1
|
Miller JML, Thompson BR, Handa JT, Luthert P, Chakravarthy U, Csaky KG, Bird A, Young BK, Iyengar SK, Baek J, Zouache MA, Richards BT, Hageman GS, Rodrigues G, Bharti K, Flannery JG, Gorin MB, Bowes Rickman C. Dissecting the biological complexity of age-related macular degeneration: Is it one disease, multiple separate diseases, or a spectrum? Exp Eye Res 2025; 254:110304. [PMID: 39983974 DOI: 10.1016/j.exer.2025.110304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Clinicians recognize the heterogeneity of age-related macular degeneration (AMD) in presentation, progression, and treatment response, as well as the challenges in distinguishing it from other macular degenerations. As part of the 2024 Ryan Initiative for Macular Research meeting, a group of clinician-scientists and basic scientists were convened to consider the question of whether AMD should be classified as a single disorder or a spectrum of conditions. To answer this question, we reviewed research on several "dimensions" that constitute AMD risk or pathogenesis: genetics, ancestry, retinal imaging findings, diet and environment, aging, and outer retinal molecular and cellular pathways. The group reached a consensus that AMD represents a heterogeneous collection of disease states arising from the interplay of these dimensions. This heterogeneity can be conceived of as a "cloud" of AMD phenotypes. Defining subtypes within this "cloud" requires longitudinal cohorts of well-genotyped and phenotyped patients who progress from no AMD through late AMD, analyzed by unsupervised learning. Comparing the AMD subtypes that emerge from this analysis, especially -omics data from each subtype, will illuminate biology that is applicable to certain subtypes of AMD patients and molecular pathogenic mechanisms that universally apply to all AMD. This knowledge will, in turn, drive improved drug development.
Collapse
Affiliation(s)
- Jason M L Miller
- University of Michigan, Kellogg Eye Center and Cellular and Molecular Biology Program, Ann Arbor, MI, USA.
| | - Benjamin R Thompson
- Northwestern University Feinberg School of Medicine, Department of Ophthalmology and Feinberg Cardiovascular and Renal Research Inst., Chicago, IL, USA
| | - James T Handa
- Johns Hopkins Medical School, Wilmer Eye Institute, Baltimore, MD, USA
| | | | - Usha Chakravarthy
- Queens University of Belfast, Center for Public Health, Belfast, Ireland
| | - Karl G Csaky
- Retina Foundation of the Southwest, Dallas, TX, USA
| | - Alan Bird
- UCL Institute of Ophthalmology, London, United Kingdom
| | - Benjamin K Young
- Oregon Health Sciences University, Department of Ophthalmology, Portland, OR, USA
| | - Sudha K Iyengar
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, OH, USA
| | - Jiwon Baek
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, South Korea
| | - Moussa A Zouache
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Burt T Richards
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Gregory S Hageman
- University of Utah, Department of Ophthalmology and Visual Sciences, Salt Lake City, UT, USA
| | - Gerry Rodrigues
- Allergan/Abbvie, Ophthalmology Discovery Research, Laguna Niguel, CA, USA
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - John G Flannery
- University of California, Berkeley, Department of Neuroscience, Berkeley, CA, USA
| | - Michael B Gorin
- David Geffen School of Medicine, Stein Eye Institute, Los Angeles, CA, USA.
| | - Catherine Bowes Rickman
- Duke University Medical Center, Departments of Ophthalmology and of Cell Biology, Durham, NC, USA.
| |
Collapse
|
2
|
Biswas P, Woodard DR, Hollingsworth T, Khan NW, Lazaro DR, Berry AM, Dagar M, Pan Y, Garland D, Shaw PX, Oka C, Iwata T, Jablonski MM, Ayyagari R. Ablation of Htra1 leads to sub-RPE deposits and photoreceptor abnormalities. JCI Insight 2025; 10:e178827. [PMID: 39927462 PMCID: PMC11948579 DOI: 10.1172/jci.insight.178827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 12/13/2024] [Indexed: 02/11/2025] Open
Abstract
The high-temperature requirement A1 (HTRA1), a serine protease, has been demonstrated to play a pivotal role in the extracellular matrix (ECM) and has been reported to be associated with the pathogenesis of age-related macular degeneration (AMD). To delineate its role in the retina, the phenotype of homozygous Htra1-KO (Htra1-/-) mice was characterized to examine the effect of Htra1 loss on the retina and retinal pigment epithelium (RPE) with age. The ablation of Htra1 led to a significant reduction in rod and cone photoreceptor function, primary cone abnormalities followed by rods, and atrophy in the RPE compared with WT mice. Ultrastructural analysis of Htra1-/- mice revealed RPE and Bruch's membrane (BM) abnormalities, including the presence of sub-RPE deposits at 5 months (m) that progressed with age accompanied by increased severity of pathology. Htra1-/- mice also displayed alterations in key markers for inflammation, autophagy, and lipid metabolism in the retina. These results highlight the crucial role of HTRA1 in the retina and RPE. Furthermore, this study allows for the Htra1-/- mouse model to be utilized for deciphering mechanisms that lead to sub-RPE deposit phenotypes including AMD.
Collapse
Affiliation(s)
- Pooja Biswas
- Shiley Eye Institute, UCSD, La Jolla, California, USA
| | | | - T.J. Hollingsworth
- The Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Naheed W. Khan
- Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Manisha Dagar
- Shiley Eye Institute, UCSD, La Jolla, California, USA
| | - Yang Pan
- The National Institute of Sensory Organs (NISO), NHO Tokyo Medical Center, Tokyo, Japan
| | | | - Peter X. Shaw
- Shiley Eye Institute, UCSD, La Jolla, California, USA
| | - Chio Oka
- Functional Genomics and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Takeshi Iwata
- The National Institute of Sensory Organs (NISO), NHO Tokyo Medical Center, Tokyo, Japan
| | - Monica M. Jablonski
- The Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | |
Collapse
|
3
|
Domalpally A, Haas AM, Chandra S, VanderZee B, S Dimopoulos I, D L Keenan T, W Pak J, G Csaky K, A Blodi B, Sivaprasad S. Photoreceptor assessment in age-related macular degeneration. Eye (Lond) 2025; 39:284-295. [PMID: 39578549 PMCID: PMC11751396 DOI: 10.1038/s41433-024-03462-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/02/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
Clinical trials investigating drugs for various stages of age-related macular degeneration (AMD) are actively underway and there is a strong interest in outcomes that demonstrate a structure-function-correlation. The ellipsoid zone (EZ), a crucial anatomical feature affected in this disease, has emerged as a strong contender. There is significant interest in evaluating EZ metrics on Optical Coherence Tomography (OCT), such as integrity and reflectivity, as disruption of this photoreceptor-rich layer may indicate disease progression. Loss of photoreceptor integrity in the junctional zone of geographic atrophy (GA) has been shown to exceed the areas of retinal pigment epithelial (RPE) atrophy, thus predicting future GA expansion. Furthermore, reduced visual acuity and retinal sensitivity have been correlated with loss of EZ integrity, underscoring a structure-function relationship. Photoreceptor integrity has also recently been acknowledged by the Food and Drug Administration (FDA), supporting its use as a primary endpoint in clinical trials investigating treatments for GA. However, the segmentation of this EZ still poses challenges. Continuous enhancements in OCT resolution and advancements in automated segmentation algorithms contribute to improved assessment of the EZ, strengthening its potential as an imaging biomarker for assessing photoreceptor function. It remains to be seen whether the EZ will serve as a surrogate marker for intermediate AMD. This article aims to provide an overview of the current understanding and knowledge of the EZ, while addressing ongoing challenges encountered in its assessment and interpretation.
Collapse
Affiliation(s)
- Amitha Domalpally
- Wisconsin Reading Center, Dept of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, USA.
| | - Anna-Maria Haas
- Wisconsin Reading Center, Dept of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, USA
- Karl Landsteiner Institute for Retinal Research and Imaging, Juchgasse 25, 1030, Vienna, Austria
- Department of Ophthalmology, Clinic Landstraße, Vienna Healthcare Group, Juchgasse 25, 1030, Vienna, Austria
| | - Shruti Chandra
- Moorfields Clinical Research Facility, NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Brandon VanderZee
- Wisconsin Reading Center, Dept of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, USA
| | | | - Tiarnan D L Keenan
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeong W Pak
- Wisconsin Reading Center, Dept of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, USA
| | - Karl G Csaky
- Retina Foundation of the Southwest, Dallas, TX, USA
| | - Barbara A Blodi
- Wisconsin Reading Center, Dept of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, USA
| | - Sobha Sivaprasad
- Moorfields Clinical Research Facility, NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
4
|
Antropoli A, Bianco L, Romano F, Trinco A, Arrigo A, Benadji A, Atia R, Palacci O, Dagostinoz D, Devisme C, Condroyer C, Antonio A, Bosello F, Casati S, Salvetti AP, Zaffalon C, Gaudric A, Sahel JA, Staurenghi G, Bandello F, Sennlaub F, Zeitz C, Meunier I, Battaglia Parodi M, Audo I. Extensive macular atrophy with pseudodrusen-like appearance (EMAP) clinical characteristics, diagnostic criteria, and insights from allied inherited retinal diseases and age-related macular degeneration. Prog Retin Eye Res 2025; 104:101320. [PMID: 39603590 DOI: 10.1016/j.preteyeres.2024.101320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Extensive macular atrophy with pseudodrusen-like appearance (EMAP) was first described in France in 2009 as a symmetric and rapidly progressive form of macular atrophy primarily affecting middle-aged individuals. Despite the recent identification of a significant number of cases in Italy and worldwide, EMAP remains an underrecognized condition. The clinical triad typical of EMAP consists of vertically oriented macular atrophy with multilobular borders, pseudodrusen-like deposits across the posterior pole and mid-periphery, and peripheral pavingstone degeneration. Nonetheless, recent research has portrayed EMAP as a highly stage-dependent condition, allowing the identification of novel disease hallmarks, including a diffuse separation between the Bruch's membrane and the retinal pigment epithelium, along with consistent sparing of a region temporal to the macula. Additionally, retinal electrophysiology is particularly useful in distinguishing EMAP from age-related macular degeneration (AMD). Supported by unpublished data from the largest EMAP cohorts worldwide, this review aims to provide a comprehensive and updated description of EMAP, now recognized as a severely blinding disease characterized by diffuse chorioretinal atrophy and photoreceptor dysfunction. Furthermore, we propose a set of diagnostic criteria that incorporate clinical, imaging, and functional tests, to facilitate the recognition of this clinical entity. Lastly, we aim to shed light on its pathogenesis by comparing it with AMD and monogenic retinal disorders exhibiting similar phenotypes.
Collapse
Affiliation(s)
- Alessio Antropoli
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France; Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France; Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Romano
- Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Trinco
- Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Arrigo
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Clinic, Department of Biomedical and Clinical Science, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Amine Benadji
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Raphaël Atia
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Oana Palacci
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Dorothée Dagostinoz
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | - Céline Devisme
- CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France
| | | | - Aline Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Francesca Bosello
- Ophthalmic Unit, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Stefano Casati
- Ophthalmic Unit, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna Paola Salvetti
- Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Zaffalon
- Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Ophthalmic Unit, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Alain Gaudric
- Ophthalmology Center for Imaging and Laser, Paris, France; Department of Ophthalmology, AP-HP, Hôpital Lariboisière, Université Paris Cité, Paris, France
| | - José-Alain Sahel
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Giovanni Staurenghi
- Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Bandello
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Isabelle Meunier
- National Reference Center for Inherited Sensory Diseases, University Hospital of Montpellier, University of Montpellier, Montpellier, France
| | | | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and DHU Sight Restore, INSERM-DGOS CIC1423, Paris, France.
| |
Collapse
|
5
|
Pu J, Zhuang X, Li M, Hao X, He G, Su Y, Gan Y, Zhang X, Ji Y, Mi L, Zhang Y, Yang R, Chen X, Wen F. Prognostic value of macular neovascularisation characteristics for photoreceptor integrity in nAMD: a prospective observational study. Br J Ophthalmol 2024:bjo-2024-326319. [PMID: 39694604 DOI: 10.1136/bjo-2024-326319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE To explore the relationship between characteristics of macular neovascularisation (MNV) and photoreceptor integrity in patients with neovascular age-related macular degeneration (nAMD). METHODS This prospective study enrolled treatment-naïve nAMD eyes and conducted a 3-month follow-up. 16 quantitative MNV features were evaluated using optical coherence tomography angiography, and the impaired areas of ellipsoid zone (EZ), external limiting membrane (ELM) and outer nuclear layer (ONL) were obtained using optical coherence tomography. Correlation and regression analyses assessed the relationships between MNV features and photoreceptor integrity. RESULTS 110 nAMD eyes from 110 patients (73.64% men) were included. Baseline MNV characteristics, including MNV perimeter, maxFeret, minFeret, vessel area, total vessel length, total number of junctions and endpoints, and mean E lacunarity, were positively correlated with photoreceptor damage areas (r ranging from 0.227 to 0.558, p<0.05 for all). Meanwhile, vessel density negatively correlated with photoreceptor damage (r=-0.468 for EZ, -0.394 for ELM and -0.538 for ONL, all p<0.05). After the loading phase, the EZ prognosis was independently associated with baseline MNV minFeret (Std β=0.362, p=0.011) and mean E lacunarity (Std β=0.130, p=0.041). The prognosis for ELM was independently linked to baseline MNV minFeret (Std β=0.373, p=0.014), while no significant factors were found to influence ONL prognosis (p>0.05 for all). CONCLUSION A strong correlation was observed between MNV features and photoreceptor integrity, with larger and more complex vascular networks associated with greater photoreceptor damage.
Collapse
Affiliation(s)
- Jiaxin Pu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Xuenan Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510060, Guangdong, China
| | - Miaoling Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Xinlei Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Guiqin He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Yongyue Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Yuhong Gan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Xiongze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Yuying Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Lan Mi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Yining Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Ruijun Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Xuelin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou 510060, Guangdong, China
| |
Collapse
|
6
|
Goerdt L, Swain TA, Kar D, McGwin G, Berlin A, Clark ME, Owsley C, Sloan KR, Curcio CA. Band Visibility in High-Resolution Optical Coherence Tomography Assessed With a Custom Review Tool and Updated, Histology-Derived Nomenclature. Transl Vis Sci Technol 2024; 13:19. [PMID: 39671227 PMCID: PMC11645748 DOI: 10.1167/tvst.13.12.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/16/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose For structure-function research at the transition of aging to age-related macular degeneration, we refined the current consensus optical coherence tomography (OCT) nomenclature and evaluated a novel review software for investigational high-resolution OCT imaging (HR-OCT; <3 µm axial resolution). Method Volume electron microscopy, immunolocalizations, histology, and investigational devices informed a refined OCT nomenclature for a custom ImageJ-based review tool to assess retinal band visibility. We examined effects on retinal band visibility of automated real-time averaging (ART) 9 and 100 (11 eyes of 10 healthy young adults), aging (10 young vs 22 healthy aged), and age-related macular degeneration (AMD; 22 healthy aged, 17 early (e)AMD, 15 intermediate (i)AMD). Intrareader reliability was assessed. Results Bands not included in consensus nomenclature are now visible using HR-OCT: inner plexiform layer (IPL) 1-5, outer plexiform layer (OPL) 1-2, outer segment interdigitation zone 1-2 (OSIZ, including hyporeflective outer segments), and retinal pigment epithelium (RPE) 1-5. Cohen's kappa was 0.54-0.88 for inner and 0.67-0.83 for outer retinal bands in a subset of 10 eyes. IPL-3-5 and OPL-2 visibility benefitted from increased ART. OSIZ-2 and RPE-1,2,3,5 visibility was worse in aged eyes than in young eyes. OSIZ-1-2, RPE-1, and RPE-5 visibility decreased in eAMD and iAMD compared to healthy aged eyes. Conclusions We reliably identified 28 retinal bands using a novel review tool for HR-OCT. Image averaging improved inner retinal band visibility. Aging and AMD development impacted outer retinal band visibility. Translational Significance Detailed knowledge of anatomic structures visible on OCT will enhance precision in research, including AI training and structure-function analyses.
Collapse
Affiliation(s)
- Lukas Goerdt
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Thomas A. Swain
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Deepayan Kar
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Gerald McGwin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, AL, USA
| | - Andreas Berlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
- Department of Ophthalmology, University of Würzburg, Würzburg, Germany
| | - Mark E. Clark
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Cynthia Owsley
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Kenneth R. Sloan
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| |
Collapse
|
7
|
Kumar H, Guymer RH, Hodgson LA, Hadoux X, Jannaud M, van Wijngaarden P, Luu CD, Wu Z. Reticular Pseudodrusen: Impact of Their Presence and Extent on Local Rod Function in Age-Related Macular Degeneration. OPHTHALMOLOGY SCIENCE 2024; 4:100551. [PMID: 39161750 PMCID: PMC11331943 DOI: 10.1016/j.xops.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 08/21/2024]
Abstract
Purpose To understand the spatial relationship between local rod-mediated visual function and reticular pseudodrusen (RPD) in eyes with large drusen. Design Retrospective cross-sectional study. Participants One eye with large drusen (>125 μm) each from 91 individuals with intermediate age-related macular degeneration, with and without RPD. Methods All participants underwent dark adaptation testing using a dark-adapted chromatic perimeter, where visual sensitivities were measured over 30 minutes of dark adaptation after photobleach. The rod intercept time (RIT; a measure of dynamic rod function) and pointwise sensitivity difference (PWSD; a relative measure of rod- compared with cone-mediated function) was determined at multiple retinal locations, and their association with the overall (central 20° × 20° region) and local (2° diameter region centered on the location tested) extent of RPD and drusen (quantified using multimodal imaging) was examined. Main Outcome Measures Association between overall and local extent of RPD and drusen with RIT and PWSD at each retinal location tested. Results In a multivariable analysis, delayed RIT was associated with an increasing overall (P < 0.001), but not local (P = 0.884), extent of RPD. In contrast, the increasing local (P < 0.001), but not overall (P = 0.475), extent of drusen was associated with delayed RIT. Furthermore, only an increasing overall extent of RPD (P < 0.001) was associated with reduced PWSD (or worse rod compared with cone function), but not the local extent of RPD and drusen, or overall extent of drusen (P ≥ 0.344). Conclusions Local rod-mediated function was associated with the overall, rather than local, extent of RPD in eyes with large drusen, suggesting that there may be widespread pathologic changes in eyes with RPD that account for this. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Himeesh Kumar
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Robyn H. Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Lauren A.B. Hodgson
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Xavier Hadoux
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Maxime Jannaud
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Chi D. Luu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Zhichao Wu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| |
Collapse
|
8
|
Sacconi R, Fazzari G, Capuano V, Menean M, Beretta F, El Matri K, Bandello F, Souied E, Querques G. Pachy-Reticular Pseudodrusen. Ophthalmol Retina 2024; 8:1066-1073. [PMID: 38844200 DOI: 10.1016/j.oret.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE To characterize the features of a peculiar association between reticular pseudodrusen (RPD) and pachychoroid (pachy-RPD) and to compare them with eyes affected by RPD and normal/leptochoroid. DESIGN Observational, retrospective, case-control study. PARTICIPANTS Among a cohort of patients with intermediate age-related macular degeneration (AMD), we selected eyes with RPD and pachychoroid (i.e., choroidal thickness of >50 μm). A control group of RPD eyes but without pachychoroid (i.e., a choroidal thickness of <250 μm) was included. METHODS Number and stages of RPD were evaluated in each ETDRS subfield. Furthermore, choroidal perfusion was investigated using the choroidal vascularity index (CVI), and choriocapillaris perfusion density (PD) on structural OCT and OCT angiography. MAIN OUTCOME MEASURES Description of the multimodal imaging features of pachy-RPD and differences with RPD associated with normal/leptochoroid. RESULTS Among 111 RPD eyes, 37 were included in the pachy-RPD group and 74 in the control group. Patients with pachy-RPD were significantly younger than patients with RPD and normal/leptochoroid (mean age, 75 ± 16 and 82 ± 7 years, respectively; P = 0.002). Total RPD number was comparable between the 2 groups (P = 0.220). However, pachy-RPD eyes showed a significantly higher number of stage 1 RPD in comparison to the controls (P < 0.001), and a lower number of stage 3 (P < 0.001) and stage 4 RPD (P = 0.052). The CVI and choriocapillaris PD were greater in pachy-RPD than in the control group (P < 0.001 and P= 0.010, respectively). CONCLUSIONS Pachy-RPD are characterized by a different distribution of RPD stages (i.e., more early stages and fewer advanced stages) in comparison to RPD with normal/leptochoroid. Furthermore, pachy-RPD eyes showed greater perfusion indices of the choroid. These features suggest that the presence of pachychoroid could be a protective factor in the RPD evolution to the advanced AMD forms. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Riccardo Sacconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giusi Fazzari
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Department of Ophthalmology, Hospital Intercommunal de Creteil, University Paris Est Creteil, Creteil, France
| | - Vittorio Capuano
- Department of Ophthalmology, Hospital Intercommunal de Creteil, University Paris Est Creteil, Creteil, France
| | - Matteo Menean
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Beretta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Khaled El Matri
- Department of Ophthalmology, Hospital Intercommunal de Creteil, University Paris Est Creteil, Creteil, France; Faculté de Médecine de Tunis, Université Tunis - El Manar, Tunis, Tunisia
| | - Francesco Bandello
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eric Souied
- Department of Ophthalmology, Hospital Intercommunal de Creteil, University Paris Est Creteil, Creteil, France
| | - Giuseppe Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
9
|
Hyttinen JMT, Koskela A, Blasiak J, Kaarniranta K. Autophagy in drusen biogenesis secondary to age-related macular degeneration. Acta Ophthalmol 2024; 102:759-772. [PMID: 39087629 DOI: 10.1111/aos.16744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Age-related macular degeneration (AMD) is an emerging cause of blindness in aged people worldwide. One of the key signs of AMD is the degeneration of the retinal pigment epithelium (RPE), which is indispensable for the maintenance of the adjacent photoreceptors. Because of impaired energy metabolism resulting from constant light exposure, hypoxia, and oxidative stress, accumulation of drusen in AMD-affected eyes is observed. Drusen contain damaged cellular proteins, lipoprotein particles, lipids and carbohydrates and they are related to impaired protein clearance, inflammation, and extracellular matrix modification. When autophagy, a major cellular proteostasis pathway, is impaired, the accumulations of intracellular lipofuscin and extracellular drusen are detected. As these aggregates grow over time, they finally cause the disorganisation and destruction of the RPE and photoreceptors leading to visual loss. In this review, the role of autophagy in drusen biogenesis is discussed since impairment in removing cellular waste in RPE cells plays a key role in AMD progression. In the future, means which improve intracellular clearance might be of use in AMD therapy to slow the progression of drusen formation.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ali Koskela
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, Plock, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
10
|
Lishinsky-Fischer N, Misgav K, Chowers I, Tiosano L, Shwartz Y, Levy J. Association of subretinal drusenoid deposits and cardiovascular disease. Sci Rep 2024; 14:25569. [PMID: 39462007 PMCID: PMC11513000 DOI: 10.1038/s41598-024-76342-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The presence of subretinal drusenoid deposits (SDDs) in patients with age-related macular degeneration (AMD) appears to be correlated with cardiovascular disease (CVD) and may serve as a useful tool for predicting certain forms of CVD. Here, we tested this hypothesis by examining whether patients with AMD with SDDs are more likely to have a cardiovascular-related condition and/or undergo a cardiovascular procedure. This is a retrospective cohort study. We included a total of 597 patients with AMD either with SDDs (n = 332) or without SDDs (n = 265). All patients underwent spectral-domain optical coherence tomography (SD-OCT). The SD-OCT scans were annotated by masked, experienced graders. We also extracted data from the patients' electronic medical records (EMRs), including patient demographics, cardiovascular diagnoses, and CVD-related procedures based on ICD-9 codes. AMD patients with SDDs were more likely to be diagnosed with CVD or undergo a cardiovascular procedure compared to AMD patients without SDDs, particularly percutaneous transluminal coronary angioplasty (PTCA; OR 2.73, 95% CI [1.21, 6.13], p = 0.02). Multivariate analysis confirmed the association between the presence of SDDs and a background of PTCA in the presence of other covariates. These data suggest that the presence of SDDs in patients with AMD correlates with certain severe cardiovascular conditions; SDDs and CVD may share common pathogenic pathways.
Collapse
Affiliation(s)
| | - Kinneret Misgav
- Data Research Unit, Hadassah Research Fund, Hadassah Medical Center, Jerusalem, Israel
| | - Itay Chowers
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Liran Tiosano
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Yahel Shwartz
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Jaime Levy
- Ophthalmology Department, Hadassah Medical Center, Jerusalem, Israel.
- Ophthalmology Department, Faculty of Medicine, Hadassah Medical Center, Hebrew University, Kiryat Hadassah, POB 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
11
|
Menean M, Sacconi R, Tombolini B, L'abbate G, Beretta F, Bandello F, Querques G. RETICULAR PSEUDODRUSEN DISAPPEARANCE AFTER DEVELOPMENT OF MACULAR NEOVASCULARIZATION. Retina 2024; 44:1688-1695. [PMID: 39287531 DOI: 10.1097/iae.0000000000004173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
PURPOSE To explore changes in reticular pseudodrusen (RPD) number and location after the development of macular neovascularization (MNV) in eyes with prior intermediate age-related macular degeneration, focusing on different retinal regions differently affected by MNV. METHODS This retrospective longitudinal study included intermediate age-related macular degeneration eyes with RPD that developed MNV. Reticular pseudodrusen were assessed at baseline when MNV was diagnosed (MNV stage) and after anti-vascular endothelial growth factor treatment. Three regions of interest were considered: MNV area, subretinal fluid (SRF) area, and a marginal area of 1,000 µm around SRF (marginal zone). Reticular pseudodrusen counts were compared with age- and sex-matched control eyes with RPD that did not develop MNV. RESULTS Reticular pseudodrusen number exhibited a significant decrease after MNV development in the MNV area (P = 0.048) and in the area with SRF (P = 0.078). A statistically significant decrease was also disclosed in the marginal area around SRF (P = 0.002), associated with larger SRF areas. Control eyes did not show any significant change in the RPD count. CONCLUSION Reticular pseudodrusen reduction after MNV development suggests a complex interplay involving the MNV itself, the presence of SRF, and trophic changes. The results of this study highlight the role of MNV in retinal nutritional balance and provide intriguing results in the RPD life cycle.
Collapse
Affiliation(s)
- Matteo Menean
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Riccardo Sacconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Beatrice Tombolini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gaia L'abbate
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federico Beretta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Bandello
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giuseppe Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; and
- Department of Ophthalmology, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
12
|
Lindenberg S, Mahmoudi A, Oncel D, Corradetti G, Oncel D, Emamverdi M, Almidani L, Farahani A, Wakatsuki Y, He Y, Saju M S, Lee WK, Wykoff CC, Sarraf D, Freund KB, Sadda SR. Acquired Vitelliform Lesions in Intermediate Age-Related Macular Degeneration: A Cross Sectional Study. Ophthalmol Retina 2024; 8:854-862. [PMID: 38631656 DOI: 10.1016/j.oret.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE This study aims to define the characteristics of acquired vitelliform lesions (AVLs) in patients with intermediate age-related macular degeneration (iAMD). DESIGN Retrospective, observational, cross sectional study. SUBJECTS This study included 217 eyes with AVLs associated with iAMD, and an equivalent number of control patients. METHODS OCT scans were evaluated for qualitative and quantitative parameters at both the eye and lesion level. Eye-level parameters included the presence of: hyporeflective core drusen, intraretinal hyperreflective foci (IHRF), subretinal drusenoid deposits, macular pachyvessels, central retinal thickness, and central choroidal thickness. Lesion-level qualitative parameters included the presence of ellipsoid zone (EZ) and external limiting membrane disruption overlying the AVL, IHRF overlying the AVL, AVL overlying drusen, pachyvessels under the AVL, a solid core within AVL, and AVL location. Lesion-level quantitative characteristics included AVL height and width, AVL distance from the fovea, and sub-AVL choroidal thickness. MAIN OUTCOME MEASURES The primary outcomes assessed included the frequency of IHRF, the presence of macular pachyvessels, central choroidal thickness, and the dimensions (both height and width) of AVLs. RESULTS Comparing the AVL and control groups, the frequency of IHRF (AVL: 49.3% vs. control: 26.3%) and macular pachyvessels (37.3% vs. 6.9%) was significantly higher in the AVL case group, and the central choroidal thickness (256.8 ± 88 μm vs. 207.1± 45 μm) was thicker in the AVL group. Acquired vitelliform lesions located over drusen, with overlying IHRF, or situated subfoveally, and AVL lesions with EZ disruption were found to have a greater lesion height and width compared with AVL lesions lacking these characteristics (P value < 0.001 for all). Additionally, a significant negative correlation was observed between the distance from the fovea and AVL height (Spearman rho: -0.19, P = 0.002) and width (Spearman rho: -0.30, P = 0.001). CONCLUSIONS This study represents the largest reported cohort of AVL lesions associated with iAMD. Novel findings include the higher frequency of pachyvessels in addition to the presence of a thicker choroid in these eyes, as well as the greater height and width of AVL closer to the foveal center. These findings may offer insights into pathophysiologic mechanisms underlying the development of AVL. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Sophiana Lindenberg
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California
| | - Alireza Mahmoudi
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Deniz Oncel
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Giulia Corradetti
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Damla Oncel
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Mehdi Emamverdi
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Louay Almidani
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alireza Farahani
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Yu Wakatsuki
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California
| | - Ye He
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Stanley Saju M
- Retina Consultants of Texas, Retina Consultants of America, Houston, Texas
| | - Won Ki Lee
- Nune Eye hospital, Seoul, Republic of South Korea
| | - Charles C Wykoff
- Retina Consultants of Texas, Retina Consultants of America, Houston, Texas
| | - David Sarraf
- Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - K Bailey Freund
- Vitreous Retina Macula Consultants of New York, New York, New York; Department of Ophthalmology, New York University Grossman School of Medicine, New York, New York
| | - Srinivas R Sadda
- Doheny Image Reading and Research Laboratory, Doheny Eye Institute, Pasadena, California; Department of Ophthalmology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California.
| |
Collapse
|
13
|
Saßmannshausen M, Sautbaeva L, von der Emde LA, Vaisband M, Sloan KR, Hasenauer J, Holz FG, Ach T. Retro Mode Imaging for Detection and Quantification of Sub-RPE Drusen and Subretinal Drusenoid Deposits in Age-Related Macular Degeneration. J Clin Med 2024; 13:4131. [PMID: 39064170 PMCID: PMC11278487 DOI: 10.3390/jcm13144131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Drusen and drusenoid deposits are a hallmark of age-related macular degeneration (AMD). Nowadays, a multimodal retinal imaging approach enables the detection of these deposits. However, quantitative data on subretinal drusenoid deposits (SDDs) are still missing. Here, we compare the capability of en-face drusen and SDD area detection in eyes with non-exudative AMD using conventional imaging modalities versus Retro mode imaging. We also quantitatively assess the topographic distribution of drusen and SDDs. Methods: In total, 120 eyes of 90 subjects (mean age ± standard deviation = 74.6 ± 8.6 years) were included. Coherent en-face drusen and SDD areas were measured via near-infrared reflectance, green (G-) and blue (B-) fundus autofluorescence (AF), and Retro mode imaging. Drusen phenotypes were classified by correlating en-face drusen areas using structural high-resolution spectral domain optical coherence tomography. The topographic distribution of drusen was analyzed according to a modified ETDRS (Early Treatment of Diabetic Retinopathy Study) grid. Intraclass correlation coefficient (ICC) analysis was applied to determine the inter-reader agreement in the SDD en-face area assessment. Results: The largest coherent en-face drusen area was found using Retro mode imaging with a mean area of 105.2 ± 45.9 mm2 (deviated left mode (DL)) and 105.4 ± 45.5 mm2 (deviated right mode (DR)). The smallest en-face drusen areas were determined by GAF (50.9 ± 42.6 mm2) and BAF imaging (49.1 ± 42.9 mm2) (p < 0.001). The inter-reader agreement for SDD en-face areas ranged from 0.93 (DR) to 0.70 (BAF). The topographic analysis revealed the highest number of SDDs in the superior peripheral retina, whereas sub-retinal pigment epithelium drusen were mostly found in the perifoveal retina. Retro mode imaging further enabled the detection of the earliest SDD stages. Conclusions: Retro mode imaging allows for a detailed detection of drusen phenotypes. While hundreds/thousands of SDDs can be present in one eye, the impact of SDD number or volume on AMD progression still needs to be evaluated. However, this new imaging modality can add important knowledge on drusen development and the pathophysiology of AMD.
Collapse
Affiliation(s)
- Marlene Saßmannshausen
- Department of Ophthalmology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany (L.A.v.d.E.)
| | - Leyla Sautbaeva
- Department of Ophthalmology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany (L.A.v.d.E.)
| | | | - Marc Vaisband
- Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute—Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Kenneth R. Sloan
- Department of Computer Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jan Hasenauer
- Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
- Helmholtz Center Munich—German Research Center for Environmental Health, Institute of Computational Biology, 85764 Neuherberg, Germany
| | - Frank G. Holz
- Department of Ophthalmology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany (L.A.v.d.E.)
| | - Thomas Ach
- Department of Ophthalmology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany (L.A.v.d.E.)
| |
Collapse
|
14
|
Cheng Y, Hiya F, Li J, Shen M, Liu J, Herrera G, Berni A, Morin R, Joseph J, Zhang Q, Gregori G, Rosenfeld PJ, Wang RK. Calcified Drusen Prevent the Detection of Underlying Choriocapillaris Using Swept-Source Optical Coherence Tomography Angiography. Invest Ophthalmol Vis Sci 2024; 65:26. [PMID: 38884553 PMCID: PMC11185265 DOI: 10.1167/iovs.65.6.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/28/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose In age-related macular degeneration (AMD), choriocapillaris flow deficits (CCFDs) under soft drusen can be measured using established compensation strategies. This study investigated whether CCFDs can be quantified under calcified drusen (CaD). Methods CCFDs were measured in normal eyes (n = 30) and AMD eyes with soft drusen (n = 30) or CaD (n = 30). CCFD density masks were generated to highlight regions with higher CCFDs. Masks were also generated for soft drusen and CaD based on both structural en face OCT images and corresponding B-scans. Dice similarity coefficients were calculated between the CCFD density masks and both the soft drusen and CaD masks. A phantom experiment was conducted to simulate the impact of light scattering that arises from CaD. Results Area measurements of CCFDs were highly correlated with those of CaD but not soft drusen, suggesting an association between CaD and underlying CCFDs. However, unlike soft drusen, the detected optical coherence tomography (OCT) signals underlying CaD did not arise from the defined CC layer but were artifacts caused by the multiple scattering property of CaD. Phantom experiments showed that the presence of highly scattering material similar to the contents of CaD caused an artifactual scattering tail that falsely generated a signal in the CC structural layer but the underlying flow could not be detected. Similarly, CaD also caused an artifactual scattering tail and prevented the penetration of light into the choroid, resulting in en face hypotransmission defects and an inability to detect blood flow within the choriocapillaris. Upon resolution of the CaD, the CC perfusion became detectable. Conclusions The high scattering property of CaD leads to a scattering tail under these drusen that gives the illusion of a quantifiable optical coherence tomography angiography signal, but this signal does not contain the angiographic information required to assess CCFDs. For this reason, CCFDs cannot be reliably measured under CaD, and CaD must be identified and excluded from macular CCFD measurements.
Collapse
Affiliation(s)
- Yuxuan Cheng
- Department of Bioengineering, University of Washington, Seattle, Washington, United States
| | - Farhan Hiya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jianqing Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Mengxi Shen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jeremy Liu
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Gissel Herrera
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Alessandro Berni
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosalyn Morin
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Joan Joseph
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Qinqin Zhang
- Research and Development, Carl Zeiss Meditec, Inc., Dublin, California, United States
| | - Giovanni Gregori
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Philip J. Rosenfeld
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Ruikang K. Wang
- Department of Bioengineering, University of Washington, Seattle, Washington, United States
- Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| |
Collapse
|
15
|
Voichanski S, Bousquet E, Abraham N, Santina A, Mafi M, Fossataro C, Sadda S, Sarraf D. En Face Optical Coherence Tomography Illustrates the Trizonal Distribution of Drusen and Subretinal Drusenoid Deposits in the Macula. Am J Ophthalmol 2024; 261:187-198. [PMID: 38218515 DOI: 10.1016/j.ajo.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/15/2024]
Abstract
PURPOSE To analyze the topographic distribution of macular drusen and subretinal drusenoid deposits (SDDs) using single-capture en face spectral domain optical coherence tomography (SD-OCT) imaging. DESIGN Retrospective case series. METHODS Analysis of 33 eyes of 20 patients with evidence of SDDs. Structural en face OCT images were reconstructed using a 40-µm-thick slab positioned from 48 to 88 µm above the Bruch membrane. The Early Treatment of Diabetic Retinopathy Study (ETDRS) grid and a rod/cone density map were overlaid on the en face OCT images, and the distribution of different subtypes of SDDs and macular drusen were assessed. RESULTS A total of 31 eyes (94%) showed a trizonal distribution pattern of drusen and SDDs. Whereas small to large drusen tended to aggregate in the central circle, dot SDDs predominated in the inner ring and the inner portion of the outer ring of the ETDRS grid and ribbon SDDs localized to the outer ring and outside the ETDRS grid. Of note, drusen colocalized to the region of greatest cone density, whereas ribbon SDDs colocalized to the area of greatest rod density. The dot SDDs mapped to the intermediate region with mixed rod and cone representation. CONCLUSION Dot and ribbon subtypes of SDDs and macular drusen show a characteristic trizonal distribution. The locations of these lesions colocalize according to the different densities of the cones and rods in the retina and may reflect varying pathophysiological activities of these photoreceptor subtypes.
Collapse
Affiliation(s)
- Shilo Voichanski
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA; Vitreoretinal Division, Ophthalmology Department, Shaare Zedek Medical Center (S.V.), Jerusalem, Israel
| | - Elodie Bousquet
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA; University of Paris Cité; Department of Ophthalmology, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (E.B.), Paris, France
| | - Neda Abraham
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA
| | - Ahmad Santina
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA
| | - Mostafa Mafi
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA
| | - Claudia Fossataro
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA; Ophthalmology Unit, Catholic University of the Sacred Heart (C.F.), Rome, Italy; Ophthalmology Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS (C.F.), Rome, Italy
| | - SriniVas Sadda
- Doheny Eye Institute, Department of Ophthalmology, University of California Los Angeles (S.S.), Los Angeles, California, USA
| | - David Sarraf
- Retinal Disorders and Ophthalmic Genetics Division, Stein Eye Institute, University of California Los Angeles, David Geffen School of Medicine at UCLA (S.V., E.B., N.A., A.S., M.M., C.F., D.S.), Los Angeles, California, USA; Greater Los Angeles VA Healthcare Center (D.S.), Los Angeles, California, USA.
| |
Collapse
|
16
|
Emamverdi M, Vatanatham C, Fasih-Ahmad S, Wang Z, Mishra Z, Jain A, Ganegoda A, Clark ME, Habibi A, Ashrafkhorasani M, Owsley C, Curcio CA, Hu ZJ, Sadda SR. Probing Deposit-Driven Age-Related Macular Degeneration Via Thicknesses of Outer Retinal Bands and Choroid: ALSTAR2 Baseline. Invest Ophthalmol Vis Sci 2024; 65:17. [PMID: 38717424 PMCID: PMC11090139 DOI: 10.1167/iovs.65.5.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose We aimed to identify structural differences in normal eyes, early age-related macular degeneration (AMD), and intermediate AMD eyes using optical coherence tomography (OCT) in a well-characterized, large cross-sectional cohort. Methods Subjects ≥ 60 years with healthy normal eyes, as well as early or intermediate AMD were enrolled in the Alabama Study on Age-related Macular Degeneration 2 (ALSTAR2; NCT04112667). Using Spectralis HRA + OCT2, we obtained macular volumes for each participant. An auto-segmentation software was used to segment six layers and sublayers: photoreceptor inner and outer segments, subretinal drusenoid deposits (SDDs), retinal pigment epithelium + basal lamina (RPE + BL), drusen, and choroid. After manually refining the segmentations of all B-scans, mean thicknesses in whole, central, inner and outer rings of the ETDRS grid were calculated and compared among groups. Results This study involved 502 patients, 252 were healthy, 147 had early AMD, and 103 had intermediate AMD eyes (per Age-Related Eye Disease Study [AREDS] 9-step). Intermediate AMD eyes exhibited thicker SDD and drusen, thinner photoreceptor inner segments, and RPE compared to healthy and early AMD eyes. They also had thicker photoreceptor outer segments than early AMD eyes. Early AMD eyes had thinner photoreceptor outer segments than normal eyes but a thicker choroid than intermediate AMD eyes. Using the Beckman scale, 42% of the eyes initially classified as early AMD shifted to intermediate AMD, making thickness differences for photoreceptor outer segments and choroid insignificant. Conclusions With AMD stages, the most consistent structural differences involve appearance of drusen and SDD, followed by RPE + BL thickness, and then thickness of photoreceptor inner and outer segments. Structural changes in the transition from aging to intermediate AMD include alterations in the outer retinal bands, including the appearance of deposits on either side of the RPE.
Collapse
Affiliation(s)
- Mehdi Emamverdi
- Doheny Eye Institute, Pasadena, California, United States
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| | | | | | - Ziyuan Wang
- Doheny Eye Institute, Pasadena, California, United States
| | - Zubin Mishra
- Doheny Eye Institute, Pasadena, California, United States
| | - Anjal Jain
- Doheny Eye Institute, Pasadena, California, United States
| | | | - Mark E. Clark
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Abbas Habibi
- Doheny Eye Institute, Pasadena, California, United States
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| | - Maryam Ashrafkhorasani
- Doheny Eye Institute, Pasadena, California, United States
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| | - Cynthia Owsley
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zhihong J. Hu
- Doheny Eye Institute, Pasadena, California, United States
| | - SriniVas R. Sadda
- Doheny Eye Institute, Pasadena, California, United States
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| |
Collapse
|
17
|
Dubey SK, Dubey R, Prajapati SC, Jung K, Mohan K, Liu X, Roney J, Tian W, Abney J, Giarmarco MM, Hernandez AG, Liu J, Kleinman ME. Histone deficiency and hypoacetylation in the aging retinal pigment epithelium. Aging Cell 2024; 23:e14108. [PMID: 38408164 PMCID: PMC11113634 DOI: 10.1111/acel.14108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Histones serve as a major carrier of epigenetic information in the form of post-translational modifications which are vital for controlling gene expression, maintaining cell identity, and ensuring proper cellular function. Loss of histones in the aging genome can drastically impact the epigenetic landscape of the cell leading to altered chromatin structure and changes in gene expression profiles. In this study, we investigated the impact of age-related changes on histone levels and histone acetylation in the retinal pigment epithelium (RPE) and retina of mice. We observed a global reduction of histones H1, H2A, H2B, H3, and H4 in aged RPE/choroid but not in the neural retina. Transcriptomic analyses revealed significant downregulation of histones in aged RPE/choroid including crucial elements of the histone locus body (HLB) complex involved in histone pre-mRNA processing. Knockdown of HINFP, a key HLB component, in human RPE cells induced histone loss, senescence, and the upregulation of senescence-associated secretory phenotype (SASP) markers. Replicative senescence and chronological aging in human RPE cells similarly resulted in progressive histone loss and acquisition of the SASP. Immunostaining of human retina sections revealed histone loss in RPE with age. Acetyl-histone profiling in aged mouse RPE/choroid revealed a specific molecular signature with loss of global acetyl-histone levels, including H3K14ac, H3K56ac, and H4K16ac marks. These findings strongly demonstrate histone loss as a unique feature of RPE aging and provide critical insights into the potential mechanisms linking histone dynamics, cellular senescence, and aging.
Collapse
Affiliation(s)
- Sushil K. Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Rashmi Dubey
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Subhash C. Prajapati
- Department of Biochemistry and Molecular GeneticsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Kyungsik Jung
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Kabhilan Mohan
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Xinan Liu
- Department of Computer ScienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Jacob Roney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Wenjian Tian
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| | - Jennifer Abney
- Department of Ophthalmology and Visual SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Alvaro G. Hernandez
- Roy J. Carver Biotechnology CenterUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jinze Liu
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mark E. Kleinman
- Department of SurgeryEast Tennessee State UniversityJohnson CityTennesseeUSA
| |
Collapse
|
18
|
Iliescu DA, Ghita AC, Ilie LA, Voiculescu SE, Geamanu A, Ghita AM. Non-Neovascular Age-Related Macular Degeneration Assessment: Focus on Optical Coherence Tomography Biomarkers. Diagnostics (Basel) 2024; 14:764. [PMID: 38611677 PMCID: PMC11011935 DOI: 10.3390/diagnostics14070764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
The imagistic evaluation of non-neovascular age-related macular degeneration (AMD) is crucial for diagnosis, monitoring progression, and guiding management of the disease. Dry AMD, characterized primarily by the presence of drusen and retinal pigment epithelium atrophy, requires detailed visualization of the retinal structure to assess its severity and progression. Several imaging modalities are pivotal in the evaluation of non-neovascular AMD, including optical coherence tomography, fundus autofluorescence, or color fundus photography. In the context of emerging therapies for geographic atrophy, like pegcetacoplan, it is critical to establish the baseline status of the disease, monitor the development and expansion of geographic atrophy, and to evaluate the retina's response to potential treatments in clinical trials. The present review, while initially providing a comprehensive description of the pathophysiology involved in AMD, aims to offer an overview of the imaging modalities employed in the evaluation of non-neovascular AMD. Special emphasis is placed on the assessment of progression biomarkers as discerned through optical coherence tomography. As the landscape of AMD treatment continues to evolve, advanced imaging techniques will remain at the forefront, enabling clinicians to offer the most effective and tailored treatments to their patients.
Collapse
Affiliation(s)
- Daniela Adriana Iliescu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Bld., 050474 Bucharest, Romania; (S.E.V.); (A.M.G.)
- Ocularcare Ophthalmology Clinic, 128 Ion Mihalache Bld., 012244 Bucharest, Romania; (A.C.G.); (L.A.I.)
| | - Ana Cristina Ghita
- Ocularcare Ophthalmology Clinic, 128 Ion Mihalache Bld., 012244 Bucharest, Romania; (A.C.G.); (L.A.I.)
| | - Larisa Adriana Ilie
- Ocularcare Ophthalmology Clinic, 128 Ion Mihalache Bld., 012244 Bucharest, Romania; (A.C.G.); (L.A.I.)
| | - Suzana Elena Voiculescu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Bld., 050474 Bucharest, Romania; (S.E.V.); (A.M.G.)
| | - Aida Geamanu
- Ophthalmology Department, Bucharest University Emergency Hospital, 169 Independence Street, 050098 Bucharest, Romania;
| | - Aurelian Mihai Ghita
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Bld., 050474 Bucharest, Romania; (S.E.V.); (A.M.G.)
- Ocularcare Ophthalmology Clinic, 128 Ion Mihalache Bld., 012244 Bucharest, Romania; (A.C.G.); (L.A.I.)
- Ophthalmology Department, Bucharest University Emergency Hospital, 169 Independence Street, 050098 Bucharest, Romania;
| |
Collapse
|
19
|
Saßmannshausen M, Döngelci S, Vaisband M, von der Emde L, Sloan KR, Hasenauer J, Holz FG, Schmitz-Valckenberg S, Ach T. Spatially Resolved Association of Structural Biomarkers on Retinal Function in Non-Exudative Age-Related Macular Degeneration Over 4 Years. Invest Ophthalmol Vis Sci 2024; 65:45. [PMID: 38687492 PMCID: PMC11067547 DOI: 10.1167/iovs.65.4.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Purpose To longitudinally assess the impact of high-risk structural biomarkers for natural disease progression in non-exudative age-related macular degeneration (AMD) on spatially resolved mesopic and scotopic fundus-controlled perimetry testing. Methods Multimodal retinal imaging data and fundus-controlled perimetry stimuli points were semiautomatically registered according to landmark correspondences at each annual visit over a period of up to 4 years. The presence of sub-RPE drusen, subretinal drusenoid deposits, pigment epithelium detachments (PEDs), hyper-reflective foci (HRF), vitelliform lesions, refractile deposits, and incomplete RPE and outer retinal atrophy (iRORA) and complete RPE and outer retinal atrophy (cRORA) were graded at each stimulus position and visit. Localized retinal layer thicknesses were extracted. Mixed-effect models were used for structure-function correlation. Results Fifty-four eyes of 49 patients with non-exudative AMD (mean age, 70.7 ± 9.1 years) and 27 eyes of 27 healthy controls (mean age, 63.4 ± 8.9 years) were included. During study course, presence of PED had the highest functional impact with a mean estimated loss of -1.30 dB (P < 0.001) for mesopic and -1.23 dB (P < 0.001) for scotopic testing, followed by HRF with -0.89 dB (mesopic, P = 0.001) and -0.87 dB (scotopic, P = 0.005). Subretinal drusenoid deposits were associated with a stronger visual impairment (mesopic, -0.38 dB; P = 0.128; scotopic, -0.37 dB; P = 0.172) compared with sub-RPE drusen (-0.22 dB, P = 0.0004; -0.18 dB, P = 0.006). With development of c-RORA, scotopic retinal sensitivity further significantly decreased (-2.15 dB; P = 0.02). Thickening of the RPE-drusen-complex and thinning of the outer nuclear layer negatively impacted spatially resolved retinal sensitivity. Conclusions The presence of PED and HRF had the greatest prognostic impact on progressive point-wise sensitivity losses. Higher predominant rod than cone-mediated localized retinal sensitivity losses with early signs of retinal atrophy development indicate photoreceptor preservation as a potential therapeutic target for future interventional AMD trials.
Collapse
Affiliation(s)
| | - Senem Döngelci
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| | - Marc Vaisband
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Leon von der Emde
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| | - Kenneth R. Sloan
- Department of Computer Science, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jan Hasenauer
- Department of Internal Medicine III With Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg, Austria
- Helmholtz Center Munich–German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Frank G. Holz
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Steffen Schmitz-Valckenberg
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Thomas Ach
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
20
|
Fu DJ, Glinton S, Lipkova V, Faes L, Liefers B, Zhang G, Pontikos N, McKeown A, Scheibler L, Patel PJ, Keane PA, Balaskas K. Deep-learning automated quantification of longitudinal OCT scans demonstrates reduced RPE loss rate, preservation of intact macular area and predictive value of isolated photoreceptor degeneration in geographic atrophy patients receiving C3 inhibition treatment. Br J Ophthalmol 2024; 108:536-545. [PMID: 37094835 PMCID: PMC10958254 DOI: 10.1136/bjo-2022-322672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/15/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE To evaluate the role of automated optical coherence tomography (OCT) segmentation, using a validated deep-learning model, for assessing the effect of C3 inhibition on the area of geographic atrophy (GA); the constituent features of GA on OCT (photoreceptor degeneration (PRD), retinal pigment epithelium (RPE) loss and hypertransmission); and the area of unaffected healthy macula.To identify OCT predictive biomarkers for GA growth. METHODS Post hoc analysis of the FILLY trial using a deep-learning model for spectral domain OCT (SD-OCT) autosegmentation. 246 patients were randomised 1:1:1 into pegcetacoplan monthly (PM), pegcetacoplan every other month (PEOM) and sham treatment (pooled) for 12 months of treatment and 6 months of therapy-free monitoring. Only participants with Heidelberg SD-OCT were included (n=197, single eye per participant).The primary efficacy endpoint was the square root transformed change in area of GA as complete RPE and outer retinal atrophy (cRORA) in each treatment arm at 12 months, with secondary endpoints including RPE loss, hypertransmission, PRD and intact macular area. RESULTS Eyes treated PM showed significantly slower mean change of cRORA progression at 12 and 18 months (0.151 and 0.277 mm, p=0.0039; 0.251 and 0.396 mm, p=0.039, respectively) and RPE loss (0.147 and 0.287 mm, p=0.0008; 0.242 and 0.410 mm, p=0.00809). PEOM showed significantly slower mean change of RPE loss compared with sham at 12 months (p=0.0313). Intact macular areas were preserved in PM compared with sham at 12 and 18 months (p=0.0095 and p=0.044). PRD in isolation and intact macula areas was predictive of reduced cRORA growth at 12 months (coefficient 0.0195, p=0.01 and 0.00752, p=0.02, respectively) CONCLUSION: The OCT evidence suggests that pegcetacoplan slows progression of cRORA overall and RPE loss specifically while protecting the remaining photoreceptors and slowing the progression of healthy retina to iRORA.
Collapse
Affiliation(s)
- Dun Jack Fu
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Sophie Glinton
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Veronika Lipkova
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Livia Faes
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Bart Liefers
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
- Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gongyu Zhang
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Nikolas Pontikos
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Alex McKeown
- Apellis Pharmaceuticals Inc, Waltham, Massachusetts, USA
| | | | - Praveen J Patel
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Pearse A Keane
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
| | - Konstantinos Balaskas
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL, Institute of Ophthalmology, London, UK
- University College London, London, UK
| |
Collapse
|
21
|
Bellis RM, Fei Y, Le B, Ledesma-Gil G, Otero-Marquez O, Tong Y, Tai K, Rosen RB, Lema GMC, Smith RT. Correlation between ellipsoid zone thickness and the presence of subretinal drusenoid deposits in age-related macular degeneration. BMJ Open Ophthalmol 2024; 9:e001622. [PMID: 38460964 DOI: 10.1136/bmjophth-2023-001622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/14/2024] [Indexed: 03/11/2024] Open
Abstract
PURPOSE Subretinal drusenoid deposits (SDDs) in age-related macular degeneration (AMD) are associated with systemic vascular diseases that compromise ocular perfusion. We demonstrate that SDDs are associated with decreased ellipsoid zone (EZ) thickness, further evidence of hypoxic damage. METHODS Post hoc analysis of a cross-sectional study. 165 AMD subjects (aged 51-100; 61% women). Spectral-domain optical coherence tomography was obtained in both eyes. Masked readers assigned subjects to three groups: drusen only, SDD+drusen (SDD+D) and SDD only. EZ thickness was measured subfoveally and 2000 µm nasally, temporally, superiorly and inferiorly from the fovea. Univariate testing was performed using two-tailed t-tests with Bonferroni correction. RESULTS The mean EZ thickness differences between the SDD+D and drusen-only groups were (in μm) 1.10, 0.67, 1.21, 1.10 and 0.50 at the foveal, nasal, temporal, superior and inferior locations, respectively (p=0.08 inferiorly, otherwise p≤0.01); between the SDD-only and drusen-only groups, the differences were 3.48, 2.48, 2.42, 2.08 and 1.42 (p≤0.0002). Differences in EZ thicknesses across all subjects and between groups were not significantly different based on gender, race or age. CONCLUSION Subjects with SDDs (±drusen) had thinner EZs than those with drusen only, and the inferior EZ was least affected. EZs were thinnest in SDD-only subjects. This thinning gradation is consistent with progressive destruction of highly oxygen-sensitive mitochondria in the EZ from hypoxia. These findings support the reduced ophthalmic perfusion hypothesis for the formation of SDDs secondary to high-risk systemic vasculopathy.
Collapse
Affiliation(s)
| | - Yang Fei
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Brandon Le
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Gerardo Ledesma-Gil
- Retina, Instituto de Oftalmologia Fundacion Conde de Valenciana IAP, Mexico City, Mexico
| | - Oscar Otero-Marquez
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Yuehong Tong
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Katy Tai
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Richard B Rosen
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Gareth M C Lema
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| | - Roland Theodore Smith
- Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York, USA
| |
Collapse
|
22
|
Zhao Q, Lai K. Role of immune inflammation regulated by macrophage in the pathogenesis of age-related macular degeneration. Exp Eye Res 2024; 239:109770. [PMID: 38145794 DOI: 10.1016/j.exer.2023.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Age-related macular degeneration (AMD) can lead to irreversible impairment of visual function, and the number of patients with AMD has been increasing globally. The immunoinflammatory theory is an important pathogenic mechanism of AMD, with macrophages serving as the primary inflammatory infiltrating cells in AMD lesions. Its powerful immunoinflammatory regulatory function has attracted considerable attention. Herein, we provide an overview of the involvement of macrophage-regulated immunoinflammation in different stages of AMD. Additionally, we summarize novel therapeutic approaches for AMD, focusing on targeting macrophages, such as macrophage/microglia modulators, reduction of macrophage aggregation in the subretinal space, modulation of macrophage effector function, macrophage phenotypic alterations, and novel biomimetic nanocomposites development based on macrophage-associated functional properties. We aimed to provide a basis and reference for the further exploration of AMD pathogenesis, developmental influences, and new therapeutic approaches.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China.
| |
Collapse
|
23
|
Levine DA, Mathew NE, Jung EH, Yan J, Newman NJ, Thulasi P, Yeh S, Ziegler TR, Wells J, Jain N. Characteristics of Vitamin A Deficiency Retinopathy at a Tertiary Referral Center in the United States. Ophthalmol Retina 2024; 8:126-136. [PMID: 37673395 DOI: 10.1016/j.oret.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
PURPOSE To explore the risk factors and fundus imaging features of vitamin A deficiency retinopathy (VADR) in an academic tertiary referral center in Atlanta, GA, United States, and to propose guidance regarding diagnostic workup and management of affected patients. DESIGN Single-center retrospective case series. SUBJECTS Nine patients seen between 2015 and 2021 at the Emory Eye Center diagnosed with VADR. METHODS Retrospective chart review. MAIN OUTCOME MEASURES Baseline serum retinol level, Snellen visual acuity, multimodal fundus imaging findings, and electroretinography findings. RESULTS Nine patients, 4 (44.4%) female, with a median (range) age of 68 (50-75) years were identified. The most common underlying etiologies for vitamin A deficiency included history of gastrointestinal surgery (55.6%), liver disease (44.4%), and nutritional depletion due to low-quality diet (44.4%). Only 1 (11.1%) patient had a history of bariatric surgery. Four (44.4%) patients were on some form of vitamin A supplementation before the diagnosis of VADR. Median (range) serum retinol level was 0.06 (< 0.06-0.19) mg/L. All patients had macular subretinal hyperreflective deposits resembling subretinal drusenoid deposits, although in some cases, these were scant and sparsely distributed. Six eyes of 3 patients with longstanding deficiency had defects in the external limiting membrane (ELM). Three of these eyes additionally had macular areas of complete retinal pigment epithelium and outer retinal atrophy (cRORA). Full-field electroretinography demonstrated severe rod dysfunction and mild to moderate cone system dysfunction. Many findings of VADR were reversible with vitamin A repletion. However, all eyes with ELM defects or cRORA had persistence or continued growth of these lesions. CONCLUSION Vitamin A deficiency retinopathy is uncommon in the developed world. However, given that early intervention can lead to dramatic visual improvement and avoid potentially permanent retinal damage, retina specialists should be familiar with its clinical presentation. The presence of nyctalopia and subretinal hyperreflective deposits in a patient with a history of gastrointestinal surgery, liver disease, and/or poor diet can be suggestive of this diagnosis, even in the presence of ongoing vitamin A supplementation. Vitamin A supplementation can vary in route and dosage and can be tailored to the individual with serial testing of serum retinol. FINANCIAL DISCLOSURE(S) The authors have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- David A Levine
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Namita E Mathew
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Emily H Jung
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jiong Yan
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Nancy J Newman
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia; Department of Neurology, Emory University School of Medicine, Atlanta, Georgia; Department of Neurological Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Praneetha Thulasi
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Steven Yeh
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Thomas R Ziegler
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jill Wells
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Nieraj Jain
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
24
|
Greferath U, Fletcher E, Savige J, Mack HG. Drusen and Other Retinal Findings in People With IgA Glomerulonephritis. Am J Ophthalmol 2024; 257:247-253. [PMID: 37757996 DOI: 10.1016/j.ajo.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE Retinal drusen have been described in people with IgA nephropathy. We examined the frequency of drusen in IgA nephropathy and compared their location and composition with those for drusen in age-related macular degeneration. DESIGN Immunohistological case series of eyes of patients with IgA nephropathy, and a comparison eye with age-related macular degeneration. METHODS Donor eyes from 4 individuals (3 male, 1 female, aged 40-80 years) with biopsy-proven IgA nephropathy and kidney failure were examined for the presence of drusen, and location and composition using antibodies for vitronectin, IgA, IgM, IgG, C3, and C1q. Results were compared with those for drusen in macular degeneration without IgA nephropathy. RESULTS All 4 donors had sparse, subretinal pigment epithelium drusen of 55-65 mm diameter that stained for vitronectin but not for IgA or complement. All donors had retinal capillaries and choriocapillaris staining for IgA. The youngest donor (female, 40) had rare deposits in the outer nuclear layer that stained for IgA, but not for vitronectin. The oldest donor (male, 82) had large cystlike spaces in the inner nuclear and plexiform layers, and smaller cysts in the outer nuclear layer, with no staining for IgA or complement. CONCLUSIONS Retinal drusen are uncommon in IgA nephropathy, even with kidney failure. Drusen in IgA nephropathy resemble drusen found in age-related macular degeneration. IgA-staining deposits in the outer nuclear layer were likely due to systemic deposition of IgA and complement activation. The nature of cystic spaces is unknown. Further analysis of the retinas of people with glomerulonephritis is recommended.
