101
|
Ying Y, Ueta T, Jiang S, Lin H, Wang Y, Vavvas D, Wen R, Chen YG, Luo Z. Metformin inhibits ALK1-mediated angiogenesis via activation of AMPK. Oncotarget 2018; 8:32794-32806. [PMID: 28427181 PMCID: PMC5464828 DOI: 10.18632/oncotarget.15825] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/12/2017] [Indexed: 11/25/2022] Open
Abstract
Anti-VEGF therapy has been proven to be effective in the treatment of pathological angiogenesis. However, therapy resistance often occurs, leading to development of alternative approaches. The present study examines if AMPK negatively regulates ALK1-mediated signaling events and associated angiogenesis. Thus, we treated human umbilical vein endothelial cells with metformin as well as other pharmacological AMPK activators and showed that activation of AMPK inhibited Smad1/5 phosphorylation and tube formation induced by BMP9. This event was mimicked by expression of the active mutant of AMPKα1 and prevented by the dominant negative AMPKα1. Metformin inhibition of BMP9 signaling is possibly mediated by upregulation of Smurf1, leading to degradation of ALK1. Furthermore, metformin suppressed BMP9-induced angiogenesis in mouse matrigel plug. In addition, laser photocoagulation was employed to evaluate the effect of metformin. The data revealed that metformin significantly reduced choroidal neovascularization to a level comparable to LDN212854, an ALK1 specific inhibitor. In conjunction, metformin diminished expression of ALK1 in endothelium of the lesion area. Collectively, our study for the first time demonstrates that AMPK inhibits ALK1 and associated angiogenesis/neovascularization. This may offer us a new avenue for the treatment of related diseases using clinically used pharmacological AMPK activators like metformin in combination with other strategies to enhance the treatment efficacy or in the case of anti-VEGF resistance.
Collapse
Affiliation(s)
- Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Takashi Ueta
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Shanshan Jiang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Yuanyuan Wang
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Demetrios Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami Miller Medical School, Miami, FL, USA
| | - Ye-Guang Chen
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing, China
| | - Zhijun Luo
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.,Windsor University School of Medicine, Brighton's Estate, Cayon, St. Kitts
| |
Collapse
|
102
|
'Statins in retinal disease'. Eye (Lond) 2018; 32:981-991. [PMID: 29556012 DOI: 10.1038/s41433-018-0066-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/05/2018] [Accepted: 02/12/2018] [Indexed: 01/14/2023] Open
Abstract
Statins are known for their blood cholesterol-lowering effect and are widely used in patients with cardiovascular and metabolic diseases. Research over the past three decades shows that statins have diverse effects on different pathophysiological pathways involved in angiogenesis, inflammation, apoptosis, and anti-oxidation, leading to new therapeutic options. Recently, statins have attracted considerable attention for their immunomodulatory effect. Since immune reactivity has been implicated in a number of retinal diseases, such as uveitis, age-related macular degeneration (AMD) and diabetic retinopathy, there is now a growing body of evidence supporting the beneficial effects of statins in these retinopathies. This review evaluates the relationship between statins and the pathophysiological basis of these diseases, focusing on their potential role in treatment. A PubMed database search and literature review was conducted. Among AMD patients, there is inconsistent evidence regarding protection against development of early AMD or delaying disease progression; though they have been found to reduce the risk of developing choroidal neovascular membranes (CNV). In patients with retinal vein occlusion, there was no evidence to support a therapeutic benefit or a protective role with statins. In patients with diabetic retinopathy, statins demonstrate a reduction in disease progression and improved resolution of diabetic macular oedema (DMO). Among patients with uveitis, statins have a protective effect by reducing the likelihood of uveitis development.
Collapse
|
103
|
Inana G, Murat C, An W, Yao X, Harris IR, Cao J. RPE phagocytic function declines in age-related macular degeneration and is rescued by human umbilical tissue derived cells. J Transl Med 2018. [PMID: 29534722 PMCID: PMC5851074 DOI: 10.1186/s12967-018-1434-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Age-related macular degeneration (AMD) is a leading cause of blindness among the elderly characterized by retinal pigment epithelium (RPE) degeneration with accumulation of abnormal intracellular deposits (lipofuscin) and photoreceptor death. RPE is vital for the retina and integrity of photoreceptors through its phagocytic function which is closely linked to formation of lipofuscin through daily phagocytosis of discarded photoreceptor outer segments (POS). Although phagocytosis has been implicated in AMD, it has not been directly shown to be altered in AMD. RPE phagocytic defect was previously shown to be rescued by subretinal injection of human umbilical tissue derived cells (hUTC) in a rodent model of retinal degeneration (RCS rat) through receptor tyrosine kinase (RTK) ligands and bridge molecules. Here, we examined RPE phagocytic function directly in the RPE from AMD patients and the ability and mechanisms of hUTC to affect phagocytosis in the human RPE. Methods Human RPE was isolated from the post-mortem eyes of normal and AMD-affected subjects and cultured. RPE phagocytic function was measured in vitro using isolated POS. The effects of hUTC conditioned media, recombinant RTK ligands brain-derived neurotrophic factor (BDNF), hepatocyte growth factor (HGF), and glial cell-derived neurotrophic factor (GDNF), as well as bridge molecules milk-fat-globule-EGF-factor 8 (MFG-E8), thrombospondin (TSP)-1, and TSP-2 on phagocytosis were also examined in phagocytosis assays using isolated POS. RNA was isolated from normal and AMD RPE treated with hUTC conditioned media and subjected to transcriptome profiling by RNA-Seq and computational analyses. Results RPE phagocytosis, while showing a moderate decline with age, was significantly reduced in AMD RPE, more than expected for age. hUTC conditioned media stimulated phagocytosis in the normal human RPE and significantly rescued the phagocytic dysfunction in the AMD RPE. RTK ligands and bridge molecules duplicated the rescue effect. Moreover, multiple molecular pathways involving phagocytosis, apoptosis, oxidative stress, inflammation, immune activation, and cholesterol transport were affected by hUTC in the RPE. Conclusions We demonstrated for the first time RPE phagocytic dysfunction in AMD, highlighting its likely importance in AMD, and the ability of hUTC to correct this dysfunction, providing insights into the therapeutic potential of hUTC for AMD. Electronic supplementary material The online version of this article (10.1186/s12967-018-1434-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- George Inana
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, 1638 N.W. 10th Avenue, Miami, FL, 33136, USA.
| | - Christopher Murat
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, 1638 N.W. 10th Avenue, Miami, FL, 33136, USA
| | - Weijun An
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, 1638 N.W. 10th Avenue, Miami, FL, 33136, USA
| | - Xiang Yao
- Janssen Research & Development, LLC, San Diego, CA, 92121, USA
| | - Ian R Harris
- Janssen Research & Development, LLC, Spring House, PA, 19477, USA
| | - Jing Cao
- Janssen Research & Development, LLC, Spring House, PA, 19477, USA.
| |
Collapse
|
104
|
Choudhary M, Safe S, Malek G. Suppression of aberrant choroidal neovascularization through activation of the aryl hydrocarbon receptor. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1583-1595. [PMID: 29481912 DOI: 10.1016/j.bbadis.2018.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand activated transcription factor, initially discovered for its role in regulating xenobiotic metabolism. There is extensive evidence supporting a multi-faceted role for AhR, modulating physiological pathways important in cell health and disease. Recently we demonstrated that the AhR plays a role in the pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. We found that loss of AhR exacerbates choroidal neovascular (CNV) lesion formation in a murine model. Herein we tested the therapeutic impact of AhR activation on CNV lesion formation and factors associated with aberrant neovascularization. We screened a panel of synthetic drugs and endogenous AhR ligands, assessed their ability to activate AhR in choroidal endothelial cells, and inhibit angiogenesis in vitro. Drugs with an anti-angiogenic profile were then administered to a murine model of CNV. Two compounds, leflunomide and flutamide, significantly inhibited CNV formation concurrent with positive modifying effects on angiogenesis, inflammation, extracellular matrix remodeling, and fibrosis. These results validate the role of the AhR pathway in regulating CNV pathogenesis, identify mechanisms of AhR-based therapies in the eye, and argue in favor of developing AhR as a drug target for the treatment of neovascular AMD.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Stephen Safe
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
105
|
Laíns I, Kelly RS, Miller JB, Silva R, Vavvas DG, Kim IK, Murta JN, Lasky-Su J, Miller JW, Husain D. Human Plasma Metabolomics Study across All Stages of Age-Related Macular Degeneration Identifies Potential Lipid Biomarkers. Ophthalmology 2018; 125:245-254. [PMID: 28916333 PMCID: PMC8077680 DOI: 10.1016/j.ophtha.2017.08.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To characterize the plasma metabolomic profile of patients with age-related macular degeneration (AMD) using mass spectrometry (MS). DESIGN Cross-sectional observational study. PARTICIPANTS We prospectively recruited participants with a diagnosis of AMD and a control group (>50 years of age) without any vitreoretinal disease. METHODS All participants underwent color fundus photography, used for AMD diagnosis and staging, according to the Age-Related Eye Disease Study classification scheme. Fasting blood samples were collected and plasma was analyzed by Metabolon, Inc. (Durham, NC), using ultrahigh-performance liquid chromatography (UPLC) and high-resolution MS. Metabolon's hardware and software were used to identify peaks and control quality. Principal component analysis and multivariate regression were performed to assess differences in the metabolomic profiles of AMD patients versus controls, while controlling for potential confounders. For biological interpretation, pathway enrichment analysis of significant metabolites was performed using MetaboAnalyst. MAIN OUTCOME MEASURES The primary outcome measures were levels of plasma metabolites in participants with AMD compared with controls and among different AMD severity stages. RESULTS We included 90 participants with AMD (30 with early AMD, 30 with intermediate AMD, and 30 with late AMD) and 30 controls. Using UPLC and MS, 878 biochemicals were identified. Multivariate logistic regression identified 87 metabolites with levels that differed significantly between AMD patients and controls. Most of these metabolites (82.8%; n = 72), including the most significant metabolites, belonged to the lipid pathways. Analysis of variance revealed that of the 87 metabolites, 48 (55.2%) also were significantly different across the different stages of AMD. A significant enrichment of the glycerophospholipids pathway was identified (P = 4.7 × 10-9) among these metabolites. CONCLUSIONS Participants with AMD have altered plasma metabolomic profiles compared with controls. Our data suggest that the most significant metabolites map to the glycerophospholipid pathway. These findings have the potential to improve our understanding of AMD pathogenesis, to support the development of plasma-based metabolomics biomarkers of AMD, and to identify novel targets for treatment of this blinding disease.
