1
|
Li B, George EW, Vachali P, Chang FY, Gorusupudi A, Arunkumar R, Giauque NA, Wan Z, Frederick JM, Bernstein PS. Mechanism for the selective uptake of macular carotenoids mediated by the HDL cholesterol receptor SR-BI. Exp Eye Res 2023; 229:109429. [PMID: 36863431 PMCID: PMC10076185 DOI: 10.1016/j.exer.2023.109429] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/24/2022] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
The macular carotenoids lutein and zeaxanthin are taken up from the bloodstream into the human retina through a selective process, for which the HDL cholesterol receptor scavenger receptor BI (SR-BI) in the cells of retinal pigment epithelium (RPE) is thought to be a key mediator. However, the mechanism of SR-BI-mediated selective uptake of macular carotenoids is still not fully understood. Here, we investigate possible mechanisms using biological assays and cultured HEK293 cells, a cell line without endogenous SR-BI expression. Binding affinities between SR-BI and various carotenoids were measured by surface plasmon resonance (SPR) spectroscopy, which shows that SR-BI cannot bind lutein or zeaxanthin specifically. Overexpression of SR-BI in HEK293 cells results in more lutein and zeaxanthin taken up than β-carotene, and this effect can be eliminated by an SR-BI mutant (C384Y) whose cholesterol uptake tunnel is blocked. Next, we determined the effects of HDL and hepatic lipase (LIPC), SR-BI's partners in HDL cholesterol transport, on SR-BI-mediated carotenoid uptake. HDL addition dramatically reduced lutein, zeaxanthin, and β-carotene in HEK293 cells expressing SR-BI, but the cellular lutein and zeaxanthin are higher than β-carotene. LIPC addition increases the uptake of all three carotenoids in HDL-treated cells, and promotes the transport of lutein and zeaxanthin better than β-carotene. Our results suggest that SR-BI and its HDL cholesterol partner HDL and LIPC may be involved in the selective uptake of macular carotenoids.
Collapse
Affiliation(s)
- Binxing Li
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Evan W George
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Preejith Vachali
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Fu-Yen Chang
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Aruna Gorusupudi
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Ranganathan Arunkumar
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Nathan A Giauque
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Zihe Wan
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Jeanne M Frederick
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Paul S Bernstein
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
2
|
Li B, Vachali P, Chang FY, Gorusupudi A, Arunkumar R, Shi L, Rognon GT, Frederick JM, Bernstein PS. HDL is the primary transporter for carotenoids from liver to retinal pigment epithelium in transgenic ApoA-I -/-/Bco2 -/- mice. Arch Biochem Biophys 2022; 716:109111. [PMID: 34942193 PMCID: PMC8792244 DOI: 10.1016/j.abb.2021.109111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 11/02/2022]
Abstract
Supplementation with antioxidant carotenoids is a therapeutic strategy to protect against age-related macular degeneration (AMD); however, the transport mechanism of carotenoids from the liver to the retina is still not fully understood. Here, we investigate if HDL serves as the primary transporter for the macular carotenoids. ApoA-I, the key apolipoprotein of HDL, was genetically deleted from BCO2 knockout (Bco2-/-) mice, a macular pigment mouse model capable of accumulating carotenoids in the retina. We then conducted a feeding experiment with a mixed carotenoid chow (lutein:zeaxanthin:β-carotene = 1:1:1) for one month. HPLC data demonstrated that the total carotenoids were increased in the livers but decreased in the serum, retinal pigment epithelium (RPE)/choroids, and retinas of ApoA-I-/-/Bco2-/- mice compared to Bco2-/- mice. In detail, ApoA-I deficiency caused a significant increase of β-carotene but not lutein and zeaxanthin in the liver, decreased all three carotenoids in the serum, blocked the majority of zeaxanthin and β-carotene transport to the RPE/choroid, and dramatically reduced β-carotene and zeaxanthin but not lutein in the retina. Furthermore, surface plasmon resonance spectroscopy (SPR) data showed that the binding affinity between ApoA-I and β-carotene ≫ zeaxanthin > lutein. Our results show that carotenoids are transported from the liver to the eye mainly by HDL, and ApoA-I may be involved in the selective delivery of macular carotenoids to the RPE.
Collapse
Affiliation(s)
- Binxing Li
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Preejith Vachali
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Fu-Yen Chang
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Aruna Gorusupudi
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Ranganathan Arunkumar
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Linjia Shi
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Gregory T Rognon
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Jeanne M Frederick
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA
| | - Paul S Bernstein
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
3
|
Zou Y, Li JJ, Xue W, Kong X, Duan H, Li Y, Wei L. Epigenetic Modifications and Therapy in Uveitis. Front Cell Dev Biol 2021; 9:758240. [PMID: 34869347 PMCID: PMC8636745 DOI: 10.3389/fcell.2021.758240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Uveitis is a sight-threatening intraocular inflammation, and the exact pathogenesis of uveitis is not yet clear. Recent studies, including multiple genome-wide association studies (GWASs), have identified genetic variations associated with the onset and progression of different types of uveitis, such as Vogt–Koyanagi–Harada (VKH) disease and Behcet’s disease (BD). However, epigenetic regulation has been shown to play key roles in the immunoregulation of uveitis, and epigenetic therapies are promising treatments for intraocular inflammation. In this review, we summarize recent advances in identifying epigenetic programs that cooperate with the physiology of intraocular immune responses and the pathology of intraocular inflammation. These attempts to understand the epigenetic mechanisms of uveitis may provide hope for the future development of epigenetic therapies for these devastating intraocular inflammatory conditions.
Collapse
Affiliation(s)
- Yanli Zou
- Department of Ophthalmology, Affiliated Foshan Hospital, Southern Medical University, Foshan, China.,State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Jing Jing Li
- State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Wei Xue
- State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Xiangbin Kong
- Department of Ophthalmology, Affiliated Foshan Hospital, Southern Medical University, Foshan, China
| | - Hucheng Duan
- Department of Ophthalmology, Affiliated Foshan Hospital, Southern Medical University, Foshan, China
| | - Yiqun Li
- Department of Orthopaedics, Affiliated Foshan Hospital, Southern Medical University, Foshan, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Immunoaffinity LC–MS/MS is more suitable than ELISA to quantify a PEGylated molecule in cynomolgus monkey serum. Bioanalysis 2020; 12:1061-1069. [DOI: 10.4155/bio-2020-0097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: Polyethylene glycolylation (PEGylation) technology is a long-acting delivery platform used to increase the half-life of protein therapeutics. Quantitation of PEGylated anti-Factor D Fab (PEG-aFD) poses bioanalytical challenges. Results: An ELISA was developed to determine total Fab concentration in cynomolgus monkey serum following intravitreal administration of PEG-aFD. However, assay characterization showed a low recovery of about 25% for free unconjugated Fab whereas recovery for PEG-conjugated Fab was within 80–120%. To overcome this challenge, an immunoaffinity liquid chromatography tandem mass spectrometry (IA LC–MS/MS) assay was developed, achieving recovery within 80–120% for both free and conjugated Fab. Conclusion: Immunoaffinity LC–MS/MS is more suitable than ELISA to accurately quantify the total protein concentration of PEG-aFD in cynomolgus monkey serum.
Collapse
|
5
|
Loyet KM, Hass PE, Sandoval WN, Morando A, Liu P, Shatz W, Dickmann L, Kenrick M, Good J, Davancaze T, Morimoto AM, Kelley RF, Scheer JM. In Vivo Stability Profiles of Anti-factor D Molecules Support Long-Acting Delivery Approaches. Mol Pharm 2018; 16:86-95. [PMID: 30444371 DOI: 10.1021/acs.molpharmaceut.8b00871] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The collection of aqueous humor (phase 1 b/2 Mahalo study) from patients dosed intravitreally with anti-factor D (AFD; FCFD4514S, lampalizumab), a humanized antibody fragment previously under investigation to treat geographic atrophy (GA) secondary to age-related macular degeneration, presented a unique opportunity to examine AFD properties in clinical samples. We investigated AFD stability and target-binding characteristics to set up strategies for engineering and evaluating optimized molecules that enable less frequent dosing. Two variants, AFD.v8 and AFD.v14, were evaluated as alternatives to AFD for longer-acting treatments. Mass spectrometry, surface plasmon resonance, and immunoassay were used to assess AFD stability and binding activity in aqueous humor samples from Mahalo patients. In vitro stability and binding activity of AFD, AFD.v8, and AFD.v14 were assessed in human vitreous humor versus buffer at 37 °C over 16 weeks and in vivo in rabbits over 28 days along with pharmacokinetic determinations. In human aqueous humor, AFD specific binding was >85% through 30 days, and deamidation was <3% through 60 days, consistent with the AFD stability and binding activity in vitreous humor from humans in vitro and rabbits in vivo. Target binding, stability, and rabbit pharmacokinetic parameters of AFD.v8 and AFD.v14 were similar to those of AFD. Physiological stability and activity of AFD translated across in vitro and in vivo studies in humans and rabbits. The two variants AFD.v8 and AFD.v14 demonstrated comparable potency and pharmacokinetics. These findings, along with previously demonstrated improved solubility of AFD.v8 and AFD.v14, provide proof-of-concept for developing other similar long-acting therapeutic variants.
Collapse
Affiliation(s)
- Kelly M Loyet
- Department of Biochemical and Cellular Pharmacology , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Philip E Hass
- Department of Protein Chemistry , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Wendy N Sandoval
- Department of Microchemistry, Proteomics, & Lipidomics , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Ashley Morando
- Department of Biochemical and Cellular Pharmacology , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Peter Liu
- Department of Microchemistry, Proteomics, & Lipidomics , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Whitney Shatz
- Department of Protein Chemistry , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Leslie Dickmann
- Department of Preclinical and Translational Pharmacokinetics , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Margaret Kenrick
- Department of Preclinical and Translational Pharmacokinetics , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Jeremy Good
- Department of Assay Development and Technology , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Teresa Davancaze
- Department of Assay Development and Technology , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Alyssa M Morimoto
- Department of Assay Development and Technology , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Robert F Kelley
- Department of Drug Delivery , Genentech, Inc. , South San Francisco , California 94080 , United States
| | - Justin M Scheer
- Department of Protein Chemistry , Genentech, Inc. , South San Francisco , California 94080 , United States
| |
Collapse
|
6
|
Kim KL, Park SP. Association between serum vitamin D deficiency and age-related macular degeneration in Koreans: Clinical case-control pilot study. Medicine (Baltimore) 2018; 97:e11908. [PMID: 30113489 PMCID: PMC6112886 DOI: 10.1097/md.0000000000011908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
The objective of this study was to evaluate the association between serum vitamin D deficiency and age-related macular degeneration (AMD) in Koreans through a clinical case-control pilot study. The study included 96 patients: 30 with late AMD, 32 with early AMD, and 34 normal controls. The patients with late AMD were divided into 2 subgroups based on the presence or absence of subretinal fibrosis on optical coherence tomography (OCT) images. We measured 25-hydroxyvitamin D levels in the serum of all patients during the same season to rule out seasonal variation of serum vitamin D level. Serum vitamin D deficiency was defined as a serum 25-hydroxyvitamin D level below 20 ng/mL. Serum vitamin D deficiency had a tendency to increase the risk of early AMD, although with borderline significance [odds ratio (OR) = 3.59; 95% confidence interval (95% CI) 0.95-13.58; P = .060]. It was significantly associated with a greater risk of late AMD (OR = 3.61; 95%CI 1.04-12.51; P = .043). Among the 2 subgroups of patients with late AMD, those with subretinal fibrosis present on the OCT images showed a greater risk of serum vitamin D deficiency than the normal controls (OR = 7.54; 95% CI 1.34-42.51). However, there was no significant association between serum vitamin D deficiency and late AMD without subretinal fibrosis (OR = 1.89; 95% CI 0.40-8.92). Serum vitamin D deficiency may increase the risk of early and late AMD in Koreans, and may also be associated with subretinal fibrosis in this population.
Collapse
|
7
|
Epigenetics, microbiota, and intraocular inflammation: New paradigms of immune regulation in the eye. Prog Retin Eye Res 2018; 64:84-95. [DOI: 10.1016/j.preteyeres.2018.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 01/07/2018] [Accepted: 01/11/2018] [Indexed: 01/15/2023]
|
8
|
Graham BC, Pulido JS, Winters JL. Seeing is believing: A review of apheresis therapy in the treatment of ophthalmologic disease. J Clin Apher 2017; 33:380-392. [PMID: 29150864 DOI: 10.1002/jca.21607] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 12/27/2022]
Abstract
Apheresis procedures have a role in treatment of disparate diseases involving many different organ systems. Often the disease processes where apheresis plays a role in treatment are considered "orphan diseases"-relatively rare disease processes that lack specific pharmaceutical agents or established treatment protocols. Many of these disease processes can affect the eye with devastating results for the eyesight of these patients. The unique ability of apheresis to affect disease by modifying blood plasma and modulating disease-causing agents therein renders apheresis procedures valuable tools in the treatment of certain ophthalmologic diseases. This review comprehensively evaluates the role of apheresis in the treatment of ophthalmologic diseases of the eye and surrounding orbit including age-related macular degeneration, bilateral diffuse uveal melanocytic proliferation, paraneoplastic retinopathy, atopic keratoconjunctivitis, sympathetic ophthalmia, and endocrine-associated ophthalmopathy. Apheresis procedure parameters are provided for the apheresis practitioner based on review of the relevant literature.
Collapse
Affiliation(s)
- Brendan C Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota
| | - Jeffrey L Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
9
|
Adamus G. Can innate and autoimmune reactivity forecast early and advance stages of age-related macular degeneration? Autoimmun Rev 2017; 16:231-236. [PMID: 28137479 PMCID: PMC5334174 DOI: 10.1016/j.autrev.2017.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/03/2016] [Indexed: 01/17/2023]
Abstract
Age-related macular degeneration (AMD) is a major cause of central vision loss in persons over 55years of age in developed countries. AMD is a complex disease in which genetic, environmental and inflammatory factors influence its onset and progression. Elevation in serum anti-retinal autoantibodies, plasma and local activation of complement proteins of the alternative pathway, and increase in secretion of proinflammatory cytokines have been seen over the course of disease. Genetic studies of AMD patients confirmed that genetic variants affecting the alternative complement pathway have a major influence on AMD risk. Because the heterogeneity of this disease, there is no sufficient strategy to identify the disease onset and progression sole based eye examination, thus identification of reliable serological biomarkers for diagnosis, prognosis and response to treatment by sampling patient's blood is necessary. This review provides an outline of the current knowledge on possible serological (autoantibodies, complement factors, cytokines, chemokines) and related genetic biomarkers relevant to the pathology of AMD, and discusses their application for prediction of disease activity and prognosis in AMD.
Collapse
Affiliation(s)
- Grazyna Adamus
- Ocular Immunology Laboratory, Casey Eye Institute, School of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Iannaccone A, Hollingsworth TJ, Koirala D, New DD, Lenchik NI, Beranova-Giorgianni S, Gerling IC, Radic MZ, Giorgianni F. Retinal pigment epithelium and microglia express the CD5 antigen-like protein, a novel autoantigen in age-related macular degeneration. Exp Eye Res 2016; 155:64-74. [PMID: 27989757 DOI: 10.1016/j.exer.2016.12.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 11/19/2022]
Abstract
We report on a novel autoantigen expressed in human macular tissues, identified following an initial Western blot (WB)-based screening of sera from subjects with age-related macular degeneration (AMD) for circulating auto-antibodies (AAbs) recognizing macular antigens. Immunoprecipitation, 2D-gel electrophoresis (2D-GE) and liquid chromatography-tandem mass spectrometry (LC-MS/MS), direct enzyme-linked immunosorbent assays (ELISA), WBs, immunohistochemistry (IHC), human primary and ARPE-19 immortalized cell cultures were used to characterize this novel antigen. An approximately 40-kDa autoantigen in AMD was identified as the scavenger receptor CD5 antigen-like protein (CD5L), also known as apoptosis inhibitor of macrophage (AIM). CD5L/AIM was localized to human RPE by IHC and WB methods and to retinal microglial cells by IHC. ELISAs with recombinant CD5L/AIM on a subset of AMD sera showed a nearly 2-fold higher anti-CD5L/AIM reactivity in AMD vs. Control sera (p = 0.000007). Reactivity ≥0.4 was associated with 18-fold higher odds of having AMD (χ2 = 21.42, p = 0.00063). Circulating CD5L/AIM levels were also nearly 2-fold higher in AMD sera compared to controls (p = 0.0052). The discovery of CD5L/AIM expression in the RPE and in retinal microglial cells adds to the known immunomodulatory roles of these cells in the retina. The discovery of AAbs recognizing CD5L/AIM identifies a possible novel disease biomarker and suggest a potential role for CD5L/AIM in the pathogenesis of AMD in situ. The possible mechanisms via which anti-CD5L/AIM AAbs may contribute to AMD pathogenesis are discussed. In particular, since CD5L is known to stimulate autophagy and to participate in oxidized LDL uptake in macrophages, we propose that anti-CD5L/AIM auto-antibodies may play a role in drusen biogenesis and inflammatory RPE damage in AMD.
Collapse
Affiliation(s)
- Alessandro Iannaccone
- University of Tennessee Health Science Center, Department of Ophthalmology, Hamilton Eye Institute, Memphis, TN, USA; Department of Ophthalmology, Duke Eye Center, Duke University School of Medicine, Durham, NC, USA.
| | - T J Hollingsworth
- University of Tennessee Health Science Center, Department of Ophthalmology, Hamilton Eye Institute, Memphis, TN, USA
| | - Diwa Koirala
- University of Tennessee Health Science Center, Department of Ophthalmology, Hamilton Eye Institute, Memphis, TN, USA; University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, Memphis, TN, USA
| | - David D New
- University of Tennessee Health Science Center, Department of Ophthalmology, Hamilton Eye Institute, Memphis, TN, USA
| | - Nataliya I Lenchik
- University of Tennessee Health Science Center, Department of Ophthalmology, Hamilton Eye Institute, Memphis, TN, USA; University of Tennessee Health Science Center, Department of Medicine, Division of Endocrinology, Memphis, TN, USA
| | - Sarka Beranova-Giorgianni
- University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, Memphis, TN, USA
| | - Ivan C Gerling
- University of Tennessee Health Science Center, Department of Medicine, Division of Endocrinology, Memphis, TN, USA
| | - Marko Z Radic
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, TN, USA
| | - Francesco Giorgianni
- University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, Memphis, TN, USA
| |
Collapse
|
11
|
Kepez Yildiz B, Ozdek S, Ergun MA, Ergun S, Yaylacioglu Tuncay F, Elbeg S. CFH Y402H and VEGF Polymorphisms and Anti-VEGF Treatment Response in Exudative Age-Related Macular Degeneration. Ophthalmic Res 2016; 56:132-8. [PMID: 27404493 DOI: 10.1159/000446186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/13/2016] [Indexed: 11/19/2022]
Abstract
PURPOSE The aim of this study was to evaluate the prevalence of single nucleotide polymorphisms (SNPs) in complement factor H (CFH) Y402H and VEGF rs2146323 and rs699947 in exudative age-related macular degeneration (AMD) and their relationship with intravitreal anti-VEGF treatment response. METHODS A total of 109 exudative AMD patients and 70 controls were included. Patients were classified as 'good responders' and 'nonresponders' based on the changes in best corrected visual acuity, central foveal thickness, lesion size, and the persistence of retinal hemorrhage after three dosages of anti-VEGF. We examined CFH, VEGF rs2146323 and rs699947 SNPs, and plasma interleukin-6 (IL-6) levels in both groups. RESULTS In total, 42 patients (38.5%) and 11 controls (15.7%) had homozygote wild genotype TT (p = 0.002). The variant C allele frequency was 45% in controls and 31.7% in patients (p = 0.011). A and C allele frequencies for VEGF rs699947 and rs2416323 were similar between the control and patient groups (p = 0.947, p = 0.378). Both SNPs were similar in responders and nonresponders. No significant difference was detected between plasma IL-6 levels of the control and AMD groups (p = 0.594), but the levels were higher in good responders than nonresponders (p < 0.001). CONCLUSION CFH Y402H SNP might be protective for AMD in the Turkish population. VEGF rs2146323 and rs699947 SNPs have no relationship to exudative AMD formation, and none of these seem to have any effect on anti-VEGF response.
Collapse
Affiliation(s)
- Burcin Kepez Yildiz
- Department of Ophthalmology, Gazi University School of Medicine, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
12
|
Ferrington DA, Kapphahn RJ, Leary MM, Atilano SR, Terluk MR, Karunadharma P, Chen GKJ, Ratnapriya R, Swaroop A, Montezuma SR, Kenney MC. Increased retinal mtDNA damage in the CFH variant associated with age-related macular degeneration. Exp Eye Res 2016; 145:269-277. [PMID: 26854823 DOI: 10.1016/j.exer.2016.01.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 11/17/2022]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness among the elderly in the developed world. Genetic analysis of AMD has identified 34 high-risk loci associated with AMD. The genes at these high risk loci belong to diverse biological pathways, suggesting different mechanisms leading to AMD pathogenesis. Thus, therapies targeting a single pathway for all AMD patients will likely not be universally effective. Recent evidence suggests defects in mitochondria (mt) of the retinal pigment epithelium (RPE) may constitute a key pathogenic event in some AMD patients. The purpose of this study is to determine if individuals with a specific genetic background have a greater propensity for mtDNA damage. We used human eyebank tissues from 76 donors with AMD and 42 age-matched controls to determine the extent of mtDNA damage in the RPE that was harvested from the macula using a long extension polymerase chain reaction assay. Genotype analyses were performed for ten common AMD-associated nuclear risk alleles (ARMS2, TNFRSF10A, CFH, C2, C3, APOE, CETP, LIPC, VEGF and COL10A1) and mtDNA haplogroups. Sufficient samples were available for genotype association with mtDNA damage for TNFRSF10A, CFH, CETP, VEGFA, and COL10A1. Our results show that AMD donors carrying the high risk allele for CFH (C) had significantly more mtDNA damage compared with donors having the wild-type genetic profile. The data from an additional 39 donors (12 controls and 27 AMD) genotyped for CFH alleles further supported these findings. Taken together, these studies provide the rationale for a more personalized approach for treating AMD by uncovering a significant correlation between the CFH high risk allele and accelerated mtDNA damage. Patients harboring this genetic risk factor may benefit from therapies that stabilize and protect the mt in the RPE.
Collapse
Affiliation(s)
- Deborah A Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Rebecca J Kapphahn
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Michaela M Leary
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Shari R Atilano
- Gavin Herbert Eye Institute, University of California, Irvine, CA, 92697, USA
| | - Marcia R Terluk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Pabalu Karunadharma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Rinki Ratnapriya
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, Bethesda, MD 20892, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, Bethesda, MD 20892, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California, Irvine, CA, 92697, USA
| |
Collapse
|
13
|
Bernstein PS, Li B, Vachali PP, Gorusupudi A, Shyam R, Henriksen BS, Nolan JM. Lutein, zeaxanthin, and meso-zeaxanthin: The basic and clinical science underlying carotenoid-based nutritional interventions against ocular disease. Prog Retin Eye Res 2016; 50:34-66. [PMID: 26541886 PMCID: PMC4698241 DOI: 10.1016/j.preteyeres.2015.10.003] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/04/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
The human macula uniquely concentrates three carotenoids: lutein, zeaxanthin, and meso-zeaxanthin. Lutein and zeaxanthin must be obtained from dietary sources such as green leafy vegetables and orange and yellow fruits and vegetables, while meso-zeaxanthin is rarely found in diet and is believed to be formed at the macula by metabolic transformations of ingested carotenoids. Epidemiological studies and large-scale clinical trials such as AREDS2 have brought attention to the potential ocular health and functional benefits of these three xanthophyll carotenoids consumed through the diet or supplements, but the basic science and clinical research underlying recommendations for nutritional interventions against age-related macular degeneration and other eye diseases are underappreciated by clinicians and vision researchers alike. In this review article, we first examine the chemistry, biochemistry, biophysics, and physiology of these yellow pigments that are specifically concentrated in the macula lutea through the means of high-affinity binding proteins and specialized transport and metabolic proteins where they play important roles as short-wavelength (blue) light-absorbers and localized, efficient antioxidants in a region at high risk for light-induced oxidative stress. Next, we turn to clinical evidence supporting functional benefits of these carotenoids in normal eyes and for their potential protective actions against ocular disease from infancy to old age.
Collapse
Affiliation(s)
- Paul S Bernstein
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Binxing Li
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Preejith P Vachali
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Aruna Gorusupudi
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Rajalekshmy Shyam
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - Bradley S Henriksen
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT, 84132, USA.
| | - John M Nolan
- Macular Pigment Research Group, Vision Research Centre, School of Health Science, Carriganore House, Waterford Institute of Technology West Campus, Carriganore, Waterford, Ireland.
| |
Collapse
|
14
|
Haas P, Kubista KE, Krugluger W, Huber J, Binder S. Impact of visceral fat and pro-inflammatory factors on the pathogenesis of age-related macular degeneration. Acta Ophthalmol 2015; 93:533-8. [PMID: 25683020 DOI: 10.1111/aos.12670] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 12/24/2014] [Indexed: 01/06/2023]
Abstract
PURPOSE Previous studies have indicated that the immune system is involved in the pathogenesis of the AMD. Increased visceral fat, in addition, has a pro-inflammatory effect on the organism by producing or influencing different kinds of inflammatory factors. The aim of this study is to determine the relationship of body fat distribution in patients with age-related macula degeneration in comparison to a control group in the Austrian population. METHODS In this case-control study, body weight and height, and body mass index (BMI) were measured for each subject in 54 patients with exudative AMD and compared to 46 gender- and age-matched healthy control subjects. Body composition and abdominal fat areas were measured using dual-energy X-ray absorptiometry (DEXA). Data on age, gender distribution, smoking history and systemic diseases, respectively, were compared. The inflammatory markers CRP, tumour necrosis factor-alpha (TNF-alpha), leptin, amyloid A, amyloid beta and interleukin-6 (IL-6) were assayed by ELISA (R&D). RESULTS DEXA revealed central-abdominal-to-total body fat ratio of 0.073 +/- 0.011 in AMD patients compared to 0.061 +/- 0.013 in the controls (p <0.001; d = 0.98). The calculation of BMI has provided a significant result (p =0.045). U-test results for Aß1-42, IL-6, SAA and CRP each were significant (p < 0.05), with higher values in AMD patients. Leptin, TNF-alpha and Aß1-40 showed no significant differences between the groups. CONCLUSION Our results suggest that abdominal fat distribution is significantly associated with age-related macular degeneration. Analysis of patients with exudative AMD revealed higher levels of CRP, amyloid ß1-42, IL-6 and amyloid alpha.
Collapse
Affiliation(s)
- Paulina Haas
- Department of Ophthalmology; Ludwig Boltzmann Institute for Retinology and Biomicroscopic Lasersurgery; Rudolf Foundation Clinic; Vienna Austria
| | - Katharina E. Kubista
- Department of Ophthalmology; Ludwig Boltzmann Institute for Retinology and Biomicroscopic Lasersurgery; Rudolf Foundation Clinic; Vienna Austria
| | - Walter Krugluger
- Department of Laboratory Medicines; Social Medical Center East; Vienna Austria
| | - Johannes Huber
- Gynecology; Medical University of Vienna; Vienna Austria
| | - Susanne Binder
- Department of Ophthalmology; Ludwig Boltzmann Institute for Retinology and Biomicroscopic Lasersurgery; Rudolf Foundation Clinic; Vienna Austria
| |
Collapse
|
15
|
Atilano SR, Malik D, Chwa M, Cáceres-Del-Carpio J, Nesburn AB, Boyer DS, Kuppermann BD, Jazwinski SM, Miceli MV, Wallace DC, Udar N, Kenney MC. Mitochondrial DNA variants can mediate methylation status of inflammation, angiogenesis and signaling genes. Hum Mol Genet 2015; 24:4491-503. [PMID: 25964427 PMCID: PMC4512622 DOI: 10.1093/hmg/ddv173] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial (mt) DNA can be classified into haplogroups representing different geographic and/or racial origins of populations. The H haplogroup is protective against age-related macular degeneration (AMD), while the J haplogroup is high risk for AMD. In the present study, we performed comparison analyses of human retinal cell cybrids, which possess identical nuclei, but mtDNA from subjects with either the H or J haplogroups, and demonstrate differences in total global methylation, and expression patterns for two genes related to acetylation and five genes related to methylation. Analyses revealed that untreated-H and -J cybrids have different expression levels for nuclear genes (CFH, EFEMP1, VEGFA and NFkB2). However, expression levels for these genes become equivalent after treatment with a methylation inhibitor, 5-aza-2'-deoxycytidine. Moreover, sequencing of the entire mtDNA suggests that differences in epigenetic status found in cybrids are likely due to single nucleotide polymorphisms (SNPs) within the haplogroup profiles rather than rare variants or private SNPs. In conclusion, our findings indicate that mtDNA variants can mediate methylation profiles and transcription for inflammation, angiogenesis and various signaling pathways, which are important in several common diseases.
Collapse
Affiliation(s)
| | | | | | | | - Anthony B Nesburn
- Gavin Herbert Eye Institute and Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David S Boyer
- Retina-Vitreous Associates Medical Group, Beverly Hills, CA 90211, USA
| | | | - S Michal Jazwinski
- Tulane Center for Aging and Department of Medicine, Tulane University, New Orleans, LA 70118, USA and
| | - Michael V Miceli
- Tulane Center for Aging and Department of Medicine, Tulane University, New Orleans, LA 70118, USA and
| | - Douglas C Wallace
- Center of Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - M Cristina Kenney
- Gavin Herbert Eye Institute and Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92697, USA,
| |
Collapse
|
16
|
Hwang HS, Lee SB, Jee D. Association between Blood Lead Levels and Age-Related Macular Degeneration. PLoS One 2015; 10:e0134338. [PMID: 26252225 PMCID: PMC4529082 DOI: 10.1371/journal.pone.0134338] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 07/08/2015] [Indexed: 12/30/2022] Open
Abstract
Purpose To investigate the association between blood lead levels and prevalence of age-related macular degeneration (AMD). Methods A nationwide population-based cross-sectional study included 4,933 subjects aged over 40 years who participated in the 2008–2012 Korean National Health and Nutrition Examination Survey, and for whom fundus photographs were available. All participants underwent a standardized interview, evaluation of blood lead concentration, and a comprehensive ophthalmic examination. Digital fundus photographs (45°) were taken of both eyes under physiological mydriasis. All fundus photographs were graded using an international classification and grading system. Results Mean blood lead levels were 3.15 μg/dL in men and 2.27 μg/dL in women (P < 0.001). After adjusting for potential confounders including age, gender, smoking status, total cholesterol levels, triglyceride levels, heart problems and strokes, the adjusted odds ratio (OR) in women for any AMD was 1.86 (95% Confidence Interval [CI], 1.03–3.36) and for early AMD was 1.92 (95% CI, 1.06–3.48), for those in the highest quintile of lead level compared with the lowest quintile. In men, however, blood lead level was not significantly associated with AMD. Conclusions Blood lead levels were higher in men, but were only associated with AMD in women. Increased levels of blood lead may be involved in the pathogenesis of AMD development in women.
