Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.343
Filtrer
1.
Zhonghua Yan Ke Za Zhi ; 60(7): 601-610, 2024 Jul 11.
Article de Chinois | MEDLINE | ID: mdl-38955762

RÉSUMÉ

Objective: To investigate the characteristics of posterior segment lesions in Marfan syndrome (MFS) patients and their relationship with anterior segment biometric parameters and FBN1 genotype. Methods: A cross-sectional study was conducted. A total of 121 MFS patients, 76 males and 45 females, with an average age of (11.72±11.66) years, who visited the Department of Ophthalmology, Eye & ENT Hospital of Fudan University from January 2013 to March 2023 were included. The presence of posterior scleral staphyloma was observed using B-mode ultrasound, and macular lesions were identified and classified using the atrophy-traction-neovascularization system based on ultra-widefield fundus images, color fundus images, and optical coherence tomography scans. Anterior segment biometric parameters, including axial length of the eye, average corneal curvature, corneal astigmatism, horizontal corneal diameter, anterior chamber depth, and lens thickness, were collected, and the direction and extent of lens dislocation were observed. Molecular genetic analysis of FBN1 gene mutations in patients was performed using next-generation sequencing based on a panel of ocular genetic diseases, and the impact of the genotype and anterior segment biometric parameters on the posterior segment manifestations was analyzed. Results: Sixty patients exhibited posterior segment lesions, including retinal detachment (4 cases, 3.31%), macular lesions (47 cases, 38.84%), and posterior scleral staphyloma (54 cases, 44.63%). There was statistically significant difference in axial length of the eye between patients with and without posterior scleral staphyloma [23.09 (22.24, 24.43) and 27.04 (25.44, 28.88) mm], between patients with and without macular lesions [23.16 (22.24, 24.61) and 27.04 (25.74, 28.78) mm], and between patients with and without atrophic macular lesions [23.16 (22.24, 24.61) and 27.04 (25.74, 28.79) mm] (all P<0.001). There was statistically significant difference in anterior chamber depth between patients with and without macular lesions [3.11 (2.75, 3.30) and 3.34 (3.09, 3.60) mm] (P<0.05). There was also statistically significant difference in corneal astigmatism between patients with and without posterior scleral staphyloma [2.15 (1.20, 2.93) and 1.40 (1.00, 2.20) diopters] (P<0.05). The location and region of the FBN1 gene mutation not only showed statistically significant difference from the positive rates of posterior scleral staphyloma and macular lesions (all P<0.05), but also influenced the occurrence of atrophic macular lesions (both P<0.05). Patients with FBN1 mutations located in the transforming growth factor ß regulatory sequence had the highest proportion of posterior scleral staphyloma and macular lesions (both 10/11). Conclusions: Posterior scleral staphyloma and macular lesions have a relatively high incidence in MFS patients and tend to progress to more severe grades. The age, axial length of the eye, anterior chamber depth, corneal astigmatism, and location and region of the FBN1 gene mutation are factors affecting the posterior segment lesions in MFS patients.


Sujet(s)
Fibrilline-1 , Génotype , Syndrome de Marfan , Humains , Mâle , Femelle , Fibrilline-1/génétique , Études transversales , Syndrome de Marfan/génétique , Enfant , Adolescent , Pôle antérieur du bulbe oculaire , Segment postérieur de l'oeil/anatomopathologie , Mutation , Biométrie , Jeune adulte , Dégénérescence maculaire/génétique , Adipokines
2.
Lipids Health Dis ; 23(1): 206, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951820

RÉSUMÉ

BACKGROUND: An imbalance in lipid metabolism has been linked to the development of AMD, but the causal relationship between AMD and plasma fatty acids (FAs) remains controversial. Using a two-sample Mendelian randomization (MR) approach, we sought to evaluate the impact of specific FA plasma levels on the risk of different AMD subtypes. METHODS: We analysed genome-wide association data of circulating FAs from 115,006 European-descended individuals in the UK Biobank. These data were used in a two-sample MR framework to assess the potential role of circulating FAs in developing wet and dry AMD. Sensitivity analyses were conducted to ensure the robustness of our findings. Additional multivariable and locus-specific MR analyses were conducted to evaluate direct effects of FA on AMD subtypes, minimizing biases from lipoprotein-related traits and triglycerides. RESULTS: Mendelian randomization revealed associations of omega-3 was associated with decreased wet (OR 0.78, 95%CI 0.66-0.92) and dry AMD (0.85, 0.74-0.97) risk, showed a protective effect on AMD. Notably, the omega-6 to omega-3 ratio showed potential causal effects on both wet (1.27, 1.03-1.56) and dry AMD (1.18, 1.02-1.37). Multivariable MR suggested that the causal relationship of omega-3, omega-6 to omega-3 ratio on wet AMD persists after conditioning on HDL, LDL and triglycerides, albeit with slightly diminished evidence strength. Locus-specific MR linked to omega-3(FADS1, 0.89, 0.82-0.98; FADS2, 0.88, 0.81-0.96) and omega-6 to omega-3 ratio (FADS1, 1.10, 1.02-1.20; FADS2, 1.11, 1.03-1.20) suggests causal effects of these factors on wet AMD. CONCLUSIONS: The associations between plasma FA concentrations and AMD, suggest potential causal role of omega-3, and the omega-6 to omega-3 ratio in wet AMD. These results underscore the impact of an imbalanced circulating omega-3 and omega-6 FA ratio on AMD pathophysiology from MR perspective.