Collapse
Affiliation(s)
- Inês Laíns
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Association for Innovation and Biomedical Research on Light, Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Rachel S Kelly
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - John B Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Rufino Silva
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Association for Innovation and Biomedical Research on Light, Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Demetrios G Vavvas
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ivana K Kim
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Joaquim N Murta
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Association for Innovation and Biomedical Research on Light, Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Jessica Lasky-Su
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joan W Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Deeba Husain
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
106
|
Wykoff CC, Clark WL, Nielsen JS, Brill JV, Greene LS, Heggen CL. Optimizing Anti-VEGF Treatment Outcomes for Patients with Neovascular Age-Related Macular Degeneration. J Manag Care Spec Pharm 2018; 24:S3-S15. [PMID: 29383980 PMCID: PMC10408401 DOI: 10.18553/jmcp.2018.24.2-a.s3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The introduction of anti-vascular endothelial growth factor (anti-VEGF) drugs to ophthalmology has revolutionized the treatment of neovascular age-related macular degeneration (nAMD). Despite this significant progress, gaps and challenges persist in the diagnosis of nAMD, initiation of treatment, and management of frequent intravitreal injections. Thus, nAMD remains a leading cause of blindness in the United States. OBJECTIVE To present current knowledge, evidence, and expert perspectives on anti-VEGF therapies in nAMD to support managed care professionals and providers in decision making and collaborative strategies to overcome barriers to optimize anti-VEGF treatment outcomes among nAMD patients. SUMMARY Three anti-VEGF therapies currently form the mainstay of treatment for nAMD, including 2 therapies approved by the FDA for treatment of nAMD (aflibercept and ranibizumab) and 1 therapy approved by the FDA for oncology indications and used off-label for treatment of nAMD (bevacizumab). In clinical trials, each of the 3 agents maintained visual acuity (VA) in approximately 90% or more of nAMD patients over 2 years. However, in long-term and real-world settings, significant gaps and challenges in diagnosis, treatment, and management pose barriers to achieving optimal outcomes for patients with nAMD. Many considerations, including individual patient characteristics, on-label versus off-label treatment, repackaging, and financial considerations, add to the complexity of nAMD decision making and management. Many factors may contribute to additional challenges leading to suboptimal long-term outcomes among nAMD patients, such as delays in diagnosis and/or treatment approval and initiation, individual patient response to different anti-VEGF therapies, lapses in physician regimentation of anti-VEGF injection and monitoring, and inadequate patient adherence to treatment and monitoring. These latter factors highlight the considerable logistical, emotional, and financial burdens of long-term, frequent intravitreal injections and the vital importance of personalized approaches to anti-VEGF treatment decision making and management for patients with nAMD. To address these challenges and reduce the number of yearly injections, studies have examined alternative dosing regimens, including extended fixed intervals, as needed, and treat-and-extend strategies in specific nAMD patient populations. New clinical evidence and insights into expert clinical practice discussed in this article can support managed care professionals in the key role they play in addressing challenges in nAMD treatment and management and optimizing patient outcomes through appropriate management of anti-VEGF treatment. DISCLOSURES PRIME Education is an independent medical education company and has been an accredited provider of continuing education for 23 years. There is no fee for this activity as it is sponsored by PRIME through an educational grant from Regeneron. All authors contributed to the writing and reviewing of the article. Wykoff reports consultancies/research grants from Alcon Laboratories, Genentech/Roche, Clearside, and Iconic Therapeutics; consultancies/honoraria, research grants, and speaker fees from Allergan and Regeneron; research grants from Allegro, Apellis, Aura, NEI, NIH, Novartis, OHR Pharmaceuticals, Ophthotech, pSivida, Roche, Santen, SciFluor, Tyrogenex; and consultancies for Alimera Sciences, Alnylam Pharmaceuticals, Bayer, DORC, ONL Therapeutics, Thrombogenics, and Valeant. Clark reports advisory board work, consultancies, research grants, and speaker fees from Genentech/Roche and Regeneron and consultancy for Bayer. Brill reports consultancies for Aries Pharma, Avella, BaroNova, Braeburn Pharmaceuticals, Cardinal Health, Endogastric Solutions, GeneNews, Halt Medical, Lumendi, Medtronic, Monteris Medical, Natera, Phosphorus, Rebiotix, Seno Medical, UCB, Vermillion, Echosens, and HAP Innovations. Brill is a shareholder in EndoChoice, GeneNews, SonarMD, and SynerZ and reports advisory board work with Nestle Health Sciences, Indivior Pharmaceuticals, Eli Lilly, Blue Earth Diagnostics, Bayer, and AstraZeneca. Nielson reports advisory board work/consultancy and research grants for Genentech/Roche; advisory board work and research grants from Regeneron; and research grants from Alcon and Ophthotech.
Collapse
|
107
|
Generative mathematical modelling to demonstrate virtual simulations of neovascular age related macular degeneration. PLoS One 2017; 12:e0189053. [PMID: 29211782 PMCID: PMC5718607 DOI: 10.1371/journal.pone.0189053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/17/2017] [Indexed: 11/26/2022] Open
Abstract
Purpose To develop a generative mathematical model of wet age-related macular degeneration (AMD) and model the impact of injections of anti-vascular endothelial growth factor to virtual patients with the condition. Methods We isolated key pathophysiological components of macular degeneration in terms of macular edema development and response to anti-vascular endothelial growth factor (VEGF) agents. We developed mathematical models for each of these components using constants determined from published biological experimentation. Consequently, we combined the mathematical models of the separate components to arrive at an end-to-end model of the evolution of macular edema size and its response to treatment. Results We present a series of simulations based upon our idealised model. Initially, we demonstrate the theoretical change in macular edema height in wet macular degeneration over time without and with anti-VEGF interventions. In our final simulation, we demonstrate the powerful possibilities of virtual clinical trials by simulating a virtual model of a landmark study using our existing mathematical AMD model. Conclusions Using our mathematical modelling based upon known pathological and pharmacological processes we have been able to model the effect of intravitreal injection of an anti-VEGF agent on macular edema from age related macular degeneration. We were subsequently able to mathematically simulate a major clinical trial with results that mirror many key features of the clinical established study. We anticipate that the generative model presented here can evolve to be a useful supportive tool in the challenge to deliver optimal therapy for patients with wet macular degeneration.
Collapse
|
108
|
Kaya F. Change in choroidal thickness after intravitreal injection for treatment of neovascular age-related macular degeneration: Ranibizumab versus aflibercept. J Fr Ophtalmol 2017; 40:832-838. [PMID: 29113742 DOI: 10.1016/j.jfo.2017.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 04/10/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE To compare the changes in subfoveal choroidal thickness after intravitreal ranibizumab or aflibercept injections for neovascular age-related macular degeneration (nAMD). METHODS In this retrospective study, 28 eyes with nAMD treated with 3 consecutive monthly injections of ranibizumab (IVR) and 24 eyes with nAMD treated with 3 consecutive monthly injections of aflibercept (IVA) between September 2012 and June 2016 were reviewed. The follow-up time was 6 months. Changes in two groups' best-corrected visual acuity (BCVA) and subfoveal choroidal thickness by using enhanced depth imaging optical coherence tomography at 1st, 3rd and 6th months were recorded and compared. RESULTS Choroidal thickness decreased significantly in eyes treated with IVR (P=0.015, 0.01 and 0.01, respectively) or IVA (P=0.001, 0.001 and ˂0.001, respectively) at 1, 3 and 6 months examination but IVA treated eyes presented a significantly further reduction in choroidal thickness when compared with ranibizumab (P=0.03, 0,04 and 0.03, respectively). There was no significant difference between aflibercept and ranibizumab group when change in BCVA from baseline compared at 1, 3 and 6th months (P=0.54, 0.06 and 0.37, respectively). There was no correlation between change in choroidal thickness and the BCVA outcomes in either group. CONCLUSIONS Subfoveal choroidal thickness decreased significantly after both of IVR and IVA injections in patients with nAMD. In conclusion, intravitreal injections of ranibizumab or aflibercept affect not only neovascular lesion but also the underlying choroid.
Collapse
Affiliation(s)
- F Kaya
- Service d'ophtalmologie, hôpital de Nisa, 34196 Istanbul, Turkey; Clinique universitaire d'ophtalmologie, faculté de médecine, université d'Istanbul Medipol, 34196 Istanbul, Turkey.
| |
Collapse
|
109
|
The ‘Displacing Foods of Modern Commerce’ Are the Primary and Proximate Cause of Age-Related Macular Degeneration: A Unifying Singular Hypothesis. Med Hypotheses 2017; 109:184-198. [DOI: 10.1016/j.mehy.2017.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 01/17/2023]
|
110
|
Abstract
PDGFs and their receptors are critical regulators of numerous tissues and organs, including the eye. Extensive studies have shown that PDGFs and their receptors play critical roles in many ocular neovascular diseases, such as neovascular age-related macular degeneration, retinopathy of prematurity, and proliferative vitreoretinopathy. In addition, PDGFs and PDGFRs are also important players in ocular diseases involving the degeneration of retinal neuronal and vascular cells, such as glaucoma and retinitis pigmentosa. Due to their critical roles in the pathogenesis of many blinding ocular diseases, the PDGFs and PDGFRs have been considered as important target molecules for the treatment of eye diseases. PDGF-C and PDGF-D are relatively new members of the PDGF family and are potent angiogenic and survival factors. Recent studies have demonstrated their important roles in different types of eye diseases. Thus, modulating PDGF-C and PDGF-D activities may have therapeutic values for the treatment of ocular neovascular and degenerative diseases. This review mainly summarizes the recent advances on PDGF-C and PDGF-D biology in relationship to some major ocular diseases.
Collapse
Affiliation(s)
- Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 South Xianlie Road, Guangzhou 510060, Guangdong, PR China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 South Xianlie Road, Guangzhou 510060, Guangdong, PR China.
| |
Collapse
|
111
|
Layana AG, Minnella AM, Garhöfer G, Aslam T, Holz FG, Leys A, Silva R, Delcourt C, Souied E, Seddon JM. Vitamin D and Age-Related Macular Degeneration. Nutrients 2017; 9:nu9101120. [PMID: 29027953 PMCID: PMC5691736 DOI: 10.3390/nu9101120] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/03/2017] [Accepted: 10/06/2017] [Indexed: 12/12/2022] Open
Abstract
In recent years, the relationship between vitamin D and health has received growing attention from the scientific and medical communities. Vitamin D deficiencies have been repeatedly associated with various acute and chronic diseases, including age-related macular degeneration (AMD). Its active metabolite, 1α,25-dihydoxy vitamin D, acts as a modulator of cell proliferation, differentiation and apoptosis, and cumulative data from experimental and observational studies suggest that relatively a lower vitamin D status could be a potential risk factor for the development of early and/or late AMD. Herein, we made a narrative review of the mechanisms linking a potential role of vitamin D with the current concepts of AMD pathophysiology.
Collapse
Affiliation(s)
| | - Angelo Maria Minnella
- Dipartimento di Scienze Otorinolaringoiatriche e Oftalmologiche, Universita' Cattolica del Sacro Cuore, Lgo F. Vito 1, 00168 Roma, Italy.
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology, University of Vienna, 1090 Vienna, Austria.
| | - Tariq Aslam
- School of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, M13 9WL Manchester, and Heriot Watt University, Edinburgh EH14 4AS, UK.
| | - Frank G Holz
- Department of Ophthalmology, University of Bonn, D-53107 Bonn, Germany.
| | - Anita Leys
- Department of Ophthalmology, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Rufino Silva
- Faculty of Medicine, Institute for Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Ophthalmology, 3000-548 Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra (CHUC), Faculty of Medicine, Institute for Biomedical Imaging and Life Sciences (IBILI-FMUC), University of Coimbra, 3000-548 Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra (CHUC), Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal.
| | - Cécile Delcourt
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Team LEHA, UMR 1219, F-33000 Bordeaux, France.
| | - Eric Souied
- Hôpital Intercommunal de Créteil, University Paris Est, 94010 Créteil, France.
| | - Johanna M Seddon
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA.
- Ophthalmic Epidemiology and Genetics Service, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
112
|
Rudolf M, Mir Mohi Sefat A, Miura Y, Tura A, Raasch W, Ranjbar M, Grisanti S, Aherrahrou Z, Wagner A, Messinger JD, Garber DW, Anantharamaiah GM, Curcio CA. ApoA-I Mimetic Peptide 4F Reduces Age-Related Lipid Deposition in Murine Bruch's Membrane and Causes Its Structural Remodeling. Curr Eye Res 2017; 43:135-146. [PMID: 28972410 DOI: 10.1080/02713683.2017.1370118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Accumulation of lipoprotein-derived lipids including esterified and unesterified cholesterol in Bruch's membrane of human eyes is a major age-related change involved in initiating and sustaining soft drusen in age-related macular degeneration (AMD). The apolipoprotein (apo) A-I mimetic peptide 4F is a small anti-inflammatory and anti-atherogenic agent, and potent modifier of plasma membranes. We evaluated the effect of intravitreally-injected 4F on murine Bruch's membrane. METHODS We tested single intravitreal injections of 4F doses (0.6 µg, 1.2 µg, 2.4 µg, and placebo scrambled peptide) in ApoEnull mice ≥10 months of age. After 30 days, mice were euthanized. Eyes were processed for either direct immunofluorescence detection of esterified cholesterol (EC) in Bruch's membrane whole mounts via a perfringolysin O-based marker linked to green fluorescent protein or by transmission electron microscopic visualization of Bruch's membrane integrity. Fluorescein isothiocyanate-conjugated 4F was traced after injection. RESULTS All injected eyes showed a dose-dependent reduction of Bruch's membrane EC with a concomitant ultrastructural improvement compared to placebo treated eyes. At a 2.4 µg dose of 4F, EC was reduced on average by ~60% and Bruch's membrane returned to a regular pentalaminar structure and thickness. Tracer studies confirmed that injected 4F reached intraocular targets. CONCLUSION We demonstrated a highly effective pharmacological reduction of EC and restoration of Bruch's membrane ultrastructure. The apoA-I mimetic peptide 4F is a novel way to treat a critical AMD disease process and thus represents a new candidate for treating the underlying cause of AMD.
