Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.366
Filter
1.
BMC Ophthalmol ; 24(1): 327, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107704

ABSTRACT

BACKGROUND: Occult Macular Dystrophy (OMD), primarily caused by retinitis pigmentosa 1-like 1 (RP1L1) variants, is a complex retinal disease characterised by progressive vision loss and a normal fundus appearance. This study aims to investigate the diverse phenotypic expressions and genotypic correlations of OMD in Chinese patients, including a rare case of Vitelliform Macular Dystrophy (VMD) associated with RP1L1. METHODS: We analysed seven OMD patients and one VMD patient, all with heterozygous pathogenic RP1L1 variants. Clinical assessments included Best Corrected Visual Acuity (BCVA), visual field testing, Spectral Domain Optical Coherence Tomography (SD-OCT), multifocal Electroretinograms (mfERGs), and microperimetry. Next-generation sequencing was utilised for genetic analysis. RESULTS: The OMD patients displayed a range of phenotypic variability. Most (5 out of 7) had the RP1L1 variant c.133 C > T; p.R45W, associated with central vision loss and specific patterns in SD-OCT and mfERG. Two patients exhibited different RP1L1 variants (c.3599G > T; p.G1200V and c.2880G > C; p.W960C), presenting milder phenotypes. SD-OCT revealed photoreceptor layer changes, with most patients showing decreased mfERG responses in the central rings. Interestingly, a unique case of VMD linked to the RP1L1 variant was observed, distinct from traditional OMD presentations. CONCLUSIONS: This study highlights the phenotypic diversity within OMD and the broader spectrum of RP1L1-associated macular dystrophies, including a novel association with VMD. The findings emphasise the complexity of RP1L1 variants in determining clinical manifestations, underscoring the need for comprehensive genetic and clinical evaluations in macular dystrophies.


Subject(s)
Electroretinography , Eye Proteins , Microtubule-Associated Proteins , Tomography, Optical Coherence , Visual Acuity , Vitelliform Macular Dystrophy , Humans , Male , Female , Tomography, Optical Coherence/methods , Adult , Middle Aged , Eye Proteins/genetics , Visual Acuity/physiology , Vitelliform Macular Dystrophy/genetics , Vitelliform Macular Dystrophy/physiopathology , Vitelliform Macular Dystrophy/diagnosis , Microtubule-Associated Proteins/genetics , Visual Fields/physiology , China/epidemiology , Young Adult , Visual Field Tests , Pedigree , Adolescent , Phenotype , Mutation , Macular Degeneration/genetics , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Asian People/genetics , Aged , East Asian People
2.
Mol Genet Genomic Med ; 12(8): e2505, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39108195

ABSTRACT

BACKGROUND: Biallelic variants in the major facilitator superfamily domain containing 8 gene (MFSD8) are associated with distinct clinical presentations that range from typical late-infantile neuronal ceroid lipofuscinosis type 7 (CLN7 disease) to isolated adult-onset retinal dystrophy. Classic late-infantile CLN7 disease is a severe, rare neurological disorder with an age of onset typically between 2 and 6 years, presenting with seizures and/or cognitive regression. Its clinical course is progressive, leading to premature death, and often includes visual loss due to severe retinal dystrophy. In rare cases, pathogenic variants in MFSD8 can be associated with isolated non-syndromic macular dystrophy with variable age at onset, in which the disease process predominantly or exclusively affects the cones of the macula and where there are no neurological or neuropsychiatric manifestations. METHODS: Here we present longitudinal studies on four adult-onset patients who were biallelic for four MFSD8 variants. RESULTS: Two unrelated patients who presented with adult-onset ataxia and had macular dystrophy on examination were homozygous for a novel variant in MFSD8 NM_152778.4: c.935T>C p.(Ile312Thr). Two other patients presented in adulthood with visual symptoms, and one of these developed mild to moderate cerebellar ataxia years after the onset of visual symptoms. CONCLUSIONS: Our observations expand the knowledge on biallelic pathogenic MFSD8 variants and confirm that these are associated with a spectrum of more heterogeneous clinical phenotypes. In MFSD8-related disease, adult-onset recessive ataxia can be the presenting manifestation or may occur in combination with retinal dystrophy.


Subject(s)
Macular Degeneration , Humans , Adult , Male , Female , Macular Degeneration/genetics , Macular Degeneration/pathology , Age of Onset , Ataxia/genetics , Ataxia/pathology , Alleles , Middle Aged , Mutation , Membrane Transport Proteins/genetics , Phenotype
3.
JCI Insight ; 9(15)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39114980

ABSTRACT

Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) is an age-related macular degeneration-like (AMD-like) retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production in retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus that enabled simple, sensitive, and high-throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix, reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium-derived factor). In vivo, treatment of 8-month-old R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is an important demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of retinal degenerative diseases, including potentially AMD itself.


Subject(s)
Extracellular Matrix Proteins , Extracellular Matrix , Macular Degeneration , Retinal Pigment Epithelium , Animals , Mice , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Macular Degeneration/pathology , Macular Degeneration/genetics , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Humans , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Disease Models, Animal , Retinal Dystrophies/metabolism , Retinal Dystrophies/pathology , Retinal Dystrophies/genetics , Optic Disk Drusen/congenital
4.
J Mol Neurosci ; 74(3): 74, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39107525

ABSTRACT

Age-related macular degeneration (AMD) is one of the most common causes of irreversible vision loss in the elderly. Its pathogenesis is likely multifactorial, involving a complex interaction of metabolic and environmental factors, and remains poorly understood. Previous studies have shown that mitochondrial dysfunction and oxidative stress play a crucial role in the development of AMD. Oxidative damage to the retinal pigment epithelium (RPE) has been identified as one of the major mediators in the pathogenesis of age-related macular degeneration (AMD). Therefore, this article combines transcriptome sequencing (RNA-seq) and single-cell sequencing (scRNA-seq) data to explore the role of mitochondria-related genes (MRGs) in AMD. Firstly, differential expression analysis was performed on the raw RNA-seq data. The intersection of differentially expressed genes (DEGs) and MRGs was performed. This paper proposes a deep subspace nonnegative matrix factorization (DS-NMF) algorithm to perform a multi-layer nonlinear transformation on the intersection of gene expression profiles corresponding to AMD samples. The age of AMD patients is used as prior information at the network's top level to change the data distribution. The classification is based on reconstructed data with altered distribution. The types obtained significantly differ in scores of multiple immune-related pathways and immune cell infiltration abundance. Secondly, an optimal AMD diagnosis model was constructed using multiple machine learning algorithms for external and qRT-PCR verification. Finally, ten potential therapeutic drugs for AMD were identified based on cMAP analysis. The AMD subtypes identified in this article and the diagnostic model constructed can provide a reference for treating AMD and discovering new drug targets.


Subject(s)
Biomarkers , Macular Degeneration , Transcriptome , Humans , Macular Degeneration/genetics , Macular Degeneration/metabolism , Biomarkers/metabolism , Machine Learning , Single-Cell Analysis/methods , Mitochondria/genetics , Mitochondria/metabolism , Multiomics
5.
Transl Vis Sci Technol ; 13(8): 2, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087930

ABSTRACT

Purpose: Homozygous hypomorphic variants of the RP1 gene, including c.5797C>T, p.Arg1933Ter, have traditionally been considered non-pathogenic. This study aimed to elucidate the clinical manifestations of late-onset, slowly progressive cone/macular dystrophy in patients homozygous for p.Arg1933Ter in the RP1 gene. Methods: Five patients with biallelic p.Arg1933Ter in RP1 were retrospectively recruited, and their clinical profiles were analyzed. Copy number variation analysis and Alu insertion assessment of genes associated with inherited retinal diseases were conducted. The results of comprehensive ophthalmological examinations, multimodal imaging, and full-field electroretinogram tests were analyzed. Results: No specific sequencing errors or structural variations associated with the clinical phenotypes were identified. Alu element insertion in RP1 was not detected. The mean ± SD age at the first visit was 62.2 ± 9.8 years, with symptoms typically starting between 45 and 50 years of age. Two patients exhibited a mild form of cone/macular dystrophy, characterized by a relatively preserved fundus appearance and blurring of the ellipsoid zone on optical coherence tomography. Three patients had late-onset cone/macular dystrophy with significant atrophy. Conclusions: To our knowledge, this study is the first to report that a homozygous hypomorphic variant of RP1, previously considered non-pathogenic, leads to cone/macular dystrophy. Translational Relevance: The study introduces novel possibilities suggesting that the homozygous hypomorphic variant of RP1 may be linked to variant pathogenicity.


Subject(s)
Electroretinography , Eye Proteins , Tomography, Optical Coherence , Humans , Male , Female , Middle Aged , Retrospective Studies , Aged , Eye Proteins/genetics , Visual Acuity , DNA Copy Number Variations/genetics , Disease Progression , Cone Dystrophy/genetics , Cone Dystrophy/diagnostic imaging , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/diagnostic imaging , Macular Degeneration/congenital , Pedigree , Homozygote , Phenotype , Mutation , Adult , Age of Onset , Microtubule-Associated Proteins
6.
Int Ophthalmol ; 44(1): 348, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141020

ABSTRACT

PURPOSE: This study aimed to investigate the association between the CC-cytokine ligand-2 (CCL2) 2518A/G (rs1024611) single nucleotide polymorphism (SNP) and susceptibility to age-related macular degeneration (AMD). METHODS: PubMed, Embase, Web of Science, and other databases were searched for articles published before August 24, 2023. After searching, data extraction, and quality assessment, meta-analysis and trial sequential analysis were conducted using RevMan 5.4, Stata 17.0, and TSA 0.9.5.10 Beta software. Combined OR, P values, and 95% confidence intervals (CIs) were calculated. Sensitivity analysis, subgroup analysis and publication bias assessment were also performed. RESULTS: Six articles, comprising 1186 cases and 1124 controls, were included. No significant statistical difference was found in six main outcomes. However, due to observed heterogeneity and high sensitivity, subgroup analysis was performed, revealing statistically significant differences across different regions. No significant publication bias was observed. Trial sequential analysis suggested the need for additional follow-up case-control studies to further validate the findings. CONCLUSION: The CCL2 gene 2518A/G (rs1024611) polymorphism is associated with AMD susceptibility. Among Caucasian populations in West Asia and Europe, the G allele is protective against AMD, whereas in East and South Asia, it poses a risk factor.


Subject(s)
Chemokine CCL2 , Genetic Predisposition to Disease , Macular Degeneration , Polymorphism, Single Nucleotide , Humans , Macular Degeneration/genetics , Chemokine CCL2/genetics
7.
Transl Vis Sci Technol ; 13(8): 14, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39115837

ABSTRACT

Purpose: Emerging research indicates a link between the intake of fatty fish and age-related macular degeneration (AMD). However, observational studies fall short in establishing a direct causal link between oily fish intake and AMD. We wanted to determine whether causal association lies between oily fish intake and age-related macular degeneration (AMD) risk in human beings. Methods: This two-sample mendelian randomization (MR) study used the MR method to probe the genetic causality in the relationship between oily fish intake and AMD. The genome-wide association study (GWAS) data for AMD were acquired from a Finnish database, whereas the data on fish oil intake came from the UK Biobank. The analysis used several approaches such as inverse-variance weighted (IVW), MR Egger, weighted median, simple mode, and weighted mode MR. In addition, the Cochran's Q test was used to evaluate heterogeneity in the MR data. The MR-Egger intercept and MR-pleiotropy residual sum and outlier (MR-PRESSO) tests were used to assess the presence of horizontal pleiotropy. A leave-one-out sensitivity analysis was conducted to determine the reliability of the association. Results: The IVW method revealed that the intake of oily fish is an independent risk factor for AMD (P = 0.034). It also suggested a minimal likelihood of horizontal pleiotropy affecting the causality (P > 0.05), with no substantial heterogeneity detected in the genetic variants (P > 0.05). The leave-one-out analysis confirmed the reliability and stability of this correlation. Conclusions: This research used a two-sample MR analysis to provide evidence of a genetic causal relationship between the eating of oily fish and AMD. This discovery held potential significance in AMD prevention.


Subject(s)
Fish Oils , Genome-Wide Association Study , Macular Degeneration , Mendelian Randomization Analysis , Mendelian Randomization Analysis/methods , Macular Degeneration/genetics , Macular Degeneration/epidemiology , Macular Degeneration/etiology , Humans , Fish Oils/administration & dosage , Risk Factors , Polymorphism, Single Nucleotide , Animals , Genetic Predisposition to Disease , Fishes/genetics , Finland/epidemiology
8.
Lipids Health Dis ; 23(1): 206, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951820

ABSTRACT

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.


Subject(s)
Delta-5 Fatty Acid Desaturase , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Genome-Wide Association Study , Macular Degeneration , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Macular Degeneration/blood , Macular Degeneration/genetics , Fatty Acids, Omega-3/blood , Male , Female , Fatty Acids, Omega-6/blood , Aged , Fatty Acid Desaturases/genetics , Middle Aged , Triglycerides/blood , Fatty Acids/blood , Risk Factors
9.
Front Immunol ; 15: 1421012, 2024.
Article in English | MEDLINE | ID: mdl-38979414

ABSTRACT

Objective: This study revealed a core regulator and common upstream mechanisms for the multifaceted pathological processes of age-related macular degeneration (AMD) and provided proof-of-concept for this new therapeutic target. Methods: Comprehensive gene expression analysis was performed using RNA sequencing of eye cup from old mice as well as laser-induced choroidal neovascularization (CNV) mouse model. Through integrative analysis and protein-protein interaction (PPI) analysis, common pathways and key transcription factor was identified simultaneously engaged in age-related retinal degeneration and CNV, the two typical pathological process of AMD. Subsequently, the expression changes of Spi1, the key regulator, as well as the alternation of the downstream mechanisms were validated in both models through qRT-PCR, Elisa, flow cytometry and immunofluorescence. Further, we assessed the impact of Spi1 knockdown in vitro and in vivo using gene intervention vectors carried by adeno-associated virus or lentivirus to test its potential as a therapeutic target. Results: Compared to corresponding controls, we found 1,939 and 1,319 genes differentially expressed in eye cups of old and CNV mice respectively. The integrative analysis identified a total of 275 overlapping DEGs, of which 150 genes were co-upregulated. PPI analysis verified a central transcription factor, SPI1. The significant upregulation of Spi1 expression was then validated in both models, accompanied by macrophage polarization towards the M1 phenotype. Finally, SPI1 suppression significantly inhibited M1 polarization of BMDMs and attenuated neovascularization in CNV mice. Conclusion: This study demonstrates that SPI1 exerts a pivotal role in AMD by regulation of macrophage polarization and innate immune response, offering promise as an innovative target for treating AMD.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Macrophages , Macular Degeneration , Trans-Activators , Animals , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Macrophages/immunology , Macrophages/metabolism , Choroidal Neovascularization/immunology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Mice, Inbred C57BL , Macrophage Activation/genetics , Humans , Gene Expression Profiling , Male
10.
Zhonghua Yan Ke Za Zhi ; 60(7): 601-610, 2024 Jul 11.
Article in Chinese | MEDLINE | ID: mdl-38955762

ABSTRACT

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.


Subject(s)
Fibrillin-1 , Genotype , Marfan Syndrome , Adolescent , Child , Female , Humans , Male , Young Adult , Adipokines , Anterior Eye Segment , Biometry , Cross-Sectional Studies , Fibrillin-1/genetics , Macular Degeneration/genetics , Marfan Syndrome/genetics , Mutation , Posterior Eye Segment/pathology , Infant, Newborn , Infant , Child, Preschool
11.
Invest Ophthalmol Vis Sci ; 65(8): 34, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39028977

ABSTRACT

Purpose: A single-nucleotide polymorphism in HTRA1 has been linked to age-related macular degeneration (AMD). Here we investigated the potential links between age-related retinal changes, elastin turnover, elastin autoantibody production, and complement C3 deposition in a mouse model with RPE-specific human HTRA1 overexpression. Methods: HTRA1 transgenic mice and age-matched CD1 wild-type mice were analyzed at 6 weeks and 4, 6, and 12 to 14 months of age using in vivo retinal imaging by optical coherence tomography (OCT) and fundus photography, as well as molecular readouts, focusing on elastin and elastin-derived peptide quantification, antielastin autoantibody, and total Ig antibody measurements and immunohistochemistry to examine elastin, IgG, and C3 protein levels in retinal sections. Results: OCT imaging indicated thinning of inner nuclear layer as an early phenotype in HTRA1 mice, followed by age and age/genotype-related thinning of the photoreceptor layer, RPE, and total retina. HTRA1 mice exhibited reduced elastin protein levels in the RPE/choroid and increased elastin breakdown products in the retina and serum. A corresponding age-dependent increase of serum antielastin IgG and IgM autoantibodies and total Ig antibody levels was observed. In the RPE/choroid, these changes were associated with an age-related increase of IgG and C3 deposition. Conclusions: Our results confirm that RPE-specific overexpression of human HTRA1 induces certain AMD-like phenotypes in mice. This includes altered elastin turnover, immune response, and complement deposition in the RPE/choroid in addition to age-related outer retinal and photoreceptor layer thinning. The identification of elastin-derived peptides and corresponding antielastin autoantibodies, together with increased C3 deposition in the RPE/choroid, provides a rationale for an overactive complement system in AMD irrespective of the underlying genetic risk.


Subject(s)
Disease Models, Animal , Elastin , High-Temperature Requirement A Serine Peptidase 1 , Macular Degeneration , Mice, Transgenic , Retinal Pigment Epithelium , Tomography, Optical Coherence , Animals , Humans , Mice , Aging , Autoantibodies/blood , Complement C3/genetics , Complement C3/metabolism , Elastin/metabolism , Elastin/genetics , Gene Expression Regulation , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Immunoglobulin G/blood , Immunohistochemistry , Macular Degeneration/genetics , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
12.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999967

ABSTRACT

Age-related macular degeneration (AMD) is a progressive neurodegenerative condition leading to vision loss and eventual blindness, with exudative AMD posing a heightened risk due to choroidal neovascularization and localized edema. Therapies targeting the VEGF pathway aim to address this mechanism for treatment effectiveness. Our study aimed to evaluate associations between specific genetic variants (RAD51B rs8017304, rs2588809; TRIB1 rs6987702, rs4351379; COL8A1 rs13095226; COL10A1 rs1064583; IL-9 rs1859430, rs2069870, rs11741137, rs2069885, rs2069884; IL-10 rs1800871, rs1800872, rs1800896; VEGFA rs1570360, rs699947, rs3025033, rs2146323) and the response to anti-VEGF treatment for exudative AMD. We enrolled 119 patients with exudative AMD categorized as responders or non-responders based on their response to anti-VEGF treatment. Statistical analysis revealed that RAD51B rs8017304 heterozygous and homozygous minor allele carriers had increased CMT before treatment compared to wild-type genotype carriers (p = 0.004). Additionally, TRIB1 rs4351379 heterozygous and homozygous minor allele carriers exhibited a greater decrease in central macular thickness (CMT) after 6 months of treatment than wild-type genotype carriers (p = 0.030). IL-9 rs1859430, rs2069870, and rs2069884 heterozygous and homozygous minor allele carriers had worse BCVA before treatment than wild-type genotype carriers (p = 0.018, p = 0.012, p = 0.041, respectively). Conversely, IL-9 rs2069885 heterozygous and homozygous minor allele carriers showed greater improvement in BCVA after 6 months compared to wild-type genotype carriers (p = 0.032). Furthermore, VEGFA rs699947 heterozygous and homozygous minor allele carriers had better BCVA before treatment and after 3 and 6 months of treatment than wild-type genotype carriers (p = 0.003, p = 0.022, respectively), with these carriers also exhibiting higher CMT after 6 months of anti-VEGF treatment (p = 0.032). Not all results remained statistically significant under this stringent correction for multiple comparisons. The comparisons of the serum concentrations of IL-10, VEGF-A, and VEGF-R2/KDR between non-responders and responders did not yield statistically significant differences. Our study identified significant associations between genetic variants, including RAD51B rs8017304, TRIB1 rs4351379, IL-9 rs1859430, rs2069870, rs2069884, rs2069885, and VEGFA rs699947, and parameters related to the efficacy of exudative AMD treatment, such as BCVA and CMT.


Subject(s)
Collagen Type X , Interleukin-10 , Interleukin-9 , Intracellular Signaling Peptides and Proteins , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/genetics , Male , Female , Aged , Interleukin-10/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Interleukin-9/genetics , Collagen Type X/genetics , Treatment Outcome , Macular Degeneration/genetics , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Aged, 80 and over , DNA-Binding Proteins/genetics , Middle Aged , Genotype , Collagen Type VIII
13.
Int Immunopharmacol ; 139: 112678, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39068753

ABSTRACT

Previous observational studies have indicated a correlation between circulating inflammatory proteins and age-related macular degeneration (AMD), yet the causal nature of this relationship remains uncertain. This study aims to investigate the causal link between circulating inflammatory proteins and AMD utilizing a bidirectional two-sample Mendelian randomization approach. The findings indicated that elevated levels of four circulating inflammatory proteins, including C-C Motif Chemokine Ligand 11 (CCL11), Signaling Lymphocytic Activation Molecule Family Member 1 (SLAMF1), TNF Superfamily Member 11 (TRANCE) and Vascular Endothelial Growth Factor A (VEGF-A) lead to the increased risk of AMD, while increased levels of two circulating inflammatory proteins, including Fibroblast Growth Factor 19 (FGF-19) and Interleukin 10 Receptor Subunit Alpha (IL-10RA), resulted in the decreased risk of AMD. Conversely, the results from reverse Mendelian randomization suggested that the presence of AMD lead to the reduction in levels of 15 circulating inflammatory proteins. The findings of this study support the association between elevated levels of circulating inflammatory proteins and the risk of AMD, as well as the potential impact of AMD on reducing circulating inflammatory protein levels. CCL11, SLAMF1, TRANCE and VEGF-A are identified as potential molecular markers in the progression of AMD. These results offer a novel molecular therapeutic target for the prevention and treatment of AMD.


Subject(s)
Macular Degeneration , Mendelian Randomization Analysis , Humans , Macular Degeneration/blood , Macular Degeneration/genetics , Biomarkers/blood , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Polymorphism, Single Nucleotide
14.
PLoS One ; 19(7): e0305466, 2024.
Article in English | MEDLINE | ID: mdl-38990973

ABSTRACT

In previous animal model studies, we demonstrated the potential of rAAV2-sVEGFRv-1, which encodes a truncated variant of the alternatively spliced soluble version of VEGF receptor-1 (VEGFR1), as a human gene therapy for age-related macular degeneration (AMD) and diabetic retinopathy (DR). Here, we elucidate in vitro some of the mechanisms by which rAAV2-sVEGFRv-1 exerts its therapeutic effects. Human umbilical vein endothelial cells (HUVECs) were infected with rAAV2-sVEGFRv-1 or a control virus vector in the presence of members of the VEGF family to identify potential binding partners via ELISA, which showed that VEGF-A, VEGF-B, and placental growth factor (PlGF) are all ligands of its transgene product. In order to determine the effects of rAAV2-sVEGFRv-1 on cell proliferation and permeability, processes that are important to the progression AMD and DR, HUVECs were infected with the therapeutic virus vector under the stimulation of VEGF-A, the major driver of the neovascularization that characterizes the forms of these conditions most associated with vision loss. rAAV2-sVEGFRv-1 treatment, as a result, markedly reduced the extent to which these processes occurred, with the latter determined by measuring zonula occludens 1 expression. Finally, the human microglial HMC3 cell line was used to show the effects of the therapeutic virus vector upon inflammatory processes, another major contributor to angiogenic eye disease pathophysiology, with rAAV2-sVEGFRv-1 reducing therein the secretion of pro-inflammatory cytokines interleukin (IL)-1ß and IL-6. Combined with our previously published in vivo data, the in vitro activity of the expressed transgene here further demonstrates the great promise of rAAV2-sVEGFRv-1 as a potential human gene therapeutic for addressing angiogenic ocular conditions.


Subject(s)
Dependovirus , Genetic Therapy , Human Umbilical Vein Endothelial Cells , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Humans , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Dependovirus/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Cell Proliferation , Macular Degeneration/therapy , Macular Degeneration/genetics , Macular Degeneration/metabolism , Diabetic Retinopathy/therapy , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism , Placenta Growth Factor/genetics , Placenta Growth Factor/metabolism
15.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000382

ABSTRACT

Alzheimer's disease (AD) represents a prominent neurodegenerative disorder (NDD), accounting for the majority of dementia cases worldwide. In addition to memory deficits, individuals with AD also experience alterations in the visual system. As the retina is an extension of the central nervous system (CNS), the loss in retinal ganglion cells manifests clinically as decreased visual acuity, narrowed visual field, and reduced contrast sensitivity. Among the extensively studied retinal disorders, age-related macular degeneration (AMD) shares numerous aging processes and risk factors with NDDs such as cognitive impairment that occurs in AD. Histopathological investigations have revealed similarities in pathological deposits found in the retina and brain of patients with AD and AMD. Cellular aging processes demonstrate similar associations with organelles and signaling pathways in retinal and brain tissues. Despite these similarities, there are distinct genetic backgrounds underlying these diseases. This review comprehensively explores the genetic similarities and differences between AMD and AD. The purpose of this review is to discuss the parallels and differences between AMD and AD in terms of pathophysiology, genetics, and epigenetics.


Subject(s)
Alzheimer Disease , Biomarkers , Epigenesis, Genetic , Macular Degeneration , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Biomarkers/metabolism , Animals , Genetic Predisposition to Disease , Retina/metabolism , Retina/pathology
16.
Nat Commun ; 15(1): 6150, 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39034314

ABSTRACT

Non-neovascular or dry age-related macular degeneration (AMD) is a multi-factorial disease with degeneration of the aging retinal-pigmented epithelium (RPE). Lysosomes play a crucial role in RPE health via phagocytosis and autophagy, which are regulated by transcription factor EB/E3 (TFEB/E3). Here, we find that increased AKT2 inhibits PGC-1α to downregulate SIRT5, which we identify as an AKT2 binding partner. Crosstalk between SIRT5 and AKT2 facilitates TFEB-dependent lysosomal function in the RPE. AKT2/SIRT5/TFEB pathway inhibition in the RPE induced lysosome/autophagy signaling abnormalities, disrupted mitochondrial function and induced release of debris contributing to drusen. Accordingly, AKT2 overexpression in the RPE caused a dry AMD-like phenotype in aging Akt2 KI mice, as evident from decline in retinal function. Importantly, we show that induced pluripotent stem cell-derived RPE encoding the major risk variant associated with AMD (complement factor H; CFH Y402H) express increased AKT2, impairing TFEB/TFE3-dependent lysosomal function. Collectively, these findings suggest that targeting the AKT2/SIRT5/TFEB pathway may be an effective therapy to delay the progression of dry AMD.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Lysosomes , Macular Degeneration , Proto-Oncogene Proteins c-akt , Retinal Pigment Epithelium , Signal Transduction , Sirtuins , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Animals , Proto-Oncogene Proteins c-akt/metabolism , Sirtuins/metabolism , Sirtuins/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Humans , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Lysosomes/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Disease Models, Animal , Induced Pluripotent Stem Cells/metabolism , Male
17.
Invest Ophthalmol Vis Sci ; 65(6): 38, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38935028

ABSTRACT

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.


Subject(s)
Disease Progression , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Macular Degeneration , Humans , Male , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Aged , Macular Degeneration/genetics , Follow-Up Studies , Risk Factors , Middle Aged , Aged, 80 and over , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease
18.
Stem Cell Res ; 78: 103458, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870564

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters , Codon, Nonsense , Exons , Induced Pluripotent Stem Cells , Stargardt Disease , Induced Pluripotent Stem Cells/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Stargardt Disease/pathology , Humans , Homozygote , Cell Line , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/metabolism
19.
Cell Death Dis ; 15(6): 385, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824126

ABSTRACT

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.


Subject(s)
Lipid Droplets , Lipid Metabolism , Macular Degeneration , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Retinal Pigment Epithelium , Retinal Pigment Epithelium/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Humans , Mice , Lipid Droplets/metabolism , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Male , Oxidative Stress
20.
Niger Postgrad Med J ; 31(2): 93-101, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38826012

ABSTRACT

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.


Subject(s)
Macular Degeneration , Sirtuin 1 , Humans , Sirtuin 1/genetics , Macular Degeneration/genetics , Macular Degeneration/epidemiology , Genetic Predisposition to Disease , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL