1
|
Contreras EO, Dearing CG, Ashinhurst CA, Fish BA, Hossain SN, Rey AM, Silva PD, Thompson S. Pupillary reflex and behavioral masking responses to light as functional measures of retinal degeneration in mice. PLoS One 2021; 16:e0244702. [PMID: 33493166 PMCID: PMC7833141 DOI: 10.1371/journal.pone.0244702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/09/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Pre-clinical testing of retinal pathology and treatment efficacy depends on reliable and valid measures of retinal function. The electroretinogram (ERG) and tests of visual acuity are the ideal standard, but can be unmeasurable while useful vision remains. Non-image-forming responses to light such as the pupillary light reflex (PLR) are attractive surrogates. However, it is not clear how accurately such responses reflect changes in visual capability in specific disease models. The purpose of this study was to test whether measures of non-visual responses to light correlate with previously determined visual function in two photoreceptor degenerations. METHODS The sensitivity of masking behavior (light induced changes in running wheel activity) and the PLR were measured in 3-month-old wild-type mice (WT) with intact inner retinal circuitry, Pde6b-rd1/rd1 mice (rd1) with early and rapid loss of rods and cones, and Prph2-Rd2/Rd2 mice (Rd2) with a slower progressive loss of rods and cones. RESULTS In rd1 mice, negative masking had increased sensitivity, positive masking was absent, and the sensitivity of the PLR was severely reduced. In Rd2 mice, positive masking identified useful vision at higher light levels, but there was a limited decrease in the irradiance sensitivity of negative masking and the PLR, and the amplitude of change for both underestimated the reduction in irradiance sensitivity of image-forming vision. CONCLUSIONS Together these data show that in a given disease, two responses to light can be affected in opposite ways, and that for a given response to light, the change in the response does not accurately represent the degree of pathology. However, the extent of the deficit in the PLR means that even a limited rescue of rod/cone function might be measured by increased PLR amplitude. In addition, positive masking has the potential to measure effective treatment in both models by restoring responses or shifting thresholds to lower irradiances.
Collapse
Affiliation(s)
- Ethan O. Contreras
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Carley G. Dearing
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
- College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO, United States of America
| | - Crystal A. Ashinhurst
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Betty A. Fish
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Sajila N. Hossain
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Ariana M. Rey
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Primal D. Silva
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| | - Stewart Thompson
- Department of Psychology, New Mexico Tech, Socorro, NM, United States of America
- Department of Biology, New Mexico Tech, Socorro, NM, United States of America
| |
Collapse
|
2
|
Tam WY, Cheung KK. Phenotypic characteristics of commonly used inbred mouse strains. J Mol Med (Berl) 2020; 98:1215-1234. [PMID: 32712726 DOI: 10.1007/s00109-020-01953-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022]
Abstract
The laboratory mouse is the most commonly used mammalian model for biomedical research. An enormous number of mouse models, such as gene knockout, knockin, and overexpression transgenic mice, have been created over the years. A common practice to maintain a genetically modified mouse line is backcrossing with standard inbred mice over several generations. However, the choice of inbred mouse for backcrossing is critical to phenotypic characterization because phenotypic variabilities are often observed between mice with different genetic backgrounds. In this review, the major features of commonly used inbred mouse lines are discussed. The aim is to provide information for appropriate selection of inbred mouse lines for genetic and behavioral studies.
Collapse
Affiliation(s)
- Wing Yip Tam
- University Research Facility in Behavioral and Systems Neuroscience, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Kwok-Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China.
| |
Collapse
|
3
|
Schnichels S, Paquet-Durand F, Löscher M, Tsai T, Hurst J, Joachim SC, Klettner A. Retina in a dish: Cell cultures, retinal explants and animal models for common diseases of the retina. Prog Retin Eye Res 2020; 81:100880. [PMID: 32721458 DOI: 10.1016/j.preteyeres.2020.100880] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022]
Abstract
For many retinal diseases, including age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), the exact pathogenesis is still unclear. Moreover, the currently available therapeutic options are often unsatisfactory. Research designed to remedy this situation heavily relies on experimental animals. However, animal models often do not faithfully reproduce human disease and, currently, there is strong pressure from society to reduce animal research. Overall, this creates a need for improved disease models to understand pathologies and develop treatment options that, at the same time, require fewer or no experimental animals. Here, we review recent advances in the field of in vitro and ex vivo models for AMD, glaucoma, and DR. We highlight the difficulties associated with studies on complex diseases, in which both the initial trigger and the ensuing pathomechanisms are unclear, and then delineate which model systems are optimal for disease modelling. To this end, we present a variety of model systems, ranging from primary cell cultures, over organotypic cultures and whole eye cultures, to animal models. Specific advantages and disadvantages of such models are discussed, with a special focus on their relevance to putative in vivo disease mechanisms. In many cases, a replacement of in vivo research will mean that several different in vitro models are used in conjunction, for instance to analyze and validate causative molecular pathways. Finally, we argue that the analytical decomposition into appropriate cell and tissue model systems will allow making significant progress in our understanding of complex retinal diseases and may furthermore advance the treatment testing.
Collapse
Affiliation(s)
- Sven Schnichels
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany.
| | - François Paquet-Durand
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Germany
| | - Marina Löscher
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany
| | - Teresa Tsai
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Germany
| | - José Hurst
- University Eye Hospital, Centre for Ophthalmology, University of Tübingen, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Germany
| | - Alexa Klettner
- Department of Ophthalmology, University Medical Center, University of Kiel, Kiel, Germany
| |
Collapse
|
4
|
Simanaviciute U, Ahmed J, Brown RE, Connor-Robson N, Farr TD, Fertan E, Gambles N, Garland H, Morton AJ, Staiger JF, Skillings EA, Trueman RC, Wade-Martins R, Wood NI, Wong AA, Grant RA. Recommendations for measuring whisker movements and locomotion in mice with sensory, motor and cognitive deficits. J Neurosci Methods 2020; 331:108532. [PMID: 31785300 DOI: 10.1016/j.jneumeth.2019.108532] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/30/2019] [Accepted: 11/25/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Previous studies have measured whisker movements and locomotion to characterise mouse models of neurodegenerative disease. However, these studies have always been completed in isolation, and do not involve standardized procedures for comparisons across multiple mouse models and background strains. NEW METHOD We present a standard method for conducting whisker movement and locomotion studies, by carrying out qualitative scoring and quantitative measurement of whisker movements from high-speed video footage of mouse models of Amyotrophic Lateral Sclerosis, Huntington's disease, Parkinson's disease, Alzheimer's disease, Cerebellar Ataxia, Somatosensory Cortex Development and Ischemic stroke. RESULTS Sex, background strain, source breeder and genotype all affected whisker movements. All mouse models, apart from Parkinson's disease, revealed differences in whisker movements during locomotion. R6/2 CAG250 Huntington's disease mice had the strongest behavioural phenotype. Robo3R3-5-CKO and RIM-DKOSert mouse models have abnormal somatosensory cortex development and revealed significant changes in whisker movements during object exploration. COMPARISON WITH EXISTING METHOD(S) Our results have good agreement with past studies, which indicates the robustness and reliability of measuring whisking. We recommend that differences in whisker movements of mice with motor deficits can be captured in open field arenas, but that mice with impairments to sensory or cognitive functioning should also be filmed investigating objects. Scoring clips qualitatively before tracking will help to structure later analyses. CONCLUSIONS Studying whisker movements provides a quantitative measure of sensing, motor control and exploration. However, the effect of background strain, sex and age on whisker movements needs to be better understood.
Collapse
Affiliation(s)
- Ugne Simanaviciute
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK; School of Biological Sciences, Manchester University, Manchester, M13 9PL, UK
| | - Jewel Ahmed
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Natalie Connor-Robson
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Tracy D Farr
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Nikki Gambles
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK; Public Health Institute, Liverpool John Moores University, Liverpool, L2 2QP, UK
| | - Huw Garland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Göttingen, 37075, Germany
| | - Elizabeth A Skillings
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Rebecca C Trueman
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Nigel I Wood
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Aimee A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Robyn A Grant
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK.
| |
Collapse
|
5
|
Tsai T, Reinehr S, Maliha AM, Joachim SC. Immune Mediated Degeneration and Possible Protection in Glaucoma. Front Neurosci 2019; 13:931. [PMID: 31543759 PMCID: PMC6733056 DOI: 10.3389/fnins.2019.00931] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
The underlying pathomechanisms for glaucoma, one of the most common causes of blindness worldwide, are still not identified. In addition to increased intraocular pressure (IOP), oxidative stress, excitotoxicity, and immunological processes seem to play a role. Several pharmacological or molecular/genetic methods are currently investigated as treatment options for this disease. Altered autoantibody levels were detected in serum, aqueous humor, and tissue sections of glaucoma patients. To further analyze the role of the immune system, an IOP-independent, experimental autoimmune glaucoma (EAG) animal model was developed. In this model, immunization with ocular antigens leads to antibody depositions, misdirected T-cells, retinal ganglion cell death and degeneration of the optic nerve, similar to glaucomatous degeneration in patients. Moreover, an activation of the complement system and microglia alterations were identified in the EAG as well as in ocular hypertension models. The inhibition of these factors can alleviate degeneration in glaucoma models with and without high IOP. Currently, several neuroprotective approaches are tested in distinct models. It is necessary to have systems that cover underlying pathomechanisms, but also allow for the screening of new drugs. In vitro models are commonly used, including single cell lines, mixed-cultures, and even organoids. In ex vivo organ cultures, pathomechanisms as well as therapeutics can be investigated in the whole retina. Furthermore, animal models reveal insights in the in vivo situation. With all these models, several possible new drugs and therapy strategies were tested in the last years. For example, hypothermia treatment, neurotrophic factors or the blockage of excitotoxity. However, further studies are required to reveal the pressure independent pathomechanisms behind glaucoma. There is still an open issue whether immune mechanisms directly or indirectly trigger cell death pathways. Hence, it might be an imbalance between protective and destructive immune mechanisms. Moreover, identified therapy options have to be evaluated in more detail, since deeper insights could lead to better treatment options for glaucoma patients.
Collapse
Affiliation(s)
| | | | | | - Stephanie C. Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
6
|
Power M, Das S, Schütze K, Marigo V, Ekström P, Paquet-Durand F. Cellular mechanisms of hereditary photoreceptor degeneration - Focus on cGMP. Prog Retin Eye Res 2019; 74:100772. [PMID: 31374251 DOI: 10.1016/j.preteyeres.2019.07.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022]
Abstract
The cellular mechanisms underlying hereditary photoreceptor degeneration are still poorly understood, a problem that is exacerbated by the enormous genetic heterogeneity of this disease group. However, the last decade has yielded a wealth of new knowledge on degenerative pathways and their diversity. Notably, a central role of cGMP-signalling has surfaced for photoreceptor cell death triggered by a subset of disease-causing mutations. In this review, we examine key aspects relevant for photoreceptor degeneration of hereditary origin. The topics covered include energy metabolism, epigenetics, protein quality control, as well as cGMP- and Ca2+-signalling, and how the related molecular and metabolic processes may trigger photoreceptor demise. We compare and integrate evidence on different cell death mechanisms that have been associated with photoreceptor degeneration, including apoptosis, necrosis, necroptosis, and PARthanatos. A special focus is then put on the mechanisms of cGMP-dependent cell death and how exceedingly high photoreceptor cGMP levels may cause activation of Ca2+-dependent calpain-type proteases, histone deacetylases and poly-ADP-ribose polymerase. An evaluation of the available literature reveals that a large group of patients suffering from hereditary photoreceptor degeneration carry mutations that are likely to trigger cGMP-dependent cell death, making this pathway a prime target for future therapy development. Finally, an outlook is given into technological and methodological developments that will with time likely contribute to a comprehensive overview over the entire metabolic complexity of photoreceptor cell death. Building on such developments, new imaging technology and novel biomarkers may be used to develop clinical test strategies, that fully consider the genetic heterogeneity of hereditary retinal degenerations, in order to facilitate clinical testing of novel treatment approaches.
Collapse
Affiliation(s)
- Michael Power
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany; Centre for Integrative Neurosciences (CIN), University of Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, Germany
| | - Soumyaparna Das
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, Germany
| | | | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Italy
| | - Per Ekström
- Ophthalmology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sweden
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany.
| |
Collapse
|
7
|
Park PSH. Rhodopsin Oligomerization and Aggregation. J Membr Biol 2019; 252:413-423. [PMID: 31286171 DOI: 10.1007/s00232-019-00078-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/25/2019] [Indexed: 12/18/2022]
Abstract
Rhodopsin is the light receptor in photoreceptor cells of the retina and a prototypical G protein-coupled receptor. Two types of quaternary structures can be adopted by rhodopsin. If rhodopsin folds and attains a proper tertiary structure, it can then form oligomers and nanodomains within the photoreceptor cell membrane. In contrast, if rhodopsin misfolds, it cannot progress through the biosynthetic pathway and instead will form aggregates that can cause retinal degenerative disease. In this review, emerging views are highlighted on the supramolecular organization of rhodopsin within the membrane of photoreceptor cells and the aggregation of rhodopsin that can lead to retinal degeneration.
Collapse
Affiliation(s)
- Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
Abstract
Approximately 93 years ago at the zoological laboratories of Harvard University, Keeler, a medical geneticist, discovered a retina from a male albino mouse that was completely devoid of visual cells (rods). This rodless mouse was to be the first ever reported murine model of retinal degeneration. Over the years, naturally occurring retinal degeneration mouse mutants have been identified in several common laboratory inbred lines including FVB/NJ (Pde6brd1) and C57BL/6N (Crb1rd8). It is therefore imperative that vision researchers employing other genetically induced retinal degeneration models and experimental models such as laser-induced choroidal neovascularization (CNV) or bright white-light exposure screen for such naturally occurring mutations to prevent costly misinterpretations. In this regard, we describe herein simple molecular-based techniques for screening the presence of some commonly encountered rd mutations (Pde6brd1, Crb1rd8, Pde6brd10, and Rpe65rd12).
Collapse
|
9
|
Schreiber S, Petrasch-Parwez E, Porrmann-Kelterbaum E, Förster E, Epplen JT, Gerding WM. Neurodegeneration in the olfactory bulb and olfactory deficits in the Ccdc66 -/- mouse model for retinal degeneration. IBRO Rep 2018; 5:43-53. [PMID: 30211337 PMCID: PMC6132079 DOI: 10.1016/j.ibror.2018.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022] Open
Abstract
The Ccdc66-deficient (Ccdc66 -/-) mouse model exhibits slow progressive retinal degeneration. It is unclear whether CCDC66 protein also plays a role in the wildtype (WT; Ccdc66 +/+) mouse brain and whether the lack of Ccdc66 gene expression in the Ccdc66 -/- mouse brain may result in morphological and behavioral alterations. CCDC66 protein expression in different brain regions of the adult WT mouse and in whole brain during postnatal development was quantified by SDS-PAGE and Western blot. Ccdc66 reporter gene expression was visualized by X-gal staining. Selected brain regions were further analyzed by light and electron microscopy. In order to correlate anatomical with behavioral data, an olfactory habituation/dishabituation test was performed. CCDC66 protein was expressed throughout the early postnatal development in the WT mouse brain. In adult mice, the main olfactory bulb exhibited high CCDC66 protein levels comparable to the expression in the retina. Additionally, the Ccdc66 -/- mouse brain showed robust Ccdc66 reporter gene expression especially in adult olfactory bulb glomeruli, the olfactory nerve layer and the olfactory epithelium. Degeneration was detected in the Ccdc66 -/- olfactory bulb glomeruli at advanced age. This degeneration was also reflected in behavioral alterations; compared to the WT, Ccdc66 -/- mice spent significantly less time sniffing at the initial presentation of unknown, neutral odors and barely responded to social odors. Ccdc66 -/- mice develop substantial olfactory nerve fiber degeneration and alteration of olfaction-related behavior at advanced age. Thus, the Ccdc66 -/- mouse model for retinal degeneration adds the possibility to study mechanisms of central nervous system degeneration.
Collapse
Key Words
- AG, astroglia
- CTX, cortex
- Ccdc66
- Ccdc66 +/+, WT, wildtype
- Ccdc66 -/-, Ccdc66-deficient
- De, dendrite
- EPL, external plexiform layer
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GL, glomerular layer
- IPL, internal plexiform layer
- M, mitochondrion
- ML, mitral cell layer
- MOB, main olfactory bulb
- Mouse model
- Neurodegeneration
- OE, olfactory epithelium
- ONF, olfactory nerve fibers
- ONL, olfactory nerve layer
- ORN, olfactory receptor neuron(s)
- Olfactory bulb
- P, postnatal day
- PBS, phosphate-buffered saline
- PG, periglomerular cells
- RIPA, radioimmunoprecipitation assay
- RMS, rostral migratory stream
- RP, retinitis pigmentosa
- Retinitis pigmentosa
- SC, supporting cell
- SEZ, subependymal zone
- SVZ, subventricular zone
- gPRA, generalized progressive retinal atrophy
- ioD, integrated optic density
- m, month/s
Collapse
Affiliation(s)
- Sabrina Schreiber
- Department of Human Genetics, Ruhr-University, 44780 Bochum, Germany
| | | | | | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University, 44780 Bochum, Germany
| | - Jörg T. Epplen
- Department of Human Genetics, Ruhr-University, 44780 Bochum, Germany
- Department of Biochemistry and Molecular Medicine, University of Witten-Herdecke, ZBAF, 58453 Witten, Germany
| | - Wanda M. Gerding
- Department of Human Genetics, Ruhr-University, 44780 Bochum, Germany
| |
Collapse
|
10
|
Grant RA, Wong AA, Fertan E, Brown RE. Whisker exploration behaviours in the 5xFAD mouse are affected by sex and retinal degeneration. GENES BRAIN AND BEHAVIOR 2018; 19:e12532. [DOI: 10.1111/gbb.12532] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Robyn A. Grant
- Division of Biology and Conservation EcologyManchester Metropolitan University Manchester UK
| | - Aimee A. Wong
- Department of Psychology and NeuroscienceDalhousie University Halifax Nova Scotia Canada
| | - Emre Fertan
- Department of Psychology and NeuroscienceDalhousie University Halifax Nova Scotia Canada
| | - Richard E. Brown
- Department of Psychology and NeuroscienceDalhousie University Halifax Nova Scotia Canada
| |
Collapse
|
11
|
Wisely CE, Sayed JA, Tamez H, Zelinka C, Abdel-Rahman MH, Fischer AJ, Cebulla CM. The chick eye in vision research: An excellent model for the study of ocular disease. Prog Retin Eye Res 2017; 61:72-97. [PMID: 28668352 PMCID: PMC5653414 DOI: 10.1016/j.preteyeres.2017.06.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023]
Abstract
The domestic chicken, Gallus gallus, serves as an excellent model for the study of a wide range of ocular diseases and conditions. The purpose of this manuscript is to outline some anatomic, physiologic, and genetic features of this organism as a robust animal model for vision research, particularly for modeling human retinal disease. Advantages include a sequenced genome, a large eye, relative ease of handling and maintenance, and ready availability. Relevant similarities and differences to humans are highlighted for ocular structures as well as for general physiologic processes. Current research applications for various ocular diseases and conditions, including ocular imaging with spectral domain optical coherence tomography, are discussed. Several genetic and non-genetic ocular disease models are outlined, including for pathologic myopia, keratoconus, glaucoma, retinal detachment, retinal degeneration, ocular albinism, and ocular tumors. Finally, the use of stem cell technology to study the repair of damaged tissues in the chick eye is discussed. Overall, the chick model provides opportunities for high-throughput translational studies to more effectively prevent or treat blinding ocular diseases.
Collapse
Affiliation(s)
- C Ellis Wisely
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Javed A Sayed
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Heather Tamez
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Chris Zelinka
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Mohamed H Abdel-Rahman
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 333 West 10th Avenue, Columbus, OH 43210, USA.
| | - Colleen M Cebulla
- Havener Eye Institute, Department of Ophthalmology and Visual Science, The Ohio State University Wexner Medical Center, 915 Olentangy River Rd, Columbus, OH 43212, USA.
| |
Collapse
|
12
|
Yan W, Yao L, Liu W, Sun K, Zhang Z, Zhang L. A kind of rd1 mouse in C57BL/6J mice from crossing with a mutated Kunming mouse. Gene 2017; 607:9-15. [PMID: 28077313 DOI: 10.1016/j.gene.2017.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 01/01/2017] [Accepted: 01/06/2017] [Indexed: 11/18/2022]
Abstract
We occasionally discovered a mouse with spontaneous retinitis pigmentosa (RP) from Kunming (KM) mouse breeding colony, with no obvious waveforms in ERG recordings. The aim of this study is to cross the spontaneously hereditary retinal degeneration mice (temporarily designated as KM/rd mice) derived from KM mice with C57BL/6J mice to establish a congenic inbred strain (temporarily designated as the B6/rd mice), and study the ocular phenotype and genotype of the mice. Fundus photography, tissue morphology, electroretinography (ERG), qRT-PCR, western blot and DNA sequence analysis were performed to observe the ocular phenotype and genotype of KM/rd and B6/rd mice. The fundus photography showed progressive retinal vascular degeneration and depigmentation in KM/rd and B6/rd mice. Compared to wild-type mice, the histological analysis revealed that the outer nuclear layer of the mutated mice was significantly reduced at 14days post born (P14), and almost disappeared by P21. No obvious waveforms were detected at P14 and P21 in the ERG from KM/rd and B6/rd mice. qRT-PCR results showed that the expression quantities of mRNA of pde6b gene in KM/rd and B6/rd mice were significantly lower compared with those of wild-type controls at P21. Western blot results confirmed an abnormal protein expression of pde6b gene in KM/rd and B6/rd mice with no protein products, while there was an obvious protein expression in wild-type mice. The nonsense mutation in exon 7 (a mutation that changes the codon 347 from TAC to TAA) in the pde6b gene of KM/rd and B6/rd mice was identified by genomic DNA sequence analysis. All these findings revealed that the ocular phenotype and genotype of KM/rd and B6/rd mice were similar to those of rd1 mice, which indicates that KM/rd and B6/rd mice can be used as an RP mouse model.
Collapse
Affiliation(s)
- Weiming Yan
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, The Fourth Military University, Xi'an, Shaanxi Province, China
| | - Lu Yao
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, The Fourth Military University, Xi'an, Shaanxi Province, China
| | - Wei Liu
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-Xianyang New Ecomic Zone, Xi'an, Shaanxi Province, China
| | - Kai Sun
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, The Fourth Military University, Xi'an, Shaanxi Province, China
| | - ZuoMing Zhang
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, The Fourth Military University, Xi'an, Shaanxi Province, China.
| | - Lei Zhang
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, The Fourth Military University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
13
|
Boya P, Esteban-Martínez L, Serrano-Puebla A, Gómez-Sintes R, Villarejo-Zori B. Autophagy in the eye: Development, degeneration, and aging. Prog Retin Eye Res 2016; 55:206-245. [PMID: 27566190 DOI: 10.1016/j.preteyeres.2016.08.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic pathway that promotes the degradation and recycling of cellular components. Proteins, lipids, and even whole organelles are engulfed in autophagosomes and delivered to the lysosome for elimination. In response to stress, autophagy mediates the degradation of cell components, which are recycled to generate the nutrients and building blocks required to sustain cellular homeostasis. Moreover, it plays an important role in cellular quality control, particularly in neurons, in which the total burden of altered proteins and damaged organelles cannot be reduced by redistribution to daughter cells through cell division. Research has only begun to examine the role of autophagy in the visual system. The retina, a light-sensitive tissue, detects and transmits electrical impulses through the optic nerve to the visual cortex in the brain. Both the retina and the eye are exposed to a variety of environmental insults and stressors, including genetic mutations and age-associated alterations that impair their function. Here, we review the main studies that have sought to explain autophagy's importance in visual function. We describe the role of autophagy in retinal development and cell differentiation, and discuss the implications of autophagy dysregulation both in physiological aging and in important diseases such as age-associated macular degeneration and glaucoma. We also address the putative role of autophagy in promoting photoreceptor survival and discuss how selective autophagy could provide alternative means of protecting retinal cells. The findings reviewed here underscore the important role of autophagy in maintaining proper retinal function and highlight novel therapeutic approaches for blindness and other diseases of the eye.
Collapse
Affiliation(s)
- Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Lorena Esteban-Martínez
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Ana Serrano-Puebla
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| |
Collapse
|
14
|
Yao L, Zhang L, Qi LS, Liu W, An J, Wang B, Xue JH, Zhang ZM. The Time Course of Deafness and Retinal Degeneration in a Kunming Mouse Model for Usher Syndrome. PLoS One 2016; 11:e0155619. [PMID: 27186975 PMCID: PMC4871471 DOI: 10.1371/journal.pone.0155619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 05/02/2016] [Indexed: 01/13/2023] Open
Abstract
Usher syndrome is a group of autosomal recessive diseases characterized by congenital deafness and retinitis pigmentosa. In a mouse model for Usher syndrome, KMush/ush, discovered in our laboratory, we measured the phenotypes, characterized the architecture and morphology of the retina, and quantified the level of expression of pde6b and ush2a between postnatal (P) days 7, and 56. Electroretinograms and auditory brainstem response were used to measure visual and auditory phenotypes. Fundus photography and light microscopy were used to measure the architecture and morphology of the retina. Quantitative real-time PCR was used to measure the expression levels of mRNA. KMush/ush mice had low amplitudes and no obvious waveforms of Electroretinograms after P14 compared with controls. Thresholds of auditory brainstem response in our model were higher than those of controls after P14. By P21, the retinal vessels of KMush/ush mice were attenuated and their optic discs had a waxy pallor. The retinas of KMush/ush mice atrophied and the choroidal vessels were clearly visible. Notably, the architecture of each retinal layer was not different as compared with control mice at P7, while the outer nuclear layer (ONL) and other retinal layers of KMush/ush mice were attenuated significantly between P14 and P21. ONL cells were barely seen in KMush/ush mice at P56. As compared with control mice, the expression of pde6b and ush2a in KMush/ush mice declined significantly after P7. This study is a first step toward characterizing the progression of disease in our mouse model. Future studies using this model may provide insights about the etiology of the disease and the relationships between genotypes and phenotypes providing a valuable resource that could contribute to the foundation of knowledge necessary to develop therapies to prevent the retinal degeneration in patients with Usher Syndrome.
Collapse
Affiliation(s)
- Lu Yao
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
| | - Lei Zhang
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
| | - Lin-Song Qi
- Department of Physical Examination, Air Force General Hospital, 30 Fucheng Road, Beijing, China
| | - Wei Liu
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-Xianyang New Ecomic Zone, 712046, Xi'an, China
| | - Jing An
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
| | - Bin Wang
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
| | - Jun-Hui Xue
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
- * E-mail: (JHX); (ZMZ)
| | - Zuo-Ming Zhang
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, 169 West Changle Road, Xi'an, China
- * E-mail: (JHX); (ZMZ)
| |
Collapse
|
15
|
Sothilingam V, Garcia Garrido M, Jiao K, Buena-Atienza E, Sahaboglu A, Trifunović D, Balendran S, Koepfli T, Mühlfriedel R, Schön C, Biel M, Heckmann A, Beck SC, Michalakis S, Wissinger B, Seeliger MW, Paquet-Durand F. Retinitis pigmentosa: impact of different Pde6a point mutations on the disease phenotype. Hum Mol Genet 2015; 24:5486-99. [PMID: 26188004 DOI: 10.1093/hmg/ddv275] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/09/2015] [Indexed: 11/13/2022] Open
Abstract
Mutations in the PDE6A gene can cause rod photoreceptors degeneration and the blinding disease retinitis pigmentosa (RP). While a number of pathogenic PDE6A mutations have been described, little is known about their impact on compound heterozygous situations and potential interactions of different disease-causing alleles. Here, we used a novel mouse model for the Pde6a R562W mutation in combination with an existing line carrying the V685M mutation to generate compound heterozygous Pde6a V685M/R562W animals, exactly homologous to a case of human RP. We compared the progression of photoreceptor degeneration in these compound heterozygous mice with the homozygous V685M and R562W mutants, and additionally with the D670G line that is known for a relatively mild phenotype. We investigated PDE6A expression, cyclic guanosine mono-phosphate accumulation, calpain and caspase activity, in vivo retinal function and morphology, as well as photoreceptor cell death and survival. This analysis confirms the severity of different Pde6a mutations and indicates that compound heterozygous mutants behave like intermediates of the respective homozygous situations. Specifically, the severity of the four different Pde6a situations may be categorized by the pace of photoreceptor degeneration: V685M (fastest) > V685M/R562W > R562W > D670G (slowest). While calpain activity was strongly increased in all four mutants, caspase activity was not. This points to the execution of non-apoptotic cell death and may lead to the identification of new targets for therapeutic interventions. For individual RP patients, our study may help to predict time-courses for Pde6a-related retinal degeneration and thereby facilitate the definition of a window-of-opportunity for clinical interventions.
Collapse
Affiliation(s)
- Vithiyanjali Sothilingam
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Schleichstr.4/3, Tuebingen 72076, Germany
| | - Marina Garcia Garrido
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Schleichstr.4/3, Tuebingen 72076, Germany
| | - Kangwei Jiao
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany, Second People's Hospital of Yunnan Province and Fourth Affiliated Hospital of Kunming Medical University, 176 Qingnian Road, Wuhua, Kunming, Yunnan 650021, China
| | - Elena Buena-Atienza
- Molecular Genetics Laboratory, Centre for Ophthalmology, University Clinics Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Ayse Sahaboglu
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Dragana Trifunović
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Sukirthini Balendran
- Molecular Genetics Laboratory, Centre for Ophthalmology, University Clinics Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Tanja Koepfli
- Molecular Genetics Laboratory, Centre for Ophthalmology, University Clinics Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Regine Mühlfriedel
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Schleichstr.4/3, Tuebingen 72076, Germany
| | - Christian Schön
- Center for Integrated Protein Science Munich (CIPSM) at the Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich 81377, Germany and
| | - Martin Biel
- Center for Integrated Protein Science Munich (CIPSM) at the Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich 81377, Germany and
| | | | - Susanne C Beck
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Schleichstr.4/3, Tuebingen 72076, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich (CIPSM) at the Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich 81377, Germany and
| | - Bernd Wissinger
- Molecular Genetics Laboratory, Centre for Ophthalmology, University Clinics Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany
| | - Mathias W Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Schleichstr.4/3, Tuebingen 72076, Germany
| | - François Paquet-Durand
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tuebingen, Roentgenweg 11, Tuebingen 72076, Germany,
| |
Collapse
|
16
|
Volland S, Esteve-Rudd J, Hoo J, Yee C, Williams DS. A comparison of some organizational characteristics of the mouse central retina and the human macula. PLoS One 2015; 10:e0125631. [PMID: 25923208 PMCID: PMC4414478 DOI: 10.1371/journal.pone.0125631] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/18/2015] [Indexed: 11/24/2022] Open
Abstract
Mouse models have greatly assisted our understanding of retinal degenerations. However, the mouse retina does not have a macula, leading to the question of whether the mouse is a relevant model for macular degeneration. In the present study, a quantitative comparison between the organization of the central mouse retina and the human macula was made, focusing on some structural characteristics that have been suggested to be important in predisposing the macula to stresses leading to degeneration: photoreceptor density, phagocytic load on the RPE, and the relative thinness of Bruch’s membrane. Light and electron microscopy measurements from retinas of two strains of mice, together with published data on human retinas, were used for calculations and subsequent comparisons. As in the human retina, the central region of the mouse retina possesses a higher photoreceptor cell density and a thinner Bruch’s membrane than in the periphery; however, the magnitudes of these periphery to center gradients are larger in the human. Of potentially greater relevance is the actual photoreceptor cell density, which is much greater in the mouse central retina than in the human macula, underlying a higher phagocytic load for the mouse RPE. Moreover, at eccentricities that correspond to the peripheral half of the human macula, the rod to cone ratio is similar between mouse and human. Hence, with respect to photoreceptor density and phagocytic load of the RPE, the central mouse retina models at least the more peripheral part of the macula, where macular degeneration is often first evident.
Collapse
Affiliation(s)
- Stefanie Volland
- Departments of Ophthalmology and Neurobiology, Stein Eye Institute, Molecular Biology Institute, Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Julian Esteve-Rudd
- Departments of Ophthalmology and Neurobiology, Stein Eye Institute, Molecular Biology Institute, Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Juyea Hoo
- Departments of Ophthalmology and Neurobiology, Stein Eye Institute, Molecular Biology Institute, Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Claudine Yee
- Departments of Ophthalmology and Neurobiology, Stein Eye Institute, Molecular Biology Institute, Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - David S. Williams
- Departments of Ophthalmology and Neurobiology, Stein Eye Institute, Molecular Biology Institute, Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Arango-Gonzalez B, Trifunović D, Sahaboglu A, Kranz K, Michalakis S, Farinelli P, Koch S, Koch F, Cottet S, Janssen-Bienhold U, Dedek K, Biel M, Zrenner E, Euler T, Ekström P, Ueffing M, Paquet-Durand F. Identification of a common non-apoptotic cell death mechanism in hereditary retinal degeneration. PLoS One 2014; 9:e112142. [PMID: 25392995 PMCID: PMC4230983 DOI: 10.1371/journal.pone.0112142] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/13/2014] [Indexed: 01/10/2023] Open
Abstract
Cell death in neurodegenerative diseases is often thought to be governed by apoptosis; however, an increasing body of evidence suggests the involvement of alternative cell death mechanisms in neuronal degeneration. We studied retinal neurodegeneration using 10 different animal models, covering all major groups of hereditary human blindness (rd1, rd2, rd10, Cngb1 KO, Rho KO, S334ter, P23H, Cnga3 KO, cpfl1, Rpe65 KO), by investigating metabolic processes relevant for different forms of cell death. We show that apoptosis plays only a minor role in the inherited forms of retinal neurodegeneration studied, where instead, a non-apoptotic degenerative mechanism common to all mutants is of major importance. Hallmark features of this pathway are activation of histone deacetylase, poly-ADP-ribose-polymerase, and calpain, as well as accumulation of cyclic guanosine monophosphate and poly-ADP-ribose. Our work thus demonstrates the prevalence of alternative cell death mechanisms in inherited retinal degeneration and provides a rational basis for the design of mutation-independent treatments.
Collapse
Affiliation(s)
| | - Dragana Trifunović
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Ayse Sahaboglu
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Katharina Kranz
- Department of Neurobiology, University of Oldenburg, Oldenburg, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Pietro Farinelli
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Division of Ophthalmology, Department of Clinical Sciences, University of Lund, Lund, Sweden
| | - Susanne Koch
- Center for Integrated Protein Science Munich and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fred Koch
- Center for Integrated Protein Science Munich and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sandra Cottet
- Institute for Research in Ophthalmology, Sion, Switzerland
| | | | - Karin Dedek
- Department of Neurobiology, University of Oldenburg, Oldenburg, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Eberhart Zrenner
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Centre for Integrative Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Centre for Integrative Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Per Ekström
- Division of Ophthalmology, Department of Clinical Sciences, University of Lund, Lund, Sweden
| | - Marius Ueffing
- Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | | |
Collapse
|
18
|
Shen SQ, Turro E, Corbo JC. Hybrid mice reveal parent-of-origin and Cis- and trans-regulatory effects in the retina. PLoS One 2014; 9:e109382. [PMID: 25340786 PMCID: PMC4207689 DOI: 10.1371/journal.pone.0109382] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/02/2014] [Indexed: 11/30/2022] Open
Abstract
A fundamental challenge in genomics is to map DNA sequence variants onto changes in gene expression. Gene expression is regulated by cis-regulatory elements (CREs, i.e., enhancers, promoters, and silencers) and the trans factors (e.g., transcription factors) that act upon them. A powerful approach to dissecting cis and trans effects is to compare F1 hybrids with F0 homozygotes. Using this approach and taking advantage of the high frequency of polymorphisms in wild-derived inbred Cast/EiJ mice relative to the reference strain C57BL/6J, we conducted allele-specific mRNA-seq analysis in the adult mouse retina, a disease-relevant neural tissue. We found that cis effects account for the bulk of gene regulatory divergence in the retina. Many CREs contained functional (i.e., activating or silencing) cis-regulatory variants mapping onto altered expression of genes, including genes associated with retinal disease. By comparing our retinal data with previously published liver data, we found that most of the cis effects identified were tissue-specific. Lastly, by comparing reciprocal F1 hybrids, we identified evidence of imprinting in the retina for the first time. Our study provides a framework and resource for mapping cis-regulatory variants onto changes in gene expression, and underscores the importance of studying cis-regulatory variants in the context of retinal disease.
Collapse
Affiliation(s)
- Susan Q. Shen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ernest Turro
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant, Cambridge, United Kingdom
| | - Joseph C. Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
19
|
Ex vivo ERG analysis of photoreceptors using an in vivo ERG system. Vision Res 2014; 101:108-17. [PMID: 24959652 DOI: 10.1016/j.visres.2014.06.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 11/21/2022]
Abstract
The Function of the retina and effects of drugs on it can be assessed by recording transretinal voltage across isolated retina that is perfused with physiological medium. However, building ex vivo ERG apparatus requires substantial amount of time, resources and expertise. Here we adapted a commercial in vivo ERG system for transretinal ERG recordings from rod and cone photoreceptors and compared rod and cone signaling between ex vivo and in vivo environments. We found that the rod and cone a- and b-waves recorded with the transretinal ERG adapter and a standard in vivo ERG system are comparable to those obtained from live anesthetized animals. However, ex vivo responses are somewhat slower and their oscillatory potentials are suppressed as compared to those recorded in vivo. We found that rod amplification constant (A) was comparable between ex vivo and in vivo conditions, ∼10-30s(-2) depending on the choice of response normalization. We estimate that the A in cones is between 3 and 6s(-2) in ex vivo conditions and by assuming equal A in vivo we arrive to light funnelling factor of 3 for cones in the mouse retina. The ex vivo ERG adapter provides a simple and affordable alternative to designing a custom-built transretinal recordings setup for the study of photoreceptors. Our results provide a roadmap to the rigorous quantitative analysis of rod and cone responses made possible with such a system.
Collapse
|
20
|
Park PSH. Constitutively active rhodopsin and retinal disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:1-36. [PMID: 24931191 DOI: 10.1016/b978-0-12-417197-8.00001-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rhodopsin is the light receptor in rod photoreceptor cells of the retina that initiates scotopic vision. In the dark, rhodopsin is bound to the chromophore 11-cis retinal, which locks the receptor in an inactive state. The maintenance of an inactive rhodopsin in the dark is critical for rod photoreceptor cells to remain highly sensitive. Perturbations by mutation or the absence of 11-cis retinal can cause rhodopsin to become constitutively active, which leads to the desensitization of photoreceptor cells and, in some instances, retinal degeneration. Constitutive activity can arise in rhodopsin by various mechanisms and can cause a variety of inherited retinal diseases including Leber congenital amaurosis, congenital night blindness, and retinitis pigmentosa. In this review, the molecular and structural properties of different constitutively active forms of rhodopsin are overviewed, and the possibility that constitutive activity can arise from different active-state conformations is discussed.
Collapse
Affiliation(s)
- Paul Shin-Hyun Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
21
|
Shukolyukov SA. Rhodopsin, Zn2+, and retinitis pigmentosa: a Short tale requiring continuation. BIOCHEMISTRY (MOSCOW) 2013; 78:660-6. [DOI: 10.1134/s0006297913060114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Longitudinal fundus and retinal studies with SD-OCT: a comparison of five mouse inbred strains. Mamm Genome 2013; 24:198-205. [PMID: 23681115 DOI: 10.1007/s00335-013-9457-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 04/24/2013] [Indexed: 02/05/2023]
Abstract
Spectral domain optical coherence tomography (SD-OCT) has recently been established as a method for in vivo imaging of fundus and retina in the mouse. It enables more effective studies of retinal diseases including investigations of etiopathologic mechanisms. In order to learn more about longitudinal fundus development and to enable recognition of disease-associated irregularities, we performed confocal scanning laser ophthalmoscopy (cSLO) and SD-OCT measurements in the inbred strains C57BL/6J, C3HeB/FeJ, FVB/NCrl, BALB/cByJ, and 129S2/SvJ when they were between 2 and 6 months of age. In general, cSLO and SD-OCT data did not reveal sex-specific or unilateral differences. C3HeB/FeJ and FVB/NCrl mice showed diffuse choroidal dysplasia. Choroidal vein-like structures appeared as dark fundus stripes in C3HeB/FeJ. In FVB/NCrl, fundus fleck accumulation was found. In contrast, only minor time-dependent changes of fundus appearance were observed in C57BL/6J, BALB/cByJ, and 129S2/SvJ. This was also found for individual fundic main blood vessel patterns in all inbred strains. Vessel numbers varied between 6 and 13 in C57BL/6J. This was comparable in most cases. We further found that retinae were significantly thicker in C57BL/6J compared to the other strains. Total retinal thickness generally did not change between 2 and 6 months of age. As a conclusion, our results indicate lifelong pathologic processes in C3HeB/FeJ and FVB/NCrl that affect choroid and orbital tissues. Inbred strains with regular retinal development did not reveal major time-dependent variations of fundus appearance, blood vessel pattern, or retinal thickness. Consequently, progressive changes of these parameters are suitable indicators for pathologic outliers.
Collapse
|
23
|
Abstract
The Eye chapter of the 3rd edition of Haschek and Rousseaux’s Handbook of Toxicologic Pathology brings a comprehensive description of pathological processes affecting the ocular tissues in the most commonly used laboratory animals and their correlations with human diseases of interest in toxicology. Also presented are detailed descriptions of the structure and function of the different ocular tissues, the most advanced techniques applied in the toxicological evaluation of the eye, useful animal models of human disease, and known mechanisms of ocular toxicity. The introductory sections of the chapter also feature such essential topics as ocular embryology, an overview of clinical ophthalmic evaluation, and eye-specific techniques of tissue processing.
Collapse
|
24
|
Sortilin participates in light-dependent photoreceptor degeneration in vivo. PLoS One 2012; 7:e36243. [PMID: 22558402 PMCID: PMC3338683 DOI: 10.1371/journal.pone.0036243] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 04/03/2012] [Indexed: 11/20/2022] Open
Abstract
Both proNGF and the neurotrophin receptor p75 (p75NTR) are known to regulate photoreceptor cell death caused by exposure of albino mice to intense illumination. ProNGF-induced apoptosis requires the participation of sortilin as a necessary p75NTR co-receptor, suggesting that sortilin may participate in the photoreceptor degeneration triggered by intense lighting. We report here that light-exposed albino mice showed sortilin, p75NTR, and proNGF expression in the outer nuclear layer, the retinal layer where photoreceptor cell bodies are located. In addition, cone progenitor-derived 661W cells subjected to intense illumination expressed sortilin and p75NTR and released proNGF into the culture medium. Pharmacological blockade of sortilin with either neurotensin or the “pro” domain of proNGF (pro-peptide) favored the survival of 661W cells subjected to intense light. In vivo, the pro-peptide attenuated retinal cell death in light-exposed albino mice. We propose that an auto/paracrine proapoptotic mechanism based on the interaction of proNGF with the receptor complex p75NTR/sortilin participates in intense light-dependent photoreceptor cell death. We therefore propose sortilin as a putative target for intervention in hereditary retinal dystrophies.
Collapse
|
25
|
McUsic AC, Lamba DA, Reh TA. Guiding the morphogenesis of dissociated newborn mouse retinal cells and hES cell-derived retinal cells by soft lithography-patterned microchannel PLGA scaffolds. Biomaterials 2011; 33:1396-405. [PMID: 22115999 DOI: 10.1016/j.biomaterials.2011.10.083] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 10/30/2011] [Indexed: 11/26/2022]
Abstract
Embryonic stem (ES) cell-derived photoreceptors are a promising cell source for enhanced in vitro models of retinal degenerative diseases, but the more differentiated characteristics of retinal cells do not typically develop in dissociated cell cultures. Therefore, we have reconstructed organized retinal tissue by seeding dissociated cells into an array of aligned units that more faithfully mimics the retina. We solvent-processed poly(lactic-co-glycolic acid) (PLGA) into a microchannel scaffold format to achieve this geometric constraint. We compared the effect of PLGA concentration on channel morphology and, along with other culture conditions, on the infiltration of dissociated newborn mouse retinal cells into the channels. Culturing scaffolds at the gas-liquid interface with low serum media increased infiltrated rod photoreceptor viability 18-fold over submerged, high serum cultures when evaluated after seven days. Rod photoreceptors and Müller glia aligned processes parallel to the microchannel walls. Otx2+ and Pax6+ subpopulations recapitulated lamination behavior. Further, we constructed scaffold/retinal pigment epithelium (RPE) co-cultures and observed rods extending rhodopsin-positive processes toward RPE cells, mimicking normal rod polarization and morphology. Finally, human embryonic stem cell-derived photoreceptors exhibited infiltration and morphological characteristics similar to mouse retinal cells inside the scaffolds. These findings constitute an important advance in generating tissue-level retinal models from dissociated cells for use as drug screening platforms and in regenerative medicine.
Collapse
Affiliation(s)
- Andrew C McUsic
- Department of Bioengineering, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | | | | |
Collapse
|
26
|
Wang W, Fernandez de Castro J, Vukmanic E, Zhou L, Emery D, Demarco PJ, Kaplan HJ, Dean DC. Selective rod degeneration and partial cone inactivation characterize an iodoacetic acid model of Swine retinal degeneration. Invest Ophthalmol Vis Sci 2011; 52:7917-23. [PMID: 21896868 DOI: 10.1167/iovs.11-7849] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE. Transgenic pigs carrying a mutant human rhodopsin transgene have been developed as a large animal model of retinitis pigmentosa (RP). This model displays some key features of human RP, but the time course of disease progression makes this model costly, time consuming, and difficult to study because of the size of the animals at end-stage disease. Here, the authors evaluate an iodoacetic acid (IAA) model of photoreceptor degeneration in the pig as an alternative model that shares features of the transgenic pig and human RP. METHODS. IAA blocks glycolysis, thereby inhibiting photoreceptor function. The effect of the intravenous injection of IAA on swine rod and cone photoreceptor viability and morphology was followed by histologic evaluation of different regions of the retina using hematoxylin and eosin and immunostaining. Rod and cone function was analyzed by full-field electroretinography and multifocal electroretinography. RESULTS. IAA led to specific loss of rods in a central-to-peripheral retinal gradient. Although cones were resistant, they showed shortened outer segments, loss of bipolar cell synaptic connections, and a diminished flicker ERG, hallmarks of transition to cone dysfunction in RP patients. CONCLUSIONS. IAA provides an alternative rod-dominant model of retinal damage that shares a surprising number of features with the pig transgenic model of RP and with human RP. This IAA model is cost-effective and rapid, ensuring that the size of the animals does not become prohibitive for end-stage evaluation or therapeutic intervention.
Collapse
Affiliation(s)
- Wei Wang
- Departments of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Kentucky, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Yoshizawa K, Kuro-Kuwata M, Sasaki T, Lai YCC, Kanematsu S, Miki H, Kimura-Kawanaka A, Uehara N, Yuri T, Tsubura A. Retinal degeneration induced in adult mice by a single intraperitoneal injection of N-ethyl-N-nitrosourea. Toxicol Pathol 2011; 39:606-13. [PMID: 21498792 DOI: 10.1177/0192623311402221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Seven-week-old female BALB/c mice received a single intraperitoneal injection of N-ethyl-N-nitrosourea (ENU) (50, 100, 200, 400, or 600 mg/kg), and retinal damage was evaluated after 7 days. Sequential morphological features of the retina and retinal apoptosis, as determined by the TUNEL assay, were analyzed 6, 12, 24, and 72 hr and 7 days after treatment with 600 mg/kg of ENU. Moreover, older mice (25 to 34 weeks of age) received an intraperitoneal injection of 600 mg/kg ENU and were sacrificed 7 days later. All animals were necropsied, and both eyes were examined histopathologically. Two of the 5 mice that received 600 mg/kg ENU died during the experimental period. Histopathologically, all mice that received 600 mg/kg of ENU experienced retinal degeneration characterized by the loss of photoreceptor cells (disappearance of the outer nuclear layer and photoreceptor layer) in both the central and peripheral retina within 7 days. One of 5 mice treated with 400 mg/kg ENU exhibited retinal damage that was restricted to the central retina. Older mice treated with 600 mg/kg ENU exhibited retinal damage that was similar to the retinal damage in younger mice. In the 600 mg/kg ENU-treated mice, TUNEL-positive photoreceptor cells peaked 72 hr after ENU treatment. Retinal thickness and the photoreceptor cell ratio in the central and peripheral retina were significantly decreased, and the retinal damage ratio was significantly increased 7 days after treatment. In conclusion, ENU induces retinal degeneration in adult mice that is characterized by photoreceptor cell apoptosis.
Collapse
Affiliation(s)
- Katsuhiko Yoshizawa
- Department of Pathology II, Kansai Medical University, Moriguchi, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Del Río P, Irmler M, Arango-González B, Favor J, Bobe C, Bartsch U, Vecino E, Beckers J, Hauck SM, Ueffing M. GDNF-induced osteopontin from Müller glial cells promotes photoreceptor survival in the Pde6brd1 mouse model of retinal degeneration. Glia 2011; 59:821-32. [DOI: 10.1002/glia.21155] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 01/18/2011] [Indexed: 01/13/2023]
|
29
|
Fuchs H, Gailus-Durner V, Adler T, Aguilar-Pimentel JA, Becker L, Calzada-Wack J, Da Silva-Buttkus P, Neff F, Götz A, Hans W, Hölter SM, Horsch M, Kastenmüller G, Kemter E, Lengger C, Maier H, Matloka M, Möller G, Naton B, Prehn C, Puk O, Rácz I, Rathkolb B, Römisch-Margl W, Rozman J, Wang-Sattler R, Schrewe A, Stöger C, Tost M, Adamski J, Aigner B, Beckers J, Behrendt H, Busch DH, Esposito I, Graw J, Illig T, Ivandic B, Klingenspor M, Klopstock T, Kremmer E, Mempel M, Neschen S, Ollert M, Schulz H, Suhre K, Wolf E, Wurst W, Zimmer A, Hrabě de Angelis M. Mouse phenotyping. Methods 2010; 53:120-35. [PMID: 20708688 DOI: 10.1016/j.ymeth.2010.08.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 08/06/2010] [Accepted: 08/06/2010] [Indexed: 12/13/2022] Open
Abstract
Model organisms like the mouse are important tools to learn more about gene function in man. Within the last 20 years many mutant mouse lines have been generated by different methods such as ENU mutagenesis, constitutive and conditional knock-out approaches, knock-down, introduction of human genes, and knock-in techniques, thus creating models which mimic human conditions. Due to pleiotropic effects, one gene may have different functions in different organ systems or time points during development. Therefore mutant mouse lines have to be phenotyped comprehensively in a highly standardized manner to enable the detection of phenotypes which might otherwise remain hidden. The German Mouse Clinic (GMC) has been established at the Helmholtz Zentrum München as a phenotyping platform with open access to the scientific community (www.mousclinic.de; [1]). The GMC is a member of the EUMODIC consortium which created the European standard workflow EMPReSSslim for the systemic phenotyping of mouse models (http://www.eumodic.org/[2]).
Collapse
Affiliation(s)
- Helmut Fuchs
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 München/Neuherberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Miyamoto M, Aoki M, Sugimoto S, Kawasaki K, Imai R. IRD1 and IRD2 Mice, Naturally Occurring Models of Hereditary Retinal Dysfunction, Show Late-Onset and Progressive Retinal Degeneration. Curr Eye Res 2010; 35:137-45. [DOI: 10.3109/02713680903447900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Baehr W, Frederick JM. Naturally occurring animal models with outer retina phenotypes. Vision Res 2009; 49:2636-52. [PMID: 19375447 PMCID: PMC2782402 DOI: 10.1016/j.visres.2009.04.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 04/07/2009] [Accepted: 04/07/2009] [Indexed: 01/28/2023]
Abstract
Naturally occurring and laboratory generated animal models serve as powerful tools with which to investigate the etiology of human retinal degenerations, especially retinitis pigmentosa (RP), Leber congenital amaurosis (LCA), cone dystrophies (CD) and macular degeneration (MD). Much progress has been made in elucidating gene defects underlying disease, in understanding mechanisms leading to disease, and in designing molecules for translational research and gene-based therapy to interfere with the progression of disease. Key to this progress has been study of naturally occurring murine and canine retinal degeneration mutants. This article will review the history, phenotypes and gene defects of select animal models with outer retina (photoreceptor and retinal pigment epithelium) degeneration phenotypes.
Collapse
Affiliation(s)
- Wolfgang Baehr
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
32
|
Abstract
There are four members of the bestrophin family of proteins in the human genome, of which two are known to be expressed in the eye. The gene BEST1 (formerly VMD2) which encodes the protein bestrophin-1 (Best1) was first identified in 1998. Mutations in this gene have now been associated with four clinically distinguishable human eye diseases, collectively referred to as "bestrophinopathies". Over the last decade, laboratories have sought to understand how Best1 mutations could result in eye diseases that range in presentation from macular degeneration to nanophthalmos. The majority of our knowledge comes from studies that have sought to understand how Best1 mutations or dysfunction could induce the classical symptoms of the most common of these diseases: Best vitelliform macular dystrophy (BVMD). BVMD is a dominant trait that is characterized electrophysiologically by a diminished electrooculogram light peak with a normal clinical electroretinogram. This together with the localization of Best1 to the retinal pigment epithelium (RPE) basolateral plasma membrane and data from heterologous expression studies, have led to the proposal that Best1 generates the light peak, and that bestrophins are a family of Ca(2+) activated Cl(-) channels (CaCCs). However, data from Best1 knock-out and knock-in mice, coupled with the recent discovery of a recessive bestrophinopathy suggest that Best1 does not generate the light peak. Recently Best2 was found to be expressed in non-pigmented epithelia in the ciliary body. However, aqueous dynamics in Best2 knock-out mice do not support a role for Best2 as a Cl(-) channel. Thus, the purported CaCC function of the bestrophins and how loss of this function relates to clinical disease needs to be reassessed. In this article, we examine data obtained from tissue-type and animal models and discuss the current state of bestrophin research, what roles Best1 and Best2 may play in ocular epithelia and ocular electrophysiology, and how perturbation of these functions may result in disease.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ 85711, USA.
| | | | | |
Collapse
|
33
|
Paquet-Durand F, Hauck SM, van Veen T, Ueffing M, Ekström P. PKG activity causes photoreceptor cell death in two retinitis pigmentosa models. J Neurochem 2009; 108:796-810. [PMID: 19187097 DOI: 10.1111/j.1471-4159.2008.05822.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Photoreceptor degeneration in retinitis pigmentosa is one of the leading causes of hereditary blindness in the developed world. Although causative genetic mutations have been elucidated in many cases, the underlying neuronal degeneration mechanisms are still unknown. Here, we show that activation of cGMP-dependent protein kinase (PKG) hallmarks photoreceptor degeneration in rd1 and rd2 human homologous mouse models. When induced in wild-type retinae, PKG activity was both necessary and sufficient to trigger cGMP-mediated photoreceptor cell death. Target-specific, pharmacological inhibition of PKG activity in both rd1 and rd2 retinae strongly reduced photoreceptor cell death in organotypic retinal explants. Likewise, inhibition of PKG in vivo, using three different application paradigms, resulted in robust photoreceptor protection in the rd1 retina. These findings suggest a pivotal role for PKG activity in cGMP-mediated photoreceptor degeneration mechanisms and highlight the importance of PKG as a novel target for the pharmacological intervention in RP.
Collapse
Affiliation(s)
- François Paquet-Durand
- University of Tübingen, Centre for Ophthalmology, Institute for Ophthalmic Research, Division of Experimental Ophthalmology, Röntgenweg 11, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
34
|
|
35
|
Colella P, Cotugno G, Auricchio A. Ocular gene therapy: current progress and future prospects. Trends Mol Med 2009; 15:23-31. [PMID: 19097940 DOI: 10.1016/j.molmed.2008.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 11/04/2008] [Accepted: 11/04/2008] [Indexed: 12/16/2022]
Abstract
As gene therapy begins to produce its first clinical successes, interest in ocular gene transfer has grown owing to the favorable safety and efficacy characteristics of the eye as a target organ for drug delivery. Important advances also include the availability of viral and non-viral vectors that are able to efficiently transduce various ocular cell types, the use of intraocular delivery routes and the development of transcriptional regulatory elements that allow sustained levels of gene transfer in small and large animal models after a single administration. Here, we review recent progress in the field of ocular gene therapy. The first experiments in humans with severe inherited forms of blindness seem to confirm the good safety and efficacy profiles observed in animal models and suggest that gene transfer has the potential to become a valuable therapeutic strategy for otherwise untreatable blinding diseases.
Collapse
Affiliation(s)
- Pasqualina Colella
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, 80131 Naples, Italy
| | | | | |
Collapse
|
36
|
Gailus-Durner V, Fuchs H, Adler T, Aguilar Pimentel A, Becker L, Bolle I, Calzada-Wack J, Dalke C, Ehrhardt N, Ferwagner B, Hans W, Hölter SM, Hölzlwimmer G, Horsch M, Javaheri A, Kallnik M, Kling E, Lengger C, Mörth C, Mossbrugger I, Naton B, Prehn C, Puk O, Rathkolb B, Rozman J, Schrewe A, Thiele F, Adamski J, Aigner B, Behrendt H, Busch DH, Favor J, Graw J, Heldmaier G, Ivandic B, Katus H, Klingenspor M, Klopstock T, Kremmer E, Ollert M, Quintanilla-Martinez L, Schulz H, Wolf E, Wurst W, de Angelis MH. Systemic first-line phenotyping. Methods Mol Biol 2009; 530:463-509. [PMID: 19266331 DOI: 10.1007/978-1-59745-471-1_25] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
With the completion of the mouse genome sequence an essential task for biomedical sciences in the twenty-first century will be the generation and functional analysis of mouse models for every gene in the mammalian genome. More than 30,000 mutations in ES cells will be engineered and thousands of mouse disease models will become available over the coming years by the collaborative effort of the International Mouse Knockout Consortium. In order to realize the full value of the mouse models proper characterization, archiving and dissemination of mouse disease models to the research community have to be performed. Phenotyping centers (mouse clinics) provide the necessary capacity, broad expertise, equipment, and infrastructure to carry out large-scale systemic first-line phenotyping. Using the example of the German Mouse Clinic (GMC) we will introduce the reader to the different aspects of the organization of a mouse clinic and present selected methods used in first-line phenotyping.
Collapse
|
37
|
Sancho-Pelluz J, Arango-Gonzalez B, Kustermann S, Romero FJ, van Veen T, Zrenner E, Ekström P, Paquet-Durand F. Photoreceptor cell death mechanisms in inherited retinal degeneration. Mol Neurobiol 2008; 38:253-69. [PMID: 18982459 DOI: 10.1007/s12035-008-8045-9] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 10/16/2008] [Indexed: 02/24/2023]
Abstract
Photoreceptor cell death is the major hallmark of a group of human inherited retinal degenerations commonly referred to as retinitis pigmentosa (RP). Although the causative genetic mutations are often known, the mechanisms leading to photoreceptor degeneration remain poorly defined. Previous research work has focused on apoptosis, but recent evidence suggests that photoreceptor cell death may result primarily from non-apoptotic mechanisms independently of AP1 or p53 transcription factor activity, Bcl proteins, caspases, or cytochrome c release. This review briefly describes some animal models used for studies of retinal degeneration, with particular focus on the rd1 mouse. After outlining the major features of different cell death mechanisms in general, we then compare them with results obtained in retinal degeneration models, where photoreceptor cell death appears to be governed by, among other things, changes in cyclic nucleotide metabolism, downregulation of the transcription factor CREB, and excessive activation of calpain and PARP. Based on recent experimental evidence, we propose a putative non-apoptotic molecular pathway for photoreceptor cell death in the rd1 retina. The notion that inherited photoreceptor cell death is driven by non-apoptotic mechanisms may provide new ideas for future treatment of RP.
Collapse
Affiliation(s)
- Javier Sancho-Pelluz
- Institute for Ophthalmic Research, University of Tübingen, Centre for Ophthalmology, Röntgenweg 11, 72076, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Greferath U, Kambourakis M, Barth C, Fletcher EL, Murphy M. Characterization of histamine projections and their potential cellular targets in the mouse retina. Neuroscience 2008; 158:932-44. [PMID: 19015005 DOI: 10.1016/j.neuroscience.2008.10.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 10/14/2008] [Accepted: 10/20/2008] [Indexed: 11/19/2022]
Abstract
The vertebrate retina receives histaminergic input from the brain via retinopetal axons that originate from perikarya in the posterior hypothalamus. In the nervous system, histamine acts on three G-protein-coupled receptors, histamine receptor (HR) 1, HR2 and HR3. In order to look for potential cellular targets of histamine in the mouse retina, we have examined the retina for the expression of histamine and the presence of these three receptors. Consistent with studies of retina from other vertebrates, histamine was only found in retinopetal axons, which coursed extensively through the ganglion cell and inner plexiform layers. mRNA for all three receptors was expressed in the mouse retina, and immunohistochemical studies further localized HR1 and HR2. HR1 immunoreactivity was observed on dopaminergic amacrine cells, calretinin-positive ganglion cells and axon bundles in the ganglion cell layer. Furthermore, a distinct group of processes in the inner plexiform layer was labeled, which most likely represents the processes of cholinergic amacrine cells. HR2 immunoreactivity was observed on the processes and cell bodies of the primary glial cells of the mammalian retina, the Müller cells. This distribution of histamine and its receptors is consistent with a brain-derived source of histamine acting on diverse populations of cells in the retina, including both neurons and glia.
Collapse
Affiliation(s)
- U Greferath
- Department of Anatomy and Cell Biology, University of Melbourne, Grattan Street, Melbourne, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
39
|
Stenkamp DL, Satterfield R, Muhunthan K, Sherpa T, Vihtelic TS, Cameron DA. Age-related cone abnormalities in zebrafish with genetic lesions in sonic hedgehog. Invest Ophthalmol Vis Sci 2008; 49:4631-40. [PMID: 18502998 PMCID: PMC2584603 DOI: 10.1167/iovs.07-1224] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Sonic hedgehog (Shh) signaling is essential for photoreceptor differentiation and retinal cell survival in embryonic zebrafish. The study was conducted to determine whether adult heterozygous carriers of mutant alleles for the shh gene display retinal abnormalities. METHODS Retinal cryosections from young, middle-aged, and senescent wild-type and sonic-you(+/-) (syu(+/-)) zebrafish were probed with retinal cell type-specific markers. Contralateral retinal flatmounts from these fish, and from adult albino zebrafish subjected to light-induced photoreceptor damage followed by regeneration, were hybridized with blue cone opsin cRNA for quantitative analysis of the blue cone pattern. Retinal expression of shh mRNA was measured by quantitative RT-PCR. RESULTS Regions of cone loss and abnormal cone morphology were observed in the oldest syu(+/-) zebrafish, although no other retinal cell type was affected. This phenotype was age-related and genotype-specific. Cone distribution in the oldest syu(+/-) zebrafish was predominantly random, as assessed by measuring the short-range pattern, whereas that of wild-type fish and the younger syu(+/-) zebrafish was statistically regular. A measure of long-range pattern revealed atypical cone aggregation in the oldest syu(+/-) zebrafish. The light-treated albino zebrafish displayed random cone patterns immediately after light toxicity, but showed cone aggregation on regeneration. Retinas from the syu(+/-) fish showed reduced expression of shh mRNA compared with those of wild-type siblings. CONCLUSIONS The syu(+/-) zebrafish presents a model for the study of hereditary age-related cone abnormalities. The syu(+/-) retinas most likely experience progressive cone photoreceptor loss, accompanied by cone regeneration. Shh signaling may be required to maintain cone viability throughout life.
Collapse
Affiliation(s)
- Deborah L Stenkamp
- Department of Biological Sciences, University of Idaho, Moscow, Idaho 83844-3051, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Bhogal N. Genetically altered mice, man and medicine. J Appl Biomed 2008. [DOI: 10.32725/jab.2008.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
41
|
Hashizume K, Hirasawa M, Imamura Y, Noda S, Shimizu T, Shinoda K, Kurihara T, Noda K, Ozawa Y, Ishida S, Miyake Y, Shirasawa T, Tsubota K. Retinal dysfunction and progressive retinal cell death in SOD1-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1325-31. [PMID: 18372426 DOI: 10.2353/ajpath.2008.070730] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The superoxide dismutase (SOD) family is a major antioxidant system, and deficiency of Cu,Zn-superoxide dismutase (SOD1) in mice leads to many different phenotypes that resemble accelerated aging. The purpose of this study was to examine the morphology and physiology of the sensory retina in Sod1(-/-) mice. The amplitudes of the a- and b-waves of electroretinograms elicited by stimuli of different intensity were reduced in senescent Sod1(-/-) mice, and this reduction in amplitude was more pronounced with increasing age. Retinal morphometric analyses showed a reduced number of nuclei in both the inner nuclear cell layer and outer nuclear cell layer. Electron microscopy revealed swollen cells and degenerated mitochondria in the inner nuclear cell and outer nuclear cell layer of senescent Sod1(-/-) mice indicating necrotic cell death. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling revealed no significant differences in the number of apoptotic cells between Sod1(-/-) and wild-type mice, and activated caspase-3 could not be detected in the retina of Sod1(-/-) mice. In addition to the age-related macular degeneration-like phenotypes previously reported, Sod1(-/-) mice also present progressive retinal degeneration. Our results indicate that Sod1(-/-) mice may be a good model system in which to study the mechanism of reactive oxygen species-mediated retinal degeneration.
Collapse
Affiliation(s)
- Kouhei Hashizume
- Department of Ophthalmology, Inaida Laboratory, Keio University School of Medicine, 35-Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ramalingam A, Duhadaway JB, Sutanto-Ward E, Wang Y, Dinchuk J, Huang M, Donover PS, Boulden J, McNally LM, Soler AP, Muller AJ, Duncan MK, Prendergast GC. Bin3 deletion causes cataracts and increased susceptibility to lymphoma during aging. Cancer Res 2008; 68:1683-90. [PMID: 18339847 DOI: 10.1158/0008-5472.can-07-6072] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bin3 encodes an evolutionarily conserved and ubiquitously expressed member of the BAR superfamily of curved membrane and GTPase-binding proteins, which includes the BAR, PCH/F-BAR, and I-BAR adapter proteins implicated in signal transduction and vesicular trafficking. In humans, Bin3 maps to chromosome 8p21.3, a region widely implicated in cancer suppression that is often deleted in non-Hodgkin's lymphomas and various epithelial tumors. Yeast studies have suggested roles for this gene in filamentous actin (F-actin) organization and cell division but its physiologic functions in mammals have not been investigated. Here we report that homozygous inactivation of Bin3 in the mouse causes cataracts and an increased susceptibility to lymphomas during aging. The cataract phenotype was marked by multiple morphologic defects in lens fibers, including the development of vacuoles in cortical fibers and a near total loss of F-actin in lens fiber cells but not epithelial cells. Through 1 year of age, no other phenotypes were apparent; however, by 18 months of age, Bin3(-/-) mice exhibited a significantly increased incidence of lymphoma. Bin3 loss did not affect normal cell proliferation, F-actin organization, or susceptibility to oncogenic transformation. In contrast, it increased the proliferation and invasive motility of cells transformed by SV40 large T antigen plus activated ras. Our findings establish functions for Bin3 in lens development and cancer suppression during aging. Further, they define Bin3 as a candidate for an unidentified tumor suppressor that exists at the human chromosome 8p21.3 locus.
Collapse
|
43
|
Thiersch M, Raffelsberger W, Frigg R, Samardzija M, Wenzel A, Poch O, Grimm C. Analysis of the retinal gene expression profile after hypoxic preconditioning identifies candidate genes for neuroprotection. BMC Genomics 2008; 9:73. [PMID: 18261226 PMCID: PMC2270833 DOI: 10.1186/1471-2164-9-73] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 02/08/2008] [Indexed: 01/03/2023] Open
Abstract
Background Retinal degeneration is a main cause of blindness in humans. Neuroprotective therapies may be used to rescue retinal cells and preserve vision. Hypoxic preconditioning stabilizes the transcription factor HIF-1α in the retina and strongly protects photoreceptors in an animal model of light-induced retinal degeneration. To address the molecular mechanisms of the protection, we analyzed the transcriptome of the hypoxic retina using microarrays and real-time PCR. Results Hypoxic exposure induced a marked alteration in the retinal transcriptome with significantly different expression levels of 431 genes immediately after hypoxic exposure. The normal expression profile was restored within 16 hours of reoxygenation. Among the differentially regulated genes, several candidates for neuroprotection were identified like metallothionein-1 and -2, the HIF-1 target gene adrenomedullin and the gene encoding the antioxidative and cytoprotective enzyme paraoxonase 1 which was previously not known to be a hypoxia responsive gene in the retina. The strongly upregulated cyclin dependent kinase inhibitor p21 was excluded from being essential for neuroprotection. Conclusion Our data suggest that neuroprotection after hypoxic preconditioning is the result of the differential expression of a multitude of genes which may act in concert to protect visual cells against a toxic insult.
Collapse
Affiliation(s)
- Markus Thiersch
- Lab of Retinal Cell Biology, Dept Ophthalmology, University of Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
44
|
Karan S, Zhang H, Li S, Frederick JM, Baehr W. A model for transport of membrane-associated phototransduction polypeptides in rod and cone photoreceptor inner segments. Vision Res 2008; 48:442-52. [PMID: 17949773 PMCID: PMC2262953 DOI: 10.1016/j.visres.2007.08.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 08/21/2007] [Accepted: 08/23/2007] [Indexed: 01/29/2023]
Abstract
We discuss putative mechanisms of membrane protein transport in photoreceptors based on Pde6d and Gucy2e/Gucy2f knockout mice. Knockout of the Pde6d gene encoding PrBP/delta, a prenyl binding protein present in the retina at relatively high levels, was shown to impair transport of G-protein coupled receptor kinase 1 (GRK1) and cone phosphodiesterase alpha' subunit (PDE6alpha') to the rod and cone outer segments. Other prenylated proteins are minimally affected, suggesting some specificity of interaction. Knockout of the Gucy2e gene encoding guanylate cyclase 1 (GC1) disrupted transport of G-protein coupled receptor kinase 1 (GRK1), cone PDE6alpha', cone transducin alpha and gamma subunits (cTalpha and cTgamma) to the cone outer segments, while a GC1/GC2 double knockout prevented transport of rod PDE6, but not transducin, GRK1, or rhodopsin, to the rod outer segments. These knockout phenotypes suggest that PrBP/delta functions in extracting prenylated proteins from the endoplasmic reticulum (ER) where they dock after prenylation, and that GC-bearing membranes may co-transport peripheral membrane proteins in vesicles. We conclude that distinct pathways have evolved in rods and cones for transport of integral and peripherally membrane-associated proteins.
Collapse
Affiliation(s)
- Sukanya Karan
- Department of Biology, University of Utah, Salt Lake City, UT 84132
| | - Houbin Zhang
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Sha Li
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Jeanne M. Frederick
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Wolfgang Baehr
- Department of Biology, University of Utah, Salt Lake City, UT 84132
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132
| |
Collapse
|
45
|
Pardue MT, Walker TA, Faulkner AE, Kim MK, Bonner CM, McLean GY. Implantation of mouse eyes with a subretinal microphotodiode array. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 613:377-82. [PMID: 18188967 PMCID: PMC3001275 DOI: 10.1007/978-0-387-74904-4_44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/30/2025]
|
46
|
Abstract
Two-photon microscopy (TPM) has come to occupy a prominent place in modern biological research with its ability to resolve the three-dimensional distribution of molecules deep inside living tissue. TPM can employ two different types of signals, fluorescence and second harmonic generation, to image biological structures with subcellular resolution. Two-photon excited fluorescence imaging is a powerful technique with which to monitor the dynamic behavior of the chemical components of tissues, whereas second harmonic imaging provides novel ways to study their spatial organization. Using TPM, great strides have been made toward understanding the metabolism, structure, signal transduction, and signal transmission in the eye. These include the characterization of the spatial distribution, transport, and metabolism of the endogenous retinoids, molecules essential for the detection of light, as well as the elucidation of the architecture of the living cornea. In this review, we present and discuss the current applications of TPM for the chemical and structural imaging of the eye. In addition, we address what we see as the future potential of TPM for eye research. This relatively new method of microscopy has been the subject of numerous technical improvements in terms of the optics and indicators used, improvements that should lead to more detailed biochemical characterizations of the eyes of live animals and even to imaging of the human eye in vivo.
Collapse
Affiliation(s)
- Yoshikazu Imanishi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Kerrie Lodowski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Yiannis Koutalos
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South, Carolina
| |
Collapse
|
47
|
Calza S, Raffelsberger W, Ploner A, Sahel J, Leveillard T, Pawitan Y. Filtering genes to improve sensitivity in oligonucleotide microarray data analysis. Nucleic Acids Res 2007; 35:e102. [PMID: 17702762 PMCID: PMC2018638 DOI: 10.1093/nar/gkm537] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 06/29/2007] [Accepted: 07/03/2007] [Indexed: 12/13/2022] Open
Abstract
Many recent microarrays hold an enormous number of probe sets, thus raising many practical and theoretical problems in controlling the false discovery rate (FDR). Biologically, it is likely that most probe sets are associated with un-expressed genes, so the measured values are simply noise due to non-specific binding; also many probe sets are associated with non-differentially-expressed (non-DE) genes. In an analysis to find DE genes, these probe sets contribute to the false discoveries, so it is desirable to filter out these probe sets prior to analysis. In the methodology proposed here, we first fit a robust linear model for probe-level Affymetrix data that accounts for probe and array effects. We then develop a novel procedure called FLUSH (Filtering Likely Uninformative Sets of Hybridizations), which excludes probe sets that have statistically small array-effects or large residual variance. This filtering procedure was evaluated on a publicly available data set from a controlled spiked-in experiment, as well as on a real experimental data set of a mouse model for retinal degeneration. In both cases, FLUSH filtering improves the sensitivity in the detection of DE genes compared to analyses using unfiltered, presence-filtered, intensity-filtered and variance-filtered data. A freely-available package called FLUSH implements the procedures and graphical displays described in the article.
Collapse
Affiliation(s)
- Stefano Calza
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| | - Wolfgang Raffelsberger
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| | - Alexander Ploner
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| | - Jose Sahel
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| | - Thierry Leveillard
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics - Karolinska Institute, Stockholm, Sweden, Section of Medical Statistics and Biometry, Department of Biomedical Sciences and Biotechnology - University of Brescia, Italy, Laboratoire de Bioinformatique et Génomique Intégratives, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch Strasbourg, France and Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Retine - Faculté de Médecine, Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
48
|
Nickell S, Park PSH, Baumeister W, Palczewski K. Three-dimensional architecture of murine rod outer segments determined by cryoelectron tomography. J Cell Biol 2007; 177:917-25. [PMID: 17535966 PMCID: PMC2064290 DOI: 10.1083/jcb.200612010] [Citation(s) in RCA: 184] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 05/03/2007] [Indexed: 12/13/2022] Open
Abstract
The rod outer segment (ROS) of photoreceptor cells houses all components necessary for phototransduction, a set of biochemical reactions that amplify and propagate a light signal. Theoretical approaches to quantify this process require precise information about the physical boundaries of the ROS. Dimensions of internal structures within the ROS of mammalian species have yet to be determined with the precision required for quantitative considerations. Cryoelectron tomography was utilized to obtain reliable three-dimensional morphological information about this important structure from murine retina. Vitrification of samples permitted imaging of the ROS in a minimally perturbed manner and the preservation of substructures. Tomograms revealed the characteristic highly organized arrangement of disc membranes stacked on top of one another with a surrounding plasma membrane. Distances among the various membrane components of the ROS were measured to define the space available for phototransduction to occur. Reconstruction of segments of the ROS from single-axis tilt series images provided a glimpse into the three-dimensional architecture of this highly differentiated neuron. The reconstructions revealed spacers that likely maintain the proper distance between adjacent discs and between discs and the plasma membrane. Spacers were found distributed throughout the discs, including regions that are distant from the rim region of discs.
Collapse
Affiliation(s)
- Stephan Nickell
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | | | | | | |
Collapse
|
49
|
Marmorstein AD, Marmorstein LY. The challenge of modeling macular degeneration in mice. Trends Genet 2007; 23:225-31. [PMID: 17368622 DOI: 10.1016/j.tig.2007.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 01/08/2007] [Accepted: 03/01/2007] [Indexed: 01/17/2023]
Abstract
Macular degenerations (MD), age-related or inherited, interfere with the ability to read, drive and recognize faces. Understanding this class of diseases has been challenging because the mouse, the mammal most amenable to genetic manipulation, lacks a macula. Here we discuss whether we can model MD in the mouse, present criteria for an 'ideal' mouse model of MD and discuss how mouse models have contributed to our knowledge of MD by contrasting how well they meet the 'ideal' criteria with how informative they have actually been. By modeling MD in mice, we can learn about aspects of MD that an animal with a macula would be unable to teach us.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, 655 North Alvernon Way, Suite 108, Tucson, AZ 85711, USA.
| | | |
Collapse
|
50
|
Perveen R, Favor J, Jamieson RV, Ray DW, Black GCM. A heterozygous c-Maf transactivation domain mutation causes congenital cataract and enhances target gene activation. Hum Mol Genet 2007; 16:1030-8. [PMID: 17374726 DOI: 10.1093/hmg/ddm048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
MAF, one of a family of large Maf bZIP transcription factors, is mutated in human developmental ocular disorders that include congenital cataract, microcornea, coloboma and anterior segment dysgenesis. Expressed early in the developing lens vesicle, it is central to regulation of lens crystallin gene expression. We report a semi-dominant mouse c-Maf mutation recovered after ENU mutatgenesis which results in the substitution, D90V, at a highly conserved residue within the N-terminal 35 amino-acid minimal transactivation domain (MTD). Unlike null and loss-of-function c-Maf mutations, which cause severe runting and renal abnormalities, the phenotype caused by the D90V mutation is isolated cataract. In reporter assays, D90V results in increased promoter activation, a situation similar to MTD mutations of NRL that also cause human disease. In contrast to wild-type protein, the c-Maf D90V mutant protein is not inhibited by protein kinase A-dependent pathways. The MTD of large Maf proteins has been shown to interact with the transcriptional co-activator p300 and we demonstrate that c-Maf D90V enhances p300 recruitment in a cell-type dependent manner. We observed the same for the pathogenic human NRL MTD mutation S50T, which suggests a common mechanism of action.
Collapse
Affiliation(s)
- R Perveen
- Academic Unit of Medical Genetics and Regional Genetics Service Department of Clinical Genetics, Central Manchester and Manchester Children's University Hospitals NHS Trust, St Mary's Hospital, Hathersage Road, Manchester M13 0JH, UK
| | | | | | | | | |
Collapse
|