Collapse
Affiliation(s)
- Martin Rudolf
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | | | - Yoko Miura
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Aysegül Tura
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Walter Raasch
- b Department of Experimental and Clinical Pharmacology and Toxicology , University of Lübeck , Lübeck , Germany
| | - Mahdy Ranjbar
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany.,c Laboratory for Angiogenesis & Ocular Cell Transplantation , University of Lübeck , Lübeck , Germany
| | | | - Zouhair Aherrahrou
- d Institute of Integrative and Experimental Genomics , University of Lübeck , Lübeck , Germany
| | - Anna Wagner
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Jeffrey D Messinger
- e Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - David W Garber
- f Atherosclerosis Research Unit , University of Alabama at Birmingham , Birmingham , AL , USA
| | - G M Anantharamaiah
- f Atherosclerosis Research Unit , University of Alabama at Birmingham , Birmingham , AL , USA.,g Department of Medicine, Biochemistry and Molecular Genetics , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Christine A Curcio
- e Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , AL , USA
| |
Collapse
|
113
|
Weinberger D, Bor-Shavit E, Barliya T, Dahbash M, Kinrot O, Gaton DD, Nisgav Y, Livnat T. Mobile Laser Indirect Ophthalmoscope: For the Induction of Choroidal Neovascularization in a Mouse Model. Curr Eye Res 2017; 42:1545-1551. [PMID: 28933966 DOI: 10.1080/02713683.2017.1349154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE This study aims to evaluate and standardize the reliability of a mobile laser indirect ophthalmoscope in the induction of choroidal neovascularization (CNV) in a mouse model. MATERIALS & METHODS A diode laser indirect ophthalmoscope was used to induce CNV in pigmented male C57BL/6J mice. Standardization of spot size and laser intensity was determined using different aspheric lenses with increasing laser intensities applied around the optic disc. Development of CNV was evaluated 1, 5, and 14 days post laser application using fluorescein angiography (FA), histology, and choroidal flat mounts stained for the endothelial marker CD31 and FITC-dextran. Correlation between the number of laser hits to the number and size of developed CNV lesions was determined using flat mount choroid staining. The ability of intravitreally injected anti-human and anti-mouse VEGF antibodies to inhibit CNV induced by the mobile laser was evaluated. RESULTS Laser parameters were standardized on 350 mW for 100 msec, using the 90 diopter lens to accomplish the highest incidence of Bruch's membrane rupture. CNV lesions' formation was validated on days 5 and 14 post laser injury, though FA showed leakage on as early as day 1. The number of laser hits was significantly correlated with the CNV area. CNV growth was successfully inhibited by both anti-human and mouse VEGF antibodies. CONCLUSION The mobile laser indirect ophthalmoscope can serve as a feasible and a reliable alternative method for the CNV induction in a mouse model.
Collapse
Affiliation(s)
- Dov Weinberger
- a Laboratory of Eye Research , Felsenstein Medical Research Center , Beilinson Campus, Petach Tikva , Israel.,b Department of Ophthalmology , Rabin Medical Center , Beilinson Campus, Petach Tikva , Israel.,c Sackler Faculty of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Elite Bor-Shavit
- b Department of Ophthalmology , Rabin Medical Center , Beilinson Campus, Petach Tikva , Israel
| | - Tilda Barliya
- a Laboratory of Eye Research , Felsenstein Medical Research Center , Beilinson Campus, Petach Tikva , Israel
| | - Mor Dahbash
- a Laboratory of Eye Research , Felsenstein Medical Research Center , Beilinson Campus, Petach Tikva , Israel
| | | | - Dan D Gaton
- b Department of Ophthalmology , Rabin Medical Center , Beilinson Campus, Petach Tikva , Israel.,c Sackler Faculty of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Yael Nisgav
- a Laboratory of Eye Research , Felsenstein Medical Research Center , Beilinson Campus, Petach Tikva , Israel
| | - Tami Livnat
- a Laboratory of Eye Research , Felsenstein Medical Research Center , Beilinson Campus, Petach Tikva , Israel.,c Sackler Faculty of Medicine , Tel Aviv University , Tel Aviv , Israel.,e The Israeli National Hemophilia Center and Thrombosis Unit , Sheba Medical Center , Tel Hashomer , Israel
| |
Collapse
|
114
|
Hasegawa E, Inafuku S, Mulki L, Okunuki Y, Yanai R, Smith KE, Kim CB, Klokman G, Bielenberg DR, Puli N, Falck JR, Husain D, Miller JW, Edin ML, Zeldin DC, Lee KSS, Hammock BD, Schunck WH, Connor KM. Cytochrome P450 monooxygenase lipid metabolites are significant second messengers in the resolution of choroidal neovascularization. Proc Natl Acad Sci U S A 2017; 114:E7545-E7553. [PMID: 28827330 PMCID: PMC5594641 DOI: 10.1073/pnas.1620898114] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of blindness for individuals age 50 and above in the developed world. Abnormal growth of choroidal blood vessels, or choroidal neovascularization (CNV), is a hallmark of the neovascular (wet) form of advanced AMD and leads to significant vision loss. A growing body of evidence supports a strong link between neovascular disease and inflammation. Metabolites of long-chain polyunsaturated fatty acids derived from the cytochrome P450 (CYP) monooxygenase pathway serve as vital second messengers that regulate a number of hormones and growth factors involved in inflammation and vascular function. Using transgenic mice with altered CYP lipid biosynthetic pathways in a mouse model of laser-induced CNV, we characterized the role of these lipid metabolites in regulating neovascular disease. We discovered that the CYP-derived lipid metabolites epoxydocosapentaenoic acids (EDPs) and epoxyeicosatetraenoic acids (EEQs) are vital in dampening CNV severity. Specifically, overexpression of the monooxygenase CYP2C8 or genetic ablation or inhibition of the soluble epoxide hydrolase (sEH) enzyme led to increased levels of EDP and EEQ with attenuated CNV development. In contrast, when we promoted the degradation of these CYP-derived metabolites by transgenic overexpression of sEH, the protective effect against CNV was lost. We found that these molecules work in part through their ability to regulate the expression of key leukocyte adhesion molecules, on both leukocytes and endothelial cells, thereby mediating leukocyte recruitment. These results suggest that CYP lipid signaling molecules and their regulators are potential therapeutic targets in neovascular diseases.
Collapse
Affiliation(s)
- Eiichi Hasegawa
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Saori Inafuku
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Lama Mulki
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Yoko Okunuki
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Ryoji Yanai
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Kaylee E Smith
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Clifford B Kim
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Garrett Klokman
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Narender Puli
- Department of Biochemistry, University of Texas Southwestern, Dallas, TX 75390
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern, Dallas, TX 75390
| | - Deeba Husain
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Joan W Miller
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114
| | - Matthew L Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Kin Sing Stephen Lee
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616
| | - Bruce D Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616;
| | | | - Kip M Connor
- Angiogenesis Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114;
| |
Collapse
|
115
|
Laíns I, Miller JB, Park DH, Tsikata E, Davoudi S, Rahmani S, Pierce J, Silva R, Chen TC, Kim IK, Vavvas D, Miller JW, Husain D. Structural Changes Associated with Delayed Dark Adaptation in Age-Related Macular Degeneration. Ophthalmology 2017; 124:1340-1352. [PMID: 28501377 DOI: 10.1016/j.ophtha.2017.03.061] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/27/2017] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To examine the relationship between dark adaptation (DA) and optical coherence tomography (OCT)-based macular morphology in age-related macular degeneration (AMD). DESIGN Prospective, cross-sectional study. PARTICIPANTS Patients with AMD and a comparison group (>50 years) without any vitreoretinal disease. METHODS All participants were imaged with spectral-domain OCT and color fundus photographs, and then staged for AMD (Age-related Eye Disease Study system). Both eyes were tested with the AdaptDx (MacuLogix, Middletown, PA) DA extended protocol (20 minutes). A software program was developed to map the DA testing spot (2° circle, 5° superior to the fovea) to the OCT B-scans. Two independent graders evaluated the B-scans within this testing spot, as well as the entire macula, recording the presence of several AMD-associated abnormalities. Multilevel mixed-effects models (accounting for correlated outcomes between 2 eyes) were used for analyses. MAIN OUTCOME MEASURES The primary outcome was rod-intercept time (RIT), defined in minutes, as a continuous variable. For subjects unable to reach RIT within the 20 minutes of testing, the value of 20 was assigned. RESULTS We included 137 eyes (n = 77 subjects), 72.3% (n = 99 eyes) with AMD and the remainder belonging to the comparison group. Multivariable analysis revealed that even after adjusting for age and AMD stage, the presence of any abnormalities within the DA testing spot (ß = 4.8, P < 0.001), as well as any abnormalities in the macula (ß = 2.4, P = 0.047), were significantly associated with delayed RITs and therefore impaired DA. In eyes with no structural changes within the DA testing spot (n = 76, 55.5%), the presence of any abnormalities in the remaining macula was still associated with delayed RITs (ß = 2.00, P = 0.046). Presence of subretinal drusenoid deposits and ellipsoid zone disruption were a consistent predictor of RIT, whether located within the DA testing spot (P = 0.001 for both) or anywhere in the macula (P < 0.001 for both). Within the testing spot, the presence of classic drusen or serous pigment epithelium detachment was also significantly associated with impairments in DA (P ≤ 0.018). CONCLUSIONS Our results suggest a significant association between macular morphology evaluated by OCT and time to dark-adapt. Subretinal drusenoid deposits and ellipsoid zone changes seem to be strongly associated with impaired dark adaptation.
Collapse
Affiliation(s)
- Inês Laíns
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Association for Innovation and Biomedical Research on Light, AIBILI, Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - John B Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Dong H Park
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, School of Medicine, Kyungpook National University, South Korea
| | - Edem Tsikata
- Glaucoma Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Samaneh Davoudi
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Safa Rahmani
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Jonathan Pierce
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Rufino Silva
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Association for Innovation and Biomedical Research on Light, AIBILI, Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Teresa C Chen
- Glaucoma Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Ivana K Kim
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Demetrios Vavvas
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Joan W Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Deeba Husain
- Retina Service, Massachusetts Eye and Ear, Harvard Ophthalmology AMD Center of Excellence, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
116
|
Schlecht A, Leimbeck SV, Jägle H, Feuchtinger A, Tamm ER, Braunger BM. Deletion of Endothelial Transforming Growth Factor-β Signaling Leads to Choroidal Neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2570-2589. [PMID: 28823871 DOI: 10.1016/j.ajpath.2017.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 12/13/2022]
Abstract
The molecular pathogenesis of choroidal neovascularization (CNV), an angiogenic process that critically contributes to vision loss in age-related macular degeneration, is unclear. Herein, we analyzed the role of transforming growth factor (TGF)-β signaling for CNV formation by generating a series of mutant mouse models with induced conditional deletion of TGF-β signaling in the entire eye, the retinal pigment epithelium (RPE), or the vascular endothelium. Deletion of TGF-β signaling in the eye caused CNV, irrespectively if it was ablated in newborn or 3-week-old mice. Areas of CNV showed photoreceptor degeneration, multilayered RPE, basal lamina deposits, and accumulations of monocytes/macrophages. The changes progressed, leading to marked structural and functional alterations of the retina. Although the specific deletion of TGF-β signaling in the RPE caused no obvious changes, specific deletion in vascular endothelial cells caused CNV and a phenotype similar to that observed after the deletion in the entire eye. We conclude that impairment of TGF-β signaling in the vascular endothelium of the eye is sufficient to trigger CNV formation. Our findings highlight the importance of TGF-β signaling as a key player in the development of ocular neovascularization and indicate a fundamental role of TGF-β signaling in the pathogenesis of age-related macular degeneration.
Collapse
Affiliation(s)
- Anja Schlecht
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Sarah V Leimbeck
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University Clinic Regensburg, Regensburg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum Munich, Munich, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| | - Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
117
|
Inner nuclear layer cystoid spaces are a poor prognostic factor in typical age-related macular degeneration and polypoidal choroidal vasculopathy. Graefes Arch Clin Exp Ophthalmol 2017; 255:2157-2163. [DOI: 10.1007/s00417-017-3776-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/27/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022] Open
|
118
|
DeAngelis MM, Owen LA, Morrison MA, Morgan DJ, Li M, Shakoor A, Vitale A, Iyengar S, Stambolian D, Kim IK, Farrer LA. Genetics of age-related macular degeneration (AMD). Hum Mol Genet 2017; 26:R45-R50. [PMID: 28854576 PMCID: PMC5886461 DOI: 10.1093/hmg/ddx228] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022] Open
Abstract
Age-related macular degeneration (AMD) is a progressive blinding disease and represents the leading cause of visual impairment in the aging population. AMD affects central vision which impairs one's ability to drive, read and recognize faces. There is no cure for this disease and current treatment modalities for the exudative form of the disease require repeated intravitreal injections which may be painful, are incompletely efficacious, and represent a significant treatment burden for both the patient and physician. As such, AMD represents a significant and important clinical problem.It is anticipated that in three years' time, 196 million individuals will be affected with AMD. Over 250 billion dollars per year are spent on care for AMD patients in the US. Over half of the heritability is explained by two major loci, thus AMD is considered the most well genetically defined of the complex disorders. A recent GWAS on 43,566 subjects identified novel loci and pathways associated with AMD risk, which has provided an excellent platform for additional functional studies. Genetic variants have been investigated, particularly with respect to anti-VEGF treatment, however to date, no pharmacogenomic associations have been consistently identified across these studies. It may be that if the goal of personalized medicine is to be realized and biomarkers are to have predictive value for determining the magnitude of risk for AMD at the genetic level, one will need to examine the relationships between these pathways across disease state and relative to modifiable risk factors such as hypertension, smoking, body mass index, and hypercholesterolemia. Further studies investigating protective alleles in populations with low AMD prevalence may lead to this goal.