Collapse
Affiliation(s)
- Ursula Greferath
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Erica Fletcher
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Judy Savige
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.)
| | - Heather G Mack
- University of Melbourne, Parkville, Victoria, Australia (U.G., E.F., J.S., H.G.M.).
| |
Collapse
|
25
|
Anderson DM, Kotnala A, Migas LG, Patterson NH, Tideman L, Cao D, Adhikari B, Messinger JD, Ach T, Tortorella S, Van de Plas R, Curcio CA, Schey KL. Lysolipids are prominent in subretinal drusenoid deposits, a high-risk phenotype in age-related macular degeneration. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1258734. [PMID: 38186747 PMCID: PMC10769005 DOI: 10.3389/fopht.2023.1258734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Introduction Age related macular degeneration (AMD) causes legal blindness worldwide, with few therapeutic targets in early disease and no treatments for 80% of cases. Extracellular deposits, including drusen and subretinal drusenoid deposits (SDD; also called reticular pseudodrusen), disrupt cone and rod photoreceptor functions and strongly confer risk for advanced disease. Due to the differential cholesterol composition of drusen and SDD, lipid transfer and cycling between photoreceptors and support cells are candidate dysregulated pathways leading to deposit formation. The current study explores this hypothesis through a comprehensive lipid compositional analysis of SDD. Methods Histology and transmission electron microscopy were used to characterize the morphology of SDD. Highly sensitive tools of imaging mass spectrometry (IMS) and nano liquid chromatography tandem mass spectrometry (nLC-MS/MS) in positive and negative ion modes were used to spatially map and identify SDD lipids, respectively. An interpretable supervised machine learning approach was utilized to compare the lipid composition of SDD to regions of uninvolved retina across 1873 IMS features and to automatically discern candidate markers for SDD. Immunohistochemistry (IHC) was used to localize secretory phospholipase A2 group 5 (PLA2G5). Results Among the 1873 detected features in IMS data, three lipid classes, including lysophosphatidylcholine (LysoPC), lysophosphatidylethanolamine (LysoPE) and lysophosphatidic acid (LysoPA) were observed nearly exclusively in SDD while presumed precursors, including phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidic acid (PA) lipids were detected in SDD and adjacent photoreceptor outer segments. Molecular signals specific to SDD were found in central retina and elsewhere. IHC results indicated abundant PLA2G5 in photoreceptors and retinal pigment epithelium (RPE). Discussion The abundance of lysolipids in SDD implicates lipid remodeling or degradation in deposit formation, consistent with ultrastructural evidence of electron dense lipid-containing structures distinct from photoreceptor outer segment disks and immunolocalization of secretory PLA2G5 in photoreceptors and RPE. Further studies are required to understand the role of lipid signals observed in and around SDD.
Collapse
Affiliation(s)
| | - Ankita Kotnala
- Department of Biochemistry, Vanderbilt University, Nashville TN
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham AL
| | - Lukasz G. Migas
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Delft, Netherlands
| | | | - Léonore Tideman
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Delft, Netherlands
| | - Dongfeng Cao
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham AL
| | - Bibek Adhikari
- Vision Science Graduate Program, University of Alabama at Birmingham, Birmingham AL
| | - Jeffrey D. Messinger
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham AL
| | - Thomas Ach
- Department of Ophthalmology, University Hospital Bonn, Bonn, Germany
| | - Sara Tortorella
- Molecular Horizon Srl, Via Montelino 30, 06084 Bettona, Perugia, Italy
| | - Raf Van de Plas
- Department of Biochemistry, Vanderbilt University, Nashville TN
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Delft, Netherlands
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham AL
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University, Nashville TN
| |
Collapse
|
26
|
Kang D, Lee YJ, Nam KT, Choi M, Yun C. Hyperreflective foci distribution in eyes with dry age-related macular degeneration with subretinal drusenoid deposits. Graefes Arch Clin Exp Ophthalmol 2023; 261:2821-2828. [PMID: 37231279 DOI: 10.1007/s00417-023-06127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023] Open
Abstract
PURPOSE To investigate the distribution of hyperreflective foci (HRF) in eyes with dry age-related macular degeneration (AMD). METHODS We retrospectively reviewed optical coherence tomography (OCT) images of 58 dry AMD eyes presenting HRF. The distribution of HRF according to the early treatment diabetic retinopathy study area was analyzed according to the presence of subretinal drusenoid deposits (SDDs). RESULTS We classified 32 eyes and 26 eyes into the dry AMD with SDD group (SDD group) and dry AMD without SDD group (non-SDD group), respectively. The non-SDD group had higher prevalence and density of HRF at the fovea (65.4% and 1.71 ± 1.48) than the SDD group (37.5% and 0.48 ± 0.63, P = 0.035 and P < 0.001, respectively). However, the prevalence and density of HRF in the outer circle area of the SDD group (81.3% and 0.11 ± 0.09) were greater than those of the non-SDD group (53.8% and 0.05 ± 0.06, p = 0.025 and p = 0.004, respectively). The SDD group showed higher prevalence and mean densities of HRF in the superior and temporal area than in the non-SDD group (all, p < 0.05). CONCLUSION HRF distributions in dry AMD varied according to the presence of SDDs. This might support that the degenerative features may be different between dry AMD eyes with and without SDDs.
Collapse
Affiliation(s)
| | - Young Joo Lee
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Ki Tae Nam
- Department of Ophthalmology, Jeju National University College of Medicine, Jeju, Korea
| | - Mihyun Choi
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Cheolmin Yun
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
27
|
Kar D, Corradetti G, Swain TA, Clark ME, McGwin G, Owsley C, Sadda SR, Curcio CA. Choriocapillaris Impairment Is Associated With Delayed Rod-Mediated Dark Adaptation in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2023; 64:41. [PMID: 37768273 PMCID: PMC10540875 DOI: 10.1167/iovs.64.12.41] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Purpose Progress toward treatment and prevention of age-related macular degeneration (AMD) requires imaging end points that relate to vision. We investigated choriocapillaris flow signal deficits (FD%) and visual function in eyes of individuals aged ≥60 years, with and without AMD. Methods One eye of each participant in the baseline visit of the Alabama Study on Early Age-Related Macular Degeneration 2 (ALSTAR2; NCT04112667) was studied. AMD presence and severity was determined using the Age-Related Eye Disease Study (AREDS) grading system. FD% was quantified using macular spectral domain optical coherence tomography angiography (OCTA) scans. Vision tests included rod-mediated dark adaptation (RMDA), best-corrected visual acuity, and contrast sensitivity (photopic and mesopic), and microperimetric light sensitivity (scotopic, mesopic, and photopic). Presence of subretinal drusenoid deposits (SDD) was determined using multimodal imaging. Results In 410 study eyes of 410 participants (mean [SD] age = 71.7 years [5.9]), FD% was higher in early AMD (mean [SD] = 54.0% [5.5], N = 122) and intermediate AMD (59.8% [7.4], N = 92), compared to normal (52.1% [5.3], N = 196) eyes. Among visual functions evaluated, RMDA showed the strongest association with FD% (r = 0.35, P < 0.0001), followed by contrast sensitivity (r = -0.22, P < 0.0001). Eyes with SDD had worse FD% (58.3% [7.4], N = 87), compared to eyes without SDD (53.4% [6.0], N = 323, P = < 0.0001). Conclusions Choriocapillaris FD% were associated with AMD severity and with impaired vision, especially RMDA. Reduced metabolic transport and exchange across the choriocapillaris-Bruch's membrane retinal pigment epithelium (RPE) complex, a causal factor for high-risk soft drusen formation, also may impair photoreceptor sustenance from the circulation. This includes retinoid resupply, essential to dynamic rod function.
Collapse
Affiliation(s)
- Deepayan Kar
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Giulia Corradetti
- Doheny Eye Institute, Los Angeles, California, United States
- Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, California, United States
| | - Thomas A. Swain
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Mark E. Clark
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Gerald McGwin
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Cynthia Owsley
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - SriniVas R. Sadda
- Doheny Eye Institute, Los Angeles, California, United States
- Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, California, United States
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
28
|
Breazzano MP, Oh JK, Batson SA, Kucherich JA, Karani R, Rohrmann CM, Sparrow JR, Fragiotta S, Tsang SH. Vitamin A deficiency and the retinal "double carrot" sign with optical coherence tomography. Eye (Lond) 2023; 37:1489-1495. [PMID: 35840717 PMCID: PMC10169789 DOI: 10.1038/s41433-022-02137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 04/30/2022] [Accepted: 06/10/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Spectral-domain optical coherence tomography (SD-OCT) and full-field electroretinography (ERG) allow retinal assessment with vitamin A deficiency (VAD). Using SD-OCT, this study aimed to characterize and follow a novel retinal abnormality in patients with VAD and intramuscular supplementation. METHODS Patients with VAD were retrospectively reviewed, including SD-OCT and electroretinography. RESULTS Three patients had VAD following bariatric or colon surgery and varying supplementation. All had nyctalopia, extinguished scotopic rod-specific function with ERG, and decreased serum vitamin A. None demonstrated surface abnormalities. All received intramuscular vitamin A with subjective resolution of symptoms. On SD-OCT, four of six eyes exhibited homogenous foveal hyperreflectivity anterior to retinal pigment epithelium-Bruch complex, reminiscent of a "double carrot", which improved following supplementation. ERG findings demonstrated improved scotopic rod-specific function in all cases; however, photopic function remained diminished in two cases. CONCLUSIONS Structural improvement of the proposed "double carrot" sign occurs soon after vitamin A supplementation. While scotopic function improves rapidly following supplementation, cone function recovers more slowly. Therefore, foveal changes such as the "double carrot" sign suggest that structural recovery of cones precedes functional recovery.
Collapse
Grants
- R01 EY024091 NEI NIH HHS
- R21 AG050437 NIA NIH HHS
- R01 EY018213 NEI NIH HHS
- U01 EY030580 NEI NIH HHS
- U54 OD020351 NIH HHS
- R01 EY026682 NEI NIH HHS
- R24 EY027285 NEI NIH HHS
- P30 CA013696 NCI NIH HHS
- R24 EY028758 NEI NIH HHS
- R01 EY024698 NEI NIH HHS
- P30 EY019007 NEI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Eye Institute (NEI)
- Foundation Fighting Blindness (Foundation Fighting Blindness, Inc.)
- Research to Prevent Blindness (RPB) Physician-Scientist Award, and unrestricted funds from RPB, New York, NY, USA
- U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- U.S. Department of Health & Human Services | NIH | NIH Office of the Director (OD)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- The Schneeweiss Stem Cell Fund, New York State [SDHDOH01-C32590GG-3450000], Nancy & Kobi Karp, the Crowley Family Funds, The Rosenbaum Family Foundation, Alcon Research Institute, the Gebroe Family Foundation, the Research to Prevent Blindness (RPB) Physician-Scientist Award, and unrestricted funds from RPB, New York, NY, USA.
Collapse
Affiliation(s)
- Mark P Breazzano
- Jonas Children's Vision Care, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA
- Department of Ophthalmology, New York University Langone Health, New York, NY, USA
- Manhattan Eye, Ear and Throat Hospital, Lenox Hill Hospital, Northwell Health, New York, NY, USA
- Wilmer Eye Institute, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Retina-Vitreous Surgeons of Central New York, Liverpool, NY, USA
- Department of Ophthalmology & Visual Sciences, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Jin Kyun Oh
- Jonas Children's Vision Care, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA
| | | | - Julia A Kucherich
- Department of Nutrition & Therapeutic Services, Morgan Stanley Children's Hospital at New York-Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, USA
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Johns Hopkins Hospital, Johns Hopkins Children's Center, Baltimore, MD, USA
| | - Rabia Karani
- Jonas Children's Vision Care, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA
| | - Caitlin M Rohrmann
- Department of Nutrition & Therapeutic Services, Morgan Stanley Children's Hospital at New York-Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, USA
| | - Janet R Sparrow
- Jonas Children's Vision Care, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | - Serena Fragiotta
- Department of Medical-Surgical Sciences and Biotechnologies, U.O.S.D. Ophthalmology, Sapienza University of Rome, Rome, Italy
| | - Stephen H Tsang
- Jonas Children's Vision Care, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA.
- Columbia Stem Cell Initiative, and Institute of Human Nutrition, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Dhingra A, Tobias JW, Philp NJ, Boesze-Battaglia K. Transcriptomic Changes Predict Metabolic Alterations in LC3 Associated Phagocytosis in Aged Mice. Int J Mol Sci 2023; 24:6716. [PMID: 37047689 PMCID: PMC10095460 DOI: 10.3390/ijms24076716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
LC3b (Map1lc3b) plays an essential role in canonical autophagy and is one of several components of the autophagy machinery that mediates non-canonical autophagic functions. Phagosomes are often associated with lipidated LC3b to promote phagosome maturation in a process called LC3-associated phagocytosis (LAP). Specialized phagocytes, such as mammary epithelial cells, retinal pigment epithelial (RPE) cells, and sertoli cells, utilize LAP for optimal degradation of phagocytosed material, including debris. In the visual system, LAP is critical to maintain retinal function, lipid homeostasis, and neuroprotection. In a mouse model of retinal lipid steatosis-mice lacking LC3b (LC3b-/-), we observed increased lipid deposition, metabolic dysregulation, and enhanced inflammation. Herein, we present a non-biased approach to determine if loss of LAP mediated processes modulate the expression of various genes related to metabolic homeostasis, lipid handling, and inflammation. A comparison of the RPE transcriptome of WT and LC3b-/- mice revealed 1533 DEGs, with ~73% upregulated and 27% downregulated. Enriched gene ontology (GO) terms included inflammatory response (upregulated DEGs), fatty acid metabolism, and vascular transport (downregulated DEGs). Gene set enrichment analysis (GSEA) identified 34 pathways; 28 were upregulated (dominated by inflammation/related pathways) and 6 were downregulated (dominated by metabolic pathways). Analysis of additional gene families identified significant differences for genes in the solute carrier family, RPE signature genes, and genes with a potential role in age-related macular degeneration. These data indicate that loss of LC3b induces robust changes in the RPE transcriptome contributing to lipid dysregulation and metabolic imbalance, RPE atrophy, inflammation, and disease pathophysiology.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John W. Tobias
- Penn Genomics and Sequencing Core, Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy J. Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
Wang X, Sadda SR, Ip MS, Sarraf D, Zhang Y. In Vivo Longitudinal Measurement of Cone Photoreceptor Density in Intermediate Age-Related Macular Degeneration. Am J Ophthalmol 2023; 248:60-75. [PMID: 36436549 PMCID: PMC10038851 DOI: 10.1016/j.ajo.2022.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE To evaluate cone photoreceptor density in clinically unremarkable retinal regions in patients with age-related macular degeneration (AMD) using adaptive optics scanning laser ophthalmoscopy (AOSLO). DESIGN Prospective case series with normal comparison group. METHODS Ten eyes of 7 patients with intermediate AMD were studied, including 4 with predominantly subretinal drusenoid deposits (SDD) and 3 without SDD. Macular regions with a clinical absence of AMD-associated lesions were identified by cone packing structure on AOSLO and optical coherence tomography. Cone density was measured in 1174 clinically unremarkable regions within the central subfield (CSF), the inner (IR), and outer rings (OR) of the Early Treatment Diabetic Retinopathy Study grid over 39.6 ± 3.3 months and compared with age-matched normal values obtained in 17 participants. RESULTS Cone density decreased at 98.3% of the examined locations over time in the eyes with AMD. In the CSF, IR, and OR, cones declined by -255 ± 135, -133 ± 45, and -59 ± 24 cones/degree2/year, respectively, in eyes with SDD, and by -212 ± 89, -83 ± 37, and -27 ± 18 cones/degree2/year, respectively, in eyes without SDD. The percentage of retinal loci with cone density lower than normal (Z score < -2) increased over the follow-up: from 42% at the baseline to 80% at the last visit in eyes with SDD and from 31% to 70% in eyes without SDD. CONCLUSIONS AOSLO revealed cone photoreceptor loss in regions that appear otherwise unremarkable clinically. These findings may help explain the loss of mesopic sensitivity reported in these areas in eyes with intermediate AMD.
