1
|
Ong J, Selvam A, Driban M, Zarnegar A, Morgado Mendes Antunes Da Silva SI, Joy J, Rossi EA, Vande Geest JP, Sahel JA, Chhablani J. Characterizing Bruch's membrane: State-of-the-art imaging, computational segmentation, and biologic models in retinal disease and health. Prog Retin Eye Res 2025; 106:101358. [PMID: 40254245 DOI: 10.1016/j.preteyeres.2025.101358] [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: 02/02/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
The Bruch's membrane (BM) is an acellular, extracellular matrix that lies between the choroid and retinal pigment epithelium (RPE). The BM plays a critical role in retinal health, performing various functions including biomolecule diffusion and RPE support. The BM is also involved in many retinal diseases, and insights into BM dysfunction allow for further understanding of the pathophysiology of various chorioretinal pathologies. Thus, characterization of the BM serves as an important area of research to further understand its involvement in retinal disease. In this article, we provide a review of various advancements in characterizing and visualizing the BM. We provide an overview of the BM in retinal health, as well as changes observed in aging and disease. We then describe current state-of-the-art imaging modalities and advances to further visualize the BM including various types of optical coherence tomography imaging, near-infrared reflectance (NIR), and autofluorescence imaging and tissue matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS). Following advances in imaging of the BM, we describe animal, cellular, and synthetic models that have been developed to further visualize the BM. Following this section, we provide an overview of deep learning in retinal imaging and describe advances in computational and artificial intelligence (AI) techniques to provide automated segmentation of the BM and BM opening. We conclude this section considering the clinical implications of these segmentation techniques. Ultimately, the diverse advances aimed to further characterize the BM may allow for deeper insights into the involvement of this critical structure in retinal health and disease.
Collapse
Affiliation(s)
- Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, United States
| | - Amrish Selvam
- Illinois Eye and Ear Infirmary, University of Illinois College of Medicine, Chicago, IL, United States
| | - Matthew Driban
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, United States
| | - Arman Zarnegar
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Jincy Joy
- Karunya Eye Hospital, Kottarakara, Kerala, India
| | - Ethan A Rossi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
2
|
Schumann U, Liu L, Aggio-Bruce R, Cioanca AV, Shariev A, Madigan MC, Valter K, Wen J, Natoli R. Spatial transcriptomics reveals regionally altered gene expression that drives retinal degeneration. Commun Biol 2025; 8:629. [PMID: 40251274 PMCID: PMC12008306 DOI: 10.1038/s42003-025-07887-2] [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: 04/06/2024] [Accepted: 03/05/2025] [Indexed: 04/20/2025] Open
Abstract
Photoreceptor cell death is a hallmark of age-related macular degeneration. Environmental, lifestyle and genetic risk factors are known contributors to disease progression, whilst at the molecular level, oxidative stress and inflammation are central pathogenetic drivers. However, the spatial and cellular origins of these molecular mechanisms remain unclear. We used spatial transcriptomics to investigate the spatio-temporal gene expression changes in the adult mouse retina in response to photo-oxidative stress. We identify regionally distinct transcriptomes, with higher expression of immunity related genes in the superior retina. Exposure to stress induced expression of genes involved in inflammatory processes, innate immune responses, and cytokine production in a highly localised manner. A distinct region ~800 µm superior from the optic nerve head seems a key driver of these molecular changes. Further, we show highly localised early molecular changes in the superior mouse retina during retinal stress and identify novel genes drivers. We provide evidence of angiogenic changes in response to photo-oxidative stress and suggest additional angiogenic signalling pathways within the retina including VEGF, pleiotrophin and midkine. These new insights into retinal angiogenesis pave the way to identify novel drivers of retinal neovascularisation with an opportunity for therapeutic development.
Collapse
Affiliation(s)
- Ulrike Schumann
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- The Shine-Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- The Save Sight Institute, The University of Sydney, Sydney, Australia.
| | - Lixinyu Liu
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The Shine-Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The Centre for Computational Biomedical Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), The Australian National University, Canberra, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The School of Medicine and Psychology, The Australian National University, Canberra, Australia
| | - Adrian V Cioanca
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The School of Medicine and Psychology, The Australian National University, Canberra, Australia
| | - Artur Shariev
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Michele C Madigan
- The Save Sight Institute, The University of Sydney, Sydney, Australia
- The School of Optometry and Vision Science, The University of New South Wales, Sydney, Australia
| | - Krisztina Valter
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The School of Medicine and Psychology, The Australian National University, Canberra, Australia
| | - Jiayu Wen
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- The Shine-Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- The Centre for Computational Biomedical Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), The Australian National University, Canberra, Australia.
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The Shine-Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- The School of Medicine and Psychology, The Australian National University, Canberra, Australia
| |
Collapse
|
3
|
Shaw EM, Anderson DM, Periasamy R, Schey KL, Curcio CA, Lipinski DM. Porcine Sub-Retinal Pigment Epithelium Deposits: A Model for Dry Age-Related Macular Degeneration With Comparison to Human Drusen. Invest Ophthalmol Vis Sci 2025; 66:18. [PMID: 40048184 PMCID: PMC11895847 DOI: 10.1167/iovs.66.3.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
Purpose Due to the slowly progressing nature of age-related macular degeneration (AMD) and critical differences in ocular anatomy between humans and animals, it has been difficult to model disease progression, hampering the development of novel therapeutics aimed at impacting drusen biogenesis. To determine whether "drusen-in-a-dish" model systems are of utility in screening potential therapeutics aimed at early-intermediate dry AMD, we developed a detailed characterization of the protein, glycoprotein, and lipid composition of sub-retinal pigment epithelium (RPE) deposits grown by monolayers of ex vivo porcine RPE with human drusen in AMD globes. Methods Immunohistochemistry and imaging mass spectrometry (IMS) were performed on 20-week aged monolayers of porcine RPE and human donor globes recovered from an 81-year-old non-transplant donor with confirmed diagnosis of bilateral dry AMD. The presence of major protein, glycoprotein, and lipid species was compared between porcine sub-RPE deposits and human drusen with reference to macular/peripheral eccentricity. Results The protein and glycoprotein composition of porcine sub-RPE deposits closely mimics human drusen identified in donor globes with dry AMD, including the presence of major complement components (C9, CFH, CHI), apolipoproteins (ApoE, ApoJ), extracellular matrix proteins (vitronectin, collagen VI), and calcification (hydroxyapatite). Sub-RPE deposits were additionally rich in long-chain ceramide species (Cer, CerPE, PI), which have only recently been described in human drusen. Conclusions Due to their compositional similarity to human drusen, ex vivo "drusen-in-a-dish" systems represent a potentially robust and cost-effective model for both studying the pathobiology of drusen biogenesis and screening novel therapeutics aimed at limiting drusen formation.
Collapse
Affiliation(s)
- Erika M. Shaw
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - David M. Anderson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Ramesh Periasamy
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Christine A. Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States
| | - Daniel M. Lipinski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Fernandes AG, Poirier AC, Veilleux CC, Melin AD. Contributions and future potential of animal models for geroscience research on sensory systems. GeroScience 2025; 47:61-83. [PMID: 39312151 PMCID: PMC11872837 DOI: 10.1007/s11357-024-01327-5] [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: 04/15/2024] [Accepted: 08/23/2024] [Indexed: 03/04/2025] Open
Abstract
Sensory systems mediate our social interactions, food intake, livelihoods, and other essential daily functions. Age-related decline and disease in sensory systems pose a significant challenge to healthy aging. Research on sensory decline in humans is informative but can often be difficult, subject to sampling bias, and influenced by environmental variation. Study of animal models, including mice, rats, rabbits, pigs, cats, dogs, and non-human primates, plays a complementary role in biomedical research, offering advantages such as controlled conditions and shorter lifespans for longitudinal study. Various species offer different advantages and limitations but have provided key insights in geroscience research. Here we review research on age-related decline and disease in vision, hearing, olfaction, taste, and touch. For each sense, we provide an epidemiological overview of impairment in humans, describing the physiological processes and diseases for each sense. We then discuss contributions made by research on animal models and ideas for future research. We additionally highlight the need for integrative, multimodal research across the senses as well as across disciplines. Long-term studies spanning multiple generations, including on species with longer life spans, are also highly valuable. Overall, integrative studies of appropriate animal models have high translational potential for clinical applications, the development of novel diagnostics, therapies, and medical interventions and future research will continue to close gaps in these areas. Research on animal models to improve understanding of the biology of the aging senses and improve the healthspan and additional research on sensory systems hold special promise for new breakthroughs.
Collapse
Affiliation(s)
- Arthur G Fernandes
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada.
| | - Alice C Poirier
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Carrie C Veilleux
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 4N1, Canada.
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Lenin RR, Koh YH, Zhang Z, Yeo YZ, Parikh BH, Seah I, Wong W, Su X. Dysfunctional Autophagy, Proteostasis, and Mitochondria as a Prelude to Age-Related Macular Degeneration. Int J Mol Sci 2023; 24:ijms24108763. [PMID: 37240109 DOI: 10.3390/ijms24108763] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Retinal pigment epithelial (RPE) cell dysfunction is a key driving force of AMD. RPE cells form a metabolic interface between photoreceptors and choriocapillaris, performing essential functions for retinal homeostasis. Through their multiple functions, RPE cells are constantly exposed to oxidative stress, which leads to the accumulation of damaged proteins, lipids, nucleic acids, and cellular organelles, including mitochondria. As miniature chemical engines of the cell, self-replicating mitochondria are heavily implicated in the aging process through a variety of mechanisms. In the eye, mitochondrial dysfunction is strongly associated with several diseases, including age-related macular degeneration (AMD), which is a leading cause of irreversible vision loss in millions of people globally. Aged mitochondria exhibit decreased rates of oxidative phosphorylation, increased reactive oxygen species (ROS) generation, and increased numbers of mitochondrial DNA mutations. Mitochondrial bioenergetics and autophagy decline during aging because of insufficient free radical scavenger systems, the impairment of DNA repair mechanisms, and reductions in mitochondrial turnover. Recent research has uncovered a much more complex role of mitochondrial function and cytosolic protein translation and proteostasis in AMD pathogenesis. The coupling of autophagy and mitochondrial apoptosis modulates the proteostasis and aging processes. This review aims to summarise and provide a perspective on (i) the current evidence of autophagy, proteostasis, and mitochondrial dysfunction in dry AMD; (ii) current in vitro and in vivo disease models relevant to assessing mitochondrial dysfunction in AMD, and their utility in drug screening; and (iii) ongoing clinical trials targeting mitochondrial dysfunction for AMD therapeutics.
Collapse
Affiliation(s)
- Raji Rajesh Lenin
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
- Department of Medical Research, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Yi Hui Koh
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
| | - Zheting Zhang
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), 11 Mandalay Road, Experimental Medicine Building, Singapore 308232, Singapore
| | - Yan Zhuang Yeo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Bhav Harshad Parikh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Ivan Seah
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
| | - Wendy Wong
- Department of Ophthalmology, National University Hospital (NUH), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
| | - Xinyi Su
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Ophthalmology, National University Hospital (NUH), 1E Kent Ridge Road, NUHS Tower Block Level 7, Singapore 119228, Singapore
- Singapore Eye Research Institute (SERI), The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| |
Collapse
|
6
|
Su N, Hansen U, Plagemann T, Gäher K, Leclaire MD, König J, Höhn A, Grune T, Uhlig CE, Eter N, Heiduschka P. Sub-Retinal Injection of Human Lipofuscin in the Mouse - A Model of "Dry" Age-Related Macular Degeneration? Aging Dis 2023; 14:184-203. [PMID: 36818570 PMCID: PMC9937713 DOI: 10.14336/ad.2022.0626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/26/2022] [Indexed: 11/18/2022] Open
Abstract
Lipofuscin (LF) accumulates during lifetime in the retinal pigment epithelium (RPE) and is thought to play a crucial role in intermediate and late age-related macular degeneration (AMD). In an attemt to simulate aged retina and to study response of retinal microglia and RPE cells to LF, we injected a suspension of LF into the subretinal space of adult mice. LF suspension was obtained from human donor eyes. Subretinal injection of PBS or sham injection served as a control. Eyes were inspected by autofluorescence and optical coherence tomography, by electroretinography and on histological and ultrastructural levels. Levels of cytokine mRNA were determined by quantitative PCR separately in the RPE/choroid complex and in the retina. After injection of LF, microglial cells migrated quickly into the subretinal space to close proximity to RPE cells and phagocytosed LF particles. Retinal function was affected only slightly by LF within the first two weeks. After longer time, RPE cells showed clear signs of melanin loss and degradation. Levels of mRNA of inflammatory cytokines increased sharply after injection of both PBS and LF and were higher in the RPE/choroid complex than in the retina and were slightly higher after LF injection. In conclusion, subretinal injection of LF causes an activation of microglial cells and their migration into subretinal space, enhanced expression of inflammatory cytokines and a gradual degradation of RPE cells. These features are found also in an aging retina, and subretinal injection of LF could be a model for intermediate and late AMD.
Collapse
Affiliation(s)
- Nan Su
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
- Department of Ophthalmology, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, China.
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - Tanja Plagemann
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Karin Gäher
- Institute of Musculoskeletal Medicine, Medical Faculty,
University of Münster, Münster, Germany.
| | - M. Dominik Leclaire
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| | - Jeannette König
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Annika Höhn
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Tilman Grune
- German Institute of Human Nutrition,
Potsdam-Rehbrücke, Germany.
| | - Constantin E. Uhlig
- Cornea Bank Münster, Department of Ophthalmology,
University Medical Center, Münster, Germany.
| | - Nicole Eter
- Department of Ophthalmology, University Medical Center,
Münster, Germany.
| | - Peter Heiduschka
- Research Laboratory, Department of Ophthalmology, University
Medical Center, Münster, Germany.
| |
Collapse
|
7
|
Logan C, Lyzogubov V, Bora N, Bora P. Role of Adiponectin Peptide I (APNp1) in Age-Related Macular Degeneration. Biomolecules 2022; 12:biom12091232. [PMID: 36139070 PMCID: PMC9496372 DOI: 10.3390/biom12091232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is an eye disease that can cause central vision loss, particularly in the elderly population. There are 2 classes of AMD, wet-type and dry-type. Wet-type involves excess angiogenesis around the macula, referred to as choroidal neovascularization (CNV). This can result in leaky vessels, often causing more severe vision loss than dry-type AMD. Adiponectin peptide 1 (APNp1) has been shown to slow the progression of CNV. Here, we used a mouse model and FITC-labeled APNp1 to determine if APNp1 could be delivered effectively as an eye drop. Our experiment revealed that topically applied FITC-APNp1 could reach the macula of the eye, which is crucial for treating wet-type AMD. We also tested delivery of APNp1 via injection of an adeno-associated virus (AAV) vector in a mouse model of CNV. AAV is a harmless virus easy to manipulate and is very often used for protein or peptide deliveries. Results revealed an increase in the expression of APNp1 in the retina and choroid over a 28-day period. Finally, we investigated the mechanism by which APNp1 affects CNV by examining the expression of adiponectin receptor 1 (AdipoR1) and proliferating cell nuclear antigen (PCNA) in the retinal and choroidal tissue of the mouse eyes. AdipoR1 and PCNA were overexpressed in these tissues in mice with laser-induced CNV compared to naïve mice. Based on our data shown here, we think it will enhance our understanding of APNp1 as a therapeutic agent for wet-type AMD and possible treatment alternatives that could be more beneficial for patients.
Collapse
Affiliation(s)
| | | | | | - Puran Bora
- Correspondence: ; Tel.: +1-(501)-352-7191
| |
Collapse
|
8
|
Molins B, Mesquida M, Adan A. Bioengineering approaches for modelling retinal pathologies of the outer blood-retinal barrier. Prog Retin Eye Res 2022:101097. [PMID: 35840488 DOI: 10.1016/j.preteyeres.2022.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/31/2022] [Accepted: 06/29/2022] [Indexed: 11/18/2022]
Abstract
Alterations of the junctional complex of the outer blood-retinal barrier (oBRB), which is integrated by the close interaction of the retinal pigment epithelium, the Bruch's membrane, and the choriocapillaris, contribute to the loss of neuronal signalling and subsequent vision impairment in several retinal inflammatory disorders such as age-related macular degeneration and diabetic retinopathy. Reductionist approaches into the mechanisms that underlie such diseases have been hindered by the absence of adequate in vitro models using human cells to provide the 3D dynamic architecture that enables expression of the in vivo phenotype of the oBRB. Conventional in vitro cell models are based on 2D monolayer cellular cultures, unable to properly recapitulate the complexity of living systems. The main drawbacks of conventional oBRB models also emerge from the cell sourcing, the lack of an appropriate Bruch's membrane analogue, and the lack of choroidal microvasculature with flow. In the last years, the advent of organ-on-a-chip, bioengineering, and stem cell technologies is providing more advanced 3D models with flow, multicellularity, and external control over microenvironmental properties. By incorporating additional biological complexity, organ-on-a-chip devices can mirror physiologically relevant properties of the native tissue while offering additional set ups to model and study disease. In this review we first examine the current understanding of oBRB biology as a functional unit, highlighting the coordinated contribution of the different components to barrier function in health and disease. Then we describe recent advances in the use of pluripotent stem cells-derived retinal cells, Bruch's membrane analogues, and co-culture techniques to recapitulate the oBRB. We finally discuss current advances and challenges of oBRB-on-a-chip technologies for disease modelling.
Collapse
Affiliation(s)
- Blanca Molins
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain.
| | - Marina Mesquida
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Alfredo Adan
- Group of Ocular Inflammation: Clinical and Experimental Studies, Institut d'Investigacions Biomèdiques Agustí Pi I Sunyer (IDIBAPS), C/ Sabino de Arana 1, 08028, Barcelona, Spain; Instituto Clínic de Oftalmología, Hospital Clínic Barcelona, C/ Sabino de Arana 1, 08028, Barcelona, Spain
| |
Collapse
|
9
|
Koster C, van den Hurk KT, ten Brink JB, Lewallen CF, Stanzel BV, Bharti K, Bergen AA. Sodium-Iodate Injection Can Replicate Retinal Degenerative Disease Stages in Pigmented Mice and Rats: Non-Invasive Follow-Up Using OCT and ERG. Int J Mol Sci 2022; 23:ijms23062918. [PMID: 35328338 PMCID: PMC8953416 DOI: 10.3390/ijms23062918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: The lack of suitable animal models for (dry) age-related macular degeneration (AMD) has hampered therapeutic research into the disease, so far. In this study, pigmented rats and mice were systematically injected with various doses of sodium iodate (SI). After injection, the retinal structure and visual function were non-invasively characterized over time to obtain in-depth data on the suitability of these models for studying experimental therapies for retinal degenerative diseases, such as dry AMD. Methods: SI was injected into the tail vein (i.v.) using a series of doses (0–70 mg/kg) in adolescent C57BL/6J mice and Brown Norway rats. The retinal structure and function were assessed non-invasively at baseline (day 1) and at several time points (1–3, 5, and 10-weeks) post-injection by scanning laser ophthalmoscopy (SLO), optical coherence tomography (OCT), and electroretinography (ERG). Results: After the SI injection, retinal degeneration in mice and rats yielded similar results. The lowest dose (10 mg/kg) resulted in non-detectable structural or functional effects. An injection with 20 mg/kg SI did not result in an evident retinal degeneration as judged from the OCT data. In contrast, the ERG responses were temporarily decreased but returned to baseline within two-weeks. Higher doses (30, 40, 50, and 70 mg/kg) resulted in moderate to severe structural RPE and retinal injury and decreased the ERG amplitudes, indicating visual impairment in both mice and rat strains. Conclusions: After the SI injections, we observed dose-dependent structural and functional pathological effects on the retinal pigment epithelium (RPE) and retina in the pigmented mouse and rat strains that were used in this study. Similar effects were observed in both species. In particular, a dose of 30 mg/kg seems to be suitable for future studies on developing experimental therapies. These relatively easily induced non-inherited models may serve as useful tools for evaluating novel therapies for RPE-related retinal degenerations, such as AMD.
Collapse
Affiliation(s)
- Céline Koster
- Department of Human Genetics, Section Ophthalmogenetics, Amsterdam University Medical Centers (AUMC), University of Amsterdam (UvA), Location AMC, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (C.K.); (K.T.v.d.H.); (J.B.t.B.)
| | - Koen T. van den Hurk
- Department of Human Genetics, Section Ophthalmogenetics, Amsterdam University Medical Centers (AUMC), University of Amsterdam (UvA), Location AMC, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (C.K.); (K.T.v.d.H.); (J.B.t.B.)
| | - Jacoline B. ten Brink
- Department of Human Genetics, Section Ophthalmogenetics, Amsterdam University Medical Centers (AUMC), University of Amsterdam (UvA), Location AMC, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (C.K.); (K.T.v.d.H.); (J.B.t.B.)
| | - Colby F. Lewallen
- Georgia Institute of Technology, G.W. Woodruff School of Mechanical Engineering, Atlanta, GA 30332, USA;
| | - Boris V. Stanzel
- Eye Clinic Sulzbach, Knappschaft Hospital Saar, 66280 Sulzbach/Saar, Germany;
- Department of Ophthalmology, University of Bonn, 53113 Bonn, Germany
| | - Kapil Bharti
- Ocular and Stem Cell Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Arthur A. Bergen
- Department of Human Genetics, Section Ophthalmogenetics, Amsterdam University Medical Centers (AUMC), University of Amsterdam (UvA), Location AMC, Meibergdreef, 1105 AZ Amsterdam, The Netherlands; (C.K.); (K.T.v.d.H.); (J.B.t.B.)
