1
|
Nouri N, Gussler BH, Stockwell A, Truong T, Kang GJ, Browder KC, Malato Y, Sene A, Van Everen S, Wykoff CC, Brown D, Fu A, Palmer JD, Lima de Carvalho JR, Ullah E, Al Rawi R, Chew EY, Zein WM, Guan B, McCarthy MI, Hofmann JW, Chaney SY, Jasper H, Yaspan BL. SLC16A8 is a causal contributor to age-related macular degeneration risk. NPJ Genom Med 2024; 9:50. [PMID: 39468037 PMCID: PMC11519927 DOI: 10.1038/s41525-024-00442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Age-related macular degeneration (AMD), a complex neurodegenerative disease, is a leading cause of visual impairment worldwide with a strong genetic component. Genetic studies have identified several loci, but few causal genes with functional characterization. Here we highlight multiple lines of evidence which show a causal role in AMD for SLC16A8, which encodes MCT3, a retinal pigment epithelium (RPE) specific lactate transporter. First, in an unbiased, genome-wide analysis of rare coding variants we show multiple SLC16A8 rare variants are associated with AMD risk, corroborating previous borderline significant reports from AMD rare variant studies. Second, we report a novel SLC16A8 mutation in a three-generation family with early onset macular degeneration. Finally, mis-expression in multiple model organisms shows functional and anatomic retinal consequences. This study highlights the important role for SLC16A8 and lactate regulation towards outer retina/RPE health and highlights a potential new therapeutic opportunity for the treatment of AMD.
Collapse
Affiliation(s)
- Navid Nouri
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | - Tom Truong
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | - Yann Malato
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | - Charles C Wykoff
- Retina Consultants of Texas, Retina Consultants of America, Houston, TX, USA
| | - David Brown
- Retina Consultants of Texas, Retina Consultants of America, Houston, TX, USA
| | - Arthur Fu
- West Coast Retina Medical Group, San Francisco, CA, USA
| | - James D Palmer
- Northern California Retina Vitreous Associates, San Jose, CA, USA
| | - Jose Ronaldo Lima de Carvalho
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
- Hospital das Clinicas de Pernambuco-Empresa Brasileira de Servicos Hospitalares, Federal University of Pernambuco, Recife, PE, Brazil
| | - Ehsan Ullah
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ranya Al Rawi
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bin Guan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
2
|
Wojciechowski AM, Bell BA, Song Y, Anderson BD, Conomikes A, Petruconis C, Dunaief JL. Inducible RPE-specific GPX4 knockout causes oxidative stress and retinal degeneration with features of age-related macular degeneration. Exp Eye Res 2024; 247:110028. [PMID: 39128667 PMCID: PMC11392608 DOI: 10.1016/j.exer.2024.110028] [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: 06/19/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of vision loss in the elderly. This disease involves oxidative stress burden in the retina leading to death of retinal pigment epithelial (RPE) cells and photoreceptors. The retina is susceptible to oxidative stress, in part due to high metabolic activity and high concentration of polyunsaturated fatty acids that undergo lipid peroxidation chain reactions. Antioxidant enzymes exist in the retina to combat this stress, including glutathione peroxidase 4 (GPX4). GPX4 specifically reduces oxidized lipids, protecting against lipid peroxidation-induced oxidative stress, which is noted in dry AMD. We hypothesize that Gpx4 knockout within the RPE will result in an environment of chronic oxidative stress yielding degeneration akin to AMD. C57BL/6J mice with a floxed Gpx4 gene were mated with Rpe65Cre/ER mice. Offspring containing Rpe65Cre ± alleles and either Gpx4 WT or Gpx4 fl/fl alleles were administered tamoxifen to induce Gpx4 knockout in Gpx4 fl/fl mice. At sequential timepoints, retinal phenotypes were assessed via in vivo imaging utilizing confocal scanning laser ophthalmoscopy and optical coherence tomography (OCT), and visual function was probed by electroretinography. Retinas were studied post-mortem by immunohistochemical analyses, electron microscopy, plastic sectioning, and quantitative polymerase chain reaction and Western analyses. The RPE-specific Gpx4 knockout model was validated via Western analysis indicating diminished GPX4 protein only within the RPE and not the neural retina. Following Gpx4 knockout, RPE cells became dysfunctional and died, with significant cell loss occurring 2 weeks post-knockout. Progressive thinning of the photoreceptor layer followed RPE degeneration and was accompanied by loss of visual function. OCT and light microscopy showed hyperreflective foci and enlarged, pigmented cells in and above the RPE layer. Electron microscopy revealed decreased mitochondrial cristae and loss of basal and apical RPE ultrastructure. Finally, there was increased carboxyethylpyrrole staining, indicating oxidation of docosahexaenoic acid, and increased levels of mRNAs encoding oxidative stress-associated genes in the RPE and photoreceptors. Overall, we show that RPE-localized GPX4 is necessary for the health of the RPE and outer retina, and that knockout recapitulates phenotypes of dry AMD.
Collapse
Affiliation(s)
- Alaina M Wojciechowski
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brent A Bell
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ying Song
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Brandon D Anderson
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Alexa Conomikes
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Cecilia Petruconis
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Chucair-Elliott AJ, Ocañas SR, Pham K, Machalinski A, Plafker S, Stout MB, Elliott MH, Freeman WM. Age- and sex- divergent translatomic responses of the mouse retinal pigmented epithelium. Neurobiol Aging 2024; 140:41-59. [PMID: 38723422 PMCID: PMC11173338 DOI: 10.1016/j.neurobiolaging.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Aging is the main risk factor for age-related macular degeneration (AMD), a retinal neurodegenerative disease that leads to irreversible blindness, particularly in people over 60 years old. Retinal pigmented epithelium (RPE) atrophy is an AMD hallmark. Genome-wide chromatin accessibility, DNA methylation, and gene expression studies of AMD and control RPE demonstrate epigenomic/transcriptomic changes occur during AMD onset and progression. However, mechanisms by which molecular alterations of normal aging impair RPE function and contribute to AMD pathogenesis are unclear. Here, we specifically interrogate the RPE translatome with advanced age and across sexes in a novel RPE reporter mouse model. We find differential age- and sex- associated transcript expression with overrepresentation of pathways related to inflammation in the RPE. Concordant with impaired RPE function, the phenotypic changes in the aged translatome suggest that aged RPE becomes immunologically active, in both males and females, with some sex-specific signatures, which supports the need for sex representation for in vivo studies.
Collapse
Affiliation(s)
- Ana J Chucair-Elliott
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Adeline Machalinski
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Scott Plafker
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael H Elliott
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
4
|
Kretschmer V, Schneider S, Matthiessen PA, Reichert D, Hotaling N, Glasßer G, Lieberwirth I, Bharti K, De Cegli R, Conte I, Nandrot EF, May-Simera HL. Deletion of IFT20 exclusively in the RPE ablates primary cilia and leads to retinal degeneration. PLoS Biol 2023; 21:e3002402. [PMID: 38048369 PMCID: PMC10721183 DOI: 10.1371/journal.pbio.3002402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/14/2023] [Accepted: 10/26/2023] [Indexed: 12/06/2023] Open
Abstract
Vision impairment places a serious burden on the aging society, affecting the lives of millions of people. Many retinal diseases are of genetic origin, of which over 50% are due to mutations in cilia-associated genes. Most research on retinal degeneration has focused on the ciliated photoreceptor cells of the retina. However, the contribution of primary cilia in other ocular cell types has largely been ignored. The retinal pigment epithelium (RPE) is a monolayer epithelium at the back of the eye intricately associated with photoreceptors and essential for visual function. It is already known that primary cilia in the RPE are critical for its development and maturation; however, it remains unclear whether this affects RPE function and retinal tissue homeostasis. We generated a conditional knockout mouse model, in which IFT20 is exclusively deleted in the RPE, ablating primary cilia. This leads to defective RPE function, followed by photoreceptor degeneration and, ultimately, vision impairment. Transcriptomic analysis offers insights into mechanisms underlying pathogenic changes, which include transcripts related to epithelial homeostasis, the visual cycle, and phagocytosis. Due to the loss of cilia exclusively in the RPE, this mouse model enables us to tease out the functional role of RPE cilia and their contribution to retinal degeneration, providing a powerful tool for basic and translational research in syndromic and non-syndromic retinal degeneration. Non-ciliary mechanisms of IFT20 in the RPE may also contribute to pathogenesis and cannot be excluded, especially considering the increasing evidence of non-ciliary functions of ciliary proteins.
Collapse
Affiliation(s)
- Viola Kretschmer
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Sandra Schneider
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Peter Andreas Matthiessen
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Reichert
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nathan Hotaling
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gunnar Glasßer
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- University of Naples “Federico II”, Naples, Italy
| | | | - Helen Louise May-Simera
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
5
|
García-Llorca A, Ólafsson KH, Sigurdsson AT, Eysteinsson T. Progressive Cone-Rod Dystrophy and RPE Dysfunction in Mitfmi/+ Mice. Genes (Basel) 2023; 14:1458. [PMID: 37510362 PMCID: PMC10379086 DOI: 10.3390/genes14071458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Mutations in the mouse microphthalmia-associated transcription factor (Mitf) gene affect retinal pigment epithelium (RPE) differentiation and development and can lead to hypopigmentation, microphthalmia, deafness, and blindness. For instance, an association has been established between loss-of-function mutations in the mouse Mitf gene and a variety of human retinal diseases, including Waardenburg type 2 and Tietz syndromes. Although there is evidence showing that mice with the homozygous Mitfmi mutation manifest microphthalmia and osteopetrosis, there are limited or no data on the effects of the heterozygous condition in the eye. Mitf mice can therefore be regarded as an important model system for the study of human disease. Thus, we characterized Mitfmi/+ mice at 1, 3, 12, and 18 months old in comparison with age-matched wild-type mice. The light- and dark-adapted electroretinogram (ERG) recordings showed progressive cone-rod dystrophy in Mitfmi/+ mice. The RPE response was reduced in the mutant in all age groups studied. Progressive loss of pigmentation was found in Mitfmi/+ mice. Histological retinal sections revealed evidence of retinal degeneration in Mitfmi/+ mice at older ages. For the first time, we report a mouse model of progressive cone-rod dystrophy and RPE dysfunction with a mutation in the Mitf gene.
Collapse
Affiliation(s)
- Andrea García-Llorca
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | | | - Arnór Thorri Sigurdsson
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | - Thor Eysteinsson
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
- Department of Ophthalmology, Landspitali—National University Hospital, 101 Reykjavík, Iceland
| |
Collapse
|
6
|
Nguyen MN, Chakraborty D, Rao SR, Onysk A, Radkiewicz M, Surmacz L, Swiezewska E, Soubeyrand E, Akhtar TA, Kraft TW, Sherry DM, Fliesler SJ, Pittler SJ. A Dhdds K42E knock-in RP59 mouse model shows inner retina pathology and defective synaptic transmission. Cell Death Dis 2023; 14:420. [PMID: 37443173 PMCID: PMC10345138 DOI: 10.1038/s41419-023-05936-4] [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: 09/11/2022] [Revised: 06/07/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Abstract
Retinitis pigmentosa (RP) defines a group of hereditary progressive rod-cone degenerations that exhibit a common phenotype caused by variants in over 70 genes. While most variants in the dehydrodolichyl diphosphate synthase (DHDDS) gene result in syndromic abnormalities, some variants cause non-syndromic RP (RP59). DHDDS encodes one subunit of the enzyme cis-prenyltransferase (CPT), which is required for the synthesis of dolichol (Dol), that is a necessary protein glycosylation cofactor. We previously reported the creation and initial characterization of a knock-in (KI) mouse model harboring the most prevalent RP59-associated DHDDS variant (K42E) to understand how defects in DHDDS lead to retina-specific pathology. This model exhibited no profound retinal degeneration, nor protein N-glycosylation defects. Here, we report that the Dol isoprenylogue species in retina, liver, and brain of the K42E mouse model are statistically shorter than in the corresponding tissues of age-matched controls, as reported in blood and urine of RP59 patients. Retinal transcriptome analysis demonstrated elevation of many genes encoding proteins involved in synaptogenesis and synaptic function. Quantitative retinal cell layer thickness measurements demonstrated a significant reduction in the inner nuclear layer (INL) and total retinal thickness (TRT) beginning at postnatal (PN) ∼2 months, progressively increasing to PN 18-mo. Histological analysis revealed cell loss in the INL, outer plexiform layer (OPL) disruption, and ectopic localization of outer nuclear layer (ONL) nuclei into the OPL of K42E mutant retinas, relative to controls. Electroretinograms (ERGs) of mutant mice exhibited reduced b-wave amplitudes beginning at PN 1-mo, progressively declining through PN 18-mo, without appreciable a-wave attenuation, relative to controls. Our results suggest that the underlying cause of DHDDS K42E variant driven RP59 retinal pathology is defective synaptic transmission from outer to inner retina.