Collapse
Affiliation(s)
- Margaret M. DeAngelis
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Pharmacotherapy, University of Utah, College of Pharmacy, Salt Lake City, UT 84132, USA
| | - Leah A. Owen
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Margaux A. Morrison
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Denise J. Morgan
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Akbar Shakoor
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Albert Vitale
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Sudha Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ivana K. Kim
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics)
- Department of Neurology
- Department of Ophthalmology, Boston University Schools of Medicine, Boston, MA 02118, USA
- Department of Biostatistics
- Department of Epidemiology, Boston University Schools of Public Health, Boston, MA 02118, USA
| |
Collapse
|
119
|
Chiu CJ, Chang ML, Li T, Gensler G, Taylor A. Visualization of Dietary Patterns and Their Associations With Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2017; 58:1404-1410. [PMID: 28253403 PMCID: PMC5361454 DOI: 10.1167/iovs.16-20454] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Purpose We aimed to visualize the relationship of predominant dietary patterns and their associations with AMD. Methods A total of 8103 eyes from 4088 participants in the baseline Age-Related Eye Disease Study (AREDS) were classified into three groups: control (n = 2739), early AMD (n = 4599), and advanced AMD (n = 765). Using principle component analysis, two major dietary patterns and eight minor dietary patterns were characterized. Applying logistic regression in our analysis, we related dietary patterns to the prevalence of AMD. Qualitative comparative analysis by operating Boolean algebra and drawing Venn diagrams was used to visualize our findings. Results In general, the eight minor patterns were subsets or extensions of either one of the two major dietary patterns (Oriental and Western patterns) and consisted of fewer characteristic foods than the two major dietary patterns. Unlike the two major patterns, which were more strongly associated with both early and advanced AMD, none of the eight minors were associated with early AMD and only four minor patterns, including the Steak pattern (odds ratio comparing the highest to lowest quintile of the pattern score = 1.73 [95% confidence interval: 1.24 to 2.41; Ptrend = 0.02]), the Breakfast pattern (0.60 [0.44 to 0.82]; Ptrend = 0.004]), the Caribbean pattern (0.64 [0.47 to 0.89; Ptrend = 0.009]), and the Peanut pattern (0.64 [0.46 to 0.89; Ptrend = 0.03]), were significantly associated with advanced AMD. Our data also suggested several potential beneficial (peanuts, pizza, coffee, and tea) and harmful (salad dressing) foods for AMD. Conclusions Our data indicate that a diet of various healthy foods may be optimal for reducing AMD risk. The effects of some specific foods in the context of overall diet warrant further study.
Collapse
Affiliation(s)
- Chung-Jung Chiu
- Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States 2Department of Ophthalmology, School of Medicine, Tufts University, Boston, Massachusetts, United States
| | - Min-Lee Chang
- Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States
| | - Tricia Li
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Gary Gensler
- Age-Related Eye Disease Study Coordinating Center, The Emmes Corporation, Rockville, Maryland, United States
| | - Allen Taylor
- Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, United States 2Department of Ophthalmology, School of Medicine, Tufts University, Boston, Massachusetts, United States
| |
Collapse
|
120
|
Evaluation of intraretinal migration of retinal pigment epithelial cells in age-related macular degeneration using polarimetric imaging. Sci Rep 2017; 7:3150. [PMID: 28600515 PMCID: PMC5466639 DOI: 10.1038/s41598-017-03529-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/28/2017] [Indexed: 02/07/2023] Open
Abstract
The purpose of the present study was to evaluate the intraretinal migration of the retinal pigment epithelium (RPE) cells in age-related macular degeneration (AMD) using polarimetry. We evaluated 155 eyes at various AMD stages. Depolarized light images were computed using a polarization-sensitive scanning laser ophthalmoscope (PS-SLO), and the degree of polarization uniformity was calculated using polarization-sensitive optical coherence tomography (OCT). Each polarimetry image was compared with the corresponding autofluorescence (AF) images at 488 nm (SW-AF) and at 787 nm (NIR-AF). Intraretinal RPE migration was defined by the presence of depolarization at intraretinal hyperreflective foci on PS-SLO and PS-OCT images, and by the presence of hyper-AF on both NIR-AF and SW-AF images. RPE migration was detected in 52 of 155 eyes (33.5%) and was observed in drusenoid pigment epithelial detachment (PED) and serous PED with significantly higher frequencies than in other groups (P = 0.015). The volume of the migrated RPE cluster in serous PED was significantly correlated with the volume of the PED (R2 = 0.26; P = 0.011). Overall, our results showed that intraretinal RPE migrations occurred in various AMD stages, and that they occurred more commonly in eyes with serous and drusenoid PED.
Collapse
|
121
|
Thakur SS, Ward MS, Popat A, Flemming NB, Parat MO, Barnett NL, Parekh HS. Stably engineered nanobubbles and ultrasound - An effective platform for enhanced macromolecular delivery to representative cells of the retina. PLoS One 2017; 12:e0178305. [PMID: 28542473 PMCID: PMC5444814 DOI: 10.1371/journal.pone.0178305] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/10/2017] [Indexed: 01/10/2023] Open
Abstract
Herein we showcase the potential of ultrasound-responsive nanobubbles in enhancing macromolecular permeation through layers of the retina, ultimately leading to significant and direct intracellular delivery; this being effectively demonstrated across three relevant and distinct retinal cell lines. Stably engineered nanobubbles of a highly homogenous and echogenic nature were fully characterised using dynamic light scattering, B-scan ultrasound and transmission electron microscopy (TEM). The nanobubbles appeared as spherical liposome-like structures under TEM, accompanied by an opaque luminal core and darkened corona around their periphery, with both features indicative of efficient gas entrapment and adsorption, respectively. A nanobubble +/- ultrasound sweeping study was conducted next, which determined the maximum tolerated dose for each cell line. Detection of underlying cellular stress was verified using the biomarker heat shock protein 70, measured before and after treatment with optimised ultrasound. Next, with safety to nanobubbles and optimised ultrasound demonstrated, each human or mouse-derived cell population was incubated with biotinylated rabbit-IgG in the presence and absence of ultrasound +/- nanobubbles. Intracellular delivery of antibody in each cell type was then quantified using Cy3-streptavidin. Nanobubbles and optimised ultrasound were found to be negligibly toxic across all cell lines tested. Macromolecular internalisation was achieved to significant, yet varying degrees in all three cell lines. The results of this study pave the way towards better understanding mechanisms underlying cellular responsiveness to ultrasound-triggered drug delivery in future ex vivo and in vivo models of the posterior eye.
Collapse
Affiliation(s)
- Sachin S. Thakur
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Micheal S. Ward
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Nicole B. Flemming
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Marie-Odile Parat
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Nigel L. Barnett
- Queensland Eye Institute, South Brisbane, Queensland, Australia
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane Queensland, Australia
| | - Harendra S. Parekh
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
- * E-mail:
| |
Collapse
|
122
|
Tian B, Al-Moujahed A, Bouzika P, Hu Y, Notomi S, Tsoka P, Miller JW, Lin H, Vavvas DG. Atorvastatin Promotes Phagocytosis and Attenuates Pro-Inflammatory Response in Human Retinal Pigment Epithelial Cells. Sci Rep 2017; 7:2329. [PMID: 28539592 PMCID: PMC5443823 DOI: 10.1038/s41598-017-02407-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/25/2017] [Indexed: 11/09/2022] Open
Abstract
Phagocytosis of daily shed photoreceptor outer segments is an important function of the retinal pigment epithelium (RPE) and it is essential for retinal homeostasis. RPE dysfunction, especially impairment of its phagocytic ability, plays an essential role in the pathogenesis of age-related macular degeneration (AMD). Statins, or HMG CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase inhibitors, are drugs with multiple properties that have been extensively used to treat hyperlipidemia. However, their effect on RPE cells has not been fully elucidated. Here we report that high dose atorvastatin increased the phagocytic function of ARPE-19 cells, as well as rescue the cells from the phagocytic dysfunction induced by cholesterol crystals and oxidized low-density lipoproteins (ox-LDL), potentially by increasing the cellular membrane fluidity. Similar effects were observed when evaluating two other hydrophobic statins, lovastatin and simvastatin. Furthermore, atorvastatin was able to block the induction of interleukins IL-6 and IL-8 triggered by pathologic stimuli relevant to AMD, such as cholesterol crystals and ox-LDL. Our study shows that statins, a well-tolerated class of drugs with rare serious adverse effects, help preserve the phagocytic function of the RPE while also exhibiting anti-inflammatory properties. Both characteristics make statins a potential effective medication for the prevention and treatment of AMD.
Collapse
Affiliation(s)
- Bo Tian
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
| | - Ahmad Al-Moujahed
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
- Department of Pathology, Boston University School of Medicine, Boston, MA, 02118, United States
| | - Peggy Bouzika
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
| | - Yijun Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Shoji Notomi
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
| | - Pavlina Tsoka
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
| | - Joan W Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States
| | - Haijiang Lin
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States.
- Department of Ophthalmology & Visual Sciences, University of Massachusetts Medical School, Worcester, MA, 01605, United States.
| | - Demetrios G Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, 02114, United States.
| |
Collapse
|
123
|
Burgess S, Davey Smith G. Mendelian Randomization Implicates High-Density Lipoprotein Cholesterol-Associated Mechanisms in Etiology of Age-Related Macular Degeneration. Ophthalmology 2017; 124:1165-1174. [PMID: 28456421 PMCID: PMC5526457 DOI: 10.1016/j.ophtha.2017.03.042] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 11/28/2022] Open
Abstract
Purpose Undertake a systematic investigation into associations between genetic predictors of lipid fractions and age-related macular degeneration (AMD) risk. Design Two-sample Mendelian randomization investigation using published data. Participants A total of 33 526 individuals (16 144 cases, 17 832 controls) predominantly of European ancestry from the International Age-related Macular Degeneration Genomics Consortium. Methods We consider 185 variants previously demonstrated to be associated with at least 1 of low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, or triglycerides at a genome-wide level of significance, and test their associations with AMD. We particularly focus on variants in gene regions that are proxies for specific pharmacologic agents for lipid therapy. We then conduct a 2-sample Mendelian randomization investigation to assess the causal roles of LDL-cholesterol, HDL-cholesterol, and triglycerides on AMD risk. We also conduct parallel investigations for coronary artery disease (CAD) (viewed as a positive control) and Alzheimer's disease (a negative control) for comparison. Main Outcome Measures Diagnosis of AMD. Results We find evidence that HDL-cholesterol is a causal risk factor for AMD, with an odds ratio (OR) estimate of 1.22 (95% confidence interval [CI], 1.03–1.44) per 1 standard deviation increase in HDL-cholesterol. No causal effect of LDL-cholesterol or triglycerides was found. Variants in the CETP gene region associated with increased circulating HDL-cholesterol also associate with increased AMD risk, although variants in the LIPC gene region that increase circulating HDL-cholesterol have the opposite direction of association with AMD risk. Parallel analyses suggest that lipids have a greater role for AMD compared with Alzheimer's disease, but a lesser role than for CAD. Conclusions Some genetic evidence suggests that HDL-cholesterol is a causal risk factor for AMD risk and that increasing HDL-cholesterol (particularly via CETP inhibition) will increase AMD risk.
Collapse
Affiliation(s)
- Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom; Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom.
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
124
|
Jia YP, Sun L, Yu HS, Liang LP, Li W, Ding H, Song XB, Zhang LJ. The Pharmacological Effects of Lutein and Zeaxanthin on Visual Disorders and Cognition Diseases. Molecules 2017; 22:E610. [PMID: 28425969 PMCID: PMC6154331 DOI: 10.3390/molecules22040610] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/19/2022] Open
Abstract
Lutein (L) and zeaxanthin (Z) are dietary carotenoids derived from dark green leafy vegetables, orange and yellow fruits that form the macular pigment of the human eyes. It was hypothesized that they protect against visual disorders and cognition diseases, such as age-related macular degeneration (AMD), age-related cataract (ARC), cognition diseases, ischemic/hypoxia induced retinopathy, light damage of the retina, retinitis pigmentosa, retinal detachment, uveitis and diabetic retinopathy. The mechanism by which they are involved in the prevention of eye diseases may be due their physical blue light filtration properties and local antioxidant activity. In addition to their protective roles against light-induced oxidative damage, there are increasing evidences that L and Z may also improve normal ocular function by enhancing contrast sensitivity and by reducing glare disability. Surveys about L and Z supplementation have indicated that moderate intakes of L and Z are associated with decreased AMD risk and less visual impairment. Furthermore, this review discusses the appropriate consumption quantities, the consumption safety of L, side effects and future research directions.