Collapse
Affiliation(s)
- Xiaolin Wang
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California
| | - SriniVas R Sadda
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California
| | - Michael S Ip
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California
| | - David Sarraf
- Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California; Stein Eye Institute (David Sarraf), Los Angeles, California, USA
| | - Yuhua Zhang
- From the Doheny Eye Institute (X.W., S.R.S., M.I., Y.Z.), Pasadena, California; Department of Ophthalmology, University of California-Los Angeles (S.R.S., M.I., D.S., Y.Z.), Los Angeles, California.
| |
Collapse
|
31
|
Dhingra A, Tobias JW, Philp NJ, Boesze-Battaglia K. Transcriptomic changes predict metabolic alterations in LC3 associated phagocytosis in aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532586. [PMID: 36993501 PMCID: PMC10054970 DOI: 10.1101/2023.03.14.532586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
LC3b ( Map1lc3b ) plays an essential role in canonical autophagy and is one of several components of the autophagy machinery that mediates non-canonical autophagic functions. Phagosomes are often associated with lipidated LC3b, to pro-mote phagosome maturation in a process called LC3-associated phagocytosis (LAP). Specialized phagocytes such as mammary epithelial cells, retinal pigment epithelial (RPE) cells, and sertoli cells utilize LAP for optimal degradation of phagocytosed material, including debris. In the visual system, LAP is critical to maintain retinal function, lipid homeostasis and neuroprotection. In a mouse model of retinal lipid steatosis - mice lacking LC3b ( LC3b -/- ), we observed increased lipid deposition, metabolic dysregulation and enhanced inflammation. Herein we present a non-biased approach to determine if loss of LAP mediated processes modulate the expression of various genes related to metabolic homeostasis, lipid handling, and inflammation. A comparison of the RPE transcriptome of WT and LC3b -/- mice revealed 1533 DEGs, with ~73% upregulated and 27% down-regulated. Enriched gene ontology (GO) terms included inflammatory response (upregulated DEGs), fatty acid metabolism and vascular transport (downregulated DEGs). Gene set enrichment analysis (GSEA) identified 34 pathways; 28 were upregulated (dominated by inflammation/related pathways) and 6 were downregulated (dominated by metabolic pathways). Analysis of additional gene families identified significant differences for genes in the solute carrier family, RPE signature genes, and genes with potential role in age-related macular degeneration. These data indicate that loss of LC3b induces robust changes in the RPE transcriptome contributing to lipid dysregulation and metabolic imbalance, RPE atrophy, inflammation, and disease pathophysiology.
Collapse
|
32
|
Vasku G, Peltier C, He Z, Thuret G, Gain P, Gabrielle PH, Acar N, Berdeaux O. Comprehensive mass spectrometry lipidomics of human biofluids and ocular tissues. J Lipid Res 2023; 64:100343. [PMID: 36773847 PMCID: PMC10027555 DOI: 10.1016/j.jlr.2023.100343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/12/2023] Open
Abstract
Evaluating lipid profiles in human tissues and biofluids is critical in identifying lipid metabolites in dysregulated metabolic pathways. Due to various chemical characteristics, single-run lipid analysis has not yet been documented. Such approach is essential for analyzing pathology-related lipid metabolites. Age-related macular degeneration, the leading cause of vision loss in western countries, is emblematic of this limitation. Several studies have identified alterations in individual lipids but the majority are based on targeted approaches. In this study, we analyzed and identified approximately 500 lipid species in human biofluids (plasma and erythrocytes) and ocular tissues (retina and retinal pigment epithelium) using the complementarity of hydrophilic interaction liquid chromatography (HILIC) and reversed-phase chromatography (RPC), coupled to high-resolution mass spectrometry. For that, lipids were extracted from human eye globes and blood from 10 subjects and lipidomic analysis was carried out through analysis in HILIC and RPC, alternately. Furthermore, we illustrate the advantages and disadvantages of both techniques for lipid characterization. RPC showed greater sensitivity in hydrophobicity-based lipid separation, detecting diglycerides, triglycerides, cholesterol, and cholesteryl esters, whereas no signal of these molecules was obtained in HILIC. However, due to coelution, RPC was less effective in separating polar lipids like phospholipids, which were separated effectively in HILIC in both ionization modes. The complementary nature of these analytical approaches was essential for the detection and identification of lipid classes/subclasses, which can then provide distinct insights into lipid metabolism, a determinant of the pathophysiology of several diseases involving lipids, notably age-related macular degeneration.
Collapse
Affiliation(s)
- Glenda Vasku
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Franche-Comté, Dijon, France; ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, Institut Agro, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France; INRAE, PROBE Research infrastructure, ChemoSens facility, Dijon, France
| | - Caroline Peltier
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, Institut Agro, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France; INRAE, PROBE Research infrastructure, ChemoSens facility, Dijon, France
| | - Zhiguo He
- Department of Ophthalmology, Biology, Imaging, and Engineering of Corneal Grafts, Faculty of Medicine, Saint Etienne, France
| | - Gilles Thuret
- Department of Ophthalmology, Biology, Imaging, and Engineering of Corneal Grafts, Faculty of Medicine, Saint Etienne, France
| | - Philippe Gain
- Department of Ophthalmology, Biology, Imaging, and Engineering of Corneal Grafts, Faculty of Medicine, Saint Etienne, France
| | - Pierre-Henry Gabrielle
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Franche-Comté, Dijon, France; Department of Ophthalmology, University Hospital, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Franche-Comté, Dijon, France
| | - Olivier Berdeaux
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, Institut Agro, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France; INRAE, PROBE Research infrastructure, ChemoSens facility, Dijon, France.
| |
Collapse
|
33
|
Völkner M, Wagner F, Kurth T, Sykes AM, Del Toro Runzer C, Ebner LJA, Kavak C, Alexaki VI, Cimalla P, Mehner M, Koch E, Karl MO. Modeling inducible neuropathologies of the retina with differential phenotypes in organoids. Front Cell Neurosci 2023; 17:1106287. [PMID: 37213216 PMCID: PMC10196395 DOI: 10.3389/fncel.2023.1106287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Neurodegenerative diseases remain incompletely understood and therapies are needed. Stem cell-derived organoid models facilitate fundamental and translational medicine research. However, to which extent differential neuronal and glial pathologic processes can be reproduced in current systems is still unclear. Here, we tested 16 different chemical, physical, and cell functional manipulations in mouse retina organoids to further explore this. Some of the treatments induce differential phenotypes, indicating that organoids are competent to reproduce distinct pathologic processes. Notably, mouse retina organoids even reproduce a complex pathology phenotype with combined photoreceptor neurodegeneration and glial pathologies upon combined (not single) application of HBEGF and TNF, two factors previously associated with neurodegenerative diseases. Pharmacological inhibitors for MAPK signaling completely prevent photoreceptor and glial pathologies, while inhibitors for Rho/ROCK, NFkB, and CDK4 differentially affect them. In conclusion, mouse retina organoids facilitate reproduction of distinct and complex pathologies, mechanistic access, insights for further organoid optimization, and modeling of differential phenotypes for future applications in fundamental and translational medicine research.
Collapse
Affiliation(s)
- Manuela Völkner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Felix Wagner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Alex M. Sykes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Lynn J. A. Ebner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Cagri Kavak
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Vasileia Ismini Alexaki
- Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Peter Cimalla
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mirko Mehner
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Edmund Koch
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mike O. Karl
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- *Correspondence: Mike O. Karl, ,
| |
Collapse
|
34
|
Weber S, Simon R, Schwanengel LS, Curcio CA, Augsten R, Meller D, Hammer M. Fluorescence Lifetime and Spectral Characteristics of Subretinal Drusenoid Deposits and Their Predictive Value for Progression of Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2022; 63:23. [PMID: 36580310 PMCID: PMC9804024 DOI: 10.1167/iovs.63.13.23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose To measure fundus autofluorescence (FAF) lifetimes and peak emission wavelengths (PEW) of subretinal drusenoid deposits (SDD) in age-related macular degeneration (AMD) and their development over time. Methods Fluorescence lifetime imaging ophthalmoscopy (FLIO) was performed in 30 eyes with optical coherence tomography (OCT)-confirmed early or intermediate AMD and SDD. Contrasts of mean lifetimes in short- (SSC) and long-wavelength channels (LSC), PEW, and relative fluorescence intensity were determined as differences of the respective measures at individual SDD and their environment. Measurements were made at baseline and at follow-up intervals 1 (13-36 months) and 2 (37-72 months), respectively. Results Of 423 SDD found at baseline, 259, 47, and 117 were hypoautofluorescent, isoautofluorescent, and hyperautofluorescent, respectively. FAF lifetimes of SDD were significantly longer than those of their environment by 14.5 ps (SSC, 95% confidence interval [CI], 13.3-15.7 ps) and 3.9 ps (LSC, 3.1-4.7 ps). PEW was shorter by 1.53 nm (1.07-1.98 nm, all contrasts P < 0.001) with higher contrasts for hyperfluorescent SDD. Over follow-up, SDD tended to hyperautofluorescence (relative intensities increased by 3.4% [95% CI, 2.9%-4.1%; P < 0.001] in follow-up 2). Hyperautofluorescence was associated with disruption of the ellipsoid zone on OCT. Disease progression to late-stage AMD was associated with higher lifetime contrast in SSC (15.9ps [14.2-17.6 ps] vs. 11.7 ps [9.9-13.5 ps], P < 0.001) at baseline. Conclusions SDD show longer FAF lifetimes and shorter PEW than their environments. A high lifetime contrast of SDD in SSC might predict disease progression to late-stage AMD.
Collapse
Affiliation(s)
- Sebastian Weber
- Department of Ophthalmology, University Hospital Jena, Jena, Germany
| | - Rowena Simon
- Department of Ophthalmology, University Hospital Jena, Jena, Germany
| | | | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Regine Augsten
- Department of Ophthalmology, University Hospital Jena, Jena, Germany
| | - Daniel Meller
- Department of Ophthalmology, University Hospital Jena, Jena, Germany
| | - Martin Hammer
- Department of Ophthalmology, University Hospital Jena, Jena, Germany,Center for Medical Optics and Photonics, Univ. of Jena, Jena, Germany
| |
Collapse
|
35
|
Natural History of the Relative Ellipsoid Zone Reflectivity in Age-Related Macular Degeneration. Ophthalmol Retina 2022; 6:1165-1172. [PMID: 35709960 DOI: 10.1016/j.oret.2022.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/08/2022] [Accepted: 06/06/2022] [Indexed: 01/06/2023]
Abstract
PURPOSE Relative ellipsoid zone reflectivity (rEZR) has been reported to be reduced in intermediate age-related macular degeneration (iAMD). However, longitudinal changes in rEZR remain unknown. This study investigated the natural history of rEZR in iAMD and its association with risk factors for disease progression, including the presence or extent of drusen volume, reticular pseudodrusen (RPD), and pigmentary abnormalities (PAs). DESIGN Longitudinal observational study. PARTICIPANTS Subjects with bilateral large drusen. METHODS Spectral-domain (SD) OCT images of both eyes from each participant were obtained every 6 months for 3 years. Using an automated rEZR determination approach, the average rEZR of the central 20° macula was determined for each SD-OCT volume scan. Linear mixed models were used to determine the rate of change in rEZR with age (using the cross-sectional data at baseline) and over time (longitudinal data) and the interactions between the rate of rEZR changes with AMD risk factors at baseline. MAIN OUTCOME MEASURES Relative ellipsoid zone reflectivity and its rate of change with age and over time. RESULTS A total of 280 eyes from 140 individuals with bilateral large drusen were included in this study. Cross-sectional data showed that rEZR reduced with increasing age (-8.4 arbitrary units [AUs] per decade; 95% confidence interval [CI], -11.5 to -5.2; P < 0.001). Longitudinal data showed that, on average, rEZR declined at a rate of -2.1 AU per year (95% CI, -2.6 to -1.6 AU per year; P < 0.001). Larger RPD area (P = 0.042) at baseline was associated with a faster rate of rEZR decline over time, whereas the presence of PAs and the drusen volume at baseline showed no significant association with rEZR decline over time (P = 0.068 and P = 0.529, respectively). CONCLUSIONS The rEZR significantly reduces over 3 years in subjects with iAMD, and both the presence and increasing extent of coexistent RPD at baseline are associated with a faster rate of decline. These findings warrant further studies to understand the value of rEZR as a biomarker of AMD progression.
Collapse
|
36
|
Malek G, Campisi J, Kitazawa K, Webster C, Lakkaraju A, Skowronska-Krawczyk D. Does senescence play a role in age-related macular degeneration? Exp Eye Res 2022; 225:109254. [PMID: 36150544 PMCID: PMC10032649 DOI: 10.1016/j.exer.2022.109254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 12/29/2022]
Abstract
Advanced age is the most established risk factor for developing age-related macular degeneration (AMD), one of the leading causes of visual impairment in the elderly, in Western and developed countries. Similarly, after middle age, there is an exponential increase in pathologic molecular and cellular events that can induce senescence, traditionally defined as an irreversible loss of the cells' ability to divide and most recently reported to also occur in select post-mitotic and terminally differentiated cells, such as neurons. Together these facts raise the question as to whether or not cellular senescence, may play a role in the development of AMD. A number of studies have reported the effect of ocular-relevant inducers of senescence using primarily in vitro models of poorly polarized, actively dividing retinal pigment epithelial (RPE) cell lines. However, in interpretating the data, the fidelity of these culture models to the RPE in vivo, must be considered. Fewer studies have explored the presence and/or impact of senescent cells in in vivo models that present with phenotypic features of AMD, leaving this an open field for further investigation. The goal of this review is to discuss current thoughts on the potential role of senescence in AMD development and progression, with consideration of the model systems used and their relevance to human disease.
