Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Invest Ophthalmol Vis Sci ; 64(15): 33, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38133503

ABSTRACT

Purpose: Genome editing is an emerging group of technologies with the potential to ameliorate dominant, monogenic human diseases such as late-onset retinal degeneration (L-ORD). The goal of this study was to identify disease stages and retinal locations optimal for evaluating the efficacy of a future genome editing trial. Methods: Twenty five L-ORD patients (age range, 33-77 years; median age, 59 years) harboring the founder variant S163R in C1QTNF5 were enrolled from three centers in the United Kingdom and United States. Patients were examined with widefield optical coherence tomography (OCT) and chromatic perimetry under dark-adapted and light-adapted conditions to derive phenomaps of retinal disease. Results were analyzed with a model of a shared natural history of a single delayed exponential across all subjects and all retinal locations. Results: Critical age for the initiation of photoreceptor loss ranged from 48 years at the temporal paramacular retina to 74 years at the inferior midperipheral retina. Subretinal deposits (sRET-Ds) became more prevalent as critical age was approached. Subretinal pigment epithelial deposits (sRPE-Ds) were detectable in the youngest patients showing no other structural or functional abnormalities at the retina. The sRPE-D thickness continuously increased, reaching 25 µm in the extrafoveal retina and 19 µm in the fovea at critical age. Loss of light sensitivity preceded shortening of outer segments and loss of photoreceptors by more than a decade. Conclusions: Retinal regions providing an ideal treatment window exist across all severity stages of L-ORD.


Subject(s)
Genetic Therapy , Retinal Degeneration , Humans , Adult , Middle Aged , Aged , Late Onset Disorders/genetics , Late Onset Disorders/pathology , Late Onset Disorders/therapy , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/therapy , Collagen/genetics , Male , Female , Fovea Centralis/pathology , Tomography, Optical Coherence , Genetic Therapy/methods , Gene Editing
2.
Prog Retin Eye Res ; 96: 101154, 2023 09.
Article in English | MEDLINE | ID: mdl-36513584

ABSTRACT

Despite comprehensive research efforts over the last decades, the pathomechanisms of age-related macular degeneration (AMD) remain far from being understood. Large-scale genome wide association studies (GWAS) were able to provide a defined set of genetic aberrations which contribute to disease risk, with the strongest contributors mapping to distinct regions on chromosome 1 and 10. While the chromosome 1 locus comprises factors of the complement system with well-known functions, the role of the 10q26-locus in AMD-pathophysiology remains enigmatic. 10q26 harbors a cluster of three functional genes, namely PLEKHA1, ARMS2 and HTRA1, with most of the AMD-associated genetic variants mapping to the latter two genes. High linkage disequilibrium between ARMS2 and HTRA1 has kept association studies from reliably defining the risk-causing gene for long and only very recently the genetic risk region has been narrowed to ARMS2, suggesting that this is the true AMD gene at this locus. However, genetic associations alone do not suffice to prove causality and one or more of the 14 SNPs on this haplotype may be involved in long-range control of gene expression, leaving HTRA1 and PLEKHA1 still suspects in the pathogenic pathway. Both, ARMS2 and HTRA1 have been linked to extracellular matrix homeostasis, yet their exact molecular function as well as their role in AMD pathogenesis remains to be uncovered. The transcriptional regulation of the 10q26 locus adds an additional level of complexity, given, that gene-regulatory as well as epigenetic alterations may influence expression levels from 10q26 in diseased individuals. Here, we provide a comprehensive overview on the 10q26 locus and its three gene products on various levels of biological complexity and discuss current and future research strategies to shed light on one of the remaining enigmatic spots in the AMD landscape.