Sujet(s)
Delta-5 fatty acid desaturase , Acides gras omega-3 , Acides gras omega-6 , Étude d'association pangénomique , Dégénérescence maculaire , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Humains , Dégénérescence maculaire/sang , Dégénérescence maculaire/génétique , Acides gras omega-3/sang , Mâle , Femelle , Acides gras omega-6/sang , Sujet âgé , Fatty acid desaturases/génétique , Adulte d'âge moyen , Triglycéride/sang , Acides gras/sang , Facteurs de risque
3.
Genes (Basel) ; 15(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38927656

RÉSUMÉ

Gene therapy holds promise as a transformative approach in the treatment landscape of age-related macular degeneration (AMD), diabetic retinopathy (DR), and diabetic macular edema (DME), aiming to address the challenges of frequent intravitreal anti-vascular endothelial growth factor (VEGF) injections. This manuscript reviews ongoing gene therapy clinical trials for these disorders, including ABBV-RGX-314, ixoberogene soroparvovec (ixo-vec), and 4D-150. ABBV-RGX-314 utilizes an adeno-associated virus (AAV) vector to deliver a transgene encoding a ranibizumab-like anti-VEGF antibody fragment, demonstrating promising results in Phase 1/2a and ongoing Phase 2b/3 trials. Ixo-vec employs an AAV2.7m8 capsid for intravitreal delivery of a transgene expressing aflibercept, showing encouraging outcomes in Phase 1 and ongoing Phase 2 trials. 4D-150 utilizes an evolved vector to express both aflibercept and a VEGF-C inhibitory RNAi, exhibiting positive interim results in Phase 1/2 studies. Other therapies reviewed include EXG102-031, FT-003, KH631, OLX10212, JNJ-1887, 4D-175, and OCU410. These therapies offer potential advantages of reduced treatment frequency and enhanced safety profiles, representing a paradigm shift in management towards durable and efficacious cellular-based biofactories. These advancements in gene therapy hold promise for improving outcomes in AMD and addressing the complex challenges of DME and DR, providing new avenues for the treatment of diabetic eye diseases.


Sujet(s)
Rétinopathie diabétique , Thérapie génétique , Dégénérescence maculaire , Humains , Rétinopathie diabétique/thérapie , Rétinopathie diabétique/génétique , Thérapie génétique/méthodes , Dégénérescence maculaire/thérapie , Dégénérescence maculaire/génétique , Vecteurs génétiques/génétique , Dependovirus/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Animaux
4.
Genes (Basel) ; 15(6)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38927702

RÉSUMÉ

Inherited retinal diseases (IRDs) represent a frequent cause of blindness in children and adults. As a consequence of the phenotype and genotype heterogeneity of the disease, it is difficult to have a specific diagnosis without molecular testing. To date, over 340 genes and loci have been associated with IRDs. We present the molecular finding of 191 individuals with IRD, analyzed by targeted next-generation sequencing (NGS). For 67 of them, we performed a family segregation study, considering a total of 126 relatives. A total of 359 variants were identified, 44 of which were novel. Genetic diagnostic yield was 41%. However, after stratifying the patients according to their clinical suspicion, diagnostic yield was higher for well-characterized diseases such as Stargardt disease (STGD), at 65%, and for congenital stationary night blindness 2 (CSNB2), at 64%. Diagnostic yield was higher in the patient group where family segregation analysis was possible (68%) and it was higher in younger (55%) than in older patients (33%). The results of this analysis demonstrated that targeted NGS is an effective method for establishing a molecular genetic diagnosis of IRDs. Furthermore, this study underlines the importance of segregation studies to understand the role of genetic variants with unknow pathogenic role.