Collapse
Affiliation(s)
- Goldis Malek
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA; Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Koji Kitazawa
- Buck Institute for Research on Aging, Novato, CA, USA; Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Corey Webster
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Aparna Lakkaraju
- Departments of Ophthalmology and Anatomy, School of Medicine, University of California, San Francisco, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
37
|
Sasaki M, Kawasaki R, Yanagi Y. Early Stages of Age-Related Macular Degeneration: Racial/Ethnic Differences and Proposal of a New Classification Incorporating Emerging Concept of Choroidal Pathology. J Clin Med 2022; 11:6274. [PMID: 36362505 PMCID: PMC9657039 DOI: 10.3390/jcm11216274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/08/2022] [Accepted: 10/18/2022] [Indexed: 01/20/2025] Open
Abstract
The progression of age-related macular degeneration (AMD) is determined by environmental and genetic factors, and phenotypic or molecular risk factors have been investigated extensively. Interestingly, risk factor profiles for advanced AMD differ among individuals, and one of the causes of variation may be explained by their ethnic background. Recent advances in retinal imaging technology have led to the identification of previously unrecognized risk factors for advanced AMD on optical coherence tomography (OCT) and OCT angiography, which expands the concept of traditional imaging risk factors such as drusen and pigmentary abnormalities visible on color fundus photographs. This OCT imaging modality has identified novel pathognomonic changes for early AMD, including the associated photoreceptor, retinal pigment epithelium, and underlying choroidal changes. Regarding features of multimodal imaging associated with the presence or progression of geographic atrophy, there is an international expert consensus classification system; however, features associated with the progression of macular neovascularization (MNV) are still obscure. To make a consensus towards understanding features associated with the risk of MNV, this review focuses on the early stages of AMD by summarizing imaging characteristics and early signs and classifications in view of advanced multimodal imaging technology. Recent evidence suggests that neovascular AMD is not a single disease entity but a heterogeneous disease characterized by MNV. Besides drusen, OCT features associated with pigment abnormalities, such as shallow irregular RPE elevation (SIRE, also known as double-layer sign), pachychoroid pigment epitheliopathy, and choriocapillaris ischemia, seem to confer a high risk of MNV developing, especially for Asian populations.
Collapse
Affiliation(s)
- Mariko Sasaki
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Tokyo Medical Center, Tokyo 152-8902, Japan
| | - Ryo Kawasaki
- Department of Vision Informatics (Topcon), Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yasuo Yanagi
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanazawa 236-0027, Japan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169608, Singapore
| |
Collapse
|
38
|
Pfau M, Schmitz-Valckenberg S, Ribeiro R, Safaei R, McKeown A, Fleckenstein M, Holz FG. Association of complement C3 inhibitor pegcetacoplan with reduced photoreceptor degeneration beyond areas of geographic atrophy. Sci Rep 2022; 12:17870. [PMID: 36284220 PMCID: PMC9596427 DOI: 10.1038/s41598-022-22404-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/14/2022] [Indexed: 01/20/2023] Open
Abstract
Preservation of photoreceptors beyond areas of retinal pigment epithelium atrophy is a critical treatment goal in eyes with geographic atrophy (GA) to prevent vision loss. Thus, we assessed the association of treatment with the complement C3 inhibitor pegcetacoplan with optical coherence tomography (OCT)-based photoreceptor laminae thicknesses in this post hoc analysis of the FILLY trial (NCT02503332). Retinal layers in OCT were segmented using a deep-learning-based pipeline and extracted along evenly spaced contour-lines surrounding areas of GA. The primary outcome measure was change from baseline in (standardized) outer nuclear layer (ONL) thickness at the 5.16°-contour-line at month 12. Participants treated with pegcetacoplan monthly had a thicker ONL along the 5.16° contour-line compared to the pooled sham arm (mean difference [95% CI] + 0.29 z-score units [0.16, 0.42], P < 0.001). The same was evident for eyes treated with pegcetacoplan every other month (+ 0.26 z-score units [0.13, 0.4], P < 0.001). Additionally, eyes treated with pegcetacoplan exhibited a thicker photoreceptor inner segment layer along the 5.16°-contour-line at month 12. These findings suggest that pegcetacoplan could slow GA progression and lead to reduced thinning of photoreceptor layers beyond the GA boundary. Future trials in earlier disease stages, i.e., intermediate AMD, aiming to slow photoreceptor degeneration warrant consideration.
Collapse
Affiliation(s)
- Maximilian Pfau
- Department of Ophthalmology, University of Bonn, Bonn, Germany
- GRADE Reading Center, Bonn, Germany
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Steffen Schmitz-Valckenberg
- Department of Ophthalmology, University of Bonn, Bonn, Germany.
- GRADE Reading Center, Bonn, Germany.
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, 65 North Mario Capecchi Drive, Salt Lake City, UT, 84312, USA.
| | | | | | | | - Monika Fleckenstein
- GRADE Reading Center, Bonn, Germany
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah, 65 North Mario Capecchi Drive, Salt Lake City, UT, 84312, USA
| | - Frank G Holz
- Department of Ophthalmology, University of Bonn, Bonn, Germany
- GRADE Reading Center, Bonn, Germany
| |
Collapse
|
39
|
Völkner M, Wagner F, Steinheuer LM, Carido M, Kurth T, Yazbeck A, Schor J, Wieneke S, Ebner LJA, Del Toro Runzer C, Taborsky D, Zoschke K, Vogt M, Canzler S, Hermann A, Khattak S, Hackermüller J, Karl MO. HBEGF-TNF induce a complex outer retinal pathology with photoreceptor cell extrusion in human organoids. Nat Commun 2022; 13:6183. [PMID: 36261438 PMCID: PMC9581928 DOI: 10.1038/s41467-022-33848-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Human organoids could facilitate research of complex and currently incurable neuropathologies, such as age-related macular degeneration (AMD) which causes blindness. Here, we establish a human retinal organoid system reproducing several parameters of the human retina, including some within the macula, to model a complex combination of photoreceptor and glial pathologies. We show that combined application of TNF and HBEGF, factors associated with neuropathologies, is sufficient to induce photoreceptor degeneration, glial pathologies, dyslamination, and scar formation: These develop simultaneously and progressively as one complex phenotype. Histologic, transcriptome, live-imaging, and mechanistic studies reveal a previously unknown pathomechanism: Photoreceptor neurodegeneration via cell extrusion. This could be relevant for aging, AMD, and some inherited diseases. Pharmacological inhibitors of the mechanosensor PIEZO1, MAPK, and actomyosin each avert pathogenesis; a PIEZO1 activator induces photoreceptor extrusion. Our model offers mechanistic insights, hypotheses for neuropathologies, and it could be used to develop therapies to prevent vision loss or to regenerate the retina in patients suffering from AMD and other diseases.
Collapse
Affiliation(s)
- Manuela Völkner
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Felix Wagner
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lisa Maria Steinheuer
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Madalena Carido
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Ali Yazbeck
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Jana Schor
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Stephanie Wieneke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lynn J A Ebner
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | | | - David Taborsky
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Katja Zoschke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Marlen Vogt
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sebastian Canzler
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Andreas Hermann
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- Department of Neurology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Shahryar Khattak
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Jörg Hackermüller
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Department of Computer Science, Leipzig University, Leipzig, Germany
| | - Mike O Karl
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| |
Collapse
|
40
|
Saßmannshausen M, Behning C, Isselmann B, Schmid M, Finger RP, Holz FG, Schmitz-Valckenberg S, Pfau M, Thiele S. Relative ellipsoid zone reflectivity and its association with disease severity in age-related macular degeneration: a MACUSTAR study report. Sci Rep 2022; 12:14933. [PMID: 36056113 PMCID: PMC9440143 DOI: 10.1038/s41598-022-18875-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Quantification of the relative ellipsoid zone reflectivity (rEZR) might be a structural surrogate parameter for an early disease progression in the context of age-related macular degeneration (AMD). Within the European multicenter, cross-sectional MACUSTAR study, we have devised an automatic approach to determine the mean rEZR [arbitrary units, AU] at two independent visits in SD-OCT volume scans in study participants. Linear mixed-effects models were applied to analyze the association of AMD stage and AMD associated high-risk features including presence of pigmentary abnormalities, reticular pseudodrusen (RPD), volume of the retinal-pigment-epithelial-drusenoid-complex (RPEDC) with the rEZR. Intra-class correlation coefficients (ICC) were determined for rEZR reliability analysis. Within the overall study cohort (301 participants), we could observe decreased rEZR values (coefficient estimate ± standard error) of - 8.05 ± 2.44 AU (p = 0.0011) in the intermediate and of - 22.35 ± 3.28 AU (p < 0.0001) in the late AMD group. RPD presence was significantly associated with the rEZR in iAMD eyes (- 6.49 ± 3.14 AU; p = 0.0403), while there was a good ICC of 0.846 (95% confidence interval: 0.809; 0.876) in the overall study cohort. This study showed an association of rEZR with increasing disease severity and the presence of iAMD high-risk features. Further studies are necessary to evaluate the rEZR's value as a novel biomarker for AMD and disease progression.
Collapse
Affiliation(s)
- Marlene Saßmannshausen
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- GRADE Reading Center, University of Bonn, Bonn, Germany
| | - Charlotte Behning
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Ben Isselmann
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Matthias Schmid
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Robert P Finger
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Frank G Holz
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- GRADE Reading Center, University of Bonn, Bonn, Germany
| | - Steffen Schmitz-Valckenberg
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- GRADE Reading Center, University of Bonn, Bonn, Germany
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Maximilian Pfau
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- GRADE Reading Center, University of Bonn, Bonn, Germany
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, MD, USA
| | - Sarah Thiele
- Department of Ophthalmology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- GRADE Reading Center, University of Bonn, Bonn, Germany.
| |
Collapse
|
41
|
Brinkmann M, Bacci T, Kar D, Messinger JD, Sloan KR, Chen L, Hamann T, Wiest M, Freund KB, Zweifel S, Curcio CA. Histology and Clinical Lifecycle of Acquired Vitelliform Lesion, a Pathway to Advanced Age-Related Macular Degeneration. Am J Ophthalmol 2022; 240:99-114. [PMID: 35192790 PMCID: PMC9592119 DOI: 10.1016/j.ajo.2022.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE To evaluate hypotheses about the role of acquired vitelliform lesion (AVL) in age-related macular degeneration pathophysiology. DESIGN Laboratory histology study; retrospective, observational case series. METHODS Two donor eyes in a research archive with AVL and age-related macular degeneration were analyzed with light and electron microscopy for AVL content at locations matched to ex vivo B-scans. A retrospective, observational clinical cohort study of 42 eyes of 30 patients at 2 referral clinics determined the frequency of optical coherence tomography features stratified by AVL fate. RESULTS Histologic and clinical cases showed subretinal drusenoid deposit and drusen. Ultrastructural AVL components in 2 donor eyes included retinal pigment epithelium (RPE) organelles (3%-22% of volume), outer segments (2%-10%), lipid droplets (0.2%-12%), and a flocculent material (57%-59%). Of 48 AVLs (mean follow-up 46 ± 39 months), 50% collapsed to complete RPE and outer retinal atrophy, 38% were stable, 10% resorbed, and 2% developed neovascularization. The Early Treatment Diabetic Retinopathy Study grid central subfield contained 77% of AVLs. Hyperreflective foci, ellipsoid zone disruption, and hyperreflective thickening of the RPE-basal lamina-Bruch membrane band were common at maximum AVL expansion. Collapsing and noncollapsing AVLs had different growth rates (rapid vs slow, respectively). CONCLUSIONS AVL deposits contain unexpectedly low levels of RPE organelles and outer segments. Subfoveal predilection, reflectivity on optical coherence tomography, hyperautofluorescence, yellow color, and growth-regression phases suggest dysregulation of lipid transfer pathways specific to cone photoreceptors and supporting cells in formation of AVL deposit, analogous to drusen and subretinal drusenoid deposit. Prediction of AVL outcomes via growth rates should be confirmed in larger clinical studies.
Collapse
Affiliation(s)
- Max Brinkmann
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA; Department of Ophthalmology (M.B., T.H., M.W., S.Z.), University Hospital Zurich, Zurich, Switzerland
| | - Tommaso Bacci
- Vitreous Retina Macula Consultants of New York (T.B., K.B.F.), New York University School of Medicine, New York, New York, USA
| | - Deepayan Kar
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Jeffrey D Messinger
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Kenneth R Sloan
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Ling Chen
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA; First Affiliated Hospital of Chongqing Medical University (L.C.), Chongqing Key Laboratory of Ophthalmology, and Chongqing Eye Institute, Chongqing, China
| | - Timothy Hamann
- Department of Ophthalmology (M.B., T.H., M.W., S.Z.), University Hospital Zurich, Zurich, Switzerland
| | - Maximilian Wiest
- Department of Ophthalmology (M.B., T.H., M.W., S.Z.), University Hospital Zurich, Zurich, Switzerland
| | - K Bailey Freund
- Vitreous Retina Macula Consultants of New York (T.B., K.B.F.), New York University School of Medicine, New York, New York, USA; LuEsther T. Mertz Retinal Research Center (K.B.F.), New York University School of Medicine, New York, New York, USA; Manhattan Eye, Ear and Throat Hospital, and the Department of Ophthalmology (K.B.F.), New York University School of Medicine, New York, New York, USA
| | - Sandrine Zweifel
- Department of Ophthalmology (M.B., T.H., M.W., S.Z.), University Hospital Zurich, Zurich, Switzerland; Department of Ophthalmology, University of Zurich (S.Z.), Zurich, Switzerland
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences (M.B., D.K., J.D.M., K.R.S., L.C., C.A.C.), University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA.
| |
Collapse
|
42
|
Mainster MA, Findl O, Dick HB, Desmettre T, Ledesma-Gil G, Curcio CA, Turner PL. The Blue Light Hazard Versus Blue Light Hype. Am J Ophthalmol 2022; 240:51-57. [PMID: 35227699 PMCID: PMC10243475 DOI: 10.1016/j.ajo.2022.02.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE The blue light hazard is the experimental finding that blue light is highly toxic to the retina (photic retinopathy), in brief abnormally intense exposures, including sungazing or vitreoretinal endoillumination. This term has been misused commercially to suggest, falsely, that ambient environmental light exposure causes phototoxicity to the retina, leading to age-related macular degeneration (AMD). We analyze clinical, epidemiologic, and biophysical data regarding blue-filtering optical chromophores. DESIGN Perspective. METHODS Analysis and integration of data regarding the blue light hazard and blue-blocking filters in ophthalmology and related disciplines. RESULTS Large epidemiologic studies show that blue-blocking intraocular lenses (IOLs) do not decrease AMD risk or progression. Blue-filtering lenses cannot reduce disability glare because image and glare illumination are decreased in the same proportion. Blue light essential for optimal rod and retinal ganglion photoreception is decreased by progressive age-related crystalline lens yellowing, pupillary miosis, and rod and retinal ganglion photoreceptor degeneration. Healthful daily environmental blue light exposure decreases in older adults, especially women. Blue light is important in dim environments where inadequate illumination increases risk of falls and associated morbidities. CONCLUSIONS The blue light hazard is misused as a marketing stratagem to alarm people into using spectacles and IOLs that restrict blue light. Blue light loss is permanent for pseudophakes with blue-blocking IOLs. Blue light hazard misrepresentation flourishes despite absence of proof that environmental light exposure or cataract surgery causes AMD or that IOL chromophores provide clinical protection. Blue-filtering chromophores suppress blue light critical for good mental and physical health and for optimal scotopic and mesopic vision.