Subject(s)
Macular Degeneration , Serine Endopeptidases , Humans , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Genome-Wide Association Study , Proteins/genetics , Proteins/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Gene Expression Regulation , Polymorphism, Single Nucleotide , Genotype , Genetic Predisposition to Disease
3.
BMC Ophthalmol ; 22(1): 140, 2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35346118

ABSTRACT

BACKGROUND: Usher Syndrome is the commonest cause of inherited blindness and deafness. The condition is clinically and genetically heterogeneous, with no current treatment. We report a case carrying novel biallelic variants in USH2A causing progressive early adolescent onset visual and hearing impairment consistent with Usher Syndrome Type IIA. CASE PRESENTATION: Our patient presented at age 13 with progressive visual field loss and hearing loss, associated with early onset of cataract in her 40s requiring lens extraction. Now 52 years old, latest best corrected visual acuity (BCVA) stands at Logmar Right Eye (RE) 0.8 and Left Eye (LE) 0.2, with significantly constricted visual fields bilaterally. She was registered partially sighted age 46. Clinical and molecular genetic assessment of the proband was consistent with a diagnosis of Usher Syndrome Type IIA. Genetic testing identified two novel USH2A variants, resulting in the premature termination codon p.Leu30Ter and a missense mutation p.Cys3251Tyr. Segregation analysis confirmed that these variants were biallelic in the affected case. Comprehensive in silico analysis confirmed that these mutations are the probable cause of Usher Syndrome Type IIA in this individual. CONCLUSIONS: The identification of novel mutations in USH2A increases the spectrum of genetic variations that lead to Usher Syndrome, aiding genetic diagnosis, assessment of patient prognosis, and emphasising the importance of genetic testing to identify new mutations in patients with undiagnosed progressive visual loss.


Subject(s)
Usher Syndromes , Adolescent , Codon, Nonsense , Extracellular Matrix Proteins/genetics , Female , Humans , Middle Aged , Mutation , Mutation, Missense , Usher Syndromes/diagnosis , Usher Syndromes/genetics
4.
Dis Model Mech ; 14(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34368841

ABSTRACT

Brittle cornea syndrome (BCS) is a rare recessive condition characterised by extreme thinning of the cornea and sclera. BCS results from loss-of-function mutations in the poorly understood genes ZNF469 or PRDM5. In order to determine the function of ZNF469 and to elucidate pathogenic mechanisms, we used genome editing to recapitulate a human ZNF469 BCS mutation in the orthologous mouse gene Zfp469. Ophthalmic phenotyping showed that homozygous Zfp469 mutation causes significant central and peripheral corneal thinning arising from reduced stromal thickness. Expression of key components of the corneal stroma in primary keratocytes from Zfp469BCS/BCS mice is affected, including decreased Col1a1 and Col1a2 expression. This alters the collagen type I/collagen type V ratio and results in collagen fibrils with smaller diameter and increased fibril density in homozygous mutant corneas, correlating with decreased biomechanical strength in the cornea. Cell-derived matrices generated by primary keratocytes show reduced deposition of collagen type I, offering an in vitro model for stromal dysfunction. Work remains to determine whether modulating ZNF469 activity will have therapeutic benefit in BCS or in conditions such as keratoconus in which the cornea thins progressively. This article has an associated First Person interview with the first author of the paper.


Subject(s)
DNA-Binding Proteins , Skin Abnormalities , Animals , Cornea , DNA-Binding Proteins/genetics , Eye Abnormalities , Humans , Joint Instability/congenital , Mice , Mutation/genetics , Skin Abnormalities/genetics , Transcription Factors/genetics , Zinc Fingers
5.
Sci Adv ; 6(8): eaax0301, 2020 02.
Article in English | MEDLINE | ID: mdl-32128391

ABSTRACT

Effector functions of immunoglobulin G (IgG) are regulated by the composition of a glycan moiety, thus affecting activity of the immune system. Aberrant glycosylation of IgG has been observed in many diseases, but little is understood about the underlying mechanisms. We performed a genome-wide association study of IgG N-glycosylation (N = 8090) and, using a data-driven network approach, suggested how associated loci form a functional network. We confirmed in vitro that knockdown of IKZF1 decreases the expression of fucosyltransferase FUT8, resulting in increased levels of fucosylated glycans, and suggest that RUNX1 and RUNX3, together with SMARCB1, regulate expression of glycosyltransferase MGAT3. We also show that variants affecting the expression of genes involved in the regulation of glycoenzymes colocalize with variants affecting risk for inflammatory diseases. This study provides new evidence that variation in key transcription factors coupled with regulatory variation in glycogenes modifies IgG glycosylation and has influence on inflammatory diseases.