Collapse
Affiliation(s)
- Yu-Ping Jia
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Lei Sun
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - He-Shui Yu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Li-Peng Liang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Wei Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Hui Ding
- Tianjin Zhongyi Pharmaceutical Co., Ltd., Tianjin 300193, China.
| | - Xin-Bo Song
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
- Tianjin Zhongyi Pharmaceutical Co., Ltd., Tianjin 300193, China.
| | - Li-Juan Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
125
|
SanGiovanni JP, SanGiovanni PM, Sapieha P, De Guire V. miRNAs, single nucleotide polymorphisms (SNPs) and age-related macular degeneration (AMD). Clin Chem Lab Med 2017; 55:763-775. [PMID: 28343170 DOI: 10.1515/cclm-2016-0898] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 02/27/2017] [Indexed: 12/15/2022]
Abstract
Advanced age-related macular degeneration (AAMD) is a complex sight-threating disease of public health significance. Micro RNAs (miRNAs) have been proposed as biomarkers for AAMD. The presence of certain single nucleotide polymorphisms (SNPs) may influence the explanatory value of these biomarkers. Here we present findings from an integrated approach used to determine whether AAMD-associated SNPs have the capacity to influence miRNA-mRNA pairing and, if so, to what extent such pairing may be manifested in a discrete AAMD transcriptome. Using a panel of 8854 SNPs associated with AAMD at p-values ≤5.0E-7 from a cohort of >30,000 elderly people, we identified SNPs in miRNA target-encoding constituents of: (1) regulator of complement activation (RCA) genes (rs390679, CFHR1, p≤2.14E-214 | rs12140421, CFHR3, p≤4.63E-29); (2) genes of major histocompatibility complex (MHC) loci (rs4151672, CFB, p≤8.91E-41 | rs115404146, HLA-C, p≤6.32E-12 | rs1055821, HLA-B, p≤1.93E-9 | rs1063355, HLA-DQB1, p≤6.82E-14); and (3) genes of the 10q26 AAMD locus (rs1045216, PLEKHA1, p≤4.17E-142 | rs2672603, ARMS2, p≤7.14E-46). We used these findings with existing data on AAMD-related retinal miRNA and transcript profiles for the purpose of making inferences on SNP-mRNA-miRNA-AAMD relationships. Four of 12 miRNAs significantly elevated in AAMD retina (hsa-miR-155-5p, hsa-let-7a-5p, hsa-let-7b-5p hsa-let-7d-5p) also showed strong pairing capacity (TarBase 7.1 context++ score <-0.2, miRanda 3.3 pairing score >150) with miRNA target transcripts encoded by AAMD-associated SNPs resident in HLA-DQB1 (rs1063355, hsa-miR-155-5p) and TGFBR1 (rs868, hsa-let-7). Three of the 12 miRNAs overexpressed in AAMD retina are inducible by NFkB and have high affinity targets in the complement factor H (CFH) mRNA 3' UTR. We used ENSEMBL to identify polymorphic regions in the CFH mRNA 3' UTR with the capacity to disrupt miRNA-mRNA pairing. Two variants (rs766666504 and rs459598) existed in DNA sequence encoding the seed region of hsa-miR-146a-5p in the CFH mRNA 3' UTR - as this miRNA is also elevated in both vitreous and serum of people with AAMD, it shows great value as a biomarker. Our findings suggest that knowledge on the nature of DNA sequence variation may increase the explanatory power of miRNA biomarkers in genetically diverse populations, while yielding information with which to develop: (1) mechanistic tests on processes implicated in AMD pathogenesis; and, (2) site-specific small molecules (synthetic mimetics or anti-miRNAs) with preventive or therapeutic efficacy for AAMD.
Collapse
Affiliation(s)
- John Paul SanGiovanni
- Laboratory of Membrane Biochemistry and Biophysics, Section on Nutritional Neuroscience, NIAAA/NIH, Bethesda, MD
| | | | - Przemysław Sapieha
- Departments of Biochemistry and Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec
| | - Vincent De Guire
- Division of Clinical Biochemistry, Maisonneuve-Rosemont Hospital, University of Montreal, Department of Biochemistry and Molecular Medicine, Montreal, Quebec
| |
Collapse
|
126
|
Potential protective function of the sterol regulatory element binding factor 1-fatty acid desaturase 1/2 axis in early-stage age-related macular degeneration. Heliyon 2017; 3:e00266. [PMID: 28367511 PMCID: PMC5362043 DOI: 10.1016/j.heliyon.2017.e00266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of vision loss in elderly individuals throughout the developed world. Inhibitors of vascular endothelial growth factor have been successfully used to treat choroidal neovascularization in late-stage AMD. The pathogenesis of early-stage AMD, however, remains largely unknown, impairing efforts to develop effective therapies that prevent progression to late-stage AMD. To address this, we performed comparative transcriptomics of macular and extramacular retinal pigmented epithelium-choroid (RPE-choroid) tissue from early-stage AMD patients. We found that expression of fatty acid desaturase 1 (FADS1), FADS2, and acetyl-CoA acetyltransferase 2 (ACAT2) is increased in macular but not extramacular tissue, possibly through activation of sterol regulatory element binding factor 1 (SREBF1). Consistent with this, we also found that expression of Fads1 is increased in RPE-choroid in a mouse model of early-stage AMD. In zebrafish, deletion of fads2, which encodes a protein that functions as both Fads1 and Fads2 in other species, enhanced apoptosis in the retina upon exposure to intense light. Similarly, pharmacological inhibition of Srebf1 enhanced apoptosis and reduced fads2 expression in zebrafish exposed to intense light. These results suggest that the SREBF1-FADS1/2 axis may be activated in macular RPE-choroid as a protective response during early-stage AMD and could thus be a therapeutic target for early-stage AMD.
Collapse
|
127
|
Calabrèse A, Liu T, Legge GE. Does Vertical Reading Help People with Macular Degeneration: An Exploratory Study. PLoS One 2017; 12:e0170743. [PMID: 28114373 PMCID: PMC5256925 DOI: 10.1371/journal.pone.0170743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/10/2017] [Indexed: 11/26/2022] Open
Abstract
Individuals with macular degeneration often develop a Preferred Retinal Locus (PRL) used in place of the impaired fovea. It is known that many people adopt a PRL left of the scotoma, which is likely to affect reading by occluding text to the right of fixation. For such individuals, we examined the possibility that reading vertical text, in which words are rotated 90° with respect to the normal horizontal orientation, would be beneficial for reading. Vertically oriented words would be tangential to the scotoma instead of being partially occluded by it. Here we report the results of an exploratory study that aimed at investigating this hypothesis. We trained individuals with macular degeneration who had PRLs left of their scotoma to read text rotated 90° clockwise and presented using rapid serial visual presentation (RSVP). Although training resulted in improved reading of vertical text, the training did not result in reading speeds that appreciably exceeded reading speeds following training with horizontal text. These results do not support the hypothesis that people with left PRLs read faster with vertical text.
Collapse
Affiliation(s)
- Aurélie Calabrèse
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Tingting Liu
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Gordon E. Legge
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
128
|
Ali ZC, Silvioli R, Rajai A, Aslam TM. Feasibility of Use of a Mobile Application for Nutrition Assessment Pertinent to Age-Related Macular Degeneration (MANAGER2). Transl Vis Sci Technol 2017; 6:4. [PMID: 28138414 PMCID: PMC5270627 DOI: 10.1167/tvst.6.1.4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/16/2016] [Indexed: 01/20/2023] Open
Abstract
Purpose This is a feasibility study assessing use of a mobile phone application (app.) to measure nutrient intake relevant to age-related macular degeneration (AMD). Methods Inclusion criteria were age over 40 and ownership of a smartphone. Participants included healthy volunteers and those with ophthalmic conditions. They were asked to record daily food intake for a minimum of 3 days in a paper food diary and the app. A dietician analyzed the food diaries, and an independent researcher analyzed data from the app. Average daily intake of nutrients relevant to AMD (docosahexaenoic acid [DHA], eicosapentaenoic acid [EPA], vitamins E and C, copper, zinc, and lutein + zeaxanthin) were calculated for both and then compared. Results A total of 54 participants completed the app. and food diary. Male-to-female ratio was 7:20. Median (interquartile range [IQR]) age was 57 years (45.3–68.7 years). More than 90% of all values were within the limits of agreement for all micronutrients. Bland Altman agreement plots demonstrated clinically acceptable agreement between the two systems of analysis. Conclusions This study has demonstrated that the app. is a feasible alternative to the food diary for assessing nutrient intake relevant to AMD. Further studies are suggested to assess long-term adherence and effect of the app. on nutrient intake in AMD patients. Translational Relevance After smoking, nutritional modification is the key modifiable factor to reduce incidence of AMD. Use of the app. could be an efficient, easy way to monitor and improve dietary intake of required nutrients pertinent to AMD.
Collapse
Affiliation(s)
- Zaria Christine Ali
- Manchester Royal Eye Hospital, CMFT, Manchester Academic Health Sciences Centre, Manchester, UK
| | | | - Azita Rajai
- Institute of Population Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK ; Research & Innovation, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Tariq Mehmood Aslam
- Manchester Royal Eye Hospital, CMFT, Manchester Academic Health Sciences Centre, Manchester, UK ; Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK ; University of Manchester, Manchester, UK ; Heriot Watt University, Edinburgh Campus, Edinburgh, UK
| |
Collapse
|
129
|
Zieger M, Punzo C. Improved cell metabolism prolongs photoreceptor survival upon retinal-pigmented epithelium loss in the sodium iodate induced model of geographic atrophy. Oncotarget 2016; 7:9620-33. [PMID: 26883199 PMCID: PMC4891071 DOI: 10.18632/oncotarget.7330] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/23/2016] [Indexed: 01/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is characterized by malfunction and loss of retinal-pigmented epithelium (RPE) cells. Because the RPE transfers nutrients from the choriocapillaris to photoreceptor (PR), PRs are affected as well. Geographic atrophy (GA) is an advanced form of AMD characterized by severe vision impairment due to RPE loss over large areas. Currently there is no treatment to delay the degeneration of nutrient deprived PRs once RPE cells die. Here we show that cell-autonomous activation of the key regulator of cell metabolism, the kinase mammalian target of rapamycin complex 1 (mTORC1), delays PR death in the sodium iodate induced model of RPE atrophy. Consistent with this finding loss of mTORC1 in cones accelerates cone death as cones fail to balance demand with supply. Interestingly, promoting rod survival does not promote cone survival in this model of RPE atrophy as both, rods and cones suffer from a sick and dying RPE. The findings suggest that activation of metabolic genes downstream of mTORC1 can serve as a strategy to prolong PR survival when RPE cells malfunction or die.
Collapse
Affiliation(s)
- Marina Zieger
- Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Claudio Punzo
- Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
130
|
Miller JW. Beyond VEGF-The Weisenfeld Lecture. Invest Ophthalmol Vis Sci 2016; 57:6911-6918. [PMID: 28027565 PMCID: PMC5214225 DOI: 10.1167/iovs.16-21201] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 02/05/2023] Open
Abstract
Purpose To review advances made in the treatment of age-related macular degeneration (AMD) and share perspectives on the future of AMD treatment. Methods Review of published clinical and experimental studies. Results Inhibitors of vascular endothelial growth factor (VEGF) truly revolutionized the treatment of AMD. However, available results from longer-term studies suggest that a degenerative process is unveiled, and continues to occur, even when neovascularization is controlled. Furthermore, anti-VEGF therapy may play a role in the development of atrophic changes. We have proposed using neuroprotection to prevent atrophy, and multiple models of retinal degeneration have shown that it is necessary to block both apoptotic and necrotic cell death pathways. Despite the success of anti-VEGF therapy and the promise of neuroprotection, neither addresses the underlying cause of AMD. It has been postulated that in early AMD, the retention and abnormal accumulation of lipids in Bruch's membrane and below the retinal pigmented epithelium (RPE) lead to drusen. Thus, it is conceivable to target the retained lipoproteins and seek to remove them. In a case study and pilot multicenter clinical trial, we observed significant regression of drusen and an improvement in visual acuity in patients taking high-dose statin therapy. These results, though preliminary, warrant further investigation. Conclusion Future treatment of AMD should be based on biology, which will require continued elucidation of the pathogenic mechanisms of AMD development. Neuroprotection represents a potential therapeutic approach, and other promising targets include immune pathways (e.g., inflammation, complement, and inflammasomes) and lipid/lipoprotein accumulation. Finally, due to the heterogeneity of AMD, future progress in therapy will benefit from improved phenotyping and classification.