Collapse
Affiliation(s)
- Mai N Nguyen
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Dibyendu Chakraborty
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sriganesh Ramachandra Rao
- Research Service, VA Western New York Healthcare System, Buffalo, NY, 14215, USA
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, 14203, USA
| | - Agnieszka Onysk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02106, Poland
| | - Mariusz Radkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02106, Poland
| | - Liliana Surmacz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02106, Poland
| | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02106, Poland
| | - Eric Soubeyrand
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G2W1, Canada
| | - Tariq A Akhtar
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G2W1, Canada
| | - Timothy W Kraft
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David M Sherry
- Departments of Cell Biology, Neurosurgery, and Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Steven J Fliesler
- Research Service, VA Western New York Healthcare System, Buffalo, NY, 14215, USA
- Departments of Ophthalmology and Biochemistry and Neuroscience Graduate Program, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, 14203, USA
| | - Steven J Pittler
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
7
|
Landowski M, Grindel S, Hao Y, Ikeda S, Bowes Rickman C, Ikeda A. A Protocol to Evaluate and Quantify Retinal Pigmented Epithelium Pathologies in Mouse Models of Age-Related Macular Degeneration. J Vis Exp 2023:10.3791/64927. [PMID: 36971449 PMCID: PMC10311451 DOI: 10.3791/64927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
Age-related macular degeneration (AMD) is a debilitating retinal disorder in aging populations. It is widely believed that dysfunction of the retinal pigmented epithelium (RPE) is a key pathobiological event in AMD. To understand the mechanisms that lead to RPE dysfunction, mouse models can be utilized by researchers. It has been established by previous studies that mice can develop RPE pathologies, some of which are observed in the eyes of individuals diagnosed with AMD. Here, we describe a phenotyping protocol to assess RPE pathologies in mice. This protocol includes the preparation and evaluation of retinal cross-sections using light microscopy and transmission electron microscopy, as well as that of RPE flat mounts by confocal microscopy. We detail the common types of murine RPE pathologies observed by these techniques and ways to quantify them through unbiased methods for statistical testing. As proof of concept, we use this RPE phenotyping protocol to quantify the RPE pathologies observed in mice overexpressing transmembrane protein 135 (Tmem135) and aged wild-type C57BL/6J mice. The main goal of this protocol is to present standard RPE phenotyping methods with unbiased quantitative assessments for scientists using mouse models of AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison; McPherson Eye Research Institute, University of Wisconsin-Madison
| | - Samuel Grindel
- Department of Medical Genetics, University of Wisconsin-Madison
| | - Ying Hao
- Department of Ophthalmology, Duke University
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison; McPherson Eye Research Institute, University of Wisconsin-Madison
| | | | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison; McPherson Eye Research Institute, University of Wisconsin-Madison;
| |
Collapse
|
8
|
Use of an alternating current amplifier when recording the ERG c-wave to evaluate the function of retinal pigment epithelial cells in rats. Doc Ophthalmol 2022; 145:147-155. [PMID: 35895211 DOI: 10.1007/s10633-022-09890-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 07/08/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE We studied the conditions under which c-waves of the electroretinogram (ERG), that represent retinal pigment epithelium (RPE) function, were detectable using an alternating current (AC) amplifier and whether the c-wave recorded using an AC amplifier was useful for evaluating RPE function. METHODS We recorded ERG responses in rats to 5 s stimuli under the conditions in which the low-cut frequency and the stimulus luminance were varied. In addition, changes in ERGs were studied after intravenous injection of sodium iodate (SI) to induce RPE degeneration. RESULTS The c-wave was detected clearly when the frequency of the low-cut filter was set at 0.01 Hz and light stimulus luminances were ≥ - 1.0 log cd/m2. The c-wave was attenuated earlier than other waves (e.g., a-wave and b-wave) after SI administration. CONCLUSIONS The c-wave was easily detectable using an AC amplifier with the low-cut filter set at 0.01 Hz. Using the AC amplifier may allow easier c-wave recording, compared with the conventional use of a direct current (DC) amplifier, and could be useful for evaluating RPE function.
Collapse
|
9
|
Sterling JK, Baumann B, Foshe S, Voigt A, Guttha S, Alnemri A, McCright SJ, Li M, Zauhar RJ, Montezuma SR, Kapphahn RJ, Chavali VRM, Hill DA, Ferrington DA, Stambolian D, Mullins RF, Merrick D, Dunaief JL. Inflammatory adipose activates a nutritional immunity pathway leading to retinal dysfunction. Cell Rep 2022; 39:110942. [PMID: 35705048 PMCID: PMC9248858 DOI: 10.1016/j.celrep.2022.110942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/24/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1β (IL-1β). IL-1β upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.
Collapse
Affiliation(s)
- Jacob K Sterling
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bailey Baumann
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sierra Foshe
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Andrew Voigt
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Samyuktha Guttha
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ahab Alnemri
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sam J McCright
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Randy J Zauhar
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, PA 19104, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rebecca J Kapphahn
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Venkata R M Chavali
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David A Hill
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Deborah A Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dwight Stambolian
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Robert F Mullins
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - David Merrick
- Department of Medicine, Division of Endocrinology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Collin GB, Shi L, Yu M, Akturk N, Charette JR, Hyde LF, Weatherly SM, Pera MF, Naggert JK, Peachey NS, Nishina PM, Krebs MP. A Splicing Mutation in Slc4a5 Results in Retinal Detachment and Retinal Pigment Epithelium Dysfunction. Int J Mol Sci 2022; 23:2220. [PMID: 35216333 PMCID: PMC8875008 DOI: 10.3390/ijms23042220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/29/2022] Open
Abstract
Fluid and solute transporters of the retinal pigment epithelium (RPE) are core components of the outer blood-retinal barrier. Characterizing these transporters and their role in retinal homeostasis may provide insights into ocular function and disease. Here, we describe RPE defects in tvrm77 mice, which exhibit hypopigmented patches in the central retina. Mapping and nucleotide sequencing of tvrm77 mice revealed a disrupted 5' splice donor sequence in Slc4a5, a sodium bicarbonate cotransporter gene. Slc4a5 expression was reduced 19.7-fold in tvrm77 RPE relative to controls, and alternative splice variants were detected. SLC4A5 was localized to the Golgi apparatus of cultured human RPE cells and in apical and basal membranes. Fundus imaging, optical coherence tomography, microscopy, and electroretinography (ERG) of tvrm77 mice revealed retinal detachment, hypopigmented patches corresponding to neovascular lesions, and retinal folds. Detachment worsened and outer nuclear layer thickness decreased with age. ERG a- and b-wave response amplitudes were initially normal but declined in older mice. The direct current ERG fast oscillation and light peak were reduced in amplitude at all ages, whereas other RPE-associated responses were unaffected. These results link a new Slc4a5 mutation to subretinal fluid accumulation and altered light-evoked RPE electrophysiological responses, suggesting that SLC4A5 functions at the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Lanying Shi
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; (M.Y.); (N.S.P.)
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Nurten Akturk
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Jeremy R. Charette
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Sonia M. Weatherly
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Martin F. Pera
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Neal S. Peachey
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; (M.Y.); (N.S.P.)
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Research Service, Louis Stokes Cleveland VA Medical Center, 10701 East Boulevard, Cleveland, OH 44106, USA
| | - Patsy M. Nishina
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| | - Mark P. Krebs
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; (G.B.C.); (L.S.); (N.A.); (J.R.C.); (L.F.H.); (S.M.W.); (M.F.P.); (J.K.N.)
| |
Collapse
|
11
|
Erythropoietin Gene Therapy Delays Retinal Degeneration Resulting from Oxidative Stress in the Retinal Pigment Epithelium. Antioxidants (Basel) 2021; 10:antiox10060842. [PMID: 34070383 PMCID: PMC8229633 DOI: 10.3390/antiox10060842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023] Open
Abstract
Erythropoietin (EPO) plays an important role in erythropoiesis by its action in blocking apoptosis of progenitor cells and protects both photoreceptors and retinal ganglion cells from induced or inherited degeneration. A modified form of EPO, EPO-R76E has attenuated erythropoietic activity but is effective in inhibiting apoptosis, oxidative stress, and inflammation in several models of retinal degeneration. In this study, we used recombinant Adeno Associated Virus (AAV) to provide long-term sustained delivery of EPO-R76E and demonstrated its effects in a mouse model of dry-AMD in which retinal degeneration is induced by oxidative stress in the retinal pigment epithelial (RPE) cells. Experimental vector AAV-EPO-R76E and control vector AAV-GFP were packaged into serotype-1 (AAV1) to enable RPE selective expression. RPE oxidative stress-mediated retinal degeneration was induced by exon specific deletion of the protective enzyme MnSOD (encoded by Sod2) by cre/lox mechanism. Experimental mice received subretinal injection of AAV-EPO-R76E in the right eye and AAV-GFP in the left eye. Western blotting of RPE/choroid protein samples from AAV-EPO-R76E injected eyes showed RPE specific EPO expression. Retinal function was monitored by electroretinography (ERG). EPO-R76E over-expression in RPE delayed the retinal degeneration as measured by light microscopy in RPE specific Sod2 knockout mice. Delivery of EPO-R76E vector can be used as a tool to prevent retinal degeneration induced by RPE oxidative stress, which is implicated as a potential cause of Age-Related Macular Degeneration.
Collapse
|
12
|
Landowski M, Grindel S, Shahi PK, Johnson A, Western D, Race A, Shi F, Benson J, Gao M, Santoirre E, Lee WH, Ikeda S, Pattnaik BR, Ikeda A. Modulation of Tmem135 Leads to Retinal Pigmented Epithelium Pathologies in Mice. Invest Ophthalmol Vis Sci 2020; 61:16. [PMID: 33064130 PMCID: PMC7581492 DOI: 10.1167/iovs.61.12.16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Aging is a critical risk factor for the development of retinal diseases, but how aging perturbs ocular homeostasis and contributes to disease is unknown. We identified transmembrane protein 135 (Tmem135) as a gene important for regulating retinal aging and mitochondrial dynamics in mice. Overexpression of Tmem135 causes mitochondrial fragmentation and pathologies in the hearts of mice. In this study, we examine the eyes of mice overexpressing wild-type Tmem135 (Tmem135 TG) and compare their phenotype to Tmem135 mutant mice. Methods Eyes were collected for histology, immunohistochemistry, electron microscopy, quantitative PCR, and Western blot analysis. Before tissue collection, electroretinography (ERG) was performed to assess visual function. Mouse retinal pigmented epithelium (RPE) cultures were established to visualize mitochondria. Results Pathologies were observed only in the RPE of Tmem135 TG mice, including degeneration, migratory cells, vacuolization, dysmorphogenesis, cell enlargement, and basal laminar deposit formation despite similar augmented levels of Tmem135 in the eyecup (RPE/choroid/sclera) and neural retina. We observed reduced mitochondria number and size in the Tmem135 TG RPE. ERG amplitudes were decreased in 365-day-old mice overexpressing Tmem135 that correlated with reduced expression of RPE cell markers. In Tmem135 mutant mice, RPE cells are thicker, smaller, and denser than their littermate controls without any signs of degeneration. Conclusions Overexpression and mutation of Tmem135 cause contrasting RPE abnormalities in mice that correlate with changes in mitochondrial shape and size (overfragmented in TG vs. overfused in mutant). We conclude proper regulation of mitochondrial homeostasis by TMEM135 is critical for RPE health.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Pediatrics, Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Samuel Grindel
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Pawan K. Shahi
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Pediatrics, Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Abigail Johnson
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Daniel Western
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Adrienne Race
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Franky Shi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Jonathan Benson
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Marvin Gao
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Evelyn Santoirre
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Wei-Hua Lee
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Bikash R. Pattnaik
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Pediatrics, Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
13
|
Cao X, Soleimani M, Hughes BA. SLC26A7 constitutes the thiocyanate-selective anion conductance of the basolateral membrane of the retinal pigment epithelium. Am J Physiol Cell Physiol 2020; 319:C641-C656. [PMID: 32726161 DOI: 10.1152/ajpcell.00027.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anion channels in the retinal pigment epithelium (RPE) play an essential role in the transport of Cl- between the outer retina and the choroidal blood to regulate the ionic composition and volume of the subretinal fluid that surrounds the photoreceptor outer segments. Recently, we reported that the anion conductance of the mouse RPE basolateral membrane is highly selective for the biologically active anion thiocyanate (SCN-), a property that does not correspond with any of the Cl- channels that have been found to be expressed in the RPE to date. The purpose of this study was to determine the extent to which SLC26A7, a SCN- permeable-anion exchanger/channel that was reported to be expressed in human RPE, contributes to the RPE basolateral anion conductance. We show by quantitative RT-PCR that Slc26a7 is highly expressed in mouse RPE compared with other members of the Slc26 gene family and Cl- channel genes known to be expressed in the RPE. By applying immunofluorescence microscopy to mouse retinal sections and isolated cells, we localized SLC26A7 to the RPE basolateral membrane. Finally, we performed whole cell and excised patch recordings from RPE cells acutely isolated from Slc26a7 knockout mice to show that the SCN- conductance and permeability of its basolateral membrane are dramatically smaller relative to wild-type mouse RPE cells. These findings establish SLC26A7 as the SCN--selective conductance of the RPE basolateral membrane and provide new insight into the physiology of an anion channel that may participate in anion transport and pH regulation by the RPE.
Collapse
Affiliation(s)
- Xu Cao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | | | - Bret A Hughes
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
14
|
Miyagishima KJ, Zhang C, Malechka VV, Bharti K, Li W. Direct-Coupled Electroretinogram (DC-ERG) for Recording the Light-Evoked Electrical Responses of the Mouse Retinal Pigment Epithelium. J Vis Exp 2020. [PMID: 32744516 DOI: 10.3791/61491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The retinal pigment epithelium (RPE) is a specialized monolayer of cells strategically located between the retina and the choriocapillaris that maintain the overall health and structural integrity of the photoreceptors. The RPE is polarized, exhibiting apically and basally located receptors or channels, and performs vectoral transport of water, ions, metabolites, and secretes several cytokines. In vivo noninvasive measurements of RPE function can be made using direct-coupled ERGs (DC-ERGs). The methodology behind the DC-ERG was pioneered by Marmorstein, Peachey, and colleagues using a custom-built stimulation recording system and later demonstrated using a commercially available system. The DC-ERG technique uses glass capillaries filled with Hank's buffered salt solution (HBSS) to measure the slower electrical responses of the RPE elicited from light-evoked concentration changes in the subretinal space due to photoreceptor activity. The prolonged light stimulus and length of the DC-ERG recording make it vulnerable to drift and noise resulting in a low yield of useable recordings. Here, we present a fast, reliable method for improving the stability of the recordings while reducing noise by using vacuum pressure to reduce/eliminate bubbles that result from outgassing of the HBSS and electrode holder. Additionally, power line artifacts are attenuated using a voltage regulator/power conditioner. We include the necessary light stimulation protocols for a commercially available ERG system as well as scripts for analysis of the DC-ERG components: c-wave, fast oscillation, light peak, and off response. Due to the improved ease of recordings and rapid analysis workflow, this simplified protocol is particularly useful in measuring age-related changes in RPE function, disease progression, and in the assessment of pharmacological intervention.
Collapse
Affiliation(s)
| | - Congxiao Zhang
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health
| | - Volha V Malechka
- Human Visual Function Core, National Eye Institute, National Institutes of Health
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health;
| |
Collapse
|
15
|
Zhang C, Miyagishima KJ, Dong L, Rising A, Nimmagadda M, Liang G, Sharma R, Dejene R, Wang Y, Abu-Asab M, Qian H, Li Y, Kopera M, Maminishkis A, Martinez J, Miller S. Regulation of phagolysosomal activity by miR-204 critically influences structure and function of retinal pigment epithelium/retina. Hum Mol Genet 2020; 28:3355-3368. [PMID: 31332443 DOI: 10.1093/hmg/ddz171] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/04/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
MicroRNA-204 (miR-204) is expressed in pulmonary, renal, mammary and eye tissue, and its reduction can result in multiple diseases including cancer. We first generated miR-204-/- mice to study the impact of miR-204 loss on retinal and retinal pigment epithelium (RPE) structure and function. The RPE is fundamentally important for maintaining the health and integrity of the retinal photoreceptors. miR-204-/- eyes evidenced areas of hyper-autofluorescence and defective photoreceptor digestion, along with increased microglia migration to the RPE. Migratory Iba1+ microglial cells were localized to the RPE apical surface where they participated in the phagocytosis of photoreceptor outer segments (POSs) and contributed to a persistent build-up of rhodopsin. These structural, molecular and cellular outcomes were accompanied by decreased light-evoked electrical responses from the retina and RPE. In parallel experiments, we suppressed miR-204 expression in primary cultures of human RPE using anti-miR-204. In vitro suppression of miR-204 in human RPE similarly showed abnormal POS clearance and altered expression of autophagy-related proteins and Rab22a, a regulator of endosome maturation. Together, these in vitro and in vivo experiments suggest that the normally high levels of miR-204 in RPE can mitigate disease onset by preventing generation of oxidative stress and inflammation originating from intracellular accumulation of undigested photoreactive POS lipids. More generally, these results implicate RPE miR-204-mediated regulation of autophagy and endolysosomal interaction as a critical determinant of normal RPE/retina structure and function.
Collapse
Affiliation(s)
- Congxiao Zhang
- Ophthalmic Genetics and Visual Function Branch, Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD USA
| | - Kiyoharu J Miyagishima
- Ophthalmic Genetics and Visual Function Branch, Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD USA
| | - Lijin Dong
- Genetic Engineering Facility, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aaron Rising
- Ophthalmic Genetics and Visual Function Branch, Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Malika Nimmagadda
- Ophthalmic Genetics and Visual Function Branch, Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Genqing Liang
- Ophthalmic Genetics and Visual Function Branch, Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ruchi Sharma
- Ophthalmic Genetics and Visual Function Branch, Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Roba Dejene
- Ophthalmic Genetics and Visual Function Branch, Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuan Wang
- Ophthalmic Genetics and Visual Function Branch, Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD USA
| | - Mones Abu-Asab
- Section of Histopathology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yichao Li
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Megan Kopera
- Genetic Engineering Facility, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arvydas Maminishkis
- Ophthalmic Genetics and Visual Function Branch, Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD USA
| | - Jennifer Martinez
- Inflammation and Autoimmunity, National Institute of Environmental Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Sheldon Miller
- Ophthalmic Genetics and Visual Function Branch, Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
16
|
Young BM, Jones K, Massengill MT, Walsh E, Li H, Lewin AS, Ildefonso CJ. Expression of a CARD Slows the Retinal Degeneration of a Geographic Atrophy Mouse Model. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:113-125. [PMID: 31334304 PMCID: PMC6624323 DOI: 10.1016/j.omtm.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) has been linked to oxidative damage and para-inflammation, an activation of inflammasome signaling in the retinal pigment epithelium (RPE) and the underlying choriocapillaris. Herein, we tested the efficacy of a gene-delivered caspase-1 inhibitor in controlling the retinal degeneration observed in two models of RPE-choroid oxidative damage. In an acute model of oxidative stress (NaIO3 injection), eyes pre-treated with the sGFP-TatCARD (trans-activator of transcription; caspase activation and recruitment domain) vector demonstrated a recovery of retinal function and partial protection of RPE structure 1 month after damage, in contrast with control-treated eyes. In a model of chronic oxidative stress (RPE-specific deletion of Sod2), eyes treated with the sGFP-TatCARD vector after the onset of degeneration had a significantly slower decline in retinal function when compared to control-treated eyes. Earlier treatment of this model with the same adeno-associated virus (AAV) vector resulted in a greater protection of RPE function in eyes treated with the TatCARD when compared to control-treated eyes. Our results demonstrate that intravitreal delivery of sGFP-TatCARD reduces inflammation and can protect the retina from both acute and sustained oxidative damage within the RPE and choroid. Therefore, gene therapy with a cell-penetrating inflammasome inhibitor such as CARD may stem the progression of AMD.
Collapse
Affiliation(s)
- Brianna M Young
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL 32610-0284, USA
| | - Kyle Jones
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michael T Massengill
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Erin Walsh
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL 32610-0284, USA
| | - Hong Li
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alfred S Lewin
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Cristhian J Ildefonso
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL 32610-0284, USA.,Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
17
|
Rosales MAB, Shu DY, Iacovelli J, Saint-Geniez M. Loss of PGC-1α in RPE induces mesenchymal transition and promotes retinal degeneration. Life Sci Alliance 2019; 2:2/3/e201800212. [PMID: 31101737 PMCID: PMC6526284 DOI: 10.26508/lsa.201800212] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 01/07/2023] Open
Abstract
Sustained loss of PGC-1α in RPE cells triggers mitochondrial/autophagic dysfunction and oxidative damage resulting in epithelial dedifferentiation and mesenchymal transition. RPE dysfunction caused by deletion of the PGC-1 coactivators in vivo causes retinal degeneration. The retinal pigment epithelium (RPE) supports visual processing and photoreceptor homeostasis via energetically demanding cellular functions. Here, we describe the consequences of repressing peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α), a master regulator of mitochondrial function and biogenesis, on RPE epithelial integrity. The sustained silencing of PGC-1α in differentiating human RPE cells affected mitochondria/autophagy function, redox state, and impaired energy sensor activity ultimately inducing epithelial to mesenchymal transition (EMT). Adult conditional knockout of PGC-1 coactivators in mice resulted in rapid RPE dysfunction and transdifferentiation associated with severe photoreceptor degeneration. RPE anomalies were characteristic of autophagic defect and mesenchymal transition comparable with the ones observed in age-related macular degeneration. These findings demonstrate that PGC-1α is required to maintain the functional and phenotypic status of RPE by supporting the cells’ oxidative metabolism and autophagy-mediated repression of EMT.
Collapse
Affiliation(s)
- Mariana Aparecida Brunini Rosales
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA.,Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Daisy Y Shu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA.,Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Jared Iacovelli
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA.,Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA .,Department of Ophthalmology, Harvard Medical School, Boston, MA
| |
Collapse
|
18
|
Umeya N, Yoshizawa Y, Fukuda K, Ikeda K, Kamada M, Miyawaki I. Availability of multistep light stimulus method for evaluation of visual dysfunctions. J Pharmacol Toxicol Methods 2019; 96:27-33. [DOI: 10.1016/j.vascn.2018.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/18/2018] [Indexed: 11/28/2022]
|
19
|
Yu M, Peachey NS. Use of Direct Current Electroretinography for Analysis of Retinal Pigment Epithelium Function in Mouse Models. Methods Mol Biol 2019; 1753:103-113. [PMID: 29564784 DOI: 10.1007/978-1-4939-7720-8_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
A monolayer of pigmented epithelial cells, the retinal pigment epithelium (RPE), supports photoreceptor function in many ways. Consistent with these roles, RPE dysfunction underlies a number of hereditary retinal disorders. To monitor RPE function in vivo models for these conditions, we adapted an electroretinographic (ERG) technique based on direct current amplification (DC-ERG). This chapter describes the main features of this approach and its application to mouse models involving the RPE.