Subject(s)
Gene Expression Regulation , Immunoglobulin G/metabolism , Inflammation/genetics , Inflammation/metabolism , Algorithms , Alleles , Computational Biology/methods , Genetic Loci , Genome-Wide Association Study , Glycosylation , Humans , Immunoglobulin G/immunology , Linkage Disequilibrium , Models, Genetic , Phenotype , Polymorphism, Single Nucleotide , Polysaccharides/metabolism
6.
Ophthalmic Genet ; 39(6): 763-770, 2018 12.
Article in English | MEDLINE | ID: mdl-30451557

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is a common sight threatening condition. However, there are a number of monogenic macular dystrophies that are clinically similar to AMD, which can potentially provide pathogenetic insights. METHODS: Three siblings from a non-consanguineous Greek-Cypriot family reported central visual disturbance and nyctalopia. The patients had full ophthalmic examinations and color fundus photography, spectral-domain ocular coherence tomography and scanning laser ophthalmoscopy. Targeted polymerase chain reaction (PCR) was performed as a first step to attempt to identify suspected mutations in C1QTNF5 and TIMP3 followed by whole genome sequencing. RESULTS: The three patients were noted to have symptoms of nyctalopia, early paracentral visual field loss and, in older patients, central vision loss. Imaging identified pseudodrusen, retinal atrophy and RPE-Bruch's membrane separation. Whole genome sequencing of the proband revealed two novel heterozygous variants in C1QTNF5, c.556C>T, and c.569C>G. The mutation segregated with disease in this family, occurred in cis, and resulted in missense amino acid changes P186S and S190W in C1QTNF5. In silico modeling of the variants revealed that the S190W mutations was likely to have the greatest pathologic effect and that the combination of the mutations was likely to have an additive effect. CONCLUSIONS: The novel mutations in C1QTNF5 identified here expand the genotypic spectrum of mutations causing late-onset retinal dystrophy.


Subject(s)
Collagen/genetics , Macular Degeneration/genetics , Mutation, Missense , Whole Genome Sequencing , Aged , DNA Mutational Analysis , Electroretinography , Female , Fluorescein Angiography , Genes, Dominant , Humans , Macular Degeneration/diagnosis , Male , Middle Aged , Pedigree , Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-3/genetics , Tomography, Optical Coherence , Visual Acuity
7.
Sci Rep ; 7(1): 12147, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28939808

ABSTRACT

Late-onset retinal degeneration (L-ORD) is a rare autosomal dominant retinal dystrophy, characterised by extensive sub-retinal pigment epithelium (RPE) deposits, RPE atrophy, choroidal neovascularisation and photoreceptor cell death associated with severe visual loss. L-ORD shows striking phenotypic similarities to age-related macular degeneration (AMD), a common and genetically complex disorder, which can lead to misdiagnosis in the early stages. To date, a single missense mutation (S163R) in the C1QTNF5 gene, encoding C1q And Tumor Necrosis Factor Related Protein 5 (C1QTNF5) has been shown to cause L-ORD in a subset of affected families. Here, we describe the identification and characterisation of three novel pathogenic mutations in C1QTNF5 in order to elucidate disease mechanisms. In silico and in vitro characterisation show that these mutations perturb protein folding, assembly or polarity of secretion of C1QTNF5 and, importantly, all appear to destabilise the wildtype protein in co-transfection experiments in a human RPE cell line. This suggests that the heterozygous mutations in L-ORD show a dominant negative, rather than a haploinsufficient, disease mechanism. The function of C1QTNF5 remains unclear but this new insight into the pathogenetic basis of L-ORD has implications for future therapeutic strategies such as gene augmentation therapy.