Collapse
Affiliation(s)
- Joan W. Miller
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, Massachusetts, United States
| |
Collapse
|
131
|
Electrophysiological testing of visual function after mirror telescope implantation: a case report. Doc Ophthalmol 2016; 133:171-181. [PMID: 27832406 DOI: 10.1007/s10633-016-9563-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE The implantation of an intraocular telescope increases life quality in patients with end-stage age-related macular degeneration (AMD). The present study monitored changes in electrophysiological markers of visual processing before and during seventeen months after a novel mirror telescope implantation in two patients (OV-male 90 years, MZ-female 70 years) with the final-stage form of AMD. METHODS Visual evoked potentials were recorded to high-contrast pattern-reversal (PR-VEP for check size 40' and 10'), low-contrast motion-onset stimuli (in visual periphery M-VEP M20°, and in central part M-VEP C8°), and event-related potentials (ERPs) in the oddball visual paradigm. RESULTS MZ's more systematic responses showed attenuation and prolongation of the M-VEP M20° and the PR-VEP 40' immediately after the telescope implantation with a slow amplitude recovery with unchanged prolonged latency. The implantation completely eradicated the M-VEP C8° without any restoration. The PR-VEP 10' were not readable. Only a part of OV's PR-VEP 40' and M-VEP M20' were of a repeatable and expected morphology. These OV's VEPs were consistent with MZ's findings. The ERPs did not show any effect of implantation in both patients. Post-implantation visual acuity and reaction time overcame the pre-implantation levels. CONCLUSIONS The mirror telescope preserved peripheral vision in contrast to classic telescopes; however, the telescope concurrently reduced the luminance of the magnified retinal image, which was likely responsible for the prolongation of the VEP latencies.
Collapse
|
132
|
Hassan M, Afridi R, Sadiq MA, Soliman MK, Agarwal A, Sepah YJ, Do DV, Nguyen QD. The role of Aflibercept in the management of age-related macular degeneration. Expert Opin Biol Ther 2016; 16:699-709. [PMID: 26982640 DOI: 10.1517/14712598.2016.1167182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION During the past decade, significant advances have occurred in the management of neovascular age-related macular degeneration (NV-AMD). The advent of anti-vascular endothelial growth factor (anti-VEGF) therapy has shifted the treatment goal of NV-AMD from merely salvaging vision to improving visual acuity and maintaining a good quality of life. Aflibercept (AFL) is a significant addition to the arsenal of anti-VEGF therapies against the NV-AMD. In the index review, pharmacology and efficacy of AFL has been reviewed. AREAS COVERED An extensive literature search was performed to identify preclinical and clinical studies performed to illustrate the role of AFL in NV-AMD. Randomized clinical trials evaluating other anti-VEGF agents were also included for comparison. Additionally, studies where AFL was employed to treat anti-VEGF-resistant cases agents have been reviewed. EXPERT OPINION AFL is an effective agent in the management of NV-AMD and its efficacy has been found to be comparable to ranibizumab (RBZ). Additionally, AFL is a good alternative agent in patients with NV-AMD resistant to RBZ and bevacizumab (BVZ), and can potentially lessen the treatment burden. As more research is conducted, the role of AFL in varying dosing regimens, as monotherapy and in combination with other agents, will become further defined.
Collapse
Affiliation(s)
- Muhammad Hassan
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Rubbia Afridi
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Mohammad Ali Sadiq
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Mohamed Kamel Soliman
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA.,b Department of Ophthalmology , Assiut University , Assiut , Egypt
| | - Aniruddha Agarwal
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Yasir Jamal Sepah
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Diana V Do
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| | - Quan Dong Nguyen
- a Ocular Imaging Research and Reading Center (OIRRC) , Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
133
|
Velazquez-Villoria A, Recalde S, Anter J, Bezunartea J, Hernandez-Sanchez M, García-García L, Alonso E, Ruiz-Moreno JM, Araiz-Iribarren J, Fernandez-Robredo P, García-Layana A. Evaluation of 10 AMD Associated Polymorphisms as a Cause of Choroidal Neovascularization in Highly Myopic Eyes. PLoS One 2016; 11:e0162296. [PMID: 27643879 PMCID: PMC5028023 DOI: 10.1371/journal.pone.0162296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/20/2016] [Indexed: 02/07/2023] Open
Abstract
Choroidal neovascularization (CNV) commonly occurs in age related macular degeneration and pathological myopia patients. In this study we conducted a case-control prospective study including 431 participants. The aim of this study was to determine the potential association between 10 single nucleotide polymorphisms (SNPs) located in 4 different genetic regions (CFI, COL8A1, LIPC, and APOE), and choroidal neovascularization in age-related macular degeneration and the development of choroidal neovascularization in highly myopic eyes of a Caucasian population. Univariate and multivariate logistic regression analysis adjusted for age, sex and hypertension was performed for each allele, genotype and haplotype frequency analysis. We found that in the univariate analysis that both single-nucleotide polymorphisms in COL8A1 gene (rs13095226 and rs669676) together with age, sex and hypertension were significantly associated with myopic CNV development in Spanish patients (p<0.05). After correcting for multiple testing none of the polymorphisms studied remained significantly associated with myopic CNV (p>0.05); however, analysis of the axial length between genotypes of rs13095226 revealed an important influence of COL8A1 in the development of CNV in high myopia. Furthermore we conducted a meta-analysis of COL8A1, CFI and LIPC genes SNPs (rs669676, rs10033900 and rs10468017) and found that only rs669676 of these SNPs were associated with high myopia neovascularization.
Collapse
Affiliation(s)
- Alvaro Velazquez-Villoria
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
- Department of Ophthalmology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sergio Recalde
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
- * E-mail:
| | - Jaouad Anter
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas and Ciber de Enfermedades Raras, Madrid, Spain
| | - Jaione Bezunartea
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
| | | | - Laura García-García
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
| | - Elena Alonso
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
- Department of Ophthalmology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Jose María Ruiz-Moreno
- Department of Ophthalmology, Castilla La Mancha University, Albacete and Baviera European Institute of Retina, Alicante, Spain
| | - Javier Araiz-Iribarren
- University of the Basque Country (Surgical-Clinical Institute of Ophthalmology) and San Eloy Hospital, Bilbao, Spain
| | | | - Alfredo García-Layana
- Ophthalmology Experimental Laboratory, Universidad de Navarra, Pamplona, Spain
- Department of Ophthalmology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
134
|
Corvi F, Souied EH, Falfoul Y, Georges A, Jung C, Querques L, Querques G. Pilot evaluation of short-term changes in macular pigment and retinal sensitivity in different phenotypes of early age-related macular degeneration after carotenoid supplementation. Br J Ophthalmol 2016; 101:770-773. [DOI: 10.1136/bjophthalmol-2016-309115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/24/2016] [Accepted: 08/14/2016] [Indexed: 11/04/2022]
|
135
|
Tan PL, Bowes Rickman C, Katsanis N. AMD and the alternative complement pathway: genetics and functional implications. Hum Genomics 2016; 10:23. [PMID: 27329102 PMCID: PMC4915094 DOI: 10.1186/s40246-016-0079-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is an ocular neurodegenerative disorder and is the leading cause of legal blindness in Western societies, with a prevalence of up to 8 % over the age of 60, which continues to increase with age. AMD is characterized by the progressive breakdown of the macula (the central region of the retina), resulting in the loss of central vision including visual acuity. While its molecular etiology remains unclear, advances in genetics and genomics have illuminated the genetic architecture of the disease and have generated attractive pathomechanistic hypotheses. Here, we review the genetic architecture of AMD, considering the contribution of both common and rare alleles to susceptibility, and we explore the possible mechanistic links between photoreceptor degeneration and the alternative complement pathway, a cascade that has emerged as the most potent genetic driver of this disorder.
Collapse
Affiliation(s)
- Perciliz L Tan
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Catherine Bowes Rickman
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, 27710, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, 27710, USA. .,Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
136
|
ASSOCIATIONS BETWEEN AGE-RELATED MACULAR DEGENERATION, OSTEOARTHRITIS AND RHEUMATOID ARTHRITIS: RECORD LINKAGE STUDY. Retina 2016; 35:2613-8. [PMID: 25996429 DOI: 10.1097/iae.0000000000000651] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE The epidemiologic relationship between age-related macular degeneration (AMD) and arthritis is unknown and has implications for understanding disease pathogenesis and treatment strategies. METHODS An AMD cohort of 245,912 people was constructed from English linked hospital episode statistics (1999-2011), principally comprising neovascular AMD patients undergoing anti-vascular endothelial growth factor therapy. We compared the AMD cohort with a reference cohort (2,134,771 people) for rates of subsequent osteoarthritis (OA) and rheumatoid arthritis. Osteoarthritis (2,032,472 people) and rheumatoid arthritis (261,232 people) cohorts were also constructed and compared with the reference cohort for rates of subsequent AMD. RESULTS Risk of arthritis after AMD was not elevated. The rate ratio for OA was 0.96 (95% confidence interval 0.95-0.97) and for rheumatoid arthritis was 0.98 (0.94-1.02). However, risk of AMD after arthritis was modestly raised. For OA, the rate ratio was 1.06 (1.04-1.08), but risk increased with longer OA duration, for example, 1.15 (1.08-1.23) for >10 years. For rheumatoid arthritis, the rate ratio was also modestly elevated at 1.15 (1.12-1.19). CONCLUSION Age-related macular degeneration and arthritis are degenerative aging conditions that share some disease mechanisms and extracellular matrix involvement. However, considering arthritis after AMD, they are not positively associated. By contrast, people with OA experience modestly increased AMD risk, perhaps owing to medical treatments for OA.
Collapse
|
137
|
Li E, Donati S, Virgili G, Krzystolik MG. Treatment schedules for administration of anti-vascular endothelial growth factor agents for neovascular age-related macular degeneration. Hippokratia 2016. [DOI: 10.1002/14651858.cd012208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Emily Li
- Signature Healthcare Brockton Hospital; Transitional Year Residency Program; 680 Centre Street Brockton Massachusetts USA 02302
| | - Simone Donati
- University of Insubria, Varese-Como; Department of Surgical and Morphological Sciences, Section of Ophthalmology; Via Guicciardini 9 Varese Italy 21100
| | - Gianni Virgili
- University of Florence; Department of Translational Surgery and Medicine, Eye Clinic; Largo Brambilla, 3 Florence Italy 50134
| | - Magdalena G Krzystolik
- Mass Eye and Ear Infirmary; Department of Ophthalmology, Retina Service; 1 Randall Square, Suite 203 Providence Rhode Island USA 02904
| |
Collapse
|
138
|
Cooke Bailey JN, Hoffman JD, Sardell RJ, Scott WK, Pericak-Vance MA, Haines JL. The Application of Genetic Risk Scores in Age-Related Macular Degeneration: A Review. J Clin Med 2016; 5:jcm5030031. [PMID: 26959068 PMCID: PMC4810102 DOI: 10.3390/jcm5030031] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/19/2016] [Accepted: 02/29/2016] [Indexed: 01/08/2023] Open
Abstract
Age-related macular degeneration (AMD), a highly prevalent and impactful disease of aging, is inarguably influenced by complex interactions between genetic and environmental factors. Various risk scores have been tested that assess measurable genetic and environmental contributions to disease. We herein summarize and review the ability and utility of these numerous models for prediction of AMD and suggest additional risk factors to be incorporated into clinically useful predictive models of AMD.
Collapse
Affiliation(s)
- Jessica N Cooke Bailey
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA.
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Joshua D Hoffman
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA.
| | - Rebecca J Sardell
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - William K Scott
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA.
