Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 406
Filter
1.
Nat Commun ; 15(1): 3562, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670966

ABSTRACT

The diagnosis of inherited retinal degeneration (IRD) is challenging owing to its phenotypic and genotypic complexity. Clinical information is important before a genetic diagnosis is made. Metabolomics studies the entire picture of bioproducts, which are determined using genetic codes and biological reactions. We demonstrated that the common diagnoses of IRD, including retinitis pigmentosa (RP), cone-rod dystrophy (CRD), Stargardt disease (STGD), and Bietti's crystalline dystrophy (BCD), could be differentiated based on their metabolite heatmaps. Hundreds of metabolites were identified in the volcano plot compared with that of the control group in every IRD except BCD, considered as potential diagnosing markers. The phenotypes of CRD and STGD overlapped but could be differentiated by their metabolomic features with the assistance of a machine learning model with 100% accuracy. Moreover, EYS-, USH2A-associated, and other RP, sharing considerable similar characteristics in clinical findings, could also be diagnosed using the machine learning model with 85.7% accuracy. Further study would be needed to validate the results in an external dataset. By incorporating mass spectrometry and machine learning, a metabolomics-based diagnostic workflow for the clinical and molecular diagnoses of IRD was proposed in our study.


Subject(s)
Machine Learning , Metabolomics , Retinal Degeneration , Retinitis Pigmentosa , Stargardt Disease , Humans , Metabolomics/methods , Diagnosis, Differential , Retinal Degeneration/diagnosis , Retinal Degeneration/blood , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Male , Female , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/blood , Retinitis Pigmentosa/metabolism , Stargardt Disease/genetics , Adult , Middle Aged , Adolescent , Young Adult , Biomarkers/blood , Metabolome , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/blood , Cone-Rod Dystrophies/metabolism , Mass Spectrometry , Macular Degeneration/blood , Macular Degeneration/diagnosis , Macular Degeneration/genetics
2.
Cells ; 10(11)2021 11 12.
Article in English | MEDLINE | ID: mdl-34831363

ABSTRACT

To characterize metabolites and metabolic pathways altered in intermediate and neovascular age-related macular degeneration (IAMD and NVAMD), high resolution untargeted metabolomics was performed via liquid chromatography-mass spectrometry on plasma samples obtained from 91 IAMD patients, 100 NVAMD patients, and 195 controls. Plasma metabolite levels were compared between: AMD patients and controls, IAMD patients and controls, and NVAMD and IAMD patients. Partial least-squares discriminant analysis and linear regression were used to identify discriminatory metabolites. Pathway analysis was performed to determine metabolic pathways altered in AMD. Among the comparisons, we identified 435 unique discriminatory metabolic features. Using computational methods and tandem mass spectrometry, we identified 11 metabolic features whose molecular identities had been previously verified and confirmed the molecular identities of three additional discriminatory features. Included among the discriminatory metabolites were acylcarnitines, phospholipids, amino acids, and steroid metabolites. Pathway analysis revealed that lipid, amino acid, and vitamin metabolism pathways were altered in NVAMD, IAMD, or AMD in general, including the carnitine shuttle pathway which was significantly altered in all comparisons. Finally, few discriminatory features were identified between IAMD patients and controls, suggesting that plasma metabolic profiles of IAMD patients are more similar to controls than to NVAMD patients.


Subject(s)
Macular Degeneration/blood , Macular Degeneration/metabolism , Metabolomics , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/metabolism , Aged , Carnitine/analogs & derivatives , Carnitine/blood , Case-Control Studies , Discriminant Analysis , Female , Humans , Least-Squares Analysis , Linear Models , Lipids/blood , Male , Metabolic Networks and Pathways , Metabolome , Middle Aged
3.
Sci Rep ; 11(1): 19987, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620972

ABSTRACT

Pachychoroid neovasculopathy (PNV) is a new concept of macular disorder. Some cases diagnosed as age-related macular degeneration (AMD) have been re-diagnosed as PNV. However, the biological features of PNV are still uncertain. The purpose of this study was to compare PNV and AMD by analyses focusing on von Willebrand factor (VWF) and complement factor H (CFH). Ninety-seven patients who were previously diagnosed with treatment naïve AMD were enrolled in this study. They were re-classified as either PNV or AMD based on the clinical criteria and 33 patients were classified as PNV and 64 patients as AMD. We examined the clinical data, analyzed VWF multimer and two genetic polymorphisms (I62V and Y402H) in the CFH. PNV group was significantly younger than AMD group (P = 0.001). In both I62V and Y402H, there were no significant differences between PNV and AMD while the recessive homozygous (AA) was found only in PNV group in I62V. The presence of unusually large VWF multimers (UL-VWFMs) and subretinal hemorrhages were significantly higher in PNV than in AMD (P = 0.045, P = 0.020, respectively). Thus, the residual UL-VWFMs may result in platelet thrombosis and hemorrhages in the choriocapillaris of PNV. In conclusion, our results suggest the biological differences between PNV and AMD.


Subject(s)
Choroidal Neovascularization/genetics , Macular Degeneration/genetics , von Willebrand Factor/analysis , Aged , Aged, 80 and over , Choroidal Neovascularization/blood , Choroidal Neovascularization/pathology , Complement Factor H/analysis , Complement Factor H/genetics , Cross-Sectional Studies , Female , Humans , Japan , Macular Degeneration/blood , Macular Degeneration/pathology , Male , Polymorphism, Genetic , Retinal Hemorrhage , von Willebrand Factor/genetics
4.
Nutrients ; 13(6)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203817

ABSTRACT

Lutein and zeaxanthin may lower the risk of age-related macular degeneration (AMD). We evaluated the associations of plasma lutein and zeaxanthin with the incidence of advanced AMD in the Alienor study (Antioxydants Lipides Essentiels Nutrition et Maladies Oculaires). Alienor study is a prospective population-based cohort of 963 residents of Bordeaux, France, who were 73 years or older at baseline (2006-2008). The present study included 609 participants with complete ophthalmologic and plasma carotenoids data. Examinations were performed every two years over an eight-year period (2006 to 2017). Plasma lutein and zeaxanthin were determined at baseline from fasting blood samples using high-performance liquid chromatography. Cox proportional hazard models were used to assess associations between plasma lutein, zeaxanthin, and their (total cholesterol (TC) + triglycerides (TG)) ratios with AMD. Among the 609 included participants, 54 developed advanced incident AMD during a median follow-up time of 7.6 years (range 0.7 to 10.4). Participants with higher plasma lutein had a reduced risk for incident advanced AMD in the fully adjusted model (HR = 0.63 per 1-SD increase (95% CI, 0.41-0.97), p = 0.03). A similar association was observed using the lutein/(TC + TG) ratio (HR = 0.59 (95% CI, 0.39-0.90), p = 0.01). No associations were evidenced for other carotenoids. Higher plasma lutein was associated with a 37% reduced risk of incident advanced AMD.


Subject(s)
Biomarkers/blood , Lutein/blood , Macular Degeneration/blood , Macular Degeneration/epidemiology , Aged , Aged, 80 and over , Carotenoids/blood , Cholesterol/blood , Cohort Studies , Female , France , Humans , Incidence , Logistic Models , Male , Nutritional Status , Prospective Studies , Risk Factors , Triglycerides , Zeaxanthins/blood
5.
Am J Hum Genet ; 108(8): 1385-1400, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34260948

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of vision loss; there is strong genetic susceptibility at the complement factor H (CFH) locus. This locus encodes a series of complement regulators: factor H (FH), a splice variant factor-H-like 1 (FHL-1), and five factor-H-related proteins (FHR-1 to FHR-5), all involved in the regulation of complement factor C3b turnover. Little is known about how AMD-associated variants at this locus might influence FHL-1 and FHR protein concentrations. We have used a bespoke targeted mass-spectrometry assay to measure the circulating concentrations of all seven complement regulators and demonstrated elevated concentrations in 352 advanced AMD-affected individuals for all FHR proteins (FHR-1, p = 2.4 × 10-10; FHR-2, p = 6.0 × 10-10; FHR-3, p = 1.5 × 10-5; FHR-4, p = 1.3 × 10-3; FHR-5, p = 1.9 × 10-4) and FHL-1 (p = 4.9 × 10-4) when these individuals were compared to 252 controls, whereas no difference was seen for FH (p = 0.94). Genome-wide association analyses in controls revealed genome-wide-significant signals at the CFH locus for all five FHR proteins, and univariate Mendelian-randomization analyses strongly supported the association of FHR-1, FHR-2, FHR-4, and FHR-5 with AMD susceptibility. These findings provide a strong biochemical explanation for how genetically driven alterations in circulating FHR proteins could be major drivers of AMD and highlight the need for research into FHR protein modulation as a viable therapeutic avenue for AMD.


Subject(s)
Complement C3b Inactivator Proteins/metabolism , Complement Factor H/genetics , Genetic Predisposition to Disease , Macular Degeneration/blood , Polymorphism, Single Nucleotide , Aged , Case-Control Studies , Complement C3b Inactivator Proteins/genetics , Female , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Male , Risk Factors
6.
Curr Neurovasc Res ; 18(2): 181-188, 2021.
Article in English | MEDLINE | ID: mdl-34060991

ABSTRACT

BACKGROUND: Many factors including genetic and environmental are responsible for the incidence of Age-related Macular Degeneration (AMD). However, its pathogenesis has not been clearly elucidated yet. OBJECTIVE: This study aimed to estimate the Age-Related Maculopathy Susceptibility 2 (ARMS2), Collagen type VIII Alpha 1 chain (COL8A1), Rad 51 paralog(RAD51B), and Vascular Endothelial Growth Factor (VEGF) protein levels in serum of AMD and control participants and to further investigate their correlation to understand AMD pathogenesis. METHODS: For this case-control study, 31 healthy control and 57 AMD patients were recruited from Advanced Eye Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India. A blood sample was taken and serum was isolated from it. ELISA (enzyme-linked immunosorbent assay) was used for the estimation of proteins in the serum of patients. RESULTS: ARMS2 and COL8A1 levels were significantly elevated in the AMD group than in the control group. The highest levels of ARMS2, COL8A1, and VEGF proteins were recorded for the wet AMD sub-group. The study results endorsed significant positive correlation between these following molecules; ARMS2 and COL8A1 (r = 0.933, p < 0.0001), ARMS2 and RAD51B (r = 0.704, p < 0.0001), ARMS2 and VEGF (r = 0.925, p < 0.0001), COL8A1 and RAD51B (r = 0.736, p < 0.0001), COL8A1 and VEGF (r = 0.879, p < 0.0001), and RAD51B and VEGF (r = 0.691, p < 0.0001). CONCLUSION: The ARMS2 and COL8A1 levels were significantly higher and RAD51B was significantly lower in the AMD group than controls. Also, a significant statistical correlation was detected between these molecules, indicating that their interaction may be involved in the pathogenesis of AMD.


Subject(s)
Collagen Type VIII/blood , DNA-Binding Proteins/blood , Macular Degeneration/blood , Proteins/metabolism , Vascular Endothelial Growth Factor A/blood , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged
7.
Aging (Albany NY) ; 13(10): 13968-14000, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33946050

ABSTRACT

Wet age-related macular degeneration (wAMD) causes central vision loss and represents a major health problem in elderly people. Here we have used untargeted metabolomics using UHPLC-MS to profile plasma from 127 patients with wAMD (67 choroidal neovascularization (CNV) and 60 polypoidal choroidal vasculopathy (PCV)) and 50 controls. A total of 545 biochemicals were detected. Among them, 17 metabolites presented difference between patients with wAMD and controls. Most of them were oxidized lipids (N=6, 35.29%). Comparing to controls, 28 and 18 differential metabolites were identified in patients with CNV and PCV, respectively. Two metabolites, hyodeoxycholic acid and L-tryptophanamide, were differently distributed between PCV and CNV. We first investigated the genetic association with metabolites in wet AMD (CFH rs800292 and HTRA1 rs10490924). We identified six differential metabolites between the GG and AA genotypes of CFH rs800292, five differential metabolites between the GG and AA genotypes of HTRA1 rs10490924, and four differential metabolites between the GG and GA genotypes of rs10490924. We selected four metabolites (cyclamic acid, hyodeoxycholic acid, L-tryptophanamide and O-phosphorylethanolamine) for in vitro experiments. Among them, cyclamic acid reduced the activity, inhibited the proliferation, increased the apoptosis and necrosis in human retinal pigment epithelial cells (HRPECs). L-tryptophanamide affected the proliferation, apoptosis and necrosis in HRPECs, and promoted the tube formation and migration in primary human retinal endothelial cells (HRECs). Hyodeoxycholic acid and O-phosphorylethanolamine inhibited the tube formation and migration in HRECs. The results suggested that differential metabolites have certain effects on wAMD pathogenesis-related HRPECs and HRECs.


Subject(s)
Biomarkers/blood , Macular Degeneration/blood , Macular Degeneration/metabolism , Metabolomics , Apoptosis , Bacteria/metabolism , Cell Proliferation , Choroidal Neovascularization/metabolism , Epithelial Cells/metabolism , Genetic Predisposition to Disease , Humans , Macular Degeneration/genetics , Metabolome , Molecular Sequence Annotation , Necrosis , Neovascularization, Physiologic , Polymorphism, Single Nucleotide/genetics , Principal Component Analysis , Retinal Pigment Epithelium/pathology , Signal Transduction
8.
Nutrients ; 13(4)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920232

ABSTRACT

The purpose of this study is evaluate the efficacy and safety of medicinal products containing the original Age-Related Eye Disease group (AREDS) formulation at doses approved in Europe (EU, control group; n = 59) with a product that adds DHA, lutein, zeaxanthin, resveratrol and hydroxytyrosol to the formula (intervention group; n = 50). This was a multicenter, randomized, observer-blinded trial conducted in patients aged 50 years or older diagnosed with unilateral exudative Age related Macular Degeneration AMD. At month 12, the intervention did not have a significant differential effect on visual acuity compared with the control group, with an estimated treatment difference in Early Treatment Diabetic Retinopathy Study (ETDRS) of -1.63 (95% CI -0.83 to 4.09; p = 0.192). The intervention exhibited a significant and, in most cases, relevant effect in terms of a reduction in some inflammatory cytokines and a greater improvement in the fatty acid profile and serum lutein and zeaxantin concentration. In patients with unilateral wet AMD, the addition of lutein, zeaxanthin, resveratrol, hydroxytyrosol and DHA to the AREDS EU recommended doses in the short-term did not have a differential effect on visual acuity compared to a standard AREDS EU formula but, in addition to improving the fatty acid profile and increasing carotenoid serum levels, may provide a beneficial effect in improving the proinflammatory and proangiogenic profile of patients with AMD.