- Department of Ophthalmology, AUMC, UvA, Location AMC, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
10
|
Pankova N, Baek DSH, Zhao X, Wang H, Reyad MM, Liang H, Joshi R, Boyd SR. Evolving Patterns of Hyperfluorescent Fundus Autofluorescence Accompany Retinal Atrophy in the Rat and Mimic Atrophic Age-Related Macular Degeneration. Transl Vis Sci Technol 2022; 11:3. [PMID: 35254423 PMCID: PMC8914569 DOI: 10.1167/tvst.11.3.3] [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 Complex two-dimensional (2D) patterns of hyperfluorescent short-wave fundus autofluorescence (FAF) at the border of geographic atrophy (GA) can predict its expansion in patients with late non-exudative “dry” AMD. However, preclinical models do not phenocopy this important feature of disease. We sought to describe the spatiotemporal changes in hyperfluorescent FAF patterns that occur following acute oxidative stress, potentially in association with GA expansion. Methods Sprague Dawley rats (n = 54) received systemic sodium iodate (25–45 mg/kg, n = 90 eyes) or saline (n = 18 eyes) and underwent serial full fundus imaging by confocal scanning laser ophthalmoscopy, including blue FAF and delayed near-infrared analysis. Composite images of the fundus were assembled, and the 2D patterns were described qualitatively and quantitatively. A subset of eyes underwent tissue analysis, and four underwent optical coherence tomography (OCT) imaging. Results Reproducibly changing, complex patterns of hyperfluorescent FAF emerge at the borders of toxin-induced damage; however, in the absence of GA expansion, they percolate inward within the region of retinal pigment epithelium loss, evolving, maturing, and senescing in situ over time. Unexpectedly, the late FAF patterns most closely resemble the diffuse tricking form of clinical disease. A five-stage classification system is presented. Conclusions Longitudinal, full-fundus imaging of outer retinal atrophy in the rat eye identifies evolving, complex patterns of hyperfluorescent FAF that phenocopy aspects of disease. Translational Relevance This work provides a novel tool to assess hyperfluorescent FAF in association with progressive retinal atrophy, a therapeutic target in late AMD.
Collapse
Affiliation(s)
- Natalie Pankova
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - David Sung Hyeon Baek
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Xu Zhao
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Hai Wang
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Matthew-Mina Reyad
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Huiyuan Liang
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Rahul Joshi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Shelley Romayne Boyd
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Wozar F, Seitz I, Reichel F, Fischer MD. Importance of Nonhuman Primates as a Model System for Gene Therapy Development in Ophthalmology. Klin Monbl Augenheilkd 2022; 239:270-274. [PMID: 35189657 DOI: 10.1055/a-1777-5033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Gene therapy is a treatment concept that uses, in most cases, viral vectors to deliver a therapeutic transgene to target cells. Although the idea of gene therapy dates back over 50 years ago, due to the complexity of the treatment concept, it took until the last decade for the responsible agencies like FDA and EMA to recommend the first gene therapy products for clinical use. The development of these therapies relies on molecular engineering of specifically designed vectors and models to test the effectiveness and safety of the treatment. Despite an increasing effort to find effective surrogates, animal models are still irreplaceable in gene therapy development. Rodents are important for exploring pathways and disease mechanisms and identifying potential treatment targets. However, only the primate eye resembles the human eye to a degree where most structures are nearly identical. Some research questions can therefore only be answered using a nonhuman primate (NHP) model. In this review, we want to summarize these key features and highlight the importance of the NHP model for gene therapy development in ophthalmology.
Collapse
Affiliation(s)
- Fabian Wozar
- University Eye Hospital, University Hospital Tübingen Centre of Ophthalmology, Tübingen, Germany
| | - Immanuel Seitz
- University Eye Hospital, University Hospital Tübingen Centre of Ophthalmology, Tübingen, Germany
| | - Felix Reichel
- University Eye Hospital, University Hospital Tübingen Centre of Ophthalmology, Tübingen, Germany
| | - M Dominik Fischer
- University Eye Hospital, University Hospital Tübingen Centre of Ophthalmology, Tübingen, Germany.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom of Great Britain and Northern Ireland.,Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
12
|
Colman K, Andrews RN, Atkins H, Boulineau T, Bradley A, Braendli-Baiocco A, Capobianco R, Caudell D, Cline M, Doi T, Ernst R, van Esch E, Everitt J, Fant P, Gruebbel MM, Mecklenburg L, Miller AD, Nikula KJ, Satake S, Schwartz J, Sharma A, Shimoi A, Sobry C, Taylor I, Vemireddi V, Vidal J, Wood C, Vahle JL. International Harmonization of Nomenclature and Diagnostic Criteria (INHAND): Non-proliferative and Proliferative Lesions of the Non-human Primate ( M. fascicularis). J Toxicol Pathol 2021; 34:1S-182S. [PMID: 34712008 PMCID: PMC8544165 DOI: 10.1293/tox.34.1s] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions Project (www.toxpath.org/inhand.asp) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in most tissues and organs from the nonhuman primate used in nonclinical safety studies. Some of the lesions are illustrated by color photomicrographs. The standardized nomenclature presented in this document is also available electronically on the internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature for lesions in laboratory animals will provide a common language among regulatory and scientific research organizations in different countries and increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
- Karyn Colman
- Novartis Institutes for BioMedical Research, Cambridge, MA,
USA
| | - Rachel N. Andrews
- Wake Forest School of Medicine, Department of Radiation
Oncology, Winston-Salem, NC, USA
| | - Hannah Atkins
- Penn State College of Medicine, Department of Comparative
Medicine, Hershey, PA, USA
| | | | - Alys Bradley
- Charles River Laboratories Edinburgh Ltd., Tranent,
Scotland, UK
| | - Annamaria Braendli-Baiocco
- Roche Pharma Research and Early Development, Pharmaceutical
Sciences, Roche Innovation Center Basel, Switzerland
| | - Raffaella Capobianco
- Janssen Research & Development, a Division of Janssen
Pharmaceutica NV, Beerse, Belgium
| | - David Caudell
- Department of Pathology, Section on Comparative Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mark Cline
- Department of Pathology, Section on Comparative Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Takuya Doi
- LSIM Safety Institute Corporation, Ibaraki, Japan
| | | | | | - Jeffrey Everitt
- Department of Pathology, Duke University School of
Medicine, Durham, NC, USA
| | | | | | | | - Andew D. Miller
- Cornell University College of Veterinary Medicine, Ithaca,
NY, USA
| | | | - Shigeru Satake
- Shin Nippon Biomedical Laboratories, Ltd., Kagoshima and
Tokyo, Japan
| | | | - Alok Sharma
- Covance Laboratories, Inc., Madison, WI, USA
| | | | | | | | | | | | - Charles Wood
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT,
USA
| | | |
Collapse
|
13
|
Alfonsetti M, Castelli V, d’Angelo M, Benedetti E, Allegretti M, Barboni B, Cimini A. Looking for In Vitro Models for Retinal Diseases. Int J Mol Sci 2021; 22:10334. [PMID: 34638674 PMCID: PMC8508697 DOI: 10.3390/ijms221910334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Retina is a layered structure of the eye, composed of different cellular components working together to produce a complex visual output. Because of its important role in visual function, retinal pathologies commonly represent the main causes of visual injury and blindness in the industrialized world. It is important to develop in vitro models of retinal diseases to use them in first screenings before translating in in vivo experiments and clinics. For this reason, it is important to develop bidimensional (2D) models that are more suitable for drug screening and toxicological studies and tridimensional (3D) models, which can replicate physiological conditions, for investigating pathological mechanisms leading to visual loss. This review provides an overview of the most common retinal diseases, relating to in vivo models, with a specific focus on alternative 2D and 3D in vitro models that can replicate the different cellular and matrix components of retinal layers, as well as injury insults that induce retinal disease and loss of the visual function.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
| | | | - Barbara Barboni
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.A.); (V.C.); (M.d.); (E.B.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
14
|
Soundara Pandi SP, Rajendran A, Radha Krishnan S, Anto MJ, Gardiner T, Chakravarthy U, Veerappan M. Characterization of age-related macular degeneration in Indian donor eyes. Indian J Ophthalmol 2021; 69:642-646. [PMID: 33595493 PMCID: PMC7942134 DOI: 10.4103/ijo.ijo_291_20] [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] [Indexed: 11/07/2022] Open
Abstract
Purpose: The purpose of this study was to test the reliability of fundus stereomicroscopy in postmortem eyes to assign severity of age-related macular degeneration (AMD) using the Minnesota grading and confirmation by histology using Alabama and Sarks grading scales and to assess the incidence of AMD pathology in donor eyes from a South Indian population. Methods: Eyes (199) from 153 donors (55–95 years) after obtaining fundus images were processed for histology. Fundus images were graded according to the Minnesota grading system based on drusen size, area of depigmentation, and atrophy. At least one eye from each donor displaying the AMD phenotypes were subjected to histological examination. The fundus grading was correlated with histology and the stages of AMD assigned for early AMD by the Alabama AMD grading system and for both early and advanced AMD by the Sarks classification. Results: Stereoscopic examination of the fundus found that 10 of the 153 donors had features of early AMD and 3 advanced AMD. Following histological examination, one of the early AMD eyes was reclassified as advanced AMD. Early AMD features that were observed on histology included soft drusen (>63 μm), basal laminar deposits, photoreceptor outer segment degeneration, disorganization of retinal pigment epithelium (RPE), Bruch's membrane thickening. Advanced AMD features observed in histology are extensive atrophy of RPE, choroidal neovascularization and disciform scar formation. Conclusion: Identification of either early or advanced AMD using stereomicroscopic assessment (SMA) showed high sensitivity and specificity. However, misclassification between AMD stages can occur when only SMA is used.
Collapse
Affiliation(s)
- Sudha Priya Soundara Pandi
- Department of Stem Cell Biology, Aravind Medical Research Foundation, Dr G. Venkataswamy Eye Research Institute, Madurai, Tamil Nadu, India; Centre for Vision Sciences, Queen's University of Belfast, Belfast; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, U.K
| | - Anand Rajendran
- Vitreo-Retinal Services, Aravind Eye Hospital, Chennai, Tamil Nadu, India
| | - Santhi Radha Krishnan
- Department of Pathology, Aravind Eye Hospital and Postgraduate Institute of Ophthalmology, Madurai, Tamil Nadu, India
| | - Minu Jenifer Anto
- Department of Stem Cell Biology, Aravind Medical Research Foundation, Dr G. Venkataswamy Eye Research Institute, Madurai; Department of Human Genetics and Molecular Biology, Bharathiyar University, Coimbatore, Tamil Nadu, India
| | - Tom Gardiner
- Centre for Vision Sciences, Queen's University of Belfast, Belfast, U.K
| | - Usha Chakravarthy
- Centre for Vision Sciences, Queen's University of Belfast, Belfast, U.K
| | - Muthukkaruppan Veerappan
- Department of Stem Cell Biology, Aravind Medical Research Foundation, Dr G. Venkataswamy Eye Research Institute, Madurai, Tamil Nadu, India
| |
Collapse
|
15
|
Muste JC, Kalur A, Iyer A, Valentim CCS, Singh RP. Photobiomodulation therapy in age-related macular degeneration. Curr Opin Ophthalmol 2021; 32:225-232. [PMID: 33606405 DOI: 10.1097/icu.0000000000000742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To review the available data supporting the use of photobiomodulation therapy (PBT) in the treatment of age-related macular degeneration (AMD). RECENT FINDINGS PBT might be used in treating nonexudative AMD. Limited evidence suggests that exudative AMD may also benefit from PBT. SUMMARY The optimal device would deliver doses of 60 J/cm2 or more with a multiwavelength composition through the pupil over short treatment intervals. Safe upper limits have not been established. More studies are needed to evaluate the efficacy of PBT in treating exudative and nonexudative AMD.
