1
|
Engfer ZJ, Palczewski K. The multifaceted roles of retinoids in eye development, vision, and retinal degenerative diseases. Curr Top Dev Biol 2024; 161:235-296. [PMID: 39870435 DOI: 10.1016/bs.ctdb.2024.10.003] [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] [Indexed: 01/29/2025]
Abstract
Vitamin A (all-trans-retinol; at-Rol) and its derivatives, known as retinoids, have been adopted by vertebrates to serve as visual chromophores and signaling molecules, particularly in the eye/retina. Few tissues rely on retinoids as heavily as the retina, and the study of genetically modified mouse models with deficiencies in specific retinoid-metabolizing proteins has allowed us to gain insight into the unique or redundant roles of these proteins in at-Rol uptake and storage, or their downstream roles in retinal development and function. These processes occur during embryogenesis and continue throughout life. This review delves into the role of these genes in supporting retinal function and maps the impact that genetically modified mouse models have had in studying retinoid-related genes. These models display distinct perturbations in retinoid biochemistry, physiology, and metabolic flux, mirroring human ocular diseases.
Collapse
Affiliation(s)
- Zachary J Engfer
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States.
| | - Krzysztof Palczewski
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States; Department of Chemistry, University of California Irvine, Irvine, CA, United States; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
2
|
Abstract
The continuous function of vertebrate photoreceptors requires regeneration of their visual pigment following its destruction upon activation by light (photobleaching). For rods, the chromophore required for the regeneration of rhodopsin is derived from the adjacent retinal pigmented epithelium (RPE) cells through a series of reactions collectively known as the RPE visual cycle. Mounting biochemical and functional evidence demonstrates that, for cones, pigment regeneration is supported by the parallel supply with chromophore by two pathways-the canonical RPE visual cycle and a second, cone-specific retina visual cycle that involves the Müller glial cells in the neural retina. In this article, we review historical information that led to the discovery of the retina visual cycle and discuss what is currently known about the reactions and molecular components of this pathway and its functional role in supporting cone-mediated vision.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, California, USA; ,
| | - Vladimir J Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, California, USA; ,
| |
Collapse
|
3
|
Getz TE, Chrenek MA, Papania JT, Shelton DA, Markand S, Iuvone PM, Kozmik Z, Boatright JH, Nickerson JM. Conditional Knockouts of Interphotoreceptor Retinoid Binding Protein Suggest Two Independent Mechanisms for Retinal Degeneration and Myopia. Invest Ophthalmol Vis Sci 2024; 65:32. [PMID: 38904640 PMCID: PMC11193143 DOI: 10.1167/iovs.65.6.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
Purpose Interphotoreceptor retinoid-binding protein's (IRBP) role in eye growth and its involvement in cell homeostasis remain poorly understood. One hypothesis proposes early conditional deletion of the IRBP gene could lead to a myopic response with retinal degeneration, whereas late conditional deletion (after eye size is determined) could cause retinal degeneration without myopia. Here, we sought to understand if prior myopia was required for subsequent retinal degeneration in the absence of IRBP. This study investigates if any cell type or developmental stage is more important in myopia or retinal degeneration. Methods IBRPfl/fl mice were bred with 5 Cre-driver lines: HRGP-Cre, Chx10-Cre, Rho-iCre75, HRGP-Cre Rho-iCre75, and Rx-Cre. Mice were analyzed for IRBP gene expression through digital droplet PCR (ddPCR). Young adult (P30) mice were tested for retinal degeneration and morphology using spectral-domain optical coherence tomography (SD-OCT) and hematoxylin and eosin (H&E) staining. Function was analyzed using electroretinograms (ERGs). Eye sizes and axial lengths were compared through external eye measurements and whole eye biometry. Results Across all outcome measures, when bred to IRBPfl/fl, HRGP-Cre and Chx10-Cre lines showed no differences from IRBPfl/fl alone. With the Rho-iCre75 line, small but significant reductions were seen in retinal thickness with SD-OCT imaging and postmortem H&E staining without increased axial length. Both the HRGP-Cre+Rho-iCre75 and the Rx-Cre lines showed significant decreases in retinal thickness and outer nuclear layer cell counts. Using external eye measurements and SD-OCT imaging, both lines showed an increase in eye size. Finally, function in both lines was roughly halved across scotopic, photopic, and flicker ERGs. Conclusions Our studies support hypotheses that for both eye size determination and retinal homeostasis, there are two critical timing windows when IRBP must be expressed in rods or cones to prevent myopia (P7-P12) and degeneration (P21 and later). The rod-specific IRBP knockout (Rho-iCre75) showed significant retinal functional losses without myopia, indicating that the two phenotypes are independent. IRBP is needed for early development of photoreceptors and eye size, whereas Rho-iCre75 IRBPfl/fl knockout results in retinal degeneration without myopia.
Collapse
Affiliation(s)
- Tatiana E. Getz
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Jack T. Papania
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Debresha A. Shelton
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Shanu Markand
- Kirksville College of Osteopathic Medicine, A.T. Still University, Kirksville, Missouri, United States
| | - P. Michael Iuvone
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Jeffrey H. Boatright
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Emory University, Department of Ophthalmology, Atlanta, Georgia, United States
| |
Collapse
|
4
|
Georgiou M, Fujinami K, Robson AG, Fujinami-Yokokawa Y, Shakarchi AF, Ji MH, Uwaydat SH, Kim A, Kolesnikova M, Arno G, Pontikos N, Mahroo OA, Tsang SH, Webster AR, Michaelides M. RBP3-Retinopathy-Inherited High Myopia and Retinal Dystrophy: Genetic Characterization, Natural History, and Deep Phenotyping. Am J Ophthalmol 2024; 258:119-129. [PMID: 37806543 PMCID: PMC11139644 DOI: 10.1016/j.ajo.2023.09.025] [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] [Received: 05/08/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE To examine the genetic and clinical features and the natural history of RBP3-associated retinopathy. DESIGN Multi-center international, retrospective, case series of adults and children, with moleculraly confirmed RBP3-asociated retinopathy. METHODS The genetic, clinical, and retinal imaging findings, including optical coherence tomography (OCT) and fundus autofluorescence (FAF), were investigated both cross-sectionally and longitudinally. The results of international standard full-field electroretinography (ERG) and pattern electroretinography (PERG) were reviewed. RESULTS We ascertained 12 patients (5 female and 7 male) from 10 families (4 patients previously reported). Ten novel disease-causing RBP3 variants were identified. Ten patients were homozygous. The mean age (±SD, range) of the group was 21.4 years (±19.1, 2.9-60.5 years) at baseline evaluation. All 12 patients were highly myopic, with a mean spherical equivalent of -16.0D (range, -7.0D to -33.0D). Visual acuity was not significantly different between eyes, and no significant anisometropia was observed. Mean best-corrected visual acuity (BCVA) was 0.48 logMAR (SD, ±0.29; range, 0.2-1.35 logMAR); at baseline. Eleven patients had longitudinal BCVA assessment, with a mean BCVA of 0.46 logMAR after a mean follow-up of 12.6 years. All patients were symptomatic with reduced VA and myopia by the age of 7 years old. All patients had myopic fundi and features in keeping with high myopia on OCT, including choroidal thinning. The 4 youngest patients had no fundus pigmentary changes, with the rest of the patients presenting with a variable degree of mid-peripheral pigmentation and macular changes. FAF showed variable phenotypes, ranging from areas of increased signal to advanced atrophy in older patients. OCT showed cystoid macular edema at presentation in 3 patients, which persisted during follow-up in 2 patients and resolved to atrophy in the third patient. The ERGs were abnormal in 9 of 9 cases, revealing variable relative involvement of rod and cone photoreceptors with additional milder dysfunction post-phototransduction in some. All but 1 patient had PERG evidence of macular dysfunction, which was severe in most cases. CONCLUSIONS This study details the clinical and functional phenotype of RBP3-retinopathy in the largest cohort reported to date. RBP3-retinopathy is a disease characterized by early onset, slow progression over decades, and high myopia. The phenotypic spectrum and natural history as described herein has prognostic and counseling implications. RBP3-related disease should be considered in children with high myopia and retinal dystrophy.
Collapse
Affiliation(s)
- Michalis Georgiou
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK; Jones Eye Institute (M.G., A.F.S., M.H.J., S.H.U.), University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kaoru Fujinami
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK; Laboratory of Visual Physiology (K.F., Y.F.-Y.), Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Anthony G Robson
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK
| | - Yu Fujinami-Yokokawa
- Laboratory of Visual Physiology (K.F., Y.F.-Y.), Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan; Department of Health Policy and Management (Y.F.-Y.), Keio University School of Medicine, Tokyo, Japan
| | - Ahmed F Shakarchi
- Jones Eye Institute (M.G., A.F.S., M.H.J., S.H.U.), University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Marco H Ji
- Jones Eye Institute (M.G., A.F.S., M.H.J., S.H.U.), University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Sami H Uwaydat
- Jones Eye Institute (M.G., A.F.S., M.H.J., S.H.U.), University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Angela Kim
- Jonas Children's Vision Care (A.K., M.K., S.H.T.), Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Columbia University, and Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| | - Masha Kolesnikova
- Jonas Children's Vision Care (A.K., M.K., S.H.T.), Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Columbia University, and Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| | - Gavin Arno
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK
| | - Nikolas Pontikos
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK
| | - Omar A Mahroo
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK
| | - Stephen H Tsang
- Jonas Children's Vision Care (A.K., M.K., S.H.T.), Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Columbia University, and Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| | - Andrew R Webster
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK
| | - Michel Michaelides
- From Moorfields Eye Hospital (M.G., K.F., A.G.R., G.A., N.P., O.A.M., A.R.W., M.M.), London, UK; UCL Institute of Ophthalmology (M.G., K.F., A.G.R.m G.A., N.P., O.A.M., A.R.W., M.M.), University College London, London, UK.
| |
Collapse
|
5
|
Gombosh M, Yogev Y, Hadar N, Proskorovski-Ohayon R, Aharoni S, Gradstein L, Birk OS. De-novo "germline second hit" loss-of-heterozygosity RBP3 deletion mutation causing recessive high myopia. Clin Genet 2023; 104:571-576. [PMID: 37308324 DOI: 10.1111/cge.14384] [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: 01/11/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/14/2023]
Abstract
Knudson's "two hit" hypothesis, mostly associated with cancer, relates to a primary heterozygous germline mutation complemented by a somatic mutation in the second allele. When the somatic "second hit" is a deletion mutation, the heterozygosity due to the first hit is lost ("loss of heterozygosity"). As the rate of germline mutations is almost two orders of magnitude lower than that of somatic mutations, de-novo germline mutations causing autosomal recessive diseases in carriers of inherited heterozygous mutations are not common. We delineate a case of high myopia presenting at infancy with mild diminution of retinal responses. Exome sequencing identified a paternally inherited apparently homozygous missense mutation in RBP3. Chromosomal microarrays delineated a de-novo germline heterozygous deletion encompassing RBP3, verified through revision of WES data. Thus, we demonstrate an inherited RBP3 missense mutation complemented by a de-novo germline RBP3 deletion, causing loss of heterozygosity of the inherited mutation. We describe a novel RBP3 missense mutation, report the first isolated RBP3 deletion, and demonstrate infantile high myopia as an initial presentation of RBP3 disease. Notably, we highlight de-novo germline deletion mutations causing "loss of heterozygosity" of inherited heterozygous mutations, culminating in autosomal recessive diseases, and discuss the scarce literature.
Collapse
Affiliation(s)
- Maya Gombosh
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Yuval Yogev
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Noam Hadar
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Regina Proskorovski-Ohayon
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Sarit Aharoni
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Libe Gradstein
- Department of Ophthalmology, Soroka Medical Center and Clalit Health Services, Ben-Gurion University, Beer-Sheva, Israel
| | - Ohad S Birk
- The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
- Genetics Institute, Soroka Medical Center, Beer-Sheva, Israel
| |
Collapse
|
6
|
DeRamus ML, Jasien JV, Eppstein JM, Koala P, Kraft TW. Retinal Responses to Visual Stimuli in Interphotoreceptor Retinoid Binding-Protein Knock-Out Mice. Int J Mol Sci 2023; 24:10655. [PMID: 37445836 PMCID: PMC10341985 DOI: 10.3390/ijms241310655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) is an abundant glycoprotein in the subretinal space bound by the photoreceptor (PR) outer segments and the processes of the retinal pigmented epithelium (RPE). IRBP binds retinoids, including 11-cis-retinal and all-trans-retinol. In this study, visual function for demanding visual tasks was assessed in IRBP knock-out (KO) mice. Surprisingly, IRBP KO mice showed no differences in scotopic critical flicker frequency (CFF) compared to wildtype (WT). However, they did have lower photopic CFF than WT. IRBP KO mice had reduced scotopic and photopic acuity and contrast sensitivity compared to WT. IRBP KO mice had a significant reduction in outer nuclear layer (ONL) thickness, PR outer and inner segment, and full retinal thickness (FRT) compared to WT. There were fewer cones in IRBP KO mice. Overall, these results confirm substantial loss of rods and significant loss of cones within 30 days. Absence of IRBP resulted in cone circuit damage, reducing photopic flicker, contrast sensitivity, and spatial frequency sensitivity. The c-wave was reduced and accelerated in response to bright steps of light. This result also suggests altered retinal pigment epithelium activity. There appears to be a compensatory mechanism such as higher synaptic gain between PRs and bipolar cells since the loss of the b-wave did not linearly follow the loss of rods, or the a-wave. Scotopic CFF is normal despite thinning of ONL and reduced scotopic electroretinogram (ERG) in IRBP KO mice, suggesting either a redundancy or plasticity in circuits detecting (encoding) scotopic flicker at threshold even with substantial rod loss.
Collapse
Affiliation(s)
| | | | | | | | - Timothy W. Kraft
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.V.J.); (J.M.E.); (P.K.)
| |
Collapse
|
7
|
Towards a New Biomarker for Diabetic Retinopathy: Exploring RBP3 Structure and Retinoids Binding for Functional Imaging of Eyes In Vivo. Int J Mol Sci 2023; 24:ijms24054408. [PMID: 36901838 PMCID: PMC10002987 DOI: 10.3390/ijms24054408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Diabetic retinopathy (DR) is a severe disease with a growing number of afflicted patients, which places a heavy burden on society, both socially and financially. While there are treatments available, they are not always effective and are usually administered when the disease is already at a developed stage with visible clinical manifestation. However, homeostasis at a molecular level is disrupted before visible signs of the disease are evident. Thus, there has been a constant search for effective biomarkers that could signal the onset of DR. There is evidence that early detection and prompt disease control are effective in preventing or slowing DR progression. Here, we review some of the molecular changes that occur before clinical manifestations are observable. As a possible new biomarker, we focus on retinol binding protein 3 (RBP3). We argue that it displays unique features that make it a very good biomarker for non-invasive, early-stage DR detection. Linking chemistry to biological function and focusing on new developments in eye imaging and two-photon technology, we describe a new potential diagnostic tool that would allow rapid and effective quantification of RBP3 in the retina. Moreover, this tool would also be useful in the future to monitor therapeutic effectiveness if levels of RBP3 are elevated by DR treatments.
Collapse
|
8
|
Preclinical Models of Retinitis Pigmentosa. Methods Mol Biol 2022; 2560:181-215. [PMID: 36481897 DOI: 10.1007/978-1-0716-2651-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Retinitis pigmentosa (RP) is the name for a group of phenotypically-related heritable retinal degenerative disorders. Many genes have been implicated as causing variants of RP, and while the clinical phenotypes are remarkably similar, they may differ in age of onset, progression, and severity. Common inheritance patterns for specific genes connected with the development of the disorder include autosomal dominant, autosomal recessive, and X-linked. Modeling the disease in animals and other preclinical systems offers a cost-conscious, ethical, and time-efficient method for studying the disease subtypes. The history of RP models is briefly examined, and both naturally occurring and transgenic preclinical models of RP in many different organisms are discussed. Syndromic forms of RP and models thereof are reviewed as well.
Collapse
|
9
|
Fan J, Rajapakse D, Peterson K, Lerner J, Parsa S, Ponduri A, Sagar V, Duncan T, Dong L, Wistow G. Retbindin mediates light-damage in mouse retina while its absence leads to premature retinal aging. Exp Eye Res 2021; 209:108698. [PMID: 34228964 DOI: 10.1016/j.exer.2021.108698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
Vision requires the transport and recycling of the pigment 11-cis retinaldehyde (retinal) between the retinal pigment epithelium (RPE) and photoreceptors. 11-cis retinal is also required for light-mediated photoreceptor death in dark-adapted mouse eye, probably through overstimulation of rod cells adapted for low light. Retbindin is a photoreceptor-specific protein, of unclear function, that is localized between the RPE and the tips of the photoreceptors. Unexpectedly, young Rtbdn-KO mice, with targeted deletion (KO) of retbindin, showed delayed regeneration of retinal function after bleaching and were strongly resistant to light-induced photoreceptor death. Furthermore, bio-layer interferometry binding studies showed recombinant retbindin had significant affinity for retinoids, most notably 11-cis retinal. This suggests that retbindin mediates light damage, probably through a role in transport of 11-cis retinal. In Rtbdn-KO mice, retinal development was normal, as were amplitudes of rod and cone electroretinograms (ERG) up to 4 months, although implicit times and c-waves were affected. However, with aging, both light- and dark-adapted ERG amplitudes declined significantly and photoreceptor outer segments became disordered, However, in contrast to other reports, there was little retinal degeneration or drop in flavin levels. The RPE developed vacuoles and lipid, protein and calcium deposits reminiscent of age-related macular degeneration. Other signs of premature aging included loss of OPN4+ retinal ganglion cells and activation of microglia. Thus, retbindin plays an unexpected role in the mammalian visual cycle, probably as an adaptation for vision in dim light. It mediates light damage in the dark-adapted eye, but also plays a role in light-adapted responses and in long term retinal homeostasis.
Collapse
Affiliation(s)
- Jianguo Fan
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dinusha Rajapakse
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katherine Peterson
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Lerner
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shabnam Parsa
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arjun Ponduri
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vatsala Sagar
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Todd Duncan
- Laboratory of Retinal Cell and Molecular Biology, 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
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Diabetic retinal neurodegeneration as a form of diabetic retinopathy. Int Ophthalmol 2021; 41:3223-3248. [PMID: 33954860 DOI: 10.1007/s10792-021-01864-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To review the evidence supporting diabetic retinal neurodegeneration (DRN) as a form of diabetic retinopathy. METHOD Review of literature. RESULTS DRN is recognized to be a part of retinopathy in patients with diabetes mellitus (DM), in addition to the well-established diabetic retinal vasculopathy (DRV). DRN has been noted in the early stages of DM, before the onset of clinically evident diabetic retinopathy. The occurrence of DRN has been confirmed in animal models of DM, histopathological examination of donor's eyes from diabetic individuals and assessment of neural structure and function in humans. DRN involves alterations in retinal ganglion cells, photoreceptors, amacrine cells and bipolar cells, and is thought to be driven by glutamate, oxidative stress and dysregulation of neuroprotective factors in the retina. Potential therapeutic options for DRN are under evaluation. CONCLUSIONS Literature is divided on the temporal relation between DRN and DRV, with evidence of both precedence and simultaneous occurrence. The relationship between DRN and multi-system neuropathy in DM is yet to be evaluated critically.
Collapse
|
11
|
Zeng S, Zhang T, Madigan MC, Fernando N, Aggio-Bruce R, Zhou F, Pierce M, Chen Y, Huang L, Natoli R, Gillies MC, Zhu L. Interphotoreceptor Retinoid-Binding Protein (IRBP) in Retinal Health and Disease. Front Cell Neurosci 2020; 14:577935. [PMID: 33328889 PMCID: PMC7710524 DOI: 10.3389/fncel.2020.577935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP), also known as retinol binding protein 3 (RBP3), is a lipophilic glycoprotein specifically secreted by photoreceptors. Enriched in the interphotoreceptor matrix (IPM) and recycled by the retinal pigment epithelium (RPE), IRBP is essential for the vision of all vertebrates as it facilitates the transfer of retinoids in the visual cycle. It also helps to transport lipids between the RPE and photoreceptors. The thiol-dependent antioxidant activity of IRBP maintains the delicate redox balance in the normal retina. Thus, its dysfunction is suspected to play a role in many retinal diseases. We have reviewed here the latest research on IRBP in both retinal health and disease, including the function and regulation of IRBP under retinal stress in both animal models and the human retina. We have also explored the therapeutic potential of targeting IRBP in retinal diseases. Although some technical barriers remain, it is possible that manipulating the expression of IRBP in the retina will rescue or prevent photoreceptor degeneration in many retinal diseases.
Collapse
Affiliation(s)
- Shaoxue Zeng
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Zhang
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Michele C Madigan
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia.,The Australian National University Medical School, The Australian National University, Acton, ACT, Australia
| | - Fanfan Zhou
- Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
| | - Matthew Pierce
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Yingying Chen
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Lianlin Huang
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Acton, ACT, Australia.,The Australian National University Medical School, The Australian National University, Acton, ACT, Australia
| | - Mark C Gillies
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Liu YH, Mölzer C, Makinen K, Kamoi K, Corbett CLC, Klaska IP, Reid DM, Wilson HM, Kuffová L, Cornall RJ, Forrester JV. Treatment With FoxP3+ Antigen-Experienced T Regulatory Cells Arrests Progressive Retinal Damage in a Spontaneous Model of Uveitis. Front Immunol 2020; 11:2071. [PMID: 33013877 PMCID: PMC7498671 DOI: 10.3389/fimmu.2020.02071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/29/2020] [Indexed: 01/17/2023] Open
Abstract
We specify the clinical features of a spontaneous experimental autoimmune uveitis (EAU) model, in which foreign hen-egg lysozyme (HEL) is expressed in the retina, controlled by the promoter for interphotoreceptor retinol binding protein (IRBP). We previously reported 100% P21 (post-partum day) IRBP:HEL single transgenic (sTg) mice, when crossed to transgenic T cell receptor mice (3A9) generating the double transgenic (dTg) genotype, develop EAU despite profound lymphopenia (thymic HEL-specific T cell deletion). In this work, we characterized the immune component of this model and found conventional dTg CD4+ T cells were less anergic than those from 3A9 controls. Furthermore, prior in vitro HEL-activation of 3A9 anergic T cells (Tan) rendered them uveitogenic upon adoptive transfer (Tx) to sTg mice, while antigen-experienced (AgX, dTg), but not naïve (3A9) T cells halted disease in P21 dTg mice. Flow cytometric analysis of the AgX cells elucidated the underlying pathology: FoxP3+CD25hiCD4+ T regulatory cells (Treg) comprised ∼18%, while FR4+CD73+FoxP3-CD25lo/–CD4+ Tan comprised ∼1.2% of total cells. Further Treg-enrichment (∼80%) of the AgX population indicated FoxP3+CD25hiCD4+ Treg played a key role in EAU-suppression while FoxP3-CD25lo/–CD4+ T cells did not. Here we present the novel concept of dual immunological tolerance where spontaneous EAU is due to escape from anergy with consequent failure of Treg induction and subsequent imbalance in the [Treg:Teffector] cell ratio. The reduced numbers of Tan, normally sustaining Treg to prevent autoimmunity, are the trigger for disease, while immune homeostasis can be restored by supplementation with AgX, but not naïve, antigen-specific Treg.
Collapse
Affiliation(s)
- Yi-Hsia Liu
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Christine Mölzer
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Kimmo Makinen
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Koju Kamoi
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Clare L C Corbett
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Izabela P Klaska
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Delyth M Reid
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Heather M Wilson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Lucia Kuffová
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Richard J Cornall
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
13
|
Yokomizo H, Maeda Y, Park K, Clermont AC, Hernandez SL, Fickweiler W, Li Q, Wang CH, Paniagua SM, Simao F, Ishikado A, Sun B, Wu IH, Katagiri S, Pober DM, Tinsley LJ, Avery RL, Feener EP, Kern TS, Keenan HA, Aiello LP, Sun JK, King GL. Retinol binding protein 3 is increased in the retina of patients with diabetes resistant to diabetic retinopathy. Sci Transl Med 2020; 11:11/499/eaau6627. [PMID: 31270273 DOI: 10.1126/scitranslmed.aau6627] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/18/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
The Joslin Medalist Study characterized people affected with type 1 diabetes for 50 years or longer. More than 35% of these individuals exhibit no to mild diabetic retinopathy (DR), independent of glycemic control, suggesting the presence of endogenous protective factors against DR in a subpopulation of patients. Proteomic analysis of retina and vitreous identified retinol binding protein 3 (RBP3), a retinol transport protein secreted mainly by the photoreceptors, as elevated in Medalist patients protected from advanced DR. Mass spectrometry and protein expression analysis identified an inverse association between vitreous RBP3 concentration and DR severity. Intravitreal injection and photoreceptor-specific overexpression of RBP3 in rodents inhibited the detrimental effects of vascular endothelial growth factor (VEGF). Mechanistically, our results showed that recombinant RBP3 exerted the therapeutic effects by binding and inhibiting VEGF receptor tyrosine phosphorylation. In addition, by binding to glucose transporter 1 (GLUT1) and decreasing glucose uptake, RBP3 blocked the detrimental effects of hyperglycemia in inducing inflammatory cytokines in retinal endothelial and Müller cells. Elevated expression of photoreceptor-secreted RBP3 may have a role in protection against the progression of DR due to hyperglycemia by inhibiting glucose uptake via GLUT1 and decreasing the expression of inflammatory cytokines and VEGF.
Collapse
Affiliation(s)
- Hisashi Yokomizo
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Yasutaka Maeda
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Kyoungmin Park
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Allen C Clermont
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | - Ward Fickweiler
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Qian Li
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Chih-Hao Wang
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | - Fabricio Simao
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Atsushi Ishikado
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Bei Sun
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - I-Hsien Wu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Sayaka Katagiri
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - David M Pober
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Liane J Tinsley
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Robert L Avery
- California Retina Consultants, Santa Barbara, CA 93103, USA
| | - Edward P Feener
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA 92697, USA
| | - Hillary A Keenan
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Lloyd Paul Aiello
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Jennifer K Sun
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA.,Beetham Eye Institute, Joslin Diabetes Center, Boston, MA 02215, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - George L King
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA. .,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
14
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:cells9040931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| |
Collapse
|
15
|
Blond F, Léveillard T. Functional Genomics of the Retina to Elucidate its Construction and Deconstruction. Int J Mol Sci 2019; 20:E4922. [PMID: 31590277 PMCID: PMC6801968 DOI: 10.3390/ijms20194922] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
The retina is the light sensitive part of the eye and nervous tissue that have been used extensively to characterize the function of the central nervous system. The retina has a central position both in fundamental biology and in the physiopathology of neurodegenerative diseases. We address the contribution of functional genomics to the understanding of retinal biology by reviewing key events in their historical perspective as an introduction to major findings that were obtained through the study of the retina using genomics, transcriptomics and proteomics. We illustrate our purpose by showing that most of the genes of interest for retinal development and those involved in inherited retinal degenerations have a restricted expression to the retina and most particularly to photoreceptors cells. We show that the exponential growth of data generated by functional genomics is a future challenge not only in terms of storage but also in terms of accessibility to the scientific community of retinal biologists in the future. Finally, we emphasize on novel perspectives that emerge from the development of redox-proteomics, the new frontier in retinal biology.
Collapse
Affiliation(s)
- Frédéric Blond
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
16
|
Liu YH, Corbett C, Klaska IP, Makinen K, Nickerson JM, Cornall RJ, Kuffova L, Forrester JV. Partial retinal photoreceptor loss in a transgenic mouse model associated with reduced levels of interphotoreceptor retinol binding protein (IRBP, RBP3). Exp Eye Res 2018; 172:54-65. [PMID: 29571629 DOI: 10.1016/j.exer.2018.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Organ-specific transgenic membrane expression of hen egg lysozyme (HEL) as a "neo-self antigen" has been used in several models to study immunological tolerance. In this study we report the changes which occur in the B10.BR mouse retina when membrane-bound HEL is expressed in photoreceptors under the control of the promoter for interphotoreceptor retinoid binding protein (IRBP, RBP3). On direct clinical examination of the single transgenic (sTg-IRBP:HEL) mouse fundus, a low-level increase in retinal degeneration compared to non-transgenic controls was observed, presenting as drusenoid deposits and occasional small patches of atrophy. On histological examination, there was an overall shortening of outer segments and loss of photoreceptor nuclei in sTg-IRBP:HEL mice, which was more pronounced in the retinal periphery, particularly inferiorly. The fundoscopically observed lesions did not correlate with the photoreceptor shortening/loss but appeared to be located at the level of the retinal pigment epithelium/choriocapillaris layer and were an exaggeration in size and number of similar age-related changes found in wild type (WT) mice. In addition, neither the atrophic lesions nor the photoreceptor shortening were associated with common retinal degeneration genes, nor were they caused by exposure to light damage since mice housed at both high and low ambient light levels had similar degrees of retinal degeneration. Instead, sTg-IRBP:HEL mice expressed reduced levels of soluble retinal IRBP compared to WT mice which were present from postnatal day16 (P16) and preceded development of photoreceptor shortening (onset P21). We propose that insertion of the HEL transgene in the photoreceptor membrane disrupted normal photoreceptor function and led to reduced levels of soluble IRBP and retinal thinning. A similar phenotype has been observed in IRBP deficient mice. Despite the retinal thinning, the amount of HEL expressed in the retina was sufficient to act as an autoantigenic target when the mice were crossed to the HEL T cell receptor Tg mouse, since double transgenic (dTg-IRBP:HEL) mice spontaneously developed a severe uveoretinitis with onset at weaning. We suggest that, although membrane expression of foreign transgene products is likely to modify the structure and function of tissues and cells, the technology provides useful models to investigate mechanisms of antigen-specific immunological tolerance.
Collapse
Affiliation(s)
- Yi-Hsia Liu
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Clare Corbett
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Izabela P Klaska
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Institute of Ophthalmology, University College London, London, UK
| | - Kimmo Makinen
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Human Health, Novozymes A/S, Bagsvaerd, Denmark
| | | | | | - Lucia Kuffova
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; Department of Ophthalmology, NHS Grampian, Aberdeen, UK
| | - John V Forrester
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; University of Western Australia, Lions Eye Institute, Perth, Western Australia, Australia.
| |
Collapse
|
17
|
Abstract
Genetic mouse models mimicking human diseases have been developed and utilized for retinal research in various topics, involving anatomy, physiology, biochemistry, and pathology. The main reasons why mouse models are important for retinal research include that rodents share a key retinal homology with humans and that genetic manipulation is relatively easily applicable for mice. Here, we describe genetic mouse models, which are categorized with functions in the retina and relationship with human diseases.
Collapse
Affiliation(s)
- Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Tadao Maeda
- Research Division, Kobe Research Institute, HEALIOS K.K., Kobe, Japan.
| |
Collapse
|
18
|
Cook JD, Ng SY, Lloyd M, Eddington S, Sun H, Nathans J, Bok D, Radu RA, Travis GH. Peropsin modulates transit of vitamin A from retina to retinal pigment epithelium. J Biol Chem 2017; 292:21407-21416. [PMID: 29109151 DOI: 10.1074/jbc.m117.812701] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/31/2017] [Indexed: 11/06/2022] Open
Abstract
Peropsin is a non-visual opsin in both vertebrate and invertebrate species. In mammals, peropsin is present in the apical microvilli of retinal pigment epithelial (RPE) cells. These structures interdigitate with the outer segments of rod and cone photoreceptor cells. RPE cells play critical roles in the maintenance of photoreceptors, including the recycling of visual chromophore for the opsin visual pigments. Here, we sought to identify the function of peropsin in the mouse eye. To this end, we generated mice with a null mutation in the peropsin gene (Rrh). These mice exhibited normal retinal histology, normal morphology of outer segments and RPE cells, and no evidence of photoreceptor degeneration. Biochemically, Rrh-/- mice had ∼2-fold higher vitamin A (all-trans-retinol (all-trans-ROL)) in the neural retina following a photobleach and 5-fold lower retinyl esters in the RPE. This phenotype was similar to those reported in mice that lack interphotoreceptor retinoid-binding protein (IRBP) or cellular retinol-binding protein, suggesting that peropsin plays a role in the movement of all-trans-ROL from photoreceptors to the RPE. We compared the phenotypes in mice lacking both peropsin and IRBP with those of mice lacking peropsin or IRBP alone and found that the retinoid phenotype was similarly severe in each of these knock-out mice. We conclude that peropsin controls all-trans-ROL movement from the retina to the RPE or may regulate all-trans-ROL storage within the RPE. We propose that peropsin affects light-dependent regulation of all-trans-ROL uptake from photoreceptors into RPE cells through an as yet undefined mechanism.
Collapse
Affiliation(s)
- Jeremy D Cook
- From the Department of Ophthalmology, Stein Eye Institute
| | - Sze Yin Ng
- From the Department of Ophthalmology, Stein Eye Institute
| | - Marcia Lloyd
- From the Department of Ophthalmology, Stein Eye Institute
| | | | - Hui Sun
- From the Department of Ophthalmology, Stein Eye Institute.,Department of Physiology, and
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Neuroscience, and Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, and.,Howard Hughes Medical Institute, Baltimore, Maryland 21205
| | - Dean Bok
- From the Department of Ophthalmology, Stein Eye Institute
| | - Roxana A Radu
- From the Department of Ophthalmology, Stein Eye Institute
| | - Gabriel H Travis
- From the Department of Ophthalmology, Stein Eye Institute, .,Department of Biological Chemistry, School of Medicine, UCLA, Los Angeles, California 90095
| |
Collapse
|
19
|
Malechka VV, Moiseyev G, Takahashi Y, Shin Y, Ma JX. Impaired Rhodopsin Generation in the Rat Model of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2222-2231. [PMID: 28734946 PMCID: PMC5809515 DOI: 10.1016/j.ajpath.2017.06.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy is a common complication of diabetes mellitus. Diabetic patients experience functional deficits in dark adaptation, contrast sensitivity, and color perception before microvascular pathologies become apparent. Herein, we evaluated early changes in neural retinal function and in retinoid metabolism in the eye in diabetes. Streptozotocin-induced diabetic rats showed decreased a- and b-wave amplitudes of scotopic and photopic electroretinography responses 4 months after diabetes induction compared to nondiabetic controls. Although Western blot analysis revealed no difference in opsin expression, rhodopsin content was decreased in diabetic retinas, as shown by a difference in absorbance. Consistently, levels of 11-cis-retinal, the chromophore for visual pigments, were significantly lower in diabetic retinas compared to those in controls, suggesting a retinoid deficiency. Among visual cycle proteins, interphotoreceptor retinoid-binding protein and stimulated by retinoic acid 6 protein showed significantly lower levels in diabetic rats than those in nondiabetic controls. Similarly, serum levels of retinol-binding protein 4 and retinoids were significantly lower in diabetic rats. Overall, these results suggest that retinoid metabolism in the eye is impaired in type 1 diabetes, which leads to deficient generation of visual pigments and neural retinal dysfunction in early diabetes.
Collapse
Affiliation(s)
- Volha V Malechka
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Gennadiy Moiseyev
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Yusuke Takahashi
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Younghwa Shin
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
20
|
Chen C, Adler L, Goletz P, Gonzalez-Fernandez F, Thompson DA, Koutalos Y. Interphotoreceptor retinoid-binding protein removes all- trans-retinol and retinal from rod outer segments, preventing lipofuscin precursor formation. J Biol Chem 2017; 292:19356-19365. [PMID: 28972139 DOI: 10.1074/jbc.m117.795187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/22/2017] [Indexed: 11/06/2022] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) is a specialized lipophilic carrier that binds the all-trans and 11-cis isomers of retinal and retinol, and this facilitates their transport between photoreceptors and cells in the retina. One of these retinoids, all-trans-retinal, is released in the rod outer segment by photoactivated rhodopsin after light excitation. Following its release, all-trans-retinal is reduced by the retinol dehydrogenase RDH8 to all-trans-retinol in an NADPH-dependent reaction. However, all-trans-retinal can also react with outer segment components, sometimes forming lipofuscin precursors, which after conversion to lipofuscin accumulate in the lysosomes of the retinal pigment epithelium and display cytotoxic effects. Here, we have imaged the fluorescence of all-trans-retinol, all-trans-retinal, and lipofuscin precursors in real time in single isolated mouse rod photoreceptors. We found that IRBP removes all-trans-retinol from individual rod photoreceptors in a concentration-dependent manner. The rate constant for retinol removal increased linearly with IRBP concentration with a slope of 0.012 min-1 μm-1 IRBP also removed all-trans-retinal, but with much less efficacy, indicating that the reduction of retinal to retinol promotes faster clearance of the photoisomerized rhodopsin chromophore. The presence of physiological IRBP concentrations in the extracellular medium resulted in lower levels of all-trans-retinal and retinol in rod outer segments following light exposure. It also prevented light-induced lipofuscin precursor formation, but it did not remove precursors that were already present. These findings reveal an important and previously unappreciated role of IRBP in protecting the photoreceptor cells against the cytotoxic effects of accumulated all-trans-retinal.
Collapse
Affiliation(s)
- Chunhe Chen
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Leopold Adler
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Patrice Goletz
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Federico Gonzalez-Fernandez
- the Departments of Ophthalmology and Pathology, University of Mississippi and G. V. (Sonny) Montgomery Veterans Affairs Medical Centers, Jackson, Mississippi 39216, and
| | - Debra A Thompson
- the Departments of Ophthalmology and Visual Sciences, and Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, Michigan 48105
| | - Yiannis Koutalos
- From the Departments of Ophthalmology and Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425,
| |
Collapse
|
21
|
Broadgate S, Yu J, Downes SM, Halford S. Unravelling the genetics of inherited retinal dystrophies: Past, present and future. Prog Retin Eye Res 2017; 59:53-96. [PMID: 28363849 DOI: 10.1016/j.preteyeres.2017.03.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 02/07/2023]
Abstract
The identification of the genes underlying monogenic diseases has been of interest to clinicians and scientists for many years. Using inherited retinal dystrophies as an example of monogenic disease we describe the history of molecular genetic techniques that have been pivotal in the discovery of disease causing genes. The methods that were developed in the 1970's and 80's are still in use today but have been refined and improved. These techniques enabled the concept of the Human Genome Project to be envisaged and ultimately realised. When the successful conclusion of the project was announced in 2003 many new tools and, as importantly, many collaborations had been developed that facilitated a rapid identification of disease genes. In the post-human genome project era advances in computing power and the clever use of the properties of DNA replication has allowed the development of next-generation sequencing technologies. These methods have revolutionised the identification of disease genes because for the first time there is no need to define the position of the gene in the genome. The use of next generation sequencing in a diagnostic setting has allowed many more patients with an inherited retinal dystrophy to obtain a molecular diagnosis for their disease. The identification of novel genes that have a role in the development or maintenance of retinal function is opening up avenues of research which will lead to the development of new pharmacological and gene therapy approaches. Neither of which can be used unless the defective gene and protein is known. The continued development of sequencing technologies also holds great promise for the advent of truly personalised medicine.
Collapse
Affiliation(s)
- Suzanne Broadgate
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Jing Yu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Susan M Downes
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, OX3 9DU, UK
| | - Stephanie Halford
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK.
| |
Collapse
|
22
|
Markand S, Baskin NL, Chakraborty R, Landis E, Wetzstein SA, Donaldson KJ, Priyadarshani P, Alderson SE, Sidhu CS, Boatright JH, Iuvone PM, Pardue MT, Nickerson JM. IRBP deficiency permits precocious ocular development and myopia. Mol Vis 2016; 22:1291-1308. [PMID: 27829784 PMCID: PMC5082647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/25/2016] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Interphotoreceptor retinoid-binding protein (IRBP) is abundant in the subretinal space and binds retinoids and lipophilic molecules. The expression of IRBP begins precociously early in mouse eye development. IRBP-deficient (KO) mice show less cell death in the inner retinal layers of the retina before eyelid opening compared to wild-type C57BL/6J (WT) controls and eventually develop profound myopia. Thus, IRBP may play a role in eye development before visually-driven phenomena. We report comparative observations during the course of the natural development of eyes in WT and congenic IRBP KO mice that suggest IRBP is necessary at the early stages of mouse eye development for correct function and development to exist in later stages. METHODS We observed the natural development of congenic WT and IRBP KO mice, monitoring several markers of eye size and development, including haze and clarity of optical components in the eye, eye size, axial length, immunohistological markers of differentiation and eye development, visually guided behavior, and levels of a putative eye growth stop signal, dopamine. We conducted these measurements at several ages. Slit-lamp examinations were conducted at post-natal day (P)21. Fundus and spectral domain optical coherence tomography (SD-OCT) images were compared at P15, P30, P45, and P80. Enucleated eyes from P5 to P10 were measured for weight, and ocular dimensions were measured with a noncontact light-emitting diode (LED) micrometer. We counted the cells that expressed tyrosine hydroxylase (TH-positive cells) at P23-P36 using immunohistochemistry on retinal flatmounts. High-performance liquid chromatography (HPLC) was used to analyze the amounts of dopamine (DA) and 3,4-dihydroxyphenylacetic acid (DOPAC) at P7-P60. Monocular form deprivation in the right eye was induced using head-mounted goggles from P28 to P56. RESULTS Eye elongation and eye size in the IRBP KO mice began to increase at P7 compared to the WT mice. This difference increased until P12, and the difference was maintained thereafter. SD-OCT images in live mice confirmed previously reported retinal thinning of the outer nuclear layer in the IRBP KO mice compared to the WT mice from P15 to P80. Slit-lamp and fundoscopy examination outcomes did not differ between the WT and KO mice. SD-OCT measurements of the optical axis components showed that the only factor contributing to excess optical axis length was the depth of the vitreous body. No other component of optical axis length (including corneal thickness, anterior chamber depth, and lens thickness) was different from that of the WT mouse. The refractive power of the IRBP KO mice did not change in response to form deprivation. The number of retinal TH-positive cells was 28% greater in the IRBP KO retinas compared to the WT mice at P30. No significant differences were observed in the steady-state retinal DA or DOPAC levels or in the DOPAC/DA ratios between the WT and IRBP KO mice. CONCLUSIONS The IRBP KO mouse eye underwent precocious development and rapid eye size growth temporally about a day sooner than the WT mouse eye. Eye size began to differ between the WT and KO mice before eyelid opening, indicating no requirement for focus-dependent vision, and suggesting a developmental abnormality in the IRBP KO mouse eye that precedes form vision-dependent emmetropization. Additionally, the profoundly myopic KO eye did not respond to form deprivation compared to the non-deprived contralateral eye. Too much growth occurred in some parts of the eye, possibly upsetting a balance among size, differentiation, and focus-dependent growth suppression. Thus, the loss of IRBP may simply cause growth that is too rapid, possibly due to a lack of sequestration or buffering of morphogens that normally would bind to IRBP but are unbound in the IRBP KO eye. Despite the development of profound myopia, the DA levels in the IRBP KO mice were not statistically different from those in the WT mice, even with the excess of TH-positive cells in the IRBP KO mice compared to the WT mice. Overall, these data suggest that abnormal eye elongation in the IRBP KO mouse is independent of, precedes, and is epistatic to the process(es) of visually-driven refractive development.
Collapse
Affiliation(s)
- Shanu Markand
- Department of Ophthalmology, Emory University, Atlanta, GA
| | | | - Ranjay Chakraborty
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA
| | - Erica Landis
- Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA,Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA
| | | | | | | | | | | | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA
| | - P. Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, GA,Department of Pharmacology, Emory University, Atlanta, GA
| | - Machelle T. Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA,Atlanta Veterans Administration Center of Visual and Neurocognitive Rehabilitation, Decatur, GA,Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA,Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | | |
Collapse
|
23
|
Ghosh D, Haswell KM, Sprada M, Gonzalez-Fernandez F. Fold conservation and proteolysis in zebrafish IRBP structure: Clues to possible enzymatic function? Exp Eye Res 2016; 147:78-84. [PMID: 27155144 DOI: 10.1016/j.exer.2016.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 12/24/2022]
Abstract
Multiple functions for Interphotoreceptor Retinoid-Binding Protein (IRBP) may explain its localization in the retina, vitreous and pineal gland and association with retinitis pigmentosa and myopia. We have been engaged in uncovering the structure-function relationships of this interesting protein long thought to bind visual-cycle retinoids and fatty acids in the subretinal space. Although hydrophobic domains capable of binding such ligands have now been found, we ask what other structural domains might be present that could predict new functions? Interestingly, IRBP possesses a fold similar to C-terminal processing proteases (CTPases) but is missing the PDZ domain. Here we present structural evidence that this fold may have a role in a recently observed autoproteolytic activity of the two-module zebrafish (z) IRBP (Ghosh et al. Exp. Eye Res., 2015). When the structure of Scenedesmus obliquus D1 CTPase (D1P) is superimposed with the first module of zIRBP (z1), the PDZ domain of D1P occupies roughly the same position in the amino acid sequence as the inter-domain tether in z1, between residues P71 and P85. The catalytic triad K397, S372 and E375 of D1P is located at the inter-domain interfacial cleft, similarly as the tetrad K241, S243, D177 and T179 of z1 residues, presumed to have proteolytic function. Packing of two adjacent symmetry-related molecules within the z1 crystal show that the helix α8 penetrates the interfacial cleft underneath the inter-domain tether, forming a simple intermolecular "knot". The full-length zIRBP is cleaved at or immediately after T309, which is located at the end of α8 and is the ninth residue of the second module z2. We propose that the helix α8 within intact zIRBP bends at P301, away from the improbable knotted fold, and positions the cleavage site T309 near the putative catalytic tetrad of the neighboring zIRBP to be proteolytically cleaved. The conservation of this functional catalytic domain suggests that possible physiological roles of IRBP as a hydrolase needs to be considered.
Collapse
Affiliation(s)
- Debashis Ghosh
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Karen M Haswell
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Molly Sprada
- SUNY Eye Institute, State University of New York, Buffalo, NY, USA
| | - Federico Gonzalez-Fernandez
- Research & Development Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA; Departments of Ophthalmology and Pathology, University of Mississippi Medical Center, Jackson, MS, USA; SUNY Eye Institute, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
24
|
Hernández C, Dal Monte M, Simó R, Casini G. Neuroprotection as a Therapeutic Target for Diabetic Retinopathy. J Diabetes Res 2016; 2016:9508541. [PMID: 27123463 PMCID: PMC4830713 DOI: 10.1155/2016/9508541] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/29/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is a multifactorial progressive disease of the retina and a leading cause of vision loss. DR has long been regarded as a vascular disorder, although neuronal death and visual impairment appear before vascular lesions, suggesting an important role played by neurodegeneration in DR and the appropriateness of neuroprotective strategies. Upregulation of vascular endothelial growth factor (VEGF), the main target of current therapies, is likely to be one of the first responses to retinal hyperglycemic stress and VEGF may represent an important survival factor in early phases of DR. Of central importance for clinical trials is the detection of retinal neurodegeneration in the clinical setting, and spectral domain optical coherence tomography seems the most indicated technique. Many substances have been tested in animal studies for their neuroprotective properties and for possible use in humans. Perhaps, the most intriguing perspective is the use of endogenous neuroprotective substances or nutraceuticals. Together, the data point to the central role of neurodegeneration in the pathogenesis of DR and indicate neuroprotection as an effective strategy for treating this disease. However, clinical trials to determine not only the effectiveness and safety but also the compliance of a noninvasive route of drug administration are needed.
Collapse
Affiliation(s)
- Cristina Hernández
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabolicas Asociadas) and Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autonoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
- *Cristina Hernández: and
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy
| | - Rafael Simó
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabolicas Asociadas) and Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autonoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Giovanni Casini
- Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy
- *Giovanni Casini:
| |
Collapse
|
25
|
Horai R, Zárate-Bladés CR, Dillenburg-Pilla P, Chen J, Kielczewski JL, Silver PB, Jittayasothorn Y, Chan CC, Yamane H, Honda K, Caspi RR. Microbiota-Dependent Activation of an Autoreactive T Cell Receptor Provokes Autoimmunity in an Immunologically Privileged Site. Immunity 2015; 43:343-53. [PMID: 26287682 DOI: 10.1016/j.immuni.2015.07.014] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/30/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022]
Abstract
Activated retina-specific T cells that have acquired the ability to break through the blood-retinal barrier are thought to be causally involved in autoimmune uveitis, a major cause of human blindness. It is unclear where these autoreactive T cells first become activated, given that their cognate antigens are sequestered within the immune-privileged eye. We demonstrate in a novel mouse model of spontaneous uveitis that activation of retina-specific T cells is dependent on gut commensal microbiota. Retina-specific T cell activation involved signaling through the autoreactive T cell receptor (TCR) in response to non-cognate antigen in the intestine and was independent of the endogenous retinal autoantigen. Our findings not only have implications for the etiology of human uveitis, but also raise the possibility that activation of autoreactive TCRs by commensal microbes might be a more common trigger of autoimmune diseases than is currently appreciated.
Collapse
Affiliation(s)
- Reiko Horai
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | | | - Patricia Dillenburg-Pilla
- Oral and Pharyngeal Cancer Branch, National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Jun Chen
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA; State Key of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | | | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | | | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Hidehiro Yamane
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Kenya Honda
- Keio University School of Medicine, Tokyo 160-8582, Japan; RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Simó R, Hernández C. Novel approaches for treating diabetic retinopathy based on recent pathogenic evidence. Prog Retin Eye Res 2015; 48:160-80. [PMID: 25936649 DOI: 10.1016/j.preteyeres.2015.04.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/13/2015] [Accepted: 04/21/2015] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy remains as a leading cause of blindness in developed countries. Current treatments target late stages of DR when vision has already been significantly affected. A better understanding of the pathogenesis of DR would permit the development of more efficient preventional/interventional strategies against early stages of DR. In this article a critical review of the state of the art of this issue is provided along with a discussion of problems which have yet to be overcome. Neuroprotection as a new approach for the treatment of the early stages of DR has been particularly emphasized. The development and progression of DR is not homogeneous and, apart from blood glucose levels and blood pressure, it depends on genetic factors which remain to be elucidated. In addition, the role of the pathogenic pathways is not the same in all patients. All these factors should be taken into account in the near future when an individualized oriented treatment for DR could become feasible. The new techniques in retinal imaging acquisition, the identification of useful circulating biomarkers and the individualized analysis of biological samples could facilitate the development of early and personalized therapy in the setting of DR. Finally, it should be noted that only a coordinated action among ophthalmologists, diabetologists, basic researchers, experts in pharmaco-economics and health care providers addressed to the design of rational strategies targeting prevention and the early stages of DR will be effective in reducing the burden and improving the clinical outcome of this devastating complication of diabetes.
Collapse
Affiliation(s)
- Rafael Simó
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain.
| | - Cristina Hernández
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain.
| |
Collapse
|
27
|
Zhu L, Shen W, Lyons B, Wang Y, Zhou F, Gillies MC. Dysregulation of inter-photoreceptor retinoid-binding protein (IRBP) after induced Müller cell disruption. J Neurochem 2015; 133:909-18. [PMID: 25692504 DOI: 10.1111/jnc.13075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 11/29/2022]
Abstract
Reduced expression of a ~150 kDa protein was unexpectedly observed while investigating Norrin protein in a transgenic murine model in which Müller cells can be selectively and inducibly disrupted. Isolation of this unknown protein via ion exchange and hydrophobic interaction chromatography followed by Tandem mass spectrometry identified it as Inter-photoreceptor retinoid-binding protein (IRBP). Significantly reduced IRBP mRNA expression was observed at the early and late stages after Müller cell disruption. IRBP protein expression was also consistently reduced to 5.7% of the control level as early as 1 week after Müller cell disruption. This down-regulation of IRBP was accompanied by focal hyperfluorescent dots and cytotoxic N-retinylidene-N-retinylethanolamine (A2E) accumulation. In vitro treatment of cone photoreceptor cell lines with conditioned medium collected from stressed Müller cells suggested that Müller cells regulated photoreceptors expression of IRBP via secreted factor(s). In vivo studies suggested that one of these secreted factors was tumour necrosis factor alpha (TNFα). These findings suggest that dysregulation of IRBP expression caused by Müller cell dysfunction may be an important early event in photoreceptor degeneration in some retinal diseases. This study reports down-regulation of inter-photoreceptor retinoid-binding protein (IRBP) in photoreceptors and retinoid cycle derangement after Müller cell disruption in a transgenic mouse model. The findings indicate that Müller cells communicate with photoreceptors in response to stress by secreting soluble protein factor(s). We propose that down-regulation of IRBP may represent an early and novel pathogenic mechanism in degenerative retinal diseases.
Collapse
Affiliation(s)
- Ling Zhu
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Weiyong Shen
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Brian Lyons
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Ying Wang
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, the University of Sydney, Sydney, NSW, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Gonzalez-Fernandez F, Betts-Obregon B, Yust B, Mimun J, Sung D, Sardar D, Tsin AT. Interphotoreceptor retinoid-binding protein protects retinoids from photodegradation. Photochem Photobiol 2015; 91:371-8. [PMID: 25565073 DOI: 10.1111/php.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022]
Abstract
Retinol degrades rapidly in light into a variety of photoproducts. It is remarkable that visual cycle retinoids can evade photodegradation as they are exchanged between the photoreceptors, retinal pigment epithelium and Müller glia. Within the interphotoreceptor matrix, all-trans retinol, 11-cis retinol and retinal are bound by interphotoreceptor retinoid-binding protein (IRBP). Apart from its role in retinoid trafficking and targeting, could IRBP have a photoprotective function? HPLC was used to evaluate the ability of IRBP to protect all-trans and 11-cis retinols from photodegradation when exposed to incandescent light (0 to 8842 μW cm(-2)); time periods of 0-60 min, and bIRBP: retinol molar ratios of 1:1 to 1:5. bIRBP afforded a significant prevention of both all-trans and 11-cis retinol to rapid photodegradation. The effect was significant over the entire light intensity range tested, and extended to the bIRBP: retinol ratio 1:5. In view of the continual exposure of the retina to light, and the high oxidative stress in the outer retina, our results suggest IRBP may have an important protective role in the visual cycle by reducing photodegradation of all-trans and 11-cis retinols. This role of IRBP is particularly relevant in the high flux conditions of the cone visual cycle.
Collapse
Affiliation(s)
- Federico Gonzalez-Fernandez
- Medical Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS; Departments of Ophthalmology & Pathology, University of Mississippi School of Medicine, Jackson, MS; SUNY Eye Institute, State University of New York, Buffalo, NY
| | | | | | | | | | | | | |
Collapse
|
29
|
Simó R, Hernández C. Neurodegeneration in the diabetic eye: new insights and therapeutic perspectives. Trends Endocrinol Metab 2014; 25:23-33. [PMID: 24183659 DOI: 10.1016/j.tem.2013.09.005] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/02/2013] [Accepted: 09/27/2013] [Indexed: 11/29/2022]
Abstract
Diabetic retinopathy (DR), one of the leading causes of preventable blindness, has been considered a microcirculatory disease of the retina. However, there is emerging evidence to suggest that retinal neurodegeneration is an early event in the pathogenesis of DR, which participates in the development of microvascular abnormalities. Therefore, the study of the underlying mechanisms leading to neurodegeneration and the identification of the mediators in the crosstalk between neurodegeneration and microangiopathy will be essential for the development of new therapeutic strategies. In this review, an updated discussion of the mechanisms involved in neurodegeneration, as well as the link between neurodegeneration and microangiopathy, is presented. Finally, the therapeutic implications and new perspectives based on identifying those patients with retinal neurodegeneration are given.
Collapse
Affiliation(s)
- Rafael Simó
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain.
| | - Cristina Hernández
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas) and Diabetes and Metabolism Research Unit, Vall Hebron Institut de Recerca (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain
| | | |
Collapse
|
30
|
Kim JY, Zhao H, Martinez J, Doggett TA, Kolesnikov AV, Tang PH, Ablonczy Z, Chan CC, Zhou Z, Green DR, Ferguson TA. Noncanonical autophagy promotes the visual cycle. Cell 2013; 154:365-76. [PMID: 23870125 DOI: 10.1016/j.cell.2013.06.012] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/06/2013] [Accepted: 06/06/2013] [Indexed: 01/08/2023]
Abstract
Phagocytosis and degradation of photoreceptor outer segments (POS) by retinal pigment epithelium (RPE) is fundamental to vision. Autophagy is also responsible for bulk degradation of cellular components, but its role in POS degradation is not well understood. We report that the morning burst of RPE phagocytosis coincided with the enzymatic conversion of autophagy protein LC3 to its lipidated form. LC3 associated with single-membrane phagosomes containing engulfed POS in an Atg5-dependent manner that required Beclin1, but not the autophagy preinitiation complex. The importance of this process was verified in mice with Atg5-deficient RPE cells that showed evidence of disrupted lysosomal processing. These mice also exhibited decreased photoreceptor responses to light stimuli and decreased chromophore levels that were restored with exogenous retinoid supplementation. These results establish that the interplay of phagocytosis and autophagy within the RPE is required for both POS degradation and the maintenance of retinoid levels to support vision.
Collapse
Affiliation(s)
- Ji-Young Kim
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
O'Byrne SM, Blaner WS. Retinol and retinyl esters: biochemistry and physiology. J Lipid Res 2013; 54:1731-43. [PMID: 23625372 PMCID: PMC3679378 DOI: 10.1194/jlr.r037648] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 04/24/2013] [Indexed: 12/23/2022] Open
Abstract
By definition, a vitamin is a substance that must be obtained regularly from the diet. Vitamin A must be acquired from the diet, but unlike most vitamins, it can also be stored within the body in relatively high levels. For humans living in developed nations or animals living in present-day vivariums, stored vitamin A concentrations can become relatively high, reaching levels that can protect against the adverse effects of insufficient vitamin A dietary intake for six months, or even much longer. The ability to accumulate vitamin A stores lessens the need for routinely consuming vitamin A in the diet, and this provides a selective advantage to the organism. The molecular processes that underlie this selective advantage include efficient mechanisms to acquire vitamin A from the diet, efficient and overlapping mechanisms for the transport of vitamin A in the circulation, a specific mechanism allowing for vitamin A storage, and a mechanism for mobilizing vitamin A from these stores in response to tissue needs. These processes are considered in this review.
Collapse
Affiliation(s)
- Sheila M. O'Byrne
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - William S. Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
32
|
Li S, Yang Z, Hu J, Gordon WC, Bazan NG, Haas AL, Bok D, Jin M. Secretory defect and cytotoxicity: the potential disease mechanisms for the retinitis pigmentosa (RP)-associated interphotoreceptor retinoid-binding protein (IRBP). J Biol Chem 2013; 288:11395-406. [PMID: 23486466 PMCID: PMC3630842 DOI: 10.1074/jbc.m112.418251] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) secreted by photoreceptors plays a pivotal role in photoreceptor survival and function. Recently, a D1080N mutation in IRBP was found in patients with retinitis pigmentosa, a frequent cause of retinal degeneration. The molecular and cellular bases for pathogenicity of the mutation are unknown. Here, we show that the mutation abolishes secretion of IRBP and results in formation of insoluble high molecular weight complexes via disulfide bonds. Co-expression of protein disulfide isomerase A2 that regulates disulfide bond formation or introduction of double Cys-to-Ala substitutions at positions 304 and 1175 in D1080N IRBP promoted secretion of the mutated IRBP. D1080N IRBP was not transported to the Golgi apparatus, but accumulated in the endoplasmic reticulum (ER), bound with the ER-resident chaperone proteins such as BiP, protein disulfide isomerase, and heat shock proteins. Splicing of X-box-binding protein-1 mRNA, expression of activating transcription factor 4 (ATF4), and cleavage of ATF6 were significantly increased in cells expressing D1080N IRBP. Moreover, D1080N IRBP induced up-regulation and nuclear translocation of the C/EBP homologous protein, a proapoptotic transcription factor associated with the unfolded protein response. These results indicate that loss of normal function (nonsecretion) and gain of cytotoxic function (ER stress) are involved in the disease mechanisms of D1080N IRBP. Chemical chaperones and low temperature, which help proper folding of many mutated proteins, significantly rescued secretion of D1080N IRBP, suggesting that misfolding is the molecular basis for pathogenicity of D1080N substitution and that chemical chaperones are therapeutic candidates for the mutation-caused blinding disease.
Collapse
Affiliation(s)
- Songhua Li
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Maeda A, Palczewski K. Retinal degeneration in animal models with a defective visual cycle. DRUG DISCOVERY TODAY. DISEASE MODELS 2013; 10:e163-e172. [PMID: 25210527 PMCID: PMC4157753 DOI: 10.1016/j.ddmod.2014.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Continuous generation of visual chromophore through the visual (retinoid) cycle is essential to maintain eyesight and retinal heath. Impairments in this cycle and related pathways adversely affect vision. In this review, we summarize the chemical reactions of vitamin A metabolites involved in the retinoid cycle and describe animal models of associated human diseases. Development of potential therapies for retinal disorders in these animal models is also introduced.
Collapse
Affiliation(s)
- Akiko Maeda
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Krzysztof Palczewski
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
34
|
Tang PH, Kono M, Koutalos Y, Ablonczy Z, Crouch RK. New insights into retinoid metabolism and cycling within the retina. Prog Retin Eye Res 2012; 32:48-63. [PMID: 23063666 DOI: 10.1016/j.preteyeres.2012.09.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 09/28/2012] [Accepted: 09/30/2012] [Indexed: 01/05/2023]
Abstract
The retinoid cycle is a series of biochemical reactions within the eye that is responsible for synthesizing the chromophore, 11-cis retinal, for visual function. The chromophore is bound to G-protein coupled receptors, opsins, within rod and cone photoreceptor cells forming the photosensitive visual pigments. Integral to the sustained function of photoreceptors is the continuous generation of chromophore by the retinoid cycle through two separate processes, one that supplies both rods and cones and another that exclusively supplies cones. Recent findings such as RPE65 localization within cones and the pattern of distribution of retinoid metabolites within mouse and human retinas have challenged previous proposed schemes. This review will focus on recent findings regarding the transport of retinoids, the mechanisms by which chromophore is supplied to both rods and cones, and the metabolism of retinoids within the posterior segment of the eye.
Collapse
Affiliation(s)
- Peter H Tang
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
35
|
Taniguchi RT, DeVoss JJ, Moon JJ, Sidney J, Sette A, Jenkins MK, Anderson MS. Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection. Proc Natl Acad Sci U S A 2012; 109:7847-52. [PMID: 22552229 PMCID: PMC3356674 DOI: 10.1073/pnas.1120607109] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The autoimmune regulator (Aire) plays a critical role in central tolerance by promoting the display of tissue-specific antigens in the thymus. To study the influence of Aire on thymic selection in a physiological setting, we used tetramer reagents to detect autoreactive T cells specific for the Aire-dependent tissue-specific antigen interphotoreceptor retinoid-binding protein (IRBP), in the polyclonal repertoire. Two class II tetramer reagents were designed to identify T cells specific for two different peptide epitopes of IRBP. Analyses of the polyclonal T-cell repertoire showed a high frequency of activated T cells specific for both IRBP tetramers in Aire(-/-) mice, but not in Aire(+/+) mice. Surprisingly, although one tetramer-binding T-cell population was efficiently deleted in the thymus in an Aire-dependent manner, the second tetramer-binding population was not deleted and could be detected in both the Aire(-/-) and Aire(+/+) T-cell repertoires. We found that Aire-dependent thymic deletion of IRBP-specific T cells relies on intercellular transfer of IRBP between thymic stroma and bone marrow-derived antigen-presenting cells. Furthermore, our data suggest that Aire-mediated deletion relies not only on thymic expression of IRBP, but also on proper antigen processing and presentation of IRBP by thymic antigen-presenting cells.
Collapse
Affiliation(s)
| | - Jason J. DeVoss
- Diabetes Center, University of California, San Francisco, CA 94143
| | - James J. Moon
- Department of Microbiology and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
- Center for Immunology and Inflammatory Diseases and Pulmonary and Critical Care Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - John Sidney
- Center for Infectious Disease, Allergy and Asthma Research, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Alessandro Sette
- Center for Infectious Disease, Allergy and Asthma Research, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Marc K. Jenkins
- Department of Microbiology and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Mark S. Anderson
- Diabetes Center, University of California, San Francisco, CA 94143
| |
Collapse
|
36
|
Arroul-Lammali A, Djeraba Z, Belkhelfa M, Belguendouz H, Hartani D, Lahlou-Boukoffa O, Touil-Boukoffa C. Early involvement of nitric oxide in mechanisms of pathogenesis of experimental autoimmune uveitis induced by interphotoreceptor retinoid-binding protein (IRBP). J Fr Ophtalmol 2012; 35:251-9. [DOI: 10.1016/j.jfo.2011.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 05/11/2011] [Accepted: 05/13/2011] [Indexed: 11/30/2022]
|
37
|
Zhou R, Horai R, Silver PB, Mattapallil MJ, Zárate-Bladés CR, Chong WP, Chen J, Rigden RC, Villasmil R, Caspi RR. The living eye "disarms" uncommitted autoreactive T cells by converting them to Foxp3(+) regulatory cells following local antigen recognition. THE JOURNAL OF IMMUNOLOGY 2012; 188:1742-50. [PMID: 22238462 DOI: 10.4049/jimmunol.1102415] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immune privilege is used by the eye, brain, reproductive organs, and gut to preserve structural and functional integrity in the face of inflammation. The eye is arguably the most vulnerable and, therefore, also the most "privileged" of tissues; paradoxically, it remains subject to destructive autoimmunity. It has been proposed, although never proven in vivo, that the eye can induce T regulatory cells (Tregs) locally. Using Foxp3-GFP reporter mice expressing a retina-specific TCR, we now show that uncommitted T cells rapidly convert in the living eye to Foxp3(+) Tregs in a process involving retinal Ag recognition, de novo Foxp3 induction, and proliferation. This takes place within the ocular tissue and is supported by retinoic acid, which is normally present in the eye because of its function in the chemistry of vision. Nonconverted T cells showed evidence of priming but appeared restricted from expressing effector function in the eye. Pre-existing ocular inflammation impeded conversion of uncommitted T cells into Tregs. Importantly, retina-specific T cells primed in vivo before introduction into the eye were resistant to Treg conversion in the ocular environment and, instead, caused severe uveitis. Thus, uncommitted T cells can be disarmed, but immune privilege is unable to protect from uveitogenic T cells that have acquired effector function prior to entering the eye. These findings shed new light on the phenomenon of immune privilege and on its role, as well as its limitations, in actively controlling immune responses in the tissue.
Collapse
Affiliation(s)
- Ru Zhou
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1857, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
den Hollander AI, Black A, Bennett J, Cremers FPM. Lighting a candle in the dark: advances in genetics and gene therapy of recessive retinal dystrophies. J Clin Invest 2010; 120:3042-53. [PMID: 20811160 DOI: 10.1172/jci42258] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Nonsyndromic recessive retinal dystrophies cause severe visual impairment due to the death of photoreceptor and retinal pigment epithelium cells. These diseases until recently have been considered to be incurable. Molecular genetic studies in the last two decades have revealed the underlying molecular causes in approximately two-thirds of patients. The mammalian eye has been at the forefront of therapeutic trials based on gene augmentation in humans with an early-onset nonsyndromic recessive retinal dystrophy due to mutations in the retinal pigment epithelium-specific protein 65kDa (RPE65) gene. Tremendous challenges still lie ahead to extrapolate these studies to other retinal disease-causing genes, as human gene augmentation studies require testing in animal models for each individual gene and sufficiently large patient cohorts for clinical trials remain to be identified through cost-effective mutation screening protocols.
Collapse
Affiliation(s)
- Anneke I den Hollander
- Department of Ophthalmology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | | | | | | |
Collapse
|
39
|
The retinal pigment epithelium: something more than a constituent of the blood-retinal barrier--implications for the pathogenesis of diabetic retinopathy. J Biomed Biotechnol 2010; 2010:190724. [PMID: 20182540 PMCID: PMC2825554 DOI: 10.1155/2010/190724] [Citation(s) in RCA: 311] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/28/2009] [Accepted: 11/16/2009] [Indexed: 12/27/2022] Open
Abstract
The retinal pigment epithelium (RPE) is an specialized epithelium lying in the interface between the neural retina and the choriocapillaris where it forms the outer blood-retinal barrier (BRB). The main functions of the RPE are the following: (1) transport of nutrients, ions, and water, (2) absorption of light and protection against photooxidation, (3) reisomerization of all-trans-retinal into 11-cis-retinal, which is crucial for the visual cycle, (4) phagocytosis of shed photoreceptor membranes, and (5) secretion of essential factors for the structural integrity of the retina. An overview of these functions will be given. Most of the research on the physiopathology of diabetic retinopathy has been focused on the impairment of the neuroretina and the breakdown of the inner BRB. By contrast, the effects of diabetes on the RPE and in particular on its secretory activity have received less attention. In this regard, new therapeutic strategies addressed to modulating RPE impairment are warranted.
Collapse
|
40
|
Parker RO, Crouch RK. The interphotoreceptor retinoid binding (IRBP) is essential for normal retinoid processing in cone photoreceptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 664:141-9. [PMID: 20238012 DOI: 10.1007/978-1-4419-1399-9_17] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
11-cis Retinal is the light-sensitive component in rod and cone photoreceptors, and its isomerization to all-trans retinal in the presence of light initiates the visual response. For photoreceptors to function normally, all-trans retinal must be converted back into 11-cis retinal through the visual cycle. While rods are primarily responsible for dim light vision, the ability of cones to function in constant light is essential to human vision and may be facilitated by cone-specific visual cycle pathways. The interphotoreceptor retinoid-binding protein (IRBP) is a proposed retinoid transporter in the visual cycle, but rods in Irbp ( -/- ) mice have a normal visual cycle. However, there is evidence that IRBP has cone-specific functions. Cone electroretinogram (ERG) responses are reduced, despite having cone densities and opsin levels similar to C57Bl/6 (WT) mice. Treatment with 9-cis retinal rescues the cone response in Irbp ( -/- ) mice and shows that retinoid deficiency underlies cone dysfunction. These data indicate that IRBP is essential to normal cone function and demonstrate that differences exist in the visual cycle of rods and cones.
Collapse
Affiliation(s)
- Ryan O Parker
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA.
| | | |
Collapse
|
41
|
Garcia-Ramírez M, Hernández C, Villarroel M, Canals F, Alonso MA, Fortuny R, Masmiquel L, Navarro A, García-Arumí J, Simó R. Interphotoreceptor retinoid-binding protein (IRBP) is downregulated at early stages of diabetic retinopathy. Diabetologia 2009; 52:2633-41. [PMID: 19823802 DOI: 10.1007/s00125-009-1548-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 09/02/2009] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Interphotoreceptor retinoid-binding protein (IRBP) plays a major role in the visual cycle and is essential to the maintenance of photoreceptors. The aim of this study was to determine whether a decrease in IRBP production exists in the early stages of diabetic retinopathy. METHODS Vitreous samples from diabetic patients with proliferative and non-proliferative diabetic retinopathy (PDR, NPDR), and from non-diabetic patients with macular hole (control group) were selected for IRBP quantitative assessment by proteomic analysis (fluorescence-based difference gel electrophoresis) and western blot. Human post mortem eyes (n = 16) from diabetic donors without clinically detectable retinopathy and from non-diabetic donors (n = 16) were used to determine IRBP (also known as RBP3) mRNA levels (RT-PCR) and protein content (western blot and confocal microscopy). Retinal neurodegeneration was assessed by measuring glial fibrillar acidic protein (GFAP) and the apoptotic rate. Y79 human retinoblastoma cells were used to test the effects of glucose, TNF-alpha and IL-1beta on IRBP expression and IRBP levels. RESULTS Intravitreous IRBP concentration was significantly lower in PDR < NPDR < control in proteomic and western blot analysis. IRBP mRNA levels and IRBP protein content were significantly lower in the retinas from diabetic donors than in those from non-diabetic donors. Increased GFAP and a higher degree of apoptosis were observed in diabetic retinas compared with non-diabetic retinas. A dose-dependent downregulation of IRBP mRNA expression and IRBP content was detected with glucose, TNF-alpha and IL-1beta in cultures of Y79 human retinoblastoma cells. CONCLUSIONS/INTERPRETATION Underproduction of IRBP is an early event in the human diabetic retina and is associated with retinal neurodegeneration. The mechanisms leading to this deficit deserve further investigation.
Collapse
Affiliation(s)
- M Garcia-Ramírez
- CIBER for Diabetes and Associated Metabolic Diseases (CIBERDEM), Diabetes and Metabolism Research Unit, Institut de Recerca Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Huang J, Possin DE, Saari JC. Localizations of visual cycle components in retinal pigment epithelium. Mol Vis 2009; 15:223-34. [PMID: 19180257 PMCID: PMC2632734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 12/31/2008] [Indexed: 10/25/2022] Open
Abstract
PURPOSE We used immunocytochemistry and confocal microscopy to determine whether enzymes of the rod visual cycle were uniformly distributed in retinal pigment epithelium (RPE) cells. The localizations of these enzymes were compared to known localizations of retinoid-binding proteins and associated proteins. METHODS Antibodies to proteins and enzymes associated with the rod visual cycle were used for fluorescence immunocytochemistry with frozen sections of albino mouse and rat retina. Images were obtained with a laser scanning confocal microscope. RESULTS Components associated with the rod visual cycle were distributed in three distinct patterns in mouse and rat RPE. Three visual cycle enzymes (RDH5, LRAT, and RPE65) were restricted to the somata of RPE cells and were not detected within apical processes. Ezrin, an actin-binding protein, and ERM-binding phosphoprotein50/sodium-hydrogen exchanger regulatory factor1 (EBP50/NHERF1), an ezrin-binding PDZ-domain protein, were largely restricted to RPE apical processes. The fluorescence intensity over Müller cell apical processes was less intense. Cellular retinaldehyde-binding protein (CRALBP), which binds to EBP50/NHERF1, and cellular retinol-binding protein type 1 (CRBP1) were found throughout RPE cells and Müller cells. CONCLUSIONS Visual cycle enzymes were confined to the somata of RPE cells and did not occur within the long apical processes, either in dark- or light-adapted animals. Other components previously linked to the visual cycle (EBP50/NHERF1 and ezrin) were largely confined to the apical processes, where they could be associated with release of 11-cis-retinal or uptake of all-trans-retinol. CRALBP and CRBP1 were distributed throughout the RPE cell, where they could mediate diffusion of retinoids between apical processes and somata.
Collapse
Affiliation(s)
- Jing Huang
- Department of Ophthalmology, University of Washington, School of Medicine, Seattle, WA, USA
| | | | | |
Collapse
|
43
|
Nickerson JM, Frey RA, Ciavatta VT, Stenkamp DL. Interphotoreceptor retinoid-binding protein gene structure in tetrapods and teleost fish. Mol Vis 2006; 12:1565-85. [PMID: 17200656 PMCID: PMC2898515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
PURPOSE The interphotoreceptor retinoid-binding protein (IRBP) gene possesses an unusual structure, encoding multiple Repeats, each consisting of about 300 amino acids. Our goals were to gain insight into the function of IRBP, and to test the current model for the evolution of IRBP, in which Repeats were replicated from a simpler ancestral gene. METHODS We employed a bioinformatics approach to analyze IRBP loci in recently completed or near-complete genome sequences of several vertebrates and nonvertebrate chordates. IRBP gene expression in zebrafish was evaluated by reverse transcriptase PCR (RT-PCR) and in situ mRNA hybridizations with gene-specific probes. RESULTS Patterns of exons and introns in the IRBP genes of tetrapods were highly similar, as were predicted amino acid sequences and Repeat structures. IRBP gene structure in teleost fish was more variable, and we report a new gene structure for two species, the Japanese puffer fish (Takifugu rubripes) and the zebrafish (Danio rerio). These teleost genomes contain a two-gene IRBP locus arranged head-to-tail in which the first gene, Gene 1, is intronless and contains a single large exon encoding three complete Repeats. It is followed by a second gene, Gene 2, which corresponds to the previously reported gene consisting of two Repeats spread across four exons and three introns. Each of the two zebrafish genes is transcribed. Gene 2 is expressed in the photoreceptors and RPE, and Gene 1 is expressed in the inner nuclear layer and weakly in the ganglion cell layer. CONCLUSIONS The tetrapod IRBP gene structure is highly conserved while the teleost fish gene structure was a surprise: It appears to be a two-gene locus with distinct Repeat organization in each open reading frame. This gene structure and gene expression data are consistent with possible neofunctionalization or sub-function partitioning of Gene 1 and Gene 2 in the zebrafish. We suggest that the two-gene locus in teleost fish arose as a consequence of either the known whole genome duplication or single gene tandem duplication.
Collapse
Affiliation(s)
- John M Nickerson
- Ophthalmology Department, Emory University, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
44
|
DeVoss J, Hou Y, Johannes K, Lu W, Liou GI, Rinn J, Chang H, Caspi RR, Caspi R, Fong L, Anderson MS. Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. J Exp Med 2006; 203:2727-35. [PMID: 17116738 PMCID: PMC2118158 DOI: 10.1084/jem.20061864] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 10/26/2006] [Indexed: 12/20/2022] Open
Abstract
The expression of self-antigen in the thymus is believed to be responsible for the deletion of autoreactive T lymphocytes, a critical process in the maintenance of unresponsiveness to self. The Autoimmune regulator (Aire) gene, which is defective in the disorder autoimmune polyglandular syndrome type 1, has been shown to promote the thymic expression of self-antigens. A clear link, however, between specific thymic self-antigens and a single autoimmune phenotype in this model has been lacking. We show that autoimmune eye disease in aire-deficient mice develops as a result of loss of thymic expression of a single eye antigen, interphotoreceptor retinoid-binding protein (IRBP). In addition, lack of IRBP expression solely in the thymus, even in the presence of aire expression, is sufficient to trigger spontaneous eye-specific autoimmunity. These results suggest that failure of thymic expression of selective single self-antigens can be sufficient to cause organ-specific autoimmune disease, even in otherwise self-tolerant individuals.
Collapse
Affiliation(s)
- Jason DeVoss
- Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Grajewski RS, Silver PB, Agarwal RK, Su SB, Chan CC, Liou GI, Caspi RR. Endogenous IRBP can be dispensable for generation of natural CD4+CD25+ regulatory T cells that protect from IRBP-induced retinal autoimmunity. ACTA ACUST UNITED AC 2006; 203:851-6. [PMID: 16585264 PMCID: PMC2118294 DOI: 10.1084/jem.20050429] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Susceptibility to experimental autoimmune uveitis (EAU), a model for human uveitis induced in mice with the retinal antigen interphotoreceptor retinoid-binding protein (IRBP), is controlled by “natural” CD4+CD25+ regulatory T (T reg) cells. To examine whether endogenous expression of IRBP is necessary to generate these T reg cells, we studied responses of IRBP knockout (KO) versus wild-type (WT) mice. Unexpectedly, not only WT but also IRBP KO mice immunized with a uveitogenic regimen of IRBP in complete Freund's adjuvant (CFA) exhibited CD25+ regulatory cells that could be depleted by PC61 treatment, which suppressed development of uveitogenic effector T cells and decreased immunological responses to IRBP. These EAU-relevant T reg cells were not IRBP specific, as their activity was not present in IRBP KO mice immunized with IRBP in incomplete Freund's adjuvant (IFA), lacking mycobacteria (whereas the same mice exhibited normal T reg cell activity to retinal arrestin in IFA). We propose that mycobacterial components in CFA activate T reg cells of other specificities to inhibit generation of IRBP-specific effector T cells in a bystander fashion, indicating that effective T reg cells can be antigen nonspecific. Our data also provide the first evidence that generation of specific T reg cells to a native autoantigen in a mouse with a diverse T cell repertoire requires a cognate interaction.
Collapse
Affiliation(s)
- Rafael S Grajewski
- Laboratory of Immunology, National Eye Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Following exposure of our eye to very intense illumination, we experience a greatly elevated visual threshold, that takes tens of minutes to return completely to normal. The slowness of this phenomenon of "dark adaptation" has been studied for many decades, yet is still not fully understood. Here we review the biochemical and physical processes involved in eliminating the products of light absorption from the photoreceptor outer segment, in recycling the released retinoid to its original isomeric form as 11-cis retinal, and in regenerating the visual pigment rhodopsin. Then we analyse the time-course of three aspects of human dark adaptation: the recovery of psychophysical threshold, the recovery of rod photoreceptor circulating current, and the regeneration of rhodopsin. We begin with normal human subjects, and then analyse the recovery in several retinal disorders, including Oguchi disease, vitamin A deficiency, fundus albipunctatus, Bothnia dystrophy and Stargardt disease. We review a large body of evidence showing that the time-course of human dark adaptation and pigment regeneration is determined by the local concentration of 11-cis retinal, and that after a large bleach the recovery is limited by the rate at which 11-cis retinal is delivered to opsin in the bleached rod outer segments. We present a mathematical model that successfully describes a wide range of results in human and other mammals. The theoretical analysis provides a simple means of estimating the relative concentration of free 11-cis retinal in the retina/RPE, in disorders exhibiting slowed dark adaptation, from analysis of psychophysical measurements of threshold recovery or from analysis of pigment regeneration kinetics.
Collapse
Affiliation(s)
- T D Lamb
- Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia.
| | | |
Collapse
|
47
|
Avichezer D, Liou GI, Chan CC, Lewis GM, Wiggert B, Donoso LA, Nickerson JM, Crawford MA, Caspi RR. Interphotoreceptor retinoid-binding protein (IRBP)-deficient C57BL/6 mice have enhanced immunological and immunopathogenic responses to IRBP and an altered recognition of IRBP epitopes. J Autoimmun 2004; 21:185-94. [PMID: 14599843 DOI: 10.1016/j.jaut.2003.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Experimental autoimmune uveitis (EAU) and pinealitis (EAP) can be induced in susceptible mice by immunization with immunologically privileged retinal antigens. In the present study, we analyzed the immunologic and immunopathologic responses of mice deficient in the retinal autoantigen interphotoreceptor retinoid-binding protein (IRBP). The consequences of IRBP deficiency on the T-cell repertoire were also investigated. IRBP+/+, IRBP+/- and IRBP-/- mice on the C57BL/6 background were immunized with IRBP or with a pathogenic epitope, IRBP(1-20) peptide in adjuvant, and were evaluated for disease severity and immunological responses. C57BL/6 IRBP-/- mice were completely resistant to EAU and EAP, and had enhanced immunological responses to IRBP and to its pathogenic peptide 1-20, as compared to their IRBP+/+ counterparts. IRBP-/- mice exhibited an altered IRBP epitope recognition. T cell epitope mapping revealed a response to IRBP peptide 271-290 in IRBP-/- mice, that was absent in the wild type. Primed T cells of IRBP-/- mice transferred an exacerbated form of EAU to nai;ve wild type recipients. A gene-dose effect was evident in that C57BL/6 IRBP+/- mice, exhibited intermediate immunological responses and lower disease scores compared to wild type. We conclude that expression of IRBP in target tissues is a necessary prerequisite for disease induction, excluding other retinoid-binding or vision-related proteins as surrogate targets. Furthermore, endogenous expression of IRBP is directly responsible for lowering the threshold of susceptibility to uveitic disease.
Collapse
Affiliation(s)
- Dody Avichezer
- Laboratory of Immunology, Section of Immunoregulation, National Institutes of Health, 10 Center Drive, 10/10N222, Bethesda, MD 20892-1857, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gonzalez-Fernandez F. Interphotoreceptor retinoid-binding protein--an old gene for new eyes. Vision Res 2004; 43:3021-36. [PMID: 14611938 DOI: 10.1016/j.visres.2003.09.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Evolving 40 times independently, eyes are striking examples of convergent evolution in that 11-cis retinaldehyde is always used for photon capture, yet the mechanism for its regeneration may be dramatically different in between systems. In particular, insects, cephalopods and vertebrates show varying physical separation of the cis-->trans photoisomerization and chromphore reisomerization. In the vertebrate retina, these two processes are actually distributed between different cells. This compartmentalization is made possible by the phylogenetic innovation of the two-layered optic cup of the vertebrate retina. This unprecedented design created the subretinal space as a novel anatomical compartment allowing photoreceptors access to the retinal pigment epithelium (RPE) and Müller cells, the two cell types which share the burden of 11-cis retinoid regeneration. To take advantage of this arrangement, early vertebrates appear to have recruited for retinoid binding, the betabetaalpha-spiral fold proven useful in enoyl-CoA isomerase/hydratases, and the carboxy-terminal proteases for stabilizing hydrophobic ligands. Quadruplication of this functional domain within a single polypeptide lead to the emergence of interphotoreceptor retinoid-binding protein (IRBP). IRBP is the main soluble component of the IPM, and is prevented from diffusing out of the subretinal space because its large size excludes it from the photoreceptor/Müller cell zonulae adheretes. Despite this physical entrapment, IRBP is rapidly turned over within the IPM through a process that coordinates secretion of the protein by the photoreceptors, and its removal from the matrix by RPE and photoreceptor endocytosis. The present review will summarize what is known about the structure and function of IRBP to anticipate future avenues of research.
Collapse
Affiliation(s)
- Federico Gonzalez-Fernandez
- Department of Ophthalmology, State University of New York at Buffalo and Medical Research Service, Veteran's Affairs Medical Center, 3495 Bailey Avenue, Buffalo, NY 14215, USA.
| |
Collapse
|
49
|
Avichezer D, Grajewski RS, Chan CC, Mattapallil MJ, Silver PB, Raber JA, Liou GI, Wiggert B, Lewis GM, Donoso LA, Caspi RR. An immunologically privileged retinal antigen elicits tolerance: major role for central selection mechanisms. J Exp Med 2003; 198:1665-76. [PMID: 14657219 PMCID: PMC2194140 DOI: 10.1084/jem.20030413] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2003] [Accepted: 09/19/2003] [Indexed: 11/04/2022] Open
Abstract
Immunologically privileged retinal antigens can serve as targets of experimental autoimmune uveitis (EAU), a model for human uveitis. The tolerance status of susceptible strains, whose target antigen is not expressed in the thymus at detectable levels, is unclear. Here, we address this issue directly by analyzing the consequences of genetic deficiency versus sufficiency of a uveitogenic retinal antigen, interphotoreceptor retinoid-binding protein (IRBP). IRBP-knockout (KO) and wild-type (WT) mice on a highly EAU-susceptible background were challenged with IRBP. The KO mice had greatly elevated responses to IRBP, an altered recognition of IRBP epitopes, and their primed T cells induced exacerbated disease in WT recipients. Ultrasensitive immunohistochemical staining visualized sparse IRBP-positive cells, undetectable by conventional assays, in thymi of WT (but not of KO) mice. IRBP message was PCR amplified from these cells after microdissection. Thymus transplantation between KO and WT hosts demonstrated that this level of expression is functionally relevant and sets the threshold of immune (and autoimmune) reactivity. Namely, KO recipients of WT thymi generated reduced IRBP-specific responses, and WT recipients of KO thymi developed enhanced responses and a highly exacerbated disease. Repertoire culling and thymus-dependent CD25+ T cells were implicated in this effect. Thus, uveitis-susceptible individuals display a detectable and functionally significant tolerance to their target antigen, in which central mechanisms play a prominent role.
Collapse
Affiliation(s)
- Dody Avichezer
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Besch D, Jägle H, Scholl HPN, Seeliger MW, Zrenner E. Inherited multifocal RPE-diseases: mechanisms for local dysfunction in global retinoid cycle gene defects. Vision Res 2003; 43:3095-108. [PMID: 14611947 DOI: 10.1016/j.visres.2003.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Alterations of retinoid cycle genes are known to cause retinal diseases characterized by focal white dot fundus lesions. Fundus appearances reveal circumscribed RPE-changes, although generalized metabolic defects and global functional abnormalities are present. As a possible explanation, topographic inhomogeneities of the human photoreceptor mosaic and the role of a cone specific visual cycle will be discussed. Due to particular characteristics of photoreceptor subtypes as well as different pathways for photopigment regeneration the metabolic demand of individual RPE cells might differ. In "flecked retina diseases" heterogeneity of metabolic demand in individual RPE cells could therefore be responsible for their multifocal appearance.
Collapse
Affiliation(s)
- Dorothea Besch
- University Eye Hospital, Schleichstr. 12-16, D-72076 Tübingen, Germany.
| | | | | | | | | |
Collapse
|