Subject(s)
Dietary Supplements/adverse effects , Macular Degeneration/diet therapy , Nutrients/administration & dosage , Aged , Aged, 80 and over , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/adverse effects , Female , Humans , Lutein/administration & dosage , Lutein/adverse effects , Macular Degeneration/blood , Macular Degeneration/diagnosis , Male , Middle Aged , Nutrients/adverse effects , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/adverse effects , Phenylethyl Alcohol/analogs & derivatives , Resveratrol/administration & dosage , Resveratrol/adverse effects , Treatment Outcome , Visual Acuity , Xanthophylls/administration & dosage , Zeaxanthins/administration & dosage , Zeaxanthins/adverse effects
9.
Sci Rep ; 11(1): 8226, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859228

ABSTRACT

Age-related macular degeneration (AMD) is a highly prevalent degenerative disease and a leading cause of vision loss worldwide. Evidence for an inflammatory component in the development of AMD exists, yet the exact mechanisms remain unclear. Bisretinoid N-retinylidene-N-retinylethanolamine (A2E) in retinal pigmental epithelial (RPE) cells, and in extracellular deposits constitutes a hallmark of AMD, but its role in the pathology of AMD is elusive. Here, we tested the hypothesis that A2E is responsible for the heightened inflammatory activity in AMD. To this end, we measured ex vivo mRNA expression of the cytokines TNF-α, IL-6, and IL-10 in whole blood samples after stimulation with A2E in a clinical sample of 27 patients with neovascular AMD and 24 patients with geographic atrophy secondary to AMD. Patients' spouses (n = 30) were included as non-affected controls. After stimulation with A2E, no statistical differences were found in the median expression level of TNF-α, IL-6, IL-10 between the control group, and the neovascular AMD and the geographic atrophy group. Our findings do not support evidence for the hypothesis, that A2E per se contributes to heightened inflammatory activity in AMD.


Subject(s)
Blood Cells/drug effects , Cytokines/metabolism , Macular Degeneration/blood , Retinoids/pharmacology , Aged , Aged, 80 and over , Blood Cells/physiology , Case-Control Studies , Female , Geographic Atrophy/blood , Geographic Atrophy/drug therapy , Geographic Atrophy/metabolism , Geographic Atrophy/pathology , Humans , In Vitro Techniques , Inflammation Mediators/metabolism , Lipopolysaccharides , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Retinoids/therapeutic use
10.
Hum Mol Genet ; 30(13): 1188-1199, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33783477

ABSTRACT

Age-related macular degeneration (AMD) is a complex neurodegenerative eye disease with behavioral and genetic etiology and is the leading cause of irreversible vision loss among elderly Caucasians. Functionally significant genetic variants in the alternative pathway of complement have been strongly linked to disease. More recently, a rare variant in the terminal pathway of complement has been associated with increased risk, Complement component 9 (C9) P167S. To assess the functional consequence of this variant, C9 levels were measured in two independent cohorts of AMD patients. In both cohorts, it was demonstrated that the P167S variant was associated with low C9 plasma levels. Further analysis showed that patients with advanced AMD had elevated sC5b-9 compared to those with non-advanced AMD, although this was not associated with the P167S polymorphism. Electron microscopy of membrane attack complexes (MACs) generated using recombinantly produced wild type or P167S C9 demonstrated identical MAC ring structures. In functional assays, the P167S variant displayed a higher propensity to polymerize and a small increase in its ability to induce hemolysis of sheep erythrocytes when added to C9-depleted serum. The demonstration that this C9 P167S AMD risk polymorphism displays increased polymerization and functional activity provides a rationale for the gene therapy trials of sCD59 to inhibit the terminal pathway of complement in AMD that are underway.


Subject(s)
Complement C9/genetics , Genetic Predisposition to Disease/genetics , Macular Degeneration/genetics , Mutation , Aged , Animals , CHO Cells , Case-Control Studies , Cohort Studies , Complement C9/metabolism , Complement Membrane Attack Complex/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , Cricetinae , Cricetulus , Female , Guinea Pigs , Hemolysis , Humans , Macular Degeneration/blood , Macular Degeneration/metabolism , Male , Polymerization , Risk Factors , Sheep
11.
J Clin Endocrinol Metab ; 106(7): e2749-e2758, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33596319