Subject(s)
Collagen/genetics , Mutation , Retinal Degeneration/genetics , Aged , Amino Acid Sequence , Cell Line , Collagen/chemistry , Collagen/metabolism , Female , Humans , Male , Middle Aged , Models, Molecular , Mutation, Missense , Pedigree , Protein Domains , Protein Folding , Retinal Degeneration/metabolism , Sequence Alignment
8.
Nat Genet ; 48(2): 134-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691988

ABSTRACT

Advanced age-related macular degeneration (AMD) is the leading cause of blindness in the elderly, with limited therapeutic options. Here we report on a study of >12 million variants, including 163,714 directly genotyped, mostly rare, protein-altering variants. Analyzing 16,144 patients and 17,832 controls, we identify 52 independently associated common and rare variants (P < 5 × 10(-8)) distributed across 34 loci. Although wet and dry AMD subtypes exhibit predominantly shared genetics, we identify the first genetic association signal specific to wet AMD, near MMP9 (difference P value = 4.1 × 10(-10)). Very rare coding variants (frequency <0.1%) in CFH, CFI and TIMP3 suggest causal roles for these genes, as does a splice variant in SLC16A8. Our results support the hypothesis that rare coding variants can pinpoint causal genes within known genetic loci and illustrate that applying the approach systematically to detect new loci requires extremely large sample sizes.


Subject(s)
Genome-Wide Association Study , Macular Degeneration/genetics , Genetic Predisposition to Disease , Humans , Mutation
9.
Adv Exp Med Biol ; 801: 251-7, 2014.
Article in English | MEDLINE | ID: mdl-24664705

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness worldwide, affecting an estimated 50 million individuals aged over 65 years.Environmental and genetic risk-factors implicate chronic inflammation in the etiology of AMD, contributing to the formation of drusen, retinal pigment epithelial cell dysfunction and photoreceptor cell death. Consistent with a role for chronic inflammation in AMD pathogenesis, several inflammatory mediators, including complement components, chemokines and cytokines, are elevated at both the local and systemic levels in AMD patients. These mediators have diverse roles in the alternative complement pathway, including recruitment of inflammatory cells, activation of the inflammasome, promotion of neovascularisation and in the resolution of inflammation. The utility of inflammatory biomarkers in assessing individual risk and progression of the disease is controversial. However, understanding the role of these inflammatory mediators in AMD onset, progression and response to treatment may increase our knowledge of disease pathogenesis and provide novel therapeutic options in the future.


Subject(s)
Macular Degeneration/immunology , Macular Degeneration/pathology , Retinitis/immunology , Retinitis/pathology , Biomarkers/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans
10.
Hum Mol Genet ; 22(23): 4857-69, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23873044

ABSTRACT

It is a longstanding puzzle why non-coding variants in the complement factor H (CFH) gene are more strongly associated with age-related macular degeneration (AMD) than functional coding variants that directly influence the alternative complement pathway. The situation is complicated by tight genetic associations across the region, including the adjacent CFH-related genes CFHR3 and CFHR1, which may themselves influence the alternative complement pathway and are contained within a common deletion (CNP147) which is associated with protection against AMD. It is unclear whether this association is mediated through a protective effect of low plasma CFHR1 concentrations, high plasma CFH or both. We examined the triangular relationships of CFH/CFHR3/CFHR1 genotype, plasma CFH or CFHR1 concentrations and AMD susceptibility in combined case-control (1256 cases, 1020 controls) and cross-sectional population (n = 1004) studies and carried out genome-wide association studies of plasma CFH and CFHR1 concentrations. A non-coding CFH SNP (rs6677604) and the CNP147 deletion were strongly correlated both with each other and with plasma CFH and CFHR1 concentrations. The plasma CFH-raising rs6677604 allele and raised plasma CFH concentration were each associated with AMD protection. In contrast, the protective association of the CNP147 deletion with AMD was not mediated by low plasma CFHR1, since AMD-free controls showed increased plasma CFHR1 compared with cases, but it may be mediated by the association of CNP147 with raised plasma CFH concentration. The results are most consistent with a regulatory locus within a 32 kb region of the CFH gene, with a major effect on plasma CFH concentration and AMD susceptibility.