Collapse
Affiliation(s)
- Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA. .,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Neal S Peachey
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.,Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
20
|
Tarchick MJ, Cutler AH, Trobenter TD, Kozlowski MR, Makowski ER, Holoman N, Shao J, Shen B, Anand-Apte B, Samuels IS. Endogenous insulin signaling in the RPE contributes to the maintenance of rod photoreceptor function in diabetes. Exp Eye Res 2018; 180:63-74. [PMID: 30543793 DOI: 10.1016/j.exer.2018.11.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/01/2023]
Abstract
In diabetes, there are two major physiological aberrations: (i) Loss of insulin signaling due to absence of insulin (type 1 diabetes) or insulin resistance (type 2 diabetes) and (ii) increased blood glucose levels. The retina has a high proclivity to damage following diabetes, and much of the pathology seen in diabetic retinopathy has been ascribed to hyperglycemia and downstream cascades activated by increased blood glucose. However, less attention has been focused on the direct role of insulin on retinal physiology, likely due to the fact that uptake of glucose in retinal cells is not insulin-dependent. The retinal pigment epithelium (RPE) is instrumental in maintaining the structural and functional integrity of the retina. Recent studies have suggested that RPE dysfunction is a precursor of, and contributes to, the development of diabetic retinopathy. To evaluate the role of insulin on RPE cell function directly, we generated a RPE specific insulin receptor (IR) knockout (RPEIRKO) mouse using the Cre-loxP system. Using this mouse, we sought to determine the impact of insulin-mediated signaling in the RPE on retinal function under physiological control conditions as well as in streptozotocin (STZ)-induced diabetes. We demonstrate that loss of RPE-specific IR expression resulted in lower a- and b-wave electroretinogram amplitudes in diabetic mice as compared to diabetic mice that expressed IR on the RPE. Interestingly, RPEIRKO mice did not exhibit significant differences in the amplitude of the RPE-dependent electroretinogram c-wave as compared to diabetic controls. However, loss of IR-mediated signaling in the RPE reduced levels of reactive oxygen species and the expression of pro-inflammatory cytokines in the retina of diabetic mice. These results imply that IR-mediated signaling in the RPE regulates photoreceptor function and may play a role in the generation of oxidative stress and inflammation in the retina in diabetes.
Collapse
Affiliation(s)
- Matthew J Tarchick
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alecia H Cutler
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Timothy D Trobenter
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Michael R Kozlowski
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Emily R Makowski
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Nicholas Holoman
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jianning Shao
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bailey Shen
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bela Anand-Apte
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Department of Molecular Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ivy S Samuels
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
21
|
Kinoshita J, Peachey NS. Noninvasive Electroretinographic Procedures for the Study of the Mouse Retina. ACTA ACUST UNITED AC 2018; 8:1-16. [PMID: 30040236 DOI: 10.1002/cpmo.39] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Overall retinal function can be monitored by recording the light-evoked response of the eye at the corneal surface. The major components of the electroretinogram (ERG) provide important information regarding the functional status of many retinal cell types including rod photoreceptors, cone photoreceptors, bipolar cells, and the retinal pigment epithelium (RPE). The ERG can be readily recorded from mice, and this unit describes procedures for mouse anesthesia and the use of stimulation and recording procedures for measuring ERGs that reflect the response properties of different retinal cell types. Through these, the mouse ERG provides a noninvasive approach to measure multiple aspects of outer retinal function, including the status of the initial rod and cone pathways, rod photoreceptor deactivation, rod dark adaptation, the photoreceptor-to-bipolar cell synapse, and the RPE. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Junzo Kinoshita
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Neal S Peachey
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio.,Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
22
|
Kiser PD, Zhang J, Sharma A, Angueyra JM, Kolesnikov AV, Badiee M, Tochtrop GP, Kinoshita J, Peachey NS, Li W, Kefalov VJ, Palczewski K. Retinoid isomerase inhibitors impair but do not block mammalian cone photoreceptor function. J Gen Physiol 2018; 150:571-590. [PMID: 29500274 PMCID: PMC5881442 DOI: 10.1085/jgp.201711815] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 12/18/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
RPE65 is a retinoid isomerase essential for rod function, but its contribution to cone vision is enigmatic. Using selective RPE65 inhibitors, Kiser et al. demonstrate that cone function depends only partially on continuous RPE65 activity, providing support for cone-specific regeneration mechanisms. Visual function in vertebrates critically depends on the continuous regeneration of visual pigments in rod and cone photoreceptors. RPE65 is a well-established retinoid isomerase in the pigment epithelium that regenerates rhodopsin during the rod visual cycle; however, its contribution to the regeneration of cone pigments remains obscure. In this study, we use potent and selective RPE65 inhibitors in rod- and cone-dominant animal models to discern the role of this enzyme in cone-mediated vision. We confirm that retinylamine and emixustat-family compounds selectively inhibit RPE65 over DES1, the putative retinoid isomerase of the intraretinal visual cycle. In vivo and ex vivo electroretinography experiments in Gnat1−/− mice demonstrate that acute administration of RPE65 inhibitors after a bleach suppresses the late, slow phase of cone dark adaptation without affecting the initial rapid portion, which reflects intraretinal visual cycle function. Acute administration of these compounds does not affect the light sensitivity of cone photoreceptors in mice during extended exposure to background light, but does slow all phases of subsequent dark recovery. We also show that cone function is only partially suppressed in cone-dominant ground squirrels and wild-type mice by multiday administration of an RPE65 inhibitor despite profound blockade of RPE65 activity. Complementary experiments in these animal models using the DES1 inhibitor fenretinide show more modest effects on cone recovery. Collectively, these studies demonstrate a role for continuous RPE65 activity in mammalian cone pigment regeneration and provide further evidence for RPE65-independent regeneration mechanisms.
Collapse
Affiliation(s)
- Philip D Kiser
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH .,Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Jianye Zhang
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aditya Sharma
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO
| | - Juan M Angueyra
- Retinal Neurophysiology Section, National Eye Institute, Bethesda, MD
| | - Alexander V Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO
| | - Mohsen Badiee
- Department of Chemistry, College of Arts and Sciences, Case Western Reserve University, Cleveland, OH
| | - Gregory P Tochtrop
- Department of Chemistry, College of Arts and Sciences, Case Western Reserve University, Cleveland, OH
| | | | - Neal S Peachey
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH.,Cole Eye Institute, Cleveland Clinic, Cleveland, OH.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Wei Li
- Retinal Neurophysiology Section, National Eye Institute, Bethesda, MD
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
23
|
Kiser PD, Zhang J, Badiee M, Kinoshita J, Peachey NS, Tochtrop GP, Palczewski K. Rational Tuning of Visual Cycle Modulator Pharmacodynamics. J Pharmacol Exp Ther 2017; 362:131-145. [PMID: 28476927 PMCID: PMC5490204 DOI: 10.1124/jpet.117.240721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 05/01/2017] [Indexed: 11/22/2022] Open
Abstract
Modulators of the visual cycle have been developed for treatment of various retinal disorders. These agents were designed to inhibit retinoid isomerase [retinal pigment epithelium-specific 65 kDa protein (RPE65)], the rate-limiting enzyme of the visual cycle, based on the idea that attenuation of visual pigment regeneration could reduce formation of toxic retinal conjugates. Of these agents, certain ones that contain primary amine groups can also reversibly form retinaldehyde Schiff base adducts, which contributes to their retinal protective activity. Direct inhibition of RPE65 as a therapeutic strategy is complicated by adverse effects resulting from slowed chromophore regeneration, whereas effective retinal sequestration can require high drug doses with potential off-target effects. We hypothesized that the RPE65-emixustat crystal structure could help guide the design of retinaldehyde-sequestering agents with varying degrees of RPE65 inhibitory activity. We found that addition of an isopropyl group to the central phenyl ring of emixustat and related compounds resulted in agents effectively lacking in vitro retinoid isomerase inhibitory activity, whereas substitution of the terminal 6-membered ring with branched moieties capable of stronger RPE65 interaction potentiated inhibition. The isopropyl derivative series produced discernible visual cycle suppression in vivo, albeit much less potently than compounds with a high affinity for the RPE65 active site. These agents were distributed into the retina and formed Schiff base adducts with retinaldehyde. Except for one compound [3-amino-1-(3-isopropyl-5-((2,6,6-trimethylcyclohex-1-en-1-yl)methoxy)phenyl)propan-1-ol (MB-007)], these agents conferred protection against retinal phototoxicity, suggesting that both direct RPE65 inhibition and retinal sequestration are mechanisms of potential therapeutic relevance.
Collapse
Affiliation(s)
- Philip D Kiser
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Jianye Zhang
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Mohsen Badiee
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Junzo Kinoshita
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Neal S Peachey
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Gregory P Tochtrop
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine (P.D.K., J.Z., K.P.), Department of Chemistry (M.B., G.P.T.), Case Western Reserve University, Cleveland, Ohio; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio (P.D.K., N.S.P.); Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio (J.K., N.S.P.); and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (N.S.P.)
| |
Collapse
|
24
|
Biswal MR, Han P, Zhu P, Wang Z, Li H, Ildefonso CJ, Lewin AS. Timing of Antioxidant Gene Therapy: Implications for Treating Dry AMD. Invest Ophthalmol Vis Sci 2017; 58:1237-1245. [PMID: 28241311 PMCID: PMC5338629 DOI: 10.1167/iovs.16-21272] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose To investigate whether antioxidant gene therapy protects the structure and function of retina in a murine model of RPE atrophy, and to determine whether antioxidant gene therapy can prevent degeneration once it has begun. Methods We induced mitochondrial oxidative stress in RPE by conditional deletion of Sod2, the gene for manganese superoxide dismutase (MnSOD). These mice exhibited localized atrophy of the RPE and overlying photoreceptors. We restored Sod2 to the RPE of one eye using adeno-associated virus (AAV) by subretinal injection at an early (6 weeks) and a late stage (6 months), injecting the other eye with an AAV vector expressing green fluorescent protein (GFP). Retinal degeneration was monitored over a period of 9 months by electroretinography (ERG) and spectral-domain optical coherence tomography (SD-OCT). Immunohistochemical and histologic analyses were conducted to measure oxidative stress markers and to visualize retinal structure. Results One month after delivery, the AAV-Sod2 injection resulted in production of MnSod in the RPE and negligible expression in the neural retina. Electroretinography and OCT suggested no adverse effects due to increased expression of MnSOD or subretinal injection. Decrease in the ERG response and thinning retinal thickness was significantly delayed in eyes with early treatment with the Sod2 vector, but treatment at 6 months of age did not affect the ERG decline seen in these mice. Conclusions We conclude that antioxidant gene therapy may be effective in preventing the detrimental effects of oxidative stress, but may not be beneficial once substantial tissue damage has occurred.
Collapse
Affiliation(s)
- Manas R Biswal
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Pingyang Han
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Ping Zhu
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Zhaoyang Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Hong Li
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Cristhian J Ildefonso
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Alfred S Lewin
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States
| |
Collapse
|
25
|
Tarchick MJ, Bassiri P, Rohwer RM, Samuels IS. Early Functional and Morphologic Abnormalities in the Diabetic Nyxnob Mouse Retina. Invest Ophthalmol Vis Sci 2017; 57:3496-508. [PMID: 27367517 PMCID: PMC4961059 DOI: 10.1167/iovs.15-18775] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose The electroretinogram c-wave is generated by the summation of the positive polarity hyperpolarization of the apical RPE membrane and a negative polarity slow PIII response of Müller glia cells. Therefore, the c-wave reduction noted in prior studies of mouse models of diabetes could reflect a reduction in the RPE component or an increase in slow PIII. The present study used a genetic approach to distinguish between these two alternatives. Methods Nyxnob mice lack the ERG b-wave, revealing the early phase of slow PIII. To visualize changes in slow PIII due to diabetes, Nyxnob mice were given streptozotocin (STZ) injections to induce diabetes or received vehicle as a control. After 1, 2, and 4 weeks of sustained hyperglycemia (>250 mg/dL), standard strobe flash ERG and dc-ERG testing were conducted. Histological analysis of the retina was performed. Results A reduced c-wave was noted at the 1 week time point, and persisted at later time points. In comparison, slow PIII amplitudes were unaffected after 1 week of hyperglycemia, but were significantly reduced in STZ mice at the 2-week time point. The decrease in amplitude occurred before any identifiable decrease to the a-wave. At the later time point, the a-wave became involved, although the slow PIII reductions were more pronounced. Morphological abnormalities in the RPE, including increased thickness and altered melanosome distribution, were identified in diabetic animals. Conclusions Because the c-wave and slow PIII were both reduced, these results demonstrated that diabetes-induced reductions to the c-wave cannot be attributed to an early increase in the Müller glia-derived potassium conductance. Furthermore, because the a-wave, slow PIII and c-wave reductions were not equivalent, and varied in their onset, the reductions cannot reflect the same mechanism, such as a change in membrane resistance. The presence of small changes to RPE architecture indicate that the c-wave reductions present in diabetic mice likely represents a primary change in the RPE induced by hyperglycemia.
Collapse
Affiliation(s)
- Matthew J Tarchick
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States 2Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Parastoo Bassiri
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Rebecca M Rohwer
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Ivy S Samuels
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States 2Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
26
|
Leinonen H, Keksa-Goldsteine V, Ragauskas S, Kohlmann P, Singh Y, Savchenko E, Puranen J, Malm T, Kalesnykas G, Koistinaho J, Tanila H, Kanninen KM. Retinal Degeneration In A Mouse Model Of CLN5 Disease Is Associated With Compromised Autophagy. Sci Rep 2017; 7:1597. [PMID: 28487519 PMCID: PMC5431647 DOI: 10.1038/s41598-017-01716-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/04/2017] [Indexed: 11/25/2022] Open
Abstract
The Finnish variant of late infantile neuronal ceroid lipofuscinosis (CLN5 disease) belongs to a family of neuronal ceroid lipofuscinosis (NCLs) diseases. Vision loss is among the first clinical signs in childhood forms of NCLs. Mutations in CLN5 underlie CLN5 disease. The aim of this study was to characterize how the lack of normal functionality of the CLN5 protein affects the mouse retina. Scotopic electroretinography (ERG) showed a diminished c-wave amplitude in the CLN5 deficient mice already at 1 month of age, indicative of pathological events in the retinal pigmented epithelium. A- and b-waves showed progressive impairment later from 2 and 3 months of age onwards, respectively. Structural and immunohistochemical (IHC) analyses showed preferential damage of photoreceptors, accumulation of autofluorescent storage material, apoptosis of photoreceptors, and strong inflammation in the CLN5 deficient mice retinas. Increased levels of autophagy-associated proteins Beclin-1 and P62, and increased LC3b-II/LC3b-I ratio, were detected by Western blotting from whole retinal extracts. Photopic ERG, visual evoked potentials, IHC and cell counting indicated relatively long surviving cone photoreceptors compared to rods. In conclusion, CLN5 deficient mice develop early vision loss that reflects the condition reported in clinical childhood forms of NCLs. The vision loss in CLN5 deficient mice is primarily caused by photoreceptor degeneration.
Collapse
Affiliation(s)
- Henri Leinonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| | - Velta Keksa-Goldsteine
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Philip Kohlmann
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Yajuvinder Singh
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ekaterina Savchenko
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Giedrius Kalesnykas
- Experimentica Ltd., Kuopio, Finland
- Research and Development Centre for Ophthalmic Innovations (SILK), Department of Ophthalmology, University of Tampere, Tampere, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Heikki Tanila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
27
|
Bonilha VL, Bell BA, Rayborn ME, Samuels IS, King A, Hollyfield JG, Xie C, Cai H. Absence of DJ-1 causes age-related retinal abnormalities in association with increased oxidative stress. Free Radic Biol Med 2017; 104:226-237. [PMID: 28088625 PMCID: PMC5328840 DOI: 10.1016/j.freeradbiomed.2017.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 01/05/2023]
Abstract
Oxidative stress alters physiological function in most biological tissues and can lead to cell death. In the retina, oxidative stress initiates a cascade of events leading to focal loss of RPE and photoreceptors, which is thought to be a major contributing factor to geographic atrophy. Despite these implications, the molecular regulation of RPE oxidative stress under normal and pathological conditions remains largely unknown. A better understanding of the mechanisms involved in regulating RPE and photoreceptors oxidative stress response is greatly needed. To this end we evaluated photoreceptor and RPE changes in mice deficient in DJ-1, a protein that is thought to be important in protecting cells from oxidative stress. Young (3 months) and aged (18 months) DJ-1 knockout (DJ-1 KO) and age-matched wild-type mice were examined. In both group of aged mice, scanning laser ophthalmoscopy (SLO) showed the presence of a few autofluorescent foci. The 18 month-old DJ-1 KO retinas were also characterized by a noticeable increase in RPE fluorescence to wild-type. Optical coherence tomography (OCT) imaging demonstrated that all retinal layers were present in the eyes of both DJ-1 KO groups. ERG comparisons showed that older DJ-1 KO mice had reduced sensitivity under dark- and light-adapted conditions compared to age-matched control. Histologically, the RPE contained prominent vacuoles in young DJ-1 KO group with the appearance of enlarged irregularly shaped RPE cells in the older group. These were also evident in OCT and in whole mount RPE/choroid preparations labeled with phalloidin. Photoreceptors in the older DJ-1 KO mice displayed decreased immunoreactivity to rhodopsin and localized reduction in cone markers compared to the wild-type control group. Lower levels of activated Nrf2 were evident in retina/RPE lysates in both young and old DJ-1 KO mouse groups compared to wild-type control levels. Conversely, higher levels of protein carbonyl derivatives and iNOS immunoreactivity were detected in retina/RPE lysates from both young and old DJ-1 KO mice. These results demonstrate that DJ-1 KO mice display progressive signs of retinal/RPE degeneration in association with higher levels of oxidative stress markers. Collectively this analysis indicates that DJ-1 plays an important role in protecting photoreceptors and RPE from oxidative damage during aging.
Collapse
Affiliation(s)
- Vera L Bonilha
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH 44195, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Brent A Bell
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mary E Rayborn
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ivy S Samuels
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Anna King
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joe G Hollyfield
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH 44195, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Chengsong Xie
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| |
Collapse
|
28
|
Mutations in CTNNA1 cause butterfly-shaped pigment dystrophy and perturbed retinal pigment epithelium integrity. Nat Genet 2015; 48:144-51. [PMID: 26691986 PMCID: PMC4787620 DOI: 10.1038/ng.3474] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/25/2015] [Indexed: 11/11/2022]
Abstract
Butterfly-shaped pigment dystrophy is an eye disease characterized by lesions in the macula that can resemble the wings of a butterfly. Here, we report the identification of heterozygous missense mutations in the α-catenin 1 (CTNNA1) gene in three families with butterfly-shaped pigment dystrophy. In addition, we identified a Ctnna1 missense mutation in a chemically induced mouse mutant, tvrm5. Parallel clinical phenotypes were observed in the retinal pigment epithelium (RPE) of individuals with butterfly-shaped pigment dystrophy and in tvrm5 mice, including pigmentary abnormalities, focal thickening and elevated lesions, and decreased light-activated responses. Morphological studies in tvrm5 mice revealed increased cell shedding and large multinucleated RPE cells, suggesting defects in intercellular adhesion and cytokinesis. This study identifies CTNNA1 gene variants as a cause of macular dystrophy, suggests that CTNNA1 is involved in maintaining RPE integrity, and suggests that other components that participate in intercellular adhesion may be implicated in macular disease.
Collapse
|
29
|
Thavikulwat AT, Lopez P, Caruso RC, Jeffrey BG. The effects of gender and age on the range of the normal human electro-oculogram. Doc Ophthalmol 2015; 131:177-88. [PMID: 26474906 DOI: 10.1007/s10633-015-9514-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/07/2015] [Indexed: 11/30/2022]
Abstract
PURPOSE To define the normal ranges for the slow oscillations (SO) and fast oscillations (FO) of the electro-oculogram (EOG) recorded to International Society for Clinical Electrophysiology of Vision (ISCEV) standards. The effects of age and gender on the EOG ranges were examined. METHODS ISCEV standard SOs and FOs were recorded from 121 subjects (51 % male) aged from 7 to 72 years. Study variables for the SO were dark trough (DT) and light peak (LP) amplitudes (µV), times to DT and LP (min), and the Arden ratio (LP/DT amplitude). The FO was fit by a sine wave and peak-to-peak amplitude (µV), phase (°), and peak-to-trough (PT) ratios derived. The effects of age, gender and pupil size on EOG parameters were examined by multiple regression analysis. RESULTS The average Arden ratio was 2.5. Arden ratio decreased with age at a rate of 0.13 per decade of age (R (2) = 0.14, P < 0.0001). The 5th percentile of the Arden ratio decreased from 2.0 to 1.7 between 10 and 60 years of age. Median time to LP was 9 min (interquartile range 8-9 min). Time to LP was age-dependent and increased by 2 min for subjects over 55 years of age compared with those less than 25 years. EOG amplitudes were greater in women than in men (P < 0.005). The average PT ratio was 1.18, which was not affected by age or gender. Time to reach the light trough of the FO was 40 s, which increased with age (1.1 s/decade). No correlation was observed between Arden ratio and PT ratio. CONCLUSIONS The major strength of this study is the definition of the normal range and associated lower limits of ISCEV standard EOGs based on recordings from 121 subjects balanced by gender and spanning the 1st through 8th decades of life. Decreased Arden ratio and increased time to LP are associated with aging, which is likely due to the intricate mechanisms involved in generation of the light rise. Differences between the FO and SO with respect to the effects of aging are consistent with separate generation of these two EOG signals.
Collapse
Affiliation(s)
| | - Patrick Lopez
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rafael C Caruso
- Department of Psychology, Princeton University, Princeton, NJ, USA
| | - Brett G Jeffrey
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA. .,Ophthalmic Genetics and Visual Function Branch, National Eye Institute, 10 Center Drive, Room 10N226, Bethesda, MD, 20892-1860, USA.
| |
Collapse
|
30
|
Xiong WH, Pang JJ, Pennesi ME, Duvoisin RM, Wu SM, Morgans CW. The Effect of PKCα on the Light Response of Rod Bipolar Cells in the Mouse Retina. Invest Ophthalmol Vis Sci 2015; 56:4961-74. [PMID: 26230760 DOI: 10.1167/iovs.15-16622] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Protein kinase C α (PKCα) is abundantly expressed in rod bipolar cells (RBCs) in the retina, yet the physiological function of PKCα in these cells is not well understood. To elucidate the role of PKCα in visual processing in the eye, we examined the effect of genetic deletion of PKCα on the ERG and on RBC light responses in the mouse. METHODS Immunofluorescent labeling was performed on wild-type (WT), TRPM1 knockout, and PKCα knockout (PKC-KO) retina. Scotopic and photopic ERGs were recorded from WT and PKC-KO mice. Light responses of RBCs were measured using whole-cell recordings in retinal slices from WT and PKC-KO mice. RESULTS Protein kinase C alpha expression in RBCs is correlated with the activity state of the cell. Rod bipolar cells dendrites are a major site of PKCα phosphorylation. Electroretinogram recordings indicated that loss of PKCα affects the scotopic b-wave, including a larger peak amplitude, longer implicit time, and broader width of the b-wave. There were no differences in the ERG a- or c-wave between PKCα KO and WT mice, indicating no measurable effect of PKCα in photoreceptors or the RPE. The photopic ERG was unaffected consistent with the lack of detectable PKCα in cone bipolar cells. Whole-cell recordings from RBCs in PKC-KO retinal slices revealed that, compared with WT, RBC light responses in the PKC-KO retina are delayed and of longer duration. CONCLUSIONS Protein kinase C alpha plays an important modulatory role in RBCs, regulating both the peak amplitude and temporal properties of the RBC light response in the rod visual pathway.
