1
|
Nguyen BN, Britten-Jones AC, Bui BV, Walker LE, Titter P. Physiological and pathological changes to the eye and vision during and after pregnancy. Clin Exp Optom 2024:1-9. [PMID: 39374945 DOI: 10.1080/08164622.2024.2410031] [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: 03/29/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
Pregnancy introduces a multitude of changes in the body, including hormonal fluctuations and metabolic changes, which can lead to atypical ocular signs and symptoms. Ocular manifestations range from fluctuations in vision, to microstructural changes in the retina and choroid, to dry eye disease. This narrative review highlights the range of pregnancy-related effects on the eye and vision that are likely to present in the context of routine eyecare. Specifically, physiological ocular changes and pathological ocular changes that manifest for the first time, or are exacerbated, in uncomplicated pregnancy are discussed. The literature has evolved from simply noting differences in the eye between pregnant and non-pregnant groups, to refining knowledge of the proposed underlying pathophysiology with the advent of newer technologies in eyecare. A particular focus of this review is navigating when pregnancy changes in the eye occur or peak during the gestational period, and whether the changes are short-lived or might extend past pregnancy. While many pregnancy-associated changes are temporary and resolve post-partum, it is also recognised that some changes persist after pregnancy, with a notable absence of literature on ocular changes with loss or termination of pregnancy. Currently or previously pregnant women (or those planning to become pregnant), and other health professionals, should be educated about the importance of seeking eyecare before, during and after pregnancy.
Collapse
Affiliation(s)
- Bao N Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Lorraine E Walker
- School of Nursing and Midwifery, Monash University, Clayton, Victoria, Australia
| | - Peta Titter
- School of Nursing, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
2
|
Sánchez-Puebla L, de Hoz R, Salobrar-García E, Arias-Vázquez A, González-Jiménez M, Ramírez AI, Fernández-Albarral JA, Matamoros JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto Vaquero C, Cuartero MI, Moro MA, Salazar JJ, López-Cuenca I, Ramírez JM. Age-Related Retinal Layer Thickness Changes Measured by OCT in APPNL-F/NL-F Mice: Implications for Alzheimer's Disease. Int J Mol Sci 2024; 25:8221. [PMID: 39125789 PMCID: PMC11312090 DOI: 10.3390/ijms25158221] [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: 05/29/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
In Alzheimer's disease (AD), transgenic mouse models have established links between abnormalities in the retina and those in the brain. APPNL-F/NL-F is a murine, humanized AD model that replicates several pathological features observed in patients with AD. Research has focused on obtaining quantitative parameters from optical coherence tomography (OCT) in AD. The aim of this study was to analyze, in a transversal case-control study using manual retinal segmentation via SD-OCT, the changes occurring in the retinal layers of the APPNL/F-NF/L AD model in comparison to C57BL/6J mice (WT) at 6, 9, 12, 15, 17, and 20 months of age. The analysis focused on retinal thickness in RNFL-GCL, IPL, INL, OPL, and ONL based on the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Both APPNL-F/NL-F-model and WT animals exhibited thickness changes at the time points studied. While WT showed significant changes in INL, OPL, and ONL, the AD model showed changes in all retinal layers analyzed. The APPNL-F/NL-F displayed significant thickness variations in the analyzed layers except for the IPL compared to related WT. These thickness changes closely resembled those found in humans during preclinical stages, as well as during mild and moderate AD stages, making this AD model behave more similarly to the disease in humans.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Brain Science Institute, RIKEN, Laboratory for Proteolytic Neuroscience, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto Vaquero
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
3
|
Linsenmeier RA, Dmitriev AV. Increased Retinal Metabolism Induced by Flicker in the Isolated Mouse Retina. eNeuro 2024; 11:ENEURO.0509-23.2024. [PMID: 38641415 PMCID: PMC11089847 DOI: 10.1523/eneuro.0509-23.2024] [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: 12/05/2023] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024] Open
Abstract
Both the retina and brain exhibit neurovascular coupling, increased blood flow during increased neural activity. In the retina increased blood flow can be evoked by flickering light, but the magnitude of the metabolic change that underlies this is not known. Local changes in oxygen consumption (QO2) are difficult to measure in vivo when both supply and demand are changing. Here we isolated the C57BL/6J mouse retina and supplied it with oxygen from both sides of the tissue. Microelectrode recordings of PO2 were made in darkness and during 20 s of high scotopic flickering light at 1 Hz. Flicker led to a PO2 increase in the outer retina and a decrease in the inner retina, indicating that outer retinal QO2 (QOR) decreased and inner retinal QO2 (QIR) increased. A four-layer oxygen diffusion model was fitted to PO2 values obtained in darkness and at the end of flicker to determine the values of QOR and QIR. QOR in flicker was 76 ± 14% (mean and SD, n = 10) of QOR in darkness. The increase in QIR was smaller, 6.4 ± 5.0%. These metabolic changes are likely smaller than the maximum changes, because with no regeneration of pigment in the isolated retina, we limited the illumination. Further modeling indicated that at high illumination, QIR could increase by up to 45%, which is comparable to the magnitude of flow changes. This suggests that the blood flow increase is at least roughly matched to the increased metabolic demands of activity in the retina.
Collapse
Affiliation(s)
- Robert A Linsenmeier
- Departments of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
- Neurobiology, Northwestern University, Evanston, Illinois 60208
- Department of Ophthalmology, Northwestern University, Chicago, Illinois 60611
| | - Andrey V Dmitriev
- Departments of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
4
|
Grannonico M, Miller DA, Liu M, Krause MA, Savier E, Erisir A, Netland PA, Cang J, Zhang HF, Liu X. Comparative In Vivo Imaging of Retinal Structures in Tree Shrews, Humans, and Mice. eNeuro 2024; 11:ENEURO.0373-23.2024. [PMID: 38538082 PMCID: PMC10972737 DOI: 10.1523/eneuro.0373-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Rodent models, such as mice and rats, are commonly used to examine retinal ganglion cell damage in eye diseases. However, as nocturnal animals, rodent retinal structures differ from primates, imposing significant limitations in studying retinal pathology. Tree shrews (Tupaia belangeri) are small, diurnal paraprimates that exhibit superior visual acuity and color vision compared with mice. Like humans, tree shrews have a dense retinal nerve fiber layer (RNFL) and a thick ganglion cell layer (GCL), making them a valuable model for investigating optic neuropathies. In this study, we applied high-resolution visible-light optical coherence tomography to characterize the tree shrew retinal structure in vivo and compare it with that of humans and mice. We quantitatively characterize the tree shrew's retinal layer structure in vivo, specifically examining the sublayer structures within the inner plexiform layer (IPL) for the first time. Next, we conducted a comparative analysis of retinal layer structures among tree shrews, mice, and humans. We then validated our in vivo findings in the tree shrew inner retina using ex vivo confocal microscopy. The in vivo and ex vivo analyses of the shrew retina build the foundation for future work to accurately track and quantify the retinal structural changes in the IPL, GCL, and RNFL during the development and progression of human optic diseases.
Collapse
Affiliation(s)
- Marta Grannonico
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - David A Miller
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Mingna Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Michael A Krause
- Departments of Ophthalmology, University of Virginia, Charlottesville, Virginia 22904
| | - Elise Savier
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Alev Erisir
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| | - Peter A Netland
- Departments of Ophthalmology, University of Virginia, Charlottesville, Virginia 22904
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
- Psychology, University of Virginia, Charlottesville, Virginia 22904
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
5
|
Chou TH, Hao Z, Alba D, Lazo A, Gallo Afflitto G, Eastwood JD, Porciatti V, Guy J, Yu H. Mitochondrially Targeted Gene Therapy Rescues Visual Loss in a Mouse Model of Leber's Hereditary Optic Neuropathy. Int J Mol Sci 2023; 24:17068. [PMID: 38069388 PMCID: PMC10707051 DOI: 10.3390/ijms242317068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Leber's hereditary optic neuropathy (LHON) is a common mitochondrial genetic disease, causing irreversible blindness in young individuals. Current treatments are inadequate, and there is no definitive cure. This study evaluates the effectiveness of delivering wildtype human NADH ubiquinone oxidoreductase subunit 4 (hND4) gene using mito-targeted AAV(MTSAAV) to rescue LHOH mice. We observed a declining pattern in electroretinograms amplitudes as mice aged across all groups (p < 0.001), with significant differences among groups (p = 0.023; Control vs. LHON, p = 0.008; Control vs. Rescue, p = 0.228). Inner retinal thickness and intraocular pressure did not change significantly with age or groups. Compared to LHON mice, those rescued with wildtype hND4 exhibited improved retinal visual acuity (0.29 ± 0.1 cy/deg vs. 0.15 ± 0.1 cy/deg) and increased functional hyperemia response (effect of flicker, p < 0.001, effect of Group, p = 0.004; Interaction Flicker × Group, p < 0.001). Postmortem analysis shows a marked reduction in retinal ganglion cell density in the LHON group compared to the other groups (Effect of Group, p < 0.001, Control vs. LHON, p < 0.001, Control vs. Rescue, p = 0.106). These results suggest that MTSAAV-delivered wildtype hND4 gene rescues, at least in part, visual impairment in an LHON mouse model and has the therapeutic potential to treat this disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (T.-H.C.); (Z.H.); (D.A.); (A.L.); (G.G.A.); (J.D.E.); (J.G.)
| | | | - Hong Yu
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (T.-H.C.); (Z.H.); (D.A.); (A.L.); (G.G.A.); (J.D.E.); (J.G.)
| |
Collapse
|
6
|
Wang Y, Halawa M, Chatterjee A, Eshwaran R, Qiu Y, Wibowo YC, Pan J, Wieland T, Feng Y. Sufficient Cav-1 levels in the endothelium are critical for the maintenance of the neurovascular unit in the retina. Mol Med 2023; 29:152. [PMID: 37923999 PMCID: PMC10623831 DOI: 10.1186/s10020-023-00749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Caveolin-1 (Cav-1) is a pivotal protein in the plasma membrane. Studies on homozygous Cav-1 deficient mice revealed that Cav-1 is essential for endothelial function and angiogenesis in the retina. However, whether a reduction in Cav-1 content hampers the neurovascular unit (NVU) in the retina is unclear. Thus, this study examines the NVU in the retinas of heterozygous Cav-1 deficient (Cav-1+/-) mice and analyzes possible underlying mechanisms. METHODS The vascular, glial and neuronal components in the retina were evaluated using retinal morphometry, whole mount retinal immunofluorescence staining, histological analysis and optical coherence tomography. In addition, immunoblotting and immunofluorescence staining, subcellular fractionation, biotin labeling of cell surface proteins, and proximity ligation assay were employed to detect expression and localization of proteins in the retina or endothelial cells (ECs) upon knockdown of Cav-1 with Cav-1 siRNA. RESULTS Cav-1+/- retinas showed a significant reduction in pericyte coverage along with an increase in acellular capillaries compared to controls at 8 months of age, but not at 1 month. A significant loss and obvious morphological abnormalities of smooth muscle cells were observed in 8-month-old Cav-1+/- retinal arterioles. Macroglial and microglial cells were activated in the Cav-1+/- retinas. A transient significant delay in retinal angiogenesis was detected in Cav-1+/- retinas at p5, which was however no longer detectable at p10. The Cav-1+/- retinas displayed increased vascular permeability and a notable reduction in VEGFR2 content at 8 months. In vitro, siRNA-mediated knockdown experiments in ECs revealed that the loss of Cav-1 in ECs resulted in decreased levels of VEGFR2, VE-Cadherin and their interaction at the plasma membrane as well. CONCLUSION Our results indicate that a sufficient Cav-1 level over 50% of its normal abundance is vital for the proper localization of VEGFR2 and VE-cadherin, likely in a complex, at the plasma membrane, which is essential for the maintenance of normal NVU in the retina.
Collapse
Affiliation(s)
- Yixin Wang
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Mahmoud Halawa
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Anupriya Chatterjee
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Rachana Eshwaran
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Yi Qiu
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Yohanes Cakrapradipta Wibowo
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Jianyuan Pan
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
7
|
Wei-Zhang S, Cui B, Xing M, Liu J, Guo Y, He K, Bai T, Dong X, Lei Y, Zhou W, Zhou H, Liu S, Wang X, Zhou D, Yan H. Chimpanzee adenovirus-mediated multiple gene therapy for age-related macular degeneration. iScience 2023; 26:107939. [PMID: 37810255 PMCID: PMC10550724 DOI: 10.1016/j.isci.2023.107939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Neovascular age-related macular degeneration AMD (nAMD) is characterized by choroidal neovascularization (CNV) and could lead to irreversible blindness. However, anti-vascular endothelial growth factor (VEGF) therapy has limited efficacy. Therefore, we generated a chimpanzee adenoviral vector (AdC68-PFC) containing three genes, pigment endothelial-derived factor (PEDF), soluble fms-like tyrosine kinase-1 (sFlt-1), and soluble forms of CD59 (sCD59), to treat nAMD. The results showed that AdC68-PFC mediated a strong onset of PEDF, sFlt-1, and sCD59 expression both in vivo and in vitro. AdC68-PFC showed preventive and therapeutic effects following intravitreal (IVT) injection in the laser-induced CNV model and very low-density lipoprotein receptor-deficient (Vldlr-/-) mouse model. In vitro assessment indicated that AdC68-PFC had a strong inhibitory effect on endothelial cells. Importantly, the safety test showed no evidence of in vivo toxicity of adenovirus in murine eyes. Our findings suggest that AdC68-PFC may be a long-acting and safe gene therapy vector for future nAMD treatments.
Collapse
Affiliation(s)
- Selena Wei-Zhang
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bohao Cui
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yingying Guo
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kai He
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
| | - Tinghui Bai
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
| | - Yi Lei
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
| | - Wei Zhou
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Hui Zhou
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Shengnan Liu
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin 300070, China
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Barroso Á, Ketelhut S, Nettels-Hackert G, Heiduschka P, del Amor R, Naranjo V, Kemper B, Schnekenburger J. Durable 3D murine ex vivo retina glaucoma models for optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2023; 14:4421-4438. [PMID: 37791268 PMCID: PMC10545187 DOI: 10.1364/boe.494271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 10/05/2023]
Abstract
Durable and standardized phantoms with optical properties similar to native healthy and disease-like biological tissues are essential tools for the development, performance testing, calibration and comparison of label-free high-resolution optical coherence tomography (HR-OCT) systems. Available phantoms are based on artificial materials and reflect thus only partially ocular properties. To address this limitation, we have performed investigations on the establishment of durable tissue phantoms from ex vivo mouse retina for enhanced reproduction of in vivo structure and complexity. In a proof-of-concept study, we explored the establishment of durable 3D models from dissected mouse eyes that reproduce the properties of normal retina structures and tissue with glaucoma-like layer thickness alterations. We explored different sectioning and preparation procedures for embedding normal and N-methyl-D-aspartate (NMDA)-treated mouse retina in transparent gel matrices and epoxy resins, to generate durable three-dimensional tissue models. Sample quality and reproducibility were quantified by thickness determination of the generated layered structures utilizing computer-assisted segmentation of OCT B-scans that were acquired with a commercial HR-OCT system at a central wavelength of 905 nm and analyzed with custom build software. Our results show that the generated 3D models feature thin biological layers close to current OCT resolution limits and glaucoma-like tissue alterations that are suitable for reliable HR-OCT performance characterization. The comparison of data from resin-embedded tissue with native murine retina in gels demonstrates that by utilization of appropriate preparation protocols, highly stable samples with layered structures equivalent to native tissues can be fabricated. The experimental data demonstrate our concept as a promising approach toward the fabrication of durable biological 3D models suitable for high-resolution OCT system performance characterization supporting the development of optimized instruments for ophthalmology applications.