Sujet(s)
Séquençage nucléotidique à haut débit , Dystrophies rétiniennes , Maladie de Stargardt , Humains , Séquençage nucléotidique à haut débit/méthodes , Mâle , Femelle , Dystrophies rétiniennes/génétique , Dystrophies rétiniennes/diagnostic , Adulte , Maladie de Stargardt/génétique , Pedigree , Enfant , Adulte d'âge moyen , Héméralopie/génétique , Maladies héréditaires de l'oeil/génétique , Adolescent , Mutation , Dégénérescence maculaire/génétique , Myopie/génétique , Enfant d'âge préscolaire , Phénotype , Jeune adulte , Sujet âgé , Maladies génétiques liées au chromosome X
5.
Invest Ophthalmol Vis Sci ; 65(6): 38, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38935028

RÉSUMÉ

Purpose: To assess the association of age-related macular degeneration (AMD) progression and statins, connected with AMD genetic risk, and if there is an interplay between statins and genetics. Methods: In this analysis, 682 subjects made two visits (6.5-year follow-up) of the Coimbra Eye Study. Subjects who started taking statins at any time point between the two visits were considered. Progressors were defined as not having AMD at baseline and having any AMD at follow-up. Genetic risk scores (GRSs) were calculated individually with 52 independent variants associated with AMD. Time to progression was estimated using unadjusted Kaplan-Meier curves. An extended Cox model was used for the association between statins and GRS with the risk for AMD progression. Multiplicative and additive interactions were assessed. Results: Median survival time was 7.50 years for subjects not taking statins and 7.62 for subjects taking statins (P < 0.001). Statin intake reduced the risk for progression to AMD in 48%, adjusting for age, sex, body mass index, smoking, and diabetes (model 1) and GRS (model 2). The combined effects of not taking statins and having high GRS increased the progression risk fourfold compared to taking statins and having low GRS (hazard ratio [HR] = 4.25; 95% confidence interval [CI], 1.62-11.16; P = 0.003). For subjects not taking statins, an increased risk of progression was found for those subjects with high GRS compared to subjects with low GRS (HR = 1.80; 95% CI, 1.13-2.85; P = 0.013). No statistically significant multiplicative or additive interactions were found. Conclusions: Statins seem to be protective against AMD progression, and genetics may play a role in treatment response.


Sujet(s)
Évolution de la maladie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Dégénérescence maculaire , Humains , Mâle , Femelle , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Sujet âgé , Dégénérescence maculaire/génétique , Études de suivi , Facteurs de risque , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Polymorphisme de nucléotide simple , Prédisposition génétique à une maladie
6.
Cell Death Dis ; 15(6): 385, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824126

RÉSUMÉ

Drusen, the yellow deposits under the retina, are composed of lipids and proteins, and represent a hallmark of age-related macular degeneration (AMD). Lipid droplets are also reported in the retinal pigment epithelium (RPE) from AMD donor eyes. However, the mechanisms underlying these disease phenotypes remain elusive. Previously, we showed that Pgc-1α repression, combined with a high-fat diet (HFD), induce drastic AMD-like phenotypes in mice. We also reported increased PGC-1α acetylation and subsequent deactivation in the RPE derived from AMD donor eyes. Here, through a series of in vivo and in vitro experiments, we sought to investigate the molecular mechanisms by which PGC-1α repression could influence RPE and retinal function. We show that PGC-1α plays an important role in RPE and retinal lipid metabolism and function. In mice, repression of Pgc-1α alone induced RPE and retinal degeneration and drusen-like deposits. In vitro inhibition of PGC1A by CRISPR-Cas9 gene editing in human RPE (ARPE19- PGC1A KO) affected the expression of genes responsible for lipid metabolism, fatty acid ß-oxidation (FAO), fatty acid transport, low-density lipoprotein (LDL) uptake, cholesterol esterification, cholesterol biosynthesis, and cholesterol efflux. Moreover, inhibition of PGC1A in RPE cells caused lipid droplet accumulation and lipid peroxidation. ARPE19-PGC1A KO cells also showed reduced mitochondrial biosynthesis, impaired mitochondrial dynamics and activity, reduced antioxidant enzymes, decreased mitochondrial membrane potential, loss of cardiolipin, and increased susceptibility to oxidative stress. Our data demonstrate the crucial role of PGC-1α in regulating lipid metabolism. They provide new insights into the mechanisms involved in lipid and drusen accumulation in the RPE and retina during aging and AMD, which may pave the way for developing novel therapeutic strategies targeting PGC-1α.


Sujet(s)
Gouttelettes lipidiques , Métabolisme lipidique , Dégénérescence maculaire , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Épithélium pigmentaire de la rétine , Épithélium pigmentaire de la rétine/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Animaux , Humains , Souris , Gouttelettes lipidiques/métabolisme , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/génétique , Souris de lignée C57BL , Mitochondries/métabolisme , Mâle , Stress oxydatif
7.
Niger Postgrad Med J ; 31(2): 93-101, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38826012