Collapse
Affiliation(s)
- Ho Sik Hwang
- Department of Ophthalmology, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Korea
| | | | - Donghyun Jee
- Department of Ophthalmology and Visual Science, St. Vincent’s Hospital, College of Medicine, Catholic University of Korea, Suwon, Korea
- * E-mail:
| |
Collapse
|
17
|
Pei X, Ma K, Xu J, Wang N, Liu N. Inhibition of cell proliferation and migration after HTRA1 knockdown in retinal pigment epithelial cells. Graefes Arch Clin Exp Ophthalmol 2015; 253:565-72. [PMID: 25550099 DOI: 10.1007/s00417-014-2901-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 12/06/2014] [Accepted: 12/15/2014] [Indexed: 10/24/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the role of HtrA serine peptidase 1 (HTRA1) in the proliferation and migration of cells of the human retinal pigment epithelial cell line ARPE-19, and the possible mechanisms involved. METHODS ARPE-19 cells were transduced by a recombinant lentiviral vector carrying HTRA1-shRNA to knockdown HTRA1 expression. Subsequent HTRA1 gene and HTRA1 protein levels in these cells and control cells were detected by quantitative real-time PCR and Western blot, respectively. Changes in cell proliferation and migration associated with the inhibition of HTRA1 expression were assessed, as well as changes in the mRNA levels of transforming growth factor beta 1 (TGFB1), bone morphogenetic protein 4 (BMP4), and bone morphogenetic protein 2 (BMP2). RESULTS The recombinant lentivirus carrying HTRA1-shRNA was successfully generated, as evidenced by reduced levels of HTRA1 mRNA and HTRA1 protein in ARPE-19 cells. The knockdown of HTRA1 in ARPE-19 cells was associated with reduced cellular proliferation and migration, and increased mRNA levels of TGF-β1, BMP4, and BMP2. CONCLUSIONS Silence of the HTRA1 gene was associated with significantly higher levels of TGF-β1, BMP4, and BMP2 mRNA and reduction in the proliferation and migration of ARPE-19 cells.
Collapse
Affiliation(s)
- Xueting Pei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, No. 1 Dongjiaominxiang, Dongcheng District, Beijing, China,
| | | | | | | | | |
Collapse
|
18
|
Joint Effect of CFH and ARMS2/HTRA1 Polymorphisms on Neovascular Age-Related Macular Degeneration in Chinese Population. J Ophthalmol 2015; 2015:821918. [PMID: 25883802 PMCID: PMC4389821 DOI: 10.1155/2015/821918] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 03/10/2015] [Indexed: 12/03/2022] Open
Abstract
Purpose. The etiology of neovascular age-related macular degeneration (nAMD) cannot be completely explained by identified environmental risk factors or single-locus gene variants. This study was to explore the potential interactions among gene variants on nAMD in Chinese population.
Methods. 43 SNPs located in different genes were genotyped in 932 Chinese individuals (464 nAMD patients and 468 controls). We explored the potential interactions among gene variants using generalized multifactor dimensionality reduction (GMDR) algorithm and the method to measure the departure from the additivity model. Results. The joint effect that involved CFH rs1061170 and HTRA1 rs3793917 was shown statistically significant (P < 0.001) with the highest cross-validation consistency (10/10) and the best testing balanced accuracy (64.50%). In addition, based on the method to measure the departure from the additivity model, the synergy index (S) was 2.63 (1.09–6.38) and the attributable proportion due to interaction (AP) was 55.7% (21.4%–89.9%), which suggested that a common pathway may exist for these genes for nAMD. Those who carried CC for rs3793917 and TC/CC for rs1061170 were at the highest risk of nAMD (OR: 9.76, 95% CI: 4.65–20.51). Conclusions. Evidence that the joint effect that involved CFH and ARMS2/HTRA1 may contribute to the risk of neovascular AMD in Chinese population was obtained.
Collapse
|
19
|
Identification of genome-wide SNP-SNP and SNP-clinical Boolean interactions in age-related macular degeneration. Methods Mol Biol 2015; 1253:217-55. [PMID: 25403535 DOI: 10.1007/978-1-4939-2155-3_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We propose here a methodology to uncover modularities in the network of SNP-SNP interactions most associated with disease. We start by computing all possible Boolean binary SNP interactions across the whole genome. By constructing a weighted graph of the most relevant interactions and via a combinatorial optimization approach, we find the most highly interconnected SNPs. We show that the method can be easily extended to find SNP/environment interactions. Using a modestly sized GWAS dataset of age-related macular degeneration (AMD), we identify a group of only 19 SNPs, which include those in previously reported regions associated to AMD. We also uncover a larger set of loci pointing to a matrix of key processes and functions that are affected. The proposed integrative methodology extends and overlaps traditional statistical analysis in a natural way. Combinatorial optimization techniques allow us to find the kernel of the most central interactions, complementing current methods of GWAS analysis and also enhancing the search for gene-environment interaction.
Collapse
|
20
|
Gemenetzi M, Lotery AJ. The role of epigenetics in age-related macular degeneration. Eye (Lond) 2014; 28:1407-17. [PMID: 25233816 DOI: 10.1038/eye.2014.225] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/31/2014] [Indexed: 12/27/2022] Open
Abstract
It is becoming increasingly evident that epigenetic mechanisms influence gene expression and can explain how interactions between genetics and the environment result in particular phenotypes during development. The extent to which this epigenetic effect contributes to phenotype heritability in age-related macular degeneration (AMD) is currently ill defined. However, emerging evidence suggests that epigenetic changes are relevant to AMD and as such provide an exciting new avenue of research for AMD. This review addresses information on the impact of posttranslational modification of the genome on the pathogenesis of AMD, such as DNA methylation changes affecting antioxidant gene expression, hypoxia-regulated alterations in chromatin structure, and histone acetylation status in relation to angiogenesis and inflammation. It also contains information on the role of non-coding RNA-mediated gene regulation in AMD at a posttranscriptional (before translation) level. Our aim was to review the epigenetic mechanisms that cause heritable changes in gene activity without changing the DNA sequence. We also describe some long-term alterations in the transcriptional potential of a cell, which are not necessarily heritable but remains to be defined in the future. Increasing understanding of the significance of common and rare genetic variants and their relationship to epigenetics and environmental influences may help in establishing methods to assess the risk of AMD. This in turn may allow new therapeutic interventions for the leading cause of central vision impairment in patients over the age of 50 years in developed countries. Search strategy We searched the MEDLINE/PubMed database following MeSH suggestions for articles including the terms: 'ocular epigenetic mechanisms', 'human disease epigenetics', and 'age-related macular degeneration genetics'. The headline used to locate related articles in PubMed was 'epigenetics in ocular disease', and to restrict search, we used the headlines 'DNA methylation in age related macular degeneration', 'altered gene expression in AMD pathogenesis'. A manual search was also based on references from these articles as well as review articles.
Collapse
Affiliation(s)
- M Gemenetzi
- Southampton Eye Unit, Southampton University Hospital, Southampton, UK
| | - A J Lotery
- 1] Southampton Eye Unit, Southampton University Hospital, Southampton, UK [2] Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| |
Collapse
|
21
|
Loyet KM, Good J, Davancaze T, Sturgeon L, Wang X, Yang J, Le KN, Wong M, Hass PE, van Lookeren Campagne M, Haughney PC, Morimoto A, Damico-Beyer LA, DeForge LE. Complement inhibition in cynomolgus monkeys by anti-factor d antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration. J Pharmacol Exp Ther 2014; 351:527-37. [PMID: 25232192 DOI: 10.1124/jpet.114.215921] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Anti-factor D (AFD; FCFD4514S, lampalizumab) is a humanized IgG Fab fragment directed against factor D (fD), a rate-limiting serine protease in the alternative complement pathway (AP). Evaluation of AFD as a potential intravitreal (IVT) therapeutic for dry age-related macular degeneration patients with geographic atrophy (GA) is ongoing. However, it is unclear whether IVT administration of AFD can affect systemic AP activation and potentially compromise host-immune responses. We characterized the pharmacologic properties of AFD and assessed the effects of AFD administered IVT (2 or 20 mg) or intravenous (0.2, 2, or 20 mg) on systemic complement activity in cynomolgus monkeys. For the IVT groups, serum AP activity was reduced for the 20 mg dose group between 2 and 6 hours postinjection. For the intravenous groups, AFD inhibited systemic AP activity for periods of time ranging from 5 minutes (0.2 mg group) to 3 hours (20 mg group). Interestingly, the concentrations of total serum fD increased up to 10-fold relative to predose levels following administration of AFD. Furthermore, AFD was found to inhibit systemic AP activity only when the molar concentration of AFD exceeded that of fD. This occurred in cynomolgus monkeys at serum AFD levels ≥2 µg/ml, a concentration 8-fold greater than the maximum serum concentration observed following a single 10 mg IVT dose in a clinical investigation in patients with GA. Based on these findings, the low levels of serum AFD resulting from IVT administration of a clinically relevant dose are not expected to appreciably affect systemic AP activity.
Collapse
Affiliation(s)
- Kelly M Loyet
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Jeremy Good
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Teresa Davancaze
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Lizette Sturgeon
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Xiangdan Wang
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Jihong Yang
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Kha N Le
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Maureen Wong
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Philip E Hass
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Menno van Lookeren Campagne
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Peter C Haughney
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Alyssa Morimoto
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Lisa A Damico-Beyer
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| | - Laura E DeForge
- Departments of Biochemical and Cellular Pharmacology (K.M.L., L.S., L.E.D.), Assay Development and Technologies (J.G., T.D., M.W., A.M.), BioAnalytical Sciences (X.W., J.Y.), Pharmacokinetics and Pharmacodynamics (K.N.L., P.C.H., L.A.D.-B.), Protein Chemistry (P.E.H.), and Immunology (M.v.L.C.), Genentech, South San Francisco, California
| |
Collapse
|
22
|
Casaroli-Marano RP, Alforja S, Giralt J, Farah ME. Epimacular brachytherapy for wet AMD: current perspectives. Clin Ophthalmol 2014; 8:1661-70. [PMID: 25210436 PMCID: PMC4155998 DOI: 10.2147/opth.s46068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Age-related macular degeneration (AMD) is considered the most common cause of blindness in the over-60 age group in developed countries. There are basically two forms of presentation: geographic (dry or atrophic) and wet (neovascular or exudative). Geographic atrophy accounts for approximately 85%–90% of ophthalmic frames and leads to a progressive degeneration of the retinal pigment epithelium and the photoreceptors. Wet AMD causes the highest percentage of central vision loss secondary to disease. This neovascular form involves an angiogenic process in which newly formed choroidal vessels invade the macular area. Today, intravitreal anti-angiogenic drugs attempt to block the angiogenic events and represent a major advance in the treatment of wet AMD. Currently, combination therapy for wet AMD includes different forms of radiation delivery. Epimacular brachytherapy (EMBT) seems to be a useful approach to be associated with current anti-vascular endothelial growth factor agents, presenting an acceptable efficacy and safety profile. However, at the present stage of research, the results of the clinical trials carried out to date are insufficient to justify extending routine use of EMBT for the treatment of wet AMD.