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
139
|
Vavvas DG, Daniels AB, Kapsala ZG, Goldfarb JW, Ganotakis E, Loewenstein JI, Young LH, Gragoudas ES, Eliott D, Kim IK, Tsilimbaris MK, Miller JW. Regression of Some High-risk Features of Age-related Macular Degeneration (AMD) in Patients Receiving Intensive Statin Treatment. EBioMedicine 2016; 5:198-203. [PMID: 27077128 PMCID: PMC4816836 DOI: 10.1016/j.ebiom.2016.01.033] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 12/26/2022] Open
Abstract
Importance Age-related macular degeneration (AMD) remains the leading cause of blindness in developed countries, and affects more than 150 million worldwide. Despite effective anti-angiogenic therapies for the less prevalent neovascular form of AMD, treatments are lacking for the more prevalent dry form. Similarities in risk factors and pathogenesis between AMD and atherosclerosis have led investigators to study the effects of statins on AMD incidence and progression with mixed results. A limitation of these studies has been the heterogeneity of AMD disease and the lack of standardization in statin dosage. Objective We were interested in studying the effects of high-dose statins, similar to those showing regression of atherosclerotic plaques, in AMD. Design Pilot multicenter open-label prospective clinical study of 26 patients with diagnosis of AMD and the presence of many large, soft drusenoid deposits. Patients received 80 mg of atorvastatin daily and were monitored at baseline and every 3 months with complete ophthalmologic exam, best corrected visual acuity (VA), fundus photographs, optical coherence tomography (OCT), and blood work (AST, ALT, CPK, total cholesterol, TSH, creatinine, as well as a pregnancy test for premenopausal women). Results Twenty-three subjects completed a minimum follow-up of 12 months. High-dose atorvastatin resulted in regression of drusen deposits associated with vision gain (+ 3.3 letters, p = 0.06) in 10 patients. No subjects progressed to advanced neovascular AMD. Conclusions High-dose statins may result in resolution of drusenoid pigment epithelial detachments (PEDs) and improvement in VA, without atrophy or neovascularization in a high-risk subgroup of AMD patients. Confirmation from larger studies is warranted. High dose lipophilic statin administration was associated with regression of large soft drusen and vision gain in 10/23 AMD patients. Duration of treatment before a positive response was observed was usually 1–1.5 years. Patients on high-dose statin appeared to be protected from progression to “wet” neovascular-AMD.
There is a lack of effective therapies for dry age-related macular degeneration (AMD), one of the leading causes of blindness affecting millions. Although AMD shares similarities with atherosclerosis, prior studies on statins and AMD have failed to show improvement. A limitation of these studies has been the heterogeneity of AMD disease and the lack of standardization in statin dosage. Here, we present for the first time evidence that treatment with high-dose atorvastatin (80 mg) is associated with regression of lipid deposits and improvement in visual acuity, without atrophy or neovascularization, in high-risk AMD patients.
Collapse
Affiliation(s)
- Demetrios G. Vavvas
- Corresponding authors at: 243 Charles St., Boston, MA 02114, USA.243 Charles St.BostonMA02114USA
| | - Anthony B. Daniels
- Corresponding authors at: 243 Charles St., Boston, MA 02114, USA.243 Charles St.BostonMA02114USA
| | | | | | | | | | | | | | | | | | - Miltiadis K. Tsilimbaris
- Corresponding authors at: 243 Charles St., Boston, MA 02114, USA.243 Charles St.BostonMA02114USA
| | - Joan W. Miller
- Corresponding authors at: 243 Charles St., Boston, MA 02114, USA.243 Charles St.BostonMA02114USA
| |
Collapse
|
140
|
Klettner A. Fucoidan as a Potential Therapeutic for Major Blinding Diseases--A Hypothesis. Mar Drugs 2016; 14:E31. [PMID: 26848666 PMCID: PMC4771984 DOI: 10.3390/md14020031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/12/2016] [Accepted: 01/22/2016] [Indexed: 12/23/2022] Open
Abstract
Fucoidan is a heterogeneous group of sulfated polysaccharide with a high content of l-fucose, which can be extracted from brown algae and marine invertebrates. It has many beneficial biological activities that make fucoidan an interesting candidate for therapeutic application in a variety of diseases. Age-related macular degeneration and diabetic retinopathy are major causes for vision loss and blindness in the industrialized countries and increasingly in the developing world. Some of the characteristics found in certain fucoidans, such as its anti-oxidant activity, complement inhibition or interaction with the Vascular Endothelial Growth factor, which would be of high interest for a potential application of fucoidan in age-related macular degeneration or diabetic retinopathy. However, the possible usage of fucoidan in ophthalmological diseases has received little attention so far. In this review, biological activities of fucoidan that could be of interest regarding these diseases will be discussed.
Collapse
Affiliation(s)
- Alexa Klettner
- Department of Ophthalmology, University Medical Center, University of Kiel, 24105 Kiel, Germany.
| |
Collapse
|
141
|
Gonçalves S, Rodrigues IP, Padrão J, Silva JP, Sencadas V, Lanceros-Mendez S, Girão H, Gama FM, Dourado F, Rodrigues LR. Acetylated bacterial cellulose coated with urinary bladder matrix as a substrate for retinal pigment epithelium. Colloids Surf B Biointerfaces 2015; 139:1-9. [PMID: 26689643 DOI: 10.1016/j.colsurfb.2015.11.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022]
Abstract
This work evaluated the effect of acetylated bacterial cellulose (ABC) substrates coated with urinary bladder matrix (UBM) on the behavior of retinal pigment epithelium (RPE), as assessed by cell adhesion, proliferation and development of cell polarity exhibiting transepithelial resistance and polygonal shaped-cells with microvilli. Acetylation of bacterial cellulose (BC) generated a moderate hydrophobic surface (around 65°) while the adsorption of UBM onto these acetylated substrates did not affect significantly the surface hydrophobicity. The ABS substrates coated with UBM enabled the development of a cell phenotype closer to that of native RPE cells. These cells were able to express proteins essential for their cytoskeletal organization and metabolic function (ZO-1 and RPE65), while showing a polygonal shaped morphology with microvilli and a monolayer configuration. The coated ABC substrates were also characterized, exhibiting low swelling effect (between 1.5-2.0 swelling/mm(3)), high mechanical strength (2048MPa) and non-pyrogenicity (2.12EU/L). Therefore, the ABC substrates coated with UBM exhibit interesting features as potential cell carriers in RPE transplantation that ought to be further explored.
Collapse
Affiliation(s)
- Sara Gonçalves
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Inês Patrício Rodrigues
- Centre of Ophthalmology and Vision Sciences, IBILI-Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Jorge Padrão
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - João Pedro Silva
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Vitor Sencadas
- Center/Department of Physics, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | | | - Henrique Girão
- Centre of Ophthalmology and Vision Sciences, IBILI-Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Francisco M Gama
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Fernando Dourado
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Lígia R Rodrigues
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
142
|
Fernandez-Godino R, Garland DL, Pierce EA. A local complement response by RPE causes early-stage macular degeneration. Hum Mol Genet 2015; 24:5555-69. [PMID: 26199322 PMCID: PMC4572070 DOI: 10.1093/hmg/ddv287] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/06/2015] [Accepted: 07/13/2015] [Indexed: 01/05/2023] Open
Abstract
Inherited and age-related macular degenerations (AMDs) are important causes of vision loss. An early hallmark of these disorders is the formation of sub-retinal pigment epithelium (RPE) basal deposits. A role for the complement system in MDs was suggested by genetic association studies, but direct functional connections between alterations in the complement system and the pathogenesis of MD remain to be defined. We used primary RPE cells from a mouse model of inherited MD due to a p.R345W mutation in EGF-containing fibulin-like extracellular matrix protein 1 (EFEMP1) to investigate the role of the RPE in early MD pathogenesis. Efemp1(R345W) RPE cells recapitulate the basal deposit formation observed in vivo by producing sub-RPE deposits in vitro. The deposits share features with basal deposits, and their formation was mediated by EFEMP1(R345W) or complement component 3a (C3a), but not by complement component 5a (C5a). Increased activation of complement appears to occur in response to an abnormal extracellular matrix (ECM), generated by the mutant EFEMP1(R345W) protein and reduced ECM turnover due to inhibition of matrix metalloproteinase 2 by EFEMP1(R345W) and C3a. Increased production of C3a also stimulated the release of cytokines such as interleukin (IL)-6 and IL-1B, which appear to have a role in deposit formation, albeit downstream of C3a. These studies provide the first direct indication that complement components produced locally by the RPE are involved in the formation of basal deposits. Furthermore, these results suggest that C3a generated by RPE is a potential therapeutic target for the treatment of EFEMP1-associated MD as well as AMD.
Collapse
Affiliation(s)
| | | | - Eric A Pierce
- Ocular Genomics Institute, Department of Ophthalmology and Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
143
|
Aredo B, Li T, Chen X, Zhang K, Wang CXZ, Gou D, Zhao B, He Y, Ufret-Vincenty RL. A chimeric Cfh transgene leads to increased retinal oxidative stress, inflammation, and accumulation of activated subretinal microglia in mice. Invest Ophthalmol Vis Sci 2015; 56:3427-40. [PMID: 26030099 DOI: 10.1167/iovs.14-16089] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Variants of complement factor H (Cfh) affecting short consensus repeats (SCRs) 6 to 8 increase the risk of age-related macular degeneration. Our aim was to explore the effect of expressing a Cfh variant on the in vivo susceptibility of the retina and RPE to oxidative stress and inflammation, using chimeric Cfh transgenic mice (chCfhTg). METHODS The chCfhTg and age-matched C57BL/6J (B6) mice were subjected to oxidative stress by either normal aging, or by exposure to a combination of oral hydroquinone (0.8% HQ) and increased light. Eyes were collected for immunohistochemistry of RPE-choroid flat mounts and of retinal sections, ELISA, electron microscopy, and RPE/microglia gene expression analysis. RESULTS Aging mice to 2 years led to an increased accumulation of basal laminar deposits, subretinal microglia/macrophages (MG/MΦ) staining for CD16 and for malondialdehyde (MDA), and MDA-modified proteins in the retina in chCfhTg compared to B6 mice. The chCfhTg mice maintained on HQ diet and increased light showed greater deposition of basal laminar deposits, more accumulation of fundus spots suggestive of MG/MΦ, and increased deposition of C3d in the sub-RPE space, compared to controls. In addition, chCfhTg mice demonstrated upregulation of NLRP3, IP-10, CD68, and TREM-2 in the RNA isolates from RPE/MG/MΦ. CONCLUSIONS Expression of a Cfh transgene introducing a variant in SCRs 6 to 8 was sufficient to lead to increased retinal/RPE susceptibility to oxidative stress, a proinflammatory MG/MΦ phenotype, and a proinflammatory RPE/MG/MΦ gene expression profile in a transgenic mouse model. Our data suggest that altered interactions of Cfh with MDA-modified proteins may be relevant in explaining the effects of the Cfh variant.
Collapse
Affiliation(s)
- Bogale Aredo
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Tao Li
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States 2Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiao Chen
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Kaiyan Zhang
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Cynthia Xin-Zhao Wang
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Darlene Gou
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Biren Zhao
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | - Yuguang He
- Department of Ophthalmology UT Southwestern Medical Center, Dallas, Texas, United States
| | | |
Collapse
|
144
|
Juel HB, Faber C, Munthe-Fog L, Bastrup-Birk S, Reese-Petersen AL, Falk MK, Singh A, Sørensen TL, Garred P, Nissen MH. Systemic and Ocular Long Pentraxin 3 in Patients with Age-Related Macular Degeneration. PLoS One 2015; 10:e0132800. [PMID: 26176960 PMCID: PMC4503310 DOI: 10.1371/journal.pone.0132800] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/19/2015] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) has been associated with both systemic and ocular alterations of the immune system. In particular dysfunction of complement factor H (CFH), a soluble regulator of the alternative pathway of the complement system, has been implicated in AMD pathogenesis. One of the ligands for CFH is long pentraxin 3 (PTX3), which is produced locally in the retinal pigment epithelium (RPE). To test the hypothesis that PTX3 is relevant to retinal immunohomeostasis and may be associated with AMD pathogenesis, we measured plasma PTX3 protein concentration and analyzed the RPE/choroid PTX3 gene expression in patients with AMD. To measure the ability of RPE cells to secrete PTX3 in vitro, polarized ARPE-19 cells were treated with activated T cells or cytokines (interferon (IFN)-gamma and/or tumor necrosis factor (TNF)-alpha) from the basolateral side; then PTX3 protein concentration in supernatants and PTX3 gene expression in tissue lysates were quantified. Plasma levels of PTX3 were generally low and did not significantly differ between patients and controls (P=0.307). No statistically significant difference was observed between dry and exudative AMD nor was there any correlation with hsCRP or CFH genotype. The gene expression of PTX3 increased in RPE/choroid with age (P=0.0098 macular; P=0.003 extramacular), but did not differ between aged controls and AMD patients. In vitro, ARPE-19 cells increased expression of the PTX3 gene as well PTX3 apical secretions after stimulation with TNF-alpha or activated T cells (P<0.01). These findings indicate that PTX3 expressed in the eye cannot be detected systemically and systemic PTX3 may have little or no impact on disease progression, but our findings do not exclude that locally produced PTX3 produced in the posterior segment of the eye may be part of the AMD immunopathogenesis.