Collapse
Affiliation(s)
- Álvaro Barroso
- Biomedical Technology Center of the Medical Faculty, University of Muenster, Mendelstr. 17, D-48149 Muenster, Germany
| | - Steffi Ketelhut
- Biomedical Technology Center of the Medical Faculty, University of Muenster, Mendelstr. 17, D-48149 Muenster, Germany
| | - Gerburg Nettels-Hackert
- Department of Ophthalmology of the Medical Faculty, University of Muenster, Domagkstr. 15, D-48149 Muenster, Germany
| | - Peter Heiduschka
- Department of Ophthalmology of the Medical Faculty, University of Muenster, Domagkstr. 15, D-48149 Muenster, Germany
| | - Rocío del Amor
- Instituto Universitario de Investigación en Tecnología Centrada en el Ser Humano, Universitat Politècnica de València, Valencia, Spain
| | - Valery Naranjo
- Instituto Universitario de Investigación en Tecnología Centrada en el Ser Humano, Universitat Politècnica de València, Valencia, Spain
| | - Björn Kemper
- Biomedical Technology Center of the Medical Faculty, University of Muenster, Mendelstr. 17, D-48149 Muenster, Germany
| | - Jürgen Schnekenburger
- Biomedical Technology Center of the Medical Faculty, University of Muenster, Mendelstr. 17, D-48149 Muenster, Germany
| |
Collapse
|
9
|
Remlinger J, Bagnoud M, Meli I, Massy M, Hoepner R, Linington C, Chan A, Bennett JL, Enzmann V, Salmen A. Modeling MOG Antibody-Associated Disorder and Neuromyelitis Optica Spectrum Disorder in Animal Models: Visual System Manifestations. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200141. [PMID: 37429715 PMCID: PMC10691219 DOI: 10.1212/nxi.0000000000200141] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Mechanisms of visual impairment in aquaporin 4 antibody (AQP4-IgG) seropositive neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody (MOG-IgG)-associated disorder (MOGAD) are incompletely understood. The respective impact of optic nerve demyelination and primary and secondary retinal neurodegeneration are yet to be investigated in animal models. METHODS Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6Jrj mice, and monoclonal MOG-IgG (8-18C5, murine), recombinant AQP4-IgG (rAb-53, human), or isotype-matched control IgG (Iso-IgG, human) was administered 10 days postimmunization. Mobility impairment was scored daily. Visual acuity by optomotor reflex and ganglion cell complex thickness (GCC, 3 innermost retinal layers) by optical coherence tomography (OCT) were longitudinally assessed. Histopathology of optic nerve and retina was investigated during presymptomatic, acute, and chronic disease phases for immune cells, demyelination, complement deposition, natural killer (NK) cell, AQP4, and astrocyte involvement, retinal ganglion cells (RGCs), and Müller cell activation. Groups were compared by nonparametric tests with a p value <0.05 indicating statistical significance. RESULTS Visual acuity decreased from baseline to chronic phase in MOG-IgG (mean ± standard error of the mean: 0.54 ± 0.01 to 0.46 ± 0.02 cycles/degree, p < 0.05) and AQP4-IgG EAE (0.54 ± 0.01 to 0.43 ± 0.02, cycles/degree, p < 0.05). Immune cell infiltration of optic nerves started in presymptomatic AQP4-IgG, but not in MOG-IgG EAE (5.85 ± 2.26 vs 0.13 ± 0.10 macrophages/region of interest [ROI] and 1.88 ± 0.63 vs 0.15 ± 0.06 T cells/ROI, both p < 0.05). Few NK cells, no complement deposition, and stable glial fibrillary acid protein and AQP4 fluorescence intensity characterized all EAE optic nerves. Lower GCC thickness (Spearman correlation coefficient r = -0.44, p < 0.05) and RGC counts (r = -0.47, p < 0.05) correlated with higher mobility impairment. RGCs decreased from presymptomatic to chronic disease phase in MOG-IgG (1,705 ± 51 vs 1,412 ± 45, p < 0.05) and AQP4-IgG EAE (1,758 ± 14 vs 1,526 ± 48, p < 0.01). Müller cell activation was not observed in either model. DISCUSSION In a multimodal longitudinal characterization of visual outcome in animal models of MOGAD and NMOSD, differential retinal injury and optic nerve involvement were not conclusively clarified. Yet optic nerve inflammation was earlier in AQP4-IgG-associated pathophysiology. Retinal atrophy determined by GCC thickness (OCT) and RGC counts correlating with mobility impairment in the chronic phase of MOG-IgG and AQP4-IgG EAE may serve as a generalizable marker of neurodegeneration.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Maud Bagnoud
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Ivo Meli
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Marine Massy
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Robert Hoepner
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Christopher Linington
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Andrew Chan
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Jeffrey L Bennett
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Volker Enzmann
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Anke Salmen
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland.
| |
Collapse
|
10
|
Che Z, Bi F, Sun Y, Xing W, Huang H, Zhang X. Dilated residual FPN-based segmentation for mouse retinal images. Heliyon 2023; 9:e18605. [PMID: 37576244 PMCID: PMC10413074 DOI: 10.1016/j.heliyon.2023.e18605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/15/2023] Open
Abstract
Background and objective Diabetes can induce diabetic retinopathy (DR), and the blindness caused by this disease is irreversible. The early analysis of mouse retinal images, including the layer and cell segmentation properties of these images, can help to effectively diagnose this disease. Method In the study, we design a dilated residual method based on a feature pyramid network (FPN), in which the FPN is adopted as the base network for solving the multiscale segmentation problem concerning mouse retinal images. In the bottom-up encoding pathway, we construct our backbone feature extraction network via the combination of dilated convolution and a residual block, further increasing the range of the receptive field to obtain more context information. At the same time, we integrate a squeeze-and-excitation (SE) attention module into the backbone network to obtain more small object details. In the top-down decoding pathway, we replace the traditional nearest-neighbor upsampling method with the transposed convolution method and add a segmentation head to obtain semantic segmentation results. Results The effectiveness of our network model is verified in two segmentation tasks: ganglion cell segmentation and mouse retinal cell and layer segmentation. The outcomes demonstrate that, compared to other supervised segmentation methods based on deep learning, our model attains the utmost precision in both binary segmentation and multiclass semantic segmentation tasks. Conclusion The dilated residual FPN is a robust method for mouse retinal image segmentation and it can effectively assist DR diagnosis.
Collapse
Affiliation(s)
- Zhihao Che
- School of Information Science and Technology, North China University of Technology, Beijing, China
| | - Fukun Bi
- School of Information Science and Technology, North China University of Technology, Beijing, China
| | - Yu Sun
- School of Information Science and Technology, North China University of Technology, Beijing, China
| | - Weiying Xing
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Huang
- Beijing Duan-Dian Pharmaceutical Research & Development Co., Ltd., Beijing, China
| | - Xinyue Zhang
- Beijing Duan-Dian Pharmaceutical Research & Development Co., Ltd., Beijing, China
| |
Collapse
|
11
|
Linsenmeier RA, Dmitriev AV, Dmitriev AA. Oxygen profiles and oxygen consumption in the isolated mouse retina. Exp Eye Res 2023; 233:109554. [PMID: 37437835 PMCID: PMC10528762 DOI: 10.1016/j.exer.2023.109554] [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: 04/12/2023] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
The retina has a large demand for oxygen, but there is only limited information on differences between oxygen utilization (QO2) in the inner and outer retina, and limited data on mouse, which has become a prevalent animal model. This study utilized the isolated mouse retina, which allowed more detailed spatial analysis of QO2 than other methods. Oxygen sensitive microelectrodes were used to obtain profiles of oxygen tension across the isolated mouse retina, and mathematical models of retinal oxygen diffusion with four and five layers were fitted to the data to obtain values for QO2 of the outer retina (QOR) and inner retina (QIR). The boundaries between layers were free parameters in these models. The five-layer model resulted in lower error between the model and data, and agreed better with known anatomy. The three layers for the outer retina occupied half of the retina, as in prior work on rat, cat, and monkey, and the inner half of the retina could be divided into two layers, in which the one closer to the vitreous (layer 5) had much lower QO2 than the more distal inner retina (layer 4). QIR in darkness was 3.9 ml O2-100 g-1-min-1, similar to the value for intact cat retina, and did not change during light. QOR in darkness was 2.4 ml O2-100 g-1-min-1, lower than previous values in cat and rat, possibly because of damage to photoreceptors during isolation. There was a tendency for QOR to be lower in light, but it was not significant in this preparation.
