1
|
Meyer KJ, Mercer HE, Roos BR, Fingert JH, Anderson MG. Minimal phenotypes in transgenic mice with the human LOXL1/LOXL1-AS1 locus associated with exfoliation glaucoma. Vision Res 2024; 223:108464. [PMID: 39151208 PMCID: PMC11381136 DOI: 10.1016/j.visres.2024.108464] [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: 05/17/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/18/2024]
Abstract
Exfoliation syndrome is a leading cause of secondary glaucoma worldwide. Among the risk-factors for exfoliation syndrome and exfoliation glaucoma that have been investigated, a genetic association with 15q24.1 is among the most striking. The leading candidates for the causal gene at this locus are LOXL1 and/or LOXL1-AS1, but studies have not yet coalesced in establishing, or ruling out, either candidate. Here, we contribute to studies of the 15q24.1 locus by making a partially humanized mouse model in which 166 kb of human genomic DNA from the 15q24.1 locus was introduced into the mouse genome via BAC transgenesis (B6-Tg(RP11-71M11)Andm). Transgenic expression of human genes in the BAC was only detectable for LOXL1-AS1. One cohort of 34 mice (21 experimental hemizygotes and 13 non-carrier control littermates) was assessed by slit-lamp exams and SD-OCT imaging at early (1-2 months) and mid (4-5 months) time points; fundus exams were performed at 5 months of age. A second smaller cohort (3 hemizygotes) were aged extensively (>12 months) to screen for overt abnormalities. Across all genotypes and ages, 136 slit-lamp exams, 128 SD-OCT exams, and 42 fundus exams detected no overt indices of exfoliation syndrome. Quantitatively, small, but statistically significant, age-related declines in ganglion cell complex thickness and total retinal thickness were detected in the hemizygotes at 4 months of age. Overall, this study demonstrates complexity in gene regulation from the 15q24.1 locus and suggests that LOXL1-AS1 is unlikely to be a monogenic cause of exfoliation syndrome but may contribute to glaucomatous retinal damage.
Collapse
Affiliation(s)
- Kacie J Meyer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States; Institute for Vision Research, University of Iowa, Iowa City, IA, United States
| | - Hannah E Mercer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States; Institute for Vision Research, University of Iowa, Iowa City, IA, United States
| | - Ben R Roos
- Institute for Vision Research, University of Iowa, Iowa City, IA, United States; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - John H Fingert
- Institute for Vision Research, University of Iowa, Iowa City, IA, United States; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Michael G Anderson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States; Institute for Vision Research, University of Iowa, Iowa City, IA, United States; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States; Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, United States.
| |
Collapse
|
2
|
Geiduschek EK, Bricco EK, McDowell CM. DAMPs Drive Fibroinflammatory Changes in the Glaucomatous ONH. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 39382882 PMCID: PMC11469284 DOI: 10.1167/iovs.65.12.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose The optic nerve head (ONH) is well known to be the initial site of glaucomatous damage; however, the molecular mechanisms initiating this pathology are not fully understood. To further understand the initiating factors in glaucomatous damage we utilized a novel mouse model of glaucoma, B6.EDA+/+ mice, which constitutively express fibronectin containing the extra domain A (FN+EDA). FN+EDA is a known damage-associated molecular pattern (DAMP) that activates Toll-like receptor 4 and elicits a fibro-inflammatory response. Methods Eyes from B6.EDA+/+ and C57BL/6J mice were evaluated for retinal ganglion cell (RGC) death, retinal nerve fiber layer (RNFL) thickness, and optic nerve (ON) damage at 12 months and 22 months of age. ONH sections were isolated using laser capture microdissection for subsequent RNA-sequencing and Gene Set Enrichment Analysis (GSEA). GSEA results were confirmed using immunohistochemical (IHC) staining. Results B6.EDA+/+ mice exhibit significantly higher intraocular pressure, loss of RGCs, thinning of the RNFL, and progressive levels of ON damage at 12 months and 22 months of age compared to C57BL/6J controls. Protein expression of DAMPs FN+EDA and biglycan was significantly increased in B6.EDA+/+ mice compared to C57BL/6J controls. GSEA analysis identified significantly up- and downregulated gene groupings at both 12 months and 22 months of age, and IHC staining at 12 and 18 months of age demonstrated significant increases of IFNα, IFNβ, and pSTAT1 expression in B6.EDA+/+ mice compared to C57BL/6J controls. Conclusions Our study characterizes glaucomatous changes to the retina, ON, and ONH over the course of 2 years and identifies novel molecular pathways associated with these pathophysiological changes. These data illustrate the effects of FN+EDA on the fibro-inflammatory response in the aging ONH in a novel mouse model of glaucoma.
Collapse
Affiliation(s)
| | - Emma K. Bricco
- University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | |
Collapse
|
3
|
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
|
4
|
Faralli JA, Filla MS, Yang YF, Sun YY, Johns K, Keller KE, Peters DM. Digital spatial profiling of segmental outflow regions in trabecular meshwork reveals a role for ADAM15. PLoS One 2024; 19:e0298802. [PMID: 38394161 PMCID: PMC10889904 DOI: 10.1371/journal.pone.0298802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
In this study we used a spatial transcriptomics approach to identify genes specifically associated with either high or low outflow regions in the trabecular meshwork (TM) that could potentially affect aqueous humor outflow in vivo. High and low outflow regions were identified and isolated from organ cultured human anterior segments perfused with fluorescently-labeled 200 nm FluoSpheres. The NanoString GeoMx Digital Spatial Profiler (DSP) platform was then used to identified genes in the paraffin embedded tissue sections from within those regions. These transcriptome analyses revealed that 16 genes were statistically upregulated in high outflow regions and 57 genes were statistically downregulated in high outflow regions when compared to low outflow regions. Gene ontology enrichment analysis indicated that the top three biological categories of these differentially expressed genes were ECM/cell adhesion, signal transduction, and transcription. The ECM/cell adhesion genes that showed the largest differential expression (Log2FC ±1.5) were ADAM15, BGN, LDB3, and CRKL. ADAM15, which is a metalloproteinase that can bind integrins, was upregulated in high outflow regions, while the proteoglycan BGN and two genes associated with integrin signaling (LDB3, and CRKL) were downregulated. Immunolabeling studies supported the differential expression of ADAM15 and showed that it was specifically upregulated in high outflow regions along the inner wall of Schlemm's canal and in the juxtacanalicular (JCT) region of the TM. In addition to these genes, the studies showed that genes for decorin, a small leucine-rich proteoglycan, and the α8 integrin subunit were enriched in high outflow regions. These studies identify several novel genes that could be involved in segmental outflow, thus demonstrating that digital spatial profiling could be a useful approach for understanding segmental flow through the TM. Furthermore, this study suggests that changes in the expression of genes involved in regulating the activity and/or organization of the ECM and integrins in the TM are likely to be key players in segmental outflow.
Collapse
Affiliation(s)
- Jennifer A. Faralli
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Mark S. Filla
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kassidy Johns
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Donna M. Peters
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Ophthalmology & Visual Sciences, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
5
|
Loo Y, Chan ASY, Khor CC, Aung T, Wang Z. Rodent genetically modified models of glaucoma. Mol Aspects Med 2024; 95:101229. [PMID: 38039744 DOI: 10.1016/j.mam.2023.101229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Glaucoma, one of the leading causes of irreversible blindness worldwide, is a complex and heterogenous disease. While environmental factors are important, it is well-recognized that the disease has a strong heritable component. With the advent of large-cohort genome wide association studies, a myriad of genetic risk loci has been linked to different forms of glaucoma. Animal models have been an indispensable tool in characterizing these loci, especially if they lie within coding regions in the genome. Not only do these models connect genotype to phenotype, advancing our understanding of glaucoma pathogenesis in the process, they also have valuable utility as a platform for the pre-clinical testing of potential therapies. In this review, we will outline genetic models used for studying the major forms of glaucoma, including primary open angle glaucoma, normal tension glaucoma, primary angle closure glaucoma, pigmentary glaucoma, pseudoexfoliation glaucoma, and early onset glaucoma, including congenital and developmental glaucoma, and how studying these models have helped shed light on human glaucoma.
Collapse
Affiliation(s)
- Yunhua Loo
- Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Anita Sook Yee Chan
- Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Chiea Chuen Khor
- Duke-NUS Medical School, Singapore; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Tin Aung
- Duke-NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Zhenxun Wang
- Duke-NUS Medical School, Singapore; Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.
| |
Collapse
|
6
|
Peng MG, Lee J, Ho W, Kim T, Yao P, Medvidovic S, Alas B, Wu V, Runner MM, Gokoffski KK. AxonQuantifier: A semi-automated program for quantifying axonal density from whole-mounted optic nerves. J Neurosci Methods 2023; 394:109895. [PMID: 37315846 PMCID: PMC10330882 DOI: 10.1016/j.jneumeth.2023.109895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Here, we present a semi-automated method for quantifying retinal ganglion cell (RGC) axon density at different distances from the optic nerve crush site using longitudinal, confocal microscopy images taken from whole-mounted optic nerves. This method employs the algorithm AxonQuantifier which operates on the freely available program, ImageJ. NEW METHOD To validate this method, seven adult male Long Evans rats underwent optic nerve crush injury followed by in vivo treatment with electric fields of varying strengths for 30 days to produce optic nerves with a wide range of axon densities distal to the optic nerve crush site. Prior to euthanasia, RGC axons were labelled with intravitreal injections of cholera toxin B conjugated to Alexa Fluor 647. After dissection, optic nerves underwent tissue clearing, were whole-mounted, and imaged longitudinally using confocal microscopy. COMPARISON WITH EXISTING METHODS Five masked raters quantified RGC axon density at 250, 500, 750, 1000, 1250, 1500, 1750, and 2000 µm distances past the optic nerve crush site for the seven optic nerves manually and using AxonQuantifier. Agreement between these methods was assessed using Bland-Altman plots and linear regression. Inter-rater agreement was assessed using the intra-class coefficient. RESULTS Semi-automated quantification of RGC axon density demonstrated improved inter-rater agreement and reduced bias values as compared to manual quantification, while also increasing time efficiency 4-fold. Relative to manual quantification, AxonQuantifier tended to underestimate axon density. CONCLUSIONS AxonQuantifier is a reliable and efficient method for quantifying axon density from whole mount optic nerves.
Collapse
Affiliation(s)
- Micalla G Peng
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jonathan Lee
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Wilson Ho
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Timothy Kim
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Petcy Yao
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Sasha Medvidovic
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Basheer Alas
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Vivian Wu
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Margaret M Runner
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA; Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Associated Retinal Consultants, PC, Department of Ophthalmology, Oakland University William Beaumont School of Medicine, Royal Oak, MI, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Keck School of Medicine, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Hedberg-Buenz A, Meyer KJ, van der Heide CJ, Deng W, Lee K, Soukup DA, Kettelson M, Pellack D, Mercer H, Wang K, Garvin MK, Abramoff MD, Anderson MG. Biological Correlations and Confounders for Quantification of Retinal Ganglion Cells by Optical Coherence Tomography Based on Studies of Outbred Mice. Transl Vis Sci Technol 2022; 11:17. [PMID: 36135979 PMCID: PMC9513741 DOI: 10.1167/tvst.11.9.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/02/2022] [Indexed: 01/28/2023] Open
Abstract
Purpose Despite popularity of optical coherence tomography (OCT) in glaucoma studies, it's unclear how well OCT-derived metrics compare to traditional measures of retinal ganglion cell (RGC) abundance. Here, Diversity Outbred (J:DO) mice are used to directly compare ganglion cell complex (GCC) thickness measured by OCT to metrics of retinal anatomy measured ex vivo with retinal wholemounts and optic nerve histology. Methods J:DO mice (n = 48) underwent fundoscopic and OCT examinations, with automated segmentation of GCC thickness. RGC axons were quantified from para-phenylenediamine-stained optic nerve cross-sections and somas from BRN3A-immunolabeled retinal wholemounts, with total inner retinal cellularity assessed by TO-PRO and subsequent hematoxylin staining. Results J:DO tissues lacked overt disease. GCC thickness, RGC abundance, and total cell abundance varied broadly across individuals. GCC thickness correlated significantly to RGC somal density (r = 0.58) and axon number (r = 0.44), but not total cell density. Retinal area and nerve cross-sectional area varied widely. No metrics were significantly influenced by sex. In bilateral comparisons, GCC thickness (r = 0.95), axon (r = 0.72), and total cell density (r = 0.47) correlated significantly within individuals. Conclusions Amongst outbred mice, OCT-derived measurements of GCC thickness correlate significantly to RGC somal and axon abundance. Factors limiting correlation are likely both biological and methodological, including differences in retinal area that distort sampling-based estimates of RGC abundance. Translational Relevance There are significant-but imperfect-correlations between GCC thickness and RGC abundance across genetic contexts in mice, highlighting valid uses and ongoing challenges for meaningful use of OCT-derived metrics.
