR thresholds, whereas J2ush/ush mice exhibited only the RP phenotype. Interestingly, J1ush/ush mice showed significantly higher ABR thresholds than wild-type mice at 28 days post born (P28), and RT-qPCR and DNA-sequencing analysis showed that Adgrv1 gene expression was significantly altered in J1ush/ush mice, but histological analysis showed no significant structural changes in the organ of Corti or spiral ganglia. Further elevation of ABR-related hearing thresholds by P56 manifested only as a reduced density of spiral ganglion cells, which differed significantly from the previous pattern of cochlear alterations in CBA-2ush/ush mice. CONCLUSIONS: We successfully introduced the hearing-loss phenotype of inbred mice with USH into CBA/J mice, which provides a good animal model for future studies on the important physiological roles of the Adgrv1 gene in inner-ear structure and for therapeutic studies targeting Adgrv1-mutated USH.


Subject(s)
Disease Models, Animal , Electroretinography , Evoked Potentials, Auditory, Brain Stem , Mice, Inbred CBA , Usher Syndromes , Animals , Usher Syndromes/genetics , Usher Syndromes/pathology , Mice , Male , Female , Phenotype , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Retina/pathology , Retina/metabolism , Crosses, Genetic
9.
BMC Med Genomics ; 17(1): 88, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627714

ABSTRACT

BACKGROUND: Liver cancer ranks sixth in incidence and third in mortality globally and hepatocellular carcinoma (HCC) accounts for 90% of it. Hypoxia, glycolysis, and lactate metabolism have been found to regulate the progression of HCC separately. However, there is a lack of studies linking the above three to predict the prognosis of HCC. The present study aimed to identify a hypoxia-glycolysis-lactate-related gene signature for assessing the prognosis of HCC. METHODS: This study collected 510 hypoxia-glycolysis-lactate genes from Molecular Signatures Database (MSigDB) and then classified HCC patients from TCGA-LIHC by analyzing their hypoxia-glycolysis-lactate genes expression. Differentially expressed genes (DEGs) were screened out to construct a gene signature by LASSO-Cox analysis. Univariate and multivariate regression analyses were used to evaluate the independent prognostic value of the gene signature. Analyses of immune infiltration, somatic cell mutations, and correlation heatmap were conducted by "GSVA" R package. Single-cell analysis conducted by "SingleR", "celldex", "Seurat", and "CellCha" R packages revealed how signature genes participated in hypoxia/glycolysis/lactate metabolism and PPI network identified hub genes. RESULTS: We classified HCC patients from TCGA-LIHC into two clusters and screened out DEGs. An 18-genes prognostic signature including CDCA8, CBX2, PDE6A, MED8, DYNC1LI1, PSMD1, EIF5B, GNL2, SEPHS1, CCNJL, SOCS2, LDHA, G6PD, YBX1, RTN3, ADAMTS5, CLEC3B, and UCK2 was built to stratify the risk of HCC. The risk score of the hypoxia-glycolysis-lactate gene signature was further identified as a valuable independent factor for estimating the prognosis of HCC. Then we found that the features of clinical characteristics, immune infiltration, somatic cell mutations, and correlation analysis differed between the high-risk and low-risk groups. Furthermore, single-cell analysis indicated that the signature genes could interact with the ligand-receptors of hepatocytes/fibroblasts/plasma cells to participate in hypoxia/glycolysis/lactate metabolism and PPI network identified potential hub genes in this process: CDCA8, LDHA, YBX1. CONCLUSION: The hypoxia-glycolysis-lactate-related gene signature we built could provide prognostic value for HCC and suggest several hub genes for future HCC studies.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Lactic Acid , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Prognosis , Hypoxia , Eye Proteins , Cyclic Nucleotide Phosphodiesterases, Type 6 , Cytoplasmic Dyneins
10.
J Med Chem ; 67(10): 8396-8405, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38688030