Collapse
Affiliation(s)
- Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA; Department of Neurobiology, Northwestern University, Evanston, Illinois, USA; Department of Ophthalmology, Northwestern University, Chicago, Illinois, USA.
| | - Andrey V Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA.
| | - Alexander A Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA.
| |
Collapse
|
12
|
Mellak Y, Achim A, Ward A, Nicholson L, Descombes X. A machine learning framework for the quantification of experimental uveitis in murine OCT. BIOMEDICAL OPTICS EXPRESS 2023; 14:3413-3432. [PMID: 37497491 PMCID: PMC10368067 DOI: 10.1364/boe.489271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 07/28/2023]
Abstract
This paper presents methods for the detection and assessment of non-infectious uveitis, a leading cause of vision loss in working age adults. In the first part, we propose a classification model that can accurately predict the presence of uveitis and differentiate between different stages of the disease using optical coherence tomography (OCT) images. We utilize the Grad-CAM visualization technique to elucidate the decision-making process of the classifier and gain deeper insights into the results obtained. In the second part, we apply and compare three methods for the detection of detached particles in the retina that are indicative of uveitis. The first is a fully supervised detection method, the second is a marked point process (MPP) technique, and the third is a weakly supervised segmentation that produces per-pixel masks as output. The segmentation model is used as a backbone for a fully automated pipeline that can segment small particles of uveitis in two-dimensional (2-D) slices of the retina, reconstruct the volume, and produce centroids as points distribution in space. The number of particles in retinas is used to grade the disease, and point process analysis on centroids in three-dimensional (3-D) shows clustering patterns in the distribution of the particles on the retina.
Collapse
Affiliation(s)
- Youness Mellak
- Université Côte d’Azur, INRIA, CNRS, I3S, Sophia Antipolis, France
| | - Alin Achim
- University of Bristol, Bristol, United Kingdom
| | - Amy Ward
- University of Bristol, Bristol, United Kingdom
| | | | - Xavier Descombes
- Université Côte d’Azur, INRIA, CNRS, I3S, Sophia Antipolis, France
| |
Collapse
|
13
|
Swirski S, May O, Ahlers M, Wissinger B, Greschner M, Jüschke C, Neidhardt J. In Vivo Efficacy and Safety Evaluations of Therapeutic Splicing Correction Using U1 snRNA in the Mouse Retina. Cells 2023; 12:cells12060955. [PMID: 36980294 PMCID: PMC10047704 DOI: 10.3390/cells12060955] [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: 01/20/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Efficacy and safety considerations constitute essential steps during development of in vivo gene therapies. Herein, we evaluated efficacy and safety of splice factor-based treatments to correct mutation-induced splice defects in an Opa1 mutant mouse line. We applied adeno-associated viruses to the retina. The viruses transduced retinal cells with an engineered U1 snRNA splice factor designed to correct the Opa1 splice defect. We found the treatment to be efficient in increasing wild-type Opa1 transcripts. Correspondingly, Opa1 protein levels increased significantly in treated eyes. Measurements of retinal morphology and function did not reveal therapy-related side-effects supporting the short-term safety of the treatment. Alterations of potential off-target genes were not detected. Our data suggest that treatments of splice defects applying engineered U1 snRNAs represent a promising in vivo therapeutic approach. The therapy increased wild-type Opa1 transcripts and protein levels without detectable morphological, functional or genetic side-effects in the mouse eye. The U1 snRNA-based therapy can be tailored to specific disease gene mutations, hence, raising the possibility of a wider applicability of this promising technology towards treatment of different inherited retinal diseases.
Collapse
Affiliation(s)
- Sebastian Swirski
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Oliver May
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Malte Ahlers
- Visual Neuroscience, Department of Neuroscience, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
| | - Martin Greschner
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
- Research Center Neurosensory Science, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Christoph Jüschke
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - John Neidhardt
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| |
Collapse
|
14
|
Ginkgo biloba extracts improve choroidal circulation leading to suppression of myopia in mice. Sci Rep 2023; 13:3772. [PMID: 36882511 PMCID: PMC9989591 DOI: 10.1038/s41598-023-30908-1] [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: 08/18/2022] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Myopia is becoming more common across the world, necessitating the development of preventive methods. We investigated the activity of early growth response 1 (EGR-1) protein and discovered that Ginkgo biloba extracts (GBEs) activated EGR-1 in vitro. In vivo, C57BL/6 J mice were fed either normal or 0.0667% GBEs (200 mg/kg) mixed chow (n = 6 each), and myopia was induced with - 30 diopter (D) lenses from 3 to 6 weeks of age. Refraction and axial length were measured by an infrared photorefractor and an SD-OCT system, respectively. In lens-induced myopia mice, oral GBEs significantly improved refractive errors (- 9.92 ± 1.53 D vs. - 1.67 ± 3.51 D, p < 0.001) and axial elongation (0.22 ± 0.02 mm vs. 0.19 ± 0.02 mm, p < 0.05). To confirm the mechanism of GBEs in preventing myopia progression, the 3-week-old mice were divided into normally fed with either myopic-induced or non-myopic-induced groups and GBEs fed with either myopic-induced or non-myopic-induced groups (n = 10 each). Choroidal blood perfusion was measured with optical coherence tomography angiography (OCTA). In both non-myopic induced groups, compared to normal chow, oral GBEs significantly improved choroidal blood perfusion (8.48 ± 15.75%Area vs. 21.74 ± 10.54%Area, p < 0.05) and expression of Egr-1 and endothelial nitric oxide synthase (eNOS) in the choroid. In both myopic-induced groups, compared to normal chow, oral GBEs also improved choroidal blood perfusion (- 9.82 ± 9.47%Area vs. 2.29 ± 11.84%Area, p < 0.05) and was positively correlated with the change in choroidal thickness. These findings suggest that GBEs may inhibit the progression of myopia by improving choroidal blood perfusion.
Collapse
|
15
|
Jeong H, Kurihara T, Jiang X, Kondo S, Ueno Y, Hayashi Y, Lee D, Ikeda SI, Mori K, Torii H, Negishi K, Tsubota K. Suppressive effects of violet light transmission on myopia progression in a mouse model of lens-induced myopia. Exp Eye Res 2023; 228:109414. [PMID: 36764596 DOI: 10.1016/j.exer.2023.109414] [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: 05/24/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The prevalence of myopia has been steadily increasing for several decades, and this condition can cause extensive medical and economic issues in society. Exposure to violet light (VL), a short wavelength (360-400 nm) of visible light from sunlight, has been suggested as an effective preventive and suppressive treatments for the development and progression of myopia. However, the clinical application of VL remains unclear. In this study, we aimed to investigate the preventive and suppressive effects of VL on myopia progression. Various transmittances of VL (40%, 70%, and 100%) were tested in C57BL/6J mice with lens-induced myopia (LIM). Changes in the refractive error, axial length, and choroid thickness during the 3-week LIM were measured. The myopic shift in refractive error and difference in axial length between the 0 and -30 diopter lens was lessened in a transmission-dependent manner. Choroidal thinning, which was observed in myopic conditions, was suppressed by VL exposure and affected by its transmission. The results suggest that myopia progression can be managed using VL transmittance. Therefore, these factors should be considered for the prevention and treatment of myopia.
Collapse
Affiliation(s)
- Heonuk Jeong
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Xiaoyan Jiang
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shinichiro Kondo
- Tsubota Laboratory, Inc., 34 Shinanomachi, 304 Toshin Shinanomachi Ekimae Building, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Yusuke Ueno
- Menicon Co., Ltd., 21-19, Aoi 3, Naka-ku, Nagoya, 460-0006, Japan
| | - Yuki Hayashi
- Menicon Co., Ltd., 21-19, Aoi 3, Naka-ku, Nagoya, 460-0006, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shin-Ichi Ikeda
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kiwako Mori
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidemasa Torii
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Tsubota Laboratory, Inc., 34 Shinanomachi, 304 Toshin Shinanomachi Ekimae Building, Shinjuku-ku, Tokyo, 160-0016, Japan.
| |
Collapse
|
16
|
Lindovsky J, Palkova M, Symkina V, Raishbrook MJ, Prochazka J, Sedlacek R. OCT and ERG Techniques in High-Throughput Phenotyping of Mouse Vision. Genes (Basel) 2023; 14:genes14020294. [PMID: 36833221 PMCID: PMC9956909 DOI: 10.3390/genes14020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
The purpose of the study is to demonstrate coherent optical tomography and electroretinography techniques adopted from the human clinical practice to assess the morphology and function of the mouse retina in a high-throughput phenotyping environment. We present the normal range of wild-type C57Bl/6NCrl retinal parameters in six age groups between 10 and 100 weeks as well as examples of mild and severe pathologies resulting from knocking out a single protein-coding gene. We also show example data obtained by more detailed analysis or additional methods useful in eye research, for example, the angiography of a superficial and deep vascular complex. We discuss the feasibility of these techniques in conditions demanding a high-throughput approach such as the systemic phenotyping carried out by the International Mouse Phenotyping Consortium.
Collapse
|
17
|
Zhou T, Yang Z, Ni B, Zhou H, Xu H, Lin X, Li Y, Liu C, Ju R, Ge J, He C, Liu X. IL-4 induces reparative phenotype of RPE cells and protects against retinal neurodegeneration via Nrf2 activation. Cell Death Dis 2022; 13:1056. [PMID: 36539414 PMCID: PMC9768119 DOI: 10.1038/s41419-022-05433-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
Retinal degeneration is a kind of neurodegeneration characterized by progressive neuronal death and dysfunction of retinal pigment epithelium (RPE) cells, leading to permanent visual impairment. It still lacks effective therapeutic options and new drugs are highly warranted. In this study, we found the expression of IL-4, a critical regulator of immunity, was reduced in both patients and mouse models. Importantly, exogenous intravitreal IL-4 application could exert a novel neuroprotective effect, characterized by well-preserved RPE layer and neuroretinal structure, as well as amplified wave-amplitudes in ERG. The RNA-seq analysis revealed that IL-4 treatment suppressed the essential oxidative and pro-inflammatory pathways in the degenerative retina. Particularly, IL-4 upregulated the IL-4Rα on RPE cells and induced a reparative phenotype via the activation of Nrf2 both in vitro and in vivo. Furthermore, the Nrf2-/- mice displayed no recovery in response to IL-4 application, highlighting a significant role of Nrf2 in IL-4-mediated protection. Our data provides evidence that IL-4 protects against retinal neurodegeneration by its antioxidant and anti-inflammatory property through IL-4Rα upregulation and Nrf2 activation in RPE cells. The IL-4/IL-4Rα-Nrf2 axis maybe the potential targets for the development of novel therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Tian Zhou
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Ziqi Yang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Biyan Ni
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Hong Zhou
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Huiyi Xu
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Xiaojing Lin
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Yingmin Li
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Chunqiao Liu
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Rong Ju
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Jian Ge
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Chang He
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| | - Xialin Liu
- grid.12981.330000 0001 2360 039XState Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 510060 Guangzhou, China
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 0] [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
|
19
|
Yu HY, Lee MW, Kim JT, Lee SC, Lee YH. Comparison of each retinal layer thicknesses between eyes with CRVO and normal contralateral eyes. KOREAN JOURNAL OF OPHTHALMOLOGY 2022; 36:274-281. [PMID: 35527524 PMCID: PMC9194737 DOI: 10.3341/kjo.2021.0174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/11/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the difference in each retinal layer thickness in central retinal vein occlusion (CRVO) with resolved macular edema after intravitreal antivascular endothelial growth factor injection and normal contralateral eyes.. Methods Patients with ischemic and nonischemic CRVO whose macular edema resolved after intravitreal antivascular endothelial growth factor injections and did not recur for at least 6 months, and a normal contralateral eye were enrolled. Each retinal layer thickness between CRVO and normal contralateral eyes was compared according to Early Treatment Diabetic Retinopathy Study subfields using spectral domain optical coherence tomography. Results The thicknesses of outer nuclear layer, photoreceptor layer, and retinal pigment epithelium in central ring, ganglion cell layer, inner plexiform layer, outer nuclear layer, and photoreceptor layer in the inner ring, and ganglion cell layer in the outer ring of CRVO eyes were significantly thinner than those of normal contralateral eyes (all p < 0.05). Whereas, inner nuclear layer and outer plexiform layer thicknesses in central ring of CRVO eyes were 23.86 ± 8.8 and 25.76 ± 7.6 μm, respectively, which was significantly thicker than those of normal contralateral eyes (19.52 ± 7.7 and 22.76 ± 6.5 μm; p = 0.019 and p = 0.043, respectively). Additionally, the mean best-corrected visual acuity of CRVO eyes were significantly correlated with photoreceptor layer thickness in central ring (p = 0.005). Conclusions In CRVO eyes with resolved macular edema, the outer retinal layers were thinner as well as inner retinal layers, whereas inner plexiform layer and outer nuclear layer were thicker than normal fellow eyes. Additionally, photoreceptor layer thickness in foveal area had a significant impact on visual acuity in CRVO.
Collapse
Affiliation(s)
- Hwa Young Yu
- Department of Ophthalmology, Konyang University College of Medicine, Daejeon, Korea
| | - Min Woo Lee
- Department of Ophthalmology, Konyang University College of Medicine, Daejeon, Korea
| | - Jung Tae Kim
- Department of Ophthalmology, Konyang University College of Medicine, Daejeon, Korea
| | - Sung Chul Lee
- Department of Ophthalmology, Konyang University College of Medicine, Daejeon, Korea
| | - Young Hoon Lee
- Department of Ophthalmology, Konyang University College of Medicine, Daejeon, Korea
| |
Collapse
|
20
|
Mischi E, Soukup P, Harman CD, Oikawa K, Kowalska ME, Hartnack S, McLellan GJ, Komáromy AM, Pot SA. Outer retinal thickness and visibility of the choriocapillaris in four distinct retinal regions imaged with spectral domain optical coherence tomography in dogs and cats. Vet Ophthalmol 2022; 25 Suppl 1:122-135. [PMID: 35611616 PMCID: PMC9246961 DOI: 10.1111/vop.12989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To evaluate the outer retinal band thickness and choriocapillaris (CC) visibility in four distinct retinal regions in dogs and cats imaged with spectral domain optical coherence tomography (SD-OCT). To attempt delineation of a fovea-like region in canine and feline SD-OCT scans, aided by the identification of outer retinal thickness differences between retinal regions. METHODS Spectralis® HRA + OCT SD-OCT scans from healthy, anesthetized dogs (n = 10) and cats (n = 12) were analyzed. Scanlines on which the CC was identifiable were counted and CC visibility was scored. Outer nuclear layer (ONL) thickness and the distances from external limiting membrane (ELM) to retinal pigment epithelium/Bruch's membrane complex (RPE/BM) and ELM to CC were measured in the area centralis (AC), a visually identified fovea-like region, and in regions superior and inferior to the optic nerve head (ONH). Measurements were analyzed using a multilevel regression. RESULTS The CC was visible in over 90% of scanlines from dogs and cats. The ONL was consistently thinnest in the fovea-like region. The outer retina (ELM-RPE and ELM-CC) was thickest within the AC compared with superior and inferior to the ONH in dogs and cats (p < .001 for all comparisons). CONCLUSIONS The CC appears a valid, albeit less than ideal outer retinal boundary marker in tapetal species. The AC can be objectively differentiated from the surrounding retina on SD-OCT images of dogs and cats; a fovea-like region was identified in dogs and its presence was suggested in cats. These findings allow targeted imaging and image evaluation of these regions of retinal specialization.
Collapse
Affiliation(s)
- Elisa Mischi
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Petr Soukup
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Christine D. Harman
- Department of Small Animal Clinical Sciences, College of Veterinary MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Kazuya Oikawa
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Malwina E. Kowalska
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
- Section of Epidemiology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Sonja Hartnack
- Section of Epidemiology, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Gillian J. McLellan
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - András M. Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Simon A. Pot
- Ophthalmology Section, Equine Department, Vetsuisse FacultyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
21
|
Quantitative Optical Coherence Tomography for Longitudinal Monitoring of Postnatal Retinal Development in Developing Mouse Eyes. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A better study of postnatal retinal development is essential for the in-depth understanding of the nature of the vision system. To date, quantitative analysis of postnatal retinal development is primarily limited to endpoint histological examination. This study is to validate in vivo optical coherence tomography (OCT) for longitudinal monitoring of postnatal retinal development in developing mouse eyes. OCT images of C57BL/6J mice were recorded from postnatal day (P) 14 to P56. Three-dimensional (3D) frame registration and super averaging were adopted to investigate the fine structure of the retina. Quantitative OCT analysis revealed distinct outer and inner retinal layer changes, corresponding to eye development. At the outer retina, external limiting membrane (ELM) and ellipsoid zone (EZ) band intensities gradually increased with aging, and the IZ band was detectable by P28. At the inner retina, a hyporeflective layer (HRL) between the nerve fiber layer (NFL) and inner plexiform layer (IPL) was observed in developing eyes and gradually disappeared with aging. Further image analysis revealed individual RGCs within the HRL layer of the young mouse retina. However, RGCs were merged with the NFL and the IPL in the aged mouse retina. Moreover, the sub-IPL layer structure was observed to be gradually enhanced with aging. To interpret the observed retinal layer kinetics, a model based on eyeball expansion, cell apoptosis, and retinal structural modification was proposed.
Collapse
|
22
|
Retinal Degeneration in a Murine Model of Retinal Ischemia by Unilateral Common Carotid Artery Occlusion. BIOMED RESEARCH INTERNATIONAL 2022; 2021:7727648. [PMID: 35005021 PMCID: PMC8741345 DOI: 10.1155/2021/7727648] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 01/21/2023]
Abstract
Retinal degeneration is a progressive retinal damage in ocular vascular diseases. There are several reasons for this, such as occlusion of arteries or veins, diabetic retinopathy, or hereditary retinal diseases. To study pathological mechanisms of retinal degeneration, it is required to develop experimentally reproducible and clinically relevant models. In our previous studies, we developed a murine model of retinal hypoperfusion by unilateral common carotid artery occlusion (UCCAO) which mimics the pathophysiology of ocular ischemic syndrome (OIS) in humans, and described broad pathological mechanisms in the retina after UCCAO. However, there still remain missing pieces of the ocular pathologic process by UCCAO. In this study, we examined those unfound mechanisms. UCCAO was performed on adult mice. Ocular dysfunctions, histological deficits, and inflammation were examined after UCCAO, compared with sham-operated mice. Evaluation values were analyzed by electrophysiological, histological, and molecular biological methods. Eyelid drooping was permanently seen after UCCAO. Induction time point of acute reversible cataract under anesthesia was shortened. Retinal/visual dysfunctions were detected 2-4 weeks after UCCAO. Specifically, scotopic b-wave was more affected than a-wave, with the dysfunction of photopic b-wave. Impaired oscillatory potentials and visual evoked potential were constantly observed. Pathological Müller gliosis/inflammation was featured with NeuN-positive cell loss in the ganglion cell layer. Axial length, intraocular pressure, pupillary light reflex, and retinal pigment epithelium/choroidal thickness were not changed by UCCAO. A murine model of retinal ischemia by UCCAO can be useful for studying a series of degenerative process in the ischemic retina.
Collapse
|
23
|
Barter KR, Paradis H, Gendron RL, Vidal JAL, Meruvia-Pastor O. Novel segmentation algorithm for high-throughput analysis of spectral domain-optical coherence tomography imaging of teleost retinas. Mol Vis 2022; 28:492-499. [PMID: 37089699 PMCID: PMC10115363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/29/2022] [Indexed: 04/25/2023] Open
Abstract
Spectral domain-optical coherence tomography (SD-OCT) has become an essential tool for assessing ocular tissues in live subjects and conducting research on ocular development, health, and disease. The processing of SD-OCT images, particularly those from non-mammalian species, is a labor-intensive manual process due to a lack of automated analytical programs. This paper describes the development and implementation of a novel computer algorithm for the quantitative analysis of SD-OCT images of live teleost eyes. Automated segmentation processing of SD-OCT images of retinal layers was developed using a novel algorithm based on thresholding. The algorithm measures retinal thickness characteristics in a large volume of imaging data of teleost ocular structures in a short time, providing increased accuracy and repeatability of SD-OCT image analysis over manual measurements. The algorithm also generates hundreds of retinal thickness measurements per image for a large number of images for a given dataset. Meanwhile, heat mapping software that plots SD-OCT image measurements as a color gradient was also created. This software directly converts the measurements of each processed image to represent changes in thickness across the whole retinal scan. It also enables 2D and 3D visualization of retinal thickness across the scan, facilitating specimen comparison and localization of areas of interest. The study findings showed that the novel algorithm is more accurate, reliable, and repeatable than manual SD-OCT analysis. The adaptability of the algorithm makes it potentially suitable for analyzing SD-OCT scans of other non-mammalian species.
Collapse
Affiliation(s)
- Kent R Barter
- Department of Computer Science, Memorial University, St. John's, A1B 3X5, NL, Canada
| | - Hélène Paradis
- Faculty of Medicine, Memorial University, St. John's, A1B 3V6, NL, Canada
| | - Robert L Gendron
- Faculty of Medicine, Memorial University, St. John's, A1B 3V6, NL, Canada
| | - Josué A Lily Vidal
- Department of Ophthalmology, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7K 0M7, Canada
| | - Oscar Meruvia-Pastor
- Department of Computer Science, Memorial University, St. John's, A1B 3X5, NL, Canada
| |
Collapse
|
24
|
Alex V, Motevasseli T, Freeman WR, Jayamon JA, Bartsch DUG, Borooah S. Assessing the validity of a cross-platform retinal image segmentation tool in normal and diseased retina. Sci Rep 2021; 11:21784. [PMID: 34750415 PMCID: PMC8575997 DOI: 10.1038/s41598-021-01105-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/18/2021] [Indexed: 11/09/2022] Open
Abstract
Comparing automated retinal layer segmentation using proprietary software (Heidelberg Spectralis HRA + OCT) and cross-platform Optical Coherence Tomography (OCT) segmentation software (Orion). Image segmentations of normal and diseased (iAMD, DME) eyes were performed using both softwares and then compared to the 'gold standard' of manual segmentation. A qualitative assessment and quantitative (layer volume) comparison of segmentations were performed. Segmented images from the two softwares were graded by two masked graders and in cases with difference, a senior retina specialist made a final independent decisive grading. Cross-platform software was significantly better than the proprietary software in the segmentation of NFL and INL layers in Normal eyes. It generated significantly better segmentation only for NFL in iAMD and for INL and OPL layers in DME eyes. In normal eyes, all retinal layer volumes calculated by the two softwares were moderate-strongly correlated except OUTLY. In iAMD eyes, GCIPL, INL, ONL, INLY, TRV layer volumes were moderate-strongly correlated between softwares. In eyes with DME, all layer volume values were moderate-strongly correlated between softwares. Cross-platform software can be used reliably in research settings to study the retinal layers as it compares well against manual segmentation and the commonly used proprietary software for both normal and diseased eyes.
Collapse
Affiliation(s)
- Varsha Alex
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | - Tahmineh Motevasseli
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | - William R Freeman
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | | | - Dirk-Uwe G Bartsch
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | - Shyamanga Borooah
- Jacobs Retina Center, Shiley Eye Institute, University of California San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
25
|
Dmitriev AV, Dmitriev AA, Linsenmeier RA. K +-dependent Müller cell-generated components of the electroretinogram. Vis Neurosci 2021; 38:E010. [PMID: 34294176 PMCID: PMC10422678 DOI: 10.1017/s0952523821000092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The electroretinogram (ERG) has been employed for years to collect information about retinal function and pathology. The usefulness of this noninvasive test depends on our understanding of the cell sources that generate the ERG. Important contributors to the ERG are glial Müller cells (MCs), which are capable of generating substantial transretinal potentials in response to light-induced changes in extracellular K+ concentration ([K+]o). For instance, the MCs generate the slow PIII (sPIII) component of the ERG as a reaction to a photoreceptor-induced [K+]o decrease in the subretinal space. Similarly, an increase of [K+]o related to activity of postreceptor retinal neurons also produces transretinal glial currents, which can potentially influence the amplitude and shape of the b-wave, one of the most frequently analyzed ERG components. Although it is well documented that the majority of the b-wave originates from On-bipolar cells, some contribution from MCs was suggested many years ago and has never been experimentally rejected. In this work, detailed information about light-evoked [K+]o changes in the isolated mouse retina was collected and then analyzed with a relatively simple linear electrical model of MCs. The results demonstrate that the cornea-positive potential generated by MCs is too small to contribute noticeably to the b-wave. The analysis also explains why MCs produce the large cornea-negative sPIII subcomponent of the ERG, but no substantial cornea-positive potential.
Collapse
Affiliation(s)
- Andrey V Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - Alexander A Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
- Department of Neurobiology, Northwestern University, Evanston, Illinois
- Department of Ophthalmology, Northwestern University, Chicago, Illinois
| |
Collapse
|
26
|
Bradley LJ, Ward A, Hsue MCY, Liu J, Copland DA, Dick AD, Nicholson LB. Quantitative Assessment of Experimental Ocular Inflammatory Disease. Front Immunol 2021; 12:630022. [PMID: 34220797 PMCID: PMC8250853 DOI: 10.3389/fimmu.2021.630022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/28/2021] [Indexed: 11/25/2022] Open
Abstract
Ocular inflammation imposes a high medical burden on patients and substantial costs on the health-care systems that mange these often chronic and debilitating diseases. Many clinical phenotypes are recognized and classifying the severity of inflammation in an eye with uveitis is an ongoing challenge. With the widespread application of optical coherence tomography in the clinic has come the impetus for more robust methods to compare disease between different patients and different treatment centers. Models can recapitulate many of the features seen in the clinic, but until recently the quality of imaging available has lagged that applied in humans. In the model experimental autoimmune uveitis (EAU), we highlight three linked clinical states that produce retinal vulnerability to inflammation, all different from healthy tissue, but distinct from each other. Deploying longitudinal, multimodal imaging approaches can be coupled to analysis in the tissue of changes in architecture, cell content and function. This can enrich our understanding of pathology, increase the sensitivity with which the impacts of therapeutic interventions are assessed and address questions of tissue regeneration and repair. Modern image processing, including the application of artificial intelligence, in the context of such models of disease can lay a foundation for new approaches to monitoring tissue health.
Collapse
Affiliation(s)
- Lydia J Bradley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Amy Ward
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Madeleine C Y Hsue
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Jian Liu
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew D Dick
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom.,Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom.,University College London, Institute of Ophthalmology, London, United Kingdom
| | - Lindsay B Nicholson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
27
|
Mesentier-Louro LA, Rangel B, Stell L, Shariati MA, Dalal R, Nathan A, Yuan K, de Jesus Perez V, Liao YJ. Hypoxia-induced inflammation: Profiling the first 24-hour posthypoxic plasma and central nervous system changes. PLoS One 2021; 16:e0246681. [PMID: 33661927 PMCID: PMC7932147 DOI: 10.1371/journal.pone.0246681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/22/2021] [Indexed: 11/18/2022] Open
Abstract
Central nervous system and visual dysfunction is an unfortunate consequence of systemic hypoxia in the setting of cardiopulmonary disease, including infection with SARS-CoV-2, high-altitude cerebral edema and retinopathy and other conditions. Hypoxia-induced inflammatory signaling may lead to retinal inflammation, gliosis and visual disturbances. We investigated the consequences of systemic hypoxia using serial retinal optical coherence tomography and by assessing the earliest changes within 24h after hypoxia by measuring a proteomics panel of 39 cytokines, chemokines and growth factors in the plasma and retina, as well as using retinal histology. We induced severe systemic hypoxia in adult C57BL/6 mice using a hypoxia chamber (10% O2) for 1 week and rapidly assessed measurements within 1h compared with 18h after hypoxia. Optical coherence tomography revealed retinal tissue edema at 18h after hypoxia. Hierarchical clustering of plasma and retinal immune molecules revealed obvious segregation of the 1h posthypoxia group away from that of controls. One hour after hypoxia, there were 10 significantly increased molecules in plasma and 4 in retina. Interleukin-1β and vascular endothelial growth factor were increased in both tissues. Concomitantly, there was significantly increased aquaporin-4, decreased Kir4.1, and increased gliosis in retinal histology. In summary, the immediate posthypoxic period is characterized by molecular changes consistent with systemic and retinal inflammation and retinal glial changes important in water transport, leading to tissue edema. This posthypoxic inflammation rapidly improves within 24h, consistent with the typically mild and transient visual disturbance in hypoxia, such as in high-altitude retinopathy. Given hypoxia increases risk of vision loss, more studies in at-risk patients, such as plasma immune profiling and in vivo retinal imaging, are needed in order to identify novel diagnostic or prognostic biomarkers of visual impairment in systemic hypoxia.
Collapse
Affiliation(s)
- Louise A. Mesentier-Louro
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Barbara Rangel
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Laurel Stell
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, California, United States of America
| | - M. Ali Shariati
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Roopa Dalal
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Abinaya Nathan
- Department of Pulmonary Medicine, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Ke Yuan
- Divisions of Pulmonary Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Vinicio de Jesus Perez
- Department of Pulmonary Medicine, Stanford University, School of Medicine, Stanford, California, United States of America
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States of America
- Department of Neurology, Stanford University, School of Medicine, Stanford, California, United States of America
| |
Collapse
|
28
|
Salobrar-García E, López-Cuenca I, Sánchez-Puebla L, de Hoz R, Fernández-Albarral JA, Ramírez AI, Bravo-Ferrer I, Medina V, Moro MA, Saido TC, Saito T, Salazar JJ, Ramírez JM. Retinal Thickness Changes Over Time in a Murine AD Model APP NL-F/NL-F. Front Aging Neurosci 2021; 12:625642. [PMID: 33542683 PMCID: PMC7852550 DOI: 10.3389/fnagi.2020.625642] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Alzheimer's disease (AD) may present retinal changes before brain pathology, suggesting the retina as an accessible biomarker of AD. The present work is a diachronic study using spectral domain optical coherence tomography (SD-OCT) to determine the total retinal thickness and retinal nerve fiber layer (RNFL) thickness in an APPNL−F/NL−F mouse model of AD at 6, 9, 12, 15, 17, and 20 months old compared to wild type (WT) animals. Methods: Total retinal thickness and RNFL thickness were determined. The mean total retinal thickness was analyzed following the Early Treatment Diabetic Retinopathy Study sectors. RNFL was measured in six sectors of axonal ring scans around the optic nerve. Results: In the APPNL−F/NL−F group compared to WT animals, the total retinal thickness changes observed were the following: (i) At 6-months-old, a significant thinning in the outer temporal sector was observed; (ii) at 15-months-old a significant thinning in the inner temporal and in the inner and outer inferior retinal sectors was noticed; (iii) at 17-months-old, a significant thickening in the inferior and nasal sectors was found in both inner and outer rings; and (iv) at 20-months-old, a significant thinning in the inner ring of nasal, temporal, and inferior retina and in the outer ring of superior and temporal retina was seen. In RNFL thickness, there was significant thinning in the global analysis and in nasal and inner-temporal sectors at 6 months old. Thinning was also found in the supero-temporal and nasal sectors and global value at 20 months old. Conclusions: In the APPNL−F/NL−F AD model, the retinal thickness showed thinning, possibly produced by neurodegeneration alternating with thickening caused by deposits and neuroinflammation in some areas of the retina. These changes over time are similar to those observed in the human retina and could be a biomarker for AD. The APPNL−F/NL−F AD model may help us better understand the different retinal changes during the progression of AD.