RÉSUMÉ

Age-related macular degeneration (AMD) is a prevalent and incurable condition affecting the central retina and posing a significant risk to vision, particularly in individuals over the age of 60. As the global population ages, the prevalence of AMD is expected to rise, leading to substantial socioeconomic impacts and increased healthcare costs. The disease manifests primarily in two forms, neovascular and non-neovascular, with genetic, environmental and lifestyle factors playing a pivotal role in disease susceptibility and progression. This review article involved conducting an extensive search across various databases, including Google Scholar, PubMed, Web of Science, ScienceDirect, Scopus and EMBASE, to compile relevant case-control studies and literature reviews from online published articles extracted using search terms related to the work. SIRT1, a key member of the sirtuin family, influences cellular processes such as ageing, metabolism, DNA repair and stress response. Its dysregulation is linked to retinal ageing and ocular conditions like AMD. This review discusses the role of SIRT1 in AMD pathology, its association with genetic variants and its potential as a biomarker, paving the way for targeted interventions and personalised treatment strategies. In addition, it highlights the findings of case-control studies investigating the relationship between SIRT1 gene polymorphisms and AMD risk. These studies collectively revealed a significant association between certain SIRT1 gene variants and AMD risk. Further studies with larger sample sizes are required to validate these findings. As the prevalence of AMD grows, understanding the role of SIRT1 and other biomarkers becomes increasingly vital for improving diagnosis, treatment and, ultimately, patient outcomes.


Sujet(s)
Dégénérescence maculaire , Sirtuine-1 , Humains , Sirtuine-1/génétique , Dégénérescence maculaire/génétique , Dégénérescence maculaire/épidémiologie , Prédisposition génétique à une maladie , Polymorphisme génétique
8.
FASEB J ; 38(11): e23720, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38837708

RÉSUMÉ

Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.


Sujet(s)
Transporteurs ABC , Cathepsine D , Lysosomes , Épithélium pigmentaire de la rétine , Maladie de Stargardt , Cathepsine D/métabolisme , Cathepsine D/génétique , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie , Maladie de Stargardt/métabolisme , Maladie de Stargardt/anatomopathologie , Maladie de Stargardt/génétique , Animaux , Humains , Souris , Lysosomes/métabolisme , Transporteurs ABC/métabolisme , Transporteurs ABC/génétique , Cellules souches pluripotentes induites/métabolisme , Souris knockout , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/génétique
9.
PLoS One ; 19(6): e0303170, 2024.
Article de Anglais | MEDLINE | ID: mdl-38857222

RÉSUMÉ

OBJECTIVE: The aim of this study is to investigate the potential causal relationship between autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, and Type 1 diabetes, and age-related macular degeneration (AMD). By utilizing the two-sample Mendelian Randomization (MR) approach, we endeavor to address this complex medical issue. METHODS: Genome-wide association study (GWAS) data for autoimmune diseases and AMD were obtained from the IEU Open GWAS database and the FinnGen consortium. A series of stringent SNP filtering steps was applied to ensure the reliability of the genetic instruments. MR analyses were conducted using the TwoSampleMR and MR-PRESSO packages in R. The inverse-variance weighted (IVW) method served as the primary analysis, complemented by multiple supplementary analyses and sensitivity tests. RESULTS: Within the discovery sample, only a statistically significant inverse causal relationship between multiple sclerosis (MS) and AMD was observed (OR = 0.92, 95% CI: 0.88-0.97, P = 0.003). This finding was confirmed in the replication sample (OR = 0.85, 95% CI: 0.80-0.89, P = 3.32×10-12). No statistically significant associations were detected between systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, and Type 1 diabetes and AMD. CONCLUSION: Strong evidence is provided by this study to support the existence of an inverse causal relationship between multiple sclerosis and age-related macular degeneration. However, no causal evidence was found linking other autoimmune diseases with AMD. These findings not only offer novel insights into the potential etiological mechanisms underlying AMD but also suggest possible directions for future clinical interventions.


Sujet(s)
Maladies auto-immunes , Étude d'association pangénomique , Dégénérescence maculaire , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Humains , Dégénérescence maculaire/génétique , Maladies auto-immunes/génétique , Maladies auto-immunes/épidémiologie , Sclérose en plaques/génétique , Polyarthrite rhumatoïde/génétique , Mâle , Diabète de type 1/génétique , Maladies inflammatoires intestinales/génétique , Femelle
10.
Sci Transl Med ; 16(750): eadi4125, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38838135

RÉSUMÉ

Chronic inflammation is a constitutive component of many age-related diseases, including age-related macular degeneration (AMD). Here, we identified interleukin-1 receptor-associated kinase M (IRAK-M) as a key immunoregulator in retinal pigment epithelium (RPE) that declines during the aging process. Rare genetic variants of IRAK3, which encodes IRAK-M, were associated with an increased likelihood of developing AMD. In human samples and mouse models, IRAK-M abundance in the RPE declined with advancing age or exposure to oxidative stress and was further reduced in AMD. Irak3-knockout mice exhibited an increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M led to a disruption in RPE cell homeostasis, characterized by compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of adeno-associated virus (AAV)-expressing human IRAK3 rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in Irak3-knockout mice. Our data show that replenishment of IRAK-M in the RPE may redress dysregulated pro-inflammatory processes in AMD, suggesting a potential treatment for retinal degeneration.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Souris knockout , Stress oxydatif , Dégénérescence de la rétine , Épithélium pigmentaire de la rétine , Animaux , Humains , Mâle , Souris , Vieillissement de la cellule , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/génétique , Souris de lignée C57BL , Mitochondries/métabolisme , Dégénérescence de la rétine/métabolisme , Dégénérescence de la rétine/anatomopathologie , Dégénérescence de la rétine/génétique , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie
11.
Sci Rep ; 14(1): 13034, 2024 06 06.
Article de Anglais | MEDLINE | ID: mdl-38844476