Collapse
Affiliation(s)
- Justin C Muste
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic
| | - Aneesha Kalur
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic
| | - Amogh Iyer
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic
| | | | - Rishi P Singh
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic
- Cole Eye Institute - Retina Service, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Van Hove I, Van Bergen T, Etienne I, Holgado A, Afonina IS, Beyaert R, Feyen JH, Hu TT. IL-33trap-mediated IL-33 neutralization does not exacerbate choroidal neovascularization, but fails to protect against retinal degeneration in a dry age-related macular degeneration model. Exp Eye Res 2021; 207:108608. [PMID: 33930400 DOI: 10.1016/j.exer.2021.108608] [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: 01/22/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 09/30/2022]
Abstract
The progressive and sight-threatening disease, age-related macular degeneration (AMD), is a growing public health concern due to ageing demographics, with the highest unmet medical need for the advanced stage of dry AMD, geographic atrophy. The pathogenesis underlying AMD is driven by a complex interplay of genetic and environmental factors. There is ample evidence that inflammation is strongly involved in AMD development. Interleukin-33 (IL-33) has been proposed to be critically involved in retinal degeneration, but a protective role in eye pathophysiology was also demonstrated. The current study investigated the therapeutic potential of IL-33trap, a novel IL-33-neutralizing biologic, in dry AMD/geographic atrophy and, based on controversial data regarding the protective versus detrimental functions of IL-33 in neovascularization, evaluated the risk of progression to wet AMD by IL-33 neutralization. Repeated intravitreal (IVT) injections of IL-33trap in the mouse laser-induced choroidal neovascularization model did not exacerbate neovascularization or leakage, while it significantly inhibited inflammatory cell infiltration in the retinal pigment epithelium and choroid. On the contrary, IVT treatment with IL-33trap significantly induced retinal inflammation and could not prevent retinopathy induction in the mouse sodium iodate (NaIO3) model. Overall, these data suggest a complex and dichotomous role of IL-33 in eye pathology and indicate that IL-33 neutralization is not able to prevent onset and progression of dry AMD pathogenesis.
Collapse
Affiliation(s)
- Inge Van Hove
- Oxurion NV, Gaston Geenslaan 1, 3001, Heverlee, Belgium.
| | | | | | - Aurora Holgado
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Inna S Afonina
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jean Hm Feyen
- Oxurion NV, Gaston Geenslaan 1, 3001, Heverlee, Belgium
| | | |
Collapse
|
17
|
Arık YB, Buijsman W, Loessberg-Zahl J, Cuartas-Vélez C, Veenstra C, Logtenberg S, Grobbink AM, Bergveld P, Gagliardi G, den Hollander AI, Bosschaart N, van den Berg A, Passier R, van der Meer AD. Microfluidic organ-on-a-chip model of the outer blood-retinal barrier with clinically relevant read-outs for tissue permeability and vascular structure. LAB ON A CHIP 2021; 21:272-283. [PMID: 33346294 DOI: 10.1039/d0lc00639d] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The outer blood-retinal barrier (oBRB) tightly controls the transport processes between the neural tissue of the retina and the underlying blood vessel network. The barrier is formed by the retinal pigment epithelium (RPE), its basal membrane and the underlying choroidal capillary bed. Realistic three-dimensional cell culture based models of the oBRB are needed to study mechanisms and potential treatments of visual disorders such as age-related macular degeneration that result from dysfunction of the barrier tissue. Ideally, such models should also include clinically relevant read-outs to enable translation of experimental findings in the context of pathophysiology. Here, we report a microfluidic organ-on-a-chip model of the oBRB that contains a monolayer of human immortalized RPE and a microvessel of human endothelial cells, separated by a semi-permeable membrane. Confluent monolayers of both cell types were confirmed by fluorescence microscopy. The three-dimensional vascular structures within the chip were imaged by optical coherence tomography: a medical imaging technique, which is routinely applied in ophthalmology. Differences in diameters and vessel density could be readily detected. Upon inducing oxidative stress by treating with hydrogen peroxide (H2O2), a dose dependent increase in barrier permeability was observed by using a dynamic assay for fluorescence tracing, analogous to the clinically used fluorescence angiography. This organ-on-a-chip of the oBRB will allow future studies of complex disease mechanisms and treatments for visual disorders using clinically relevant endpoints in vitro.
Collapse
Affiliation(s)
- Yusuf B Arık
- Applied Stem Cell Technologies, Technical Medical Centre, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Telias M, Nawy S, Kramer RH. Degeneration-Dependent Retinal Remodeling: Looking for the Molecular Trigger. Front Neurosci 2021; 14:618019. [PMID: 33390897 PMCID: PMC7775662 DOI: 10.3389/fnins.2020.618019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/27/2020] [Indexed: 11/23/2022] Open
Abstract
Vision impairment and blindness in humans are most frequently caused by the degeneration and loss of photoreceptor cells in the outer retina, as is the case for age-related macular degeneration, retinitis pigmentosa, retinal detachment and many other diseases. While inner retinal neurons survive degeneration, they undergo fundamental pathophysiological changes, collectively known as “remodeling.” Inner retinal remodeling downstream to photoreceptor death occurs across mammalian retinas from mice to humans, independently of the cause of degeneration. It results in pervasive spontaneous hyperactivity and membrane hyperpermeability in retinal ganglion cells, which funnel all retinal signals to the brain. Remodeling reduces light detection in vision-impaired patients and precludes meaningful vision restoration in blind individuals. In this review, we summarize current hypotheses proposed to explain remodeling and their potential medical significance highlighting the important role played by retinoic acid and its receptor.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States
| | - Scott Nawy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| |
Collapse
|
19
|
Abstract
Normal retina and its cell layers are essential for processing visual stimuli, and loss of its integrity has been documented in many disease processes. The numbers and the axonal processes of retinal ganglion cells are reduced substantially in glaucoma, leading to vision loss and blindness. Similarly, selective loss of photoreceptors in age-related macular degeneration and hereditary retinal dystrophies also results in the compromise of visual acuity. Development of genetically modified mice has led to increased understanding of the pathogenesis of many retinal diseases. Similarly, in this digital era, usage of modalities to quantify the retinal cell loss has grown exponentially leading to a better understanding of the suitability of animal models to study human retinal diseases. These quantification modalities provide valuable quantifiable data in studying pathogenesis and disease progression. This review will discuss the immunohistochemical markers for various retinal cells, available automated tools to quantify retinal cells, and present an example of retinal ganglion cell quantification using HALO image analysis platform. Additionally, we briefly review retinal cell types and subtypes, salient features of retina in various laboratory animal species, and a few of the main disease processes that affect retinal cell numbers in humans.
Collapse
Affiliation(s)
| | - Henry Chen
- 7845Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Ying Hu
- 7845Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Oliver C Turner
- Novartis, 98557Novartis Institutes for BioMedical Research, Preclinical Safety, East Hanover, NJ, USA
| | - Olulanu H Aina
- 426218Janssen Pharmaceutical Company of Johnson & Johnson, Spring House, PA, USA
| |
Collapse
|
20
|
Hurst J, Fietz A, Tsai T, Joachim SC, Schnichels S. Organ Cultures for Retinal Diseases. Front Neurosci 2020; 14:583392. [PMID: 33324149 PMCID: PMC7724035 DOI: 10.3389/fnins.2020.583392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
The successful development of novel therapies is closely linked with understanding the underlying pathomechanisms of a disease. To do so, model systems that reflect human diseases and allow for the evaluation of new therapeutic approaches are needed. Yet, preclinical animal studies often have limited success in predicting human physiology, pathology, and therapeutic responses. Moreover, animal testing is facing increasing ethical and bureaucratic hurdles, while human cell cultures are limited in their ability to represent in vivo situations due to the lack of the tissue microenvironment, which may alter cellular responses. To overcome these struggles, organ cultures, especially those of complex organs such as the retina, can be used to study physiological reactions to substances or stressors. Human and animal organ cultures are now well established and recognized. This mini-review discusses how retinal organ cultures can be used to preserve tissue architecture more realistically and therefore better represent disease-related changes. It also shows how molecular biological, biochemical, and histological techniques can be combined to investigate how anatomical localization may alter cellular responses. Examples for the use of retinal organ cultures, including models to study age-related macular degeneration (AMD), retinitis pigmentosa (RP), central artery occlusion (CRAO), and glaucoma are presented, and their advantages and disadvantages are discussed. We conclude that organ cultures significantly improve our understanding of complex retinal diseases and may advance treatment testing without the need for animal testing.
Collapse
Affiliation(s)
- José Hurst
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Agnes Fietz
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Teresa Tsai
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sven Schnichels
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Singh RK, Nasonkin IO. Limitations and Promise of Retinal Tissue From Human Pluripotent Stem Cells for Developing Therapies of Blindness. Front Cell Neurosci 2020; 14:179. [PMID: 33132839 PMCID: PMC7513806 DOI: 10.3389/fncel.2020.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The self-formation of retinal tissue from pluripotent stem cells generated a tremendous promise for developing new therapies of retinal degenerative diseases, which previously seemed unattainable. Together with use of induced pluripotent stem cells or/and CRISPR-based recombineering the retinal organoid technology provided an avenue for developing models of human retinal degenerative diseases "in a dish" for studying the pathology, delineating the mechanisms and also establishing a platform for large-scale drug screening. At the same time, retinal organoids, highly resembling developing human fetal retinal tissue, are viewed as source of multipotential retinal progenitors, young photoreceptors and just the whole retinal tissue, which may be transplanted into the subretinal space with a goal of replacing patient's degenerated retina with a new retinal "patch." Both approaches (transplantation and modeling/drug screening) were projected when Yoshiki Sasai demonstrated the feasibility of deriving mammalian retinal tissue from pluripotent stem cells, and generated a lot of excitement. With further work and testing of both approaches in vitro and in vivo, a major implicit limitation has become apparent pretty quickly: the absence of the uniform layer of Retinal Pigment Epithelium (RPE) cells, which is normally present in mammalian retina, surrounds photoreceptor layer and develops and matures first. The RPE layer polarize into apical and basal sides during development and establish microvilli on the apical side, interacting with photoreceptors, nurturing photoreceptor outer segments and participating in the visual cycle by recycling 11-trans retinal (bleached pigment) back to 11-cis retinal. Retinal organoids, however, either do not have RPE layer or carry patches of RPE mostly on one side, thus directly exposing most photoreceptors in the developing organoids to neural medium. Recreation of the critical retinal niche between the apical RPE and photoreceptors, where many retinal disease mechanisms originate, is so far unattainable, imposes clear limitations on both modeling/drug screening and transplantation approaches and is a focus of investigation in many labs. Here we dissect different retinal degenerative diseases and analyze how and where retinal organoid technology can contribute the most to developing therapies even with a current limitation and absence of long and functional outer segments, supported by RPE.
Collapse
|
22
|
Baradaran-Rafii A, Sarvari M, Alavi-Moghadam S, Payab M, Goodarzi P, Aghayan HR, Larijani B, Rezaei-Tavirani M, Biglar M, Arjmand B. Cell-based approaches towards treating age-related macular degeneration. Cell Tissue Bank 2020; 21:339-347. [PMID: 32157501 DOI: 10.1007/s10561-020-09826-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/05/2020] [Indexed: 12/19/2022]
Abstract
Age-related macular degeneration as one of the most common causes of worldwide vision loss needs a proper approach for treatment. Therein, cell therapy and regenerative medicine can hold a great promise to be an effective approach. Accordingly, some preclinical and clinical studies were conducted to search around the therapeutic influence of stem cells in Age-related macular degeneration models and subjects. Hereupon, the purpose of the current review is to discuss the mechanisms of age-related macular degeneration, appropriate animal models along with suitable dosage and route of stem cell administration for its treatment.
Collapse
Affiliation(s)
- Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Sarvari
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Biglar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Gong J, Cai H, Noggle S, Paull D, Rizzolo LJ, Del Priore LV, Fields MA. Stem cell-derived retinal pigment epithelium from patients with age-related macular degeneration exhibit reduced metabolism and matrix interactions. Stem Cells Transl Med 2019; 9:364-376. [PMID: 31840941 PMCID: PMC7031648 DOI: 10.1002/sctm.19-0321] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Modeling age‐related macular degeneration (AMD) is challenging, because it is a multifactorial disease. To focus on interactions between the retinal pigment epithelium (RPE) and Bruch's membrane, we generated RPE from AMD patients and used an altered extracellular matrix (ECM) that models aged Bruch's membrane. Induced pluripotent stem cells (iPSCs) were generated from fibroblasts isolated from AMD patients or age‐matched (normal) controls. RPE derived from iPSCs were analyzed by morphology, marker expression, transepithelial electrical resistance (TER), and phagocytosis of rod photoreceptor outer segments. Cell attachment and viability was tested on nitrite‐modified ECM, a typical modification of aged Bruch's membrane. DNA microarrays with hierarchical clustering and analysis of mitochondrial function were used to elucidate possible mechanisms for the observed phenotypes. Differentiated RPE displayed cell‐specific morphology and markers. The TER and phagocytic capacity were similar among iPSC‐derived RPE cultures. However, distinct clusters were found for the transcriptomes of AMD and control iPSC‐derived RPE. AMD‐derived iPSC‐RPE downregulated genes responsible for metabolic‐related pathways and cell attachment. AMD‐derived iPSC‐RPE exhibited reduced mitochondrial respiration and ability to attach and survive on nitrite‐modified ECM. Cells that did attach induced the expression of complement genes. Despite reprogramming, iPSC derived from AMD patients yielded RPE with a transcriptome that is distinct from that of age‐matched controls. When challenged with an AMD‐like modification of Bruch's membrane, AMD‐derived iPSC‐RPE activated the complement immune system.
Collapse
Affiliation(s)
- Jie Gong
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | - Hui Cai
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | -
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Scott Noggle
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Daniel Paull
- The New York Stem Cell Foundation (NYSCF) Research Institute, New York, New York
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| | - Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Elmore SA, Cesta MF, Crabbs TA, Janardhan KS, Krane GA, Mahapatra D, Quist EM, Rinke M, Schaaf GW, Travlos GS, Wang H, Willson CJ, Wolf JC. Proceedings of the 2019 National Toxicology Program Satellite Symposium. Toxicol Pathol 2019; 47:913-953. [PMID: 31645210 PMCID: PMC6911009 DOI: 10.1177/0192623319876929] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The 2019 annual National Toxicology Program Satellite Symposium, entitled "Pathology Potpourri," was held in Raleigh, North Carolina, at the Society of Toxicologic Pathology's 38th annual meeting. The goal of this symposium was to present and discuss challenging diagnostic pathology and/or nomenclature issues. This article presents summaries of the speakers' talks along with select images that were used by the audience for voting and discussion. Various lesions and topics covered during the symposium included aging mouse lesions from various strains, as well as the following lesions from various rat strains: rete testis sperm granuloma/fibrosis, ovarian cystadenocarcinoma, retro-orbital schwannoma, periductal cholangiofibrosis of the liver and pancreas, pars distalis hypertrophy, chronic progressive nephropathy, and renal tubule regeneration. Other cases included polyovular follicles in young beagle dogs and a fungal blood smear contaminant. One series of cases challenged the audience to consider how immunohistochemistry may improve the diagnosis of some tumors. Interesting retinal lesions from a rhesus macaque emphasized the difficulty in determining the etiology of any particular retinal lesion due to the retina's similar response to vascular injury. Finally, a series of lesions from the International Harmonization of Nomenclature and Diagnostic Criteria Non-Rodent Fish Working Group were presented.
Collapse
Affiliation(s)
- Susan A. Elmore
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Mark F. Cesta
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Torrie A Crabbs
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | | | - Gregory A. Krane
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Debabrata Mahapatra
- Integrated Laboratory Systems, Inc., Research Triangle Park, North Carolina, USA
| | - Erin M. Quist
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | | | - George W. Schaaf
- Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Gregory S. Travlos
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Haoan Wang
- West China-Frontier Pharma Tech Co., Ltd., Chengdu, Sichuan, China
| | - Cynthia J. Willson
- Integrated Laboratory Systems, Inc., Research Triangle Park, North Carolina, USA
| | - Jeffrey C. Wolf
- Experimental Pathology Laboratories, Inc., Sterling, Virginia, USA
| |
Collapse
|
25
|
Fields MA, Del Priore LV, Adelman RA, Rizzolo LJ. Interactions of the choroid, Bruch's membrane, retinal pigment epithelium, and neurosensory retina collaborate to form the outer blood-retinal-barrier. Prog Retin Eye Res 2019; 76:100803. [PMID: 31704339 DOI: 10.1016/j.preteyeres.2019.100803] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023]
Abstract
The three interacting components of the outer blood-retinal barrier are the retinal pigment epithelium (RPE), choriocapillaris, and Bruch's membrane, the extracellular matrix that lies between them. Although previously reviewed independently, this review integrates these components into a more wholistic view of the barrier and discusses reconstitution models to explore the interactions among them. After updating our understanding of each component's contribution to barrier function, we discuss recent efforts to examine how the components interact. Recent studies demonstrate that claudin-19 regulates multiple aspects of RPE's barrier function and identifies a barrier function whereby mutations of claudin-19 affect retinal development. Co-culture approaches to reconstitute components of the outer blood-retinal barrier are beginning to reveal two-way interactions between the RPE and choriocapillaris. These interactions affect barrier function and the composition of the intervening Bruch's membrane. Normal or disease models of Bruch's membrane, reconstituted with healthy or diseased RPE, demonstrate adverse effects of diseased matrix on RPE metabolism. A stumbling block for reconstitution studies is the substrates typically used to culture cells are inadequate substitutes for Bruch's membrane. Together with human stem cells, the alternative substrates that have been designed offer an opportunity to engineer second-generation culture models of the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Mark A Fields
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lucian V Del Priore
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Ron A Adelman
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA
| | - Lawrence J Rizzolo
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208061, New Haven, CT, 06520-8061, USA; Department of Surgery, Yale University School of Medicine, PO Box 208062, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
26
|
Rashid K, Akhtar-Schaefer I, Langmann T. Microglia in Retinal Degeneration. Front Immunol 2019; 10:1975. [PMID: 31481963 PMCID: PMC6710350 DOI: 10.3389/fimmu.2019.01975] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022] Open
Abstract
The retina is a complex tissue with multiple cell layers that are highly ordered. Its sophisticated structure makes it especially sensitive to external or internal perturbations that exceed the homeostatic range. This necessitates the continuous surveillance of the retina for the detection of noxious stimuli. This task is mainly performed by microglia cells, the resident tissue macrophages which confer neuroprotection against transient pathophysiological insults. However, under sustained pathological stimuli, microglial inflammatory responses become dysregulated, often worsening disease pathology. In this review, we provide an overview of recent studies that depict microglial responses in diverse retinal pathologies that have degeneration and chronic immune reactions as key pathophysiological components. We also discuss innovative immunomodulatory therapy strategies that dampen the detrimental immunological responses to improve disease outcome.
Collapse
Affiliation(s)
- Khalid Rashid
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Isha Akhtar-Schaefer
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Cologne, Germany
| |
Collapse
|
27
|
Towards A Microbead Occlusion Model of Glaucoma for a Non-Human Primate. Sci Rep 2019; 9:11572. [PMID: 31399621 PMCID: PMC6689098 DOI: 10.1038/s41598-019-48054-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/27/2019] [Indexed: 12/16/2022] Open
Abstract
Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). The disease causes vision loss through the degeneration of retinal ganglion cell neurons and their axons in the optic nerve. Using an inducible model of glaucoma, we elevated IOP in the squirrel monkey (Saimiri boliviensis) using intracameral injection of 35 μm polystyrene microbeads and measured common pathogenic outcomes in the optic projection. A 42% elevation in IOP over 28 weeks reduced anterograde transport of fluorescently-labeled cholera toxin beta from retina to the lateral geniculate nucleus (60% decrease), and to the superior colliculus (49% decrease). Pressure also reduced survival of ganglion cellaxons in the optic nerve by 22%. The same elevation caused upregulation of proteins associated with glaucomatous neurodegeneration in the retina and optic nerve, including complement 1q, interleukin 6, and brain-derived neurotrophic factor. That axon degeneration in the nerve lagged deficits in anterograde transport is consistent with progression in rodent models, while the observed protein changes also occur in tissue from human glaucoma patients. Thus, microbead occlusion in a non-human primate with a visual system similar to our own represents an attractive model to investigate neurodegenerative mechanisms and therapeutic interventions for glaucoma.
Collapse
|
28
|
Park SM, Lee KP, Huh MI, Eom S, Park BU, Kim KH, Park DH, Kim DS, Kim HK. Development of an in vitro 3D choroidal neovascularization model using chemically induced hypoxia through an ultra-thin, free-standing nanofiber membrane. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109964. [PMID: 31499990 DOI: 10.1016/j.msec.2019.109964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 12/18/2022]
Abstract
Choroidal neovascularization (CNV) is the pathological growth of new blood vessels in the sub-retinal pigment epithelial (RPE) space from the choroid through a break in the Bruch's membrane (BM). Despite its importance in studying biological processes and drug discovery, the development of an in vitro CNV model that achieves the physiological structures of native RPE-BM-choroidal capillaries (CC) is still challenging. Here, we develop a novel 3D RPE-BM-CC complex biomimetic system on an ultra-thin, free-standing nanofiber membrane. The thickness of the pristine nanofiber membrane is 2.17 ± 0.81 μm, and the Matrigel-coated nanofiber membrane attains a permeability coefficient of 2.95 ± 0.25 × 10-6 cm/s by 40 kDa FITC-dextran, which is similar to the physiological value of the native BM. On the in vitro 3D RPE-BM-CC complex system, we demonstrate endothelial cell invasion across the 3D RPE-BM-CC complex and the mechanism of the invasion by imposing a hypoxic condition, which is thought to be the major pathological cause of CNV. Furthermore, alleviation of the invasion is achieved by treating with chrysin and anti-VEGF antibody. Thus, the in vitro 3D RPE-BM-CC complex biomimetic system can recapitulate essential features of the pathophysiological environment and be employed for the screening of drug candidates to reduce the number of costly and time-consuming in vivo tests or clinical trials.