Subject(s)
Blood Proteins/genetics , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Alleles , Blood Proteins/metabolism , Case-Control Studies , Complement Factor H/genetics , Cross-Sectional Studies , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genotype , Humans , Introns , Macular Degeneration/immunology , Polymorphism, Single Nucleotide , Sequence Deletion
12.
Immunobiology ; 217(2): 158-61, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22024702

ABSTRACT

OBJECTIVES: Age-related macular degeneration (AMD) is the commonest cause of blindness in Western populations. Risk is influenced by age, genetic and environmental factors. Complement activation appears to be important in the pathogenesis and associations have been found between AMD and genetic variations in complement regulators such as complement factor H. We therefore investigated other complement regulators for association with AMD. METHODS: We carried out a case-control study to test for association between AMD and single nucleotide polymorphisms (SNPs) spanning the genes encoding complement factor P (CFP, properdin), CD46 (membrane cofactor protein, MCP), CD55 (decay accelerating factor, DAF) and CD59 (protectin). All cases and controls were examined by an ophthalmologist and had independent grading of fundus photographs to confirm their disease status. RESULTS: 20 SNPs were genotyped in 446 cases and 262 controls. For two SNPs with p-values approaching significance additional subjects were genotyped to increase the numbers to 622 cases and 359 controls. There was no evidence of association between AMD and any of the SNPs typed in CFP, CD46, CD55 or CD59. CONCLUSIONS: In a case-control sample that has shown the well established associations between AMD and variants in CFH, CFB and C3 there was absence of association with SNPs in CFP, CD46, CD55 and CD59. This suggests that these are not important susceptibility genes for AMD.


Subject(s)
Complement System Proteins/genetics , Genetic Predisposition to Disease , Macular Degeneration/genetics , Aged , Aged, 80 and over , Aging , CD55 Antigens/genetics , CD59 Antigens/genetics , Case-Control Studies , Female , Genetic Association Studies , Genetic Variation , Genotype , Humans , Macular Degeneration/pathology , Male , Membrane Cofactor Protein/genetics , Polymorphism, Single Nucleotide/genetics , Properdin/genetics
13.
Invest Ophthalmol Vis Sci ; 52(12): 8828-34, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22003108

ABSTRACT

PURPOSE: To examine the role of complement factor D (CFD) in age-related macular degeneration (AMD) by analysis of genetic association, copy number variation, and plasma CFD concentrations. METHODS: Single nucleotide polymorphisms (SNPs) in the CFD gene were genotyped and the results analyzed by binary logistic regression. CFD gene copy number was analyzed by gene copy number assay. Plasma CFD was measured by an enzyme-linked immunosorbent assay. RESULTS: Genetic association was found between CFD gene SNP rs3826945 and AMD (odds ratio 1.44; P = 0.028) in a small discovery case-control series (462 cases and 325 controls) and replicated in a combined cohorts meta-analysis of 4765 cases and 2693 controls, with an odds ratio of 1.11 (P = 0.032), with the association almost confined to females. Copy number variation in the CFD gene was identified in 13 out of 640 samples examined but there was no difference in frequency between AMD cases (1.3%) and controls (2.7%). Plasma CFD concentration was measured in 751 AMD cases and 474 controls and found to be elevated in AMD cases (P = 0.00025). The odds ratio for those in the highest versus lowest quartile for plasma CFD was 1.81. The difference in plasma CFD was again almost confined to females. CONCLUSIONS: CFD regulates activation of the alternative complement pathway, which is implicated in AMD pathogenesis. The authors found evidence for genetic association between a CFD gene SNP and AMD and a significant increase in plasma CFD concentration in AMD cases compared with controls, consistent with a role for CFD in AMD pathogenesis.