Collapse
Affiliation(s)
- Adam Hedberg-Buenz
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Kacie J. Meyer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Carly J. van der Heide
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Wenxiang Deng
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Kyungmoo Lee
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Dana A. Soukup
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Monica Kettelson
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Danielle Pellack
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Hannah Mercer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Mona K. Garvin
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Michael D. Abramoff
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Michael G. Anderson
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Furundaoturan O, Esen Baris M, Ayyıldız Emecen D, Demirkilinc Biler E. Retinal detachment in a child with Frank-ter Haar syndrome. Ophthalmic Genet 2022; 44:291-294. [PMID: 36004685 DOI: 10.1080/13816810.2022.2113543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
BACKGROUND To present a rare case of ocular involvement in a child with Frank-ter Haar syndrome (FTHS) presenting retinal detachment. MATERIALS AND METHODS Detailed ophthalmological evaluation including examination under general anesthesia, ocular ultrasound, and visual evoked potential testing was completed. Photographic documentation of the physical findings was obtained. RESULTS A 3-year-old female patient with FTHS was referred to evaluate for possible ophthalmic involvement. The patient presented with the classical dysmorphic abnormalities of the syndrome. Ophthalmologic evaluation revealed a high, against-the-rule corneal astigmatism in the right eye. In the left eye, the red reflex was absent with a suspicious membrane behind the lens, and a sensory exotropia was present. Ultrasonography confirmed retinal detachment with no history of previous trauma. Due to poor visual evoked potentials, no surgery was planned. Astigmatic refractive error was corrected with routine follow-up. CONCLUSIONS FTHS is associated with multiple ocular involvement such as megalocornea, congenital glaucoma, or colobomas. This case report is the first to describe a high, against-the-rule astigmatism and retinal detachment in a female child with FTHS and demonstrates that an early and detailed ophthalmological examination is essential for these patients.
Collapse
Affiliation(s)
- Onur Furundaoturan
- Department of Ophthalmology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Mine Esen Baris
- Department of Ophthalmology, Ege University Faculty of Medicine, Izmir, Turkey
| | | | | |
Collapse
|
9
|
Yoon B, Yeung P, Santistevan N, Bluhm LE, Kawasaki K, Kueper J, Dubielzig R, VanOudenhove J, Cotney J, Liao EC, Grinblat Y. Zebrafish models of alx-linked frontonasal dysplasia reveal a role for Alx1 and Alx3 in the anterior segment and vasculature of the developing eye. Biol Open 2022; 11:bio059189. [PMID: 35142342 PMCID: PMC9167625 DOI: 10.1242/bio.059189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/28/2022] [Indexed: 11/18/2022] Open
Abstract
The cellular and genetic mechanisms that coordinate formation of facial sensory structures with surrounding skeletal and soft tissue elements remain poorly understood. Alx1, a homeobox transcription factor, is a key regulator of midfacial morphogenesis. ALX1 mutations in humans are linked to severe congenital anomalies of the facial skeleton (frontonasal dysplasia, FND) with malformation or absence of eyes and orbital contents (micro- and anophthalmia). Zebrafish with loss-of-function alx1 mutations develop with craniofacial and ocular defects of variable penetrance, likely due to compensatory upregulation in expression of a paralogous gene, alx3. Here we show that zebrafish alx1;alx3 mutants develop with highly penetrant cranial and ocular defects that resemble human ALX1-linked FND. alx1 and alx3 are expressed in anterior cranial neural crest (aCNC), which gives rise to the anterior neurocranium (ANC), anterior segment structures of the eye and vascular pericytes. Consistent with a functional requirement for alx genes in aCNC, alx1; alx3 mutants develop with nearly absent ANC and grossly aberrant hyaloid vasculature and ocular anterior segment, but normal retina. In vivo lineage labeling identified a requirement for alx1 and alx3 during aCNC migration, and transcriptomic analysis suggested oxidative stress response as a key target mechanism of this function. Oxidative stress is a hallmark of fetal alcohol toxicity, and we found increased penetrance of facial and ocular malformations in alx1 mutants exposed to ethanol, consistent with a protective role for alx1 against ethanol toxicity. Collectively, these data demonstrate a conserved role for zebrafish alx genes in controlling ocular and facial development, and a novel role in protecting these key midfacial structures from ethanol toxicity during embryogenesis. These data also reveal novel roles for alx genes in ocular anterior segment formation and vascular development and suggest that retinal deficits in alx mutants may be secondary to aberrant ocular vascularization and anterior segment defects. This study establishes robust zebrafish models for interrogating conserved genetic mechanisms that coordinate facial and ocular development, and for exploring gene--environment interactions relevant to fetal alcohol syndrome.
Collapse
Affiliation(s)
- Baul Yoon
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Pan Yeung
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Nicholas Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren E. Bluhm
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Kenta Kawasaki
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Janina Kueper
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
- Institute of Human Genetics, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Richard Dubielzig
- Comparative Ocular Pathology Laboratory of Wisconsin (COPLOW), University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer VanOudenhove
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Justin Cotney
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
10
|
McDowell CM, Kizhatil K, Elliott MH, Overby DR, van Batenburg-Sherwood J, Millar JC, Kuehn MH, Zode G, Acott TS, Anderson MG, Bhattacharya SK, Bertrand JA, Borras T, Bovenkamp DE, Cheng L, Danias J, De Ieso ML, Du Y, Faralli JA, Fuchshofer R, Ganapathy PS, Gong H, Herberg S, Hernandez H, Humphries P, John SWM, Kaufman PL, Keller KE, Kelley MJ, Kelly RA, Krizaj D, Kumar A, Leonard BC, Lieberman RL, Liton P, Liu Y, Liu KC, Lopez NN, Mao W, Mavlyutov T, McDonnell F, McLellan GJ, Mzyk P, Nartey A, Pasquale LR, Patel GC, Pattabiraman PP, Peters DM, Raghunathan V, Rao PV, Rayana N, Raychaudhuri U, Reina-Torres E, Ren R, Rhee D, Chowdhury UR, Samples JR, Samples EG, Sharif N, Schuman JS, Sheffield VC, Stevenson CH, Soundararajan A, Subramanian P, Sugali CK, Sun Y, Toris CB, Torrejon KY, Vahabikashi A, Vranka JA, Wang T, Willoughby CE, Xin C, Yun H, Zhang HF, Fautsch MP, Tamm ER, Clark AF, Ethier CR, Stamer WD. Consensus Recommendation for Mouse Models of Ocular Hypertension to Study Aqueous Humor Outflow and Its Mechanisms. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 35129590 PMCID: PMC8842499 DOI: 10.1167/iovs.63.2.12] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Due to their similarities in anatomy, physiology, and pharmacology to humans, mice are a valuable model system to study the generation and mechanisms modulating conventional outflow resistance and thus intraocular pressure. In addition, mouse models are critical for understanding the complex nature of conventional outflow homeostasis and dysfunction that results in ocular hypertension. In this review, we describe a set of minimum acceptable standards for developing, characterizing, and utilizing mouse models of open-angle ocular hypertension. We expect that this set of standard practices will increase scientific rigor when using mouse models and will better enable researchers to replicate and build upon previous findings.
Collapse
Affiliation(s)
- Colleen M. McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Michael H. Elliott
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Darryl R. Overby
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - J. Cameron Millar
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences and Institute for Vision Research, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | - Gulab Zode
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Ted S. Acott
- Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Michael G. Anderson
- Department of Molecular Physiology and Biophysics and Department of Ophthalmology and Visual Sciences, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | | | - Jacques A. Bertrand
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Terete Borras
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | | | - Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - John Danias
- SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| | - Michael Lucio De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Jennifer A. Faralli
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | | | - Peter Humphries
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Simon W. M. John
- Department of Ophthalmology, Columbia University, New York, New York, United States
| | - Paul L. Kaufman
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Mary J. Kelley
- Department of Ophthalmology and Department of Integrative Biosciences, Oregon Health & Science University, Portland, Oregon, United States
| | - Ruth A. Kelly
- Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David Krizaj
- Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, University of California, Davis, Davis, California, United States
| | - Raquel L. Lieberman
- Department of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Paloma Liton
- Department of Ophthalmology and Department of Pathology, Duke University, Durham, North Carolina, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, James & Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Katy C. Liu
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Navita N. Lopez
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, United States
| | - Weiming Mao
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Timur Mavlyutov
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Fiona McDonnell
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Gillian J. McLellan
- Department of Surgical Sciences and Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Philip Mzyk
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Andrews Nartey
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Louis R. Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Gaurang C. Patel
- Ophthalmology Research, Regeneron Pharmaceuticals, Tarreytown, New York, United States
| | | | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Naga Rayana
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Urmimala Raychaudhuri
- Department of Neurobiology, University of California, Irvine, Irvine, California, United States
| | - Ester Reina-Torres
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Ruiyi Ren
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Douglas Rhee
- Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Uttio Roy Chowdhury
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - John R. Samples
- Washington State University, Floyd Elson College of Medicine, Spokane, Washington, United States
| | | | - Najam Sharif
- Santen Inc., Emeryville, California, United States
| | - Joel S. Schuman
- Department of Ophthalmology and Department of Physiology and Neuroscience, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States; Departments of Biomedical Engineering and Electrical and Computer Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States; Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States
| | - Val C. Sheffield
- Department of Pediatrics and Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Cooper H. Stevenson
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Avinash Soundararajan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | | | - Chenna Kesavulu Sugali
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Yang Sun
- Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California, United States
| | - Carol B. Toris
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States; Department of Ophthalmology and Vision Sciences, The Ohio State University, Columbus, Ohio, United States
| | | | - Amir Vahabikashi
- Cell and Developmental Biology Department, Northwestern University, Chicago, Illinois, United States
| | - Janice A. Vranka
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ting Wang
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Colin E. Willoughby
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Chen Xin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hao F. Zhang
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | - Michael P. Fautsch
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | | | - Abbot F. Clark
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - C. Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology; Emory University School of Medicine, Emory University, Atlanta, Georgia, United States
| | - W. Daniel Stamer
- Duke Ophthalmology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
11
|
Deng W, Hedberg-Buenz A, Soukup DA, Taghizadeh S, Wang K, Anderson MG, Garvin MK. AxonDeep: Automated Optic Nerve Axon Segmentation in Mice With Deep Learning. Transl Vis Sci Technol 2021; 10:22. [PMID: 34932117 PMCID: PMC8709929 DOI: 10.1167/tvst.10.14.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Optic nerve damage is the principal feature of glaucoma and contributes to vision loss in many diseases. In animal models, nerve health has traditionally been assessed by human experts that grade damage qualitatively or manually quantify axons from sampling limited areas from histologic cross sections of nerve. Both approaches are prone to variability and are time consuming. First-generation automated approaches have begun to emerge, but all have significant shortcomings. Here, we seek improvements through use of deep-learning approaches for segmenting and quantifying axons from cross-sections of mouse optic nerve. Methods Two deep-learning approaches were developed and evaluated: (1) a traditional supervised approach using a fully convolutional network trained with only labeled data and (2) a semisupervised approach trained with both labeled and unlabeled data using a generative-adversarial-network framework. Results From comparisons with an independent test set of images with manually marked axon centers and boundaries, both deep-learning approaches outperformed an existing baseline automated approach and similarly to two independent experts. Performance of the semisupervised approach was superior and implemented into AxonDeep. Conclusions AxonDeep performs automated quantification and segmentation of axons from healthy-appearing nerves and those with mild to moderate degrees of damage, similar to that of experts without the variability and constraints associated with manual performance. Translational Relevance Use of deep learning for axon quantification provides rapid, objective, and higher throughput analysis of optic nerve that would otherwise not be possible.
Collapse
Affiliation(s)
- Wenxiang Deng
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa City, IA, USA.,Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
| | - Adam Hedberg-Buenz
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA.,Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA, USA
| | - Dana A Soukup
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA.,Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA, USA
| | - Sima Taghizadeh
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa City, IA, USA
| | - Kai Wang
- Department of Biostatistics, The University of Iowa, Iowa City, IA, USA
| | - Michael G Anderson
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA.,Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, IA, USA.,Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, USA
| | - Mona K Garvin
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa City, IA, USA.,Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
12
|
Taiyab A, Akula M, Dham J, Deschamps P, Sheardown H, Williams T, Borrás T, West-Mays JA. Deletion of transcription factor AP-2β from the developing murine trabecular meshwork region leads to progressive glaucomatous changes. J Neurosci Res 2021; 100:638-652. [PMID: 34822722 DOI: 10.1002/jnr.24982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 11/08/2022]
Abstract
Glaucoma is one of the leading causes of irreversible blindness and can result from abnormalities in anterior segment structures required for aqueous humor outflow, including the trabecular meshwork (TM) and Schlemm's canal (SC). Transcription factors such as AP-2β play critical roles in anterior segment development. Here, we show that the Mgp-Cre knock-in (Mgp-Cre.KI) mouse can be used to target the embryonic periocular mesenchyme giving rise to the TM and SC. Fate mapping of male and female mice indicates that AP-2β loss causes a decrease in iridocorneal angle cells derived from Mgp-Cre.KI-expressing populations compared to controls. Moreover, histological analyses revealed peripheral iridocorneal adhesions in AP-2β mutants that were accompanied by a decrease in expression of TM and SC markers, as observed using immunohistochemistry. In addition, rebound tonometry showed significantly higher intraocular pressure (IOP) that was correlated with a progressive significant loss of retinal ganglion cells, reduced retinal thickness, and reduced retinal function, as measured using an electroretinogram, in AP-2β mutants compared with controls, reflecting pathology described in late-stage glaucoma patients. Importantly, elevated IOP in AP-2β mutants was significantly reduced by treatment with latanoprost, a prostaglandin analog that increases unconventional outflow. These findings demonstrate that AP-2β is critical for TM and SC development, and that these mutant mice can serve as a model for understanding and treating progressive human primary angle-closure glaucoma.
Collapse
Affiliation(s)
- Aftab Taiyab
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Monica Akula
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Japnit Dham
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Paula Deschamps
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado, Aurora, CO, USA
| | - Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
13
|
van der Heide CJ, Meyer KJ, Hedberg-Buenz A, Pellack D, Pomernackas N, Mercer HE, Anderson MG. Quantification and image-derived phenotyping of retinal ganglion cell nuclei in the nee mouse model of congenital glaucoma. Exp Eye Res 2021; 212:108774. [PMID: 34597676 PMCID: PMC8608716 DOI: 10.1016/j.exer.2021.108774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/31/2022]
Abstract
The nee mouse model exhibits characteristic features of congenital glaucoma, a common cause of childhood blindness. The current study of nee mice had two components. First, the time course of neurodegeneration in nee retinal flat-mounts was studied over time using a retinal ganglion cell (RGC)-marker, BRN3A; a pan-nuclear marker, TO-PRO-3; and H&E staining. Based on segmentation of nuclei using ImageJ and RetFM-J, this analysis identified a rapid loss of BRN3A+ nuclei from 4 to 15 weeks of age, with the first statistically significant difference in average density compared to age-matched controls detected in 8-week-old cohorts (49% reduction in nee). Consistent with a model of glaucoma, no reductions in BRN3A- nuclei were detected, but the combined analysis indicated that some RGCs lost BRN3A marker expression prior to actual cell loss. These results have a practical application in the design of experiments using nee mice to study mechanisms or potential therapies for congenital glaucoma. The second component of the study pertains to a discovery-based analysis of the large amount of image data with 748,782 segmented retinal nuclei. Using the automatedly collected region of interest feature data captured by ImageJ, we tested whether RGC density of glaucomatous mice was significantly correlated to average nuclear area, perimeter, Feret diameter, or MinFeret diameter. These results pointed to two events influencing nuclear size. For variations in RGC density above approximately 3000 nuclei/mm2 apparent spreading was observed, in which BRN3A- nuclei-regardless of genotype-became slightly larger as RGC density decreased. This same spreading occurred in BRN3A+ nuclei of wild-type mice. For variation in RGC density below 3000 nuclei/mm2, which only occurred in glaucomatous nee mutants, BRN3A+ nuclei became smaller as disease was progressively severe. These observations have relevance to defining RGCs of relatively higher sensitivity to glaucomatous cell death and the nuclear dynamics occurring during their demise.
Collapse
Affiliation(s)
- Carly J van der Heide
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA.
| | - Kacie J Meyer
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA.
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA; VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 601 Hwy 6 West (151), Iowa City, IA, 52246, USA.
| | - Danielle Pellack
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA.
| | - Nicholas Pomernackas
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA.
| | - Hannah E Mercer
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA.
| | - Michael G Anderson
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Rd., Iowa City, IA, 52242, USA; VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 601 Hwy 6 West (151), Iowa City, IA, 52246, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Dr., Iowa City, IA, 52242, USA.
| |
Collapse
|
14
|
Garai S, Leo LM, Szczesniak AM, Hurst DP, Schaffer PC, Zagzoog A, Black T, Deschamps JR, Miess E, Schulz S, Janero DR, Straiker A, Pertwee RG, Abood ME, Kelly MEM, Reggio PH, Laprairie RB, Thakur GA. Discovery of a Biased Allosteric Modulator for Cannabinoid 1 Receptor: Preclinical Anti-Glaucoma Efficacy. J Med Chem 2021; 64:8104-8126. [PMID: 33826336 DOI: 10.1021/acs.jmedchem.1c00040] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We apply the magic methyl effect to improve the potency/efficacy of GAT211, the prototypic 2-phenylindole-based cannabinoid type-1 receptor (CB1R) agonist-positive allosteric modulator (ago-PAM). Introducing a methyl group at the α-position of nitro group generated two diastereomers, the greater potency and efficacy of erythro, (±)-9 vs threo, (±)-10 constitutes the first demonstration of diastereoselective CB1R-allosteric modulator interaction. Of the (±)-9 enantiomers, (-)-(S,R)-13 evidenced improved potency over GAT211 as a CB1R ago-PAM, whereas (+)-(R,S)-14 was a CB1R allosteric agonist biased toward G protein- vs β-arrestin1/2-dependent signaling. (-)-(S,R)-13 and (+)-(R,S)-14 were devoid of undesirable side effects (triad test), and (+)-(R,S)-14 reduced intraocular pressure with an unprecedentedly long duration of action in a murine glaucoma model. (-)-(S,R)-13 docked into both a CB1R extracellular PAM and intracellular allosteric-agonist site(s), whereas (+)-(R,S)-14 preferentially engaged only the latter. Exploiting G-protein biased CB1R-allosteric modulation can offer safer therapeutic candidates for glaucoma and, potentially, other diseases.
Collapse
Affiliation(s)
- Sumanta Garai
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Luciana M Leo
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Anna-Maria Szczesniak
- Department of Pharmacology and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Dow P Hurst
- Center for Drug Discovery, University of North Carolina Greensboro, Greensboro, North Carolina 27402, United States
| | - Peter C Schaffer
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ayat Zagzoog
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Pl, Saskatoon, Saskatchewan S7N2Z4, Canada
| | - Tallan Black
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Pl, Saskatoon, Saskatchewan S7N2Z4, Canada
| | - Jeffrey R Deschamps
- Naval Research Laboratory, Code 6930, 4555 Overlook Avenue, Washington, District of Columbia 20375, United States
| | - Elke Miess
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, D-07747 Jena, Germany
| | - David R Janero
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Alex Straiker
- The Gill Center and the Department of Psychological & Brain Sciences, Indiana University, 1101 E. 10th St, Bloomington, Indiana 47405, United States
| | - Roger G Pertwee
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, U.K
| | - Mary E Abood
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Melanie E M Kelly
- Department of Pharmacology and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Patricia H Reggio
- Center for Drug Discovery, University of North Carolina Greensboro, Greensboro, North Carolina 27402, United States
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Pl, Saskatoon, Saskatchewan S7N2Z4, Canada
- Department of Pharmacology and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
15
|
Harper MM, Boehme N, Dutca LM, Anderson MG. The Retinal Ganglion Cell Response to Blast-Mediated Traumatic Brain Injury Is Genetic Background Dependent. Invest Ophthalmol Vis Sci 2021; 62:13. [PMID: 34106210 PMCID: PMC8196410 DOI: 10.1167/iovs.62.7.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to examine the influence of genetic background on the retinal ganglion cell (RGC) response to blast-mediated traumatic brain injury (TBI) in Jackson Diversity Outbred (J:DO), C57BL/6J and BALB/cByJ mice. Methods Mice were subject to one blast injury of 137 kPa. RGC structure was analyzed by optical coherence tomography (OCT), function by the pattern electroretinogram (PERG), and histologically using BRN3A antibody staining. Results Comparison of the change in each group from baseline for OCT and PERG was performed. There was a significant difference in the J:DOΔOCT compared to C57BL/6J mice (P = 0.004), but not compared to BALB/cByJ (P = 0.21). There was a significant difference in the variance of the ΔOCT in J:DO compared to both C57BL/6J and BALB/cByJ mice. The baseline PERG amplitude was 20.33 ± 9.32 µV, which decreased an average of −4.14 ± 12.46 µV following TBI. Baseline RGC complex + RNFL thickness was 70.92 ± 4.52 µm, which decreased an average of −1.43 ± 2.88 µm following blast exposure. There was not a significant difference in the ΔPERG between J:DO and C57BL/6J (P = 0.13), although the variances of the groups were significantly different. Blast exposure in J:DO mice results in a density change of 558.6 ± 440.5 BRN3A-positive RGCs/mm2 (mean ± SD). Conclusions The changes in retinal outcomes had greater variance in outbred mice than what has been reported, and largely replicated herein, for inbred mice. These results demonstrate that the RGC response to blast injury is highly dependent upon genetic background.
Collapse
Affiliation(s)
- Matthew M Harper
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Center for the Prevention and Treatment of Visual Loss, Iowa City VA Healthcare System, Department of Veterans Affairs, Iowa City, IA, United States
| | - Nickolas Boehme
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Center for the Prevention and Treatment of Visual Loss, Iowa City VA Healthcare System, Department of Veterans Affairs, Iowa City, IA, United States
| | - Laura M Dutca
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Center for the Prevention and Treatment of Visual Loss, Iowa City VA Healthcare System, Department of Veterans Affairs, Iowa City, IA, United States
| | - Michael G Anderson
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Center for the Prevention and Treatment of Visual Loss, Iowa City VA Healthcare System, Department of Veterans Affairs, Iowa City, IA, United States.,The Department of Molecular Physiology and Biophysics, Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
16
|
Axonopathy precedes cell death in ocular damage mediated by blast exposure. Sci Rep 2021; 11:11774. [PMID: 34083587 PMCID: PMC8175471 DOI: 10.1038/s41598-021-90412-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injuries (TBI) of varied types are common across all populations and can cause visual problems. For military personnel in combat settings, injuries from blast exposures (bTBI) are prevalent and arise from a myriad of different situations. To model these diverse conditions, we are one of several groups modeling bTBI using mice in varying ways. Here, we report a refined analysis of retinal ganglion cell (RGC) damage in male C57BL/6J mice exposed to a blast-wave in an enclosed chamber. Ganglion cell layer thickness, RGC density (BRN3A and RBPMS immunoreactivity), cellular density of ganglion cell layer (hematoxylin and eosin staining), and axon numbers (paraphenylenediamine staining) were quantified at timepoints ranging from 1 to 17-weeks. RNA sequencing was performed at 1-week and 5-weeks post-injury. Earliest indices of damage, evident by 1-week post-injury, are a loss of RGC marker expression, damage to RGC axons, and increase in glial markers expression. Blast exposure caused a loss of RGC somas and axons—with greatest loss occurring by 5-weeks post-injury. While indices of glial involvement are prominent early, they quickly subside as RGCs are lost. The finding that axonopathy precedes soma loss resembles pathology observed in mouse models of glaucoma, suggesting similar mechanisms.
Collapse
|
17
|
Taiyab A, Saraco A, Akula M, Deschamps P, Ball AK, Williams T, West-Mays JA. Progressive Loss of Retinal Ganglion Cells in Activating Protein-2β Neural Crest Cell Knockout Mice. Curr Eye Res 2021; 46:1509-1515. [PMID: 33689532 DOI: 10.1080/02713683.2021.1901939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Purpose: Our lab has shown that conditionally disrupting the transcription factor activating protein 2β (Tfap2b) gene, responsible for the activating protein-2β (AP-2β) transcription factor, exclusively in cranial neural crest cells (AP-2β NCC KO), leads to anterior segment dysgenesis and a closed angle phenotype. The purpose of the current study is to determine if there is a progressive loss of retinal ganglion cells (RGCs) in the mutant over time and whether this loss was associated with macroglial activity changes and elevated intraocular pressure (IOP).Methods: Using the Cre-loxP system, we generated a conditional knockout of Tfap2b exclusively in cranial NCC (AP-2β NCC KO). Immunohistochemistry was performed using anti-Brn3a, anti-GFAP and anti-Vimentin antibodies. IOP was measured using a tonometer and the data was analyzed using GraphPad Prism software. Brn3a and DAPI positive cells were counted using Image-J and statistical analysis was performed with GraphPad Prism software.Results: Our findings revealed that while no statistical difference in Brn3a expression was observed between wild-type and mutant mice at postnatal day (P) 4 or P10, at P40 (p < .01) and P42 (p < .0001) Brn3a expression was significantly reduced in the mutant retina at the region of the ONH. There was also increased expression of glial fibrillary acidic protein (GFAP) by Müller cells in the AP-2β NCC KO mice at P35 and P40, indicating the presence of neuroinflammation. Moreover, increased IOP was observed starting at P35 and continuing at P40 and P42 (p < .0001 for all three ages examined).Conclusions: Together, these findings suggest that the retinal damage observed in the KO mouse becomes apparent by P40 after increased IOP was observed at P35 and progressed over time. The AP-2β NCC KO mouse may therefore be a novel experimental model for glaucoma.
Collapse
Affiliation(s)
- Aftab Taiyab
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Anthony Saraco
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Monica Akula
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Paula Deschamps
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Alexander K Ball
- Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Trevor Williams
- Craniofacial Biology, University of Colorado, Aurora, Colorado, USA
| | | |
Collapse
|
18
|
de Vos IJHM, Wong ASW, Taslim J, Ong SLM, Syder NC, Goggi JL, Carney TJ, van Steensel MAM. The novel zebrafish model pretzel demonstrates a central role for SH3PXD2B in defective collagen remodelling and fibrosis in Frank-Ter Haar syndrome. Biol Open 2020; 9:bio054270. [PMID: 33234702 PMCID: PMC7790187 DOI: 10.1242/bio.054270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/05/2020] [Indexed: 11/20/2022] Open
Abstract
Frank-Ter Haar syndrome (FTHS, MIM #249420) is a rare skeletal dysplasia within the defective collagen remodelling spectrum (DECORS), which is characterised by craniofacial abnormalities, skeletal malformations and fibrotic soft tissues changes including dermal fibrosis and joint contractures. FTHS is caused by homozygous or compound heterozygous loss-of-function mutation or deletion of SH3PXD2B (Src homology 3 and Phox homology domain-containing protein 2B; MIM #613293). SH3PXD2B encodes an adaptor protein with the same name, which is required for full functionality of podosomes, specialised membrane structures involved in extracellular matrix (ECM) remodelling. The pathogenesis of DECORS is still incompletely understood and, as a result, therapeutic options are limited. We previously generated an mmp14a/b knockout zebrafish and demonstrated that it primarily mimics the DECORS-related bone abnormalities. Here, we present a novel sh3pxd2b mutant zebrafish, pretzel, which primarily reflects the DECORS-related dermal fibrosis and contractures. In addition to relatively mild skeletal abnormalities, pretzel mutants develop dermal and musculoskeletal fibrosis, contraction of which seems to underlie grotesque deformations that include kyphoscoliosis, abdominal constriction and lateral folding. The discrepancy in phenotypes between mmp14a/b and sh3pxd2b mutants suggests that in fish, as opposed to humans, there are differences in spatiotemporal dependence of ECM remodelling on either sh3pxd2b or mmp14a/b The pretzel model presented here can be used to further delineate the underlying mechanism of the fibrosis observed in DECORS, as well as screening and subsequent development of novel drugs targeting DECORS-related fibrosis.This paper has an associated First Person interview with the first author of the article.
Collapse
Affiliation(s)
- Ivo J H M de Vos
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| | - Arnette Shi Wei Wong
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| | - Jason Taslim
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| | - Sheena Li Ming Ong
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Nicole C Syder
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 308232, Singapore
| | - Julian L Goggi
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 138667, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 117593, Singapore
| | - Thomas J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), 636921, Singapore
| | - Maurice A M van Steensel
- Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 308232, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), 636921, Singapore
| |
Collapse
|
19
|
A Rare Case Report of Frank Ter Haar Syndrome in a Sibling Pair Presenting With Congenital Glaucoma. J Glaucoma 2020; 29:236-238. [PMID: 31809395 DOI: 10.1097/ijg.0000000000001420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Frank Ter Haar syndrome (FTHS) is a rare autosomal recessive disorder with characteristic skeletal, cardiac, ocular, and craniofacial abnormalities. We report a sibling pair presenting with clinical features typical of FTHS, born to consanguineous parents, with a novel mutation in the SH3PXD2B gene on chromosome 5q35.1 that results in premature truncation of the protein encoded. The children presented with brachycephaly, multiple joint contractures, cardiac valvular defects, bilateral megalocornea, and congenital glaucoma. Trabeculotomy combined with trabeculectomy was performed in both siblings to control intraocular pressure. The characteristic clinical features with the underlying genetic defects confirmed the diagnosis of FTHS. Early diagnosis and treatment of congenital glaucoma preserved vision in the children.
Collapse
|
20
|
Durand B, Stoetzel C, Schaefer E, Calmels N, Scheidecker S, Kempf N, De Melo C, Guilbert AS, Timbolschi D, Donato L, Astruc D, Sauer A, Antal MC, Dollfus H, El Chehadeh S. A severe case of Frank-ter Haar syndrome and literature review: Further delineation of the phenotypical spectrum. Eur J Med Genet 2020; 63:103857. [DOI: 10.1016/j.ejmg.2020.103857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/07/2019] [Accepted: 01/17/2020] [Indexed: 11/26/2022]
|
21
|
Evans LP, Woll AW, Wu S, Todd BP, Hehr N, Hedberg-Buenz A, Anderson MG, Newell EA, Ferguson PJ, Mahajan VB, Harper MM, Bassuk AG. Modulation of Post-Traumatic Immune Response Using the IL-1 Receptor Antagonist Anakinra for Improved Visual Outcomes. J Neurotrauma 2020; 37:1463-1480. [PMID: 32056479 PMCID: PMC7249480 DOI: 10.1089/neu.2019.6725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to characterize acute changes in inflammatory pathways in the mouse eye after blast-mediated traumatic brain injury (bTBI) and to determine whether modulation of these pathways could protect the structure and function of retinal ganglion cells (RGC). The bTBI was induced in C57BL/6J male mice by exposure to three 20 psi blast waves directed toward the head with the body shielded, with an inter-blast interval of one hour. Acute cytokine expression in retinal tissue was measured through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) four hours post-blast. Increased retinal expression of interleukin (lL)-1β, IL-1α, IL-6, and tumor necrosis factor (TNF)α was observed in bTBI mice exposed to blast when compared with shams, which was associated with activation of microglia and macroglia reactivity, assessed via immunohistochemistry with ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein, respectively, one week post-blast. Blockade of the IL-1 pathway was accomplished using anakinra, an IL-1RI antagonist, administered intra-peritoneally for one week before injury and continuing for three weeks post-injury. Retinal function and RGC layer thickness were evaluated four weeks post-injury using pattern electroretinogram (PERG) and optical coherence tomography (OCT), respectively. After bTBI, anakinra treatment resulted in a preservation of RGC function and RGC structure when compared with saline treated bTBI mice. Optic nerve integrity analysis demonstrated a trend of decreased damage suggesting that IL-1 blockade also prevents axonal damage after blast. Blast exposure results in increased retinal inflammation including upregulation of pro-inflammatory cytokines and activation of resident microglia and macroglia. This may explain partially the RGC loss we observed in this model, as blockade of the acute inflammatory response after injury with the IL-1R1 antagonist anakinra resulted in preservation of RGC function and RGC layer thickness.
Collapse
Affiliation(s)
- Lucy P Evans
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA.,Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, USA
| | - Addison W Woll
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Shu Wu
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Brittany P Todd
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Nicole Hehr
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Adam Hedberg-Buenz
- The Iowa City Department of Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa, USA.,Department of Molecular Physiology and Biophysics, and University of Iowa, Iowa City, Iowa, USA
| | - Michael G Anderson
- The Iowa City Department of Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa, USA.,Department of Molecular Physiology and Biophysics, and University of Iowa, Iowa City, Iowa, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, USA
| | | | - Polly J Ferguson
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Matthew M Harper
- The Iowa City Department of Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
22
|
Pang IH, Clark AF. Inducible rodent models of glaucoma. Prog Retin Eye Res 2020; 75:100799. [PMID: 31557521 PMCID: PMC7085984 DOI: 10.1016/j.preteyeres.2019.100799] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 11/23/2022]
Abstract
Glaucoma is one of the leading causes of vision impairment worldwide. In order to further understand the molecular pathobiology of this disease and to develop better therapies, clinically relevant animal models are necessary. In recent years, both the rat and mouse have become popular models in glaucoma research. Key reasons are: many important biological similarities shared among rodent eyes and the human eye; development of improved methods to induce glaucoma and to evaluate glaucomatous damage; availability of genetic tools in the mouse; as well as the relatively low cost of rodent studies. Commonly studied rat and mouse glaucoma models include intraocular pressure (IOP)-dependent and pressure-independent models. The pressure-dependent models address the most important risk factor of elevated IOP, whereas the pressure-independent models assess "normal tension" glaucoma and other "non-IOP" related factors associated with glaucomatous damage. The current article provides descriptions of these models, their characterizations, specific techniques to induce glaucoma, mechanisms of injury, advantages, and limitations.
Collapse
Affiliation(s)
- Iok-Hou Pang
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA; Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| |
Collapse
|
23
|
Effect of ocular hypertension on the pattern of retinal ganglion cell subtype loss in a mouse model of early-onset glaucoma. Exp Eye Res 2019; 185:107703. [PMID: 31211954 PMCID: PMC7430001 DOI: 10.1016/j.exer.2019.107703] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/06/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022]
Abstract
Glaucoma is a neurodegenerative disease with elevated intraocular pressure as one of the major risk factors. Glaucoma leads to irreversible loss of vision and its progression involves optic nerve head cupping, axonal degeneration, retinal ganglion cell (RGC) loss, and visual field defects. Despite its high global prevalence, glaucoma still remains a major neurodegenerative disease. Introduction of mouse models of experimental glaucoma has become integral to glaucoma research due to well-studied genetics as well as ease of manipulations. Many established inherent and inducible mouse models of glaucoma are used to study the molecular and physiological progression of the disease. One such model of spontaneous mutation is the nee model, which is caused by mutation of the Sh3pxd2b gene. In both humans and mice, mutations disrupting function of the SH3PXD2B adaptor protein cause a developmental syndrome including secondary congenital glaucoma. The purpose of this study was to characterize the early onset nee glaucoma phenotype on the C57BL/6J background and to evaluate the pattern of RGC loss and axonal degeneration in specific RGC subtypes. We found that the B6.Sh3pxd2bnee mutant animals exhibit glaucoma phenotypes of elevated intraocular pressure, RGC loss and axonal degeneration. Moreover, the non-image forming RGCs survived longer than the On-Off direction selective RGCs (DSGC), and the axonal death in these RGCs was independent of their respective RGC subtype. In conclusion, through this study we characterized an experimental model of early onset glaucoma on a C57BL/6J background exhibiting key glaucoma phenotypes. In addition, we describe that RGC death has subtype-specific sensitivities and follows a specific pattern of cell death under glaucomatous conditions.
Collapse
|
24
|
Common J, Barker J, Steensel M. What does acne genetics teach us about disease pathogenesis? Br J Dermatol 2019; 181:665-676. [DOI: 10.1111/bjd.17721] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2019] [Indexed: 12/18/2022]
Affiliation(s)
- J.E.A. Common
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Singapore
| | - J.N. Barker
- St John's Institute of Dermatology Faculty of Life Sciences and Medicine King's College London London U.K
| | - M.A.M. Steensel
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR) Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Clinical Sciences Building Novena Singapore
| |
Collapse
|
25
|
Abstract
Many diseases are related to age, among these neurodegeneration is particularly important. Alzheimer's disease Parkinson's and Glaucoma have many common pathogenic events including oxidative damage, Mitochondrial dysfunction, endothelial alterations and changes in the visual field. These are well known in the case of glaucoma, less in the case of neurodegeneration of the brain. Many other molecular aspects are common, such as the role of endoplasmic reticulum autophagy and neuronal apoptosis while others have been neglected due to lack of space such as inflammatory cytokine or miRNA. Moreover, the loss of specific neuronal populations, the induction of similar mechanisms of cell injury and the deposition of protein aggregates in specific anatomical areas are very similar events between these diseases. Intracellular and/or extracellular accumulation of protein aggregates is a key feature of many neurodegenerative disorders. The existence of abnormal protein aggregates has been documented in the RGCs of glaucomatous patients such as the anomalous Tau protein or the β-amyloid accumulations. Intra-cell catabolic processes also appear to be common in both glaucoma and neurodegeneration. They also help us to understand how the basis between these diseases is common and how the visual aspects can be a serious problem for those who are affected.
Collapse
Affiliation(s)
- Sergio Claudio Saccà
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy.
| | - Carlo Alberto Cutolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Science, University of Genoa, Policlinico San Martino Hospital, Eye Clinic Genoa, Genoa, Italy
| | - Tommaso Rossi
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy
| |
Collapse
|
26
|
Iizuka S, Abdullah C, Buschman MD, Diaz B, Courtneidge SA. The role of Tks adaptor proteins in invadopodia formation, growth and metastasis of melanoma. Oncotarget 2018; 7:78473-78486. [PMID: 27802184 PMCID: PMC5346654 DOI: 10.18632/oncotarget.12954] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/22/2016] [Indexed: 01/07/2023] Open
Abstract
Metastatic cancer cells are characterized by their ability to degrade and invade through extracellular matrix. We previously showed that the Tks adaptor proteins, Tks4 and Tks5, are required for invadopodia formation and/or function in Src-transformed fibroblasts and a number of human cancer cell types. In this study, we investigated the role of Tks adaptor proteins in melanoma cell invasion and metastasis. Knockdown of either Tks4 or Tks5 in both mouse and human melanoma cell lines resulted in a decreased ability to form invadopodia and degrade extracellular matrix. In addition, Tks-knockdown melanoma cells had decreased proliferation in a 3-dimensional type l collagen matrix, but not in 2-dimensional culture conditions. We also investigated the role of Tks proteins in melanoma progression in vivo using xenografts and experimental metastasis assays. Consistent with our in vitro results, reduction of Tks proteins markedly reduced subcutaneous melanoma growth as well as metastatic growth in the lung. We explored the clinical relevance of Tks protein expression in human melanoma specimens using a tissue microarray. Compared to non-malignant nevi, both Tks proteins were highly expressed in melanoma tissues. Moreover, metastatic melanoma cases showed higher expression of Tks5 than primary melanoma cases. Taken together, these findings suggest the importance of Tks adaptor proteins in melanoma growth and metastasis in vivo, likely via functional invadopodia formation.
Collapse
Affiliation(s)
- Shinji Iizuka
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Christopher Abdullah
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Matthew D Buschman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA
| | - Begoña Diaz
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sara A Courtneidge
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
27
|
Abstract
Tyrosine kinase substrate (Tks) adaptor proteins are considered important regulators of various physiological and/or pathological processes, particularly cell migration and invasion, and cancer progression. These proteins contain PX and SH3 domains, and act as scaffolds, bringing membrane and cellular components in close proximity in structures known as invadopodia or podosomes. Tks proteins, analogous to the related proteins p47phox, p40phox and NoxO1, also facilitate local generation of reactive oxygen species (ROS), which aid in signaling at invadopodia and/or podosomes to promote their activity. As their name suggests, Tks adaptor proteins are substrates for tyrosine kinases, especially Src. In this Cell Science at a Glance article and accompanying poster, we discuss the known structural and functional aspects of Tks adaptor proteins. As the science of Tks proteins is evolving, this article will point out where we stand and what still needs to be explored. We also underscore pathological conditions involving these proteins, providing a basis for future research to develop therapies for treatment of these diseases.
Collapse
Affiliation(s)
- Priyanka Saini
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
28
|
Chang TC, Bauer M, Puerta HS, Greenberg MB, Cavuoto KM. Ophthalmic findings in Frank-ter Haar syndrome: report of a sibling pair. J AAPOS 2017; 21:514-516. [PMID: 29100834 DOI: 10.1016/j.jaapos.2017.07.216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/21/2017] [Accepted: 07/29/2017] [Indexed: 11/29/2022]
Abstract
Frank-ter Haar syndrome (FTHS) is an autosomal recessive disorder characterized by abnormalities that affect the development of bone, heart, and eyes. We report a sibling pair with FTHS caused by a homozygous, novel mutation pLys133Glnfs*13 in the SH3PXD2B gene: one sibling had bilateral ocular hypertension and unilateral colobomas of iris, choroid and retina; the other, unilateral myelinated nerve fiber layer of the optic disk and papilledema due to idiopathic intracranial hypertension. Both children had refractive amblyopia and megalocornea.
Collapse
Affiliation(s)
- Ta C Chang
- Bascom Palmer Eye Institute, Miami, Florida.
| | - Mislen Bauer
- Nicklaus Children's Hospital, Division of Genetics and Metabolism, Miami, Florida
| | - Herminia S Puerta
- Nicklaus Children's Hospital, Division of Genetics and Metabolism, Miami, Florida
| | | | | |
Collapse
|
29
|
Cairns EA, Szczesniak AM, Straiker AJ, Kulkarni PM, Pertwee RG, Thakur GA, Baldridge WH, Kelly MEM. The In Vivo Effects of the CB 1-Positive Allosteric Modulator GAT229 on Intraocular Pressure in Ocular Normotensive and Hypertensive Mice. J Ocul Pharmacol Ther 2017; 33:582-590. [PMID: 28719234 DOI: 10.1089/jop.2017.0037] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Orthosteric cannabinoid receptor 1 (CB1) activation leads to decreases in intraocular pressure (IOP). However, use of orthosteric CB1 agonists chronically has several disadvantages, limiting their usefulness as clinically relevant drugs. Allosteric modulators interact with topographically distinct sites to orthosteric ligands and may be useful to circumvent some of these disadvantages. The purpose of this study was to investigate the effects of the novel CB1-positive allosteric modulator (PAM) GAT229 on IOP. METHODS IOP was measured using rebound tonometry in anesthetized normotensive C57Bl/6 mice and in a genetic model of ocular hypertension [nose, eyes, ears (nee) mice] before drug administration, and at 1, 6, and 12 h thereafter. RESULTS In normotensive mice, topical administration of 5 μL GAT229 alone at either 0.2% or 2% did not reduce IOP. However, a subthreshold dose (0.25%) of the nonselective orthosteric CB1 agonist WIN 55,212-2, when combined with 0.2% GAT229, significantly reduced IOP compared with vehicle at 6 and 12 h. Similarly, combination of subthreshold Δ9-tetrahydrocannabinol (a nonselective orthosteric CB1 agonist; 1 mg/kg) with topical 0.2% GAT229 produced IOP lowering at 6 h. In nee mice, administration of topical 0.2% GAT229 or 10 mg/kg GAT229 alone was sufficient to lower IOP at 6 and 12 h, and 12 h, respectively. CONCLUSIONS The CB1 PAM GAT229 reduces IOP in ocular hypertensive mice and enhanced CB1-mediated IOP reduction when combined with subthreshold CB1 orthosteric ligands in normotensive mice. Administration of CB1 PAMs may provide a novel approach to reduce IOP with fewer of the disadvantages associated with orthosteric CB1 activation.
Collapse
Affiliation(s)
- Elizabeth A Cairns
- 1 Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia, Canada
| | | | - Alex J Straiker
- 2 Department of Psychological and Brain Sciences, Indiana University , Bloomington, Indiana
| | - Pushkar M Kulkarni
- 3 Department of Pharmaceutical Sciences School of Pharmacy, Bouvé College of Health Sciences, Northeastern University , Boston, Massachusetts
| | - Roger G Pertwee
- 4 School of Medicine, Medical Sciences, and Nutrition, Institute of Medical Sciences, University of Aberdeen , Aberdeen, Scotland
| | - Ganesh A Thakur
- 3 Department of Pharmaceutical Sciences School of Pharmacy, Bouvé College of Health Sciences, Northeastern University , Boston, Massachusetts
| | - William H Baldridge
- 5 Department of Medical Neuroscience, Dalhousie University , Halifax, Nova Scotia, Canada .,6 Department of Ophthalmology and Visual Sciences, Dalhousie University , Halifax, Nova Scotia, Canada
| | - Melanie E M Kelly
- 1 Department of Pharmacology, Dalhousie University , Halifax, Nova Scotia, Canada .,6 Department of Ophthalmology and Visual Sciences, Dalhousie University , Halifax, Nova Scotia, Canada
| |
Collapse
|
30
|
Paterson EK, Courtneidge SA. Invadosomes are coming: new insights into function and disease relevance. FEBS J 2017; 285:8-27. [PMID: 28548369 DOI: 10.1111/febs.14123] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/09/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Invadopodia and podosomes are discrete, actin-based molecular protrusions that form in cancer cells and normal cells, respectively, in response to diverse signaling pathways and extracellular matrix cues. Although they participate in a host of different cellular processes, they share a common functional theme of controlling pericellular proteolytic activity, which sets them apart from other structures that function in migration and adhesion, including focal adhesions, lamellipodia, and filopodia. In this review, we highlight research that explores the function of these complex structures, including roles for podosomes in embryonic and postnatal development, in angiogenesis and remodeling of the vasculature, in maturation of the postsynaptic membrane, in antigen sampling and recognition, and in cell-cell fusion mechanisms, as well as the involvement of invadopodia at multiple steps of the metastatic cascade, and how all of this may apply in the treatment of human disease states. Finally, we explore recent research that implicates a novel role for exosomes and microvesicles in invadopodia-dependent and invadopodia-independent mechanisms of invasion, respectively.
Collapse
Affiliation(s)
- Elyse K Paterson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Carreon TA, Edwards G, Wang H, Bhattacharya SK. Segmental outflow of aqueous humor in mouse and human. Exp Eye Res 2017; 158:59-66. [PMID: 27498226 PMCID: PMC5290258 DOI: 10.1016/j.exer.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 07/08/2016] [Accepted: 08/01/2016] [Indexed: 12/28/2022]
Abstract
The main and only modifiable risk factor in glaucoma, the group of usually late onset progressive and irreversible blinding optic neuropathies, is elevated intraocular pressure (IOP). The increase in IOP is due to impeded aqueous humor (AH) outflow through the conventional pathway. The aberrant increased resistance at the trabecular meshwork (TM), the filter-like region in the anterior eye chamber is the major contributory factor in causing the impeded outflow. In normal as well as in glaucoma eyes the regions of the TM are divided into areas of high and low flow. The collector channels and distal outflow regions are now increasingly being recognized as potential players in contributing to impede AH outflow. Structural and molecular make-up contributing to the segmental blockage to outflow is likely to provide greater insight. Establishing segmental blockage to outflow in model systems of glaucoma such as the mouse in parallel to human eyes will expand our repertoire of tools for investigation. Further study into this area of interest has the potential to ultimately lead to the development of new therapeutics focused on lowering IOP by targeting the various components of segmental blockage of outflow in the TM and in the distal outflow region.
Collapse
Affiliation(s)
- Teresia A Carreon
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Genea Edwards
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Haiyan Wang
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Shanghai First People's Hospital Affiliated to Jiaotong University, Shanghai, 200080, China
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
32
|
Carreon T, van der Merwe E, Fellman RL, Johnstone M, Bhattacharya SK. Aqueous outflow - A continuum from trabecular meshwork to episcleral veins. Prog Retin Eye Res 2017; 57:108-133. [PMID: 28028002 PMCID: PMC5350024 DOI: 10.1016/j.preteyeres.2016.12.004] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 11/14/2016] [Accepted: 12/22/2016] [Indexed: 12/22/2022]
Abstract
In glaucoma, lowered intraocular pressure (IOP) confers neuroprotection. Elevated IOP characterizes glaucoma and arises from impaired aqueous humor (AH) outflow. Increased resistance in the trabecular meshwork (TM), a filter-like structure essential to regulate AH outflow, may result in the impaired outflow. Flow through the 360° circumference of TM structures may be non-uniform, divided into high and low flow regions, termed as segmental. After flowing through the TM, AH enters Schlemm's canal (SC), which expresses both blood and lymphatic markers; AH then passes into collector channel entrances (CCE) along the SC external well. From the CCE, AH enters a deep scleral plexus (DSP) of vessels that typically run parallel to SC. From the DSP, intrascleral collector vessels run radially to the scleral surface to connect with AH containing vessels called aqueous veins to discharge AH to blood-containing episcleral veins. However, the molecular mechanisms that maintain homeostatic properties of endothelial cells along the pathways are not well understood. How these molecular events change during aging and in glaucoma pathology remain unresolved. In this review, we propose mechanistic possibilities to explain the continuum of AH outflow control, which originates at the TM and extends through collector channels to the episcleral veins.
Collapse
Affiliation(s)
- Teresia Carreon
- Department of Ophthalmology & Bascom Palmer Eye Institute, University of Miami, Miami, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, USA
| | - Elizabeth van der Merwe
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925 Cape Town, South Africa
| | | | - Murray Johnstone
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Sanjoy K Bhattacharya
- Department of Ophthalmology & Bascom Palmer Eye Institute, University of Miami, Miami, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, USA.
| |
Collapse
|
33
|
Mao M, Kiss M, Ou Y, Gould DB. Genetic dissection of anterior segment dysgenesis caused by a Col4a1 mutation in mouse. Dis Model Mech 2017; 10:475-485. [PMID: 28237965 PMCID: PMC5399567 DOI: 10.1242/dmm.027888] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Ocular anterior segment dysgenesis (ASD) describes a spectrum of clinically and genetically heterogeneous congenital disorders affecting anterior structures that often lead to impaired vision. More importantly, 50-75% of patients with ASD develop early onset and aggressive glaucoma. Although several genes have been implicated in the etiology of ASD, the underlying mechanisms remain elusive. Type IV collagen alpha 1 (COL4A1) is an extracellular matrix protein and a critical component of nearly all basement membranes. COL4A1 mutations cause multi-system disorders in patients, including ASD (congenital cataracts, Axenfeld-Rieger's anomaly, Peter's anomaly and microphthalmia) and congenital or juvenile glaucoma. Here, we use a conditional Col4a1 mutation in mice to determine the location and timing of pathogenic events underlying COL4A1-related ocular dysgenesis. Our results suggest that selective expression of the Col4a1 mutation in neural crest cells and their derivatives is not sufficient to cause ocular dysgenesis and that selective expression of the Col4a1 mutation in vascular endothelial cells can lead to mild ASD and optic nerve hypoplasia but only on a sensitized background. In contrast, lens-specific expression of the conditional Col4a1 mutant allele led to cataracts, mild ASD and optic nerve hypoplasia, and age-related intraocular pressure dysregulation and optic nerve damage. Finally, ubiquitous expression of the conditional Col4a1 mutation at distinct developmental stages suggests that pathogenesis takes place before E12.5. Our results show that the lens and possibly vasculature play important roles in Col4a1-related ASD and that the pathogenic events occur at mid-embryogenesis in mice, during early stages of ocular development. Summary: Key pathogenic events in anterior segment dysgenesis, a congenital ocular disease with complex etiology, are recapitulated in a mouse model of Col4a1-related ASD.
Collapse
Affiliation(s)
- Mao Mao
- Department of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Márton Kiss
- Department of Genetics, University of Szeged, Középfasor 52, Szeged H-6726, Hungary
| | - Yvonne Ou
- Department of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Douglas B Gould
- Department of Ophthalmology, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA 94143, USA .,Department of Anatomy, Institute for Human Genetics, UCSF School of Medicine, San Francisco, CA 94143, USA
| |
Collapse
|
34
|
Fingert JH, Miller K, Hedberg-Buenz A, Roos BR, Lewis CJ, Mullins RF, Anderson MG. Transgenic TBK1 mice have features of normal tension glaucoma. Hum Mol Genet 2017; 26:124-132. [PMID: 28025332 PMCID: PMC6075615 DOI: 10.1093/hmg/ddw372] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/23/2016] [Accepted: 10/25/2016] [Indexed: 01/20/2023] Open
Abstract
Duplication of the TBK1 gene is associated with 1-2% of normal tension glaucoma, a common cause of vision loss and blindness that occurs without grossly abnormal intraocular pressure. We generated a transgenic mouse that has one copy of the human TBK1 gene (native promoter and gene structure) incorporated into the mouse genome (Tg-TBK1). Expression of the TBK1 transgene in the retinae of these mice was demonstrated by real-time PCR. Using immunohistochemistry TBK1 protein was predominantly localized to the ganglion cell layer of the retina, the cell type most affected by glaucoma. More intense TBK1 labelling was detected in the retinal ganglion cells (RGCs) of Tg-TBK1 mice than in wild-type littermates. Tg-TBK1 mice exhibit the cardinal sign of glaucoma, a progressive loss of RGCs. Hemizygous Tg-TBK1 mice (with one TBK1 transgene per genome) had a 13% loss of RGCs by 18 months of age (P = 1.5 × 10-8). Homozygous Tg-TBK1 mice had 7.6% fewer RGCs than hemizygous Tg-TBK1 mice and 20% fewer RGCs than wild-type mice (P = 1.9 × 10-5) at 6 months of age. No difference in intraocular pressures was detected between Tg-TBK1 mice and wild-type littermates as they aged (P > 0.05). Tg-TBK1 mice with extra doses of the TBK1 gene recapitulate the phenotype of normal tension glaucoma in human patients with a TBK1 gene duplication. Together, these studies confirm the pathogenicity of the TBK1 gene duplication in human glaucoma and suggest that excess production of TBK1 kinase may have a role in the pathology of glaucoma.
Collapse
Affiliation(s)
- John H. Fingert
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
| | - Kathy Miller
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
| | - Adam Hedberg-Buenz
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA and
| | - Ben R. Roos
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
| | - Carly J. Lewis
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA and
| | - Robert F. Mullins
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
| | - Michael G. Anderson
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA and
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
35
|
Martino VB, Sabljic T, Deschamps P, Green RM, Akula M, Peacock E, Ball A, Williams T, West-Mays JA. Conditional deletion of AP-2β in mouse cranial neural crest results in anterior segment dysgenesis and early-onset glaucoma. Dis Model Mech 2016; 9:849-61. [PMID: 27483349 PMCID: PMC5007979 DOI: 10.1242/dmm.025262] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/21/2016] [Indexed: 12/11/2022] Open
Abstract
Anterior segment dysgenesis (ASD) encompasses a group of developmental disorders in which a closed angle phenotype in the anterior chamber of the eye can occur and 50% of patients develop glaucoma. Many ASDs are thought to involve an inappropriate patterning and migration of the periocular mesenchyme (POM), which is derived from cranial neural crest cells (NCCs) and mesoderm. Although, the mechanism of this disruption is not well understood, a number of transcriptional regulatory molecules have previously been implicated in ASDs. Here, we investigate the function of the transcription factor AP-2β, encoded by Tfap2b, which is expressed in NCCs and their derivatives. Wnt1-Cre-mediated conditional deletion of Tfap2b in NCCs resulted in post-natal ocular defects typified by opacity. Histological data revealed that the conditional AP-2β NCC knockout (KO) mutants exhibited dysgenesis of multiple structures in the anterior segment of the eye including defects in the corneal endothelium, corneal stroma, ciliary body and disruption in the iridocorneal angle with adherence of the iris to the cornea. We further show that this phenotype leads to a significant increase in intraocular pressure and a subsequent loss of retinal ganglion cells and optic nerve degeneration, features indicative of glaucoma. Overall, our findings demonstrate that AP-2β is required in the POM for normal development of the anterior segment of the eye and that the AP-2β NCC KO mice might serve as a new and exciting model of ASD and glaucoma that is fully penetrant and with early post-natal onset. Summary: Tissue-specific deletion of transcription factor AP-2β in the neural-crest-derived periocular mesenchyme generates a novel model of anterior segment dysgenesis and early onset glaucoma in mice.
Collapse
Affiliation(s)
- Vanessa B Martino
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Thomas Sabljic
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Paula Deschamps
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Rebecca M Green
- Department of Craniofacial Biology, University of Colorado Denver, Anschutz Medical Campus, Mailstop 8120, RC-1 South Building, 11th Floor, Room 111, 12801 East 17th Ave. P.O., Aurora, CO 80045, USA Department of Cell and Developmental Biology, University of Colorado Denver, Anschutz Medical Campus, Mailstop 8120, RC-1 South Building, 11th Floor, Room 111, 12801 East 17th Ave. P.O., Aurora, CO 80045, USA
| | - Monica Akula
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Erica Peacock
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Alexander Ball
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Denver, Anschutz Medical Campus, Mailstop 8120, RC-1 South Building, 11th Floor, Room 111, 12801 East 17th Ave. P.O., Aurora, CO 80045, USA Department of Cell and Developmental Biology, University of Colorado Denver, Anschutz Medical Campus, Mailstop 8120, RC-1 South Building, 11th Floor, Room 111, 12801 East 17th Ave. P.O., Aurora, CO 80045, USA
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Room 4N65, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5
| |
Collapse
|
36
|
Nair KS, Cosma M, Raghupathy N, Sellarole MA, Tolman NG, de Vries W, Smith RS, John SWM. YBR/EiJ mice: a new model of glaucoma caused by genes on chromosomes 4 and 17. Dis Model Mech 2016; 9:863-71. [PMID: 27483353 PMCID: PMC5007977 DOI: 10.1242/dmm.024307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/03/2016] [Indexed: 01/23/2023] Open
Abstract
A variety of inherited animal models with different genetic causes and distinct genetic backgrounds are needed to help dissect the complex genetic etiology of glaucoma. The scarcity of such animal models has hampered progress in glaucoma research. Here, we introduce a new inherited glaucoma model: the inbred mouse strain YBR/EiJ (YBR). YBR mice develop a form of pigmentary glaucoma. They exhibit a progressive age-related pigment-dispersing iris disease characterized by iris stromal atrophy. Subsequently, these mice develop elevated intraocular pressure (IOP) and glaucoma. Genetic mapping studies utilizing YBR as a glaucoma-susceptible strain and C57BL/6J as a glaucoma-resistant strain were performed to identify genetic loci responsible for the iris disease and high IOP. A recessive locus linked to Tyrp1b on chromosome 4 contributes to iris stromal atrophy and high IOP. However, this is not the only important locus. A recessive locus on YBR chromosome 17 causes high IOP independent of the iris stromal atrophy. In specific eyes with high IOP caused by YBR chromosome 17, the drainage angle (through which ocular fluid leaves the eye) is largely open. The YBR alleles of genes on chromosomes 4 and 17 underlie the development of high IOP and glaucoma but do so through independent mechanisms. Together, these two loci act in an additive manner to increase the susceptibility of YBR mice to the development of high IOP. The chromosome 17 locus is important not only because it causes IOP elevation in mice with largely open drainage angles but also because it exacerbates IOP elevation and glaucoma induced by pigment dispersion. Therefore, YBR mice are a valuable resource for studying the genetic etiology of IOP elevation and glaucoma, as well as for testing new treatments. Summary: We identify the YBR/EiJ mouse strain as a new model of high intraocular pressure and glaucoma, and also identify genetic loci that contribute to this glaucoma.
Collapse
Affiliation(s)
- K Saidas Nair
- Department of Ophthalmology, University of California, San Francisco, USA The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | | | | | | | | | - Simon W M John
- The Jackson Laboratory, Bar Harbor, ME, USA Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
37
|
Transplantation of iPSC-derived TM cells rescues glaucoma phenotypes in vivo. Proc Natl Acad Sci U S A 2016; 113:E3492-500. [PMID: 27274060 DOI: 10.1073/pnas.1604153113] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is a common cause of vision loss or blindness and reduction of intraocular pressure (IOP) has been proven beneficial in a large fraction of glaucoma patients. The IOP is maintained by the trabecular meshwork (TM) and the elevation of IOP in open-angle glaucoma is associated with dysfunction and loss of the postmitotic cells residing within this tissue. To determine if IOP control can be maintained by replacing lost TM cells, we transplanted TM-like cells derived from induced pluripotent stem cells into the anterior chamber of a transgenic mouse model of glaucoma. Transplantation led to significantly reduced IOP and improved aqueous humor outflow facility, which was sustained for at least 9 wk. The ability to maintain normal IOP engendered survival of retinal ganglion cells, whose loss is ultimately the cause for reduced vision in glaucoma. In vivo and in vitro analyses demonstrated higher TM cellularity in treated mice compared with littermate controls and indicated that this increase is primarily because of a proliferative response of endogenous TM cells. Thus, our study provides in vivo demonstration that regeneration of the glaucomatous TM is possible and points toward novel approaches in the treatment of this disease.
Collapse
|
38
|
Zarei K, Scheetz TE, Christopher M, Miller K, Hedberg-Buenz A, Tandon A, Anderson MG, Fingert JH, Abràmoff MD. Automated Axon Counting in Rodent Optic Nerve Sections with AxonJ. Sci Rep 2016; 6:26559. [PMID: 27226405 PMCID: PMC4881014 DOI: 10.1038/srep26559] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/05/2016] [Indexed: 01/17/2023] Open
Abstract
We have developed a publicly available tool, AxonJ, which quantifies the axons in optic nerve sections of rodents stained with paraphenylenediamine (PPD). In this study, we compare AxonJ's performance to human experts on 100x and 40x images of optic nerve sections obtained from multiple strains of mice, including mice with defects relevant to glaucoma. AxonJ produced reliable axon counts with high sensitivity of 0.959 and high precision of 0.907, high repeatability of 0.95 when compared to a gold-standard of manual assessments and high correlation of 0.882 to the glaucoma damage staging of a previously published dataset. AxonJ allows analyses that are quantitative, consistent, fully-automated, parameter-free, and rapid on whole optic nerve sections at 40x. As a freely available ImageJ plugin that requires no highly specialized equipment to utilize, AxonJ represents a powerful new community resource augmenting studies of the optic nerve using mice.
Collapse
Affiliation(s)
- Kasra Zarei
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Mark Christopher
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Kathy Miller
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Adam Hedberg-Buenz
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Veterans Affairs, Iowa City VA Medical Center, 601 Highway 6 West, Iowa City, IA 55242, USA.,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Anamika Tandon
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Michael G Anderson
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA.,Department of Veterans Affairs, Iowa City VA Medical Center, 601 Highway 6 West, Iowa City, IA 55242, USA.,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - John H Fingert
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Michael David Abràmoff
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA.,Department of Veterans Affairs, Iowa City VA Medical Center, 601 Highway 6 West, Iowa City, IA 55242, USA.,Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
39
|
Hedberg-Buenz A, Christopher MA, Lewis CJ, Fernandes KA, Dutca LM, Wang K, Scheetz TE, Abràmoff MD, Libby RT, Garvin MK, Anderson MG. Quantitative measurement of retinal ganglion cell populations via histology-based random forest classification. Exp Eye Res 2016; 146:370-385. [PMID: 26474494 PMCID: PMC4841761 DOI: 10.1016/j.exer.2015.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/20/2015] [Indexed: 01/27/2023]
Abstract
The inner surface of the retina contains a complex mixture of neurons, glia, and vasculature, including retinal ganglion cells (RGCs), the final output neurons of the retina and primary neurons that are damaged in several blinding diseases. The goal of the current work was two-fold: to assess the feasibility of using computer-assisted detection of nuclei and random forest classification to automate the quantification of RGCs in hematoxylin/eosin (H&E)-stained retinal whole-mounts; and if possible, to use the approach to examine how nuclear size influences disease susceptibility among RGC populations. To achieve this, data from RetFM-J, a semi-automated ImageJ-based module that detects, counts, and collects quantitative data on nuclei of H&E-stained whole-mounted retinas, were used in conjunction with a manually curated set of images to train a random forest classifier. To test performance, computer-derived outputs were compared to previously published features of several well-characterized mouse models of ophthalmic disease and their controls: normal C57BL/6J mice; Jun-sufficient and Jun-deficient mice subjected to controlled optic nerve crush (CONC); and DBA/2J mice with naturally occurring glaucoma. The result of these efforts was development of RetFM-Class, a command-line-based tool that uses data output from RetFM-J to perform random forest classification of cell type. Comparative testing revealed that manual and automated classifications by RetFM-Class correlated well, with 83.2% classification accuracy for RGCs. Automated characterization of C57BL/6J retinas predicted 54,642 RGCs per normal retina, and identified a 48.3% Jun-dependent loss of cells at 35 days post CONC and a 71.2% loss of RGCs among 16-month-old DBA/2J mice with glaucoma. Output from automated analyses was used to compare nuclear area among large numbers of RGCs from DBA/2J mice (n = 127,361). In aged DBA/2J mice with glaucoma, RetFM-Class detected a decrease in median and mean nucleus size of cells classified into the RGC category, as did an independent confirmation study using manual measurements of nuclear area demarcated by BRN3A-immunoreactivity. In conclusion, we have demonstrated that histology-based random forest classification is feasible and can be utilized to study RGCs in a high-throughput fashion. Despite having some limitations, this approach demonstrated a significant association between the size of the RGC nucleus and the DBA/2J form of glaucoma.
Collapse
Affiliation(s)
- Adam Hedberg-Buenz
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Mark A Christopher
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Carly J Lewis
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Kimberly A Fernandes
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Laura M Dutca
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Michael D Abràmoff
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA; Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Richard T Libby
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mona K Garvin
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Michael G Anderson
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
40
|
Wholemount imaging reveals abnormalities of the aqueous outflow pathway and corneal vascularity in Foxc1 and Bmp4 heterozygous mice. Exp Eye Res 2016; 146:293-303. [PMID: 27068508 DOI: 10.1016/j.exer.2016.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 03/16/2016] [Accepted: 04/06/2016] [Indexed: 12/14/2022]
Abstract
Mutations in the FOXC1/Foxc1 gene in humans and mice and Bmp4 in mice are associated with congenital anterior segment dysgenesis (ASD) and the development of the aqueous outflow structures throughout the limbus. The aim of this study was to advance our understanding of anterior segment abnormalities in mouse models of ASD using a 3-D imaging approach. Holistic imaging information combined with quantitative measurements were carried out on PECAM-1 stained individual components of the aqueous outflow vessels and corneal vasculature of Foxc1(+/-) on the C57BL/6Jx129 and ICR backgrounds, Bmp4(+/-) ICR mice, and wildtype mice from each background. In both wildtype and heterozygotes, singular, bifurcated and plexus forms of Schlemm's canal were noted. Of note, missing portions of the canal were seen in the heterozygous groups but not in wildtype animals. In general, we found the number of collector channels to be reduced in both heterozygotes. Lastly, we found a significant increase in the complexity of the corneal arcades and their penetration into the cornea in heterozygotes as compared with wild types. In conclusion, our 3-D imaging studies have revealed a more complex arrangement of both the aqueous vessels and corneal arcades in Foxc1(+/-) and Bmp4(+/-) heterozygotes, and further advance our understanding of how such abnormalities could impact on IOP and the aetiology of glaucoma.
Collapse
|
41
|
Gramlich OW, Ding QJ, Zhu W, Cook A, Anderson MG, Kuehn MH. Adoptive transfer of immune cells from glaucomatous mice provokes retinal ganglion cell loss in recipients. Acta Neuropathol Commun 2015; 3:56. [PMID: 26374513 PMCID: PMC4591529 DOI: 10.1186/s40478-015-0234-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 08/21/2015] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION Several studies have indicated that autoimmune and neuroinflammatory processes contribute to the neurodegeneration of retinal ganglion cells in human glaucoma patients and in animal models. To test the involvement of cellular immune processes in the pathophysiology of retinal ganglion cell degeneration in vivo, we carried out adoptive transfer experiments from two independent genetic mouse models of glaucoma into normal recipient mice. RESULTS Our findings indicate that transfer results in a progressive loss of retinal ganglion cells and their axons despite normal intraocular pressure in recipient mice. Signs of pan-retinal inflammation were not detected. Similar findings were obtained following transfer of isolated T-lymphocytes, but not after transfer of splenocytes from immune deficient glaucomatous mice. Transferred lymphocytes were detected integrated in the spleen and in the retinal ganglion cell layer of recipient animals, albeit at very low frequencies. Furthermore, we observed cell-cell interaction between transferred T-cells and recipient microglia along with focal microglial activation in recipient eyes. CONCLUSION This study demonstrates that the pathophysiology of glaucomatous degeneration in the tested animal models includes T-cell mediated events that are capable of causing loss of healthy retinal ganglion cells.
Collapse
Affiliation(s)
- Oliver W Gramlich
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA
| | - Qiong J Ding
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Wei Zhu
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Amy Cook
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Michael G Anderson
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, 52242, IA, USA
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA.
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
42
|
Abstract
Purpose We have observed that the commonly used ketamine/xylazine anesthesia mix can induce a focally severe and permanent corneal opacity. The purpose of this study was to establish the clinical and histological features of this deleterious side effect, its sensitivity with respect to age and anesthesia protocol, and approaches for avoiding it. Methods Young C57BL/6J, C57BLKS/J, and SJL/J mice were treated with permutations of anesthesia protocols and compared using slit-lamp exams, optical coherence tomography, histologic analyses, and telemetric measurements of body temperature. Results Ketamine/xylazine induces corneal damage in mice with a variable frequency. Among 12 experimental cohorts, corneal damage associated with ketamine/xylazine was observed in 9 of them. Despite various treatments to avoid corneal dehydration during anesthesia, the frequency of corneas experiencing damage among responding cohorts was 42% (26% inclusive of all cohorts), which is significantly greater than the natural prevalence (5%). The damage was consistent with band keratopathy. It appeared as a white or gray horizontal band located proximal to the pupil and was positive for subepithelial calcium deposition with von Kossa stain. Conclusions The sum of our clinical and histological observations is consistent with ketamine/xylazine-induced band keratopathy in mice. This finding is relevant for mouse studies involving the eye and/or vision-dependent behavioral assays, which would both be prone to artifact without appreciation of the damage caused by ketamine/xylazine anesthesia. Use of yohimbine is suggested as a practical means of avoiding this complication.
Collapse
|
43
|
Quigley HA. The contribution of the sclera and lamina cribrosa to the pathogenesis of glaucoma: Diagnostic and treatment implications. PROGRESS IN BRAIN RESEARCH 2015; 220:59-86. [PMID: 26497785 DOI: 10.1016/bs.pbr.2015.04.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Glaucoma, the second most common cause of world blindness, results from loss of retinal ganglion cells (RGC). RGC die as a consequence of injury to their axons, as they pass through the transition between the environment within the eye and that of the retrobulbar optic nerve, as they course to central visual centers. At the optic nerve head (ONH), axonal transport becomes abnormal, at least in part due to the effect of strain induced by intraocular pressure (IOP) on the sclera and ONH. Animal glaucoma models provide the ability to study how alterations in ocular connective tissues affect this pathological process. New therapeutic interventions are being investigated to mitigate glaucoma blindness by modifying the remodeling of ocular tissues in glaucoma. Some genetically altered mice are resistant to glaucoma damage, while treatment of the sclera with cross-linking agents makes experimental mouse glaucoma damage worse. Inhibition of transforming growth factor β activity is strikingly protective. Treatments that alter the response of ocular connective tissues to IOP may be effective in protecting those with glaucoma from vision loss.
Collapse
Affiliation(s)
- Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Using genetic mouse models to gain insight into glaucoma: Past results and future possibilities. Exp Eye Res 2015; 141:42-56. [PMID: 26116903 DOI: 10.1016/j.exer.2015.06.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/16/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022]
Abstract
While all forms of glaucoma are characterized by a specific pattern of retinal ganglion cell death, they are clinically divided into several distinct subclasses, including normal tension glaucoma, primary open angle glaucoma, congenital glaucoma, and secondary glaucoma. For each type of glaucoma there are likely numerous molecular pathways that control susceptibility to the disease. Given this complexity, a single animal model will never precisely model all aspects of all the different types of human glaucoma. Therefore, multiple animal models have been utilized to study glaucoma but more are needed. Because of the powerful genetic tools available to use in the laboratory mouse, it has proven to be a highly useful mammalian system for studying the pathophysiology of human disease. The similarity between human and mouse eyes coupled with the ability to use a combination of advanced cell biological and genetic tools in mice have led to a large increase in the number of studies using mice to model specific glaucoma phenotypes. Over the last decade, numerous new mouse models and genetic tools have emerged, providing important insight into the cell biology and genetics of glaucoma. In this review, we describe available mouse genetic models that can be used to study glaucoma-relevant disease/pathobiology. Furthermore, we discuss how these models have been used to gain insights into ocular hypertension (a major risk factor for glaucoma) and glaucomatous retinal ganglion cell death. Finally, the potential for developing new mouse models and using advanced genetic tools and resources for studying glaucoma are discussed.
Collapse
|
45
|
What Animal Models Can Tell Us About Glaucoma. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:365-80. [DOI: 10.1016/bs.pmbts.2015.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Abu-Hassan DW, Acott TS, Kelley MJ. The Trabecular Meshwork: A Basic Review of Form and Function. ACTA ACUST UNITED AC 2014; 2. [PMID: 25356439 DOI: 10.13188/2334-2838.1000017] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Diala W Abu-Hassan
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, USA
| | - Ted S Acott
- Department of Biochemistry & Molecular Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Mary J Kelley
- Department of Biochemistry & Physiology, University of Jordan, Amman, Jordan
| |
Collapse
|
47
|
Mutations in SH3PXD2B cause Borrone dermato-cardio-skeletal syndrome. Eur J Hum Genet 2013; 22:741-7. [PMID: 24105366 DOI: 10.1038/ejhg.2013.229] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/08/2013] [Accepted: 08/31/2013] [Indexed: 11/08/2022] Open
Abstract
Borrone Dermato-Cardio-Skeletal (BDCS) syndrome is a severe progressive autosomal recessive disorder characterized by coarse facies, thick skin, acne conglobata, dysmorphic facies, vertebral abnormalities and mitral valve prolapse. We identified a consanguineous kindred with a child clinically diagnosed with BDCS. Linkage analysis of this family (BDCS1) identified five regions homozygous by descent with a maximum LOD score of 1.75. Linkage analysis of the family that originally defined BDCS (BDCS3) identified an overlapping linkage peak at chromosome 5q35.1. Sequence analysis identified two different homozygous mutations in BDCS1 and BDCS3, affecting the gene encoding the protein SH3 and PX domains 2B (SH3PXD2B), which localizes to 5q35.1. Western blot analysis of patient fibroblasts derived from affected individuals in both families demonstrated complete loss of SH3PXD2B. Homozygosity mapping and sequence analysis in a second published BDCS family (BDCS2) excluded SH3PXD2B. SH3PXD2B is required for the formation of functional podosomes, and loss-of-function mutations in SH3PXD2B have recently been shown to underlie 7 of 13 families with Frank-Ter Haar syndrome (FTHS). FTHS and BDCS share some overlapping clinical features; therefore, our results demonstrate that a proportion of BDCS and FTHS cases are allelic. Mutations in other gene(s) functioning in podosome formation and regulation are likely to underlie the SH3PXD2B-mutation-negative BDSC/FTHS patients.
Collapse
|
48
|
Quigley HA, Cone FE. Development of diagnostic and treatment strategies for glaucoma through understanding and modification of scleral and lamina cribrosa connective tissue. Cell Tissue Res 2013; 353:231-44. [PMID: 23535950 PMCID: PMC3716834 DOI: 10.1007/s00441-013-1603-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/28/2013] [Indexed: 12/13/2022]
Abstract
Considerable evidence indicates that the state of ocular connective tissues and their response in glaucomatous disease affect the degree of glaucoma damage. Both experimental and clinical data suggest that improved diagnostic and prognostic information can be derived from the assessment of the mechanical responsiveness of the sclera and lamina cribrosa to intraocular pressure (IOP). Controlled mutagenesis of the sclera has produced a mouse strain that is relatively resistant to increased IOP. Alteration of the baseline scleral state can be accomplished through either increased cross-linking of fibrillar components or their reduction. The sclera is a dynamic structure, altering its structure and behavior in response to IOP change. The biochemical pathways that control these responses are fertile areas for new glaucoma treatments.
Collapse
Affiliation(s)
- Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
49
|
Lu Y, Vitart V, Burdon KP, Khor CC, Bykhovskaya Y, Mirshahi A, Hewitt AW, Koehn D, Hysi PG, Ramdas WD, Zeller T, Vithana EN, Cornes BK, Tay WT, Tai ES, Cheng CY, Liu J, Foo JN, Saw SM, Thorleifsson G, Stefansson K, Dimasi DP, Mills RA, Mountain J, Ang W, Hoehn R, Verhoeven VJM, Grus F, Wolfs R, Castagne R, Lackner KJ, Springelkamp H, Yang J, Jonasson F, Leung DYL, Chen LJ, Tham CCY, Rudan I, Vatavuk Z, Hayward C, Gibson J, Cree AJ, MacLeod A, Ennis S, Polasek O, Campbell H, Wilson JF, Viswanathan AC, Fleck B, Li X, Siscovick D, Taylor KD, Rotter JI, Yazar S, Ulmer M, Li J, Yaspan BL, Ozel AB, Richards JE, Moroi SE, Haines JL, Kang JH, Pasquale LR, Allingham RR, Ashley-Koch A, Mitchell P, Wang JJ, Wright AF, Pennell C, Spector TD, Young TL, Klaver CCW, Martin NG, Montgomery GW, Anderson MG, Aung T, Willoughby CE, Wiggs JL, Pang CP, Thorsteinsdottir U, Lotery AJ, Hammond CJ, van Duijn CM, Hauser MA, Rabinowitz YS, Pfeiffer N, Mackey DA, Craig JE, Macgregor S, Wong TY. Genome-wide association analyses identify multiple loci associated with central corneal thickness and keratoconus. Nat Genet 2013; 45:155-63. [PMID: 23291589 DOI: 10.1038/ng.2506] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 11/29/2012] [Indexed: 12/13/2022]
Abstract
Central corneal thickness (CCT) is associated with eye conditions including keratoconus and glaucoma. We performed a meta-analysis on >20,000 individuals in European and Asian populations that identified 16 new loci associated with CCT at genome-wide significance (P < 5 × 10(-8)). We further showed that 2 CCT-associated loci, FOXO1 and FNDC3B, conferred relatively large risks for keratoconus in 2 cohorts with 874 cases and 6,085 controls (rs2721051 near FOXO1 had odds ratio (OR) = 1.62, 95% confidence interval (CI) = 1.4-1.88, P = 2.7 × 10(-10), and rs4894535 in FNDC3B had OR = 1.47, 95% CI = 1.29-1.68, P = 4.9 × 10(-9)). FNDC3B was also associated with primary open-angle glaucoma (P = 5.6 × 10(-4); tested in 3 cohorts with 2,979 cases and 7,399 controls). Further analyses implicate the collagen and extracellular matrix pathways in the regulation of CCT.
Collapse
Affiliation(s)
- Yi Lu
- Queensland Institute of Medical Research, Statistical Genetics, Herston, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bendon CL, Fenwick AL, Hurst JA, Nürnberg G, Nürnberg P, Wall SA, Wilkie AOM, Johnson D. Frank-ter Haar syndrome associated with sagittal craniosynostosis and raised intracranial pressure. BMC MEDICAL GENETICS 2012; 13:104. [PMID: 23140272 PMCID: PMC3532175 DOI: 10.1186/1471-2350-13-104] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 10/29/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND Frank-ter Haar syndrome is a rare disorder associated with skeletal, cardiac, ocular and craniofacial features including hypertelorism and brachycephaly. The most common underlying genetic defect in Frank-ter Haar syndrome appears to be a mutation in the SH3PXD2B gene on chromosome 5q35.1. Craniosynostosis, or premature fusion of the calvarial sutures, has not previously been described in Frank-ter Haar syndrome. CASE PRESENTATION We present a family of three affected siblings born to consanguineous parents with clinical features in keeping with a diagnosis of Frank-ter Haar syndrome. All three siblings have a novel mutation caused by the deletion of exon 13 of the SH3PXD2B gene. Two of the three siblings also have non-scaphocephalic sagittal synostosis associated with raised intracranial pressure. CONCLUSION The clinical features of craniosynostosis and raised intracranial pressure in this family with a confirmed diagnosis of Frank-ter Haar syndrome expand the clinical spectrum of the disease. The abnormal cranial proportions in a mouse model of the disease suggests that the association is not coincidental. The possibility of craniosynostosis should be considered in individuals with a suspected diagnosis of Frank-ter Haar syndrome.
Collapse
Affiliation(s)
- Charlotte L Bendon
- Oxford Craniofacial Unit, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | | | | | | | | | |
Collapse
|