RÉSUMÉ

The risk of developing age-related macular degeneration (AMD) is influenced by genetic background. In 2016, the International AMD Genomics Consortium (IAMDGC) identified 52 risk variants in 34 loci, and a polygenic risk score (PRS) from these variants was associated with AMD. The Israeli population has a unique genetic composition: Ashkenazi Jewish (AJ), Jewish non-Ashkenazi, and Arab sub-populations. We aimed to perform a genome-wide association study (GWAS) for AMD in Israel, and to evaluate PRSs for AMD. Our discovery set recruited 403 AMD patients and 256 controls at Hadassah Medical Center. We genotyped individuals via custom exome chip. We imputed non-typed variants using cosmopolitan and AJ reference panels. We recruited additional 155 cases and 69 controls for validation. To evaluate predictive power of PRSs for AMD, we used IAMDGC summary-statistics excluding our study and developed PRSs via clumping/thresholding or LDpred2. In our discovery set, 31/34 loci reported by IAMDGC were AMD-associated (P < 0.05). Of those, all effects were directionally consistent with IAMDGC and 11 loci had a P-value under Bonferroni-corrected threshold (0.05/34 = 0.0015). At a 5 × 10-5 threshold, we discovered four suggestive associations in FAM189A1, IGDCC4, C7orf50, and CNTNAP4. Only the FAM189A1 variant was AMD-associated in the replication cohort after Bonferroni-correction. A prediction model including LDpred2-based PRS + covariates had an AUC of 0.82 (95% CI 0.79-0.85) and performed better than covariates-only model (P = 5.1 × 10-9). Therefore, previously reported AMD-associated loci were nominally associated with AMD in Israel. A PRS developed based on a large international study is predictive in Israeli populations.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Dégénérescence maculaire , Polymorphisme de nucléotide simple , Humains , Dégénérescence maculaire/génétique , Dégénérescence maculaire/épidémiologie , Israël/épidémiologie , Femelle , Mâle , Sujet âgé , Facteurs de risque , Adulte d'âge moyen , Études cas-témoins , Sujet âgé de 80 ans ou plus , Hérédité multifactorielle/génétique , Juif/génétique , Génotype
12.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928273

RÉSUMÉ

Age-related macular degeneration (AMD) is a chronic disease, which often develops in older people, but this is not the rule. AMD pathogenesis changes include the anatomical and functional complex. As a result of damage, it occurs, in the retina and macula, among other areas. These changes may lead to partial or total loss of vision. This disease can occur in two clinical forms, i.e., dry (progression is slowly and gradually) and exudative (wet, progression is acute and severe), which usually started as dry form. A coexistence of both forms is possible. AMD etiology is not fully understood. Extensive genetic studies have shown that this disease is multifactorial and that genetic determinants, along with environmental and metabolic-functional factors, are important risk factors. This article reviews the impact of heavy metals, macro- and microelements, and genetic factors on the development of AMD. We present the current state of knowledge about the influence of environmental factors and genetic determinants on the progression of AMD in the confrontation with our own research conducted on the Polish population from Kuyavian-Pomeranian and Lubusz Regions. Our research is concentrated on showing how polluted environments of large agglomerations affects the development of AMD. In addition to confirming heavy metal accumulation, the growth of risk of acute phase factors and polymorphism in the genetic material in AMD development, it will also help in the detection of new markers of this disease. This will lead to a better understanding of the etiology of AMD and will help to establish prevention and early treatment.


Sujet(s)
Dégénérescence maculaire , Humains , Dégénérescence maculaire/génétique , Dégénérescence maculaire/étiologie , Facteurs de risque , Prédisposition génétique à une maladie , Métaux lourds/toxicité , Métaux lourds/effets indésirables , Exposition environnementale/effets indésirables , Immunogénétique
13.
Stem Cell Res ; 78: 103458, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870564

RÉSUMÉ

The Stargardt's Disease, Type 1 (STGD1) is associated with the loss of function mutations in ABCA4. This gene codes for a retina-specific, ATP-binding cassette (ABC) family transporter, involved in the transport of the key visual cycle intermediate, all-trans-retinaldehyde (atRAL), across the photoreceptor cell membranes. Here, we report the establishment of a patient-specific, iPSC line (LVPEIi008-A), that carries a homozygous nonsense mutation at (c.6088C > T) position, within exon 44 of ABCA4. The patient-specific skin fibroblasts were reprogrammed using episomal plasmids and the stably expanding iPSC line expressed the key stemness and pluripotency markers, maintained its chromosomal integrity and tested negative for mycoplasma.