Collapse
Affiliation(s)
- Wei-Hong Xiong
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| | - Ji-Jie Pang
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Robert M Duvoisin
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| | - Samuel M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Catherine W Morgans
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
31
|
Bonilha VL, Bell BA, Rayborn ME, Yang X, Kaul C, Grossman GH, Samuels IS, Hollyfield JG, Xie C, Cai H, Shadrach KG. Loss of DJ-1 elicits retinal abnormalities, visual dysfunction, and increased oxidative stress in mice. Exp Eye Res 2015. [PMID: 26215528 DOI: 10.1016/j.exer.2015.07.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
DJ-1/PARK7 mutations or deletions cause autosomal recessive early onset Parkinson's disease (PD). Thus, DJ-1 protein has been extensively studied in brain and neurons. PD patients display visual symptoms; however, the visual symptoms specifically attributed to PD patients carrying DJ-1/PARK7 mutations are not known. In this study, we analyzed the structure and physiology of retinas of 3- and 6-month-old DJ-1 knockout (KO) mice to determine how loss of function of DJ-1 specifically contributes to the phenotypes observed in PD patients. As compared to controls, the DJ-1 KO mice displayed an increase in the amplitude of the scotopic ERG b-wave and cone ERG, while the amplitude of a subset of the dc-ERG components was decreased. The main structural changes in the DJ-1 KO retinas were found in the outer plexiform layer (OPL), photoreceptors and retinal pigment epithelium (RPE), which were observed at 3 months and progressively increased at 6 months. RPE thinning and structural changes within the OPL were observed in the retinas in DJ-1 KO mice. DJ-1 KO retinas also exhibited disorganized outer segments, central decrease in red/green cone opsin staining, decreased labeling of ezrin, broader distribution of ribeye labeling, decreased tyrosine hydroxylase in dopaminergic neurons, and increased 7,8-dihydro-8-oxoguanine-labeled DNA oxidation. Accelerated outer retinal atrophy was observed in DJ-1 KO mice after selective oxidative damage induced by a single tail vein injection of NaIO3, exposing increased susceptibility to oxidative stress. Our data indicate that DJ-1-deficient retinas exhibit signs of morphological abnormalities and physiological dysfunction in association with increased oxidative stress. Degeneration of RPE cells in association with oxidative stress is a key hallmark of age-related macular degeneration (AMD). Therefore, in addition to detailing the visual defects that occur as a result of the absence of DJ-1, our data is also relevant to AMD pathogenesis.
Collapse
Affiliation(s)
- Vera L Bonilha
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Brent A Bell
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mary E Rayborn
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaoping Yang
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Charlie Kaul
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gregory H Grossman
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ivy S Samuels
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA; Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Joe G Hollyfield
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chengsong Xie
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Karen G Shadrach
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
32
|
Chrispell JD, Rebrik TI, Weiss ER. Electroretinogram analysis of the visual response in zebrafish larvae. J Vis Exp 2015:52662. [PMID: 25867216 PMCID: PMC4401321 DOI: 10.3791/52662] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The electroretinogram (ERG) is a noninvasive electrophysiological method for determining retinal function. Through the placement of an electrode on the surface of the cornea, electrical activity generated in response to light can be measured and used to assess the activity of retinal cells in vivo. This manuscript describes the use of the ERG to measure visual function in zebrafish. Zebrafish have long been utilized as a model for vertebrate development due to the ease of gene suppression by morpholino oligonucleotides and pharmacological manipulation. At 5-10 dpf, only cones are functional in the larval retina. Therefore, the zebrafish, unlike other animals, is a powerful model system for the study of cone visual function in vivo. This protocol uses standard anesthesia, micromanipulation and stereomicroscopy protocols that are common in laboratories that perform zebrafish research. The outlined methods make use of standard electrophysiology equipment and a low light camera to guide the placement of the recording microelectrode onto the larval cornea. Finally, we demonstrate how a commercially available ERG stimulator/recorder originally designed for use with mice can easily be adapted for use with zebrafish. ERG of larval zebrafish provides an excellent method of assaying cone visual function in animals that have been modified by morpholino oligonucleotide injection as well as newer genome engineering techniques such as Zinc Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs), and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9, all of which have greatly increased the efficiency and efficacy of gene targeting in zebrafish. In addition, we take advantage of the ability of pharmacological agents to penetrate zebrafish larvae to evaluate the molecular components that contribute to the photoresponse. This protocol outlines a setup that can be modified and used by researchers with various experimental goals.
Collapse
Affiliation(s)
- Jared D Chrispell
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | | | - Ellen R Weiss
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill;
| |
Collapse
|
33
|
Samuels IS, Bell BA, Pereira A, Saxon J, Peachey NS. Early retinal pigment epithelium dysfunction is concomitant with hyperglycemia in mouse models of type 1 and type 2 diabetes. J Neurophysiol 2014; 113:1085-99. [PMID: 25429122 DOI: 10.1152/jn.00761.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the diabetic retina, cellular changes in the retinal pigment epithelium (RPE) and neurons occur before vision loss or diabetic retinopathy can be identified clinically. The precise etiologies of retinal pathology are poorly defined, and it remains unclear if the onset and progression of cellular dysfunction differ between type 1 and type 2 diabetes. Three mouse models were used to compare the time course of RPE involvement in type 1 and type 2 diabetes. C57BL/6J mice injected with streptozotocin (STZ mice) modeled type 1 diabetes, whereas Lepr(db/db) mice on both BKS and B6.BKS background strains modeled type 2 diabetes. Electroretinogram (ERG)-based techniques were used to measure light-evoked responses of the RPE (direct current-coupled ERG, dc-ERG) and the neural retina (a-wave, b-wave). Following onset of hyperglycemia, a-wave and b-wave amplitudes of STZ mice declined progressively and by equivalent degrees. Components of the dc-ERG were also altered, with the largest reduction seen in the c-wave. Lepr(db/db) mice on the BKS strain (BKS.Lepr) displayed sustained hyperglycemia and a small increase in insulin, whereas Lepr(db/db) mice on the B6.BKS background (B6.BKS.Lepr) were transiently hyperglycemic and displayed severe hyperinsulinemia. BKS.Lepr mice exhibited sustained reductions in the dc-ERG c-wave, fast oscillation, and off response that were not attributable to reduced photoreceptor activity; B6.BKS.Lepr mice displayed transient reductions in the c-wave and fast oscillation that correlated with hyperglycemia and magnitude of photoreceptor activity. In summary, all mouse models displayed altered RPE function concomitant with the onset of hyperglycemia. These results suggest that RPE function is directly reduced by elevated blood glucose levels. That RPE dysfunction was reversible and mitigated in hyperinsulinemic B6.BKS.Lepr mice provides insight into the underlying mechanism.
Collapse
Affiliation(s)
- Ivy S Samuels
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Brent A Bell
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Ariane Pereira
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio
| | - Joseph Saxon
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Neal S Peachey
- Research Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio; Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio; and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
34
|
Patil H, Saha A, Senda E, Cho KI, Haque M, Yu M, Qiu S, Yoon D, Hao Y, Peachey NS, Ferreira PA. Selective impairment of a subset of Ran-GTP-binding domains of ran-binding protein 2 (Ranbp2) suffices to recapitulate the degeneration of the retinal pigment epithelium (RPE) triggered by Ranbp2 ablation. J Biol Chem 2014; 289:29767-89. [PMID: 25187515 DOI: 10.1074/jbc.m114.586834] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Retinal pigment epithelium (RPE) degeneration underpins diseases triggered by disparate genetic lesions, noxious insults, or both. The pleiotropic Ranbp2 controls the expression of intrinsic and extrinsic pathological stressors impinging on cellular viability. However, the physiological targets and mechanisms controlled by Ranbp2 in tissue homeostasis, such as RPE, are ill defined. We show that mice, RPE-cre::Ranbp2(-/-), with selective Ranbp2 ablation in RPE develop pigmentary changes, syncytia, hypoplasia, age-dependent centrifugal and non-apoptotic degeneration of the RPE, and secondary leakage of choriocapillaris. These manifestations are accompanied by the development of F-actin clouds, metalloproteinase-11 activation, deregulation of expression or subcellular localization of critical RPE proteins, atrophic cell extrusions into the subretinal space, and compensatory proliferation of peripheral RPE. To gain mechanistic insights into what Ranbp2 activities are vital to the RPE, we performed genetic complementation analyses of transgenic lines of bacterial artificial chromosomes of Ranbp2 harboring loss of function of selective Ranbp2 domains expressed in a Ranbp2(-/-) background. Among the transgenic lines produced, only Tg(RBD2/3*-HA)::RPE-cre::Ranbp2(-/-)-expressing mutations, which selectively impair binding of RBD2/3 (Ran-binding domains 2 and 3) of Ranbp2 to Ran-GTP, recapitulate RPE degeneration, as observed with RPE-cre::Ranbp2(-/-). By contrast, Tg(RBD2/3*-HA) expression rescues the degeneration of cone photoreceptors lacking Ranbp2. The RPE of RPE-cre::Ranbp2(-/-) and Tg(RBD2/3*-HA)::RPE-cre::Ranbp2(-/-) share proteostatic deregulation of Ran GTPase, serotransferrin, and γ-tubulin and suppression of light-evoked electrophysiological responses. These studies unravel selective roles of Ranbp2 and its RBD2 and RBD3 in RPE survival and functions. We posit that the control of Ran GTPase by Ranbp2 emerges as a novel therapeutic target in diseases promoting RPE degeneration.
Collapse
Affiliation(s)
| | - Arjun Saha
- From the Departments of Ophthalmology and
| | | | | | | | - Minzhong Yu
- the Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - Sunny Qiu
- From the Departments of Ophthalmology and
| | - Dosuk Yoon
- From the Departments of Ophthalmology and
| | - Ying Hao
- From the Departments of Ophthalmology and
| | - Neal S Peachey
- the Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, the Research Service, Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, and the Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195
| | - Paulo A Ferreira
- From the Departments of Ophthalmology and Pathology, Duke University Medical Center, Durham, North Carolina 27710,
| |
Collapse
|
35
|
Pasquay C, Wang LF, Lorenz B, Preising MN. Bestrophin 1 – Phenotypes and Functional Aspects in Bestrophinopathies. Ophthalmic Genet 2013; 36:193-212. [DOI: 10.3109/13816810.2013.863945] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Yu M, Zou W, Peachey NS, McIntyre TM, Liu J. A novel role of complement in retinal degeneration. Invest Ophthalmol Vis Sci 2012; 53:7684-92. [PMID: 23074214 DOI: 10.1167/iovs.12-10069] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The association of single nucleotide polymorphisms of components of the complement alternative pathway with the risk of age-related macular degeneration (AMD) indicates that complement signaling plays an important role in retinal physiology. How genetic variation leads to retinal degeneration is unknown. It has been assumed that complement activation augments immune responses, which in turn initiate AMD pathogenesis. To better understand the relationship between complement and the outer retina, we examined mice lacking the main complement component C3 and the receptors for complement activation fragments C3a (C3aR) and/or C5a (C5aR). METHODS Complement mutant mice were studied along with wild-type (WT) littermates from 6 weeks to 14 months of age. Strobe flash electroretinography (ERG) was used to examine outer retinal function and a dc-ERG technique was used to measure ERG components generated by the retinal pigment epithelium. Retinas were examined by histology, immunohistochemistry, and biochemistry. RESULTS Mice lacking C3aR and/or C5aR developed early onset and progressive retinal degeneration, accompanied by cleaved caspase-3 upregulation. Genetic deletion of C3aR and/or C5aR led to cell-specific defects that matched the cellular localization of these receptors in the WT retina. Compared to WT, C3aR(-/-) and C3aR(-/-)C5aR(-/-) mice showed increased retinal dysfunction upon light exposure. C3aR(-/-)C5aR(-/-) mice immunized with 4-hydroxynonenal-adducted protein developed severe retinal impairment unrelated to immune response. CONCLUSIONS C3aR- and C5aR-mediated signaling was necessary to maintain normal retinal function and structure. These receptors may be important biomarkers for predicting retinal degeneration including AMD.