Collapse
Affiliation(s)
- Ricardo P Casaroli-Marano
- Instituto Clínic de Oftalmología (Hospital Clínic de Barcelona), University of Barcelona, Barcelona, Spain ; Department of Ophthalmology and Visual Sciences, Universidade Federal de Sao Paulo, Escola Paulista de Medicina, Sao Paulo, Brazil
| | - Socorro Alforja
- Instituto Clínic de Oftalmología (Hospital Clínic de Barcelona), University of Barcelona, Barcelona, Spain
| | - Joan Giralt
- Instituto Clínic de Oftalmología (Hospital Clínic de Barcelona), University of Barcelona, Barcelona, Spain
| | - Michel E Farah
- Department of Ophthalmology and Visual Sciences, Universidade Federal de Sao Paulo, Escola Paulista de Medicina, Sao Paulo, Brazil
| |
Collapse
|
23
|
Association between polymorphism of the DNA repair SMUG1 and UNG genes and age-related macular degeneration. Retina 2014; 34:38-47. [PMID: 23714858 DOI: 10.1097/iae.0b013e31829477d8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE To investigate the association between the g.4235T>C (rs2337395) polymorphism of the UNG gene and the c.-31A>G (rs3087404) polymorphism of the SMUG1 gene and the risk of age-related macular degeneration (AMD), as well as modulation of this association by some environmental and lifestyle factors. METHODS Overall, 272 AMD patients and 105 control subjects were enrolled in this study. Both polymorphisms were genotyped by restriction fragment length polymorphism-polymerase chain reaction (PCR-RFLP). RESULTS The C/C genotype of the g.4235T>C polymorphism of the UNG gene was associated with an increased risk of dry AMD (odds ratio, 2.54), whereas the T/T genotype of this polymorphism decreased such risk (odds ratio, 0.41). The presence of the T allele of the g.4235T>C polymorphism and the A allele of the c.-31A>G polymorphism of the SMUG1 gene (odds ratio, 2.17 and 2.18, respectively) was associated with an increased risk of AMD severity, expressed by the comparison of the frequencies of genotypes in the group of patients with wet AMD versus those with dry AMD. Conversely, the C/C genotype of the g.4235T>C polymorphism, the G/G genotype of the c.-31A>G polymorphism, and the C/C-G/G combined genotype of both polymorphisms had a protective effect (odds ratio, 0.48, 0.46, and 0.18; respectively). CONCLUSION The results obtained suggest the potential role of the g.4235T>C and the c.-31A>G polymorphisms in AMD pathogenesis.
Collapse
|
24
|
Zhao L, Grob S, Avery R, Kimura A, Pieramici D, Lee J, Rabena M, Ortiz S, Quach J, Cao G, Luo H, Zhang M, Pei M, Song Y, Tornambe P, Goldbaum M, Ferreyra H, Kozak I, Zhang K. Common variant in VEGFA and response to anti-VEGF therapy for neovascular age-related macular degeneration. Curr Mol Med 2014; 13:929-34. [PMID: 23745581 DOI: 10.2174/15665240113139990048] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 04/28/2013] [Accepted: 05/28/2013] [Indexed: 11/22/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment in aging populations in industrialized countries. Here we investigated whether the genotype of vascular endothelial growth factor A (VEGFA) gene is associated with response to anti-VEGF therapy. 223 eyes with neovascular AMD were treated with intravitreal anti-VEGF therapy. Responders were defined as patients who had an improvement in best corrected visual acuity (BCVA) of at least 5 letters or one line on the EDTRS visual acuity chart along with resolution of intraretinal or subretinal fluid over 12 months. Patients who did not meet the definition of responders were classified as poor-responders. The vision of responders (n = 148) improved while the vision of poor-responders (n = 75) worsened (P<0.001). Responders on average had a decrease in central foveal thickness (CFT), while poor-responders had an increase in CFT (P <0.001). Compared with the responder group, the poor-responder group had a higher frequency of the risk (T) allele (Allelic P = 0.019) and TT genotype (P = 0.002 under a recessive model) for the VEGFA-rs943080 polymorphism. VEGFA expression was 1.8-fold higher in cells with the VEGFA rs943080 TT genotype than in cells with the VEGFA rs943080 CC genotype (P = 0.012). Age, gender, smoking, diabetes mellitus, and hypertension did not play a significant role in treatment response, but BMI was found to be significantly different between responders and poorresponders (P = 0.033). In conclusion, we demonstrated a potential pharmacogenetic relationship between the VEGFA gene and treatment response to anti-VEGF therapy.The studies are registered at ClinicalTrials.gov under the identifiers NCT00474695 (http://clinicaltrials. gov/ct2/show/NCT00474695) and NCT01464723 (http://clinicaltrials.gov/ct2/show/NCT01464723).
Collapse
Affiliation(s)
- L Zhao
- Department of Ophthalmology at Shiley Eye Center and Institute for Genomic Medicine, University of California-San Diego, 9415 Campus Point Drive #0946, La Jolla, CA 92093-0838, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Predictive value of VEGF A and VEGFR2 polymorphisms in the response to intravitreal ranibizumab treatment for wet AMD. Graefes Arch Clin Exp Ophthalmol 2014; 252:469-75. [PMID: 24522370 DOI: 10.1007/s00417-014-2585-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 12/11/2013] [Accepted: 01/28/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND To determine whether gene polymorphisms of the vascular endothelial growth factor A (VEGF A) and its receptor (VEGFR) influence the response to a variable-dosing treatment regimen with ranibizumab for age-related macular degeneration. METHODS This prospective cohort study included 94 patients (94 eyes) with exudative age-related macular degeneration (AMD) treated with ranibizumab. Patients underwent a 1-year treatment as in the Study of Ranibizumab in Patients with Subfoveal Choroidal Neovascularization Secondary to Age-Related Macular Degeneration (SUSTAIN). Injections were administered monthly during 3 months to all the patients diagnosed of neovascular AMD; reinjections were made when a patient lost 5 letters on the Early Treatment Diabetic Retinopathy Study chart or gained 100 μm in central subfield retinal thickness measured by OCT. Genotypes (VEGF A (rs 699947, rs833061) and VEGFR (rs 2071559)) were analyzed using TaqMan probes. Best-corrected visual acuity (BCVA), subjective improvement, and macular thickness measured with OCT values were compared with VEGF A and VEGFR genotypes. Multiple regression analysis was used to assess the statistical significance. RESULTS We found statistically significant differences in allelic distribution of VEGF A rs833061 polymorphism in relation with the response to intravitreal ranibizumab regarding to visual acuity improvement [p = 0,.34; OR: 1.619 (1.098-2.386)]. Patients carrying "protector" genotype CC had higher probability of best corrected visual acuity improvement. When we analyzed VEGF A rs699947 polymorphism we found that patients expressing AA genotype had a higher chance of increasing their best corrected visual acuity [p:0,022; OR 1,532 (1,015-2,313)]. We did not find statistically significant differences reagarding VEGFR rs2071559 polymorphism and treatment response. CONCLUSIONS Polymorphisms of VEGF A seem to influence the different response to antiangiogenic treatment in patients with AMD in our population, although further investigation is needed to know the mechanisms of this relationship.
Collapse
|
26
|
Klein R, Li X, Kuo JZ, Klein BEK, Cotch MF, Wong TY, Taylor KD, Rotter JI. Associations of candidate genes to age-related macular degeneration among racial/ethnic groups in the multi-ethnic study of atherosclerosis. Am J Ophthalmol 2013; 156:1010-1020.e1. [PMID: 23938121 DOI: 10.1016/j.ajo.2013.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE To describe the relationships of selected candidate genes to the prevalence of early age-related macular degeneration (AMD) in a cohort of whites, blacks, Hispanics, and Chinese Americans. DESIGN Cross-sectional study. METHODS setting: Multicenter study. study population: A total of 2456 persons aged 45-84 years with genotype information and fundus photographs. procedures: Twelve of 2862 single nucleotide polymorphisms (SNPs) from 11 of 233 candidate genes for cardiovascular disease were selected for analysis based on screening with marginal unadjusted P value <.001 within 1 or more racial/ethnic groups. Logistic regression models tested for association in case-control samples. main outcome measure: Prevalence of early AMD. RESULTS Early AMD was present in 4.0% of the cohort and varied from 2.4% in blacks to 6.0% in whites. The odds ratio increased from 2.3 for 1 to 10.0 for 4 risk alleles in a joint effect analysis of Age-Related Maculopathy Susceptibility 2 rs10490924 and Complement Factor H Y402H (P for trend = 4.2×10(-7)). Frequencies of each SNP varied among the racial/ethnic groups. Adjusting for age and other factors, few statistically significant associations of the 12 SNPs with AMD were consistent across all groups. In a multivariate model, most candidate genes did not attenuate the comparatively higher odds of AMD in whites. The higher frequency of risk alleles for several SNPs in Chinese Americans may partially explain their AMD frequency's approaching that of whites. CONCLUSIONS The relationships of 11 candidate genes to early AMD varied among 4 racial/ethnic groups, and partially explained the observed variations in early AMD prevalence among them.
Collapse
Affiliation(s)
- Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Cruz-González F, Cieza-Borrella C, Cabrillo-Estévez L, Cañete-Campos C, Escudero-Domínguez F, González-Sarmiento R. VEGF A (rs699947 and rs833061) and VEGFR2 (rs2071559) Gene Polymorphisms are not Associated with AMD Susceptibility in a Spanish Population. Curr Eye Res 2013; 38:1274-7. [DOI: 10.3109/02713683.2013.819926] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
28
|
Ansari M, McKeigue PM, Skerka C, Hayward C, Rudan I, Vitart V, Polasek O, Armbrecht AM, Yates JRW, Vatavuk Z, Bencic G, Kolcic I, Oostra BA, Van Duijn CM, Campbell S, Stanton CM, Huffman J, Shu X, Khan JC, Shahid H, Harding SP, Bishop PN, Deary IJ, Moore AT, Dhillon B, Rudan P, Zipfel PF, Sim RB, Hastie ND, Campbell H, Wright AF. Genetic influences on plasma CFH and CFHR1 concentrations and their role in susceptibility to age-related macular degeneration. Hum Mol Genet 2013; 22:4857-69. [PMID: 23873044 PMCID: PMC3820139 DOI: 10.1093/hmg/ddt336] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
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.