ABSTRACT

Retinitis pigmentosa (RP) is a form of retinal degeneration affecting a young population with an unmet medical need. Photoreceptor degeneration has been associated with increased guanosine 3',5'-cyclic monophosphate (cGMP), which reaches toxic levels for photoreceptors. Therefore, inhibitory cGMP analogues attract interest for RP treatments. Here we present the synthesis of dithio-CN03, a phosphorodithioate analogue of cGMP, prepared using the H-phosphonothioate route. Two crystal modifications were identified as a trihydrate and a tetrahydrofuran monosolvates. Dithio-CN03 featured a lower aqueous solubility than its RP-phosphorothioate counterpart CN03, a drug candidate, and this characteristic might be favorable for sustained-release formulations aimed at retinal delivery. Dithio-CN03 was tested in vitro for its neuroprotective effects in photoreceptor models of RP. The comparison of dithio-CN03 to CN03 and its diastereomer SP-CN03, and to their phosphate derivative oxo-CN03 identifies dithio-CN03 as the compound with the highest efficacy in neuroprotection and thus as a promising new candidate for the treatment of RP.


Subject(s)
Cyclic GMP , Neuroprotective Agents , Retinal Rod Photoreceptor Cells , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 6/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Retinal Degeneration/drug therapy , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/metabolism , Structure-Activity Relationship
11.
BMC Ophthalmol ; 24(1): 55, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38317096

ABSTRACT

BACKGROUND: Inherited retinal degenerations (IRDs) are a group of rare genetic conditions affecting retina of the eye that range in prevalence from 1 in 2000 to 1 in 4000 people globally. This review is based on a retrospective analysis of research articles reporting IRDs associated genetic findings in Pakistani families between 1999 and April 2023. METHODS: Articles were retrieved through survey of online sources, notably, PubMed, Google Scholar, and Web of Science. Following a stringent selection criterion, a total of 126 research articles and conference abstracts were considered. All reported variants were cross-checked and validated for their correct genomic nomenclature using different online resources/databases, and their pathogenicity scores were explained as per ACMG guidelines. RESULTS: A total of 277 unique sequence variants in 87 distinct genes, previously known to cause IRDs, were uncovered. In around 70% cases, parents of the index patient were consanguineously married, and approximately 88.81% of the detected variants were found in a homozygous state. Overall, more than 95% of the IRDs cases were recessively inherited. Missense variants were predominant (41.88%), followed by Indels/frameshift (26.35%), nonsense (19.13%), splice site (12.27%) and synonymous change (0.36%). Non-syndromic IRDs were significantly higher than syndromic IRDs (77.32% vs. 22.68%). Retinitis pigmentosa (RP) was the most frequently observed IRD followed by Leber's congenital amaurosis (LCA). Altogether, mutations in PDE6A gene was the leading cause of IRDs in Pakistani families followed by mutations in TULP1 gene. CONCLUSION: In summary, Pakistani families are notable in expressing recessively inherited monogenic disorders including IRDs likely due to the highest prevalence of consanguinity in the country that leads to expression of rare pathogenic variants in homozygous state.


Subject(s)
Retinal Dystrophies , Retinitis Pigmentosa , Humans , Pakistan/epidemiology , Retrospective Studies , Retinal Dystrophies/epidemiology , Retinal Dystrophies/genetics , Retina/pathology , Retinitis Pigmentosa/genetics , Mutation , Pedigree , Eye Proteins/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics
12.
Am J Ophthalmol ; 263: 1-10, 2024 07.
Article in English | MEDLINE | ID: mdl-38364953