ABSTRACT

CONTEXT: To enable prevention and treatment of age-related macular degeneration (AMD), understanding risk factors for AMD is important. OBJECTIVE: We tested the hypotheses that elevated plasma apolipoprotein A1 and high-density lipoprotein (HDL) cholesterol and low levels of low-density lipoprotein (LDL) cholesterol are associated with increased risk of AMD. METHODS: From the Danish general population, we studied 106 703 and 16 032 individuals in the Copenhagen General Population Study (CGPS) and the Copenhagen City Heart Study (CCHS) with median follow-up of 9 and 32 years, respectively.The main outcome measures were 1787 AMD in CGPS and 206 in CCHS. RESULTS: Higher concentrations of plasma apolipoprotein A1 and HDL cholesterol, and lower concentrations of LDL cholesterol, were associated with higher risk of AMD in CGPS. After multifactorial adjustment, individuals in the highest versus lowest quartile of plasma apolipoprotein A1 and HDL cholesterol had hazard ratios for AMD of 1.40 (95% CI: 1.20-1.63) and 1.22 (1.03-1.45). Corresponding hazard ratios for individuals in the lowest versus highest quartile of LDL cholesterol were 1.18 (1.02-1.37). Per 100 mg/dL higher plasma apolipoprotein A1, 1 mmol/L (39 mg/dL) higher HDL, and 1 mmol/L (39 mmol/L) lower LDL cholesterol, the hazard ratios for AMD were 1.53(1.31-1.80), 1.19 (1.07-1.32), and 1.05 (1.00-1.11), respectively, with similar results across strata of different risk factors. Higher concentrations of HDL cholesterol were also associated with higher risk of AMD in the CCHS. CONCLUSION: Elevated plasma apolipoprotein A1 and HDL cholesterol and lower LDL cholesterol are associated with increased risk of AMD.


Subject(s)
Apolipoprotein A-I/blood , Cholesterol, HDL/blood , Macular Degeneration/blood , Aged , Aged, 80 and over , Denmark/epidemiology , Female , Humans , Macular Degeneration/epidemiology , Male , Middle Aged , Proportional Hazards Models , Risk Factors
12.
Acta Ophthalmol ; 99(7): e1063-e1069, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33533136

ABSTRACT

PURPOSE: To examine whether serum cholesterol in early middle age is associated with age-related macular degeneration (AMD) later in life. METHODS: A group of Helsinki Businessmen Study (HBS) participants (n = 209) were recruited for the study. Total cholesterol (TC), triglyceride and body mass index (BMI) were measured at the HBS baseline visit in 1964-1973. Lipid subfractions, BMI, smoking status and statin use were recorded in 2011 and fundus photographs graded for AMD in 2005-2012. The subjects were genotyped for the main AMD risk single nucleotide polymorphisms (SNPs). RESULTS: TC measured at baseline 1964-1973 was significantly higher in subjects later developing intermediate or late AMD (6.67 mmol/l versus 6.20 mmol/l, p = 0.024) or with drusen size of ≥125 µm (6.68 mmol/l versus 6.21 mmol/l, p = 0.030) compared with the rest of the study population. TC, LDL and TG values at follow-up 2011 were lower in subjects with AMD compared to those without, whereas HDL levels showed no difference. In multivariate analysis, baseline TC associated with intermediate or late AMD (OR 1.59, p = 0.004) and drusen size ≥ 125 µm (OR 1.57, p = 0.006) when corrected for age, BMI, AMD risk SNPs and smoking. Lipid values measured 2011 had no associations after correction. CONCLUSIONS: High systemic total cholesterol in early middle age may have a role in the initial development of AMD, especially in patients later developing large drusen.


Subject(s)
Cholesterol/blood , Macular Degeneration/blood , Polymorphism, Single Nucleotide , Risk Assessment/methods , Adult , Age Distribution , Age Factors , Aged , Aged, 80 and over , Biomarkers/blood , Finland/epidemiology , Follow-Up Studies , Humans , Incidence , Macular Degeneration/epidemiology , Macular Degeneration/genetics , Male , Middle Aged , Retrospective Studies , Risk Factors , Time Factors
13.
PLoS One ; 16(1): e0246114, 2021.
Article in English | MEDLINE | ID: mdl-33513185

ABSTRACT

PURPOSE: Mitochondrial (mt) DNA damage is associated with age-related macular degeneration (AMD) and other human aging diseases. This study was designed to quantify and characterize mtDNA low-frequency heteroplasmy single nucleotide polymorphisms (SNPs) of three different tissues isolated from AMD subjects using Next Generation Sequencing (NGS) technology. METHODS: DNA was extracted from neural retina, [RPE+choroid] and blood from three deceased age-related macular degeneration (AMD) subjects. Entire mitochondrial genomes were analyzed for low-frequency heteroplasmy SNPs using NGS technology that independently sequenced both mtDNA strands. This deep sequencing method (average sequencing depth of 30,000; range 1,000-100,000) can accurately differentiate low-frequency heteroplasmy SNPs from DNA modification artifacts. Twenty-three 'hot-spot' heteroplasmy mtDNA SNPs were analyzed in 222 additional blood samples. RESULTS: Germline homoplasmy SNPs that defined mtDNA haplogroups were consistent in the three tissues of each subject. Analyses of SNPs with <40% heteroplasmy revealed the blood had significantly greater numbers of heteroplasmy SNPs than retina alone (p≤0.05) or retina+choroid combined (p = 0.008). Twenty-three 'hot-spot' mtDNA heteroplasmy SNPs were present, with three being non-synonymous (amino acid change). Four 'hot-spot' heteroplasmy SNPs (m.1120C>T, m.1284T>C, m.1556C>T, m.7256C>T) were found in additional samples (n = 222). Five heteroplasmy SNPs (m.4104A>G, m.5320C>T, m.5471G>A, m.5474A>G, m.5498A>G) declined with age. Two heteroplasmy SNPs (m.13095T>C, m.13105A>G) increased in AMD compared to Normal samples. In the heteroplasmy SNPs, very few transversion mutations (purine to pyrimidine or vice versa, associated with oxidative damage) were found and the majority were transition changes (purine to purine or pyrimidine to pyrimidine, associated with replication errors). CONCLUSION: Within an individual, the blood, retina and [RPE+choroid] contained identical homoplasmy SNPs representing inherited germline mtDNA haplogroup. NGS methodology showed significantly more mtDNA heteroplasmy SNPs in blood compared to retina and [RPE+choroid], suggesting the latter tissues have substantial protection. Significantly higher heteroplasmy levels of m.13095T>C and m.13105A>G may represent potential AMD biomarkers. Finally, high levels of transition mutations suggest that accumulation of heteroplasmic SNPs may occur through replication errors rather than oxidative damage.