Collapse
Affiliation(s)
- Sang Min Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| | - Kyoung-Pil Lee
- Bio-Medical Institute, Kyungpook National University Hospital, 807 Hoguk-ro, Buk-gu, Daegu 41404, South Korea; Department of Ophthalmology, School of Medicine, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, South Korea
| | - Man-Il Huh
- Bio-Medical Institute, Kyungpook National University Hospital, 807 Hoguk-ro, Buk-gu, Daegu 41404, South Korea; Department of Ophthalmology, School of Medicine, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, South Korea
| | - Seongsu Eom
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| | - Byeong-Ung Park
- Bio-Medical Institute, Kyungpook National University Hospital, 807 Hoguk-ro, Buk-gu, Daegu 41404, South Korea; Department of Ophthalmology, School of Medicine, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, South Korea
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, South Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea.
| | - Hong Kyun Kim
- Bio-Medical Institute, Kyungpook National University Hospital, 807 Hoguk-ro, Buk-gu, Daegu 41404, South Korea; Department of Ophthalmology, School of Medicine, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, South Korea.
| |
Collapse
|
29
|
Age-related distribution and potential role of SNCB in topographically different retinal areas of the common marmoset Callithrix jacchus, including the macula. Exp Eye Res 2019; 185:107676. [PMID: 31128101 DOI: 10.1016/j.exer.2019.05.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/08/2019] [Accepted: 05/21/2019] [Indexed: 01/24/2023]
Abstract
Evidence of an age-related increase of β-synuclein (SNCB) in several parts of the visual system including the retina has been reported. SNCB is thought to function as an antagonist of α-synuclein in neurodegenerative diseases, but the exact role of SNCB remains unclear. The presented work studies two different aspects of the onset and role of SNCB in the retinal pigment epithelium (RPE). First, the topographical and intracellular distributions of SNCB in the RPE of non-human marmoset monkey (Callithrix jacchus) were evaluated in paraffin-embedded eyes and RPE whole mounts from different developmental stages (neonatal, adolescent, and adult). Thus, revealed distinct lifetime-related alterations of the topographical and intracellular distributions of SNCB in the primate macula compared to the retinal periphery. Furthermore, the function and influences of SNCB on ARPE-19 cells and primary porcine RPE (ppRPE) cells were characterized by exposing these cells with recombinant SNCB (rSNCB) at different concentrations. Moreover, apoptosis, protein- and mRNA-expression levels of factors of the p53/MDM2 signaling cascade and inflammation- and oxidation-related genes were investigated. The observed dose-depended decreased apoptosis rates together with the PLD2 mediated activation of the p53 pathway promotes senescence-related processes in SNCB exposed common ARPE-19 cells from human origin. Further, increased HMOX1 and NOX4 levels indicate increased oxidative stress and inflammatory responses triggered by SNCB. The obtained differences in the distribution of SNCB in primate RPE together with alterations of cellular functions in rSNCB-exposed RPE cells (e.g., ARPE-19, ppRPE) support SNCB-related effects like inflammatory response and stress-related properties on RPE over lifetime. The possible functional relevance of SNCB in physiological aging converting into a pathophysiological condition should be investigated in further studies.
Collapse
|
30
|
Lai S, Wei Y, Wu Q, Zhou K, Liu T, Zhang Y, Jiang N, Xiao W, Chen J, Liu Q, Yu Y. Liposomes for effective drug delivery to the ocular posterior chamber. J Nanobiotechnology 2019; 17:64. [PMID: 31084611 PMCID: PMC6515668 DOI: 10.1186/s12951-019-0498-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/04/2019] [Indexed: 01/03/2023] Open
Abstract
Background Age-related macular degeneration (AMD) is a leading cause of severe visual deficits and blindness. Meanwhile, there is convincing evidence implicating oxidative stress, inflammation, and neovascularization in the onset and progression of AMD. Several studies have identified berberine hydrochloride and chrysophanol as potential treatments for ocular diseases based on their antioxidative, antiangiogenic, and anti-inflammatory effects. Unfortunately, their poor stability and bioavailability have limited their application. In order to overcome these disadvantages, we prepared a compound liposome system that can entrap these drugs simultaneously using the third polyamidoamine dendrimer (PAMAM G3.0) as a carrier. Results PAMAM G3.0-coated compound liposomes exhibited appreciable cellular permeability in human corneal epithelial cells and enhanced bio-adhesion on rabbit corneal epithelium. Moreover, coated liposomes greatly improved BBH bioavailability. Further, coated liposomes exhibited obviously protective effects in human retinal pigment epithelial cells and rat retinas after photooxidative retinal injury. Finally, administration of P-CBLs showed no sign of side effects on ocular surface structure in rabbits model. Conclusions The PAMAM G3.0-liposome system thus displayed a potential use for treating various ocular diseases. Electronic supplementary material The online version of this article (10.1186/s12951-019-0498-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sisi Lai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yanyan Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Quanwu Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Kang Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Tuo Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yingfeng Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Ning Jiang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Wen Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Junjie Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Qiuhong Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yang Yu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
31
|
Shah M, Cabrera-Ghayouri S, Christie LA, Held KS, Viswanath V. Translational Preclinical Pharmacologic Disease Models for Ophthalmic Drug Development. Pharm Res 2019; 36:58. [PMID: 30805711 PMCID: PMC6394514 DOI: 10.1007/s11095-019-2588-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
Preclinical models of human diseases are critical to our understanding of disease etiology, pathology, and progression and enable the development of effective treatments. An ideal model of human disease should capture anatomical features and pathophysiological mechanisms, mimic the progression pattern, and should be amenable to evaluating translational endpoints and treatment approaches. Preclinical animal models have been developed for a variety of human ophthalmological diseases to mirror disease mechanisms, location of the affected region in the eye and severity. These models offer clues to aid in our fundamental understanding of disease pathogenesis and enable progression of new therapies to clinical development by providing an opportunity to gain proof of concept (POC). Here, we review preclinical animal models associated with development of new therapies for diseases of the ocular surface, glaucoma, presbyopia, and retinal diseases, including diabetic retinopathy and age-related macular degeneration (AMD). We have focused on summarizing the models critical to new drug development and described the translational features of the models that contributed to our understanding of disease pathogenesis and establishment of preclinical POC.
Collapse
Affiliation(s)
- Mihir Shah
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Sara Cabrera-Ghayouri
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Lori-Ann Christie
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Katherine S Held
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Veena Viswanath
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA.
| |
Collapse
|
32
|
König S, Hadrian K, Schlatt S, Wistuba J, Thanos S, Böhm M. Topographic protein profiling of the age-related proteome in the retinal pigment epithelium of Callithrix jacchus with respect to macular degeneration. J Proteomics 2019; 191:1-15. [DOI: 10.1016/j.jprot.2018.05.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/12/2018] [Accepted: 05/28/2018] [Indexed: 12/27/2022]
|
33
|
Wang Y, Grenell A, Zhong F, Yam M, Hauer A, Gregor E, Zhu S, Lohner D, Zhu J, Du J. Metabolic signature of the aging eye in mice. Neurobiol Aging 2018; 71:223-233. [PMID: 30172221 DOI: 10.1016/j.neurobiolaging.2018.07.024] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/28/2018] [Accepted: 07/29/2018] [Indexed: 12/29/2022]
Abstract
Aging is a major risk factor for age-related ocular diseases including age-related macular degeneration in the retina and retinal pigment epithelium (RPE), cataracts in the lens, glaucoma in the optic nerve, and dry eye syndrome in the cornea. We used targeted metabolomics to analyze metabolites from young (6 weeks) and old (73 weeks) eyes in C57 BL6/J mice. Old mice had diminished electroretinogram responses and decreased number of photoreceptors in their retinas. Among the 297 detected metabolites, 45-114 metabolites are significantly altered in aged eye tissues, mostly in the neuronal tissues (retina and optic nerve) and less in cornea, RPE/choroid, and lens. We noted that changes of metabolites in mitochondrial metabolism and glucose metabolism are common features in the aged retina, RPE/choroid, and optic nerve. The aging retina, cornea, and optic nerve also share similar changes in Nicotinamide adenine dinucleotide (NAD), 1-methylnicotinamides, 3-methylhistidine, and other methylated metabolites. Metabolites in taurine metabolism are strikingly influenced by aging in the cornea and lens. In conclusion, the aging eye has both common and tissue-specific metabolic signatures. These changes may be attributed to dysregulated mitochondrial metabolism, reprogrammed glucose metabolism and impaired methylation in the aging eye. Our findings provide biochemical insights into the mechanisms of age-related ocular changes.
Collapse
Affiliation(s)
- Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Allison Grenell
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Fanyi Zhong
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, USA
| | - Michelle Yam
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Allison Hauer
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Elizabeth Gregor
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Siyan Zhu
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Daniel Lohner
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Jiangjiang Zhu
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, USA
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, WV, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
34
|
Ramos MF, Baker J, Atzpodien EA, Bach U, Brassard J, Cartwright J, Farman C, Fishman C, Jacobsen M, Junker-Walker U, Kuper F, Moreno MCR, Rittinghausen S, Schafer K, Tanaka K, Teixeira L, Yoshizawa K, Zhang H. Nonproliferative and Proliferative Lesions of the Ratand Mouse Special Sense Organs(Ocular [eye and glands], Olfactory and Otic). J Toxicol Pathol 2018; 31:97S-214S. [PMID: 30158741 PMCID: PMC6108092 DOI: 10.1293/tox.31.97s] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
| | - Julia Baker
- Member of eye subgroup
- Charles River Laboratories, Inc., Frederick, MD, USA
| | | | - Ute Bach
- Member of eye subgroup
- Bayer AG, Wuppertal, Germany
| | | | | | | | - Cindy Fishman
- Member of eye subgroup
- Member of glands of the eye subgroup
- GlaxoSmithKline, King of Prussia, PA, USA
| | | | | | - Frieke Kuper
- Member of olfactory subgroup
- Retired; formerly The Netherlands Organization for Applied
Scientific Research (TNO), Zeist, the Netherlands
| | | | | | - Ken Schafer
- Member of eye subgroup
- Member of otic subgroup
- Vet Path Services, Inc., Mason, OH, USA
| | - Kohji Tanaka
- Member of eye subgroup
- Nippon Boehringer Ingelheim, Japan
| | | | | | | |
Collapse
|
35
|
Malek G, Busik J, Grant MB, Choudhary M. Models of retinal diseases and their applicability in drug discovery. Expert Opin Drug Discov 2018; 13:359-377. [PMID: 29382242 DOI: 10.1080/17460441.2018.1430136] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The impact of vision debilitating diseases is a global public health concern, which will continue until effective preventative and management protocols are developed. Two retinal diseases responsible for the majority of vision loss in the working age adults and elderly populations are diabetic retinopathy (DR) and age-related macular degeneration (AMD), respectively. Model systems, which recapitulate aspects of human pathology, are valid experimental modalities that have contributed to the identification of signaling pathways involved in disease development and consequently potential therapies. Areas covered: The pathology of DR and AMD, which serve as the basis for designing appropriate models of disease, is discussed. The authors also review in vitro and in vivo models of DR and AMD and evaluate the utility of these models in exploratory and pre-clinical studies. Expert opinion: The complex nature of non-Mendelian diseases such as DR and AMD has made identification of effective therapeutic treatments challenging. However, the authors believe that while in vivo models are often criticized for not being a 'perfect' recapitulation of disease, they have been valuable experimentally when used with consideration of the strengths and limitations of the experimental model selected and have a place in the drug discovery process.