Collapse
Affiliation(s)
- Morad Ansari
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Agosta E, Lazzeri S, Orlandi P, Figus M, Fioravanti A, Di Desidero T, Sartini MS, Nardi M, Danesi R, Bocci G. Pharmacogenetics of antiangiogenic and antineovascular therapies of age-related macular degeneration. Pharmacogenomics 2013; 13:1037-53. [PMID: 22838951 DOI: 10.2217/pgs.12.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Age-related macular degeneration (AMD), the most common age-related disease causing irreversible visual loss in industrialized countries, is a complex and multifactorial illness. Researchers have found components of the complement alternative pathway inside drusen and Bruch's membrane of AMD patients, underlying a possible important role of complement factor H in the pathogenesis of AMD. The neovascular (wet) AMD is the most destructive form and it is characterized by invasion of new blood vessels into subretinal spaces with subsequent exudation and bleeding, resulting in scarring of the macular region and loss of the central vision. The hallmark of the neovascular form is the choroidal neovascularization, where VEGF-A has an important role in the pathogenesis of the disease. SNPs of these genes have recently been investigated as potential pharmacogenetic markers of the antiangiogenic and antineovascular therapy of AMD, which includes verteporfin photodynamic therapy and anti-VEGF-A drugs, such as pegaptanib, bevacizumab and ranibizumab. The CFH rs1061170 CT and TT genotypes have been associated with an improvement of visual acuity in bevacizumab or ranibizumab treated patients, whereas patients harboring VEGF-A rs699946 G allele responded better to bevacizumab-based therapy if compared with patients carrying the A allele. In conclusion, the discovery of pharmacogenetic markers for the personalization of the antiangiogenic and/or antineovascular therapy could be, in the future, a key issue in ophthalmology to obtain a personalization of the therapy and to avoid unnecessary costs and adverse drug reactions.
Collapse
Affiliation(s)
- Elisa Agosta
- Division of Pharmacology, Department of Internal Medicine, University of Pisa, Via Roma, 55-56125, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Conselho Brasileiro de Oftalmologia. Degeneração macular relacionada à idade. Rev Assoc Med Bras (1992) 2013; 59:106-11. [DOI: 10.1016/j.ramb.2012.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 08/12/2012] [Indexed: 11/26/2022] Open
|
31
|
Jünemann AGM, Stopa P, Michalke B, Chaudhri A, Reulbach U, Huchzermeyer C, Schlötzer-Schrehardt U, Kruse FE, Zrenner E, Rejdak R. Levels of aqueous humor trace elements in patients with non-exsudative age-related macular degeneration: a case-control study. PLoS One 2013; 8:e56734. [PMID: 23457607 PMCID: PMC3574106 DOI: 10.1371/journal.pone.0056734] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/14/2013] [Indexed: 11/18/2022] Open
Abstract
Trace elements might play a role in the complex multifactorial pathogenesis of age-related macular degeneration (AMD). The aim of this study was to measure alterations of trace elements levels in aqueous humor of patients with non-exsudative (dry) AMD. For this pilot study, aqueous humor samples were collected from patients undergoing cataract surgery. 12 patients with dry AMD (age 77.9±6.62, female 8, male 4) and 11 patients without AMD (age 66.6±16.7, female 7, male 4) were included. Aqueous levels of cadmium, cobalt, copper, iron, manganese, selenium, and zinc were measured by use of Flow-Injection-Inductively-Coupled-Plasma-Mass-Spectrometry (FI-ICP-MS), quality controlled with certified standards. Patients with AMD had significantly higher aqueous humor levels of cadmium (median: 0.70 µmol/L, IQR: 0.40–0.84 vs. 0.06 µmol/L; IQR: 0.01–.018; p = 0.002), cobalt (median: 3.1 µmol/L, IQR: 2.62–3.15 vs. 1.17 µmol/L; IQR: 0.95–1.27; p<0.001), iron (median: 311 µmol/L, IQR: 289–329 vs. 129 µmol/L; IQR: 111–145; p<0.001) and zinc (median: 23.1 µmol/L, IQR: 12.9–32.6 vs. 5.1 µmol/L; IQR: 4.4–9.4; p = 0.020) when compared with patients without AMD. Copper levels were significantly reduced in patients with AMD (median: 16.2 µmol/L, IQR: 11.4–31.3 vs. 49.9 µmol/L; IQR: 32.0–.142.0; p = 0.022) when compared to those without. No significant differences were observed in aqueous humor levels of manganese and selenium between patients with and without AMD. After an adjustment for multiple testing, cadmium, cobalt, copper and iron remained a significant factor in GLM models (adjusted for age and gender of the patients) for AMD. Alterations of trace element levels support the hypothesis that cadmium, cobalt, iron, and copper are involved in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Anselm G M Jünemann
- Department of Ophthalmology, University Hospital of Erlangen, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sun Y, Yu W, Huang L, Hou J, Gong P, Zheng Y, Zhao M, Zhou P, Li X. Is asthma related to choroidal neovascularization? PLoS One 2012; 7:e35415. [PMID: 22567103 PMCID: PMC3342271 DOI: 10.1371/journal.pone.0035415] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 03/15/2012] [Indexed: 11/29/2022] Open
Abstract
Background Age-related degeneration(AMD) and asthma are both diseases that are related to the activation of the complement system. The association between AMD and asthma has been debated in previous studies. The authors investigated the relationship between AMD and asthma systemically. Principal Findings The epidemiological study showed that asthma was related to choroidal neovascularization(CNV) subtype(OR = 1.721, P = 0.023). However, the meta-analysis showed there was no association between AMD and asthma. In an animal model, we found more fluoresce in leakage of CNV lesions by FA analysis and more angiogenesis by histological analysis in rats with asthma. Western blot demonstrated an elevated level of C3α-chain, C3α’-chain and VEGF. After compstatin was intravitreally injected, CNV leakage decreased according to FA analysis, with the level of C3 and VEGF protein decreasing at the same time. Significance This study first investigated the relationship between AMD and asthma systematically, and it was found that asthma could be a risk factor for the development of AMD. The study may provide a better understanding of the disease, which may advance the potential for screening asthma patients in clinical practice.
Collapse
Affiliation(s)
- Yaoyao Sun
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Wenzhen Yu
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Lvzhen Huang
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Jing Hou
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Peihua Gong
- Department of Respiration, Peking University People’s Hospital, Beijing, China
| | - Yi Zheng
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Peng Zhou
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
- Department of Ophthalmology, Eye and ENT, Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiaoxin Li
- Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
- * E-mail:
| |
Collapse
|
33
|
Jarrett SG, Boulton ME. Consequences of oxidative stress in age-related macular degeneration. Mol Aspects Med 2012; 33:399-417. [PMID: 22510306 DOI: 10.1016/j.mam.2012.03.009] [Citation(s) in RCA: 379] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 03/31/2012] [Indexed: 12/24/2022]
Abstract
The retina resides in an environment that is primed for the generation of reactive oxygen species (ROS) and resultant oxidative damage. The retina is one of the highest oxygen-consuming tissues in the human body. The highest oxygen levels are found in the choroid, but this falls dramatically across the outermost retina, creating a large gradient of oxygen towards the retina and inner segments of the photoreceptors which contain high levels of polyunsaturated fatty acids. This micro-environment together with abundant photosensitizers, visible light exposure and a high energy demand supports a highly oxidative milieu. However, oxidative damage is normally minimized by the presence of a range of antioxidant and efficient repair systems. Unfortunately, as we age oxidative damage increases, antioxidant capacity decreases and the efficiency of reparative systems become impaired. The result is retinal dysfunction and cell loss leading to visual impairment. It appears that these age-related oxidative changes are a hallmark of early age-related macular degeneration (AMD) which, in combination with hereditary susceptibility and other retinal modifiers, can progress to the pathology and visual morbidity associated with advanced AMD. This review reassesses the consequences of oxidative stress in AMD and strategies for preventing or reversing oxidative damage in retinal tissues.
Collapse
Affiliation(s)
- Stuart G Jarrett
- Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
34
|
|
35
|
Synowiec E, Blasiak J, Zaras M, Szaflik J, Szaflik JP. Association between polymorphisms of the DNA base excision repair genes MUTYH and hOGG1 and age-related macular degeneration. Exp Eye Res 2012; 98:58-66. [PMID: 22469746 DOI: 10.1016/j.exer.2012.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 01/10/2012] [Accepted: 02/15/2012] [Indexed: 12/13/2022]
Abstract
Age-Related Macular Degeneration (AMD) is an eye disease that results in progressive and irreversible loss of central vision and is considered as the primary cause of visual impairment, including blindness, in the elderly in industrialized countries. Oxidative stress has been implicated in the pathogenesis of AMD. The hOGG1 and the MUTYH genes play an important role in the repair of oxidatively damaged DNA in the base excision repair pathway. The DNA glycosylases encoded by the hOGG1 and MUTYH genes initiate this pathway by recognizing and removing 8-oxoguanine and adenine paired with 8-oxoguanine, respectively. Our study was designed to examine the association between the c.977C>G polymorphism (rs1052133) of the hOGG1 gene and the c.972G>C polymorphism (rs3219489) of the MUTYH gene and AMD as well as the modulation of this association by some clinical and lifestyle factors. Genotypes were determined in DNA from blood of 271 AMD patients, including 101 with wet and 170 with dry form of the disease and 105 sex- and age-matched individuals without AMD. We observed an association between AMD, dry and wet forms of AMD and the C/G genotype and the G allele of the c.977C>G-hOGG1 polymorphism (p 0.006; 0.009; 0.021 and 0.004; 0.005; 0.016 respectively). On the other hand, the C/C genotype and the C allele reduced the risk of AMD as well as of its dry form or wet form (p 0.002; 0.003; 0.010 and 0.004; 0.005; 0.016, respectively). Therefore, the associations we detected were driven by the dry AMD. We observed some statistically significant association between the occurrence of AMD and its dry and wet forms and genotypes of the other polymorphism, the c.972G>C-MUTYH polymorphism, but due to borderline character of all this association we do not consider them as medically relevant. Our findings suggest that the c.977C>G-hOGG1 polymorphism may be associated with dry AMD. Further studies are needed to determine possible association between AMD and the c.972G>C-MUTYH polymorphism.
Collapse
Affiliation(s)
- Ewelina Synowiec
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | | | | | | | | |
Collapse
|
36
|
Wysokinski D, Zaras M, Dorecka M, Waszczyk M, Szaflik J, Blasiak J, Szaflik JP. An association between environmental factors and the IVS4+44C>A polymorphism of the DMT1 gene in age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol 2012; 250:1057-65. [PMID: 22371024 PMCID: PMC3382657 DOI: 10.1007/s00417-012-1966-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 02/01/2012] [Accepted: 02/03/2012] [Indexed: 12/14/2022] Open
Abstract
Background Age-related macular degeneration (AMD) is an ocular disease affecting macula — the central part of the retina, resulting in the degeneration of photoreceptors and retinal epithelium and causing severe central vision impairment. The pathophysiology of the disease is not completely known, but a significant role is attributed to genetic factors. The contribution of oxidative stress in AMD as a trigger of the degenerative process is well-established. Iron ions may act as a source of reactive oxygen species; therefore, maintaining iron homeostasis is important for redox balance in the organism. Diversity in iron homeostasis genes may counterpart in unbalanced redox state, and thus be involved in AMD pathophysiology. Methods In this work, we searched for an association between some single nucleotide polymorphisms in the divalent metal transporter 1 (DMT1) gene intronic IVS4+44C>A (rs224589) and 3’-UTR c.2044T>C (rs2285230) and environmental factors and AMD. Genotyping was performed using the PCR-RFLP method. DNA was obtained from 436 AMD patients and 168 controls. Results We did not find any association between the genotypes of the two polymorphisms and AMD occurrence. However, we observed that AMD patients living in a rural environment and having the CC genotype of the IVS4+44C>A polymorphism had an increased risk of AMD, while individuals with the CA genotype or the A allele had a decreased risk of the disease. Moreover, in male AMD patients the C allele increased the risk of the disease, while the AA genotype decreased it. Conclusions These results suggest that the VS4+44C>A polymorphism of the DMT1 gene may interact with place of living and gender to modulate the risk of AMD.