Collapse
Affiliation(s)
- Helene Bæk Juel
- Eye Research Unit, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Faber
- Eye Research Unit, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Glostrup Hospital, Glostrup, Denmark
- * E-mail:
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Simone Bastrup-Birk
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alexander Lynge Reese-Petersen
- Eye Research Unit, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Krüger Falk
- Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark
| | - Amardeep Singh
- Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Copenhagen University Hospital, Roskilde, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mogens Holst Nissen
- Eye Research Unit, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
145
|
Morrison MA, Magalhaes TR, Ramke J, Smith SE, Ennis S, Simpson CL, Portas L, Murgia F, Ahn J, Dardenne C, Mayne K, Robinson R, Morgan DJ, Brian G, Lee L, Woo SJ, Zacharaki F, Tsironi EE, Miller JW, Kim IK, Park KH, Bailey-Wilson JE, Farrer LA, Stambolian D, DeAngelis MM. Ancestry of the Timorese: age-related macular degeneration associated genotype and allele sharing among human populations from throughout the world. Front Genet 2015. [PMID: 26217379 PMCID: PMC4496576 DOI: 10.3389/fgene.2015.00238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We observed that the third leading cause of blindness in the world, age-related macular degeneration (AMD), occurs at a very low documented frequency in a population-based cohort from Timor-Leste. Thus, we determined a complete catalog of the ancestry of the Timorese by analysis of whole exome chip data and haplogroup analysis of SNP genotypes determined by sequencing the Hypervariable I and II regions of the mitochondrial genome and 17 genotyped YSTR markers obtained from 535 individuals. We genotyped 20 previously reported AMD-associated SNPs in the Timorese to examine their allele frequencies compared to and between previously documented AMD cohorts of varying ethnicities. For those without AMD (average age > 55 years), genotype and allele frequencies were similar for most SNPs with a few exceptions. The major risk allele of HTRA1 rs11200638 (10q26) was at a significantly higher frequency in the Timorese, as well as 3 of the 5 protective CFH (1q32) SNPs (rs800292, rs2284664, and rs12066959). Additionally, the most commonly associated AMD-risk SNP, CFH rs1061170 (Y402H), was also seen at a much lower frequency in the Korean and Timorese populations than in the assessed Caucasian populations (C ~7 vs. ~40%, respectively). The difference in allele frequencies between the Timorese population and the other genotyped populations, along with the haplogroup analysis, also highlight the genetic diversity of the Timorese. Specifically, the most common ancestry groupings were Oceanic (Melanesian and Papuan) and Eastern Asian (specifically Han Chinese). The low prevalence of AMD in the Timorese population (2 of 535 randomly selected participants) may be due to the enrichment of protective alleles in this population at the 1q32 locus.
Collapse
Affiliation(s)
- Margaux A Morrison
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Tiago R Magalhaes
- National Children's Research Centre, Our Lady's Children's Hospital Dublin, Ireland ; Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin Dublin, Ireland
| | | | - Silvia E Smith
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Sean Ennis
- Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin Dublin, Ireland ; National Centre for Medical Genetics, Our Lady's Children's Hospital Dublin, Ireland
| | - Claire L Simpson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health Baltimore, MD, USA
| | - Laura Portas
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health Baltimore, MD, USA ; Institute of Population Genetics, The National Research Council Sassari, Italy
| | - Federico Murgia
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health Baltimore, MD, USA ; Institute of Population Genetics, The National Research Council Sassari, Italy
| | - Jeeyun Ahn
- Department of Ophthalmology, Seoul National University College of Medicine Seoul, South Korea ; Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center Seoul, South Korea
| | - Caitlin Dardenne
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Katie Mayne
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Rosann Robinson
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Denise J Morgan
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| | - Garry Brian
- The Fred Hollows Foundation New Zealand Auckland, New Zealand
| | - Lucy Lee
- The Fred Hollows Foundation New Zealand Auckland, New Zealand ; London School of Hygiene and Tropical Medicine, University of London London, UK
| | - Se J Woo
- Department of Ophthalmology, Seoul National University College of Medicine Seoul, South Korea ; Department of Ophthalmology, Seoul National University Bundang Hospital Seoungnam, South Korea
| | - Fani Zacharaki
- Department of Ophthalmology, University of Thessaly School of Medicine Larissa, Greece
| | - Evangelia E Tsironi
- Department of Ophthalmology, University of Thessaly School of Medicine Larissa, Greece
| | - Joan W Miller
- Retina Service and Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School Boston, MA, USA
| | - Ivana K Kim
- Retina Service and Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School Boston, MA, USA
| | - Kyu H Park
- Department of Ophthalmology, Seoul National University College of Medicine Seoul, South Korea ; Department of Ophthalmology, Seoul National University Bundang Hospital Seoungnam, South Korea
| | - Joan E Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health Baltimore, MD, USA
| | - Lindsay A Farrer
- Departments of Medicine, Ophthalmology, Neurology, Epidemiology, and Biostatistics, Boston University Schools of Medicine and Public Health Boston, MA, USA
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania Philadelphia, PA, USA
| | - Margaret M DeAngelis
- Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah Salt Lake City, UT, USA
| |
Collapse
|
146
|
Modifying Choroidal Neovascularization Development with a Nutritional Supplement in Mice. Nutrients 2015; 7:5423-42. [PMID: 26153682 PMCID: PMC4517006 DOI: 10.3390/nu7075229] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 12/23/2022] Open
Abstract
We examined the effect of nutritional supplements (modified Age Related Eye Disease Study (AREDS)-II formulation containing vitamins, minerals, lutein, resveratrol, and omega-3 fatty acids) on choroidal neovascularization (CNV). Supplements were administered alone and combined with intravitreal anti-VEGF in an early-CNV (diode laser-induced) murine model. Sixty mice were evenly divided into group V (oral vehicle, intravitreal saline), group S (oral supplement, intravitreal saline), group V + aVEGF (oral vehicle, intravitreal anti-VEGF), and group S + aVEGF (oral supplement, intravitreal anti-VEGF). Vehicle and nutritional supplements were administered daily for 38 days beginning 10 days before laser. Intravitreal injections were administered 48 h after laser. Fluorescein angiography (FA) and flat-mount CD31 staining evaluated leakage and CNV lesion area. Expression of VEGF, MMP-2 and MMP-9 activity, and NLRP3 were evaluated with RT-PCR, zymography, and western-blot. Leakage, CNV size, VEGF gene and protein expression were lower in groups V + aVEGF, S + aVEGF, and S than in V (all p < 0.05). Additionally, MMP-9 gene expression differed between groups S + aVEGF and V (p < 0.05) and MMP-9 activity was lower in S + aVEGF than in V and S (both p < 0.01). Levels of MMP-2 and NLRP3 were not significantly different between groups. Nutritional supplements either alone or combined with anti-VEGF may mitigate CNV development and inhibit retinal disease involving VEGF overexpression and CNV.
Collapse
|
147
|
Reprint of: Aspirin use and early age-related macular degeneration: a meta-analysis. Can J Ophthalmol 2015; 50 Suppl 1:S29-33. [DOI: 10.1016/j.jcjo.2015.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/02/2013] [Indexed: 12/20/2022]
|
148
|
SanGiovanni JP, Chen J, Gupta AS, Smith LEH, Sapieha P, Lee PH. Netrin-1 - DCC Signaling Systems and Age-Related Macular Degeneration. PLoS One 2015; 10:e0125548. [PMID: 25950802 PMCID: PMC4423995 DOI: 10.1371/journal.pone.0125548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 03/25/2015] [Indexed: 01/04/2023] Open
Abstract
We conducted a nested candidate gene study and pathway-based enrichment analysis on data from a multi-national 77,000-person project on the molecular genetics of age-related macular degeneration (AMD) to identify AMD-associated DNA-sequence variants in genes encoding constituents of a netrin-1 (NTN1)-based signaling pathway that converges on DNA-binding transcription complexes through a 3'-5'-cyclic adenosine monophosphate-calcineurin (cAMP-CN)-dependent axis. AMD-associated single nucleotide polymorphisms (SNPs) existed in 9 linkage disequilibrium-independent genomic regions; these included loci overlapping NTN1 (rs9899630, P ≤ 9.48 x 10-5), DCC (Deleted in Colorectal Cancer)—the gene encoding a primary NTN1 receptor (rs8097127, P ≤ 3.03 x 10-5), and 6 other netrin-related genes. Analysis of the NTN1-DCC pathway with exact methods demonstrated robust enrichment with AMD-associated SNPs (corrected P-value = 0.038), supporting the idea that processes driven by NTN1-DCC signaling systems operate in advanced AMD. The NTN1-DCC pathway contains targets of FDA-approved drugs and may offer promise for guiding applied clinical research on preventive and therapeutic interventions for AMD.
Collapse
Affiliation(s)
- John Paul SanGiovanni
- National Institute of Alcohol Abuse and Alcoholism, Section on Nutritional Neuroscience, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail:
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA, United States of America
| | - Ankur S. Gupta
- University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Lois E. H. Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA, United States of America
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Phil H. Lee
- Analytic & Translational Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
149
|
Faber C, Jehs T, Juel HB, Singh A, Falk MK, Sørensen TL, Nissen MH. Early and exudative age-related macular degeneration is associated with increased plasma levels of soluble TNF receptor II. Acta Ophthalmol 2015; 93:242-7. [PMID: 25363549 DOI: 10.1111/aos.12581] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/23/2014] [Indexed: 02/03/2023]
Abstract
PURPOSE We have recently identified homeostatic alterations in the circulating T cells of patients with age-related macular degeneration (AMD). In cultures of retinal pigment epithelial cells, we have demonstrated that T-cell-derived cytokines induced the upregulation of complement, chemokines and other proteins implicated in AMD pathogenesis. The purpose of this study was to test whether increased plasma levels of cytokines were present in patients with AMD. METHODS We conducted a case-control study. Age-related macular degeneration status was assessed using standardized multimodal imaging techniques. Plasma was isolated from freshly drawn peripheral venous blood samples and analysed for interleukin (IL)15, IL18, interferon (IFN)γ, soluble tumour necrosis factor (TNF) receptor II (sTNFRII) and complement factor H (CFH) Y402H genotype. RESULTS We included 136 individuals with early or late forms of AMD and 74 controls. Significantly increased levels of sTNFRII were observed in patients with early or exudative AMD (p < 0.01). After adjusting for CFH Y402H genotype, age, sex and smoking history, the level of sTNFRII remained a significant predictor for prevalence of AMD with odds ratios at 3.0 in the middle and 3.6 in the highest tertiles. Levels of IL15, IL18 and IFNγ were low and not associated with AMD. CONCLUSIONS Increased plasma level of sTNFRII is found to be associated with AMD. The data supports the observations of low-grade, systemic inflammatory alterations in patients with AMD. However, it remains to be determined whether increased levels of TNFα can be found, which directly reflects an increased activity of macrophages and T cells.
Collapse
Affiliation(s)
- Carsten Faber
- Department of International Health, Immunology and Microbiology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen N Denmark
- Department of Ophthalmology Glostrup Hospital Glostrup Denmark
| | - Tina Jehs
- Department of International Health, Immunology and Microbiology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen N Denmark
| | - Helene Bæk Juel
- Department of International Health, Immunology and Microbiology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen N Denmark
| | - Amardeep Singh
- Department of Ophthalmology Copenhagen University Hospital Roskilde Roskilde Denmark
- Faculty of Health Sciences University of Copenhagen Copenhagen Denmark
| | - Mads Krüger Falk
- Department of Ophthalmology Glostrup Hospital Glostrup Denmark
- Department of Ophthalmology Copenhagen University Hospital Roskilde Roskilde Denmark
- Faculty of Health Sciences University of Copenhagen Copenhagen Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology Copenhagen University Hospital Roskilde Roskilde Denmark
- Faculty of Health Sciences University of Copenhagen Copenhagen Denmark
| | - Mogens Holst Nissen
- Department of International Health, Immunology and Microbiology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen N Denmark
| |
Collapse
|
150
|
Venkatesh A, Ma S, Le YZ, Hall MN, Rüegg MA, Punzo C. Activated mTORC1 promotes long-term cone survival in retinitis pigmentosa mice. J Clin Invest 2015; 125:1446-58. [PMID: 25798619 DOI: 10.1172/jci79766] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inherited photoreceptor degenerative disorder that results in blindness. The disease is often caused by mutations in genes that are specific to rod photoreceptors; however, blindness results from the secondary loss of cones by a still unknown mechanism. Here, we demonstrated that the mammalian target of rapamycin complex 1 (mTORC1) is required to slow the progression of cone death during disease and that constitutive activation of mTORC1 in cones is sufficient to maintain cone function and promote long-term cone survival. Activation of mTORC1 in cones enhanced glucose uptake, retention, and utilization, leading to increased levels of the key metabolite NADPH. Moreover, cone death was delayed in the absence of the NADPH-sensitive cell death protease caspase 2, supporting the contribution of reduced NADPH in promoting cone death. Constitutive activation of mTORC1 preserved cones in 2 mouse models of RP, suggesting that the secondary loss of cones is caused mainly by metabolic deficits and is independent of a specific rod-associated mutation. Together, the results of this study address a longstanding question in the field and suggest that activating mTORC1 in cones has therapeutic potential to prolong vision in RP.
Collapse
|