1
|
Gogna N, Pinkney J, Stone L, Khorzom MM, Zhao F, Collin GB, Naggert JK, Krebs MP, Nishina PM. A biometric survey of known and prospective murine models of posterior microphthalmia-nanophthalmia. Exp Eye Res 2025; 255:110335. [PMID: 40154727 DOI: 10.1016/j.exer.2025.110335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Posterior microphthalmia and nanophthalmia are related genetic conditions that disrupt ocular growth. Here, we conducted a biometric analysis of mouse models to assess shared features of these diseases. Three known microphthalmia alleles (Mfrprd6, Prss56glcr4, and Adipor1tm1Dgen) and two prospective alleles (C1qtnf5tm1.1(KOMP)Vlcg and Prss56em2(IMPC)J) were introgressed onto the C57BL/6J (B6) genetic background and compared to B6 mice at 1 through 12 months of age. Biometric parameters obtained using optical coherence tomography were analyzed statistically to identify strain differences. Fundus imaging and histological analyses were performed to assess ocular morphology. Mfrprd6, Prss56glcr4, and Prss56em2(IMPC)J mice had significantly shorter axial and posterior lengths, and longer anterior chamber depth compared to controls at all ages studied. Adipor1tm1Dgen mice exhibited similar, but less severe, biometric changes. Axial length was not significantly changed in C1qtnf5tm1.1(KOMP)Vlcg mice, but reduced anterior chamber depth and increased lens thickness were observed at one month of age. Lens and corneal thicknesses were otherwise unchanged in the models as compared to B6 controls. Corneal radius of curvature, examined at 4 months of age, was significantly decreased in all models relative to controls. Micropthalmia was observed independent of retinal degeneration (Mfrprd6, Adipor1tm1Dgen) or retinal thickening (Prss56 mutants). Prss56 mutants developed retinal folds that were absent from other mutants and controls. We conclude that, in mice, Mfrp, Prss56, and Adipor1 mutations yield similar microphthalmia phenotypes involving both the anterior and posterior eye. Changes to anterior chamber depth, lens thickness, and corneal curvature in C1qtnf5tm1.1(KOMP)Vlcg mice suggest a role of C1qtnf5 in anterior ocular growth.
Collapse
Affiliation(s)
- Navdeep Gogna
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Jai Pinkney
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Lisa Stone
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | | | - Fuxin Zhao
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Gayle B Collin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | | | - Mark P Krebs
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - Patsy M Nishina
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
2
|
Sze YH, Zuo B, Lu DQ, Li KK, Tse DYY, Zhao Q, Lam TC. Comparative analysis of ocular biometrics using spectral domain optical coherence tomography with Purkinje image and optic nerve head alignments in mice. Eur J Med Res 2025; 30:67. [PMID: 39901220 PMCID: PMC11789383 DOI: 10.1186/s40001-025-02305-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Mice are an emerging model for experimental myopia. Due to their small eye size, non-invasive optical coherence tomography is essential for evaluating ocular biometrics. There is currently no universally accepted protocol for those measurements. This study aims to compare ocular biometric measurements using two methods: Purkinje image-based alignment and optic nerve head alignment, utilizing spectral domain optical coherence tomography. Gaining an understanding of the implications of these methods in determining axial elongation in the normal growing eyes of wild-type C57BL/6J mice would offer valuable insight into their relevance for the experimental myopia model. METHODS Ocular dimensions and refractive development were measured on postnatal days P21 (n = 10), P28 (n = 15), and P35 (n = 8). The Purkinje image-based alignment (P1) was determined using a photorefractor and aligned perpendicular to the corneal apex using SD-OCT. In comparison, due to the absence of a fovea in the mouse retina, the optic nerve head (ONH) alignment was used. Variance analysis, regression analysis, and Bland-Altman analysis were performed to compare the differences between alignment methods as well as the replication by another operator. RESULTS Mice developed hyperopic ametropia under normal visual conditions. The photorefractor measured a technical variation of 3.9 D (95% CI, n = 170, triplicates). Bland-Altman analysis revealed a shorter (mean ± SD) axial length (- 26.4 ± 18.1 μm) and vitreous chamber depth (- 39.9 ± 25.4 μm) in the Purkinje image-based alignment. There was a significant difference in the relative growth trend in VCD (linear regression, p = 0.02), which was relatively stable and showed shortening when measured with ONH alignment from postnatal age 21 to 35 days. CONCLUSIONS SD-OCT allowed precise in-vivo measurement and segmentation of ocular dimensions, regardless of the methods adopted. P1 alignment consistently resulted in significantly shorter VCD and AL compared to ONH alignment at most time points. When considering temporal changes from P21 to P35, both methods showed similar results, with significant elongation of ACD, LT, and AL as expected. However, our findings revealed a significant shortening of VCD over time with the adoption of ONH alignment, while the change in P1 alignment was relatively stable. Therefore, AL provides a better measure for evaluating ocular growth in mice using optical coherence tomography than VCD for myopia research.
Collapse
Affiliation(s)
- Ying Hon Sze
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China
| | - Bing Zuo
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Da Qian Lu
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - King Kit Li
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Dennis Yan Yin Tse
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Thomas Chuen Lam
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China.
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China.
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, 518052, China.
| |
Collapse
|
3
|
Isaacson M, Chang H, Berkowitz L, Zirkel R, Park Y, Hu D, Ellwood I, Schaffer CB. MouseGoggles: an immersive virtual reality headset for mouse neuroscience and behavior. Nat Methods 2025; 22:380-385. [PMID: 39668209 PMCID: PMC11810773 DOI: 10.1038/s41592-024-02540-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/24/2024] [Indexed: 12/14/2024]
Abstract
Small-animal virtual reality (VR) systems have become invaluable tools in neuroscience for studying complex behavior during head-fixed neural recording, but they lag behind commercial human VR systems in terms of miniaturization, immersivity and advanced features such as eye tracking. Here we present MouseGoggles, a miniature VR headset for head-fixed mice that delivers independent, binocular visual stimulation over a wide field of view while enabling eye tracking and pupillometry in VR. Neural recordings in the visual cortex validate the quality of image presentation, while hippocampal recordings, associative reward learning and innate fear responses to virtual looming stimuli demonstrate an immersive VR experience. Our open-source system's simplicity and compact size will enable the broader adoption of VR methods in neuroscience.
Collapse
Affiliation(s)
- Matthew Isaacson
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| | - Hongyu Chang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Laura Berkowitz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Rick Zirkel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Yusol Park
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Danyu Hu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Ian Ellwood
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
4
|
Zheng L, Liao Z, Zou J. Animal modeling for myopia. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2024; 4:173-181. [PMID: 39263386 PMCID: PMC11385420 DOI: 10.1016/j.aopr.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 09/13/2024]
Abstract
Background Myopia is one of the most common eye diseases globally, and has become an increasingly serious health concern among adolescents. Understanding the factors contributing to the onset of myopia and the strategies to slow its progression is critical to reducing its prevalence. Main text Animal models are key to understanding of the etiology of human diseases. Various experimental animal models have been developed to mimic human myopia, including chickens, rhesus monkeys, marmosets, mice, tree shrews, guinea pigs and zebrafish. Studies using these animal models have provided evidences and perspectives on the regulation of eye growth and refractive development. This review summarizes the characteristics of these models, the induction methods, common indicators of myopia in animal models, and recent findings on the pathogenic mechanism of myopia. Conclusions Investigations using experimental animal models have provided valuable information and insights into the pathogenic mechanisms of human myopia and its treatment strategies.
Collapse
Affiliation(s)
- Lingman Zheng
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Zhiyong Liao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Jian Zou
- Eye Center of the Second Affiliated Hospital, Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Wen Y, Li Y, Zhu L, Tang T, Yan H, Hu J, Wang K, Zhao M, Xu Q. Nonlinear pathological trajectory of a high-myopia C57/BL6J mouse model induced by form deprivation. Front Physiol 2024; 15:1442000. [PMID: 39539955 PMCID: PMC11557532 DOI: 10.3389/fphys.2024.1442000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction To establish a high myopia model in C57BL/6J mice with monocular form deprivation myopia (FDM) and investigate its ocular structure pathological trajectory. Methods Healthy 3-week-old C57BL/6J mice were divided into an FDM group (n = 36) and a control group (n = 24). The left eyes of the FDM group were patched, while the right eyes served as controls. Biometric parameters and fundus morphology were assessed at baseline and after 4, 8, and 12 weeks of form deprivation. Results Significant differences were observed in the deprived eyes, including longer axial length, higher refractive power, deeper vitreous chambers, thinner retina, choroid, and sclera, and smaller scleral fibers' diameters under a transmission electron microscope. Retinal vascular area proportion in covered eyes decreased significantly (P < 0.05), with a decline rate of 11% from weeks 4 to 8 and a faster decline of 19% from weeks 8 to 12, while this proportion increased significantly in control eyes. Discussion This study successfully induced a high myopia model in mice with long-term form deprivation. The axial length grew dramatically in FDM in the first 8 weeks, while the pathological progress of the fundus accelerated from weeks 8 to 12.
Collapse
Affiliation(s)
- Yue Wen
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Yan Li
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Li Zhu
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Tao Tang
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Huichao Yan
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Jie Hu
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Kai Wang
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| | - Qiong Xu
- Department of Ophthalmology and Clinical Center of Optometry, Peking University People’s Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and ChoroidDiseases, Beijing, China
| |
Collapse
|
6
|
Lamirande P, Gaffney EA, Gertz M, Maini PK, Crawshaw JR, Caruso A. A First-Passage Model of Intravitreal Drug Delivery and Residence Time-Influence of Ocular Geometry, Individual Variability, and Injection Location. Invest Ophthalmol Vis Sci 2024; 65:21. [PMID: 39412819 PMCID: PMC11488524 DOI: 10.1167/iovs.65.12.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/12/2024] [Indexed: 10/20/2024] Open
Abstract
Purpose Standard of care for various retinal diseases involves recurrent intravitreal injections. This motivates mathematical modeling efforts to identify influential factors for ocular drug residence time, aiming to minimize administration frequency. We sought to describe the vitreal diffusion of therapeutics in nonclinical species frequently used during drug development assessments. In human eyes, we investigated the impact of variability in vitreous cavity size and eccentricity, and in injection location, on drug disposition. Methods Using a first-passage time approach, we modeled the transport-controlled distribution of two standard therapeutic protein formats (Fab and IgG) and elimination through anterior and posterior pathways. Anatomical three-dimensional geometries of mouse, rat, rabbit, cynomolgus monkey, and human eyes were constructed using ocular images and biometry datasets. A scaling relationship was derived for comparison with experimental ocular half-lives. Results Model simulations revealed a dependence of residence time on ocular size and injection location. Delivery to the posterior vitreous resulted in increased vitreal half-life and retinal permeation. Interindividual variability in human eyes had a significant influence on residence time (half-life range of 5-7 days), showing a strong correlation to axial length and vitreal volume. Anterior exit was the predominant route of drug elimination. Contribution of the posterior pathway displayed a 3% difference between protein formats but varied between species (10%-30%). Conclusions The modeling results suggest that experimental variability in ocular half-life is partially attributed to anatomical differences and injection site location. Simulations further suggest a potential role of the posterior pathway permeability in determining species differences in ocular pharmacokinetics.
Collapse
Affiliation(s)
- Patricia Lamirande
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Eamonn A. Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Michael Gertz
- Pharmaceutical Sciences, Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Jessica R. Crawshaw
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Antonello Caruso
- Pharmaceutical Sciences, Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| |
Collapse
|
7
|
Tang T, Ren C, Cai Y, Li Y, Wang K, Zhao M. Lifelong Changes in the Choroidal Thickness, Refractive Status, and Ocular Dimensions in C57BL/6J Mouse. Invest Ophthalmol Vis Sci 2024; 65:26. [PMID: 39422919 PMCID: PMC11500047 DOI: 10.1167/iovs.65.12.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 08/25/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose To investigate the changes in choroidal thickness (ChT), refractive status, and ocular dimensions in the mouse eye in vivo using updated techniques and instrumentation. Methods High-resolution swept-source optical coherence tomography (SS-OCT), eccentric infrared photoretinoscopy, and custom real-time optical coherence tomography were used to analyze choroidal changes, refractive changes and ocular growth in C57BL/6J mice from postnatal day (P) 21 to month 22. Results The ChT gradually increased with age, with the thickest region in the para-optic nerve head and thinning outward, and the temporal ChT was globally thicker than the nasal ChT. Retinal thickness remained stable until 4 months and subsequently decreased. The average spherical equivalent refraction error was -4.81 ± 2.71 diopters (D) at P21, which developed into emmetropia by P32, reached a hyperopic peak (+5.75 ± 1.38 D) at P82 and returned to +0.66 ± 1.86 D at 22 months. Central corneal thickness, anterior chamber depth, lens thickness, and axial length (AL) increased continuously before 4 months, but subsequently exhibited subtle changes. Vitreous chamber depth decreased with lens growth. ChT was correlated significantly with the ocular parameters (except for retinal thickness) before the age of 4 months, but these correlations diminished after 4 months. Furthermore, for mice younger than 4 months, the difference in the ChT, especially temporal ChT, between the two eyes contributed most to that of axial length and spherical equivalent refraction error. Conclusions Four months could be a watershed age in the growth of mouse eyes. Large-span temporal recordings of refraction, ocular dimensions, and choroidal changes provided references for the study of the physiological and pathological mechanisms responsible for myopia.
Collapse
Affiliation(s)
- Tao Tang
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
- The Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Chi Ren
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Yi Cai
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Yan Li
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Kai Wang
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- College of Optometry, Peking University Health Science Center, Beijing, China
- Eye Disease and Optometry Institute, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of the Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| |
Collapse
|
8
|
Skelin L, Racetin A, Kelam N, Ogorevc M, Znaor L, Saraga-Babić M, Filipović N, Katsuyama Y, Pogorelić Z, Vukojević K. Connexin Expression Is Altered in the Eye Development of Yotari Mice: A Preliminary Study. Biomolecules 2024; 14:1174. [PMID: 39334940 PMCID: PMC11430515 DOI: 10.3390/biom14091174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to explore how Dab1 functional silencing influences the expression patterns of different connexins in the developing yotari (yot) mice eyes as potential determinants of retinogenesis. Using immunofluorescence staining, the protein expression of Dab1, Reelin, and connexin 37, 40, 43, and 45 (Cx37, Cx40, Cx43, and Cx45) in the wild-type (wt) and yot eyes at embryonic days 13.5 and 15.5 (E13.5 and E15.5) were analyzed. Different expression patterns of Cx37 were seen between the wt and yot groups. The highest fluorescence intensity of Cx37 was observed in the yot animals at E15.5. Cx40 had higher expression at the E13.5 when differentiation of retinal layers was still beginning, whereas it decreased at the E15.5 when differentiation was at the advanced stage. Higher expression of Cx43 was found in the yot group at both time points. Cx45 was predominantly expressed at E13.5 in both groups. Our results reveal the altered expression of connexins during retinogenesis in yot mice and their potential involvement in retinal pathology, where they might serve as prospective therapeutic targets.
Collapse
Affiliation(s)
- Ljubica Skelin
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Ljubo Znaor
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
- Department of Ophthalmology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Zenon Pogorelić
- Department of Pediatric Surgery, Split University Hospital, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| |
Collapse
|
9
|
Wilmet B, Michiels C, Zhang J, Callebert J, Sahel JA, Picaud S, Audo I, Zeitz C. Loss of ON-Pathway Function in Mice Lacking Lrit3 Decreases Recovery From Lens-Induced Myopia. Invest Ophthalmol Vis Sci 2024; 65:18. [PMID: 39250117 PMCID: PMC11385651 DOI: 10.1167/iovs.65.11.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Abstract
Purpose To determine whether the Lrit3-/- mouse model of complete congenital stationary night blindness with an ON-pathway defect harbors myopic features and whether the genetic defect influences the recovery from lens-induced myopia. Methods Retinal levels of dopamine (DA) and 3,4 dihydroxyphenylacetic acid (DOPAC) from adult isolated Lrit3-/- retinas were quantified using ultra performance liquid chromatography after light adaptation. Natural refractive development of Lrit3-/- mice was measured from three weeks to nine weeks of age using an infrared photorefractometer. Susceptibility to myopia induction was assessed using a lens-induced myopia protocol with -25 D lenses placed in front of the right eye of the animals for three weeks; the mean interocular shift was measured with an infrared photorefractometer after two and three weeks of goggling and after one and two weeks after removal of goggles. Results Compared to wild-type littermates (Lrit3+/+), both DA and DOPAC were drastically reduced in Lrit3-/- retinas. Natural refractive development was normal but Lrit3-/- mice showed a higher myopic shift and a lower ability to recover from induced myopia. Conclusions Our data consolidate the link between ON pathway defect altered dopaminergic signaling and myopia. We document for the first time the role of ON pathway on the recovery from myopia induction.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Jingyi Zhang
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburg, PA, United States
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
10
|
Navneet S, Ishii M, Rohrer B. Altered Elastin Turnover, Immune Response, and Age-Related Retinal Thinning in a Transgenic Mouse Model With RPE-Specific HTRA1 Overexpression. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 39028977 PMCID: PMC11262478 DOI: 10.1167/iovs.65.8.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
Purpose A single-nucleotide polymorphism in HTRA1 has been linked to age-related macular degeneration (AMD). Here we investigated the potential links between age-related retinal changes, elastin turnover, elastin autoantibody production, and complement C3 deposition in a mouse model with RPE-specific human HTRA1 overexpression. Methods HTRA1 transgenic mice and age-matched CD1 wild-type mice were analyzed at 6 weeks and 4, 6, and 12 to 14 months of age using in vivo retinal imaging by optical coherence tomography (OCT) and fundus photography, as well as molecular readouts, focusing on elastin and elastin-derived peptide quantification, antielastin autoantibody, and total Ig antibody measurements and immunohistochemistry to examine elastin, IgG, and C3 protein levels in retinal sections. Results OCT imaging indicated thinning of inner nuclear layer as an early phenotype in HTRA1 mice, followed by age and age/genotype-related thinning of the photoreceptor layer, RPE, and total retina. HTRA1 mice exhibited reduced elastin protein levels in the RPE/choroid and increased elastin breakdown products in the retina and serum. A corresponding age-dependent increase of serum antielastin IgG and IgM autoantibodies and total Ig antibody levels was observed. In the RPE/choroid, these changes were associated with an age-related increase of IgG and C3 deposition. Conclusions Our results confirm that RPE-specific overexpression of human HTRA1 induces certain AMD-like phenotypes in mice. This includes altered elastin turnover, immune response, and complement deposition in the RPE/choroid in addition to age-related outer retinal and photoreceptor layer thinning. The identification of elastin-derived peptides and corresponding antielastin autoantibodies, together with increased C3 deposition in the RPE/choroid, provides a rationale for an overactive complement system in AMD irrespective of the underlying genetic risk.
Collapse
Affiliation(s)
- Soumya Navneet
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Masaaki Ishii
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States
- Ralph H. Johnson VA Medical Center, Division of Research, Charleston, South Carolina, United States
| |
Collapse
|
11
|
Donaldson PJ, Petrova RS, Nair N, Chen Y, Schey KL. Regulation of water flow in the ocular lens: new roles for aquaporins. J Physiol 2024; 602:3041-3056. [PMID: 37843390 PMCID: PMC11018719 DOI: 10.1113/jp284102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023] Open
Abstract
The ocular lens is an important determinant of overall vision quality whose refractive and transparent properties change throughout life. The lens operates an internal microcirculation system that generates circulating fluxes of ions, water and nutrients that maintain the transparency and refractive properties of the lens. This flow of water generates a substantial hydrostatic pressure gradient which is regulated by a dual feedback system that uses the mechanosensitive channels TRPV1 and TRPV4 to sense decreases and increases, respectively, in the pressure gradient. This regulation of water flow (pressure) and hence overall lens water content, sets the two key parameters, lens geometry and the gradient of refractive index, which determine the refractive properties of the lens. Here we focus on the roles played by the aquaporin family of water channels in mediating lens water fluxes, with a specific focus on AQP5 as a regulated water channel in the lens. We show that in addition to regulating the activity of ion transporters, which generate local osmotic gradients that drive lens water flow, the TRPV1/4-mediated dual feedback system also modulates the membrane trafficking of AQP5 in the anterior influx pathway and equatorial efflux zone of the lens. Since both lens pressure and AQP5-mediated water permeability (P H 2 O ${P_{{{\mathrm{H}}_{\mathrm{2}}}{\mathrm{O}}}}$ ) can be altered by changes in the tension applied to the lens surface via modulating ciliary muscle contraction we propose extrinsic modulation of lens water flow as a potential mechanism to alter the refractive properties of the lens to ensure light remains focused on the retina throughout life.
Collapse
Affiliation(s)
- Paul J. Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Center, University of Auckland, Auckland, New Zealand
| | - Rosica S. Petrova
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Center, University of Auckland, Auckland, New Zealand
| | - Nikhil Nair
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Center, University of Auckland, Auckland, New Zealand
| | - Yadi Chen
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Center, University of Auckland, Auckland, New Zealand
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
12
|
Wang K, Pu Y, Chen L, Hoshino M, Uesugi K, Yagi N, Chen X, Usui Y, Hanashima A, Hashimoto K, Mohri S, Pierscionek BK. Optical development in the murine eye lens of accelerated senescence-prone SAMP8 and senescence-resistant SAMR1 strains. Exp Eye Res 2024; 241:109858. [PMID: 38467176 DOI: 10.1016/j.exer.2024.109858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
The eye lens is responsible for focusing objects at various distances onto the retina and its refractive power is determined by its surface curvature as well as its internal gradient refractive index (GRIN). The lens continues to grow with age resulting in changes to the shape and to the GRIN profile. The present study aims to investigate how the ageing process may influence lens optical development. Murine lenses of accelerated senescence-prone strain (SAMP8) aged from 4 to 50 weeks; senescence-resistant strain (SAMR1) aged from 5 to 52 weeks as well as AKR strain (served as control) aged from 6 to 70 weeks were measured using the X-ray interferometer at the SPring-8 synchrotron Japan within three consecutive years from 2020 to 2022. Three dimensional distributions of the lens GRIN were reconstructed using the measured data and the lens shapes were determined using image segmentation in MatLab. Variations in the parameters describing the lens shape and the GRIN profile with age were compared amongst three mouse strains. With advancing age, both the lens anterior and posterior surface flattens and the lens sagittal thickness increase in all three mouse strains (Anterior radius of curvature increase at 0.008 mm/week, 0.007 mm/week and 0.002 mm/week while posterior radius of curvature increase at 0.002 mm/week, 0.007 mm/week and 0.003 mm/week respectively in AKR, SAMP8 and SAMR1 lenses). Compared with the AKR strain, the SAMP8 samples demonstrate a higher rate of increase in the posterior curvature radius (0.007 mm/week) and the thickness (0.015 mm/week), whilst the SAMR1 samples show slower increases in the anterior curvature radius (0.002 mm/week) and its thickness (0.013 mm/week). There are similar age-related trends in GRIN shape in the radial direction (in all three types of murine lenses nr2 and nr6 increase with age while nr4 decrease with age consistently) but not in the axial direction amongst three mouse strains (nz1 of AKR lens decrease while of SAMP8 and SAMR1 increase with age; nz2 of all three models increase with age; nz3 of AKR lens increase while of SAMP8 and SAMR1 decrease with age). The ageing process can influence the speed of lens shape change and affect the GRIN profile mainly in the axial direction, contributing to an accelerated decline rate of the optical power in the senescence-prone strain (3.5 D/week compared to 2.3 D/week in the AKR control model) but a retardatory decrease in the senescence-resistant strain (2.1 D/week compared to the 2.3D/week in the AKR control model).
Collapse
Affiliation(s)
- Kehao Wang
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yutian Pu
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Leran Chen
- Peking University First Hospital, Beijing, China.
| | - Masato Hoshino
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Kentaro Uesugi
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Naoto Yagi
- Japan Synchrotron Radiation Research Institute (Spring-8), 1-1-1, Kouto, Sayo-cho, Sayo-gun, Hyogo, 679-5198, Japan.
| | - Xiaoyong Chen
- Department of Ophthalmology, Peking University Third Hospital, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, China.
| | - Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Barbara K Pierscionek
- Faculty of Health, Education, Medicine and Social Care, Medical Technology Research Centre, Anglia Ruskin University, Bishops Hall Lane, Chelmsford, United Kingdom.
| |
Collapse
|
13
|
Pan X, Muir ER, Sellitto C, Wang K, Cheng C, Pierscionek B, Donaldson PJ, White TW. Age-Dependent Changes in the Water Content and Optical Power of the In Vivo Mouse Lens Revealed by Multi-Parametric MRI and Optical Modeling. Invest Ophthalmol Vis Sci 2023; 64:24. [PMID: 37079314 PMCID: PMC10132318 DOI: 10.1167/iovs.64.4.24] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023] Open
Abstract
Purpose The purpose of this study was to utilize in vivo magnetic resonance imaging (MRI) and optical modeling to investigate how changes in water transport, lens curvature, and gradient refractive index (GRIN) alter the power of the mouse lens as a function of age. Methods Lenses of male C57BL/6 wild-type mice aged between 3 weeks and 12 months (N = 4 mice per age group) were imaged using a 7T MRI scanner. Measurements of lens shape and the distribution of T2 (water-bound protein ratios) and T1 (free water content) values were extracted from MRI images. T2 values were converted into the refractive index (n) using an age-corrected calibration equation to calculate the GRIN at different ages. GRIN maps and shape parameters were inputted into an optical model to determine ageing effects on lens power and spherical aberration. Results The mouse lens showed two growth phases. From 3 weeks to 3 months, T2 decreased, GRIN increased, and T1 decreased. This was accompanied by increased lens thickness, volume, and surface radii of curvatures. The refractive power of the lens also increased significantly, and a negative spherical aberration was developed and maintained. Between 6 and 12 months of age, all physiological, geometrical, and optical parameters remained constant, although the lens continued to grow. Conclusions In the first 3 months, the mouse lens power increased as a result of changes in shape and in the GRIN, the latter driven by the decreased water content of the lens nucleus. Further research into the mechanisms regulating this decrease in mouse lens water could improve our understanding of how lens power changes during emmetropization in the developing human lens.
Collapse
Affiliation(s)
- Xingzheng Pan
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Eric R. Muir
- Department of Radiology, School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Caterina Sellitto
- Department of Physiology & Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Kehao Wang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Engineering Medicine, Beihang University, Beijing, China
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, Indiana, United States
| | - Barbara Pierscionek
- Faculty of Health, Education, Medicine and Social Care, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford Campus, United Kingdom
| | - Paul J. Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Thomas W. White
- Department of Physiology & Biophysics, School of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
14
|
Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, Audo I, Picaud S, Zeitz C. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int J Mol Sci 2022; 24:ijms24010219. [PMID: 36613663 PMCID: PMC9820543 DOI: 10.3390/ijms24010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
| | - Robert Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruben Goulet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christophe Tourain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
| | - Christelle Michiels
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
- Académie des Sciences, Institut de France, 75006 Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| |
Collapse
|
15
|
Regulation of lens water content: Effects on the physiological optics of the lens. Prog Retin Eye Res 2022:101152. [DOI: 10.1016/j.preteyeres.2022.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022]
|
16
|
Li Y, Lu Y, Du K, Yin Y, Hu T, Fu Q, Zhang Y, Wen D, Wu X, Xia X. RNA-sequencing analysis reveals the long noncoding RNA profile in the mouse myopic retina. Front Genet 2022; 13:1014031. [PMID: 36313450 PMCID: PMC9606684 DOI: 10.3389/fgene.2022.1014031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2023] Open
Abstract
Aim: Myopia is a prevalent public health problem. The long noncoding RNA (lncRNA) mechanisms for dysregulated retinal signaling in the myopic eye have remained elusive. The aim of this study was to analyze the expression profiles and possible pathogenic roles of lncRNAs in mouse form-deprived myopia (FDM) retinas. Methods: A mouse FDM model was induced and retinas from the FDM right eyes and the contralateral eyes were collected for RNA sequencing. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and lncRNA-mRNA coexpression network analyses were conducted to explore the biological functions of the differentially expressed lncRNAs. In addition, the levels of differentially expressed lncRNAs in the myopic retinas were validated by quantitative real-time PCR (qRT-PCR). Fluorescence in situ hybridization (FISH) was used to detect the localization of lncRNAs in mouse retinas. Results: FDM eyes exhibited reduced refraction and increased ocular axial length compared to control fellow eyes. RNA sequencing revealed that there were 655 differentially expressed lncRNAs between the FDM and control retinas. Functional enrichment analysis indicated that the differentially expressed RNAs were mostly enriched in cellular processes, cytokine-cytokine receptor interactions, retinol metabolism, and rhythmic processes. Differentially expressed lncRNAs were validated by qRT-PCR. Additionally, RNA FISH showed that XR_384718.4 (Gm35369) localized in the ganglion cell (GCL) and inner nuclear layers (INL). Conclusion: This study identified the differential expression profiles of lncRNAs in myopic mouse retinas. Our results provide scientific evidence for investigations of myopia and the development of putative interventions in the future.
Collapse
Affiliation(s)
- Yuanjun Li
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Lu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kaixuan Du
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yewei Yin
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tu Hu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qiuman Fu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yanni Zhang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dan Wen
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoying Wu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Quantitative Optical Coherence Tomography for Longitudinal Monitoring of Postnatal Retinal Development in Developing Mouse Eyes. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041860] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A better study of postnatal retinal development is essential for the in-depth understanding of the nature of the vision system. To date, quantitative analysis of postnatal retinal development is primarily limited to endpoint histological examination. This study is to validate in vivo optical coherence tomography (OCT) for longitudinal monitoring of postnatal retinal development in developing mouse eyes. OCT images of C57BL/6J mice were recorded from postnatal day (P) 14 to P56. Three-dimensional (3D) frame registration and super averaging were adopted to investigate the fine structure of the retina. Quantitative OCT analysis revealed distinct outer and inner retinal layer changes, corresponding to eye development. At the outer retina, external limiting membrane (ELM) and ellipsoid zone (EZ) band intensities gradually increased with aging, and the IZ band was detectable by P28. At the inner retina, a hyporeflective layer (HRL) between the nerve fiber layer (NFL) and inner plexiform layer (IPL) was observed in developing eyes and gradually disappeared with aging. Further image analysis revealed individual RGCs within the HRL layer of the young mouse retina. However, RGCs were merged with the NFL and the IPL in the aged mouse retina. Moreover, the sub-IPL layer structure was observed to be gradually enhanced with aging. To interpret the observed retinal layer kinetics, a model based on eyeball expansion, cell apoptosis, and retinal structural modification was proposed.
Collapse
|
18
|
Holmgren CD, Stahr P, Wallace DJ, Voit KM, Matheson EJ, Sawinski J, Bassetto G, Kerr JND. Visual pursuit behavior in mice maintains the pursued prey on the retinal region with least optic flow. eLife 2021; 10:e70838. [PMID: 34698633 PMCID: PMC8547958 DOI: 10.7554/elife.70838] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022] Open
Abstract
Mice have a large visual field that is constantly stabilized by vestibular ocular reflex (VOR) driven eye rotations that counter head-rotations. While maintaining their extensive visual coverage is advantageous for predator detection, mice also track and capture prey using vision. However, in the freely moving animal quantifying object location in the field of view is challenging. Here, we developed a method to digitally reconstruct and quantify the visual scene of freely moving mice performing a visually based prey capture task. By isolating the visual sense and combining a mouse eye optic model with the head and eye rotations, the detailed reconstruction of the digital environment and retinal features were projected onto the corneal surface for comparison, and updated throughout the behavior. By quantifying the spatial location of objects in the visual scene and their motion throughout the behavior, we show that the prey image consistently falls within a small area of the VOR-stabilized visual field. This functional focus coincides with the region of minimal optic flow within the visual field and consequently area of minimal motion-induced image-blur, as during pursuit mice ran directly toward the prey. The functional focus lies in the upper-temporal part of the retina and coincides with the reported high density-region of Alpha-ON sustained retinal ganglion cells.
Collapse
Affiliation(s)
- Carl D Holmgren
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Paul Stahr
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Damian J Wallace
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Kay-Michael Voit
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Emily J Matheson
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Juergen Sawinski
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| | - Giacomo Bassetto
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
- Machine Learning in Science, Eberhard Karls University of TübingenTübingenGermany
| | - Jason ND Kerr
- Department of Behavior and Brain Organization, Research center caesarBonnGermany
| |
Collapse
|
19
|
Brais-Brunet S, Heckel É, Kanniyappan U, Chemtob S, Boudoux C, Joyal JS, Dehaes M. Morphometric and Microstructural Changes During Murine Retinal Development Characterized Using In Vivo Optical Coherence Tomography. Invest Ophthalmol Vis Sci 2021; 62:20. [PMID: 34698774 PMCID: PMC8556565 DOI: 10.1167/iovs.62.13.20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose The purpose of this study was to develop an in vivo optical coherence tomography (OCT) system capable of imaging the developing mouse retina and its associated morphometric and microstructural changes. Methods Thirty-four wild-type mice (129S1/SvlmJ) were anesthetized and imaged between postnatal (P) day 7 and P21. OCT instrumentation was developed to optimize signal intensity and image quality. Semi-automatic segmentation tools were developed to quantify the retinal thickness of the nerve fiber layer (NFL), inner plexiform layer (IPL), inner nuclear layer (INL), and the outer retinal layers (ORL), in addition to the total retina. The retinal maturation was characterized by comparing layer thicknesses between consecutive time points. Results From P7 to P10, the IPL increased significantly, consistent with retinal synaptogenesis. From P10 to P12, the IPL and ORL also increased, which is coherent with synaptic connectivity and photoreceptor maturation. In contrast, during these periods, the INL decreased significantly, consistent with cellular densification and selective apoptotic “pruning” of the tissue during nuclear migration. Thereafter from P12 to P21, the INL continued to thin (significantly from P17 to P21) whereas the other layers remained unchanged. No time-dependent changes were observed in the NFL. Overall, changes in the total retina were attributed to those in the IPL, INL, and ORL. Regions of the retina adjacent to the optic nerve head were thinner than distal regions during maturation. Conclusions Changes in retinal layer thickness are consistent with retinal developmental mechanisms. Accordingly, this report opens new horizons in using our system in the mouse to characterize longitudinally developmental digressions in models of human diseases.
Collapse
Affiliation(s)
- Simon Brais-Brunet
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada.,Research Center, CHU Sainte-Justine, Montréal, Canada
| | - Émilie Heckel
- Research Center, CHU Sainte-Justine, Montréal, Canada.,Department of Pharmacology, University of Montréal, Montréal, Canada
| | - Udayakumar Kanniyappan
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada.,Research Center, CHU Sainte-Justine, Montréal, Canada
| | - Sylvain Chemtob
- Research Center, CHU Sainte-Justine, Montréal, Canada.,Department of Pharmacology, University of Montréal, Montréal, Canada.,Department of Pediatrics, University of Montréal, Montréal, Canada.,Department of Ophthalmology, University of Montréal, Montréal, Canada
| | - Caroline Boudoux
- Research Center, CHU Sainte-Justine, Montréal, Canada.,Department of Engineering Physics, Polytechnique Montréal, Montréal, Canada
| | - Jean-Sébastien Joyal
- Research Center, CHU Sainte-Justine, Montréal, Canada.,Department of Pharmacology, University of Montréal, Montréal, Canada.,Department of Pediatrics, University of Montréal, Montréal, Canada.,Department of Ophthalmology, University of Montréal, Montréal, Canada
| | - Mathieu Dehaes
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada.,Research Center, CHU Sainte-Justine, Montréal, Canada.,Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montréal, Montréal, Canada
| |
Collapse
|
20
|
Zhou M, Liu Y, Ma C. Distinct Nuclear Architecture of Photoreceptors and Light-Induced Behaviors in Different Strains of Mice. Transl Vis Sci Technol 2021; 10:37. [PMID: 34003922 PMCID: PMC7910638 DOI: 10.1167/tvst.10.2.37] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The mouse retina is considered a remarkable model for studying gene functions. However, variations in genetic background influence phenotypes in the mammalian retina. Therefore this study aimed to investigate the effects of the genetic background on the nuclear architecture of photoreceptor cells and the light-induced behavior in C57BL/6, 129 × 1/svj, and ICR mice. Methods The nuclear architecture of photoreceptor cells was investigated using various staining methods on postnatal day 21 (P21). Murine behavior was observed using a light-dark compartment test. Results The outer nuclear layer and retina were significantly thicker in C57BL/6 mice than in 129 × 1/svj mice. The percentage of photoreceptors with one chromocenter was significantly higher in C57BL/6 mice than in 129 × 1/svj and ICR mice on P21. The numbers of photoreceptor cells in C57BL/6 and ICR mice were significantly higher than those in 129 × 1/svj mice. The behavior test revealed that the walking distance and velocity in the light compartment were increased in C57BL/6 and ICR mice compared to 129 × 1/svj mice. Conclusions Different mouse strains had a distinct nuclear architecture of photoreceptors on P21, and C57BL/6 and ICR mice were more active than 129 × 1/svj mice in response to light-induced stress. Translational Relevance This study demonstrates a technique for assessing retinal structures and nuclear architecture in various strains of mice, which are often used to model human retinal disease. Hence, this study may help to elucidate the effect of genetic or disease-induced variance in retinal architecture and the organization of photoreceptor nuclear content on visual function in humans.
Collapse
Affiliation(s)
- Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Yutong Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chao Ma
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
21
|
Tkatchenko TV, Tkatchenko AV. Genetic network regulating visual acuity makes limited contribution to visually guided eye emmetropization. Genomics 2021; 113:2780-2792. [PMID: 34147636 DOI: 10.1016/j.ygeno.2021.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
During postnatal development, the eye undergoes a refinement process whereby optical defocus guides eye growth towards sharp vision in a process of emmetropization. Optical defocus activates a signaling cascade originating in the retina and propagating across the back of the eye to the sclera. Several observations suggest that visual acuity might be important for optical defocus detection and processing in the retina; however, direct experimental evidence supporting or refuting the role of visual acuity in refractive eye development is lacking. Here, we used genome-wide transcriptomics to determine the relative contribution of the retinal genetic network regulating visual acuity to the signaling cascade underlying visually guided eye emmetropization. Our results provide evidence that visual acuity is regulated at the level of molecular signaling in the retina by an extensive genetic network. The genetic network regulating visual acuity makes relatively small contribution to the signaling cascade underlying refractive eye development. This genetic network primarily affects baseline refractive eye development and this influence is primarily facilitated by the biological processes related to melatonin signaling, nitric oxide signaling, phototransduction, synaptic transmission, and dopamine signaling. We also observed that the visual-acuity-related genes associated with the development of human myopia are chiefly involved in light perception and phototransduction. Our results suggest that the visual-acuity-related genetic network primarily contributes to the signaling underlying baseline refractive eye development, whereas its impact on visually guided eye emmetropization is modest.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
22
|
Tkatchenko TV, Tkatchenko AV. Genome-wide analysis of retinal transcriptome reveals common genetic network underlying perception of contrast and optical defocus detection. BMC Med Genomics 2021; 14:153. [PMID: 34107987 PMCID: PMC8190860 DOI: 10.1186/s12920-021-01005-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Refractive eye development is regulated by optical defocus in a process of emmetropization. Excessive exposure to negative optical defocus often leads to the development of myopia. However, it is still largely unknown how optical defocus is detected by the retina. METHODS Here, we used genome-wide RNA-sequencing to conduct analysis of the retinal gene expression network underlying contrast perception and refractive eye development. RESULTS We report that the genetic network subserving contrast perception plays an important role in optical defocus detection and emmetropization. Our results demonstrate an interaction between contrast perception, the retinal circadian clock pathway and the signaling pathway underlying optical defocus detection. We also observe that the relative majority of genes causing human myopia are involved in the processing of optical defocus. CONCLUSIONS Together, our results support the hypothesis that optical defocus is perceived by the retina using contrast as a proxy and provide new insights into molecular signaling underlying refractive eye development.
Collapse
Affiliation(s)
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
- Edward S. Harkness Eye Institute, Research Annex Room 415, 635 W. 165th Street, New York, NY 10032 USA
| |
Collapse
|
23
|
Li H, Yu H, Kim YK, Wang F, Teodoro G, Jiang Y, Nickerson JM, Kong J. Computational Model-Based Estimation of Mouse Eyeball Structure From Two-Dimensional Flatmount Microscopy Images. Transl Vis Sci Technol 2021; 10:25. [PMID: 34004004 PMCID: PMC8088229 DOI: 10.1167/tvst.10.4.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/17/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose Retinal pigment epithelial (RPE) cells serve as a supporter for the metabolism and visual function of photoreceptors and a barrier for photoreceptor protection. Morphology dynamics, spatial organization, distribution density, and growth patterns of RPE cells are important for further research on these RPE main functions. To enable such investigations within the authentic eyeball structure, a new method for estimating the three-dimensional (3D) eyeball sphere from two-dimensional tissue flatmount microscopy images was investigated. Methods An error-correction term was formulated to compensate for the reconstruction error as a result of tissue distortions. The effect of the tissue-distortion error was evaluated by excluding partial data points from the low- and high-latitude zones. The error-correction parameter was learned automatically using a set of samples with the ground truth eyeball diameters measured with noncontact light-emitting diode micrometry at submicron accuracy and precision. Results The analysis showed that the error-correction term in the reconstruction model is a valid method for modeling tissue distortions in the tissue flatmount preparation steps. With the error-correction model, the average relative error of the estimated eyeball diameter was reduced from 14% to 5%, and the absolute error was reduced from 0.22 to 0.03 mm. Conclusions A new method for enabling RPE morphometry analysis with respect to locations on an eyeball sphere was created, an important step in increasing RPE research and eye disease diagnosis. Translational Relevance This method enables one to derive RPE cell information from the 3D eyeball surface and helps characterize eyeball volume growth patterns under diseased conditions.
Collapse
Affiliation(s)
- Hongxiao Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hanyi Yu
- Department of Computer Science, Emory University, Atlanta, GA, USA
| | - Yong-Kyu Kim
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Fusheng Wang
- Department of Computer Science, Stony Brook University, Stony Brook, NY, USA
| | - George Teodoro
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| | | | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
- Department of Computer Science, Emory University, Atlanta, GA, USA
| |
Collapse
|
24
|
Kim YK, Yu H, Summers VR, Donaldson KJ, Ferdous S, Shelton D, Zhang N, Chrenek MA, Jiang Y, Grossniklaus HE, Boatright JH, Kong J, Nickerson JM. Morphometric Analysis of Retinal Pigment Epithelial Cells From C57BL/6J Mice During Aging. Invest Ophthalmol Vis Sci 2021; 62:32. [PMID: 33616620 PMCID: PMC7910641 DOI: 10.1167/iovs.62.2.32] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose To quantitatively evaluate the changes in orientation and morphometric features of mouse retinal pigment epithelial (RPE) cells in different regions of the eye during aging. Methods We segmented individual RPE cells from whole RPE flatmount images of C57BL/6J mice (postnatal days 30 to 720) using a machine-learning method and evaluated changes in morphometric features, including our newly developed metric combining alignment and shape of RPE cells during aging. Results Mainly, the anterior part of the RPE sheet grows during aging, while the posterior part remains constant. Changes in size and shape of the peripheral RPE cells are prominent with aging as cells become larger, elongated, and concave. Conversely, the central RPE cells maintain relatively constant size and numbers with aging. Cell count in the central area and the overall cell count (approximately 50,000) were relatively constant over different age groups. RPE cells also present a specific orientation concordance that matches the shape of the specific region of the eyeball. Those cells near the optic disc or equator have a circumferential orientation to cover the round shape of the eyeball, whereas those cells in the periphery have a radial orientation and corresponding radial elongation, the extent of which increases with aging and matches with axial elongation of the eyeball. Conclusions These results suggest that the fluid RPE morphology reflects various growth rates of underlying eyeball, and RPE cells could be classified into four regional classes (near the optic disc, central, equatorial, and peripheral) according to their morphometric features.
Collapse
Affiliation(s)
- Yong-Kyu Kim
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Ophthalmology, Hallym University College of Medicine, Kangdong Sacred Heart Hospital, Seoul, South Korea
| | - Hanyi Yu
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
| | - Vivian R. Summers
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Kevin J. Donaldson
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Salma Ferdous
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Debresha Shelton
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Nan Zhang
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States
| | - Hans E. Grossniklaus
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Jun Kong
- Department of Computer Science, Emory University, Atlanta, Georgia, United States
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States
- Department of Computer Science, Georgia State University, Atlanta, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
25
|
Three-dimensional data capture and analysis of intact eye lenses evidences emmetropia-associated changes in epithelial cell organization. Sci Rep 2020; 10:16898. [PMID: 33037268 PMCID: PMC7547080 DOI: 10.1038/s41598-020-73625-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/17/2020] [Indexed: 01/16/2023] Open
Abstract
Organ and tissue development are highly coordinated processes; lens growth and functional integration into the eye (emmetropia) is a robust example. An epithelial monolayer covers the anterior hemisphere of the lens, and its organization is the key to lens formation and its optical properties throughout all life stages. To better understand how the epithelium supports lens function, we have developed a novel whole tissue imaging system using conventional confocal light microscopy and a specialized analysis software to produce three-dimensional maps for the epithelium of intact mouse lenses. The open source software package geometrically determines the anterior pole position, the equatorial diameter, and three-dimensional coordinates for each detected cell in the epithelium. The user-friendly cell maps, which retain global lens geometry, allow us to document age-dependent changes in the C57/BL6J mouse lens cell distribution characteristics. We evidence changes in epithelial cell density and distribution in C57/BL6J mice during the establishment of emmetropia between postnatal weeks 4-6. These epithelial changes accompany a previously unknown spheroid to lentoid shape transition of the lens as detected by our analyses. When combined with key findings from previous mouse genetic and cell biological studies, we suggest a cytoskeleton-based mechanism likely underpins these observations.
Collapse
|
26
|
Fiedorowicz M, Wełniak-Kamińska M, Świątkiewicz M, Orzeł J, Chorągiewicz T, Toro MD, Rejdak R, Bogorodzki P, Grieb P. Changes of Ocular Dimensions as a Marker of Disease Progression in a Murine Model of Pigmentary Glaucoma. Front Pharmacol 2020; 11:573238. [PMID: 33013417 PMCID: PMC7500411 DOI: 10.3389/fphar.2020.573238] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose The elevation of intraocular pressure (IOP), a major risk factor in glaucoma, is an important parameter tracked in experimental models of this disease. However, IOP measurement in laboratory rodents is challenging and may not correlate with some key pathological events that occur in the development of glaucoma. The aims of this study were to quantify changes in ocular morphology in DBA/2J mice that develop spontaneous, age-dependent, pigmentary glaucoma and to check the possible correlation of these parameters with IOP. Method Eye morphology was evaluated with MRI in DBA/2J, DBA/2J-Gpnmb+/SjJ, and C57BL/6J female mice ages 3, 6, 9, 12, and 15 months. The animals were anesthetized with isoflurane. A planar receive-only surface coil (inner diameter = 10 mm) was placed over each animal’s left eye and the image was acquired with a 7T small animal-dedicated magnetic resonance tomograph and T2-weighted TurboRARE sequence. Ocular dimensions were manually quantitated using OsiriX software. IOP was measured with rebound tonometry. Results In the control animals, no age-related changes in the ocular morphology were noted. Since 6 months of age, the anterior chamber deepening and elongation of the eyeballs of DBA/2J mice was detectable. We found a significant, positive correlation between IOP and axial length, anterior chamber area, or anterior chamber width in C57BL/6J mice but not in DBA/2J mice. However, after excluding the measurements performed in the oldest DBA/2J mice (i.e. analyzing only the animals ages 3 to 12 months), we demonstrated a significant positive correlation between IOP and anterior chamber width. Conclusion High-resolution magnetic resonance imaging of the eye area in mice enables reproducible and consistent measures of key dimensions of the eyeball. We observed age-dependent alterations in the eye morphology of DBA/2J mice that mostly affected the anterior chamber. We also demonstrated a correlation between some of the ocular dimensions and the IOP of C57Bl/6J mice and DBA/2J mice with moderately advanced glaucomatous pathology.
Collapse
Affiliation(s)
- Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Świątkiewicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Orzeł
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Electronics and Information Technology, Warsaw University of Technology, Warsaw, Poland
| | - Tomasz Chorągiewicz
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Mario Damiano Toro
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland.,Faculty of Medical Sciences, Collegium Medicum, Cardinal Stefan Wyszyński University, Warsaw, Poland
| | - Robert Rejdak
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Piotr Bogorodzki
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Electronics and Information Technology, Warsaw University of Technology, Warsaw, Poland
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
27
|
Muir ER, Pan X, Donaldson PJ, Vaghefi E, Jiang Z, Sellitto C, White TW. Multi-parametric MRI of the physiology and optics of the in-vivo mouse lens. Magn Reson Imaging 2020; 70:145-154. [PMID: 32380160 DOI: 10.1016/j.mri.2020.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/30/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023]
Abstract
The optics of the ocular lens are determined by its geometry (shape and volume) and its inherent gradient of refractive index (water to protein ratio), which are in turn maintained by unique cellular physiology known as the lens internal microcirculation system. Previously, magnetic resonance imaging (MRI) has been used on ex vivo organ cultured bovine lenses to show that pharmacological perturbations to this microcirculation system disrupt ionic and fluid homeostasis and overall lens optics. In this study, we have optimised in vivo MRI protocols for use on wild-type and transgenic mouse models so that the effects of genetically perturbing the lens microcirculation system on lens properties can be studied. In vivo MRI protocols and post-analysis methods for studying the mouse lens were optimised and used to measure the lens geometry, diffusion, T1 and T2, as well as the refractive index (n) calculated from T2, in wild-type mice and the genetically modified Cx50KI46 mouse. In this animal line, gap junctional coupling in the lens is increased by knocking in the gap junction protein Cx46 into the Cx50 locus. Relative to wild-type mice, Cx50KI46 mice showed significantly reduced lens size and radius of curvature, increased T1 and T2 values, and decreased n in the lens nucleus, which was consistent with the developmental and functional changes characterised previously in this lens model. These proof of principle experiments show that in vivo MRI can be applied to transgenic mouse models to gain mechanistic insights into the relationship between lens physiology and optics, and in the future suggest that longitudinal studies can be performed to determine how this relationship is altered by age in mouse models of cataract.
Collapse
Affiliation(s)
- Eric R Muir
- Department of Radiology, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Xingzheng Pan
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Paul J Donaldson
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand; Department of Physiology, School of Medical Sciences, University of Auckland, New Zealand
| | - Ehsan Vaghefi
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Zhao Jiang
- Department of Radiology, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Caterina Sellitto
- Department of Physiology & Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Thomas W White
- Department of Physiology & Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
28
|
Paulson B, Lee S, Jue M, Lee K, Lee S, Kim GB, Moon Y, Lee JY, Kim N, Kim JK. Stereotaxic endoscopy for the ocular imaging of awake, freely moving animal models. JOURNAL OF BIOPHOTONICS 2020; 13:e201960188. [PMID: 32017450 DOI: 10.1002/jbio.201960188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/07/2020] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
Stereotaxic instruments are increasingly used in research animals for the study of disease, but typically require restraints and anesthetic procedures. A stereotaxic head mount that enables imaging of the anterior chamber of the eye in alert and freely mobile mice is presented in this study. The head mount is fitted based on computed tomography scans and manufactured using 3D printing. The system is placed noninvasively using temporal mount bars and a snout mount, without breaking the skin or risking suffocation, while an instrument channel stabilizes the ocular probes. With a flexible micro-endoscopic probe and a confocal scanning laser microscopy system, <20 μm resolution is achieved in vivo with a field of view of nearly 1 mm. Discomfort is minimal, and further adaptations for minimally invasive neuroscience, optogenetics and auditory studies are possible.
Collapse
Affiliation(s)
- Bjorn Paulson
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Sangwook Lee
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Miyeon Jue
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Kyungsung Lee
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | - Sanghwa Lee
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
| | | | - Youngjin Moon
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Joo Yong Lee
- Department of Ophthalmology, College of Medicine, University of Ulsan, Asan Medical Center, Seoul, South Korea
| | - Namkug Kim
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Jun Ki Kim
- Biomedical Engineering Research Center, Asan Institute for Life Science, Asan Medical Center, Seoul, South Korea
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, South Korea
| |
Collapse
|
29
|
Abstract
PURPOSE To analyze the hallmark features of pathologic myopia developed in animal models and compare them with those seen in patients. METHODS A literature review was performed to identify animal models that exhibited key features of pathologic myopia, namely posterior staphyloma, myopic maculopathy, lacquer cracks, and choroidal neovascularization, either spontaneously or induced by monocular deprivation. Using imaging modalities, such as optical coherence tomography, confocal scanning laser ophthalmoscopy, fluorescein angiography, and electron microscopy, these features were compared with those found in myopic maculopathy of patients. RESULTS Three types of animals were identified. The LRP2 knockout mice exhibited posterior staphylomas and chorioretinal atrophy at 21 and 60 days after birth, respectively. Retinopathy globe enlarged (rge) chicks and normal lid-sutured chicks developed lacquer cracks and chorioretinal atrophy. Lacquer cracks detected in rge chicks subsequently progressed to patchy chorioretinal atrophy, which is also commonly seen in patients with pathologic myopia. CONCLUSION The LRP2 knockout mice, retinopathy globe enlarged (rge) chicks, and normal lid-sutured chicks exhibit features typical for myopic maculopathy in patients and could serve to further elucidate the pathogenesis of myopic maculopathy.
Collapse
|
30
|
Liu AS, Brown DM, Conn RE, McNabb RP, Pardue MT, Kuo AN. Topography and pachymetry maps for mouse corneas using optical coherence tomography. Exp Eye Res 2020; 190:107868. [PMID: 31704241 PMCID: PMC6961820 DOI: 10.1016/j.exer.2019.107868] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/04/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022]
Abstract
The majority of the eye's refractive power lies in the cornea, and pathological changes in its shape can affect vision. Small animal models offer an unparalleled degree of control over genetic and environmental factors that can help elucidate mechanisms of diseases affecting corneal shape. However, there is not currently a method to characterize the corneal shape of small animal eyes with topography or pachymetry maps, as is done clinically for humans. We bridge this gap by demonstrating methods using optical coherence tomography (OCT) to generate the first topography and pachymetry (thickness) maps of mouse corneas. Radii of curvature acquired using OCT were validated using calibration spheres as well as in vivo mouse corneas with a mouse keratometer. The resulting topography and pachymetry maps are analogous to those used diagnostically in clinic and potentially allow for characterization of genetically modified mice that replicate key features of human corneal disease.
Collapse
Affiliation(s)
- Alice S Liu
- Ophthalmology, Duke University, Durham, NC, USA.
| | - Dillon M Brown
- Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | | | | | - Machelle T Pardue
- Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA; Neuroscience, Emory University, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA
| | - Anthony N Kuo
- Ophthalmology, Duke University, Durham, NC, USA; Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
31
|
Arakawa H. Sensorimotor developmental factors influencing the performance of laboratory rodents on learning and memory. Behav Brain Res 2019; 375:112140. [PMID: 31401145 PMCID: PMC6741784 DOI: 10.1016/j.bbr.2019.112140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Behavioral studies in animal models have advanced our knowledge of brain function and the neural mechanisms of human diseases. Commonly used laboratory rodents, such as mice and rats, provide a useful tool for studying the behaviors and mechanisms associated with learning and memory processes which are cooperatively regulated by multiple underlying factors, including sensory and motor performance and emotional/defense innate components. Each of these factors shows unique ontogeny and governs the sustainment of behavioral performance in learning tasks, and thus, understanding the integrative processes of behavioral development are crucial in the accurate interpretation of the functional meaning of learning and memory behaviors expressed in commonly employed behavioral test paradigms. In this review, we will summarize the major findings in the developmental processes of rodent behavior on the basis of the emergence of fundamental components for sustaining learning and memory behaviors. Briefly, most sensory modalities (except for vision) and motor abilities are functional at the juvenile stage, in which several defensive components, including active and passive defensive strategies and risk assessment behavior, emerge. Sex differences are detectable from the juvenile stage through adulthood and are considerable factors that influence behavioral tests. The test paradigms addressed in this review include associative learning (with an emphasis on fear conditioning), spatial learning, and recognition. This basic background information will aid in accurately performing behavioral studies in laboratory rodents and will therefore contribute to reducing inappropriate interpretations of behavioral data and further advance research on learning and memory in rodent models.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St. HSF2/S251, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Subramanian K, Weigert M, Borsch O, Petzold H, Garcia-Ulloa A, Myers EW, Ader M, Solovei I, Kreysing M. Rod nuclear architecture determines contrast transmission of the retina and behavioral sensitivity in mice. eLife 2019; 8:49542. [PMID: 31825309 PMCID: PMC6974353 DOI: 10.7554/elife.49542] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/11/2019] [Indexed: 01/06/2023] Open
Abstract
Rod photoreceptors of nocturnal mammals display a striking inversion of nuclear architecture, which has been proposed as an evolutionary adaptation to dark environments. However, the nature of visual benefits and the underlying mechanisms remains unclear. It is widely assumed that improvements in nocturnal vision would depend on maximization of photon capture at the expense of image detail. Here, we show that retinal optical quality improves 2-fold during terminal development, and that this enhancement is caused by nuclear inversion. We further demonstrate that improved retinal contrast transmission, rather than photon-budget or resolution, enhances scotopic contrast sensitivity by 18–27%, and improves motion detection capabilities up to 10-fold in dim environments. Our findings therefore add functional significance to a prominent exception of nuclear organization and establish retinal contrast transmission as a decisive determinant of mammalian visual perception.
Collapse
Affiliation(s)
- Kaushikaram Subramanian
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany.,Cluster of Excellence, Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Martin Weigert
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany.,Cluster of Excellence, Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Oliver Borsch
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Heike Petzold
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany.,Cluster of Excellence, Physics of Life, Technische Universität Dresden, Dresden, Germany.,Department of Computer Science, Technische Universität Dresden, Dresden, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Irina Solovei
- Biozentrum, Ludwig Maximilians Universität, München, Germany
| | - Moritz Kreysing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany.,Cluster of Excellence, Physics of Life, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
33
|
Kim TH, Son T, Le D, Yao X. Longitudinal OCT and OCTA monitoring reveals accelerated regression of hyaloid vessels in retinal degeneration 10 (rd10) mice. Sci Rep 2019; 9:16685. [PMID: 31723168 PMCID: PMC6853881 DOI: 10.1038/s41598-019-53082-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/27/2019] [Indexed: 12/12/2022] Open
Abstract
The hyaloid vascular system (HVS) is known to have an important role in eye development. However, physiological mechanisms of HVS regression and their correlation with developmental eye disorders remain unclear due to technical limitations of conventional ending point examination with fixed tissues. Here, we report comparative optical coherence tomography (OCT) and OCT angiography (OCTA) monitoring of HVS regression in wild-type and retinal degeneration 10 (rd10) mice. Longitudinal OCTA monitoring revealed accelerated regression of hyaloid vessels correlated with retinal degeneration in rd10. Quantitative OCT measurement disclosed significant distortions of both retinal thickness and the vitreous chamber in rd10 compared to WT mice. These OCT/OCTA observations confirmed the close relationship between HVS physiology and retinal neurovascular development. The distorted HVS regression might result from retinal hyperoxia or dopamine abnormality due to retinal remodeling in rd10 retina. By providing a noninvasive imaging platform for longitudinal monitoring of HVS regression, further OCT/OCTA study may lead to in-depth understanding of the physiological mechanisms of HVS regression in normal and diseased eyes, which is not only important for advanced study of the nature of the visual system but also may provide insights into the development of better treatment protocols of congenital eye disorders.
Collapse
Affiliation(s)
- Tae-Hoon Kim
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Taeyoon Son
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - David Le
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xincheng Yao
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
34
|
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
|
35
|
Tkatchenko TV, Shah RL, Nagasaki T, Tkatchenko AV. Analysis of genetic networks regulating refractive eye development in collaborative cross progenitor strain mice reveals new genes and pathways underlying human myopia. BMC Med Genomics 2019; 12:113. [PMID: 31362747 PMCID: PMC6668126 DOI: 10.1186/s12920-019-0560-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Population studies suggest that genetic factors play an important role in refractive error development; however, the precise role of genetic background and the composition of the signaling pathways underlying refractive eye development remain poorly understood. METHODS Here, we analyzed normal refractive development and susceptibility to form-deprivation myopia in the eight progenitor mouse strains of the Collaborative Cross (CC). We used RNA-seq to analyze gene expression in the retinae of these mice and reconstruct genetic networks and signaling pathways underlying refractive eye development. We also utilized genome-wide gene-based association analysis to identify mouse genes and pathways associated with myopia in humans. RESULTS Genetic background strongly influenced both baseline refractive development and susceptibility to environmentally-induced myopia. Baseline refractive errors ranged from - 21.2 diopters (D) in 129S1/svlmj mice to + 22.0 D in CAST/EiJ mice and represented a continuous distribution typical of a quantitative genetic trait. The extent of induced form-deprivation myopia ranged from - 5.6 D in NZO/HILtJ mice to - 20.0 D in CAST/EiJ mice and also followed a continuous distribution. Whole-genome (RNA-seq) gene expression profiling in retinae from CC progenitor strains identified genes whose expression level correlated with either baseline refractive error or susceptibility to myopia. Expression levels of 2,302 genes correlated with the baseline refractive state of the eye, whereas 1,917 genes correlated with susceptibility to induced myopia. Genome-wide gene-based association analysis in the CREAM and UK Biobank human cohorts revealed that 985 of the above genes were associated with myopia in humans, including 847 genes which were implicated in the development of human myopia for the first time. Although the gene sets controlling baseline refractive development and those regulating susceptibility to myopia overlapped, these two processes appeared to be controlled by largely distinct sets of genes. CONCLUSIONS Comparison with data for other animal models of myopia revealed that the genes identified in this study comprise a well-defined set of retinal signaling pathways, which are highly conserved across different vertebrate species. These results identify major signaling pathways involved in refractive eye development and provide attractive targets for the development of anti-myopia drugs.
Collapse
Affiliation(s)
| | - Rupal L. Shah
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK
| | | | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
| |
Collapse
|
36
|
Locri F, Dal Monte M, Aronsson M, Cammalleri M, De Rosa M, Pavone V, Kvanta A, Bagnoli P, André H. UPARANT is an effective antiangiogenic agent in a mouse model of rubeosis iridis. J Mol Med (Berl) 2019; 97:1273-1283. [PMID: 31243519 PMCID: PMC6713680 DOI: 10.1007/s00109-019-01794-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/08/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023]
Abstract
Abstract Puncture-induced iris neovascularization (rubeosis iridis; RI) in mice is associated with upregulation of extracellular matrix (ECM) degradation and inflammatory factors. The anti-angiogenic and anti-inflammatory efficacy of UPARANT in reducing RI was determined by noninvasive, in vivo iris vascular densitometry, and confirmed in vitro by quantitative vascular-specific immunostaining. Intravitreal administration of UPARANT successfully and rapidly reduced RI to non-induced control levels. Molecular analysis revealed that UPARANT inhibits formyl peptide receptors through a predominantly anti-inflammatory response, accompanied with a significant reduction in ECM degradation and inflammation markers. Similar results were observed with UPARANT administered systemically by subcutaneous injection. These data suggest that the tetrapeptide UPARANT is an effective anti-angiogenic agent for the treatment of RI, both by local and systemic administrations. The effectiveness of UPARANT in reducing RI in a model independent of the canonical vascular endothelial growth factor (VEGF) proposes an alternative for patients that do not respond to anti-VEGF treatments, which could improve treatment in proliferative ocular diseases. Key messages UPARANT is effective in the treatment of rubeosis iridis, both by local and systemic administrations. UPARANT can reduce VEGF-independent neovascularization.
Electronic supplementary material The online version of this article (10.1007/s00109-019-01794-w) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Filippo Locri
- Department of Clinical Neuroscience, Division of Eye and Vision, St Erik Eye Hospital, Karolinska Institutet, Polhemsgatan 50, 112 82, Stockholm, Sweden.,Department of Biology, University of Pisa, Pisa, Italy
| | | | - Monica Aronsson
- Department of Clinical Neuroscience, Division of Eye and Vision, St Erik Eye Hospital, Karolinska Institutet, Polhemsgatan 50, 112 82, Stockholm, Sweden
| | | | - Mario De Rosa
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Vincenzo Pavone
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Anders Kvanta
- Department of Clinical Neuroscience, Division of Eye and Vision, St Erik Eye Hospital, Karolinska Institutet, Polhemsgatan 50, 112 82, Stockholm, Sweden
| | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St Erik Eye Hospital, Karolinska Institutet, Polhemsgatan 50, 112 82, Stockholm, Sweden.
| |
Collapse
|
37
|
Bell BA, Bonilha VL, Hagstrom SA, Anand-Apte B, Hollyfield JG, Samuels IS. Prolonged ocular exposure leads to retinal lesions in mice. Exp Eye Res 2019; 185:107672. [PMID: 31128100 DOI: 10.1016/j.exer.2019.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/06/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023]
Abstract
Retinal lesions in the posterior pole of laboratory mice occur due to native, developmental abnormalities or as a consequence of environmental or experimental conditions. In this study, we investigated the rate and extent of retinal lesions as a result of prolonged ocular exposure following general anesthesia. Following experimental preparation induction procedures (EPIP) involving general anesthesia, mydriasis/cycloplegia, and topical anesthesia to the cornea, two ocular recovery conditions (protected and unprotected) were tested within two different animal recovery chambers (open or closed). The anterior and posterior poles were evaluated for the development of retinal lesions using digital color photography, scanning laser ophthalmoscopy, and spectral-domain optical coherence during anesthesia recovery and up to 2.5 months thereafter. In some mice, electroretinograms, histological and immunohistological evaluations were performed to assess functional and structural changes that accompanied the retinal lesions detected by in vivo imaging. Our data suggests that prolonged ocular surface exposure to circulating ambient room air leads to significant anterior and posterior segment ocular complications. The most abundant, semi-reversible complication observed was the development of lesions in the outer retina, which had a 90% probability of occurring after 45 min of exposure. The lesions mostly resolved short-term, but functional and imaging evidence suggest that some perturbations to the outer retina may persist one or more months following initial development.
Collapse
Affiliation(s)
- Brent A Bell
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States.
| | - Vera L Bonilha
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Stephanie A Hagstrom
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Bela Anand-Apte
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Joe G Hollyfield
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Ivy S Samuels
- Cole Eye Institute/Ophthalmic Research, Cleveland Clinic, Cleveland, OH, United States; Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
38
|
Troilo D, Smith EL, Nickla DL, Ashby R, Tkatchenko AV, Ostrin LA, Gawne TJ, Pardue MT, Summers JA, Kee CS, Schroedl F, Wahl S, Jones L. IMI - Report on Experimental Models of Emmetropization and Myopia. Invest Ophthalmol Vis Sci 2019; 60:M31-M88. [PMID: 30817827 PMCID: PMC6738517 DOI: 10.1167/iovs.18-25967] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 10/20/2018] [Indexed: 11/24/2022] Open
Abstract
The results of many studies in a variety of species have significantly advanced our understanding of the role of visual experience and the mechanisms of postnatal eye growth, and the development of myopia. This paper surveys and reviews the major contributions that experimental studies using animal models have made to our thinking about emmetropization and development of myopia. These studies established important concepts informing our knowledge of the visual regulation of eye growth and refractive development and have transformed treatment strategies for myopia. Several major findings have come from studies of experimental animal models. These include the eye's ability to detect the sign of retinal defocus and undergo compensatory growth, the local retinal control of eye growth, regulatory changes in choroidal thickness, and the identification of components in the biochemistry of eye growth leading to the characterization of signal cascades regulating eye growth and refractive state. Several of these findings provided the proofs of concepts that form the scientific basis of new and effective clinical treatments for controlling myopia progression in humans. Experimental animal models continue to provide new insights into the cellular and molecular mechanisms of eye growth control, including the identification of potential new targets for drug development and future treatments needed to stem the increasing prevalence of myopia and the vision-threatening conditions associated with this disease.
Collapse
Affiliation(s)
- David Troilo
- SUNY College of Optometry, State University of New York, New York, New York, United States
| | - Earl L. Smith
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Debora L. Nickla
- Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States
| | - Regan Ashby
- Health Research Institute, University of Canberra, Canberra, Australia
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | - Lisa A. Ostrin
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Timothy J. Gawne
- School of Optometry, University of Alabama Birmingham, Birmingham, Alabama, United States
| | - Machelle T. Pardue
- Biomedical Engineering, Georgia Tech College of Engineering, Atlanta, Georgia, United States31
| | - Jody A. Summers
- College of Medicine, University of Oklahoma, Oklahoma City, Oklahoma, United States
| | - Chea-su Kee
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Falk Schroedl
- Departments of Ophthalmology and Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Siegfried Wahl
- Institute for Ophthalmic Research, University of Tuebingen, Zeiss Vision Science Laboratory, Tuebingen, Germany
| | - Lyndon Jones
- CORE, School of Optometry and Vision Science, University of Waterloo, Ontario, Canada
| |
Collapse
|
39
|
Zhang X, Wang D, Dongye M, Zhu Y, Chen C, Wang R, Long E, Liu Z, Wu X, Lin D, Chen J, Lin Z, Wang J, Li W, Li Y, Li D, Lin H. Loss-of-function mutations in FREM2 disrupt eye morphogenesis. Exp Eye Res 2019; 181:302-312. [PMID: 30802441 DOI: 10.1016/j.exer.2019.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/30/2019] [Accepted: 02/17/2019] [Indexed: 10/27/2022]
Abstract
Cryptophthalmos is a rare congenital disorder characterized by ocular dysplasia with eyelid malformation. Complete cryptophthalmos is characterized by the presence of continuous skin from the forehead over the eyes and onto the cheek, along with complete fusion of the eyelids. In the present study, we characterized the clinical manifestations of three patients with isolated bilateral cryptophthalmos. These patients shared the same c.6499C > T missense mutation in the FRAS1-related extracellular matrix protein 2 (FREM2) gene, while each individual presented an additional nonsense mutation in the same gene (Patient #1, c.2206C > T; Patient #2, c.5309G > A; and Patient #3, c.4063C > T). Then, we used CRISPR/Cas9 to generate mice carrying Frem2R725X/R2156W compound heterozygous mutations, and showed that these mice recapitulated the human isolated cryptophthalmos phenotype. We detected FREM2 expression in the outer plexiform layer of the retina for the first time in the cryptophthalmic eyes, and the levels were comparable to the wild-type mice. Moreover, a set of different expressed genes that may contribute secondarily to the phenotypes were identified by performing RNA sequencing (RNA-seq) of the fetal Frem2 mutant mice. Our findings extend the spectrum of FREM2 mutations, and provide insights into opportunities for the prenatal diagnosis of isolated cryptophthalmos. Furthermore, our work highlights the importance of the FREM2 protein during the development of eyelids and the anterior segment of the eyeballs, establishes a suitable animal model for studying epithelial reopening during eyelid development and serves as a valuable reference for further mechanistic studies of the pathogenesis of isolated cryptophthalmos.
Collapse
Affiliation(s)
- Xiayin Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Dongni Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Meimei Dongye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yi Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Chuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ruixin Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Erping Long
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhenzhen Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiaohang Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Duoru Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jingjing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhuoling Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jinghui Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Wangting Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yang Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Science, Beijing, 100730, China
| | - Dongmei Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Science, Beijing, 100730, China
| | - Haotian Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
40
|
Crosbie DE, Keaney J, Tam LCS, Daniel Stamer W, Campbell M, Humphries P. Age-related changes in eye morphology and aqueous humor dynamics in DBA/2J mice using contrast-enhanced ocular MRI. Magn Reson Imaging 2019; 59:10-16. [PMID: 30660703 DOI: 10.1016/j.mri.2019.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE Here, we are testing the hypothesis that dynamic contrast enhanced MRI (DCE-MRI) is a useful approach for non-invasively evaluating age-related changes in aqueous humor outflow and its contribution to elevated intraocular pressure in the DBA/2J model of pigmentary glaucoma. METHODS A rodent-specific 7 T MRI was used to assess eye anatomy (anterior chamber (AC) and vitreous body (VB) morphology, eye size, lens size) and aqueous humor dynamics (via intravenous administration of Gd-DTPA and Gd-BOPTA contrast agents) in C57BL/6 and DBA/2J mice at 3 and 9 months of age. RESULTS Gd-MRI was used to demonstrate an anterior solute pathway into the mouse AC. Topical latanoprost treatment in C57BL/6J mice reduced Gd-BOPTA accumulation in the AC. Age-related increases in AC area, AC depth and eye size were observed in DBA/2J mice compared to C57BL/6J mice. The rate of Gd-DTPA accumulation and peak Gd-DTPA intensity was lowest in 9-month old DBA/2J mice compared to 3-month old DBA/2J mice and C57BL/6J mice at both ages. Leakage of Gd-DTPA posteriorly into the VB was also observed in 9-month old DBA/2J mice. CONCLUSIONS These studies support the idea that age-related changes in aqueous humor outflow contribute to elevated intraocular pressure (IOP) in the DBA/2J model of pigmentary glaucoma. Gd-MRI is a valuable tool for better understanding of mechanisms and dynamics of aqueous humor circulation in normal and glaucomatous mouse eyes or following topical administration of medicines to reduce IOP.
Collapse
Affiliation(s)
- Darragh E Crosbie
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - James Keaney
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland; Ross University School of Veterinary Medicine, P. O. Box 334, Basseterre, St. Kitts, Saint Kitts and Nevis
| | - Lawrence C S Tam
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland
| | - W Daniel Stamer
- Departments of Ophthalmology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Matthew Campbell
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland.
| | - Peter Humphries
- Ocular Genetics, Unit, Smurfit Institute of Genetics, Trinity College Dublin, Lincoln Place Gate, Dublin 2, Ireland.
| |
Collapse
|
41
|
Wecker T, van Oterendorp C, Reichardt W. Quantitative evaluation of an optimized Time of Flight Magnetic Resonance Imaging procedure using a phantom setup to simulate aqueous humor flow. Data Brief 2018; 18:585-589. [PMID: 29896531 PMCID: PMC5996222 DOI: 10.1016/j.dib.2018.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 11/10/2022] Open
Abstract
Preclinical Magnetic Resonance Imaging at high field strength offers the great advantage of combining anatomical information and very high resolution of down to 25 µm in mice and even higher resolutions in ex vivo settings. The presented data is Time of Flight MR imaging data using a tube phantom and a given flow-rate to determine the lower limit of the flow rate that is detectable with an experimental set-up and a specifically optimized 2D TOF sequence. In this work we present data on a phantom study which shows the ability of Time of Flight MR Imaging to detect very low flow rates down to 25 µl/h at a velocity of 0.1 mm/s non-invasively in a phantom study.
Collapse
Affiliation(s)
- Thomas Wecker
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.,Wecker Eye Center, Heilbronn, Germany
| | | | - Wilfried Reichardt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
42
|
Paylakhi S, Labelle-Dumais C, Tolman NG, Sellarole MA, Seymens Y, Saunders J, Lakosha H, deVries WN, Orr AC, Topilko P, John SWM, Nair KS. Müller glia-derived PRSS56 is required to sustain ocular axial growth and prevent refractive error. PLoS Genet 2018. [PMID: 29529029 PMCID: PMC5864079 DOI: 10.1371/journal.pgen.1007244] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A mismatch between optical power and ocular axial length results in refractive errors. Uncorrected refractive errors constitute the most common cause of vision loss and second leading cause of blindness worldwide. Although the retina is known to play a critical role in regulating ocular growth and refractive development, the precise factors and mechanisms involved are poorly defined. We have previously identified a role for the secreted serine protease PRSS56 in ocular size determination and PRSS56 variants have been implicated in the etiology of both hyperopia and myopia, highlighting its importance in refractive development. Here, we use a combination of genetic mouse models to demonstrate that Prss56 mutations leading to reduced ocular size and hyperopia act via a loss of function mechanism. Using a conditional gene targeting strategy, we show that PRSS56 derived from Müller glia contributes to ocular growth, implicating a new retinal cell type in ocular size determination. Importantly, we demonstrate that persistent activity of PRSS56 is required during distinct developmental stages spanning the pre- and post-eye opening periods to ensure optimal ocular growth. Thus, our mouse data provide evidence for the existence of a molecule contributing to both the prenatal and postnatal stages of human ocular growth. Finally, we demonstrate that genetic inactivation of Prss56 rescues axial elongation in a mouse model of myopia caused by a null mutation in Egr1. Overall, our findings identify PRSS56 as a potential therapeutic target for modulating ocular growth aimed at preventing or slowing down myopia, which is reaching epidemic proportions.
Collapse
Affiliation(s)
- Seyyedhassan Paylakhi
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Nicholas G Tolman
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Michael A. Sellarole
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Yusef Seymens
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
| | - Joseph Saunders
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Hesham Lakosha
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Wilhelmine N. deVries
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | - Andrew C. Orr
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Piotr Topilko
- Ecole Normale Supérieure, Institut de Biologie de l’ENS (IBENS), and Inserm U1024, and CNRS UMR 8197, Paris, France
| | - Simon WM. John
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, United States of America
- Department of Ophthalmology, Tufts University School of Medicine Boston, MA, United States of America
| | - K. Saidas Nair
- Department of Ophthalmology, University of California, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
43
|
Trejo-Reveles V, McTeir L, Summers K, Rainger J. An analysis of anterior segment development in the chicken eye. Mech Dev 2018. [PMID: 29526791 DOI: 10.1016/j.mod.2018.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Precise anterior segment (AS) development in the vertebrate eye is essential for maintaining ocular health throughout life. Disruptions to genetic programs can lead to severe structural AS disorders at birth, while more subtle AS defects may disrupt the drainage of ocular fluids and cause dysregulation of intraocular pressure homeostasis, leading to progressive vision loss. To date, the mouse has served as the major model to study AS development and pathogenesis. Here we present an accurate histological atlas of chick AS formation throughout eye development, with a focus on the formation of drainage structures. We performed expression analyses for a panel of known AS disorder genes, and showed that chick PAX6 was localized to cells of neural retina and surface ectoderm derived structures, displaying remarkable similarity to the mouse. We provide a comparison to mouse and humans for chick AS developmental sequences and structures and confirm that AS development shares common features in all three species, although the main AS structures in the chick are developed prior to hatching. These features enable the unique experimental advantages inherent to chick embryos, and we therefore propose the chick as an appropriate additional model for AS development and disease.
Collapse
Affiliation(s)
- Violeta Trejo-Reveles
- The Roslin Institute Chicken Embryology (RICE) group, The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Lynn McTeir
- The Roslin Institute Chicken Embryology (RICE) group, The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Kim Summers
- Mater Research Institute-UQ, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia.
| | - Joe Rainger
- The Roslin Institute Chicken Embryology (RICE) group, The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| |
Collapse
|
44
|
Abstract
Despite the global pandemic of myopia, the precise molecular mechanism of the onset of myopia remains largely unknown. This is partially because of the lack of efficient murine myopic models that allow genetic manipulation at low cost. Here we report a highly practical and reproducible lens-induced myopia model by specially designed frames and lenses for mice. A lens power dependent myopic induction in mice was shown until minus 30 diopter lenses. The phenotype was significantly stronger than form-deprivation myopia. We presented the protocol for precise evaluations of the state of myopia, including refraction, corneal curvature and axial length using up-to-date devices. We also found that myopic mouse eyes showed decreased visual acuity on optokinetic response examination. Finally, we confirmed the anti-myopic effect of 1% atropine using this model, which showed its potential in drug screening. The strong phenotype, stable evaluation and the potential for gene manipulation utilizing the presented method in mice will accelerate the translational research of myopia.
Collapse
|
45
|
Wecker T, van Oterendorp C, Reichardt W. Functional assessment of the aqueous humour distal outflow pathways in bovine eyes using time-of-flight magnetic resonance tomography. Exp Eye Res 2017; 166:168-173. [PMID: 29074388 DOI: 10.1016/j.exer.2017.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 11/29/2022]
Abstract
The major part of the aqueous humor leaves the eye through the "conventional outflow pathway", consisting of the trabecular meshwork, Schlemm's canal, collector channels, an intrasceral plexus and the episcleral veins. While the trabecular meshwork is well characterized, little is known about anatomical and functional features of the peripheral outflow tract beyond Schlemm's canal. The emergence of minimally-invasive glaucoma surgery directly targeting the outflow resistance in the trabecular meshwork has elicited growing interest in these structures. We used time-of-flight magnetic resonance imaging in ex vivo bovine eyes to map fluid flow under physiological conditions. We were able to identify the peripheral outflow vessels solely by the signal detected from the fluid flow inside their lumina. A question of clinical relevance is, whether localized opening of the trabecular meshwork leads to only localized or to a 360° increase in intrascleral flow. To address this, a goniotomy ab interno was performed in 3 eyes and the flow signal intensity was quantified sectorially. A significant increase in fluid flow was observed in the sector distal to the goniotomy (p = 0.0005) but not in the other sectors (p = 0.1). As a proof of concept we demonstrated that TOF-MRI based detection of flow in the peripheral aqueous outflow tract is feasible. The functional link observed between trabecular meshwork sectors and their distal outflow tract sectors may be relevant for minimally-invasive glaucoma surgery in humans.
Collapse
Affiliation(s)
- Thomas Wecker
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Wecker Eye Center, Heilbronn, Germany.
| | | | - Wilfried Reichardt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Germany; German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
46
|
Li L, Yuan Y, Chen L, Li M, Ji P, Gong J, Zhao Y, Zhang H. Gadolinium-enhanced 7.0 T magnetic resonance imaging assessment of the aqueous inflow in rat eyes in vivo. Exp Eye Res 2017; 162:18-26. [DOI: 10.1016/j.exer.2017.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 01/09/2023]
|
47
|
The effect of topical administration of cyclopentolate on ocular biometry: An analysis for mouse and human models. Sci Rep 2017; 7:9952. [PMID: 28855546 PMCID: PMC5577254 DOI: 10.1038/s41598-017-09924-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/01/2017] [Indexed: 11/14/2022] Open
Abstract
Mydriasis with muscarinic antagonists have been used routinely prior to retinal examination and sometimes prior to refractive measurements of the mouse eye. However, biometric changes during topical administration of muscarinic antagonists have not been fully investigated in mice and humans. We found that the mouse eyes treated with cyclopentolate developed a hyperopia with a reduction in both the vitreous chamber depth and axial length. In humans, prior to the cyclopentolate treatment, a 6D accommodative stimulus produced a myopic shift with a reduced anterior chamber depth, choroidal thickness and anterior lens radius of curvature and an increase in lens thickness. After the cyclopentolate treatment, human eyes developed a hyperopic shift with an increased anterior chamber depth and anterior lens radius of curvature and a reduced lens thickness. Therefore, the biometric changes associated with this hyperopic shift were mainly located in the posterior segment of the eye in mice. However, it is the anterior segment of the eye that plays a main role in the hyperopic shift in human subjects. These results further indicate that mouse eyes do not have accommodation which needs to be taken into account when they are used for the study of human refractive errors.
Collapse
|
48
|
Magnetic resonance imaging study of eye congenital birth defects in mouse model. Mol Vis 2017; 23:572-578. [PMID: 28848319 PMCID: PMC5561137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/08/2017] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Embryonic eyelid closure is a well-documented morphogenetic episode in mammalian eye development. Detection of eyelid closure defect in humans is a major challenge because eyelid closure and reopen occur entirely in utero. As a consequence, congenital eye defects that are associated with failure of embryonic eyelid closure remain unknown. To fill the gap, we developed a mouse model of defective eyelid closure. This preliminary work demonstrates that the magnetic resonance imaging (MRI) approach can be used for the detection of extraocular muscle abnormalities in the mouse model. METHODS Mice with either normal (Map3k1+/- ) or defective (Map3k1-/- ) embryonic eyelid closure were used in this study. Images of the extraocular muscles were obtained with a 9.4 T high resolution microimaging MRI system. The extraocular muscles were identified, segmented, and measured in each imaging slice using an in-house program. RESULTS In agreement with histological findings, the imaging data show that mice with defective embryonic eyelid closure develop less extraocular muscle than normal mice. In addition, the size of the eyeballs was noticeably reduced in mice with defective embryonic eyelid closure. CONCLUSIONS We demonstrated that MRI can potentially be used for the study of extraocular muscle in the mouse model of the eye open-at-birth defect, despite the lack of specificity of muscle group provided by the current imaging resolution.
Collapse
|
49
|
Beaujean O, Locri F, Aronsson M, Kvanta A, André H. A novel in vivo model of puncture-induced iris neovascularization. PLoS One 2017; 12:e0180235. [PMID: 28658313 PMCID: PMC5489193 DOI: 10.1371/journal.pone.0180235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/12/2017] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To assess iris neovascularization by uveal puncture of the mouse eye and determine the role of angiogenic factors during iris neovascularization. METHODS Uveal punctures were performed on BalbC mouse eyes to induce iris angiogenesis. VEGF-blockage was used as an anti-angiogenic treatment, while normoxia- and hypoxia-conditioned media from retinal pigment epithelium (RPE) cells was used as an angiogenic-inducer in this model. Iris vasculature was determined in vivo by noninvasive methods. Iris blood vessels were stained for platelet endothelial cell adhesion molecule-1 and vascular sprouts were counted as markers of angiogenesis. Expression of angiogenic and inflammatory factors in the puncture-induced model were determined by qPCR and western blot. RESULTS Punctures led to increased neovascularization and sprouting of the iris. qPCR and protein analysis showed an increase of angiogenic factors, particularly in the plasminogen-activating receptor and inflammatory systems. VEGF-blockage partly reduced iris neovascularization, and treatment with hypoxia-conditioned RPE medium led to a statistically significant increase in iris neovascularization. CONCLUSIONS This study presents the first evidence of a puncture-induced iris angiogenesis model in the mouse. In a broader context, this novel in vivo model of neovascularization has the potential for noninvasive evaluation of angiogenesis modulating substances.
Collapse
Affiliation(s)
- Ophélie Beaujean
- Department of Clinical Neuroscience, Section of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Filippo Locri
- Department of Clinical Neuroscience, Section of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Monica Aronsson
- Department of Clinical Neuroscience, Section of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anders Kvanta
- Department of Clinical Neuroscience, Section of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Helder André
- Department of Clinical Neuroscience, Section of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
50
|
Chakraborty R, Park HN, Tan CC, Weiss P, Prunty MC, Pardue MT. Association of Body Length with Ocular Parameters in Mice. Optom Vis Sci 2017; 94:387-394. [PMID: 28005683 PMCID: PMC5459603 DOI: 10.1097/opx.0000000000001036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To determine the association between changes in body length with ocular refraction, corneal radii, axial length, and lens thickness in two different mouse strains. METHODS Body length, ocular refraction, corneal radii, axial length, and lens thickness were measured for two inbred mouse strains: 129S1/SvJ (n = 7) and C57BL/6 J (n = 10) from 4 to 12 weeks of age. Body length, from tip of nose to base of tail, was obtained using a digital camera. Biometric parameters, corneal radii, and refractions were measured using spectral-domain optical coherence tomography, automated keratometry, and infrared photorefraction, respectively. A mixed-model ANOVA was performed to examine the changes in ocular parameters as a function of body length and strain in mice controlling for age, gender, and weight over time. RESULTS C57BL/6J mice had significantly longer body length (average body length at 10 weeks, 8.60 ± 0.06 cm) compared to 129S1/SvJ mice (8.31 ± 0.05 cm) during development (P < .001). C57BL/6J mice had significantly hyperopic refractions compared to 129S1/SvJ mice across age (mean refraction at 10 weeks, 129S1/SvJ: +0.99 ± 0.44D vs. C57BL/6J: +6.24 ± 0.38D, P < .001). Corneal radius of curvature, axial length, and lens thickness (except 10 weeks lens thickness) were similar between the two strains throughout the measurement. In the mixed-model ANOVA, changes in body length showed an independent and significant association with the changes in refraction (P = .002) and corneal radii (P = .016) for each mouse strain. No significant association was found between the changes in axial length (P = .925) or lens thickness (P = .973) as a function of body length and strain. CONCLUSIONS Changes in body length are significantly associated with the changes in ocular refraction and corneal radii in different mouse strains. Future studies are needed to determine if the association between body length and ocular refraction are related to changes in corneal curvature in mice.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- *PhD †BS ‡MS Rehab R&D Center of Excellence, Atlanta VA Medical Center, Decatur (RC, CCT, MCP, MTP); Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta (MTP); Department of Ophthalmology, Emory University School of Medicine, Atlanta (RC, HP, CCT, MTP); and Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Georgia (PW)
| | | | | | | | | | | |
Collapse
|