Collapse
Affiliation(s)
- Daniel Wysokinski
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, Lodz, Poland
| | | | | | | | | | | | | |
Collapse
|
37
|
Synowiec E, Pogorzelska M, Blasiak J, Szaflik J, Szaflik JP. Genetic polymorphism of the iron-regulatory protein-1 and -2 genes in age-related macular degeneration. Mol Biol Rep 2012; 39:7077-87. [PMID: 22331484 PMCID: PMC3334491 DOI: 10.1007/s11033-012-1539-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Accepted: 01/24/2012] [Indexed: 12/24/2022]
Abstract
Iron can be involved in the pathogenesis of AMD through the oxidative stress because it may catalyze the Haber–Weiss and Fenton reactions converting hydrogen peroxide to free radicals, which can induce cellular damage. We hypothesized that genetic polymorphism in genes related to iron metabolism may predispose individuals to the development of AMD and therefore we checked for an association between the g.32373708 G>A polymorphism (rs867469) of the IRP1 gene and the g.49520870 G>A (rs17483548) polymorphism of the IRP2 gene and AMD risk as well as the modulation of this association by some environmental and life-style factors. Genotypes were determined in DNA from blood of 269 AMD patients and 116 controls by the allele-specific oligonucleotide-restriction fragment length polymorphism and the polymerase chain reaction-restriction fragment length polymorphism. An association between AMD, dry and wet forms of AMD and the G/G genotype of the g.32373708 G>A-IRP1 polymorphism was found (OR 3.40, 4.15, and 2.75). On the other hand, the G/A genotype reduced the risk of AMD as well as its dry or wet form (OR 0.23, 0.21, 0.26). Moreover, the G allele of the g.49520870 G>A-IRP2 polymorphism increased the risk of the dry form of the disease (OR 1.51) and the A/A genotype and the A allele decreased such risk (OR 0.43 and 0.66). Our data suggest that the g.32373708 G>A-IRP1 and g.49520870 G>A-IRP2 polymorphisms may be associated with increased risk for AMD.
Collapse
Affiliation(s)
- Ewelina Synowiec
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | | | | | | | | |
Collapse
|
38
|
Audo I, Bujakowska KM, Léveillard T, Mohand-Saïd S, Lancelot ME, Germain A, Antonio A, Michiels C, Saraiva JP, Letexier M, Sahel JA, Bhattacharya SS, Zeitz C. Development and application of a next-generation-sequencing (NGS) approach to detect known and novel gene defects underlying retinal diseases. Orphanet J Rare Dis 2012; 7:8. [PMID: 22277662 PMCID: PMC3352121 DOI: 10.1186/1750-1172-7-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 01/25/2012] [Indexed: 12/25/2022] Open
Abstract
Background Inherited retinal disorders are clinically and genetically heterogeneous with more than 150 gene defects accounting for the diversity of disease phenotypes. So far, mutation detection was mainly performed by APEX technology and direct Sanger sequencing of known genes. However, these methods are time consuming, expensive and unable to provide a result if the patient carries a new gene mutation. In addition, multiplicity of phenotypes associated with the same gene defect may be overlooked. Methods To overcome these challenges, we designed an exon sequencing array to target 254 known and candidate genes using Agilent capture. Subsequently, 20 DNA samples from 17 different families, including four patients with known mutations were sequenced using Illumina Genome Analyzer IIx next-generation-sequencing (NGS) platform. Different filtering approaches were applied to identify the genetic defect. The most likely disease causing variants were analyzed by Sanger sequencing. Co-segregation and sequencing analysis of control samples validated the pathogenicity of the observed variants. Results The phenotype of the patients included retinitis pigmentosa, congenital stationary night blindness, Best disease, early-onset cone dystrophy and Stargardt disease. In three of four control samples with known genotypes NGS detected the expected mutations. Three known and five novel mutations were identified in NR2E3, PRPF3, EYS, PRPF8, CRB1, TRPM1 and CACNA1F. One of the control samples with a known genotype belongs to a family with two clinical phenotypes (Best and CSNB), where a novel mutation was identified for CSNB. In six families the disease associated mutations were not found, indicating that novel gene defects remain to be identified. Conclusions In summary, this unbiased and time-efficient NGS approach allowed mutation detection in 75% of control cases and in 57% of test cases. Furthermore, it has the possibility of associating known gene defects with novel phenotypes and mode of inheritance.
Collapse
|
39
|
Nguyen ATH, Campbell M, Kenna PF, Kiang AS, Tam L, Humphries MM, Humphries P. Calpain and photoreceptor apoptosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 723:547-52. [PMID: 22183376 DOI: 10.1007/978-1-4614-0631-0_69] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Anh T H Nguyen
- The Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
40
|
Stanton CM, Yates JRW, den Hollander AI, Seddon JM, Swaroop A, Stambolian D, Fauser S, Hoyng C, Yu Y, Atsuhiro K, Branham K, Othman M, Chen W, Kortvely E, Chalmers K, Hayward C, Moore AT, Dhillon B, Ueffing M, Wright AF. Complement factor D in age-related macular degeneration. Invest Ophthalmol Vis Sci 2011; 52:8828-34. [PMID: 22003108 DOI: 10.1167/iovs.11-7933] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
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.
Collapse
Affiliation(s)
- Chloe M Stanton
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bitoun P, Pipiras E, Rigaudiere F. Congenital macular dystrophy, corpus callosum agenesis, hippocampi hypoplasia--a novel neuro-ophthalmic syndrome: case report. Ophthalmic Genet 2011; 33:39-43. [PMID: 21834622 DOI: 10.3109/13816810.2011.596892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
INTRODUCTION Macular dystrophy is a cause of childhood and adult visual handicap and has been associated with multiple gene defects. Syndromic macular dystrophy is rare and a novel congenital form of syndromic macular dystrophy is presented. The authors report on a consanguineous family in which the 5-year-old female proband presented with nystagmus and low vision due to congenital macular dystrophy visible on fundus examination associated with complete corpus callosum agenesis, hippocampi hypoplasia and recurrent illnesses. MATERIALS AND METHODS Patients signed informed consent forms to participate in the research. Proband was screened for 18 recessive macular dystrophy genes and ABCA4 and had a G banded karyotype on peripheral blood lymphocytes. Patients were evaluated using ocular biomicrosopy, fluorescein retinal angiograms, electroretinograms, visual evoked potentials, retinal optical coherence tomography, brain MRI and multifocal electroretinograms. RESULTS The older brother presented with subclinical findings of bilateral absence of foveal macular peak on multifocal electroretinograms and minimal corpus callosum hypoplasia. The younger sister was recently discovered to have a similar macular dystrophy. The father showed subclinical unilateral decreased foveal macular peak and the mother showed a granular-appearing fundus. No mutations were identified in the RP and macular dystrophy genes screened. DISCUSSION A review of the literature confirms that this is the first report of a congenital and possibly developmental macular dystrophy, with neurologic syndromic features and possible autosomal recessive inheritance but varying penetrance.
Collapse
Affiliation(s)
- Pierre Bitoun
- Génétique Médicale, Hôpital Jean Verdier AP-HP, C.H.U. Paris Nord, Bondy, France.
| | | | | |
Collapse
|
42
|
Chen Y, Bedell M, Zhang K. Age-related macular degeneration: genetic and environmental factors of disease. Mol Interv 2011; 10:271-81. [PMID: 21045241 DOI: 10.1124/mi.10.5.4] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Age-related macular degeneration (AMD) is the most common cause of visual impairment among the elderly in developed countries, and its prevalence is thus increasing as the population ages; however, treatment options remain limited because the etiology and pathogenesis of AMD are incompletely defined. Recently, much progress has been made in gene discovery and mechanistic studies, which clearly indicate that AMD involves the interaction of multiple genetic and environmental factors. The identification of genes that have a substantial impact on the risk for AMD is not only facilitating the diagnosis and screening of populations at risk but is also elucidating key molecular pathways of pathogenesis. Pharmacogenetic studies of treatment responsiveness among patients with the "wet" form of AMD are increasingly proving to be clinically relevant; pharmacogenetic approaches hold great promise for both identifying patients with the best chance for vision recovery as well as tailoring individualized therapies.
Collapse
Affiliation(s)
- Yuhong Chen
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China
| | | | | |
Collapse
|
43
|
Bruban J, Maoui A, Chalour N, An N, Jonet L, Feumi C, Tréton J, Sennlaub F, Behar-Cohen F, Mascarelli F, Dinet V. CCR2/CCL2-mediated inflammation protects photoreceptor cells from amyloid-β-induced apoptosis. Neurobiol Dis 2011; 42:55-72. [PMID: 21220018 DOI: 10.1016/j.nbd.2011.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 12/22/2010] [Accepted: 01/02/2011] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration is characterized by the formation of drusen containing amyloid-β (Aβ) and the degeneration of photoreceptors. To explore the largely unknown role of Aβ in the retina, we investigated the effects on photoreceptors of the oligomeric form of Aβ(1-42). Subretinal injection of the Aβ peptide induced misplaced expression of recoverin and synaptophysin in the photoreceptors, oxidative stress in their inner and outer segments, and finally apoptosis. Aβ did not induce cell death in purified photoreceptor cell cultures, but did so in retinal cell cultures, thereby suggesting that the cellular environment plays a role in Aβ-induced photoreceptor apoptosis. Subretinal injection of Aβ was followed by activation and migration of microglial cells and then by photoreceptor apoptosis. Microglial cells phagocytosed rhodopsin-containing debris and Aβ in the subretinal space. Quantitative RT-PCR allowed us to identify a specific gene expression profile associated with the Aβ-induced progression of retinal degeneration and consistent with oxidative stress, inflammation, and an apoptotic program. The gene most highly upregulated in Aβ-injected retinas was that for the chemokine CCL2, and its absence or that of its cognate receptor CCR2 greatly reduced migration of activated microglial cells to the site of retinal injury and profoundly worsened photoreceptor degeneration and disorganization of the retinal pigment epithelium in Aβ-injected retinas. Our study pinpoints the roles of Aβ and of CCL2/CCR2 axis-dependent inflammation in photoreceptor apoptosis.