Collapse
Affiliation(s)
- Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
37
|
Exclusion of aldose reductase as a mediator of ERG deficits in a mouse model of diabetic eye disease. Vis Neurosci 2012; 29:267-74. [PMID: 23101909 DOI: 10.1017/s0952523812000326] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Streptozotocin (STZ)-induced diabetes is associated with reductions in the electrical response of the outer retina and retinal pigment epithelium (RPE) to light. Aldose reductase (AR) is the first enzyme required in the polyol-mediated metabolism of glucose, and AR inhibitors have been shown to improve diabetes-induced electroretinogram (ERG) defects. Here, we used control and AR -/- mice to determine if genetic inactivation of this enzyme likewise inhibits retinal electrophysiological defects observed in a mouse model of type 1 diabetes. STZ was used to induce hyperglycemia and type 1 diabetes. Diabetic and age-matched nondiabetic controls of each genotype were maintained for 22 weeks, after which ERGs were used to measure the light-evoked components of the RPE (dc-ERG) and the neural retina (a-wave, b-wave). In comparison to their nondiabetic controls, wildtype (WT) and AR -/- diabetic mice displayed significant decreases in the c-wave, fast oscillation, and off response components of the dc-ERG but not in the light peak response. Nondiabetic AR -/- mice displayed larger ERG component amplitudes than did nondiabetic WT mice; however, the amplitude of dc-ERG components in diabetic AR -/- animals were similar to WT diabetics. ERG a-wave amplitudes were not reduced in either diabetic group, but b-wave amplitudes were lower in WT and AR -/-diabetic mice. These findings demonstrate that the light-induced responses of the RPE and outer retina are disrupted in diabetic mice, but these defects are not due to photoreceptor dysfunction, nor are they ameliorated by deletion of AR. This latter finding suggests that benefits observed in other studies utilizing pharmacological inhibitors of AR might have been secondary to off-target effects of the drugs.
Collapse
|
38
|
Onnela N, Lehtonen L, Koski M, Hyttinen J. In vitro electroretinogram for the study of the functionality of differentiated retinal pigment epithelium cells. Med Biol Eng Comput 2012; 51:61-70. [PMID: 23065627 DOI: 10.1007/s11517-012-0968-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 09/26/2012] [Indexed: 10/27/2022]
Abstract
The purpose of this study is to develop and test a method to reveal if the retinal pigment epithelium (RPE) cells differentiated from human embryonic stem cells (hESC) support the functions of photoreceptors. hESC-derived RPE (hESC-RPE) cells offer a potent cell source for cell replacement therapy that may be used to prevent certain eye diseases. Methods to assure the functionality of the RPE cells are well warranted. Electroretinograms (ERG) measure the electrophysiological response of the retina to light stimuli. A setup was developed that enables the measurement of ERG in vitro from mice retinas cultured together with hESC-RPE cells. The co-culture of RPE and retinas seems to be a viable tool to assess the functionality of RPE in vitro. However, owing to limited sample size results were somewhat mixed, and thus it was not possible to prove that hESC-RPE cells enhance the ERG response of a mouse retina in vitro. The long-term culturing of the retinas needs to be refined to acquire more conclusive evidence of the supporting role of the RPE and to explore the full potential of the co-culture and ERG methods in assessing RPE functionality.
Collapse
Affiliation(s)
- Niina Onnela
- Department of Biomedical Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland.
| | | | | | | |
Collapse
|
39
|
Age-related changes in visual function in cystathionine-beta-synthase mutant mice, a model of hyperhomocysteinemia. Exp Eye Res 2011; 96:124-31. [PMID: 22197750 DOI: 10.1016/j.exer.2011.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 12/06/2011] [Accepted: 12/09/2011] [Indexed: 11/24/2022]
Abstract
Homocysteine is an amino acid required for the metabolism of methionine. Excess homocysteine is implicated in cardiovascular and neurological disease and new data suggest a role in various retinopathies. Mice lacking cystathionine-beta-synthase (cbs(-/-)) have an excess of retinal homocysteine and develop anatomical abnormalities in multiple retinal layers, including photoreceptors and ganglion cells; heterozygous (cbs(+/-)) mice demonstrate ganglion cell loss and mitochondrial abnormalities in the optic nerve. The purpose of the present study was to determine whether elevated homocysteine, due to absent or diminished cbs, alters visual function. We examined cbs(-/-) (3 weeks) and cbs(+/-) mice (5, 10, 15, 30 weeks) and results were compared to those obtained from wild type (WT) littermates. Conventional dark- and light-adapted ERGs were recorded, along with dc-ERG to assess retinal pigment epithelial (RPE) function. The visual evoked potential (VEP) was used to assess transmission to the visual cortex. The amplitudes of the major ERG components were reduced in cbs(-/-) mice at age 3 weeks and VEPs were delayed markedly. These findings are consistent with the early retinal disruption observed anatomically in these mice. In comparison, at 3 weeks of age, responses of cbs(+/-) mice did not differ significantly from those of WT mice. Functional abnormalities were not observed in cbs(+/-) mice until 15 weeks of age, at which time amplitude reductions were noted for the ERG a- and b-wave and the light peak component, but not for other components generated by the RPE. VEP implicit times were delayed in cbs(+/-) mice at 15 and 30 weeks, while VEP amplitudes were unaffected. The later onset of functional defects in cbs(+/-) mice is consistent with a slow loss of ganglion cells reported previously in the heterozygous mutant. Light peak abnormalities indicate that RPE function is also compromised in older cbs(+/-) mice. The data suggest that severe elevations of homocysteine are associated with marked alterations of retinal function while modest homocysteine elevation is reflected in milder and delayed alterations of retinal function. The work lays the foundation to explore the role of homocysteine in retinal diseases such as glaucoma and optic neuropathy.
Collapse
|
40
|
Bharti K, Miller SS, Arnheiter H. The new paradigm: retinal pigment epithelium cells generated from embryonic or induced pluripotent stem cells. Pigment Cell Melanoma Res 2010; 24:21-34. [PMID: 20846177 DOI: 10.1111/j.1755-148x.2010.00772.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Compared with neural crest-derived melanocytes, retinal pigment epithelium (RPE) cells in the back of the eye are pigment cells of a different kind. They are a part of the brain, form an epithelial monolayer, respond to distinct extracellular signals, and provide functions that far exceed those of a light-absorbing screen. For instance, they control nutrient and metabolite flow to and from the retina, replenish 11-cis-retinal by re-isomerizing all-trans-retinal generated during photoconversion, phagocytose daily a portion of the photoreceptors' outer segments, and secrete cytokines that locally control the innate and adaptive immune systems. Not surprisingly, RPE cell damage is a major cause of human blindness worldwide, with age-related macular degeneration a prevalent example. RPE replacement therapies using RPE cells generated from embryonic or induced pluripotent stem cells provide a novel approach to a rational treatment of such forms of blindness. In fact, RPE-like cells can be obtained relatively easily when stem cells are subjected to a two-step induction protocol, a first step that leads to a neuroectodermal fate and a second to RPE differentiation. Here, we discuss the characteristics of such cells, propose criteria they should fulfill in order to be considered authentic RPE cells, and point out the challenges one faces when using such cells in attempts to restore vision.
Collapse
Affiliation(s)
- Kapil Bharti
- Mammalian Development Section, National Institutes of Neurological Disorders and Stroke, Bethesda, MD, USA.
| | | | | |
Collapse
|
41
|
Samuels IS, Sturgill GM, Grossman GH, Rayborn ME, Hollyfield JG, Peachey NS. Light-evoked responses of the retinal pigment epithelium: changes accompanying photoreceptor loss in the mouse. J Neurophysiol 2010; 104:391-402. [PMID: 20484527 DOI: 10.1152/jn.00088.2010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mutations in genes expressed in the retinal pigment epithelium (RPE) underlie a number of human inherited retinal disorders that manifest with photoreceptor degeneration. Because light-evoked responses of the RPE are generated secondary to rod photoreceptor activity, RPE response reductions observed in human patients or animal models may simply reflect decreased photoreceptor input. The purpose of this study was to define how the electrophysiological characteristics of the RPE change when the complement of rod photoreceptors is decreased. To measure RPE function, we used an electroretinogram (dc-ERG)-based technique. We studied a slowly progressive mouse model of photoreceptor degeneration (Prph(Rd2/+)), which was crossed onto a Nyx(nob) background to eliminate the b-wave and most other postreceptoral ERG components. On this background, Prph(Rd2/+) mice display characteristic reductions in a-wave amplitude, which parallel those in slow PIII amplitude and the loss of rod photoreceptors. At 2 and 4 mo of age, the amplitude of each dc-ERG component (c-wave, fast oscillation, light peak, and off response) was larger in Prph(Rd2/+) mice than predicted by rod photoreceptor activity (Rm(P3)) or anatomical analysis. At 4 mo of age, the RPE in Prph(Rd2/+) mice showed several structural abnormalities including vacuoles and swollen, hypertrophic cells. These data demonstrate that insights into RPE function can be gained despite a loss of photoreceptors and structural changes in RPE cells and, moreover, that RPE function can be evaluated in a broader range of mouse models of human retinal disease.
Collapse
Affiliation(s)
- Ivy S Samuels
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Edwards MM, Marín de Evsikova C, Collin GB, Gifford E, Wu J, Hicks WL, Whiting C, Varvel NH, Maphis N, Lamb BT, Naggert JK, Nishina PM, Peachey NS. Photoreceptor degeneration, azoospermia, leukoencephalopathy, and abnormal RPE cell function in mice expressing an early stop mutation in CLCN2. Invest Ophthalmol Vis Sci 2010; 51:3264-72. [PMID: 20071672 DOI: 10.1167/iovs.09-4887] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine the molecular basis and the pathologic consequences of a chemically induced mutation in a mouse model of photoreceptor degeneration, nmf240. METHODS Mice from a G3 N-ethyl-N-nitrosourea mutagenesis program were screened by indirect ophthalmoscopy for abnormal fundi. A chromosomal position for the recessive nmf240 mutation was determined by a genome-wide linkage analysis by use of simple sequence length polymorphic markers in an F2 intercross. The critical region was refined, and candidate genes were screened by direct sequencing. The nmf240 phenotype was characterized by histologic analysis of the retina, brain, and male reproductive organs and by electroretinogram (ERG)-based studies of the retina and retinal pigment epithelium (RPE). RESULTS Clinically, homozygous nmf240 mutants exhibit a grainy retina that progresses to panretinal patches of depigmentation. The mutation was localized to a region on chromosome 16 containing Clcn2, a gene associated with retinal degeneration. Sequencing identified a missense C-T mutation at nucleotide 1063 in Clcn2 that converts a glutamine to a stop codon. Mice homozygous for the Clcn2(nmf240) mutation experience a severe loss of photoreceptor cells at 14 days of age that is preceded by an elongation of RPE apical microvilli. Homozygous mutants also experience leukoencephalopathy in multiple brain areas and male sterility. Despite a normal retinal histology in nmf240 heterozygotes, the ERG light peak, generated by the RPE, is reduced. CONCLUSIONS The nmf240 phenotype closely resembles that reported for Clcn2 knockout mice. The observation that heterozygous nmf240 mice present with a reduced ERG light peak component suggests that CLCN2 is necessary for the generation of this response component.
Collapse
|
43
|
Zhang Y, Stanton JB, Wu J, Yu K, Hartzell HC, Peachey NS, Marmorstein LY, Marmorstein AD. Suppression of Ca2+ signaling in a mouse model of Best disease. Hum Mol Genet 2010; 19:1108-18. [PMID: 20053664 DOI: 10.1093/hmg/ddp583] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Mutations in BEST1, encoding bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited macular degeneration characterized by a diminished electrooculogram light peak (LP), lipofuscin in retinal pigment epithelial cells (RPE), and fluid- and debris-filled retinal detachments. To understand the pathogenesis of BVMD we generated knock-in mice carrying the BVMD-causing mutation W93C in Best1. Both Best1(+/W93C)and Best1(W93C/W93C) mice had normal ERG a- and b-waves, but exhibited an altered LP luminance response reminiscent of that observed in BVMD patients. Morphological analysis identified fluid- and debris-filled retinal detachments in mice as young as 6 months of age. By 18-24 months of age Best1(+/W93C)and Best1(W93C/W93C) mice exhibited enhanced accumulation of lipofuscin in the RPE, and a significant deposition of debris composed of unphagocytosed photoreceptor outer segments and lipofuscin granules in the subretinal space. Although Best1 is thought to function as a Ca(2+)-activated Cl(-) channel, RPE cells from Best1(W93C) mice exhibited normal Cl(-) conductances. We have previously shown that Best1(-/-) mice exhibit increased [Ca(2+)](i) in response to ATP stimulation. However, ATP-stimulated changes in [Ca(2+)](i) in RPE cells from Best1(+/W93C) and Best1(W93C/W93C) mice were suppressed relative to Best1(+/+) littermates. Based on these data we conclude that mice carrying the Best1(W93C) mutation are a valid model for BVMD. Furthermore, these data suggest that BVMD is not because of Best1 deficiency, as the phenotypes of Best1(+/W93C) and Best1(W93C/W93C) mice are distinct from that of Best1(-/-) mice with regard to lipofuscin accumulation, and changes in the LP and ATP Ca(2+) responses.
Collapse
Affiliation(s)
- Youwen Zhang
- Department of Ophthalmology and Vision Science, University of Arizona, 655 N. Alvernon Way, Suite 108, Tucson, AZ 85711, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Marmorstein AD, Cross HE, Peachey NS. Functional roles of bestrophins in ocular epithelia. Prog Retin Eye Res 2009; 28:206-26. [PMID: 19398034 DOI: 10.1016/j.preteyeres.2009.04.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
There are four members of the bestrophin family of proteins in the human genome, of which two are known to be expressed in the eye. The gene BEST1 (formerly VMD2) which encodes the protein bestrophin-1 (Best1) was first identified in 1998. Mutations in this gene have now been associated with four clinically distinguishable human eye diseases, collectively referred to as "bestrophinopathies". Over the last decade, laboratories have sought to understand how Best1 mutations could result in eye diseases that range in presentation from macular degeneration to nanophthalmos. The majority of our knowledge comes from studies that have sought to understand how Best1 mutations or dysfunction could induce the classical symptoms of the most common of these diseases: Best vitelliform macular dystrophy (BVMD). BVMD is a dominant trait that is characterized electrophysiologically by a diminished electrooculogram light peak with a normal clinical electroretinogram. This together with the localization of Best1 to the retinal pigment epithelium (RPE) basolateral plasma membrane and data from heterologous expression studies, have led to the proposal that Best1 generates the light peak, and that bestrophins are a family of Ca(2+) activated Cl(-) channels (CaCCs). However, data from Best1 knock-out and knock-in mice, coupled with the recent discovery of a recessive bestrophinopathy suggest that Best1 does not generate the light peak. Recently Best2 was found to be expressed in non-pigmented epithelia in the ciliary body. However, aqueous dynamics in Best2 knock-out mice do not support a role for Best2 as a Cl(-) channel. Thus, the purported CaCC function of the bestrophins and how loss of this function relates to clinical disease needs to be reassessed. In this article, we examine data obtained from tissue-type and animal models and discuss the current state of bestrophin research, what roles Best1 and Best2 may play in ocular epithelia and ocular electrophysiology, and how perturbation of these functions may result in disease.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA.
| | | | | |
Collapse
|
45
|
Membrane frizzled-related protein is necessary for the normal development and maintenance of photoreceptor outer segments. Vis Neurosci 2009; 25:563-74. [PMID: 18764959 DOI: 10.1017/s0952523808080723] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A 4 base pair deletion in a splice donor site of the Mfrp (membrane-type frizzled-related protein) gene, herein referred to as Mfrprd6/rd6, is predicted to lead to the skipping of exon 4 and photoreceptor degeneration in retinal degeneration 6 (rd6) mutant mice. Little, however, is known about the function of the protein or how the mutation causes the degenerative retinal phenotype. Here we examine ultrastructural changes in the retina of Mfrprd6/rd6 mice to determine the earliest effects of the mutation. We also extend the reported observations of the expression pattern of the dicistronic Mfrp/C1qtnf5 message and the localization of these and other retinal pigment epithelium (RPE) and retinal proteins during development and assess the ability of RPE cells to phagocytize outer segments (OSs) in mutant and wild-type (WT) mice. At the ultrastructural level, OSs do not develop normally in Mfrprd6/rd6 mutants. They are disorganized and become progressively shorter as mutant mice age. Additionally, there are focal areas in which there is a reduction of apical RPE microvilli. At P25, the rod electroretinogram (ERG) a-wave of Mfrprd6/rd6 mice is reduced in amplitude by ~50% as are ERG components generated by the RPE. Examination of beta-catenin localization and Fos and Tcf-1 expression, intermediates of the canonical Wnt pathway, showed that they were not different between mutant and WT mice, suggesting that MFRP may operate through an alternative pathway. Finally, impaired OS phagocytosis was observed in Mfrprd6/rd6 mice both in standard ambient lighting conditions and with bright light exposure when compared to WT controls.
Collapse
|
46
|
Nöjd N, Ilmarinen T, Lehtonen L, Skottman H, Suuronen R, Hyttinen J. Using MEA system in verifying the functionality of retinal pigment epithelium cells differentiated from human embryonic stem cells. IFMBE PROCEEDINGS 2009. [DOI: 10.1007/978-3-540-89208-3_547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
47
|
Hartzell HC, Qu Z, Yu K, Xiao Q, Chien LT. Molecular physiology of bestrophins: multifunctional membrane proteins linked to best disease and other retinopathies. Physiol Rev 2008; 88:639-72. [PMID: 18391176 DOI: 10.1152/physrev.00022.2007] [Citation(s) in RCA: 258] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This article reviews the current state of knowledge about the bestrophins, a newly identified family of proteins that can function both as Cl(-) channels and as regulators of voltage-gated Ca(2+) channels. The founding member, human bestrophin-1 (hBest1), was identified as the gene responsible for a dominantly inherited, juvenile-onset form of macular degeneration called Best vitelliform macular dystrophy. Mutations in hBest1 have also been associated with a small fraction of adult-onset macular dystrophies. It is proposed that dysfunction of bestrophin results in abnormal fluid and ion transport by the retinal pigment epithelium, resulting in a weakened interface between the retinal pigment epithelium and photoreceptors. There is compelling evidence that bestrophins are Cl(-) channels, but bestrophins remain enigmatic because it is not clear that the Cl(-) channel function can explain Best disease. In addition to functioning as a Cl(-) channel, hBest1 also is able to regulate voltage-gated Ca(2+) channels. Some bestrophins are activated by increases in intracellular Ca(2+) concentration, but whether bestrophins are the molecular counterpart of Ca(2+)-activated Cl(-) channels remains in doubt. Bestrophins are also regulated by cell volume and may be a member of the volume-regulated anion channel family.
Collapse
Affiliation(s)
- H Criss Hartzell
- Department of Cell Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | |
Collapse
|
48
|
Kang Derwent JJ, Saszik SM, Maeda H, Little DM, Pardue MT, Frishman LJ, Pepperberg DR. Test of the paired-flash electroretinographic method in mice lacking b-waves. Vis Neurosci 2007; 24:141-9. [PMID: 17640404 DOI: 10.1017/s0952523807070162] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 02/14/2007] [Indexed: 11/07/2022]
Abstract
Previous studies of rod photoreceptors in vivo have employed a paired-flash electroretinographic (ERG) technique to determine rod response properties. To test whether absence versus presence of the ERG b-wave affects the photoreceptor response derived by the paired-flash method, we examined paired-flash-derived responses obtained from nob mice, a mutant strain with a defect in signal transduction between photoreceptors and ON bipolar cells that causes a lack of the b-wave. Normal littermates of the nob mice served as controls. The normalized amplitude-intensity relation of the derived response determined in nob mice at the near-peak time of 86 ms was similar to that determined for the controls. The full time course of the derived rod response was obtained for test flash strengths ranging from 0.11 to 17.38 scotopic cd s m(-2) (sc cd s m(-2)). Time-course data obtained from nob and control mice exhibited significant but generally modest differences. With saturating test flash strengths, half-recovery times for the derived response of nob versus control mice differed by approximately 60 ms or less about the combined (nob and control) average respective values. Time course data also were obtained before versus after intravitreal injection of L-2-amino-4-phosphonobutyrate (APB) (which blocks transmission from photoreceptors to depolarizing bipolar cells) and of cis 2,3-piperidine dicarboxylic acid (PDA) (which blocks transmission to OFF bipolar cells, and to horizontal, amacrine and ganglion cells). Neither APB nor PDA substantially affected derived responses obtained from nob or control mice. The results provide quantitative information on the effect of b-wave removal on the paired-flash-derived response in mouse. They argue against a substantial skewing effect of the b-wave on the paired-flash-derived response obtained in normal mice and are consistent with the notion that, to good approximation, this derived response represents the isolated flash response of the photoreceptors in both nob and normal mice.
Collapse
Affiliation(s)
- Jennifer J Kang Derwent
- Lions of Illinois Eye Research Institute, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Pinto LH, Invergo B, Shimomura K, Takahashi JS, Troy JB. Interpretation of the mouse electroretinogram. Doc Ophthalmol 2007; 115:127-36. [PMID: 17636411 PMCID: PMC3786689 DOI: 10.1007/s10633-007-9064-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 05/01/2007] [Accepted: 05/03/2007] [Indexed: 12/22/2022]
Abstract
The mouse electroretinogram (ERG) consists of a complex set of signals or "waves" generated by multiple types of retinal cell. The origins of these waves are reviewed briefly for the C57BL/6J mouse. The differences in the properties of these waves are described for 34 strains of mice and 11 F1 hybrid mice, as is the way that inter-strain genetic polymorphisms can be exploited in order to help pin-point the genes responsible for ERG differences. There are certain technical difficulties, some subtle, that can arise in recording the ERG and these are classified and illustrated in order to facilitate their diagnosis. Forward genetic screens are described, along with abnormal mice that have been generated in a large screen. Several means are suggested for determining if a mouse having an abnormal ERG is a mutant.
Collapse
Affiliation(s)
- Lawrence H Pinto
- Department of Neurobiology and Physiology and Center for Functional Genomics, Northwestern University, 2205 Tech Drive, Evanston, IL 60208, USA.
| | | | | | | | | |
Collapse
|
50
|
Bakall B, Radu RA, Stanton JB, Burke JM, McKay BS, Wadelius C, Mullins RF, Stone EM, Travis GH, Marmorstein AD. Enhanced accumulation of A2E in individuals homozygous or heterozygous for mutations in BEST1 (VMD2). Exp Eye Res 2007; 85:34-43. [PMID: 17477921 DOI: 10.1016/j.exer.2007.02.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 02/21/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
Best vitelliform macular dystrophy (BMD) is an autosomal dominant inherited macular degenerative disease caused by mutations in the gene BEST1 (formerly VMD2). Prior reports indicate that BMD is characterized histopathologically by accumulation of lipofuscin in the retinal pigment epithelium (RPE). However, this accumulation has not been quantified and the chemical composition of lipofuscin in BMD has not been examined. In this study we characterize the histopathology of a donor eye from a rare individual homozygous for a mutation (W93C) in BEST1. We find that this individual's disease was not any more severe than has been described for heterozygotes. We then used this tissue to quantify lipofuscin accumulation by enriching intracellular granules from RPE cells on sucrose gradients and counting the granules in each density fraction. Granules from the homozygous donor eye as well as a donor eye from an individual heterozygous for the mutation T6R were compared with age-matched control eyes. Interestingly, the least dense fraction, representing classical lipofuscin granules was either not present or significantly diminished in the BMD donor eyes and the autoflourescence associated with lipofuscin had shifted to denser fractions. However, a substantial enrichment for granules in fractions of higher density was also noted in the BMD samples. Inspection of granules from the homozygous donor eye by electron microscopy revealed a complex abnormal multilobular structure. Analysis of granules by HPLC indicated a approximately 1.6- and approximately fourfold overall increase in A2E in the BMD eyes versus age-matched control eyes, with a shift of A2E to more dense granules in the BMD donor eyes. Despite the increase in A2E and total intracellular granules, the RPE in the homozygous donor eyes was relatively well preserved. Based on these data we conclude that the clinical and histopathologic consequences to the homozygous donor were not any more severe than has been reported previously for individuals who are established or presumptive heterozygotes. We find that A2E is a component of the lipofuscin accumulated in BMD and that it is more abundant than in control eyes suggesting that the etiology of BMD is similar to Stargardt's disease and Stargardt-like macular dystrophy. Finally, the changes we observe in the granules suggest that the histopathology and eventual vision loss associated with BMD may be due to defects in the ability of the RPE to fully degrade phagocytosed photoreceptor outer segments.
Collapse
Affiliation(s)
- B Bakall
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|