Sujet(s)
Transporteurs ABC , Codon non-sens , Exons , Cellules souches pluripotentes induites , Maladie de Stargardt , Cellules souches pluripotentes induites/métabolisme , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Maladie de Stargardt/anatomopathologie , Humains , Homozygote , Lignée cellulaire , Dégénérescence maculaire/génétique , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/métabolisme
14.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167239, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38750770

RÉSUMÉ

Fibulin-3 (FBLN3, aka EFEMP1) is a secreted extracellular matrix (ECM) glycoprotein implicated in ocular diseases including glaucoma and age-related macular degeneration. Yet surprisingly, little is known about its native biology, expression patterns, and localization in the eye. To overcome these shortcomings, we conducted gene expression analysis and immunohistochemistry for FBLN3 in ocular tissues from mice, pigs, non-human primates, and humans. Moreover, we evaluated age-related changes in FBLN3 and FBLN3-related ECM remodeling enzymes/inhibitors in aging mice. We found that FBLN3 displayed distinct staining patterns consistent across the mouse retina, particularly in the ganglion cell layer and inner nuclear layer (INL). In contrast, human retinas exhibited a unique staining pattern, with enrichment of FBLN3 in the retinal pigment epithelium (RPE), INL, and outer nuclear layer (ONL) in the peripheral retina. This staining transitioned to the outer plexiform layer (OPL) in the central retina/macula, and was accompanied by reduced RPE immunoreactivity approaching the fovea. Surprisingly, we found significant age-related increases in FBLN3 expression and protein abundance in the mouse retina which was paralleled by reduced transcript levels of FBLN3-degrading enzymes (i.e., Mmp2 and Htra1). Our findings highlight important species-dependent, retinal region-specific, and age-related expression and localization patterns of FBLN3 which favor its accumulation during aging. These findings contribute to a better understanding of FBLN3's role in ocular pathology and provide valuable insights for future FBLN3 research.


Sujet(s)
Vieillissement , Protéines de la matrice extracellulaire , Animaux , Humains , Souris , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Vieillissement/métabolisme , Vieillissement/génétique , Rétine/métabolisme , Suidae , Mâle , Souris de lignée C57BL , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/génétique , Épithélium pigmentaire de la rétine/métabolisme , Femelle , Spécificité d'espèce , Sujet âgé
15.
Aging (Albany NY) ; 16(9): 8044-8069, 2024 05 10.
Article de Anglais | MEDLINE | ID: mdl-38742956

RÉSUMÉ

Age-related macular degeneration (AMD) is a condition causing progressive central vision loss. Growing evidence suggests a link between cellular senescence and AMD. However, the exact mechanism by which cellular senescence leads to AMD remains unclear. Employing machine learning, we established an AMD diagnostic model. Through unsupervised clustering, two distinct AMD subtypes were identified. GO, KEGG, and GSVA analyses explored the diverse biological functions associated with the two subtypes. By WGCNA, we constructed a coexpression network of differential genes between the subtypes, revealing the regulatory role of hub genes at the level of transcription factors and miRNAs. We identified 5 genes associated with inflammation for the construction of the AMD diagnostic model. Additionally, we observed that the level of cellular senescence and pathways related to programmed cell death (PCD), such as ferroptosis, necroptosis, and pyroptosis, exhibited higher expression levels in subtype B than A. Immune microenvironments also differed between the subtypes, indicating potentially distinct pathogenic mechanisms and therapeutic targets. In summary, by leveraging cellular senescence-associated gene expression, we developed an AMD diagnostic model. Furthermore, we identified two subtypes with varying expression patterns of senescence genes, revealing their differential roles in programmed cell death, disease progression, and immune microenvironments within AMD.


Sujet(s)
Vieillissement de la cellule , Biologie informatique , Dégénérescence maculaire , Vieillissement de la cellule/génétique , Dégénérescence maculaire/génétique , Dégénérescence maculaire/diagnostic , Dégénérescence maculaire/anatomopathologie , Humains , Réseaux de régulation génique , Analyse de profil d'expression de gènes , Apprentissage machine , microARN/génétique , microARN/métabolisme
16.
Ophthalmic Res ; 67(1): 358-386, 2024.
Article de Anglais | MEDLINE | ID: mdl-38754401