Collapse
Affiliation(s)
- Goldis Malek
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA.,b Department of Pathology , Duke University School of Medicine , Durham , NC , USA
| | - Julia Busik
- c Department of Physiology , Michigan State University , East Lansing , MI , USA
| | - Maria B Grant
- d Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , Al , USA
| | - Mayur Choudhary
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA
| |
Collapse
|
36
|
Abstract
The eye is susceptible to adverse toxic effects by direct application, inadvertent ocular contact, or systemic exposure to chemicals or their metabolites. Although the albino rat is a less than ideal model for ocular toxicity studies, it has gained popularity for specific applications and may be the first species in which the ocular toxicity of a systemically administered xenobiotic becomes evident. This chapter reviews the embryology, anatomy, and physiology of the eye and associated glands and describes common nonneoplastic and neoplastic lesions encountered in laboratory rats.
Collapse
|
37
|
Joyal JS, Gantner ML, Smith LEH. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog Retin Eye Res 2017; 64:131-156. [PMID: 29175509 DOI: 10.1016/j.preteyeres.2017.11.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Canada.
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, United States
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston MA 02115, United States.
| |
Collapse
|
38
|
Quantitative analysis of retinal perfusion in mice using optical coherence tomography angiography. Exp Eye Res 2017; 164:151-156. [DOI: 10.1016/j.exer.2017.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/29/2017] [Accepted: 09/05/2017] [Indexed: 12/26/2022]
|
39
|
Rudolf M, Mir Mohi Sefat A, Miura Y, Tura A, Raasch W, Ranjbar M, Grisanti S, Aherrahrou Z, Wagner A, Messinger JD, Garber DW, Anantharamaiah GM, Curcio CA. ApoA-I Mimetic Peptide 4F Reduces Age-Related Lipid Deposition in Murine Bruch's Membrane and Causes Its Structural Remodeling. Curr Eye Res 2017; 43:135-146. [PMID: 28972410 DOI: 10.1080/02713683.2017.1370118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Accumulation of lipoprotein-derived lipids including esterified and unesterified cholesterol in Bruch's membrane of human eyes is a major age-related change involved in initiating and sustaining soft drusen in age-related macular degeneration (AMD). The apolipoprotein (apo) A-I mimetic peptide 4F is a small anti-inflammatory and anti-atherogenic agent, and potent modifier of plasma membranes. We evaluated the effect of intravitreally-injected 4F on murine Bruch's membrane. METHODS We tested single intravitreal injections of 4F doses (0.6 µg, 1.2 µg, 2.4 µg, and placebo scrambled peptide) in ApoEnull mice ≥10 months of age. After 30 days, mice were euthanized. Eyes were processed for either direct immunofluorescence detection of esterified cholesterol (EC) in Bruch's membrane whole mounts via a perfringolysin O-based marker linked to green fluorescent protein or by transmission electron microscopic visualization of Bruch's membrane integrity. Fluorescein isothiocyanate-conjugated 4F was traced after injection. RESULTS All injected eyes showed a dose-dependent reduction of Bruch's membrane EC with a concomitant ultrastructural improvement compared to placebo treated eyes. At a 2.4 µg dose of 4F, EC was reduced on average by ~60% and Bruch's membrane returned to a regular pentalaminar structure and thickness. Tracer studies confirmed that injected 4F reached intraocular targets. CONCLUSION We demonstrated a highly effective pharmacological reduction of EC and restoration of Bruch's membrane ultrastructure. The apoA-I mimetic peptide 4F is a novel way to treat a critical AMD disease process and thus represents a new candidate for treating the underlying cause of AMD.
Collapse
Affiliation(s)
- Martin Rudolf
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | | | - Yoko Miura
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Aysegül Tura
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Walter Raasch
- b Department of Experimental and Clinical Pharmacology and Toxicology , University of Lübeck , Lübeck , Germany
| | - Mahdy Ranjbar
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany.,c Laboratory for Angiogenesis & Ocular Cell Transplantation , University of Lübeck , Lübeck , Germany
| | | | - Zouhair Aherrahrou
- d Institute of Integrative and Experimental Genomics , University of Lübeck , Lübeck , Germany
| | - Anna Wagner
- a Department of Ophthalmology , University of Lübeck , Lübeck , Germany
| | - Jeffrey D Messinger
- e Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , AL , USA
| | - David W Garber
- f Atherosclerosis Research Unit , University of Alabama at Birmingham , Birmingham , AL , USA
| | - G M Anantharamaiah
- f Atherosclerosis Research Unit , University of Alabama at Birmingham , Birmingham , AL , USA.,g Department of Medicine, Biochemistry and Molecular Genetics , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Christine A Curcio
- e Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , AL , USA
| |
Collapse
|
40
|
Assessment of Safety and Functional Efficacy of Stem Cell-Based Therapeutic Approaches Using Retinal Degenerative Animal Models. Stem Cells Int 2017; 2017:9428176. [PMID: 28928775 PMCID: PMC5592015 DOI: 10.1155/2017/9428176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
Dysfunction and death of retinal pigment epithelium (RPE) and or photoreceptors can lead to irreversible vision loss. The eye represents an ideal microenvironment for stem cell-based therapy. It is considered an “immune privileged” site, and the number of cells needed for therapy is relatively low for the area of focused vision (macula). Further, surgical placement of stem cell-derived grafts (RPE, retinal progenitors, and photoreceptor precursors) into the vitreous cavity or subretinal space has been well established. For preclinical tests, assessments of stem cell-derived graft survival and functionality are conducted in animal models by various noninvasive approaches and imaging modalities. In vivo experiments conducted in animal models based on replacing photoreceptors and/or RPE cells have shown survival and functionality of the transplanted cells, rescue of the host retina, and improvement of visual function. Based on the positive results obtained from these animal experiments, human clinical trials are being initiated. Despite such progress in stem cell research, ethical, regulatory, safety, and technical difficulties still remain a challenge for the transformation of this technique into a standard clinical approach. In this review, the current status of preclinical safety and efficacy studies for retinal cell replacement therapies conducted in animal models will be discussed.
Collapse
|
41
|
Prasad T, Zhu P, Verma A, Chakrabarty P, Rosario AM, Golde TE, Li Q. Amyloid β peptides overexpression in retinal pigment epithelial cells via AAV-mediated gene transfer mimics AMD-like pathology in mice. Sci Rep 2017; 7:3222. [PMID: 28607377 PMCID: PMC5468329 DOI: 10.1038/s41598-017-03397-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/27/2017] [Indexed: 01/24/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive retinal neurodegenerative disorder characterized by extracellular deposits known as drusen. A major constituent of drusen deposits are Alzheimer disease-associated amyloid β (Aβ) peptides. To understand the etiology of Aβ proteostasis in AMD, we delivered recombinant adeno-associated virus (AAV) encoding Aβ42 and Aβ40 peptides fused to BRI2 protein by intraocular injection in C57BL/6J mice. Endogenous protease cleavage of such constructs leads to production of secreted Aβ42 and Aβ40 respectively. We demonstrate that overexpression of secreted Aβ40 or Aβ42 resulted in dramatic induction of drusen-like deposits by 2 months' post-injection. These drusen-like deposits were immunopositive for Aβ and complement proteins but did not stain for conventional amyloid dyes, such as Thioflavin S. Both injected cohorts showed gliosis and degenerative changes, though ERG responses were minimally affected. Intriguingly, simultaneous overexpression of BRI-Aβ40 or BRI-Aβ42 together resulted in dose-dependent and cumulative changes reminiscent of AMD type pathology - drusen-like deposits, severe reduction in ERG responses, photoreceptor cell loss and gliosis. Here, we have established a physiological model of Aβ containing deposits in wild-type mice that recapitulates major retinal pathophysiological features of AMD and will be instrumental in mechanistic understanding and development of therapeutic strategies against AMD.
Collapse
Affiliation(s)
- Tuhina Prasad
- Department of Ophthalmology, University of Florida, Gainesville, Florida, 32610, USA
| | - Ping Zhu
- Department of Ophthalmology, University of Florida, Gainesville, Florida, 32610, USA
| | - Amrisha Verma
- Department of Ophthalmology, University of Florida, Gainesville, Florida, 32610, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative disease and McKnight Brain Institute, University of Florida, Gainesville, Florida, 32610, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative disease and McKnight Brain Institute, University of Florida, Gainesville, Florida, 32610, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative disease and McKnight Brain Institute, University of Florida, Gainesville, Florida, 32610, USA
| | - Qiuhong Li
- Department of Ophthalmology, University of Florida, Gainesville, Florida, 32610, USA.
| |
Collapse
|
42
|
Nommiste B, Fynes K, Tovell VE, Ramsden C, da Cruz L, Coffey P. Stem cell-derived retinal pigment epithelium transplantation for treatment of retinal disease. PROGRESS IN BRAIN RESEARCH 2017; 231:225-244. [PMID: 28554398 DOI: 10.1016/bs.pbr.2017.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Age-related macular degeneration remains the most common cause of blindness in the western world, severely comprising patients' and carers' quality of life and presenting a great cost to the healthcare system. As the disease progresses, the retinal pigmented epithelium (RPE) layer at the back of the eye degenerates, contributing to a series of events resulting in visual impairment. The easy accessibility of the eye has allowed for in-depth study of disease progression in patients, while in vivo studies have facilitated investigations into healthy and diseased RPE. Consequently, a number of research groups are examining different approaches for the replacement of RPE cells in age-related macular degeneration (AMD) patients. This chapter examines some of these initial proof-of-principle studies and goes on to review the use of pluripotent stem cells as a source for RPE replacement in a number of current AMD clinical trials. Finally, we consider just some of the regulatory and manufacturing challenges presented in taking a promising AMD treatment from the research bench into clinical trials in patients, and how to mitigate potential risks early in process development.
Collapse
Affiliation(s)
| | - Kate Fynes
- Institute of Ophthalmology, London, United Kingdom
| | | | - Conor Ramsden
- Institute of Ophthalmology, London, United Kingdom; NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Lyndon da Cruz
- Institute of Ophthalmology, London, United Kingdom; NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Peter Coffey
- Institute of Ophthalmology, London, United Kingdom; NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States.
| |
Collapse
|
43
|
Abstract
The innate immune system is activated in a number of degenerative and inflammatory retinal disorders such as age-related macular degeneration (AMD). Retinal microglia, choroidal macrophages, and recruited monocytes, collectively termed 'retinal mononuclear phagocytes', are critical determinants of ocular disease outcome. Many publications have described the presence of these cells in mouse models for retinal disease; however, only limited aspects of their behavior have been uncovered, and these have only been uncovered using a single detection method. The workflow presented here describes a comprehensive analysis strategy that allows characterization of retinal mononuclear phagocytes in vivo and in situ. We present standardized working steps for scanning laser ophthalmoscopy of microglia from MacGreen reporter mice (mice expressing the macrophage colony-stimulating factor receptor GFP transgene throughout the mononuclear phagocyte system), quantitative analysis of Iba1-stained retinal sections and flat mounts, CD11b-based retinal flow cytometry, and qRT-PCR analysis of key microglia markers. The protocol can be completed within 3 d, and we present data from retinas treated with laser-induced choroidal neovascularization (CNV), bright white-light exposure, and Fam161a-associated inherited retinal degeneration. The assays can be applied to any of the existing mouse models for retinal disorders and may be valuable for documenting immune responses in studies for immunomodulatory therapies.