ABSTRACT

PURPOSE: To analyze the clinical characteristics, natural history, and genetics of PDE6B-associated retinal dystrophy. DESIGN: Retrospective, observational cohort study. METHODS: Review of medical records and retinal imaging, including fundus autofluorescence (FAF) imaging and spectral-domain optical coherence tomography (SD-OCT) of patients with molecularly confirmed PDE6B-associated retinal dystrophy in a single tertiary referral center. Genetic results were reviewed, and the detected variants were assessed. RESULTS: Forty patients (80 eyes) were identified and evaluated longitudinally. The mean age (±SD, range) was 42.1 years (± 19.0, 10-86) at baseline, with a mean follow-up time of 5.2 years. Twenty-nine (72.5%) and 27 (67.5%) patients had no or mild visual acuity impairment at baseline and last visit, respectively. Best-corrected visual acuity (BCVA) was 0.56 ± 0.72 LogMAR (range -0.12 to 2.80) at baseline and 0.63 ± 0.73 LogMAR (range 0.0-2.80) at the last visit. BCVA was symmetrical in 87.5% of patients. A hyperautofluorescent ring was observed on FAF in 48 and 46 eyes at baseline and follow-up visit, respectively, with a mean area of 7.11 ± 4.13 mm2 at baseline and mean of 6.13 ± 3.62 mm2 at the follow-up visit. Mean horizontal ellipsoid zone width at baseline was 1946.1 ± 917.2 µm, which decreased to 1763.9 ± 827.9 µm at follow-up. Forty-four eyes had cystoid macular edema at baseline (55%), and 41 eyes (51.3%) at follow-up. There were statistically significant changes during the follow-up period in terms of BCVA and the ellipsoid zone width. Genetic analysis identified 43 variants in the PDE6B gene, including 16 novel variants. CONCLUSIONS: This study details the natural history of PDE6B-retinopathy in the largest cohort to date. Most patients had mild to no BCVA loss, with slowly progressive disease, based on FAF and OCT metrics. There is a high degree of disease symmetry and a wide window for intervention.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Fluorescein Angiography , Retinal Dystrophies , Tomography, Optical Coherence , Visual Acuity , Humans , Retrospective Studies , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Male , Female , Visual Acuity/physiology , Adult , Middle Aged , Adolescent , Aged , Young Adult , Child , Retinal Dystrophies/genetics , Retinal Dystrophies/physiopathology , Retinal Dystrophies/diagnosis , Aged, 80 and over , Follow-Up Studies , Mutation , Electroretinography , DNA Mutational Analysis
13.
Acta Ophthalmol ; 102(6): e893-e905, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38348755

ABSTRACT

PURPOSE: To describe the phenotype of Danish patients with genetically verified achromatopsia (ACHM) with special focus on signs of progression on structural or functional parameters, and possible genotype-phenotype correlations. METHODS: Forty-eight patients were identified, with disease-causing variants in five different genes: CNGA3, CNGB3, GNAT2, PDE6C and PDE6H. Longitudinal evaluation was possible for 11 patients and 27 patients participated in a renewed in-depth phenotyping consisting of visual acuity assessment, optical coherence tomography (OCT), fundus autofluorescence, colour vision evaluation, contrast sensitivity, mesopic microperimetry and full-field electroretinography. Foveal morphology was evaluated based on OCT images for all 48 patients using a grading system based on the integrity of the hyperreflective photoreceptor band, the inner segment ellipsoid zone (ISe). Signs of progression were evaluated based on longitudinal data and correlation with age. RESULTS: We found a statistically significant positive correlation between OCT grade and age (Spearman ρ = 0.62, p < 0.0001) and we observed changes in the foveal morphology in 2 of 11 patients with ≥5 years of follow-up. We did not find any convincing correlation between age and functional parameters (visual acuity, retinal sensitivity and contrast sensitivity) nor did we find correlation between structural and functional parameters, or any clear genotype-phenotype correlation. CONCLUSIONS: Some patients with ACHM demonstrate signs of progressive foveal changes in OCT characteristics with increasing age. This is relevant in terms of possible new treatments. However, functional characteristics, such as visual acuity, remained stable despite changing foveal structure. Thus, seen from a patient perspective, ACHM can still be considered a non-progressive condition.


Subject(s)
Color Vision Defects , Cyclic Nucleotide-Gated Cation Channels , Electroretinography , Phenotype , Tomography, Optical Coherence , Visual Acuity , Humans , Color Vision Defects/genetics , Color Vision Defects/physiopathology , Color Vision Defects/diagnosis , Male , Tomography, Optical Coherence/methods , Female , Visual Acuity/physiology , Adult , Denmark , Adolescent , Young Adult , Child , Middle Aged , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Mutation , Visual Field Tests , Contrast Sensitivity/physiology , Follow-Up Studies , Fluorescein Angiography/methods , Eye Proteins/genetics , Guanylate Cyclase/genetics , Visual Fields/physiology , Genetic Association Studies , DNA Mutational Analysis , DNA/genetics , Color Vision/physiology
14.
Exp Anim ; 73(2): 203-210, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38171880