Collapse
Affiliation(s)
- Julien Bruban
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris6, UMRS 872, F-75006 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Molday RS, Zhang K. Defective lipid transport and biosynthesis in recessive and dominant Stargardt macular degeneration. Prog Lipid Res 2010; 49:476-92. [PMID: 20633576 DOI: 10.1016/j.plipres.2010.07.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Stargardt disease is a common inherited macular degeneration characterized by a significant loss in central vision in the first or second decade of life, bilateral atrophic changes in the central retina associated with degeneration of photoreceptors and underlying retinal pigment epithelial cells, and the presence of yellow flecks extending from the macula. Autosomal recessive Stargardt disease, the most common macular dystrophy, is caused by mutations in the gene encoding ABCA4, a photoreceptor ATP binding cassette (ABC) transporter. Biochemical studies together with analysis of abca4 knockout mice and Stargardt patients have implicated ABCA4 as a lipid transporter that facilitates the removal of potentially toxic retinal compounds from photoreceptors following photoexcitation. An autosomal dominant form of Stargardt disease also known as Stargardt-like dystrophy is caused by mutations in a gene encoding ELOVL4, an enzyme that catalyzes the elongation of very long-chain fatty acids in photoreceptors and other tissues. This review focuses on the molecular characterization of ABCA4 and ELOVL4 and their role in photoreceptor cell biology and the pathogenesis of Stargardt disease.
Collapse
Affiliation(s)
- Robert S Molday
- Department of Biochemistry and Molecular Biology, Centre of Macular Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada.
| | | |
Collapse
|
45
|
Gnana-Prakasam JP, Martin PM, Smith SB, Ganapathy V. Expression and function of iron-regulatory proteins in retina. IUBMB Life 2010; 62:363-70. [PMID: 20408179 DOI: 10.1002/iub.326] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Iron is essential for cell survival and function; yet excess iron is toxic to cells. Therefore, the cellular and whole-body levels of iron are regulated exquisitely. At least a dozen proteins participate in the regulation of iron homeostasis. Hemochromatosis, a genetic disorder of iron overload, is caused by mutations in at least five genes, namely HFE, hemojuvelin, Transferrin receptor 2, ferroportin, and hepcidin. Retina is separated from systemic circulation by inner and outer blood-retinal barriers; therefore it is widely believed that this tissue is immune to changes in systemic circulation. Even though hemochromatosis is associated with iron overload and dysfunction of a variety of systemic organs, little is known on the effects of this disease on the retina. Recent studies have shown that all five genes that are associated with hemochromatosis are expressed in the retina in a cell type-specific manner. The retinal pigment epithelium, which forms the outer blood-retinal barrier, expresses all of these five genes. It is therefore clearly evident that iron homeostasis in the retina is maintained locally by active participation of various iron-regulatory proteins. Excess iron is detrimental to the retina as evidenced from human studies and from mouse models of iron overload. Retinal iron homeostasis is disrupted in various clinical conditions such as hemochromatosis, aceruloplasminemia, age-related macular degeneration, and bacterial and viral infections.
Collapse
Affiliation(s)
- Jaya P Gnana-Prakasam
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | |
Collapse
|
46
|
Gehrs KM, Jackson JR, Brown EN, Allikmets R, Hageman GS. Complement, age-related macular degeneration and a vision of the future. ACTA ACUST UNITED AC 2010; 128:349-58. [PMID: 20212207 DOI: 10.1001/archophthalmol.2010.18] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is one of the most well-characterized late-onset, complex trait diseases. Remarkable advances in our understanding of the genetic and biological foundations of this disease were derived from a recent convergence of scientific and clinical data. Importantly, the more recent identification of AMD-associated variations in a number of complement pathway genes has provided strong support for earlier, paradigm-shifting studies that suggested that aberrant function of the complement system plays a key role in disease etiology. Collectively, this wealth of information has provided an impetus for the development of powerful tools to accurately diagnose disease risk and progression and complement-based therapeutics that will ultimately delay or prevent AMD. Indeed, we are poised to witness a new era of a personalized approach toward the assessment, management, and treatment of this debilitating, chronic disease.
Collapse
Affiliation(s)
- Karen M Gehrs
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, USA
| | | | | | | | | |
Collapse
|
47
|
Ayala-Haedo JA, Gallins PJ, Whitehead PL, Schwartz SG, Kovach JL, Postel EA, Agarwal A, Wang G, Haines JL, Pericak-Vance MA, Scott WK. Analysis of single nucleotide polymorphisms in the NOS2A gene and interaction with smoking in age-related macular degeneration. Ann Hum Genet 2010; 74:195-201. [PMID: 20374233 DOI: 10.1111/j.1469-1809.2010.00570.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Age-related macular degeneration (AMD) is a complex degenerative retinal disease influenced by both genetic and environmental risk factors. We assessed whether single nucleotide polymorphisms (SNPs) in the NOS2A gene increase risk and modulate the effect of smoking in AMD. 998 Caucasian subjects (712 AMD cases and 286 controls) were genotyped for 17 SNPs in NOS2A. Multivariable logistic regression models containing SNP genotypes, age, sex, smoking status and genotype/smoking interaction were constructed. SNP rs8072199 was significantly associated with AMD (OR = 1.3; 95% CI : 1.02, 1.65; P = 0.035). A significant interaction with smoking was detected at rs2248814 (P = 0.037). Stratified data by genotypes demonstrated that the association between AMD and smoking was stronger in carriers of AA genotypes (OR = 35.98; 95% CI: 3.19, 405.98) than in carriers of the AG genotype (OR = 3.05; 95% CI: 1.36, 6.74) or GG genotype (OR = 2.1; 95% CI: 0.91, 4.84). The results suggest a possible synergistic interaction of AA genotype with smoking, although the result bears replication in larger samples. Our data suggests that SNPs in the NOS2A gene are associated with increased risk for AMD and might modulate the effect of smoking on AMD.
Collapse
Affiliation(s)
- Juan A Ayala-Haedo
- John P. Hussman Institute for Human Genomics and the Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Anderson DH, Radeke MJ, Gallo NB, Chapin EA, Johnson PT, Curletti CR, Hancox LS, Hu J, Ebright JN, Malek G, Hauser MA, Rickman CB, Bok D, Hageman GS, Johnson LV. The pivotal role of the complement system in aging and age-related macular degeneration: hypothesis re-visited. Prog Retin Eye Res 2009; 29:95-112. [PMID: 19961953 DOI: 10.1016/j.preteyeres.2009.11.003] [Citation(s) in RCA: 565] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During the past ten years, dramatic advances have been made in unraveling the biological bases of age-related macular degeneration (AMD), the most common cause of irreversible blindness in western populations. In that timeframe, two distinct lines of evidence emerged which implicated chronic local inflammation and activation of the complement cascade in AMD pathogenesis. First, a number of complement system proteins, complement activators, and complement regulatory proteins were identified as molecular constituents of drusen, the hallmark extracellular deposits associated with early AMD. Subsequently, genetic studies revealed highly significant statistical associations between AMD and variants of several complement pathway-associated genes including: Complement factor H (CFH), complement factor H-related 1 and 3 (CFHR1 and CFHR3), complement factor B (CFB), complement component 2 (C2), and complement component 3 (C3). In this article, we revisit our original hypothesis that chronic local inflammatory and immune-mediated events at the level of Bruch's membrane play critical roles in drusen biogenesis and, by extension, in the pathobiology of AMD. Secondly, we report the results of a new screening for additional AMD-associated polymorphisms in a battery of 63 complement-related genes. Third, we identify and characterize the local complement system in the RPE-choroid complex - thus adding a new dimension of biological complexity to the role of the complement system in ocular aging and AMD. Finally, we evaluate the most salient, recent evidence that bears directly on the role of complement in AMD pathogenesis and progression. Collectively, these recent findings strongly re-affirm the importance of the complement system in AMD. They lay the groundwork for further studies that may lead to the identification of a transcriptional disease signature of AMD, and hasten the development of new therapeutic approaches that will restore the complement-modulating activity that appears to be compromised in genetically susceptible individuals.
Collapse
Affiliation(s)
- Don H Anderson
- Center for the Study of Macular Degeneration, Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Janik-Papis K, Zaras M, Krzyzanowska A, Wozniak K, Blasiak J, Szaflik J, Szaflik JP. Association between vascular endothelial growth factor gene polymorphisms and age-related macular degeneration in a Polish population. Exp Mol Pathol 2009; 87:234-8. [DOI: 10.1016/j.yexmp.2009.09.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 09/07/2009] [Accepted: 09/08/2009] [Indexed: 10/20/2022]
|
50
|
Parekh N, Voland RP, Moeller SM, Blodi BA, Ritenbaugh C, Chappell RJ, Wallace RB, Mares JA. Association between dietary fat intake and age-related macular degeneration in the Carotenoids in Age-Related Eye Disease Study (CAREDS): an ancillary study of the Women's Health Initiative. ACTA ACUST UNITED AC 2009; 127:1483-93. [PMID: 19901214 DOI: 10.1001/archophthalmol.2009.130] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To evaluate the relationships between the amount and type of dietary fat and intermediate age-related macular degeneration (AMD). DESIGN Women aged 50 to 79 years with high and low lutein intake from 3 sites of the Women's Health Initiative Observational Study were recruited into the Carotenoids in Age-Related Eye Disease Study. Fat intake from 1994 through 1998 was estimated using food frequency questionnaires, and AMD was assessed photographically from 2001 through 2004. RESULTS Intakes of omega-6 and omega-3 polyunsaturated fatty acids, which were highly correlated (r = 0.8), were associated with approximately 2-fold higher prevalence of intermediate AMD in high vs low quintiles. However, monounsaturated fatty acid intake was associated with lower prevalence. Age interactions were often observed. In women younger than 75 years (n = 1325), total fat and saturated fatty acid intakes were associated with increased prevalence of AMD (multivariate adjusted odds ratios [95% confidence interval] for intermediate AMD, 1.7 [1.0-2.7] for quintile 5 vs quintile 1 for total fat [P = .10 for trend] and 1.6 [0.7-3.6] for saturated fatty acids [P = .23 for trend]). The associations were reversed in older women. CONCLUSIONS These results support a growing body of evidence suggesting that diets high in several types of fat may contribute to the risk of intermediate AMD and that diets high in monounsaturated fatty acids may be protective.
Collapse
Affiliation(s)
- Niyati Parekh
- Department of Nutrition, Food Studies, and Public Health, New York University, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|