Collapse
|
44
|
Jones MK, Lu B, Girman S, Wang S. Cell-based therapeutic strategies for replacement and preservation in retinal degenerative diseases. Prog Retin Eye Res 2017; 58:1-27. [PMID: 28111323 PMCID: PMC5441967 DOI: 10.1016/j.preteyeres.2017.01.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/08/2017] [Accepted: 01/17/2017] [Indexed: 12/13/2022]
Abstract
Cell-based therapeutics offer diverse options for treating retinal degenerative diseases, such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP). AMD is characterized by both genetic and environmental risks factors, whereas RP is mainly a monogenic disorder. Though treatments exist for some patients with neovascular AMD, a majority of retinal degenerative patients have no effective therapeutics, thus indicating a need for universal therapies to target diverse patient populations. Two main cell-based mechanistic approaches are being tested in clinical trials. Replacement therapies utilize cell-derived retinal pigment epithelial (RPE) cells to supplant lost or defective host RPE cells. These cells are similar in morphology and function to native RPE cells and can potentially supplant the responsibilities of RPE in vivo. Preservation therapies utilize supportive cells to aid in visual function and photoreceptor preservation partially by neurotrophic mechanisms. The goal of preservation strategies is to halt or slow the progression of disease and maintain remaining visual function. A number of clinical trials are testing the safety of replacement and preservation cell therapies in patients; however, measures of efficacy will need to be further evaluated. In addition, a number of prevailing concerns with regards to the immune-related response, longevity, and functionality of the grafted cells will need to be addressed in future trials. This review will summarize the current status of cell-based preclinical and clinical studies with a focus on replacement and preservation strategies and the obstacles that remain regarding these types of treatments.
Collapse
Affiliation(s)
- Melissa K Jones
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Bin Lu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Sergey Girman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Shaomei Wang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Ave., Los Angeles, CA 90095, USA.
| |
Collapse
|
45
|
Manthey AL, Chiu K, So KF. Effects of Lycium barbarum on the Visual System. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 135:1-27. [PMID: 28807155 DOI: 10.1016/bs.irn.2017.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lycium barbarum (wolfberry, gogi berry, gouqizi, ) is one of the most widely used Chinese herbal medicines (CHMs) and is also one of the most scientifically studied. Indeed, the polysaccharide component of this berry (LBP) has been shown to have antioxidant, antiinflammatory, antiexcitotoxic, and antiapoptotic properties. These properties make it a particularly useful treatment option for the ocular environment. Although there are a handful of studies investigating the use of LBP to treat diseases affecting the lens, the vast majority of the published literature investigating LBP in the visual system focus on the retina. In this chapter, we have described what is currently understood concerning the effects of LBP treatment on various retinal diseases, including glaucoma, ischemia/reperfusion, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. We then describe the functions attributed to LBP using other cellular contexts to elucidate the full mechanisms this CHM utilizes in the retina. By making connections between what is known about the function of LBP in a variety of tissues and its function as a therapy for retinal degenerative diseases, we hope to further emphasize the continued use of this CHM in clinical medicine in addition to providing a platform for additional study.
Collapse
Affiliation(s)
| | - Kin Chiu
- The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China.
| | - Kwok-Fai So
- The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration and Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China; Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
46
|
Combes RD, Shah AB. The use of in vivo, ex vivo, in vitro, computational models and volunteer studies in vision research and therapy, and their contribution to the Three Rs. Altern Lab Anim 2017; 44:187-238. [PMID: 27494623 DOI: 10.1177/026119291604400302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Much is known about mammalian vision, and considerable progress has been achieved in treating many vision disorders, especially those due to changes in the eye, by using various therapeutic methods, including stem cell and gene therapy. While cells and tissues from the main parts of the eye and the visual cortex (VC) can be maintained in culture, and many computer models exist, the current non-animal approaches are severely limiting in the study of visual perception and retinotopic imaging. Some of the early studies with cats and non-human primates (NHPs) are controversial for animal welfare reasons and are of questionable clinical relevance, particularly with respect to the treatment of amblyopia. More recently, the UK Home Office records have shown that attention is now more focused on rodents, especially the mouse. This is likely to be due to the perceived need for genetically-altered animals, rather than to knowledge of the similarities and differences of vision in cats, NHPs and rodents, and the fact that the same techniques can be used for all of the species. We discuss the advantages and limitations of animal and non-animal methods for vision research, and assess their relative contributions to basic knowledge and clinical practice, as well as outlining the opportunities they offer for implementing the principles of the Three Rs (Replacement, Reduction and Refinement).
Collapse
Affiliation(s)
| | - Atul B Shah
- Ophthalmic Surgeon, National Eye Registry Ltd, Leicester, UK
| |
Collapse
|
47
|
Lyzogubov VV, Bora PS, Wu X, Horn LE, de Roque R, Rudolf XV, Atkinson JP, Bora NS. The Complement Regulatory Protein CD46 Deficient Mouse Spontaneously Develops Dry-Type Age-Related Macular Degeneration-Like Phenotype. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2088-2104. [PMID: 27295359 PMCID: PMC4973660 DOI: 10.1016/j.ajpath.2016.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 11/20/2022]
Abstract
In the mouse, membrane cofactor protein (CD46), a key regulator of the alternative pathway of the complement system, is only expressed in the eye and on the inner acrosomal membrane of spermatozoa. We noted that although Cd46(-/-) mice have normal systemic alternative pathway activating ability, lack of CD46 leads to dysregulated complement activation in the eye, as evidenced by increased deposition of C5b-9 in the retinal pigment epithelium (RPE) and choroid. A knockout of CD46 induced the following cardinal features of human dry age-related macular degeneration (AMD) in 12-month-old male and female mice: accumulation of autofluorescent material in and hypertrophy of the RPE, dense deposits in and thickening of Bruch's membrane, loss of photoreceptors, cells in subretinal space, and a reduction of choroidal vessels. Collectively, our results demonstrate spontaneous age-related degenerative changes in the retina, RPE, and choroid of Cd46(-/-) mice that are consistent with human dry AMD. These findings provide the exciting possibility of using Cd46(-/-) mice as a convenient and reliable animal model for dry AMD. Having such a relatively straight-forward model for dry AMD should provide valuable insights into pathogenesis and a test model system for novel drug targets. More important, tissue-specific expression of CD46 gives the Cd46(-/-) mouse model of dry AMD a unique advantage over other mouse models using knockout strains.
Collapse
Affiliation(s)
- Valeriy V Lyzogubov
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Puran S Bora
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Xiaobo Wu
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Leah E Horn
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Ryan de Roque
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas; University of Arkansas for Medical Sciences, College of Medicine, Little Rock, Arkansas
| | - Xeniya V Rudolf
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Nalini S Bora
- Department of Ophthalmology, Jones Eye Institute, Pat and Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
48
|
Abstract
Mouse models are powerful tools for the study of ocular diseases. Alterations in the morphology and function of the retinal pigment epithelium (RPE) are common features shared by many ocular disorders. We report a detailed protocol to collect, seed, culture and characterize RPE cells from mice. We describe a reproducible method that we previously developed to collect and culture murine RPE cells on Transwells as functional polarized monolayers. The collection of RPE cells takes ∼3 h, and the cultures mimic in vivo RPE cell features within 1 week. This protocol also describes methods to characterize the cells on Transwells within 1-2 weeks by transmission and scanning electron microscopy (TEM and SEM, respectively), immunostaining of vibratome sections and flat mounts, and measurement of transepithelial electrical resistance. The RPE cell cultures are suitable to study the biology of the RPE from wild-type and genetically modified strains of mice between the ages of 10 d and 12 months. The RPE cells can also be manipulated to investigate molecular mechanisms underlying the RPE pathology in the numerous mouse models of ocular disorders. Furthermore, modeling the RPE pathology in vitro represents a new approach to testing drugs that will help accelerate the development of therapies for vision-threatening disorders such as macular degeneration (MD).
Collapse
|
49
|
Shafaie S, Hutter V, Cook MT, Brown MB, Chau DYS. In Vitro Cell Models for Ophthalmic Drug Development Applications. Biores Open Access 2016; 5:94-108. [PMID: 27158563 PMCID: PMC4845647 DOI: 10.1089/biores.2016.0008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tissue engineering is a rapidly expanding field that aims to establish feasible techniques to fabricate biologically equivalent replacements for diseased and damaged tissues/organs. Emerging from this prospect is the development of in vitro representations of organs for drug toxicity assessment. Due to the ever-increasing interest in ocular drug delivery as a route for administration as well as the rise of new ophthalmic therapeutics, there is a demand for physiologically accurate in vitro models of the eye to assess drug delivery and safety of new ocular medicines. This review summarizes current existing ocular models and highlights the important factors and limitations that need to be considered during their use.
Collapse
Affiliation(s)
- Sara Shafaie
- Department of Pharmacy, Pharmacology, and Postgraduate Medicine, The Research Center in Topical Drug Delivery and Toxicology, School of Life and Medical Sciences, University of Hertfordshire , Hertfordshire, United Kingdom
| | - Victoria Hutter
- Department of Pharmacy, Pharmacology, and Postgraduate Medicine, The Research Center in Topical Drug Delivery and Toxicology, School of Life and Medical Sciences, University of Hertfordshire , Hertfordshire, United Kingdom
| | - Michael T Cook
- Department of Pharmacy, Pharmacology, and Postgraduate Medicine, The Research Center in Topical Drug Delivery and Toxicology, School of Life and Medical Sciences, University of Hertfordshire , Hertfordshire, United Kingdom
| | - Marc B Brown
- Department of Pharmacy, Pharmacology, and Postgraduate Medicine, The Research Center in Topical Drug Delivery and Toxicology, School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire, United Kingdom.; MedPharm Ltd., Guildford, Surrey, United Kingdom
| | - David Y S Chau
- Department of Pharmacy, Pharmacology, and Postgraduate Medicine, The Research Center in Topical Drug Delivery and Toxicology, School of Life and Medical Sciences, University of Hertfordshire , Hertfordshire, United Kingdom
| |
Collapse
|
50
|
Forest DL, Johnson LV, Clegg DO. Cellular models and therapies for age-related macular degeneration. Dis Model Mech 2016; 8:421-7. [PMID: 26035859 PMCID: PMC4415892 DOI: 10.1242/dmm.017236] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Age-related macular degeneration (AMD) is a complex neurodegenerative visual disorder that causes profound physical and psychosocial effects. Visual impairment in AMD is caused by the loss of retinal pigmented epithelium (RPE) cells and the light-sensitive photoreceptor cells that they support. There is currently no effective treatment for the most common form of this disease (dry AMD). A new approach to treating AMD involves the transplantation of RPE cells derived from either human embryonic or induced pluripotent stem cells. Multiple clinical trials are being initiated using a variety of cell therapies. Although many animal models are available for AMD research, most do not recapitulate all aspects of the disease, hampering progress. However, the use of cultured RPE cells in AMD research is well established and, indeed, some of the more recently described RPE-based models show promise for investigating the molecular mechanisms of AMD and for screening drug candidates. Here, we discuss innovative cell-culture models of AMD and emerging stem-cell-based therapies for the treatment of this vision-robbing disease. Summary: Here, we discuss the emerging cell-culture models and potential stem-cell-based therapies for AMD, a blinding disorder that affects millions of people worldwide.
Collapse
Affiliation(s)
- David L Forest
- Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| | - Lincoln V Johnson
- Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| | - Dennis O Clegg
- Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| |
Collapse
|