ABSTRACT

In CBA/J and C3H/HeJ mice, retinitis pigmentosa is inherited as an autosomal-recessive trait due to a mutation in Pde6b, which encodes cGMP phosphodiesterase subunit b. In these strains, the Y347X mutation in Pde6b leads to the upregulation of cGMP levels, increased Ca2+ influx induces rod death, and the outer segment and rod cells entirely disappeared by 35 days after birth. In the present study, we utilized the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9-mediated gene editing to repair the Y347X mutation in CBA/J and C3H/HeJ mice. Evaluation of the established CBA/J-Pde6bY347Y/Y347X and C3H/HeJ-Pde6bY347Y/Y347X mice, which were confirmed to have normal retinal layers by live fundoscopic imaging and histopathological analysis, revealed improved visual acuity based on the visual cliff and light/dark latency tests. Furthermore, our analyses revealed that the visible platform test was a more effective tool for testing visual behavior in these mice. The results suggest that the established strains can serve as control groups for CBA/J and C3H/HeJ in ophthalmology studies involving retinitis pigmentosa.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Mice, Inbred C3H , Mice, Inbred CBA , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Retinitis Pigmentosa/genetics , Mice , Gene Editing , Mutation , Disease Models, Animal , Visual Acuity/physiology , CRISPR-Cas Systems , Retina/metabolism
15.
J Genet Genomics ; 51(2): 208-221, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38157933

ABSTRACT

Inherited retinal dystrophies (IRDs) are major causes of visual impairment and irreversible blindness worldwide, while the precise molecular and genetic mechanisms are still elusive. N6-methyladenosine (m6A) modification is the most prevalent internal modification in eukaryotic mRNA. YTH domain containing 2 (YTHDC2), an m6A reader protein, has recently been identified as a key player in germline development and human cancer. However, its contribution to retinal function remains unknown. Here, we explore the role of YTHDC2 in the visual function of retinal rod photoreceptors by generating rod-specific Ythdc2 knockout mice. Results show that Ythdc2 deficiency in rods causes diminished scotopic ERG responses and progressive retinal degeneration. Multi-omics analysis further identifies Ppef2 and Pde6b as the potential targets of YTHDC2 in the retina. Specifically, via its YTH domain, YTHDC2 recognizes and binds m6A-modified Ppef2 mRNA at the coding sequence and Pde6b mRNA at the 5'-UTR, resulting in enhanced translation efficiency without affecting mRNA levels. Compromised translation efficiency of Ppef2 and Pde6b after YTHDC2 depletion ultimately leads to decreased protein levels in the retina, impaired retinal function, and progressive rod death. Collectively, our finding highlights the importance of YTHDC2 in visual function and photoreceptor survival, which provides an unreported elucidation of IRD pathogenesis via epitranscriptomics.


Subject(s)
Photoreceptor Cells, Vertebrate , Retinal Degeneration , Animals , Humans , Mice , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , RNA Helicases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
J Biol Chem ; 300(2): 105608, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159849