RÉSUMÉ

INTRODUCTION: Anti-vascular endothelial growth factor (anti-VEGF) agents have a variable effect on patients with age-related macular degeneration (AMD) that has been attributed to several causes, including genetic factors. We evaluated the effects of Complement Factor H (CFH) rs1061170/Y402H polymorphism on the response to anti-VEGF therapy among AMD patients. METHODS: PubMed, Scopus, EMBASE, Web of Science, and Google Scholar were used for a literature search. Pooled odds ratios (ORs) and their 95% confidence intervals (CIs) were estimated to assess the effects of CFH Y402H polymorphism on the response to anti-VEGF therapy in AMD. I2 was used to present the amount of heterogeneity. We used STATA version 14.0 software. RESULTS: Twenty-five papers reporting data for 4,681 patients were included in this study. Better response to anti-VEGF therapy was seen in T over C (OR = 1.25, 95% CI = 1.04-1.50), TT over CC (OR = 1.60, 95% CI = 1.06-2.4), and TT + TC over CC (OR = 1.68, 95% CI = 1.23-2.28) genotypes. There was no significant difference in the three other genetic models (TT vs. TC, TT vs. TC + CC, TC vs. TT + CC). In Asians, no significant difference was observed in all six genetic models. Ranibizumab and bevacizumab had similar efficacy; however, conbercept was more effective in homozygous genotypes. The literature indicated that TT and TC genotypes and T allele were associated with a better functional response, while the CC genotype and C alleles had a better anatomical response. The combination of risk alleles in ARMS2 A69S (rs10490924), VEGF-A (rs699947), and VEGF-A (rs833069) with Y420H is a predictor of non-respondents. CONCLUSION: In patients with AMD, the CFH Y402H is a predictor of the response to anti-VEGF agents and should be considered in the treatment plan.


Sujet(s)
Inhibiteurs de l'angiogenèse , Facteur H du complément , Dégénérescence maculaire , Polymorphisme de nucléotide simple , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur H du complément/génétique , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/génétique , Inhibiteurs de l'angiogenèse/usage thérapeutique , Dégénérescence maculaire/génétique , Dégénérescence maculaire/traitement médicamenteux , Génotype
17.
Immunohorizons ; 8(5): 363-370, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38775688

RÉSUMÉ

Although the pathogenesis of choroidal neovascularization (CNV) is largely unknown in age-related macular degeneration (AMD), inflammasomes may contribute to CNV development and progression. To understand the role NLRP3 inflammasomes in CNV, we used Ccr2RFPCx3cr1GFP dual-reporter mice and immunostaining techniques to confirm localization of NLRP3 inflammasomes in the laser-induced CNV (LCNV) lesions. Confocal microscopy was used to image and quantify LCNV volumes. MCC950 was used as NLRP3 inhibitor. ELISA and quantitative RT-PCR were used to confirm the activation of NLRP3 by monitoring the expression of IL-1ß protein and mRNA in choroidal tissues from LCNV mice. In addition, NLRP3 (-/-) LCNV mice were used to investigate whether NLRP3 inflammasomes contribute to the development of LCNV lesions. We observed that red fluorescent protein (RFP)-positive monocyte-derived macrophages and GFP-positive microglia-derived macrophages, in addition to other cell types, were localized in LCNV lesions at day 7 post-laser injury. In addition, NLRP3 inflammasomes are associated with LCNV lesions. Inhibition of NLRP3 inflammasomes, using MCC950, caused an increased Ccr2RFP-positive macrophages, Cx3cr1GFP-positive microglia, and other cells, resulting in an increase in total lesion size. NLRP3 (-/-) LCNV mice showed significantly increased lesion size compared with age-matched controls. Inhibition of NLRP3 resulted in decreased IL-1ß mRNA and protein expression in the choroidal tissues, suggesting that increased lesion size may not be directly related to IL-1ß.


Sujet(s)
Néovascularisation choroïdienne , Indènes , Inflammasomes , Interleukine-1 bêta , Microglie , Monocytes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Néovascularisation choroïdienne/métabolisme , Néovascularisation choroïdienne/anatomopathologie , Souris , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Microglie/métabolisme , Monocytes/métabolisme , Souris knockout , Sulfones/pharmacologie , Souris de lignée C57BL , Furanes/pharmacologie , Récepteurs CCR2/métabolisme , Récepteurs CCR2/génétique , Macrophages/métabolisme , Macrophages/immunologie , Sulfonamides/pharmacologie , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Protéines de transport/métabolisme , Protéines de transport/génétique , Choroïde/métabolisme , Choroïde/anatomopathologie , Modèles animaux de maladie humaine , Lasers/effets indésirables , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/génétique
18.
Hum Cell ; 37(4): 1056-1069, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38744794

RÉSUMÉ

Epithelial mesenchymal transition (EMT) occurring in retinal pigment epithelial cells (RPE) is a crucial mechanism that contributes to the development of age-related macular degeneration (AMD), a pivotal factor leading to permanent vision impairment. Long non-coding RNAs (lncRNAs) have emerged as critical regulators orchestrating EMT in RPE cells. In this study, we explored the function of the lncRNA CYTOR (cytoskeleton regulator RNA) in EMT of RPE cells and its underlying mechanisms. Through weighted correlation network analysis, we identified CYTOR as an EMT-related lncRNA associated with AMD. Experimental validation revealed that CYTOR orchestrates TGF-ß1-induced EMT, as well as proliferation and migration of ARPE-19 cells. Further investigation demonstrated the involvement of CYTOR in regulating the WNT5A/NFAT1 pathway and NFAT1 intranuclear translocation in the ARPE-19 cell EMT model. Mechanistically, CHIP, EMSA and dual luciferase reporter assays confirmed NFAT1's direct binding to CYTOR's promoter, promoting transcription. Reciprocally, CYTOR overexpression promoted NFAT1 expression, while NFAT1 overexpression increased CYTOR transcription. These findings highlight a mutual promotion between CYTOR and NFAT1, forming a positive feedback loop that triggers the EMT phenotype in ARPE-19 cells. These discoveries provide valuable insights into the molecular mechanisms of EMT and its association with AMD, offering potential avenues for targeted therapies in EMT-related conditions, including AMD.