Subject(s)
DNA, Mitochondrial/genetics , Heteroplasmy , Macular Degeneration/genetics , Mitochondria/genetics , Polymorphism, Single Nucleotide , Aged, 80 and over , Choroid/metabolism , Choroid/pathology , DNA, Mitochondrial/blood , Female , Humans , Macular Degeneration/blood , Male , Mitochondria/metabolism , Retina/metabolism , Retina/pathology
14.
Front Immunol ; 12: 789897, 2021.
Article in English | MEDLINE | ID: mdl-35069568

ABSTRACT

Complement factor I (FI) is a central inhibitor of the complement system, and impaired FI function increases complement activation, contributing to diseases such as age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome (aHUS). Genetic variation in complement factor I (CFI) has been identified in both AMD and aHUS, with more than half of these variants leading to reduced FI secretion levels. For many of the variants with normal FI secretion, however, functional implications are not yet known. Here we studied 11 rare missense variants, with FI secretion levels comparable to wildtype, but a predicted damaging effects based on the Combined Annotation Dependent Depletion (CADD) score. Three variants (p.Pro50Ala, p.Arg339Gln, and p.Ser570Thr) were analyzed in plasma and serum samples of carriers affected by AMD. All 11 variants (nine for the first time in this study) were recombinantly expressed and the ability to degrade C3b was studied with the C3b degradation assay. The amount of degradation was determined by measuring the degradation product iC3b with ELISA. Eight of 11 (73%) mutant proteins (p.Pro50Ala, p.Arg339Gln, p.Ile340Thr, p.Gly342Glu, p.Gly349Arg, p.Arg474Gln, p.Gly487Cys, and p.Gly512Ser) showed significantly impaired C3b degradation, and were therefore classified as likely pathogenic. Our data indicate that genetic variants in CFI with a CADD score >20 are likely to affect FI function, and that monitoring iC3b in a degradation assay is a useful tool to establish the pathogenicity of CFI variants in functional studies.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Complement Factor I , Macular Degeneration , Mutation, Missense , Amino Acid Substitution , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Complement C3b/immunology , Complement C3b/metabolism , Complement Factor I/genetics , Complement Factor I/immunology , Complement Factor I/metabolism , Female , Humans , Macular Degeneration/blood , Macular Degeneration/genetics , Macular Degeneration/immunology , Male
15.
PLoS One ; 15(8): e0236283, 2020.
Article in English | MEDLINE | ID: mdl-32764794

ABSTRACT

Geographic atrophy (GA) is an advanced form of dry age-related macular degeneration (AMD), in which local inflammation and hyperactivity of the complement pathway have been implicated in its pathophysiology. This study explores whether any surrogate biomarkers are specifically associated with GA. Plasma from subjects with GA, intermediate dry AMD and non-AMD control were evaluated in 2 cohorts. Cohort 1 was assayed in a 320-analyte Luminex library. Statistical analysis was performed using non-parametric and parametric methods (Kruskal-Wallis, principal component analysis, partial least squares and multivariate analysis of variance (MANOVA) and univariate ANCOVAs). Bioinformatic analysis was conducted and identified connections to the amyloid pathway. Statistically significant biomarkers identified in Cohort 1 were then re-evaluated in Cohort 2 using individual ELISA and multiplexing. Of 320 analytes in Cohort 1, 273 were rendered measurable, of which 56 were identified as changing. Among these markers, 40 were identified in univariate ANCOVAs. Serum amyloid precursor protein (sAPP) was analyzed by a separate ELISA and included in further analyses. The 40 biomarkers, sAPP and amyloid-ß (Aß) (1-42) (included for comparison) were evaluated in Cohort 2. This resulted in 11 statistically significant biomarkers, including sAPP and Aß(1-40), but not Aß(1-42). Other biomarkers identified included serum proteases- tissue plasminogen activator, tumor-associated trypsinogen inhibitor, matrix metalloproteinases 7 and 9, and non-proteases- insulin-like growth factor binding protein 6, AXL receptor tyrosine kinase, omentin, pentraxin-3 and osteopontin. Findings suggest that there is a preferential processing of APP to Aß(1-40) over Aß(1-42), and a potential role for the carboxylase activity of the γ-secretase protein, which preferentially splices sAPPß to Aß(1-40). Other markers are associated with the breakdown and remodeling of the extracellular matrix, and loss of homeostasis, possibly within the photoreceptor-retinal pigment epithelium-choriocapillaris complex. These data suggest novel disease pathways associated with GA pathogenesis and could provide potential novel targets for treatment of GA.