ABSTRACT

Phototransduction in retinal rods occurs when the G protein-coupled photoreceptor rhodopsin triggers the activation of phosphodiesterase 6 (PDE6) by GTP-bound alpha subunits of the G protein transducin (GαT). Recently, we presented a cryo-EM structure for a complex between two GTP-bound recombinant GαT subunits and native PDE6, that included a bivalent antibody bound to the C-terminal ends of GαT and the inhibitor vardenafil occupying the active sites on the PDEα and PDEß subunits. We proposed GαT-activated PDE6 by inducing a striking reorientation of the PDEγ subunits away from the catalytic sites. However, questions remained including whether in the absence of the antibody GαT binds to PDE6 in a similar manner as observed when the antibody is present, does GαT activate PDE6 by enabling the substrate cGMP to access the catalytic sites, and how does the lipid membrane enhance PDE6 activation? Here, we demonstrate that 2:1 GαT-PDE6 complexes form with either recombinant or retinal GαT in the absence of the GαT antibody. We show that GαT binding is not necessary for cGMP nor competitive inhibitors to access the active sites; instead, occupancy of the substrate binding sites enables GαT to bind and reposition the PDE6γ subunits to promote catalytic activity. Moreover, we demonstrate by reconstituting GαT-stimulated PDE6 activity in lipid bilayer nanodiscs that the membrane-induced enhancement results from an increase in the apparent binding affinity of GαT for PDE6. These findings provide new insights into how the retinal G protein stimulates rapid catalytic turnover by PDE6 required for dim light vision.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Models, Molecular , Transducin , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Guanosine Triphosphate/metabolism , Retinal Rod Photoreceptor Cells/enzymology , Retinal Rod Photoreceptor Cells/metabolism , Transducin/chemistry , Transducin/genetics , Transducin/metabolism , Animals , Cattle , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Protein Structure, Quaternary , Protein Binding/drug effects , Catalytic Domain , 1-Methyl-3-isobutylxanthine/pharmacology , Lipid Bilayers/metabolism , Enzyme Activation
17.
J Biol Chem ; 300(1): 105576, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38110033

ABSTRACT

The sixth family phosphodiesterases (PDE6) are principal effector enzymes of the phototransduction cascade in rods and cones. Maturation of nascent PDE6 protein into a functional enzyme relies on a coordinated action of ubiquitous chaperone HSP90, its specialized cochaperone aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1), and the regulatory Pγ-subunit of PDE6. Deficits in PDE6 maturation and function underlie severe visual disorders and blindness. Here, to elucidate the roles of HSP90, AIPL1, and Pγ in the maturation process, we developed the heterologous expression system of human cone PDE6C in insect cells allowing characterization of the purified enzyme. We demonstrate that in the absence of Pγ, HSP90, and AIPL1 convert the inactive and aggregating PDE6C species into dimeric PDE6C that is predominantly misassembled. Nonetheless, a small fraction of PDE6C is properly assembled and fully functional. From the analysis of mutant mice that lack both rod Pγ and PDE6C, we conclude that, in contrast to the cone enzyme, no maturation of rod PDE6AB occurs in the absence of Pγ. Co-expression of PDE6C with AIPL1 and Pγ in insect cells leads to a fully mature enzyme that is equivalent to retinal PDE6. Lastly, using immature PDE6C and purified chaperone components, we reconstituted the process of the client maturation in vitro. Based on this analysis we propose a scheme for the PDE6 maturation process.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Retinal Cone Photoreceptor Cells , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Blindness/genetics , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 6/deficiency , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mutation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism , Retinal Cone Photoreceptor Cells/chemistry , Retinal Cone Photoreceptor Cells/metabolism
18.
Invest Ophthalmol Vis Sci ; 64(11): 18, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37578425

ABSTRACT

Purpose: Verifying whether specific genotypes causing retinitis pigmentosa (RP) show differences in the preservation of rod and cone function measured by chromatic pupil campimetry (CPC). Methods: Sixty-three RP eyes (37 male, 14-58 years) were measured using CPC with specific photopic and scotopic protocols, and the relative maximal constriction amplitudes and latencies to constriction onset were analyzed per genotype (RP due to variants in EYS, n = 14; PDE6A, n = 10; RPE65, n = 15; USH2A, n = 10; and RPGR, n = 14). Correlation analyses between the pupillary responses were performed with age, full-field stimulus threshold (FST), and optical coherence tomography (OCT) for cones and rods, respectively, to the genotype. Results: Pupillary responses were most severely reduced in RPE65-RP. Patients with disease-associated variants in EYS and USH2A were accompanied with better-preserved rod function compared with the other subgroups, reaching statistical significance between EYS and RPE65. Cone function was statistically significantly correlated with age in USH2A-RP with an annual decline of 2.4%. Correlations of pupillary responses were found with FST but barely with the ellipsoid zone area in OCT. Latency was significantly more prolonged in RPE65-RP compared with the other genotypes for cones. Conclusions: Rod and cone function measured objectively by CPC showed a different preservation between genotypes in RP. However, heterogeneity inside the same genotype was present. CPC data correlated with FST, but structural OCT parameters seem to be limited indicators for photoreceptor function in RP. Prolonged time dynamics for cones in RPE65 mutations suggest an impact on cone processing and might provide additional information in the evaluation of therapy effects.