Sujet(s)
Transition épithélio-mésenchymateuse , Rétrocontrôle physiologique , Dégénérescence maculaire , Facteurs de transcription NFATC , ARN long non codant , Épithélium pigmentaire de la rétine , Transition épithélio-mésenchymateuse/génétique , Humains , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/cytologie , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , ARN long non codant/physiologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Dégénérescence maculaire/génétique , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/étiologie , Expression des gènes/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Facteur de croissance transformant bêta-1/métabolisme , Transduction du signal/génétique , Transduction du signal/physiologie , Cellules épithéliales/métabolisme , Lignée cellulaire , Cellules cultivées
19.
Neurobiol Aging ; 140: 41-59, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38723422

RÉSUMÉ

Aging is the main risk factor for age-related macular degeneration (AMD), a retinal neurodegenerative disease that leads to irreversible blindness, particularly in people over 60 years old. Retinal pigmented epithelium (RPE) atrophy is an AMD hallmark. Genome-wide chromatin accessibility, DNA methylation, and gene expression studies of AMD and control RPE demonstrate epigenomic/transcriptomic changes occur during AMD onset and progression. However, mechanisms by which molecular alterations of normal aging impair RPE function and contribute to AMD pathogenesis are unclear. Here, we specifically interrogate the RPE translatome with advanced age and across sexes in a novel RPE reporter mouse model. We find differential age- and sex- associated transcript expression with overrepresentation of pathways related to inflammation in the RPE. Concordant with impaired RPE function, the phenotypic changes in the aged translatome suggest that aged RPE becomes immunologically active, in both males and females, with some sex-specific signatures, which supports the need for sex representation for in vivo studies.


Sujet(s)
Vieillissement , Dégénérescence maculaire , Épithélium pigmentaire de la rétine , Caractères sexuels , Animaux , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie , Femelle , Mâle , Vieillissement/génétique , Vieillissement/physiologie , Vieillissement/anatomopathologie , Dégénérescence maculaire/génétique , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/étiologie , Transcriptome , Modèles animaux de maladie humaine , Expression des gènes , Inflammation , Souris , Souris de lignée C57BL
20.
Nat Commun ; 15(1): 3780, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38710714

RÉSUMÉ

Recombinant adeno-associated viruses (rAAVs) have emerged as promising gene therapy vectors due to their proven efficacy and safety in clinical applications. In non-human primates (NHPs), rAAVs are administered via suprachoroidal injection at a higher dose. However, high doses of rAAVs tend to increase additional safety risks. Here, we present a novel AAV capsid (AAVv128), which exhibits significantly enhanced transduction efficiency for photoreceptors and retinal pigment epithelial (RPE) cells, along with a broader distribution across the layers of retinal tissues in different animal models (mice, rabbits, and NHPs) following intraocular injection. Notably, the suprachoroidal delivery of AAVv128-anti-VEGF vector completely suppresses the Grade IV lesions in a laser-induced choroidal neovascularization (CNV) NHP model for neovascular age-related macular degeneration (nAMD). Furthermore, cryo-EM analysis at 2.1 Å resolution reveals that the critical residues of AAVv128 exhibit a more robust advantage in AAV binding, the nuclear uptake and endosome escaping. Collectively, our findings highlight the potential of AAVv128 as a next generation ocular gene therapy vector, particularly using the suprachoroidal delivery route.


Sujet(s)
Néovascularisation choroïdienne , Dependovirus , Thérapie génétique , Vecteurs génétiques , Épithélium pigmentaire de la rétine , Animaux , Dependovirus/génétique , Vecteurs génétiques/génétique , Vecteurs génétiques/administration et posologie , Thérapie génétique/méthodes , Souris , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/virologie , Néovascularisation choroïdienne/thérapie , Néovascularisation choroïdienne/génétique , Lapins , Humains , Techniques de transfert de gènes , Dégénérescence maculaire/thérapie , Dégénérescence maculaire/génétique , Dégénérescence maculaire/anatomopathologie , Modèles animaux de maladie humaine , Protéines de capside/génétique , Protéines de capside/métabolisme , Transduction génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Souris de lignée C57BL , Rétine/métabolisme , Rétine/virologie , Mâle , Cellules HEK293
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...