Subject(s)
Complement Pathway, Alternative/physiology , Macular Degeneration/genetics , Polymorphism, Single Nucleotide/genetics , Aged , Aged, 80 and over , Cohort Studies , Complement Factor D/genetics , Complement Factor D/metabolism , Female , Gene Dosage/genetics , Genotype , Humans , Macular Degeneration/blood , Male , Middle Aged
14.
PLoS One ; 5(1): e8764, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20098734

ABSTRACT

BACKGROUND: The insulin-degrading enzyme gene (IDE) is a strong functional and positional candidate for late onset Alzheimer's disease (LOAD). METHODOLOGY/PRINCIPAL FINDINGS: We examined conserved regions of IDE and its 10 kb flanks in 269 AD cases and 252 controls thereby identifying 17 putative functional polymorphisms. These variants formed eleven haplotypes that were tagged with ten variants. Four of these showed significant association with IDE transcript levels in samples from 194 LOAD cerebella. The strongest, rs6583817, which has not previously been reported, showed unequivocal association (p = 1.5x10(-8), fold-increase = 2.12,); the eleven haplotypes were also significantly associated with transcript levels (global p = 0.003). Using an in vitro dual luciferase reporter assay, we found that rs6583817 increases reporter gene expression in Be(2)-C (p = 0.006) and HepG2 (p = 0.02) cell lines. Furthermore, using data from a recent genome-wide association study of two Croatian isolated populations (n = 1,879), we identified a proxy for rs6583817 that associated significantly with decreased plasma Abeta40 levels (ss = -0.124, p = 0.011) and total measured plasma Abeta levels (b = -0.130, p = 0.009). Finally, rs6583817 was associated with decreased risk of LOAD in 3,891 AD cases and 3,605 controls. (OR = 0.87, p = 0.03), and the eleven IDE haplotypes (global p = 0.02) also showed significant association. CONCLUSIONS: Thus, a previously unreported variant unequivocally associated with increased IDE expression was also associated with reduced plasma Abeta40 and decreased LOAD susceptibility. Genetic association between LOAD and IDE has been difficult to replicate. Our findings suggest that targeted testing of expression SNPs (eSNPs) strongly associated with altered transcript levels in autopsy brain samples may be a powerful way to identify genetic associations with LOAD that would otherwise be difficult to detect.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/genetics , Insulysin/genetics , RNA, Messenger/genetics , Alzheimer Disease/genetics , Case-Control Studies , Humans
15.
EMBO J ; 28(20): 3145-56, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19713932

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are best known for altering the coding sequences of mRNA through RNA editing, as in the GluR-B Q/R site. ADARs have also been shown to affect RNA interference (RNAi) and microRNA processing by deamination of specific adenosines to inosine. Here, we show that ADAR proteins can affect RNA processing independently of their enzymatic activity. We show that ADAR2 can modulate the processing of mir-376a2 independently of catalytic RNA editing activity. In addition, in a Drosophila assay for RNAi deaminase-inactive ADAR1 inhibits RNAi through the siRNA pathway. These results imply that ADAR1 and ADAR2 have biological functions as RNA-binding proteins that extend beyond editing per se and that even genomically encoded ADARs that are catalytically inactive may have such functions.


Subject(s)
Adenosine Deaminase/metabolism , MicroRNAs/genetics , RNA Editing/genetics , RNA, Small Interfering/genetics , Signal Transduction/physiology , Adenosine Deaminase/genetics , Animals , Blotting, Northern , Cell Line , Drosophila , Humans , RNA Interference , RNA-Binding Proteins , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...