Collapse
Affiliation(s)
- Martin A Mainster
- Department of Ophthalmology, University of Kansas School of Medicine, Prairie Village, Kansas, USA.
| | - Oliver Findl
- Vienna Institute for Research in Ocular Surgery, A Karl Landsteiner Institute, Hanusch Hospital, Vienna, Austria
| | - H Burkhard Dick
- Department of Ophthalmology, Ruhr University Eye Hospital, Science, Bochum, Germany
| | | | - Gerardo Ledesma-Gil
- Retina Department, Institute of Ophthalmology, Fundación Conde de Valenciana, Mexico City, Mexico
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Patricia L Turner
- Department of Ophthalmology, University of Kansas School of Medicine, Prairie Village, Kansas, USA
| |
Collapse
|
43
|
Kang D, Yoon EG, Nam KT, Yun C. Chorioretinal thickness and retinal pigment epithelial degeneration of fellow eyes in patients with unilateral neovascular age-related macular degeneration with subretinal drusenoid deposits. BMC Ophthalmol 2022; 22:304. [PMID: 35836149 PMCID: PMC9284825 DOI: 10.1186/s12886-022-02518-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background We sought to investigate the chorioretinal thickness and retinal pigment epithelial (RPE) degenerative features of eyes with early age-related macular degeneration (AMD) and subretinal drusenoid deposits (SDDs) according to the presence of macular neovascularization (MNV) in the fellow eyes. Methods We classified 70 eyes into two groups of 47 eyes with non-neovascular AMD and 23 eyes with neovascular AMD, respectively, according to the presence of MNV in the fellow eyes. The mean macular retinal, ganglion cell–inner plexiform layer (GCIPL), and choroidal thickness values and RPE features of the 6-mm-diameter zone were compared. RPE degeneration was defined as a lesion with an incomplete RPE and outer retinal atrophy (iRORA) or attenuated RPE reflectivity with diffuse basal laminar deposits, which was defined as when the eye showed an attenuated RPE line with granular features and mixed reflectivity in combination with sub-RPE deposits with a lesion ≥ 1,000 µm in length. Results Mean retinal, GCIPL, and choroidal thickness values (286.69 ± 15.02 µm, 64.36 ± 4.21 µm, and 156.11 ± 33.10 µm) of the neovascular AMD group were greater than those (278.61 ± 13.96 µm, 61.44 ± 4.63 µm, and 133.59 ± 34.33 µm) of the non-neovascular AMD group (all P < 0.05). RPE degeneration was more prevalent in the neovascular AMD group (65.2%) than the non-neovascular AMD group (38.3%; P = 0.034). Greater mean GCIPL and choroidal thickness values and the presence of RPE degeneration were associated with type 3 MNV in fellow eyes (all P < 0.05). Conclusions Different degenerative features according to MNV in fellow eyes of patients with AMD and SDDs suggest that variable degenerative features might be present during disease progression and have an association with the phenotype. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-022-02518-4.
Collapse
Affiliation(s)
- Dongwan Kang
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Eun Gyu Yoon
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Ki Tae Nam
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Cheolmin Yun
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
44
|
Chen J, Cao D, Fortmann SD, Curcio CA, Feist RM, Crosson JN. Transthyretin proteoforms of intraocular origin in human subretinal fluid. Exp Eye Res 2022; 222:109163. [PMID: 35760119 DOI: 10.1016/j.exer.2022.109163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022]
Abstract
Understanding the molecular composition of ocular tissues and fluids could inform new approaches to prevalent causes of blindness. Subretinal fluid accumulating between the photoreceptor outer segments and retinal pigment epithelium (RPE) is potentially a rich source of proteins and lipids normally cycling among outer retinal cells and choroid. Herein, intact post-translationally modified proteins (proteoforms) were extracted from subretinal fluids of five patients with rhegmatogenous retinal detachment (RRD), analyzed by tandem mass spectrometry, and compared to published data on these same proteins as synthesized by other organs. Single-nuclei transcriptomic data from non-diseased human retina/RPE were used to identify whether proteins in subretinal fluid were of potential ocular origin. Two human donor eyes with normal maculas were immunoprobed for transthyretin (TTR) with appropriate controls. The three most abundant proteins detected in subretinal fluid were albumin, TTR, and apolipoprotein A-I. Remarkably, TTR relative to the other proteins was more abundant than its serum counterpart, suggestive of TTR being synthesized predominantly locally. Six post-translationally modified protein forms (proteoforms) of TTR were detected, with the relative amount of glutathionylated TTR being much higher in the subretinal fluid (12-43%) than values reported for serum (<5%) and cerebrospinal fluid (0.4-13%). Moreover, a putative glycosylated TTR dimer of 32,428 Da was detected as the fourth most abundant protein. The high abundance of TTR and putative TTR dimer in subretinal fluid was supported by analysis of available single-nuclei transcriptomic data, which showed strong and specific signal for TTR in RPE. Immunohistochemistry further showed strong diffuse TTR immunoreactivity in choroidal stroma that contrasted with vertically aligned signal in the outer segment zone of the subretinal space and negligible signal in RPE cell bodies. These results suggest that TTR in the retina is synthesized intraocularly, and glutathionylation is crucial for its normal function. Further studies on the composition, function, and quantities of TTR and other proteoforms in subretinal fluid could inform mechanisms, diagnostic methods, and treatment strategies for age-related macular degeneration, familial amyloidosis, and other retinal diseases involving dysregulation of physiologic lipid transfer and oxidative stress.
Collapse
Affiliation(s)
- Jianzhong Chen
- Department of Optometry and Vision Science, The University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Dongfeng Cao
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Seth D Fortmann
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Richard M Feist
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason N Crosson
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
45
|
Jeffrey BG, Flynn OJ, Huryn LA, Pfau M, Cukras CA. Scotopic Contour Deformation Detection Reveals Early Rod Dysfunction in Age-Related Macular Degeneration With and Without Reticular Pseudodrusen. Invest Ophthalmol Vis Sci 2022; 63:23. [PMID: 35749129 PMCID: PMC9234356 DOI: 10.1167/iovs.63.6.23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to investigate scotopic contour deformation detection (sCDD), and its structural determinants, in participants with intermediate age-related macular degeneration (iAMD) with or without reticular pseudodrusen (RPD). Methods Forty-one participants (aged 58–89 years), including 9 with iAMD and RPD, 16 with iAMD only, and 16 controls, underwent functional testing. The sCDD was evaluated with radial frequency arcs presented at 4 loci: ±4 degrees and 8 degrees vertical eccentricity. Scotopic thresholds and dark adaptation (DA) were measured at the same loci. Retinal layers of spectral domain optical coherence tomography (SD-OCT) volume scans were segmented. To establish the concurrent validity of the functional test, we evaluated the fraction of variability in sCDD thresholds explained by SD-OCT data. Results The iAMD group had significantly worse sCDD thresholds compared with controls (8 degrees inferior retina: P = 0.004 and the 4 degrees loci: P < 0.02 for both). Elevated sCDD thresholds were observed in iAMD and RPD eyes at loci with normal scotopic thresholds; the opposite was rarely encountered. Elevated sCDD thresholds were also observed in iAMD eyes with normal DA. Elevated sCDD thresholds were associated with increased age and presence of late AMD in the fellow eye. The optimal machine learning model predicted 16% of variability (cross-validated R2) in sCDD thresholds at 8 degrees. Discussion A novel scotopic contour deformation task can provide unique information about rod dysfunction in participants with iAMD and RPD not observed with structural and other functional assessments. Rod dysfunction observed with scotopic contour deformation testing was associated with factors linked to risk of AMD progression.
Collapse
Affiliation(s)
- Brett G Jeffrey
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Oliver J Flynn
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Laryssa A Huryn
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Maximilian Pfau
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States.,University of Bonn, Bonn, Germany
| | - Catherine A Cukras
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
46
|
Young BK, Shen LL, Del Priore LV. Subretinal Drusenoid Deposit Formation: Insights From Turing Patterns. Transl Vis Sci Technol 2022; 11:5. [PMID: 35254421 PMCID: PMC8914568 DOI: 10.1167/tvst.11.3.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to demonstrate that the organized formation of subretinal drusenoid deposits (SDDs) may be a Turing pattern. Methods A Java-based computational model of an inferred reaction-diffusion system using paired partial differential equations was used to create topographic images. Reaction kinetics were varied to illustrate a spectrum of pattern development, which were then compared to dot-like, reticular, and confluent SDD patterns observed clinically. Results A reaction-diffusion system using two agents, one an “activator” that increases its own production, and the other an “inhibitor” that decreases the activator's production, can create patterns that match the spectrum of topographic appearance of organized SDD. By varying a single parameter, the strength of the activator, the full spectrum of clinically observed SDD patterns can be generated. A new pattern, confluence with holes, is predicted and identified in one case example. Conclusions The formation of clinically significant SDD and its different patterns can be explained using Turing patterns obtained by simulating a two-component reaction-diffusion system. Translational Relevance This model may be able to guide future risk stratification for patients with SDD, and provide mechanistic insights into the cause of the disease.
Collapse
Affiliation(s)
- Benjamin K Young
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT, USA.,Kellogg Eye Center, Department of Ophthalmology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Liangbo L Shen
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
47
|
A common finding in foveal-sparing extensive macular atrophy with pseudodrusen (EMAP) implicates basal laminar deposits. Retina 2022; 42:1319-1329. [DOI: 10.1097/iae.0000000000003463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Zhang Y, Sadda SR, Sarraf D, Swain TA, Clark ME, Sloan KR, Warriner WE, Owsley C, Curcio CA. Spatial Dissociation of Subretinal Drusenoid Deposits and Impaired Scotopic and Mesopic Sensitivity in AMD. Invest Ophthalmol Vis Sci 2022; 63:32. [PMID: 35212721 PMCID: PMC8883144 DOI: 10.1167/iovs.63.2.32] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Subretinal drusenoid deposits (SDD) first appear in the rod-rich perifovea and can extend to the cone-rich fovea. To refine the spatial relationship of visual dysfunction with SDD burden, we determined the topography of mesopic and scotopic light sensitivity in participants with non-neovascular AMD with and without SDD. Methods Thirty-three subjects were classified into three groups: normal (n = 9), AMD-Drusen (with drusen and without SDD; n = 12), and AMD-SDD (predominantly SDD; n = 12). Mesopic and scotopic microperimetry were performed using 68 targets within the Early Treatment Diabetic Retinopathy Study grid, including points at 1.7° from the foveal center (rod:cone ratio, 0.35). Age-adjusted linear regression was used to compare mesopic and scotopic light sensitivities across groups. Results Across the entire Early Treatment Diabetic Retinopathy Study grid and within individual subfields, the three groups differed significantly for mesopic and scotopic light sensitivities (all P < 0.05). The AMD-SDD group exhibited significantly decreased mesopic and scotopic sensitivity versus both the normal and the AMD-Drusen groups (all P < 0.05), while AMD-Drusen and normal eyes did not significantly differ (all P > 0.05). The lowest relative sensitivities were recorded for scotopic light levels, especially in the central subfield, in the AMD-SDD group. Conclusions SDD-associated decrements in rod-mediated vision can be detected close to the foveola, and these deficits are proportionately worse than functional loss in the rod-rich perifovea. This finding suggests that factors other than the previously hypothesized direct cytotoxicity to photoreceptors and local transport barrier limitations may negatively impact vision. Larger prospective studies are required to confirm these observations.
Collapse
Affiliation(s)
- Yuhua Zhang
- Doheny Eye Institute, Los Angeles, California, United States.,Department of Ophthalmology, University of California - Los Angeles, Los Angeles, California, United States
| | - SriniVas R Sadda
- Doheny Eye Institute, Los Angeles, California, United States.,Department of Ophthalmology, University of California - Los Angeles, Los Angeles, California, United States
| | - David Sarraf
- Department of Ophthalmology, University of California - Los Angeles, Los Angeles, California, United States.,Jules Stein Eye Institute, Los Angeles, California, United States
| | - Thomas A Swain
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Mark E Clark
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kenneth R Sloan
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - William E Warriner
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Research Computing, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Cynthia Owsley
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
49
|
Wu L, Monge M, Araya A. Subretinal drusenoid deposits: An update. Taiwan J Ophthalmol 2022; 12:138-146. [PMID: 35813798 PMCID: PMC9262011 DOI: 10.4103/tjo.tjo_18_22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/12/2022] [Indexed: 11/30/2022] Open
Abstract
A wide spectrum of phenotypic manifestations characterizes age-related macular degeneration (AMD). Drusen is considered the hallmark of AMD and is located underneath the retinal pigment epithelium (RPE). In contrast, subretinal drusenoid deposits (SDDs), also known as reticular pseudodrusens, are located in the subretinal space, on top of the RPE. SDDs are poorly detected by clinical examination and color fundus photography. Multimodal imaging is required for their proper diagnosis. SDDs are topographically and functionally related to rods. SDDs cause a deep impairment in retinal sensitivity and dark adaptation. SDDs are dynamic structures that may grow, fuse with each other, or regress over time. An intermediate step in some eyes is the development of an acquired vitelliform lesion. The presence of SDD confers an eye a high risk for the development of late AMD. SDD leads to macular neovascularization, particularly type 3, geographic atrophy, and outer retinal atrophy.
Collapse
|
50
|
Rossi EA, Norberg N, Eandi C, Chaumette C, Kapoor S, Le L, Snyder VC, Martel JN, Gautier J, Gocho K, Dansingani KK, Chhablani J, Arleo A, Mrejen S, Sahel JA, Grieve K, Paques M. A New Method for Visualizing Drusen and Their Progression in Flood-Illumination Adaptive Optics Ophthalmoscopy. Transl Vis Sci Technol 2021; 10:19. [PMID: 34928325 PMCID: PMC8709936 DOI: 10.1167/tvst.10.14.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Drusen are dynamic sub-RPE deposits that are risk factors for late-stage age-related macular degeneration (AMD). Here we show a new imaging method using flood-illumination adaptive optics (FIAO) that reveal drusen with high contrast and resolution. Methods A fovea-centered 4° × 4° FIAO image and eight surrounding images with gaze displaced by ±2° vertically and horizontally were acquired. Clinical color fundus and spectral-domain optical coherence tomography were acquired for clinical grading and comparison. Custom software registered overlapping FIAO images and fused the data statistically to generate a fovea-centered 4° × 4° gaze-dependent image. Our dataset included 15 controls (aged 31-72) and 182 eyes from 104 AMD patients (aged 56-92), graded as either normal aging (n = 7), and early (n = 12), intermediate (n = 108) and late AMD (n = 42); 27 had subretinal drusenoid deposits (SDDs), and 83 were imaged longitudinally. Results No gaze varying structures were detected in young eyes. In aging eyes with no evidence of age-related changes, putative drusen <20 µm in diameter were visible. Gaze-dependent images revealed more drusen and many smaller drusen than visible in color fundus images. Longitudinal images showed expansion and fusion of drusen. SDDs were lower contrast, and RPE atrophy did not yield a consistent signal. Conclusions Gaze-dependent imaging in a commercially available FIAO fundus camera combined with image registration and postprocessing permits visualization of drusen and their progression with high contrast and resolution. Translational Relevance This new technique offers promise as a robust and sensitive method to detect, map, quantify, and monitor the dynamics of drusen in aging and AMD.
Collapse
Affiliation(s)
- Ethan A Rossi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathaniel Norberg
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Chiara Eandi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France.,Department of Surgical Science, University of Torino, Turin, Italy
| | - Celine Chaumette
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Saloni Kapoor
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura Le
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Valerie C Snyder
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joseph N Martel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Josselin Gautier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Kiyoko Gocho
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Kunal K Dansingani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Angelo Arleo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Sarah Mrejen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Kate Grieve
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Michel Paques
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France and CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| |
Collapse
|