Subject(s)
Amyloid beta-Peptides/metabolism , Geographic Atrophy/blood , Macular Degeneration/complications , Retinal Pigment Epithelium/pathology , Aged , Aged, 80 and over , Biomarkers/blood , Biomarkers/metabolism , Cohort Studies , Computational Biology , Female , Fundus Oculi , Geographic Atrophy/diagnosis , Geographic Atrophy/etiology , Geographic Atrophy/pathology , Humans , Macular Degeneration/blood , Macular Degeneration/pathology , Male , Middle Aged , Optical Imaging , Signal Transduction , Tissue Plasminogen Activator
16.
Lipids ; 55(6): 693-706, 2020 11.
Article in English | MEDLINE | ID: mdl-32602621

ABSTRACT

Oxidized polyunsaturated fatty acids (PUFA) are associated to pathogenesis of diseases including cardiovascular and neurodegeneration. The novel products are not only biomarkers but also lipid mediators in gene regulation and signaling pathways. Herein, simultaneous quantitation of 28 products derived from nonenzymatic and enzymatic oxidation of PUFA i.e. 5-, 15-F2t -isoprostanes, 7-, 17-F2t -dihomo-isoprostanes, 7-, 17-F2t -dihomo-isofurans, 5-, 8-, 18-F3t -isoprostanes, 4-, 10-, 13-, 14-, 20-F4t -neuroprostanes, 5-, 8-, 9-, 11-,12-, 15-, 20-HETE, 4-, 7-, 11-, 14-, 17-HDHA, RvE1, and NPD1 using LC-(ESI)-QTOF-MS/MS was developed. These products were measurable in a single sample and the analytical time was relative short (~15 min). Furthermore, we showed that the use of internal standards is a requisite to normalize matrix effects and preparation loss for the quantitation. Validation assays indicated the method to be robust for plasma and mid-stream urine sample analysis in particular from those of age-related macular degeneration subjects, where the accuracy of quantitation displayed good repeatability.


Subject(s)
Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/urine , Macular Degeneration/metabolism , Tandem Mass Spectrometry/methods , Blood Chemical Analysis/methods , Chromatography, Liquid , Fatty Acids, Unsaturated/metabolism , Humans , Isoprostanes/blood , Isoprostanes/metabolism , Isoprostanes/urine , Limit of Detection , Macular Degeneration/blood , Macular Degeneration/urine , Neuroprostanes/blood , Neuroprostanes/metabolism , Neuroprostanes/urine , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization , Urinalysis/methods
17.
Hum Mol Genet ; 29(14): 2313-2324, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32510551

ABSTRACT

Factor I (FI) is one of the main inhibitors of complement activity, and numerous rare coding variants have been reported in patients with age-related macular degeneration, atypical hemolytic uremic syndrome and C3 glomerulopathy. Since many of these variants are of unknown clinical significance, this study aimed to determine the effect of rare coding variants in the complement factor I (CFI) gene on FI expression. We measured FI levels in plasma samples of carriers of rare coding variants and in vitro in the supernatants of epithelial cells expressing recombinant FI. FI levels were measured in 177 plasma samples of 155 individuals, carrying 24 different rare coding variants in CFI. In carriers of the variants p.Gly119Arg, p.Leu131Arg, p.Gly188Ala and c.772G>A (r.685_773del), significantly reduced FI plasma levels were detected. Furthermore, recombinant FI expression levels were determined for 126 rare coding variants. Of these variants 68 (54%) resulted in significantly reduced FI expression in supernatant compared to wildtype (WT). The recombinant protein expression levels correlated significantly with the FI level in plasma of carriers of CFI variants. In this study, we performed the most comprehensive FI expression level analysis of rare coding variants in CFI to date. More than half of CFI variants lead to reduced FI expression, which might impair complement regulation in vivo. Our study will aid the interpretation of rare coding CFI variants identified in clinical practice, which is in particular important in light of patient inclusion in ongoing clinical trials for CFI gene supplementation in AMD.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement Factor I/genetics , Fibrinogen/genetics , Macular Degeneration/genetics , Aged , Aged, 80 and over , Alleles , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/pathology , Female , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Genotype , Heterozygote , Humans , Macular Degeneration/blood , Macular Degeneration/pathology , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics
18.
Invest Ophthalmol Vis Sci ; 61(6): 18, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32516404