Subject(s)
Retinitis Pigmentosa , Visual Field Tests , Humans , Male , Pupil , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinal Cone Photoreceptor Cells/physiology , Genotype , Electroretinography/methods , Eye Proteins/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics
19.
Eur Rev Med Pharmacol Sci ; 27(11): 4876-4882, 2023 06.
Article in English | MEDLINE | ID: mdl-37318461

ABSTRACT

OBJECTIVE: The aim of this study was to identify the hub genes and uncover the molecular mechanisms of diabetic retinopathy (DR). MATERIALS AND METHODS: We used the Gene Expression Omnibus (GEO) dataset GSE60436 in our study. After screening for differentially expressed genes (DEGs), we performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis. Subsequently, a protein-protein interaction (PPI) network was constructed using the Search Tool for Retrieval of Interacting Genes (STRING) database and visualized using the Cytoscape software. Finally, we identified 10 hub genes by cytoHubba plugin. RESULTS: A total of 592 DEGs were identified, including 203 up-regulated genes and 389 downregulated genes. The DEGs were mainly enriched in visual perception, photoreceptor outer segment membrane, retinal binding, and PI3K-Akt signaling pathway. By constructing a protein-protein interaction (PPI) network, 10 central genes were finally identified, including CNGA1, PDE6G, RHO, ABCA4, PDE6A, PDE6B, NRL, RPE65, GUCA1B and AIPL1. CONCLUSIONS: CNGA1, PDE6G, RHO, ABCA4, PDE6A, PDE6B, NRL, RPE65, GUCA1B, and AIPL1 may be potential biomarkers and therapeutic targets for DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Gene Expression Profiling/methods , Gene Regulatory Networks , Diabetic Retinopathy/genetics , Phosphatidylinositol 3-Kinases , Computational Biology/methods , ATP-Binding Cassette Transporters , Eye Proteins/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6 , Adaptor Proteins, Signal Transducing
20.
Indian J Ophthalmol ; 71(6): 2512-2520, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37322672

ABSTRACT

Purpose: Inherited retinal dystrophies (IRD) are a heterogeneous group of retinal diseases leading to progressive loss of photoreceptors through apoptosis. Retinitis pigmentosa (RP) is considered the most common form of IRD. Panel-based testing in RP has proven effective in identifying the causative genetic mutations in 70% and 80% of the patients. This is a retrospective, observational, single-center study of 107 RP patients who had undergone next-generation sequencing-based targeted gene panel testing for IRD genes. These patients were inspected for common phenotypic features to arrive at meaningful genotype-phenotype correlation. Methods: Patients underwent complete ophthalmic examination, and blood was collected from the proband for DNA extraction after documenting the pedigree. Targeted Next Generation Sequencing (NGS) was done by panel-based testing for IRD genes followed by co-segregation analysis wherever applicable. Results: Of the 107 patients, 72 patients had pathogenic mutations. The mean age of onset of symptoms was 14 ± 12 years (range: 5-55). Mean (Best Corrected Visual Acuity) BCVA was 6/48 (0.9 logMAR) (range 0.0-3.0). At presentation, over one-third of eyes had BCVA worse than 6/60 (<1 logMAR). Phenotype analysis with the gene defects showed overlapping features, such as peripheral well-defined chorioretinal atrophic patches in patients with CERKL, PROM1, and RPE65 gene mutations and large macular lesions in patients with RDH12 and CRX gene mutations, respectively. Nummular or clump-like pigmentation was noted in CRB1, TTC8, PDE6A, and PDE6B. Conclusion: NGS-based genetic testing can help clinicians to diagnose RP more accurately, and phenotypic correlations can also help in better patient counselling with respect to prognosis and guidance regarding ongoing newer gene-based therapies.


Subject(s)
Retinal Dystrophies , Retinitis Pigmentosa , Humans , Genetic Testing , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Phenotype , Mutation , High-Throughput Nucleotide Sequencing/methods , Eye Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Alcohol Oxidoreductases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL