1
|
Meyer KJ, Fingert JH, Anderson MG. Lack of evidence for GWAS signals of exfoliation glaucoma working via monogenic loss-of-function mutation in the nearest gene. Hum Mol Genet 2024:ddae088. [PMID: 38770563 DOI: 10.1093/hmg/ddae088] [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: 01/17/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
PURPOSE Exfoliation syndrome (XFS) is a systemic disease of elastin-rich tissues involving a deposition of fibrillar exfoliative material (XFM) in the anterior chamber of the eye, which can promote glaucoma. The purpose of this study was to create mice with CRISPR/Cas9-induced variations in candidate genes identified from human genome-wide association studies (GWAS) and screen them for indices of XFS. METHODS Variants predicted to be deleterious were sought in the Agpat1, Cacna1a, Loxl1, Pomp, Rbms3, Sema6a, and Tlcd5 genes of C57BL/6J mice using CRISPR/Cas9-based gene editing. Strains were phenotyped by slit-lamp, SD-OCT imaging, and fundus exams at 1-5 mos of age. Smaller cohorts of 12-mos-old mice were also studied. RESULTS Deleterious variants were identified in six targets; Pomp was recalcitrant to targeting. Multiple alleles of some targets were isolated, yielding 12 strains. Across all genotypes and ages, 277 mice were assessed by 902 slit-lamp exams, 928 SD-OCT exams, and 358 fundus exams. Homozygosity for Agpat1 or Cacna1a mutations led to early lethality; homozygosity for Loxl1 mutations led to pelvic organ prolapse, preventing aging. Loxl1 homozygotes exhibited a conjunctival phenotype of potential relevance to XFS. Multiple other genotype-specific phenotypes were variously identified. XFM was not observed in any mice. CONCLUSIONS This study did not detect XFM in any of the strains. This may have been due to species-specific differences, background dependence, or insufficient aging. Alternatively, it is possible that the current candidates, selected based on proximity to GWAS signals, are not effectors acting via monogenic loss-of-function mechanisms.
Collapse
Affiliation(s)
- Kacie J Meyer
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, United States
- Institute for Vision Research, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, United States
| | - John H Fingert
- Institute for Vision Research, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, United States
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Dr, Iowa City, IA 52242, United States
| | - Michael G Anderson
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, United States
- Institute for Vision Research, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, United States
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Dr, Iowa City, IA 52242, United States
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 601 Hwy 6 W, Iowa City, IA 52246, United States
| |
Collapse
|
2
|
Pinke D, Issa JB, Dara GA, Dobos G, Dombeck DA. Full field-of-view virtual reality goggles for mice. Neuron 2023; 111:3941-3952.e6. [PMID: 38070501 PMCID: PMC10841834 DOI: 10.1016/j.neuron.2023.11.019] [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: 06/07/2023] [Revised: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023]
Abstract
Visual virtual reality (VR) systems for head-fixed mice offer advantages over real-world studies for investigating the neural circuitry underlying behavior. However, current VR approaches do not fully cover the visual field of view of mice, do not stereoscopically illuminate the binocular zone, and leave the lab frame visible. To overcome these limitations, we developed iMRSIV (Miniature Rodent Stereo Illumination VR)-VR goggles for mice. Our system is compact, separately illuminates each eye for stereo vision, and provides each eye with an ∼180° field of view, thus excluding the lab frame while accommodating saccades. Mice using iMRSIV while navigating engaged in virtual behaviors more quickly than in a current monitor-based system and displayed freezing and fleeing reactions to overhead looming stimulation. Using iMRSIV with two-photon functional imaging, we found large populations of hippocampal place cells during virtual navigation, global remapping during environment changes, and unique responses of place cell ensembles to overhead looming stimulation.
Collapse
Affiliation(s)
- Domonkos Pinke
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - John B Issa
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Gabriel A Dara
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Gergely Dobos
- 360world Ltd, Sümegvár köz 9, 1118 Budapest, Hungary
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
3
|
Nakagawa H, Alemi H, Wang S, Kahale F, Blanco T, Liu C, Yin J, Dohlman TH, Dana R. Descemet Stripping Only Technique for Corneal Endothelial Damage in Mice. Cornea 2023; 42:470-475. [PMID: 36728991 PMCID: PMC10117527 DOI: 10.1097/ico.0000000000003223] [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: 08/09/2022] [Accepted: 11/14/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Descemet stripping only is an emerging surgical technique used to remove central Descemet membrane and corneal endothelial cells in patients with corneal endothelial disease. Here, we describe a murine model of this procedure to help facilitate basic science investigation and evaluation of postoperative outcomes using this surgical technique. METHODS Slitlamp biomicroscopy, central corneal thickness assessment (by optical coherence tomography), and immunohistochemistry were used to assess the model through 7 weeks of follow-up. RESULTS Complete removal of the endothelium and Descemet membrane was confirmed by slitlamp biomicroscopy and by histology. Central corneal thickness peaked at day 1 postinjury and then declined over the course of 2 weeks to a stable level of persistent edema. Seven weeks postinjury, immunohistochemical staining for ZO-1 showed the area of Descemet stripping was fully covered by enlarged and dysmorphic corneal endothelial cell. No significant ocular complications were appreciated through the end of the follow-up. CONCLUSIONS We demonstrate the feasibility of and provide detailed instructions for a murine model of Descemet stripping only. This model provides a potential in vivo platform to investigate the mechanisms and biology of this emerging surgical procedure.
Collapse
Affiliation(s)
- Hayate Nakagawa
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Hamid Alemi
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Shudan Wang
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Francesca Kahale
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Tomas Blanco
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Catherine Liu
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Jia Yin
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Thomas H. Dohlman
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Reza Dana
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Li S, Pang K, Zhu S, Pate K, Yin J. Perfluorodecalin-based oxygenated emulsion as a topical treatment for chemical burn to the eye. Nat Commun 2022; 13:7371. [PMID: 36450767 PMCID: PMC9712419 DOI: 10.1038/s41467-022-35241-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Chemical injuries to the eye are emergencies with limited acute treatment options other than prompt irrigation and can cause permanent vision loss. We developed a perfluorodecalin-based supersaturated oxygen emulsion (SSOE) to topically deliver high concentration of oxygen to the eye. SSOE is manufactured in hyperbaric conditions and stored in a ready-to-use canister. Upon dispensation, SSOE rapidly raises partial oxygen pressure 3 times over atmospheric level. SSOE is biocompatible with human corneal cells and safe on mouse eyes in vivo. A single topical application of SSOE to the eye after alkali injury significantly promotes corneal epithelial wound healing, decreases anterior chamber exudation, and reduces optical opacity and cataract formation in mice. SSOE treatment reduces intraocular hypoxia, cell death, leukocyte infiltration, production of inflammatory mediators, and hypoxia-inducible factor 1-alpha signaling, thus hastening recovery of normal tissue integrity during the wound healing process. Here, we show that SSOE is an effective topical therapeutic in the acute treatment of ocular chemical injuries.
Collapse
Affiliation(s)
- Sanming Li
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Department of Ophthalmology, Boston, MA, USA
| | - Kunpeng Pang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Department of Ophthalmology, Boston, MA, USA
| | - Shuyan Zhu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Department of Ophthalmology, Boston, MA, USA
| | | | - Jia Yin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Department of Ophthalmology, Boston, MA, USA.
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
Miralles de Imperial-Ollero JA, Gallego-Ortega A, Norte-Muñoz M, Di Pierdomenico J, Valiente-Soriano FJ, Vidal-Sanz M. An in vivo model of focal light emitting diode-induced cone photoreceptor phototoxicity in adult pigmented mice: Protection with bFGF. Exp Eye Res 2021; 211:108746. [PMID: 34450185 DOI: 10.1016/j.exer.2021.108746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE To develop a model of focal injury by blue light-emitting diode (LED)-induced phototoxicity (LIP) in pigmented mouse retinas and to study the effects on cone, Iba-1+ cells and retinal pigment epithelium (RPE) cell populations after administration of basic fibroblast growth factor (bFGF) and minocycline, alone or combined. METHODS In anesthetized dark-adapted adult female pigmented C57BL/6 mice, left pupils were dilated and the eye exposed to LIP (500 lux, 45 s). The retina was monitored longitudinally in vivo with SD-OCT for 7 days (d). Ex vivo, the effects of LIP and its protection with bFGF (0.5 μg) administered alone or combined with minocycline (45 mg/kg) were studied in immunolabeled arrestin-cone outer segments (a+OS) and quantified within a predetermined fixed-size circular area (PCA) centered on the lesion in flattened retinas at 1, 3, 5 or 7d. Moreover, Iba-1+ cells and RPE cell morphology were analysed with Iba-1 and ZO-1 antibodies, respectively. RESULTS LIP caused a focal lesion within the superior-temporal retina with retinal thinning, particularly the outer retinal layers (116.5 ± 2.9 μm to 36.8 ± 6.3 μm at 7d), and with progressive diminution of a+OS within the PCA reaching minimum values at 7d (6218 ± 342 to 3966 ± 311). Administration of bFGF alone (4519 ± 320) or in combination with minocycline (4882 ± 446) had a significant effect on a+OS survival at 7d and Iba-1+ cell activation was attenuated in the groups treated with minocycline. In parallel, the RPE cell integrity was progressively altered after LIP and administration of neuroprotective components had no restorative effect at 7d. CONCLUSIONS LIP resulted in progressive outer retinal damage affecting the OS cone population and RPE. Administration of bFGF increased a+OS survival but did not prevent RPE deterioration.
Collapse
Affiliation(s)
- Juan A Miralles de Imperial-Ollero
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain
| | - Alejandro Gallego-Ortega
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain
| | - María Norte-Muñoz
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain
| | - Johnny Di Pierdomenico
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain
| | - Francisco J Valiente-Soriano
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain.
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca. Campus de CC de la Salud, 30120, El Palmar, Murcia, Spain.
| |
Collapse
|
7
|
Abstract
PURPOSE In humans, loss-of-function mutations in the gene encoding Chordin-like 1 (CHRDL1) cause X-linked megalocornea (MGC1), characterized by bilateral corneal enlargement, decreased corneal thickness, and increased anterior chamber depth (ACD). We sought to determine whether Chrdl1 knockout (KO) mice would recapitulate the ocular findings found in patients with MGC1. METHODS We generated mice with a Chrdl1 KO allele and confirmed that male Chrdl1 hemizygous KO mice do not express Chrdl1 mRNA. We examined the eyes of male mice that were hemizygous for either the wild-type (WT) or KO allele and measured corneal diameter, corneal area, corneal thickness, endothelial cell density, ACD, tear volume, and intraocular pressure. We also harvested retinas and counted retinal ganglion cell numbers. Eye segregation pattern in the dorsal lateral geniculate nucleus were also compared between male Chrdl1 KO and WT mice. RESULTS Male Chrdl1 KO mice do not have larger cornea diameters than WT mice. KO mice have significantly thicker central corneas (116.5 ± 3.9 vs. 100.9 ± 4.2 μm, P = 0.013) and smaller ACD (325.7 ± 5.7 vs. 405.6 ± 6.3 μm, P < 0.001) than WT mice, which is the converse of what occurs in patients who lack CHRDL1. Retinal-thalamic projections and other ocular measurements did not significantly differ between KO and WT mice. CONCLUSIONS Male Chrdl1 KO mice do not have the same anterior chamber abnormalities seen in humans with CHRDL1 mutations. Therefore, Chrdl1 KO mice do not recapitulate the human MGC1 phenotype. Nevertheless, Chrdl1 plays a role during mouse ocular development because corneas in KO mice differ from those in WT mice.
Collapse
|
8
|
Crane R, Conley SM, Al-Ubaidi MR, Naash MI. Gene Therapy to the Retina and the Cochlea. Front Neurosci 2021; 15:652215. [PMID: 33815052 PMCID: PMC8010260 DOI: 10.3389/fnins.2021.652215] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Vision and hearing disorders comprise the most common sensory disorders found in people. Many forms of vision and hearing loss are inherited and current treatments only provide patients with temporary or partial relief. As a result, developing genetic therapies for any of the several hundred known causative genes underlying inherited retinal and cochlear disorders has been of great interest. Recent exciting advances in gene therapy have shown promise for the clinical treatment of inherited retinal diseases, and while clinical gene therapies for cochlear disease are not yet available, research in the last several years has resulted in significant advancement in preclinical development for gene delivery to the cochlea. Furthermore, the development of somatic targeted genome editing using CRISPR/Cas9 has brought new possibilities for the treatment of dominant or gain-of-function disease. Here we discuss the current state of gene therapy for inherited diseases of the retina and cochlea with an eye toward areas that still need additional development.
Collapse
Affiliation(s)
- Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
9
|
Ainsbury EA, Dalke C, Hamada N, Benadjaoud MA, Chumak V, Ginjaume M, Kok JL, Mancuso M, Sabatier L, Struelens L, Thariat J, Jourdain JR. Radiation-induced lens opacities: Epidemiological, clinical and experimental evidence, methodological issues, research gaps and strategy. ENVIRONMENT INTERNATIONAL 2021; 146:106213. [PMID: 33276315 DOI: 10.1016/j.envint.2020.106213] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 06/12/2023]
Abstract
In 2011, the International Commission on Radiological Protection (ICRP) recommended reducing the occupational equivalent dose limit for the lens of the eye from 150 mSv/year to 20 mSv/year, averaged over five years, with no single year exceeding 50 mSv. With this recommendation, several important assumptions were made, such as lack of dose rate effect, classification of cataracts as a tissue reaction with a dose threshold at 0.5 Gy, and progression of minor opacities into vision-impairing cataracts. However, although new dose thresholds and occupational dose limits have been set for radiation-induced cataract, ICRP clearly states that the recommendations are chiefly based on epidemiological evidence because there are a very small number of studies that provide explicit biological and mechanistic evidence at doses under 2 Gy. Since the release of the 2011 ICRP statement, the Multidisciplinary European Low Dose Initiative (MELODI) supported in April 2019 a scientific workshop that aimed to review epidemiological, clinical and biological evidence for radiation-induced cataracts. The purpose of this article is to present and discuss recent related epidemiological and clinical studies, ophthalmic examination techniques, biological and mechanistic knowledge, and to identify research gaps, towards the implementation of a research strategy for future studies on radiation-induced lens opacities. The authors recommend particularly to study the effect of ionizing radiation on the lens in the context of the wider, systemic effects, including in the retina, brain and other organs, and as such cataract is recommended to be studied as part of larger scale programs focused on multiple radiation health effects.
Collapse
Affiliation(s)
- Elizabeth A Ainsbury
- Public Health England (PHE) Centre for Radiation, Chemical and Environmental Hazards, Oxon, United Kingdom.
| | - Claudia Dalke
- Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Germany.
| | - Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan.
| | - Mohamed Amine Benadjaoud
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), BP 17, 31 avenue de la division Leclerc, Fontenay-aux-Roses, France.
| | - Vadim Chumak
- National Research Centre for Radiation Medicine, Ukraine.
| | | | - Judith L Kok
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, (ENEA), Rome, Italy.
| | - Laure Sabatier
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Saclay, France.
| | | | - Juliette Thariat
- Laboratoire de physique corpusculaire IN2P3/ENSICAEN -UMR6534 - Unicaen - Normandie University, France
| | - Jean-René Jourdain
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), BP 17, 31 avenue de la division Leclerc, Fontenay-aux-Roses, France.
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Retina transduction by rAAV2 after intravitreal injection: comparison between mouse and rat. Gene Ther 2019; 26:479-490. [PMID: 31562387 DOI: 10.1038/s41434-019-0100-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022]
Abstract
Adeno-associated virus vectors (rAAV) are currently the most common vehicle used in clinical trials of retinal gene therapy, usually delivered through subretinal injections to target cells of the outer retina. However, targeting the inner retina requires intravitreal injections, a simple and safe procedure, which is effective for transducing the rodent retina, but still of low efficiency in the eyes of primates. We investigated whether adjuvant pharmacological agents may enhance rAAV transduction of the retinas of mouse and rat after intravitreal delivery. Tyrosine kinase inhibitors were highly efficient in mice, especially imatinib and genistein, and promoted transduction even of the outer retina. In rats, however, we report that they were not effective. Even with direct proteasomal inhibition in rats, the effects upon transduction were only minimal and restricted to the inner retina. Even tyrosine capsid mutant rAAVs in rats had a transduction profile similar to wtAAV. Thus, the differences between mouse and rat, in both eye size and the inner limiting membrane, compromise the efficiency of AAV vectors penetration from the vitreous into the retina, and impact the efficacy of strategies developed to enhance intravitreal retinal rAAV transduction. Further improvement of strategies, then are required.
Collapse
|
12
|
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: 5.8] [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
|
13
|
Cross SH, Mckie L, Keighren M, West K, Thaung C, Davey T, Soares DC, Sanchez-Pulido L, Jackson IJ. Missense Mutations in the Human Nanophthalmos Gene TMEM98 Cause Retinal Defects in the Mouse. Invest Ophthalmol Vis Sci 2019; 60:2875-2887. [PMID: 31266059 PMCID: PMC6986908 DOI: 10.1167/iovs.18-25954] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose We previously found a dominant mutation, Rwhs, causing white spots on the retina accompanied by retinal folds. Here we identify the mutant gene to be Tmem98. In humans, mutations in the orthologous gene cause nanophthalmos. We modeled these mutations in mice and characterized the mutant eye phenotypes of these and Rwhs. Methods The Rwhs mutation was identified to be a missense mutation in Tmem98 by genetic mapping and sequencing. The human TMEM98 nanophthalmos missense mutations were made in the mouse gene by CRISPR-Cas9. Eyes were examined by indirect ophthalmoscopy and the retinas imaged using a retinal camera. Electroretinography was used to study retinal function. Histology, immunohistochemistry, and electron microscopy techniques were used to study adult eyes. Results An I135T mutation of Tmem98 causes the dominant Rwhs phenotype and is perinatally lethal when homozygous. Two dominant missense mutations of TMEM98, A193P and H196P, are associated with human nanophthalmos. In the mouse these mutations cause recessive retinal defects similar to the Rwhs phenotype, either alone or in combination with each other, but do not cause nanophthalmos. The retinal folds did not affect retinal function as assessed by electroretinography. Within the folds there was accumulation of disorganized outer segment material as demonstrated by immunohistochemistry and electron microscopy, and macrophages had infiltrated into these regions. Conclusions Mutations in the mouse orthologue of the human nanophthalmos gene TMEM98 do not result in small eyes. Rather, there is localized disruption of the laminar structure of the photoreceptors.
Collapse
Affiliation(s)
- Sally H. Cross
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Lisa Mckie
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Margaret Keighren
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Katrine West
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Caroline Thaung
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, United Kingdom
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, United Kingdom
| | - Tracey Davey
- Electron Microscopy Research Services, Newcastle University, Newcastle NE2 4HH, United Kingdom
| | - Dinesh C. Soares
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Luis Sanchez-Pulido
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - Ian J. Jackson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
14
|
Mouse models for microphthalmia, anophthalmia and cataracts. Hum Genet 2019; 138:1007-1018. [PMID: 30919050 PMCID: PMC6710221 DOI: 10.1007/s00439-019-01995-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/04/2019] [Indexed: 12/21/2022]
Abstract
Mouse mutants are a long-lasting, valuable tool to identify genes underlying eye diseases, because the absence of eyes, very small eyes and severely affected, cataractous eyes are easily to detect without major technical equipment. In mice, actually 145 genes or loci are known for anophthalmia, 269 for microphthalmia, and 180 for cataracts. Approximately, 25% of the loci are not yet characterized; however, some of the ancient lines are extinct and not available for future research. The phenotypes of the mutants represent a continuous spectrum either in anophthalmia and microphthalmia, or in microphthalmia and cataracts. On the other side, mouse models are still missing for some genes, which have been identified in human families to be causative for anophthalmia, microphthalmia, or cataracts. Finally, the mouse offers the possibility to genetically test the roles of modifiers and the role of SNPs; these aspects open new avenues for ophthalmogenetics in the mouse.
Collapse
|
15
|
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: 231] [Impact Index Per Article: 46.2] [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
|
16
|
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: 1.4] [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
|
17
|
Abstract
Purpose To investigate the entire nerve architecture and content of the two main sensory neuropeptides in mouse cornea to determine if it is a good model with similarities to human corneal innervation. Methods Mice aged 1 to 24 weeks were used. The corneas were stained with neuronal-class βIII-tubulin, calcitonin gene–related peptide (CGRP), and substance P (SP) antibodies; whole-mount images were acquired to build an entire view of corneal innervation. To test the origin of CGRP and SP, trigeminal ganglia (TG) were processed for immunofluorescence. Relative corneal nerve fiber densities or neuron numbers were assessed by computer-assisted analysis. Results Between 1 and 3 weeks after birth, mouse cornea was mainly composed of a stromal nerve network. At 4 weeks, a whorl-like structure (or vortex) appeared that gradually became more defined. By 8 weeks, anatomy of corneal nerves had reached maturity. Epithelial bundles converged into the central area to form the vortex. The number and pattern of whorl-like structures were different. Subbasal nerve density and nerve terminals were greater in the center than the periphery. Nerve fibers and terminals that were CGRP-positive were more abundant than SP-positive nerves and terminals. In trigeminal ganglia, the number of CGRP-positive neurons significantly outnumbered those positive for SP. Conclusions This is the first study to show a complete map of the entire corneal nerves and CGRP and SP sensory neuropeptide distribution in the mouse cornea. This finding shows mouse corneal innervation has many similarities to human cornea and makes the mouse an appropriate model to study pathologies involving corneal nerves.
Collapse
Affiliation(s)
- Jiucheng He
- Louisiana State University Health School of Medicine, New Orleans, Louisiana, United States 2Neuroscience Center of Excellence, Louisiana State University Health, New Orleans, Louisiana, United States 3Department of Ophthalmology, Louisiana State Universi
| | - Haydee E P Bazan
- Louisiana State University Health School of Medicine, New Orleans, Louisiana, United States 2Neuroscience Center of Excellence, Louisiana State University Health, New Orleans, Louisiana, United States 3Department of Ophthalmology, Louisiana State Universi
| |
Collapse
|
18
|
The pros and cons of vertebrate animal models for functional and therapeutic research on inherited retinal dystrophies. Prog Retin Eye Res 2015; 48:137-59. [DOI: 10.1016/j.preteyeres.2015.04.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/12/2015] [Accepted: 04/16/2015] [Indexed: 01/19/2023]
|
19
|
Marcos S, González-Lázaro M, Beccari L, Carramolino L, Martin-Bermejo MJ, Amarie O, Martín DMS, Torroja C, Bogdanović O, Doohan R, Puk O, de Angelis MH, Graw J, Gomez-Skarmeta JL, Casares F, Torres M, Bovolenta P. Meis1 coordinates a network of genes implicated in eye development and microphthalmia. Development 2015; 142:3009-20. [DOI: 10.1242/dev.122176] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/17/2015] [Indexed: 01/08/2023]
Abstract
Microphthalmos is a rare congenital anomaly characterized by reduced eye size and visual deficits of variable degrees. Sporadic and hereditary microphthalmos has been associated to heterozygous mutations in genes fundamental for eye development. Yet, many cases are idiopathic or await the identification of molecular causes. Here we show that haploinsufficiency of Meis1, a transcription factor with an evolutionary conserved expression in the embryonic trunk, brain and sensory organs, including the eye, causes microphthalmic traits and visual impairment, in adult mice. By combining the analysis of Meis1 loss-of-function and conditional Meis1 functional rescue with ChIP-seq and RNA-seq approaches we show that, in contrast to Meis1 preferential association with Hox-Pbx binding sites in the trunk, Meis1 binds to Hox/Pbx-independent sites during optic cup development. In the eye primordium, Meis1 coordinates, in a dose-dependent manner, retinal proliferation and differentiation by regulating genes responsible for human microphthalmia and components the Notch signalling pathway. In addition, Meis1 is required for eye patterning by controlling a set of eye territory-specific transcription factors, so that in Meis1−/− embryos boundaries among the different eye territories are shifted or blurred. We thus propose that Meis1 is at the core of a genetic network implicated in eye patterning/microphthalmia, itself representing an additional candidate for syndromic cases of these ocular malformations.
Collapse
Affiliation(s)
- Séverine Marcos
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Monica González-Lázaro
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Leonardo Beccari
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Laura Carramolino
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Maria Jesus Martin-Bermejo
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| | - Oana Amarie
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | - Daniel Mateos-San Martín
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Ozren Bogdanović
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
- ARC Center of Excellence in Plant Energy Biology, School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, WA 6009, Australia
| | - Roisin Doohan
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Oliver Puk
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | | | - Jochen Graw
- Institute of Developmental Genetics Helmholtz Center Munich; D-85764 Neuherberg, Germany
| | - Jose Luis Gomez-Skarmeta
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
| | - Fernando Casares
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-UPO, Carretera de Utrera Km1, E-41013 Sevilla, Spain
| | - Miguel Torres
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular “Severo Ochoa”, CSIC-UAM, c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
- CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera, 1, E-28049 Madrid, Spain
| |
Collapse
|
20
|
Kraus P, V S, Yu HB, Xing X, Lim SL, Adler T, Pimentel JAA, Becker L, Bohla A, Garrett L, Hans W, Hölter SM, Janas E, Moreth K, Prehn C, Puk O, Rathkolb B, Rozman J, Adamski J, Bekeredjian R, Busch DH, Graw J, Klingenspor M, Klopstock T, Neff F, Ollert M, Stoeger T, Yildrim AÖ, Eickelberg O, Wolf E, Wurst W, Fuchs H, Gailus-Durner V, de Angelis MH, Lufkin T, Stanton LW. Pleiotropic functions for transcription factor zscan10. PLoS One 2014; 9:e104568. [PMID: 25111779 PMCID: PMC4128777 DOI: 10.1371/journal.pone.0104568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/12/2014] [Indexed: 12/17/2022] Open
Abstract
The transcription factor Zscan10 had been attributed a role as a pluripotency factor in embryonic stem cells based on its interaction with Oct4 and Sox2 in in vitro assays. Here we suggest a potential role of Zscan10 in controlling progenitor cell populations in vivo. Mice homozygous for a Zscan10 mutation exhibit reduced weight, mild hypoplasia in the spleen, heart and long bones and phenocopy an eye malformation previously described for Sox2 hypomorphs. Phenotypic abnormalities are supported by the nature of Zscan10 expression in midgestation embryos and adults suggesting a role for Zscan10 in either maintaining progenitor cell subpopulation or impacting on fate choice decisions thereof.
Collapse
Affiliation(s)
- Petra Kraus
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Sivakamasundari V
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Hong Bing Yu
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Xing Xing
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Siew Lan Lim
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Thure Adler
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Juan Antonio Aguilar Pimentel
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Klinikum rechts der Isar der Technischen Universität München, Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Munich, Germany
| | - Lore Becker
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexander Bohla
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sabine M. Hölter
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Eva Janas
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kristin Moreth
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Cornelia Prehn
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Puk
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jan Rozman
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jerzy Adamski
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Raffi Bekeredjian
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine III, Division of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Dirk H. Busch
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Microbiology, Immunology, and Hygiene, Technische Universität München, Munich, Germany
| | - Jochen Graw
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Klingenspor
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Neurology, Friedrich-Baur-Institut, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Frauke Neff
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Markus Ollert
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Klinikum rechts der Isar der Technischen Universität München, Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Munich, Germany
| | - Tobias Stoeger
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ali Önder Yildrim
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Eickelberg
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Eckhard Wolf
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Wolfgang Wurst
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Developmental Biology, Technische Universität München, Freising-Weihenstephan, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
- Deutsches Institut für Neurodegenerative Erkrankungen Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- Member of German Center for Diabetes Research, Neuherberg, Germany
| | - Thomas Lufkin
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Lawrence W. Stanton
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
21
|
Refractive index measurement of the mouse crystalline lens using optical coherence tomography. Exp Eye Res 2014; 125:62-70. [PMID: 24939747 DOI: 10.1016/j.exer.2014.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/22/2014] [Accepted: 05/24/2014] [Indexed: 11/24/2022]
Abstract
In recent years, there has been a growing interest for using mouse models in refractive development and myopia research. The crystalline lens is a critical optical component of the mouse eye that occupies greater than 50% of the ocular space, and significant increases in thickness with age. However, changes in refractive index of the mouse crystalline lens are less known. In this study, we examined the changes in thickness and refractive index of the mouse crystalline lens for two different strains, wild-type (WT) and a nyx mutant (nob) over the course of normal visual development or after form deprivation. Refractive index and lens thickness measurements were made on ex vivo lenses using spectral domain optical coherence tomography (SD-OCT). Comparison of refractive index measurements on 5 standard ball lenses using the SD-OCT and their known refractive indices (manufacturer provided) indicated good precision (intra-class correlation coefficient, 0.998 and Bland-Altman coefficient of repeatability, 0.116) of the SD-OCT to calculate mouse lens refractive index ex vivo. During normal visual development, lens thickness increased significantly with age for three different cohorts of mice, aged 4 (average thickness from both eyes; WT: 1.78 ± 0.03, nob: 1.79 ± 0.08 mm), 10 (WT: 2.02 ± 0.05, nob: 2.01 ± 0.04 mm) and 16 weeks (WT: 2.12 ± 0.06, nob: 2.09 ± 0.06 mm, p < 0.001). Lens thickness was not significantly different between the two strains at any age (p = 0.557). For mice with normal vision, refractive index for isolated crystalline lenses in nob mice was significantly greater than WT mice (mean for all ages; WT: 1.42 ± 0.01, nob: 1.44 ± 0.001, p < 0.001). After 4 weeks of form deprivation to the right eye using a skull-mounted goggling apparatus, a thinning of the crystalline lens was observed in both right and left eyes of goggled animals compared to their naïve controls (average from both the right and the left eye) for both strains (p = 0.052). In form deprived mice, lens refractive index was significantly different between the goggled animals and non-goggled naïve controls in nob mice, but not in WT mice (p = 0.009). Both eyes of goggled nob mice had significantly greater lens refractive index (goggled, 1.49 ± 0.01; opposite, 1.47 ± 0.03) compared to their naïve controls (1.45 ± 0.02, p < 0.05). The results presented here suggest that there are genetic differences in the crystalline lens refractive index of the mouse eye, and that the lens refractive index in mice significantly increase with form deprivation. Research applications requiring precise optical measurements of the mouse eye should take these lens refractive indices into account when interpreting SD-OCT data.
Collapse
|
22
|
Boussommier-Calleja A, Overby DR. The influence of genetic background on conventional outflow facility in mice. Invest Ophthalmol Vis Sci 2013; 54:8251-8. [PMID: 24235015 DOI: 10.1167/iovs.13-13025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Intraocular pressure (IOP) varies between genetically distinct strains of mice. The purpose was to test the hypothesis that strain-dependent differences in IOP are attributable to differences in conventional outflow facility (C). METHODS The IOP was measured by rebound tonometry in conscious or anesthetized BALB/cJ, C57BL/6J, and CBA/J mice (N = 6-10 per strain). Conventional outflow facility was measured by ex vivo perfusion of enucleated eyes (N = 9-10 per strain). RESULTS Conscious IOP varied between strains, being highest in CBA/J (14.5 ± 0.9 mm Hg, mean ± SD), intermediate in C57BL/6J (12.3 ± 1.0 mm Hg), and lowest in BALB/cJ (10.6 ± 1.8 mm Hg) mice. Anesthesia reduced IOP and eliminated any detectable differences between strains. Conventional outflow facility also varied between strains, but, in contrast to IOP, C was lowest in CBA/J (0.0113 ± 0.0031 μL/min/mm Hg) and highest in BALB/cJ (0.0164 ± 0.0059 μL/min/mm Hg). Like IOP, C was intermediate in C57BL/6J (0.0147 ± 0.0029 μL/min/mm Hg). There was a strong correlation between conscious IOP and outflow resistance (1/C) from individual eyes across all three strains, revealing that 70% of the variation in IOP was attributable to variation in outflow resistance. CONCLUSIONS Differences in IOP among three genetically distinct murine strains are attributable largely to differences in conventional outflow facility. These results motivate further studies using mice to identify the morphologic and genetic factors that underlie IOP regulation within the conventional outflow pathway.
Collapse
|
23
|
Longitudinal fundus and retinal studies with SD-OCT: a comparison of five mouse inbred strains. Mamm Genome 2013; 24:198-205. [PMID: 23681115 DOI: 10.1007/s00335-013-9457-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 04/24/2013] [Indexed: 02/05/2023]
Abstract
Spectral domain optical coherence tomography (SD-OCT) has recently been established as a method for in vivo imaging of fundus and retina in the mouse. It enables more effective studies of retinal diseases including investigations of etiopathologic mechanisms. In order to learn more about longitudinal fundus development and to enable recognition of disease-associated irregularities, we performed confocal scanning laser ophthalmoscopy (cSLO) and SD-OCT measurements in the inbred strains C57BL/6J, C3HeB/FeJ, FVB/NCrl, BALB/cByJ, and 129S2/SvJ when they were between 2 and 6 months of age. In general, cSLO and SD-OCT data did not reveal sex-specific or unilateral differences. C3HeB/FeJ and FVB/NCrl mice showed diffuse choroidal dysplasia. Choroidal vein-like structures appeared as dark fundus stripes in C3HeB/FeJ. In FVB/NCrl, fundus fleck accumulation was found. In contrast, only minor time-dependent changes of fundus appearance were observed in C57BL/6J, BALB/cByJ, and 129S2/SvJ. This was also found for individual fundic main blood vessel patterns in all inbred strains. Vessel numbers varied between 6 and 13 in C57BL/6J. This was comparable in most cases. We further found that retinae were significantly thicker in C57BL/6J compared to the other strains. Total retinal thickness generally did not change between 2 and 6 months of age. As a conclusion, our results indicate lifelong pathologic processes in C3HeB/FeJ and FVB/NCrl that affect choroid and orbital tissues. Inbred strains with regular retinal development did not reveal major time-dependent variations of fundus appearance, blood vessel pattern, or retinal thickness. Consequently, progressive changes of these parameters are suitable indicators for pathologic outliers.
Collapse
|
24
|
Khan A, Fu H, Tan LA, Harper JE, Beutelspacher SC, Larkin DFP, Lombardi G, McClure MO, George AJT. Dendritic cell modification as a route to inhibiting corneal graft rejection by the indirect pathway of allorecognition. Eur J Immunol 2013; 43:734-46. [PMID: 23212959 PMCID: PMC3615172 DOI: 10.1002/eji.201242914] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/30/2012] [Accepted: 11/28/2012] [Indexed: 01/08/2023]
Abstract
Dendritic cell (DC) modification is a potential strategy to induce clinical transplantation tolerance. We compared two DC modification strategies to inhibit allogeneic T-cell proliferation. In the first strategy, murine DCs were transduced with a lentiviral vector expressing CTLA4-KDEL, a fusion protein that prevents surface CD80/86 expression by retaining the co-stimulatory molecules within the ER. In the second approach, DCs were transduced to express the tryptophan-catabolising enzyme IDO. CTLA4-KDEL-expressing DCs induced anergy in alloreactive T cells and generated both CD4+CD25+ and CD4+CD25− Treg cells (with direct and indirect donor allospecificity and capacity for linked suppression) both in vitro and in vivo. In contrast, T-cell unresponsiveness induced by IDO+ DCs lacked donor specificity. In the absence of any immunosuppressive treatment, i.v. administration of CTLA4-KDEL-expressing DCs resulted in long-term survival of corneal allografts only when the DCs were capable of indirect presentation of alloantigen. This study demonstrates the therapeutic potential of CTLA4-KDEL-expressing DCs in tolerance induction.
Collapse
Affiliation(s)
- Adnan Khan
- Section of Molecular Immunology, Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pardue MT, Stone RA, Iuvone PM. Investigating mechanisms of myopia in mice. Exp Eye Res 2013; 114:96-105. [PMID: 23305908 DOI: 10.1016/j.exer.2012.12.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/19/2012] [Accepted: 12/25/2012] [Indexed: 11/25/2022]
Abstract
Genetic and environmental factors have been shown to control visually-guided eye growth and influence myopia development. However, investigations into the intersection of these two factors in controlling refractive development have been limited by the lack of a genetically modifiable animal model. Technological advances have now made it possible to assess refractive state and ocular biometry in the small mouse eye and therefore to exploit the many genetic mouse mutants to investigate mechanisms of visually-guided eye growth. This review considers the benefits and challenges of studying refractive development in mice, compares the results of refractive error and ocular biometry from wild-type strains and genetic models in normal laboratory visual environments or with disrupted visual input, and discusses some of the remaining challenges in interpreting data from the mouse to validate and standardize methods between labs.
Collapse
Affiliation(s)
- Machelle T Pardue
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA.
| | | | | |
Collapse
|
26
|
Comparative Ocular Anatomy in Commonly Used Laboratory Animals. ASSESSING OCULAR TOXICOLOGY IN LABORATORY ANIMALS 2012. [DOI: 10.1007/978-1-62703-164-6_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
27
|
Puk O, Möller G, Geerlof A, Krowiorz K, Ahmad N, Wagner S, Adamski J, de Angelis MH, Graw J. The pathologic effect of a novel neomorphic Fgf9(Y162C) allele is restricted to decreased vision and retarded lens growth. PLoS One 2011; 6:e23678. [PMID: 21858205 PMCID: PMC3157460 DOI: 10.1371/journal.pone.0023678] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 07/25/2011] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor (Fgf) signalling plays a crucial role in many developmental processes. Among the Fgf pathway ligands, Fgf9 (UniProt: P54130) has been demonstrated to participate in maturation of various organs and tissues including skeleton, testes, lung, heart, and eye. Here we establish a novel Fgf9 allele, discovered in a dominant N-ethyl-N-nitrosourea (ENU) screen for eye-size abnormalities using the optical low coherence interferometry technique. The underlying mouse mutant line Aca12 was originally identified because of its significantly reduced lens thickness. Linkage studies located Aca12 to chromosome 14 within a 3.6 Mb spanning interval containing the positional candidate genes Fgf9 (MGI: 104723), Gja3 (MGI: 95714), and Ift88 (MGI: 98715). While no sequence differences were found in Gja3 and Ift88, we identified an A→G missense mutation at cDNA position 770 of the Fgf9 gene leading to an Y162C amino acid exchange. In contrast to previously described Fgf9 mutants, Fgf9Y162C carriers were fully viable and did not reveal reduced body-size, male-to-female sexual reversal or skeletal malformations. The histological analysis of the retina as well as its basic functional characterization by electroretinography (ERG) did not show any abnormality. However, the analysis of head-tracking response of the Fgf9Y162C mutants in a virtual drum indicated a gene-dosage dependent vision loss of almost 50%. The smaller lenses in Fgf9Y162C suggested a role of Fgf9 during lens development. Histological investigations showed that lens growth retardation starts during embryogenesis and continues after birth. Young Fgf9Y162C lenses remained transparent but developed age-related cataracts. Taken together, Fgf9Y162C is a novel neomorphic allele that initiates microphakia and reduced vision without effects on organs and tissues outside the eye. Our data point to a role of Fgf9 signalling in primary and secondary lens fiber cell growth. The results underline the importance of allelic series to fully understand multiple functions of a gene.
Collapse
MESH Headings
- Alleles
- Amino Acid Sequence
- Animals
- Base Sequence
- Binding, Competitive
- Cataract/genetics
- Female
- Fibroblast Growth Factor 9/chemistry
- Fibroblast Growth Factor 9/genetics
- Fibroblast Growth Factor 9/metabolism
- Genotype
- Haplotypes
- Heparin/metabolism
- Lens, Crystalline/embryology
- Lens, Crystalline/growth & development
- Lens, Crystalline/metabolism
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Models, Molecular
- Molecular Sequence Data
- Mutation, Missense
- Protein Binding
- Protein Structure, Tertiary
- Sequence Homology, Amino Acid
- Vision, Ocular/genetics
- Visual Acuity/genetics
Collapse
Affiliation(s)
- Oliver Puk
- German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wang L, Považay B, Chen YP, Hofer B, Drexler W, Guggenheim JA. Heritability of ocular component dimensions in mice phenotyped using depth-enhanced swept source optical coherence tomography. Exp Eye Res 2011; 93:482-90. [PMID: 21726551 DOI: 10.1016/j.exer.2011.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 06/02/2011] [Accepted: 06/14/2011] [Indexed: 10/18/2022]
Abstract
The range of genetic and genomic resources available makes the mouse a powerful model for the genetic dissection of complex traits. Because accurate, high-throughput phenotypic characterisation is crucial to the success of such endeavours, we recently developed an optical coherence tomography (OCT) system with extended depth range scanning capability for measuring ocular component dimensions in mice. In order to test whether the accuracy and reproducibility of our OCT system was sufficient for gene mapping studies, we carried out an experiment designed to estimate the heritability of mouse ocular component dimensions. High-resolution, two dimensional tomograms were obtained for both eyes of 11 pairs of 8 week-old outbred MF1 mice. Subsequently, images were obtained when their offspring were aged 8 weeks. Biometric data were extracted after image segmentation, reconstruction of the geometric shape of each surface, and calculation of intraocular distances. The repeatability of measurements was evaluated for 12 mice scanned on consecutive days. Heritability estimates were calculated using variance components analysis. Sets of tomograms took ∼2 s to acquire. Biometric data could be obtained for 98% of the 130 eyes scanned. The 95% limits of repeatability ranged from ±6 to ±16 μm for the axial ocular component dimensions. The heritability of the axial ocular components was 0.6-0.8, except for corneal thickness, which had a heritability not significantly different from zero. In conclusion, axial ocular component dimensions are highly heritable in mice, as they are in humans. OCT with extended depth range scanning can be used to rapidly phenotype individual mice with sufficient accuracy and precision to permit gene mapping studies.
Collapse
Affiliation(s)
- Ling Wang
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff CF24 4LU, Wales, UK
| | | | | | | | | | | |
Collapse
|
29
|
Chou TH, Kocaoglu OP, Borja D, Ruggeri M, Uhlhorn SR, Manns F, Porciatti V. Postnatal elongation of eye size in DBA/2J mice compared with C57BL/6J mice: in vivo analysis with whole-eye OCT. Invest Ophthalmol Vis Sci 2011; 52:3604-12. [PMID: 21372015 DOI: 10.1167/iovs.10-6340] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To characterize postnatal changes in eye size in glaucomatous DBA/2J (D2) mice and in nonglaucomatous C57BL/6J mice (B6) in vivo by means of whole-eye optical coherence tomography (OCT). METHODS D2 (n = 32) and B6 (n = 36) mice were tested between 2 and 20 months of age in eight age bins. A custom time-domain OCT system with a center wavelength of 825 nm and an axial scan length of 7.1 mm produced axial A-scan interferograms at a rate of 20 A-lines/s with a resolution of 8 μm. Axial length (AL), corneal thickness (CT), anterior chamber depth (ACD), lens thickness (LT), vitreous chamber depth (VCD), and retinal thickness (RT) were measured in the optical axis and adjusted with corresponding refractive indices. Corneal curvature (CC) and IOP were also measured. RESULTS AL increased (P < 0.001) more in the D2 (21%) than in the B6 (9%) mice. There was an interaction effect (two-way ANOVA, P < 0.001) between age and strain for AL, CT, ACD, and VCD. In the D2 mice, the lens became dislocated posteriorly. Multiple regression analysis in the D2 mice revealed an independent effect of age and IOP (P ≤ 0.01) on axial length. CC steepened in the older D2 mice, whereas it flattened in the B6 mice. CONCLUSIONS In D2 mice, postnatal elongation of AL is larger than that in B6 mice and is associated with a greater increase in ACD and IOP, which seems to be a causal factor. The ease of use, short acquisition time, and noninvasiveness of whole-eye OCT make it suitable for routine use in longitudinal studies of mouse models.
Collapse
Affiliation(s)
- Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Gabriele ML, Ishikawa H, Schuman JS, Ling Y, Bilonick RA, Kim JS, Kagemann L, Wollstein G. Optic nerve crush mice followed longitudinally with spectral domain optical coherence tomography. Invest Ophthalmol Vis Sci 2011; 52:2250-4. [PMID: 21398282 DOI: 10.1167/iovs.10-6311] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To investigate the longitudinal effect of optic nerve crush injury in mice by measuring retinal thickness with spectral-domain optical coherence tomography (SD-OCT). METHODS Optic nerves of one eye from each C57Bl/6 mouse were crushed under direct visualization for 3 seconds, 1 mm posterior to the globe. The optic nerve head (ONH) was imaged with SD-OCT (1.5 × 1.5 × 2.0 mm scan) before the surgical intervention and repeated subsequently for up to 32 days postinjury. A cohort of mice not exposed to the nerve crush procedure served as control. En face SD-OCT images were used to manually align subsequent scans to the baseline en face image. Total retinal thickness (TRT) (along a sampling band with radii 0.33-0.42 mm centered on the ONH) from each follow-up day was automatically quantified for global and sectoral measurements using custom software. Linear mixed-effects models with quadratic terms were fitted to compare TRT of nerve-crushed and control eyes over time. RESULTS Eleven eyes from 11 nerve crush mice (baseline age 76 ± 11.8 days) and eight eyes from four healthy mice (baseline age 64 ± 0 days) were included. The control eyes showed a small, gradual, and consistent TRT increase throughout follow-up. Nerve-crushed eyes showed an initial period of thickening, followed by thinning and slight rebound after day 21. The decrease in thickness observed after the early thickening resolved was statistically significantly different from the control eyes (P < 0.05 for global and sectoral measurements). CONCLUSIONS SD-OCT can be used to quantitatively monitor changes in retinal thickness in mice over time.
Collapse
Affiliation(s)
- Michelle L Gabriele
- UPMC Eye Center, Eye and Ear Institute, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine, 203 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Wang L, Hofer B, Chen YP, Guggenheim JA, Drexler W, Povazay B. Highly reproducible swept-source, dispersion-encoded full-range biometry and imaging of the mouse eye. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:046004. [PMID: 20799806 DOI: 10.1117/1.3463480] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We report a high-speed, dispersion-encoded, full-range (DEFR) swept-source optical coherence tomography system for in vivo ocular imaging and biometry of small animals. The fast DEFR algorithm removes the depth ambiguity, gives access to objects located at the zero delay position, and doubles the sampling depth to 2x5.0 mm (at -101 to -71 dB sensitivity) in a single scan using 2048 samples/depth scan 0.43 nm line width of a light source operating at 1056 nm with 70 nm tuning range. The acquisition speed (frames of 512 depth scans in 18.3 ms) permits precise on-line monitoring during positioning and provides cross-sectional views of the mouse eye. Preliminary studies demonstrate high-throughput, reproducible assessment of multiple biometric features (e.g., day-to-day reproducibility of axial length measurement +/-5.3 microm) that is insensitive to eye motion sufficient for long-term monitoring.
Collapse
Affiliation(s)
- Ling Wang
- Cardiff University, School of Optometry and Vision Sciences, Wales, United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Lively GD, Koehn D, Hedberg-Buenz A, Wang K, Anderson MG. Quantitative trait loci associated with murine central corneal thickness. Physiol Genomics 2010; 42:281-6. [PMID: 20423963 DOI: 10.1152/physiolgenomics.00140.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The cornea is a specialized transparent tissue responsible for refracting light, serving as a protective barrier, and lending structural support to eye shape. Given its importance, the cornea exhibits a surprising amount of phenotypic variability in some traits, including central corneal thickness (CCT). More than a mere anatomic curiosity, differences in CCT have recently been associated with risk for glaucoma. Although multiple lines of evidence support a strong role for heredity in regulating CCT, the responsible genes remain unknown. To better understand the genetic basis of CCT variability, we conducted a genomewide quantitative trait locus (QTL) analysis with (C57BLKS/J x SJL/J) F(2) mice. This experiment identified a locus, Cctq1 (central corneal thickness QTL 1) on chromosome 7 (Chr 7; peak, 105 Mb), that is significantly associated with CCT. To independently test the biological significance of these results, (C57BLKS/J x NZB/B1NJ) F(2) mice were generated and analyzed for associations with Chr 7. This experiment identified a significant association at 131 Mb. Furthermore, low-generation congenic mice in which the Chr 7 QTL interval from the SJL strain was transferred onto the KS background had CCT values significantly higher than inbred KS mice. These results demonstrate that the genetic dependence of CCT in mice is a multigenic trait, which in these contexts is significantly regulated by a region on Chr 7. Future identification of the genes for these QTL will provide improved understanding of the processes regulating CCT and the pathophysiology of glaucoma.
Collapse
Affiliation(s)
- Geoffrey D Lively
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
33
|
Wisard J, Chrenek MA, Wright C, Dalal N, Pardue MT, Boatright JH, Nickerson JM. Non-contact measurement of linear external dimensions of the mouse eye. J Neurosci Methods 2010; 187:156-66. [PMID: 20067806 PMCID: PMC2832717 DOI: 10.1016/j.jneumeth.2010.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 01/04/2010] [Accepted: 01/05/2010] [Indexed: 11/25/2022]
Abstract
Biometric analyses of quantitative traits in eyes of mice can reveal abnormalities related to refractive or ocular development. Due to the small size of the mouse eye, highly accurate and precise measurements are needed to detect meaningful differences. We sought a non-contact measuring technique to obtain highly accurate and precise linear dimensions of the mouse eye. Laser micrometry was validated with gauge block standards. Simple procedures to measure eye dimensions on three axes were devised. Mouse eyes from C57BL/6J and rd10 on a C57BL/6J background were dissected and extraocular muscle and fat removed. External eye dimensions of axial length (anterior-posterior (A-P) axis) and equatorial diameter (superior-inferior (S-I) and nasal-temporal (N-T) axes) were obtained with a laser micrometer. Several approaches to prevent or ameliorate evaporation due to room air were employed. The resolution of the laser micrometer was less than 0.77 microm, and it provided accurate and precise non-contact measurements of eye dimensions on three axes. External dimensions of the eye strongly correlated with eye weight. The N-T and S-I dimensions of the eye correlated with each other most closely from among the 28 pair-wise combinations of the several parameters that were collected. The equatorial axis measurements correlated well from the right and left eye of each mouse. The A-P measurements did not correlate or correlated poorly in each pair of eyes. The instrument is well suited for the measurement of enucleated eyes and other structures from most commonly used species in experimental vision research and ophthalmology.
Collapse
Affiliation(s)
- Jeffrey Wisard
- Department of Ophthalmology, Emory University, Atlanta, GA
| | | | - Charles Wright
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - Nupur Dalal
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - Machelle T. Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA
- Rehabilitation Research and Development Center of Excellence, Atlanta VA Hospital, Decatur, GA
| | | | | |
Collapse
|
34
|
Lively GD, Jiang B, Hedberg-Buenz A, Chang B, Petersen GE, Wang K, Kuehn MH, Anderson MG. Genetic dependence of central corneal thickness among inbred strains of mice. Invest Ophthalmol Vis Sci 2009; 51:160-71. [PMID: 19710407 DOI: 10.1167/iovs.09-3429] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Central corneal thickness (CCT) exhibits broad variability. For unknown reasons, CCT also associates with diseases not typically considered corneal, particularly glaucoma. The purpose of this study was to test the strain dependence of CCT variability among inbred mice and identify cellular and molecular factors associated with differing CCT. METHODS Methodology for measuring murine CCT with ultrasound pachymetry was developed and used to measure CCT among 17 strains of mice. Corneas from three strains with nonoverlapping differences in CCT (C57BLKS/J, C57BL/6J, and SJL/J) were compared by histology, transmission electron microscopy, and expression profiling with gene microarrays. RESULTS CCT in mice was highly strain dependent. CCT exhibited continuous variation from 89.2 microm in C57BLKS/J to 123.8 microm in SJL/J. Stromal thickness was the major determinant of the varying murine CCT, with epithelial thickness also contributing. Corneal expression levels of many genes differed between strains with differing CCT, but most of these changes did not correlate with the changes observed in previously studied corneal diseases nor did they correlate with genes encoding major structural proteins of the cornea. CONCLUSIONS Murine CCT has been measured with a variety of different techniques, but only among a limited number of different strains. Here, pachymetry was established as an additional tool and used to conduct a broad survey of different strains of inbred mice. These results demonstrated that murine CCT was highly influenced by genetic background and established a baseline for future genetic approaches to further elucidate mechanisms regulating CCT and its disease associations.
Collapse
Affiliation(s)
- Geoffrey D Lively
- Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, Iowa, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Tkatchenko TV, Shen Y, Tkatchenko AV. Analysis of postnatal eye development in the mouse with high-resolution small animal magnetic resonance imaging. Invest Ophthalmol Vis Sci 2009; 51:21-7. [PMID: 19661239 DOI: 10.1167/iovs.08-2767] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Studies of myopia in mice have been complicated by the difficulty in obtaining accurate measurements of small changes observed in the growing mouse eye in vivo and the lack of data on refractive eye development. The purpose of this study was to carry out an in vivo high-resolution analysis of mouse eye growth and refractive development. METHODS High-resolution small animal magnetic resonance imaging and high-resolution infrared photorefraction were used to analyze refractive development in postnatal day (P)21 to P89 C57BL/6J mice. RESULTS The growth of the mouse eye decelerated after P40. The eye maintained a slightly prolate shape during growth. The anterior chamber growth exhibited a similar pattern, whereas the corneal radius of curvature (CRC) increased linearly. The growth rate of the lens remained constant until P89. The lens "overgrew" the eye at P40, resulting in a decline in vitreous chamber depth. Mice showed myopic refractive errors at a younger age (-13.2 +/- 2.0 D; mean +/- SD, P21). The refractive errors stabilized around emmetropic values by P32 and remained emmetropic until P40. Mice became progressively hyperopic with age (+1.2 +/- 1.7 D, P67; +3.6 +/- 2.3 D, P89). CONCLUSIONS Development of ocular components in the mouse is similar to that of the tree shrew but different from that of higher primates and humans. Primary differences can be attributed to the age-related changes of the crystalline lens and CRC. In spite of these differences, mice appear to be able to achieve and maintain emmetropic refractive status at P32 to P40.
Collapse
Affiliation(s)
- Tatiana V Tkatchenko
- Department of Anatomy and Cell Biology, Wayne State University, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
36
|
Hewitt AW, Kearns LS, Jamieson RV, Williamson KA, van Heyningen V, Mackey DA. PAX6Mutations May Be Associated with High Myopia. Ophthalmic Genet 2009; 28:179-82. [PMID: 17896318 DOI: 10.1080/13816810701356676] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PAX6 is a key regulator of eye development and there are many well recognized ophthalmic sequelae of mutations at this locus. The 14 exon PAX6 gene is well conserved across species and phyla. Coding region mutations manifest in a variety of phenotypes. Predicted premature protein truncations are generally associated with classical aniridia. Missense mutations are often found in cases with variant phenotypes such as ectopia pupillae; isolated foveal hypoplasia; nystagmus and hyaloid vessel proliferation. The locus has also been implicated, through a genome-wide sib-pair scan, to be important in the normal variation of myopia. We investigated the association between identified PAX6 mutations and refractive error in Australian patients from four pedigrees. Two of eight subjects with a 1410delC PAX6 mutation had a mean spherical equivalence < -9D, whilst a mean spherical equivalence < or = -5D was recorded in two from four subjects with an Arg240Stop PAX6 mutation and one of two subjects with a Glu93Stop mutation. One individual identified with a Pro346Ala PAX6 mutation had a mean spherical equivalence of +2.8 D. Thus, our observations generally support other incidental findings, that PAX6 mutation, particularly predicted haploinsufficiency, may be associated with extreme refractive error, although the mechanism by which this occurs is not clear.
Collapse
Affiliation(s)
- Alex W Hewitt
- Department of Ophthalmology, Flinders University, Adelaide, Australia
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
PURPOSE Pupillometry is used as a phenotyping assay for investigating pupil responses in normal and genetically engineered mice. The mice used most often in contemporary ophthalmic research have not been adequately investigated by pupillometry. METHODS An infrared-video camera was used to assess pupillary functioning under standard dark-adapted conditions (30 min acclimatization in a ganzfeld chamber) and 30 min after the installation of either 10 microl tropicamide (1.0%) or 10 microl pilocarpine (5.0%) into the conjunctival cul-de-sac. The experiment was self-controlled using a repeated-measures analysis of variance to analyze 60 mice (30 males, 24 weeks of age) from three strains (C57BL/6, 129SvJ, and F1 hybrid). RESULTS The dark-adapted pupillary diameter of mice ranged from 2.3 mm (SD = 0.14) in female C57BL/6 to 2.9 mm (SD = 0.05) in male 129SvJ. Under dark-adapted conditions, all mice examined were sexual dimorphic (F = 19.5, dF = 2, 119, p < 0.001) and strain-dependent differences were observed between male C57BL/6-129SvJ and C57BL/6-F1 and female C57BL/6-F1 (F = 82.32, dF = 1, 119, p < 0.001). The mean pupillary diameter 30 min after the application of tropicamide ranged from 2.5 mm (SD = 0.16) in female F1 to 3.0 mm (SD = 0.13) in male C57BL/6. Tropicamide produced a sexual dimorphism in mydriasis for all mice (F = 56.30, dF = 1, 59, p < 0.001); however, strain-dependent differences were not observed (F = 1.31, dF = 2, 59, p < 0.280). The mean pupillary diameter 30 min after the application of pilocarpine ranged from 2.6 mm (SD = 0.28) in female C57BL/6 to 3.2 mm in both male F1 (SD = 0.22) and 129SvJ (SD = 0.04). Pilocarpine produced a sexual dimorphism in mydriasis for 129SvJ and F1 (F = 106.70, dF = 1, 59, p < 0.001) and strain-dependent differences were observed between female C57BL/6-F1 (F = 17.25, dF = 2, 59, p < 0.001). CONCLUSIONS The experiment demonstrates that mice respond idiosyncratically in their pupillary response under standard dark-adapted conditions and to either tropicamide or pilocarpine depending on strain and/or sex of the mouse. The characteristic responses observed are likely due to subtle differences in the genetic expression of phenotype.
Collapse
|
38
|
Gailus-Durner V, Fuchs H, Adler T, Aguilar Pimentel A, Becker L, Bolle I, Calzada-Wack J, Dalke C, Ehrhardt N, Ferwagner B, Hans W, Hölter SM, Hölzlwimmer G, Horsch M, Javaheri A, Kallnik M, Kling E, Lengger C, Mörth C, Mossbrugger I, Naton B, Prehn C, Puk O, Rathkolb B, Rozman J, Schrewe A, Thiele F, Adamski J, Aigner B, Behrendt H, Busch DH, Favor J, Graw J, Heldmaier G, Ivandic B, Katus H, Klingenspor M, Klopstock T, Kremmer E, Ollert M, Quintanilla-Martinez L, Schulz H, Wolf E, Wurst W, de Angelis MH. Systemic first-line phenotyping. Methods Mol Biol 2009; 530:463-509. [PMID: 19266331 DOI: 10.1007/978-1-59745-471-1_25] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
With the completion of the mouse genome sequence an essential task for biomedical sciences in the twenty-first century will be the generation and functional analysis of mouse models for every gene in the mammalian genome. More than 30,000 mutations in ES cells will be engineered and thousands of mouse disease models will become available over the coming years by the collaborative effort of the International Mouse Knockout Consortium. In order to realize the full value of the mouse models proper characterization, archiving and dissemination of mouse disease models to the research community have to be performed. Phenotyping centers (mouse clinics) provide the necessary capacity, broad expertise, equipment, and infrastructure to carry out large-scale systemic first-line phenotyping. Using the example of the German Mouse Clinic (GMC) we will introduce the reader to the different aspects of the organization of a mouse clinic and present selected methods used in first-line phenotyping.
Collapse
|
39
|
Söker T, Dalke C, Puk O, Floss T, Becker L, Bolle I, Favor J, Hans W, Hölter SM, Horsch M, Kallnik M, Kling E, Moerth C, Schrewe A, Stigloher C, Topp S, Gailus-Durner V, Naton B, Beckers J, Fuchs H, Ivandic B, Klopstock T, Schulz H, Wolf E, Wurst W, Bally-Cuif L, de Angelis MH, Graw J. Pleiotropic effects in Eya3 knockout mice. BMC DEVELOPMENTAL BIOLOGY 2008; 8:118. [PMID: 19102749 PMCID: PMC2653502 DOI: 10.1186/1471-213x-8-118] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 12/22/2008] [Indexed: 01/29/2023]
Abstract
BACKGROUND In Drosophila, mutations in the gene eyes absent (eya) lead to severe defects in eye development. The functions of its mammalian orthologs Eya1-4 are only partially understood and no mouse model exists for Eya3. Therefore, we characterized the phenotype of a new Eya3 knockout mouse mutant. RESULTS Expression analysis of Eya3 by in-situ hybridizations and beta-Gal-staining of Eya3 mutant mice revealed abundant expression of the gene throughout development, e.g. in brain, eyes, heart, somites and limbs suggesting pleiotropic effects of the mutated gene. A similar complex expression pattern was observed also in zebrafish embryos. The phenotype of young adult Eya3 mouse mutants was systematically analyzed within the German Mouse Clinic. There was no obvious defect in the eyes, ears and kidneys of Eya3 mutant mice. Homozygous mutants displayed decreased bone mineral content and shorter body length. In the lung, the tidal volume at rest was decreased, and electrocardiography showed increased JT- and PQ intervals as well as decreased QRS amplitude. Behavioral analysis of the mutants demonstrated a mild increase in exploratory behavior, but decreased locomotor activity and reduced muscle strength. Analysis of differential gene expression revealed 110 regulated genes in heart and brain. Using real-time PCR, we confirmed Nup155 being down regulated in both organs. CONCLUSION The loss of Eya3 in the mouse has no apparent effect on eye development. The wide-spread expression of Eya3 in mouse and zebrafish embryos is in contrast to the restricted expression pattern in Xenopus embryos. The loss of Eya3 in mice leads to a broad spectrum of minor physiological changes. Among them, the mutant mice move less than the wild-type mice and, together with the effects on respiratory, muscle and heart function, the mutation might lead to more severe effects when the mice become older. Therefore, future investigations of Eya3 function should focus on aging mice.
Collapse
Affiliation(s)
- Torben Söker
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pardue MT, Faulkner AE, Fernandes A, Yin H, Schaeffel F, Williams RW, Pozdeyev N, Iuvone PM. High susceptibility to experimental myopia in a mouse model with a retinal on pathway defect. Invest Ophthalmol Vis Sci 2008; 49:706-12. [PMID: 18235018 DOI: 10.1167/iovs.07-0643] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Nob mice share the same mutation in the Nyx gene that is found in humans with complete congenital stationary night blindness (CSNB1). Nob mutant mice were studied to determine whether this defect resulted in myopia, as it does in humans. METHODS Refractive development was measured in unmanipulated wild-type C57BL/6J (WT) and nob mice from 4 to 12 weeks of age by using an infrared photorefractor. The right eye was form deprived by means of a skull-mounted goggling apparatus at 4 weeks of age. Refractive errors were recorded every 2 weeks after goggling. The content of dopamine and the dopamine metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) were measured by HPLC with electrochemical detection (HPLC-ECD) in retinas of nob and WT mice under light- and dark-adapted conditions. RESULTS The nob mice had greater hyperopic refractive errors than did the WT mice under normal visual conditions, until 12 weeks of age when both strains had similar refractions. At 6 weeks of age, refractions became less hyperopic in the nob mice but continued to become more hyperopic in the WT mice. After 2 weeks of form deprivation (6 weeks of age), the nob mice displayed a significant myopic shift (~4 D) in refractive error relative to the opposite and control eyes, whereas WT mice required 6 weeks of goggling to elicit a similar response. As expected with loss of ON pathway transmission, light exposure did not alter DOPAC levels in the nob mice. However, dopamine and DOPAC levels were significantly lower in the nob mice compared with WT. CONCLUSIONS Under normal laboratory visual conditions, only minor differences in refractive development were observed between the nob and WT mice. The largest myopic shift in the nob mice resulted after form deprivation, suggesting that visual pathways dependent on nyctalopin and/or abnormally low dopaminergic activity play a role in regulating refractive development. These findings demonstrate an interaction of genetics and environment in refractive development.
Collapse
Affiliation(s)
- Machelle T Pardue
- Rehabilitation Research and Development Center, Atlanta VA Medical Center, Decatur, GA 30033, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Qiao-Grider Y, Hung LF, Kee CS, Ramamirtham R, Smith EL. A comparison of refractive development between two subspecies of infant rhesus monkeys (Macaca mulatta). Vision Res 2007; 47:1668-81. [PMID: 17442365 PMCID: PMC1950249 DOI: 10.1016/j.visres.2007.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 03/06/2007] [Accepted: 03/09/2007] [Indexed: 12/22/2022]
Abstract
PURPOSE Different subspecies of rhesus monkeys (Macaca mulatta) that are derived from different geographical locations, primarily Indian and China, are commonly employed in vision research. Substantial morphological and behavioral differences have been reported between Chinese- and Indian-derived subspecies. The purpose of this study was to compare refractive development in Chinese- and Indian-derived rhesus monkeys. METHODS The subjects were 216 Indian-derived and 78 Chinese-derived normal infant rhesus monkeys. Cross-sectional data were obtained at 3 weeks of age for all subjects. In addition, longitudinal data were obtained from 10 Indian-derived (male=5, female=5) and 5 Chinese-derived monkeys (male=3, female=2) that were reared with unrestricted vision. Ocular and refractive development was assessed by retinoscopy, keratometry, video-based ophthalmophakometry, and A-scan ultrasonography. RESULTS Although the course of emmetropization was very similar in these two groups of rhesus monkeys, there were consistent and significant inter-group differences in ocular dimensions and refractive error. Throughout the observation period, the Chinese-derived monkeys were on average about 0.4D less hyperopic than the Indian-derived monkeys and the Chinese-derived monkeys had longer overall axial lengths, deeper anterior and vitreous chamber depths, thicker crystalline lenses, flatter corneas and lower powered crystalline lenses. CONCLUSIONS The ocular differences observed in this study presumably reflect genetic differences between subspecies but could reflect the differences in the genetic pool between isolated colonies rather than true subspecies differences. Nonetheless, the substantial ocular differences that we observed emphasize that caution must be exercised when comparing and/or pooling data from rhesus monkeys obtained from different colonies. These inter-subspecies differences might be analogous to the ethnic differences in ocular parameters that have been observed in humans.
Collapse
Affiliation(s)
- Ying Qiao-Grider
- College of Optometry, University of Houston, Houston, TX 77204-2020
- The Vision CRC, The University of New South Wales, Sydney NSW 2052, Australia
| | - Li-Fang Hung
- College of Optometry, University of Houston, Houston, TX 77204-2020
- The Vision CRC, The University of New South Wales, Sydney NSW 2052, Australia
| | - Chea-su Kee
- Department of Optometry & Radiography, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Ramkumar Ramamirtham
- College of Optometry, University of Houston, Houston, TX 77204-2020
- The Vision CRC, The University of New South Wales, Sydney NSW 2052, Australia
| | - Earl L. Smith
- College of Optometry, University of Houston, Houston, TX 77204-2020
- The Vision CRC, The University of New South Wales, Sydney NSW 2052, Australia
| |
Collapse
|