ABSTRACT

Purpose: Rare genetic variants in complement factor I (CFI) that cause low systemic levels of the protein (FI) have been reported as a strong risk factor for advanced age-related macular degeneration (AMD). This study set out to replicate these findings. Methods: FI levels were measured by sandwich ELISA in an independent cohort of 276 patients with AMD and 205 elderly controls. Single-nucleotide polymorphism genotyping and Sanger sequencing were used to assess genetic variability. Results: The median FI level was significantly lower in those individuals with AMD and a rare CFI variant (28.3 µg/mL) compared to those with AMD without a rare CFI variant (38.8 µg/mL, P = 0.004) or the control population with (41.7 µg/mL, P = 0.0085) or without (41.5 µg/mL, P < 0.0001) a rare CFI variant. Thirty-six percent of patients with AMD with a rare CFI variant had levels below the fifth percentile, compared to 6% in controls with CFI variants. Multiple regression analyses revealed a decreased FI level associated with a rare CFI variant was a risk factor for AMD (early or late AMD: odds ratio [OR] 12.05, P = 0.03; early AMD: OR 30.3, P = 0.02; late AMD: OR 10.64, P < 0.01). Additionally, measurement of FI in aqueous humor revealed a large FI concentration gradient between systemic circulation and the eye (∼286-fold). Conclusions: Rare genetic variants in CFI causing low systemic FI levels are strongly associated with AMD. The impermeability of the Bruch's membrane to FI will have implications for therapeutic replacement of FI in individuals with CFI variants and low FI levels at risk of AMD.


Subject(s)
Complement Factor I/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Macular Degeneration/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Biomarkers , Case-Control Studies , Complement Factor H/genetics , Complement Factor I/metabolism , Female , Genetic Testing , Genotyping Techniques , Humans , Macular Degeneration/blood , Male , Middle Aged , Proportional Hazards Models , Proteins/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Retrospective Studies , Risk Factors , Vascular Endothelial Growth Factor A/genetics
19.
Proc Natl Acad Sci U S A ; 117(18): 9952-9963, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32345717

ABSTRACT

Genetic polymorphisms in the region of the trimeric serine hydrolase high-temperature requirement 1 (HTRA1) are associated with increased risk of age-related macular degeneration (AMD) and disease progression, but the precise biological function of HtrA1 in the eye and its contribution to disease etiologies remain undefined. In this study, we have developed an HtrA1-blocking Fab fragment to test the therapeutic hypothesis that HtrA1 protease activity is involved in the progression of AMD. Next, we generated an activity-based small-molecule probe (ABP) to track target engagement in vivo. In addition, we used N-terminomic proteomic profiling in preclinical models to elucidate the in vivo repertoire of HtrA1-specific substrates, and identified substrates that can serve as robust pharmacodynamic biomarkers of HtrA1 activity. One of these HtrA1 substrates, Dickkopf-related protein 3 (DKK3), was successfully used as a biomarker to demonstrate the inhibition of HtrA1 activity in patients with AMD who were treated with the HtrA1-blocking Fab fragment. This pharmacodynamic biomarker provides important information on HtrA1 activity and pharmacological inhibition within the ocular compartment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antibodies, Anti-Idiotypic/pharmacology , Geographic Atrophy/drug therapy , High-Temperature Requirement A Serine Peptidase 1/genetics , Macular Degeneration/drug therapy , Adaptor Proteins, Signal Transducing/isolation & purification , Aged , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Biomarkers/blood , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Geographic Atrophy/blood , Geographic Atrophy/genetics , Geographic Atrophy/immunology , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Macular Degeneration/blood , Macular Degeneration/genetics , Macular Degeneration/immunology , Male , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Proteome/immunology , Rats , Retina/drug effects , Retina/immunology , Retina/pathology , Small Molecule Libraries/pharmacology
20.
Sci Rep ; 10(1): 5958, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32249850

ABSTRACT

This prospective randomized double-masked study investigated the effects of 20 mg lutein supplementation with two different capsules (beeswax or glycerol fatty acid esters) for 6 months on the fellow eyes of 39 Japanese patients with unilateral age-related macular degeneration, and assessed the factors associated with baseline plasma lutein concentration via lifestyle interviews. Macular pigment optical density (MPOD), determined with the two-wavelength autofluorescence method, increased over time in the beeswax group (ANOVA, p = 0.0451), although the increase from 3 months to 6 months was only marginally significant. No significant increase was observed in the glycerol fatty acid esters group (ANOVA, p = 0.7396). Plasma lutein concentrations significantly increased at 3 and 6 months from baseline in both groups (both p < 0.01). In a multiple regression model, age was negatively associated with higher plasma lutein concentration (p = 0.0305), while consumption of green vegetables was positively associated with baseline plasma lutein concentration (p = 0.0322). In conclusion, a significant increase in MPOD was not fully confirmed with 6 months intake duration despite a significant increase in plasma lutein concentrations. Consumption of green vegetable was confirmed to be associated with plasma lutein concentration after adjusting for other potential factors including age.


Subject(s)
Dietary Supplements , Lutein/administration & dosage , Lutein/blood , Macular Degeneration/physiopathology , Macular Pigment , Vegetables , Age Factors , Aged , Diet , Double-Blind Method , Female , Humans , Japan , Macular Degeneration/blood , Male , Middle Aged , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...