Collapse
Affiliation(s)
- Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Lídia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Ana I Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Isabel Bravo-Ferrer
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain.,Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Violeta Medina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María A Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Juan J Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
29
|
Atkinson R, Leung J, Bender J, Kirkcaldie M, Vickers J, King A. TDP-43 mislocalization drives neurofilament changes in a novel model of TDP-43 proteinopathy. Dis Model Mech 2021; 14:dmm.047548. [PMID: 33408125 PMCID: PMC7888715 DOI: 10.1242/dmm.047548] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Mislocalization of the TAR DNA-binding protein 43 (TDP-43) from the nucleus to the cytoplasm is a common feature of neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). The downstream in vivo cellular effects of this mislocalization are not well understood. To investigate the impact of mislocalized TDP-43 on neuronal cell bodies, axons and axonal terminals, we utilized the mouse visual system to create a new model of TDP-43 proteinopathy. Mouse (C57BL/6J) retinal ganglion cells (RGCs) were transduced with GFP-tagged human wildtype TDP-43 (hTDP-WT-GFP) and human TDP-43 with a mutation in the nuclear localization sequence (hTDP-ΔNLS-GFP), to cause TDP-43 mislocalization, with ∼60% transduction efficiency achieved. Expression of both hTDP-WT-GFP and hTDP-ΔNLS-GFP resulted in changes to neurofilament expression, with cytoplasmic TDP-43 being associated with significantly (p<0.05) increased neurofilament heavy expression in the cell soma, and both forms of altered TDP-43 leading to significantly (p<0.05) decreased numbers of neurofilament-positive axons within the optic nerve. Alterations to neurofilament proteins were associated with significantly (p<0.05) increased microglial density in the optic nerve and retina. Furthermore expression of hTDP-WT-GFP was associated with a significant (p<0.05) increase in pre-synaptic input into RGCs in the retina. The current study has developed a new model allowing detailed examination of alterations to TDP-43 and will contribute to the knowledge of TDP-43-mediated neuronal alterations and degeneration.
Collapse
Affiliation(s)
- Rachel Atkinson
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Jacqueline Leung
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - James Bender
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Matthew Kirkcaldie
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - James Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Anna King
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| |
Collapse
|
30
|
Gu J, Jiang T, Yu M, Yu J, Li W, Liu S, Zhang P, Chen W, Chang Q. A Novel Approach to Quantitative Evaluation of Outer Retinal Lesions Via a New Parameter "Integral" in Spectral Domain Optical Coherence Tomography. Transl Vis Sci Technol 2020; 9:8. [PMID: 33200049 PMCID: PMC7645250 DOI: 10.1167/tvst.9.12.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 10/03/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to design a new parameter "integral" to quantitatively evaluate the spatial cumulative reflectivity of the outer retinal layers in optical coherence tomography (OCT), and to investigate its role in the detection of outer retinal diseases. Methods This was a cross-sectional study. Fovea-centered line OCT scans were performed on 60 eyes of 60 healthy volunteers and 44 eyes of 44 patients diagnosed with outer retinal diseases. The integrals of the ellipsoid zone (EZ) and interdigitation zone (IZ) were measured by respectively accumulating the grayscale values of all the pixels within the EZ and IZ at specified locations on the scanning lines, and were then adjusted by calculating their percentages on the outer retina. The integrals of the EZ and IZ were compared between the two groups. Results The integrals of the EZ and IZ were stably and normally distributed in the healthy eyes, and were significantly lower in eyes with outer retinal lesions than in healthy ones (P < 0.05). Moreover, the integrals of the EZ and IZ were correlated with best corrected visual acuity (BCVA; adjusted R2 = 0.620) and the presence of outer retinal lesions (Nagelkerke R2 = 0.767). The area under the receiver operating characteristic (ROC) curve was 0.954 (95% confidence interval [CI] = 0.918-0.990) when the integral was selected as a diagnostic variable. Conclusions Obtained from this novel quantification method, the new parameter integral was comparable between different individuals and had the potential to detect outer retinal abnormalities in reflectivity through OCT. Translational Relevance Our work verified the feasibility of the new image analysis technique in the detection of the diseases affecting the outer retina.
Collapse
Affiliation(s)
- Junxiang Gu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Tingting Jiang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Mingrong Yu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jian Yu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wenting Li
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Shixue Liu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Peijun Zhang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wenwen Chen
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Qing Chang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital of Fudan University, Fudan University, Shanghai, China.,Key NHC Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
31
|
Rincón Montes V, Gehlen J, Ingebrandt S, Mokwa W, Walter P, Müller F, Offenhäusser A. Development and in vitro validation of flexible intraretinal probes. Sci Rep 2020; 10:19836. [PMID: 33199768 PMCID: PMC7669900 DOI: 10.1038/s41598-020-76582-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/28/2020] [Indexed: 12/01/2022] Open
Abstract
The efforts to improve the treatment efficacy in blind patients with retinal degenerative diseases would greatly benefit from retinal activity feedback, which is lacking in current retinal implants. While the door for a bidirectional communication device that stimulates and records intraretinally has been opened by the recent use of silicon-based penetrating probes, the biological impact induced by the insertion of such rigid devices is still unknown. Here, we developed for the first time, flexible intraretinal probes and validated in vitro the acute biological insertion impact in mouse retinae compared to standard silicon-based probes. Our results show that probes based on flexible materials, such as polyimide and parylene-C, in combination with a narrow shank design 50 µm wide and 7 µm thick, and the use of insertion speeds as high as 187.5 µm/s will successfully penetrate the retina, reduce the footprint of the insertion to roughly 2 times the cross-section of the probe, and induce low dead cell counts, while keeping the vitality of the tissue and recording the neural activity at different depths.
Collapse
Affiliation(s)
- V Rincón Montes
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- RWTH Aachen University, Aachen, Germany
| | - J Gehlen
- Molecular and Cellular Physiology, Institute of Biological Information Processing-1, Forschungszentrum Jülich, Jülich, Germany
| | - S Ingebrandt
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - W Mokwa
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - P Walter
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany
| | - F Müller
- Molecular and Cellular Physiology, Institute of Biological Information Processing-1, Forschungszentrum Jülich, Jülich, Germany
| | - A Offenhäusser
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany.
- RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
32
|
Lavaud A, Soukup P, Martin L, Hartnack S, Pot S. Spectral Domain Optical Coherence Tomography in Awake Rabbits Allows Identification of the Visual Streak, a Comparison with Histology. Transl Vis Sci Technol 2020; 9:13. [PMID: 32821485 PMCID: PMC7401941 DOI: 10.1167/tvst.9.5.13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/13/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose To evaluate visual streak (VS) identification on spectral-domain optical coherence tomography (SD-OCT) scans in awake rabbits. To report thickness measurements in the VS and adjacent retina on OCT B-scans and histologic sections and to assess inter-method bias, precision and repeatability between OCT and histology. Methods Vertical SD-OCT B-scan images through the optic nerve head and VS were acquired from 16 awake, ophthalmologically healthy experimental rabbits. Scans were acquired from both eyes, which were later enucleated and processed for light microscopy. Inner retina, inner nuclear layer, outer nuclear layer, outer retina (OR) and photoreceptor outer segment (PROS) thickness were measured on OCT images and digitalized microscopy slides in- and outside of the VS, and compared using linear mixed effects models. Results Both SD-OCT and histology allowed retinal layer identification and measurement. On OCT, OR and PROS were thickest in the central VS and thinnest outside the VS. Histology mirrored OCT results for central outer retinal layers but shows discrepancies for other layers likely because of postmortem processing artifacts. The method comparison demonstrated better repeatability for OCT measurements compared with histology. Conclusions Increased OR and PROS thickness compared with the adjacent retina allowed identification of the VS on SD-OCT in awake rabbits. OCT allows measurements devoid of processing artifacts in contrast to histology. Translational Relevance SD-OCT is possible in awake rabbits. Easy and reliable identification of the VS may facilitate the positioning and use of rabbits as model species in human macular and generalized retinal disease research.
Collapse
Affiliation(s)
- Arnold Lavaud
- Ophthalmology Section, Equine Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Petr Soukup
- Ophthalmology Section, Equine Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Louise Martin
- Clinic for Zoo Animals, Exotic Pets and Wildlife, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sonja Hartnack
- Section of Epidemiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Simon Pot
- Ophthalmology Section, Equine Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Zhao D, He Z, Wang L, Fortune B, Lim JKH, Wong VHY, Nguyen CTO, Bui BV. Response of the Trilaminar Retinal Vessel Network to Intraocular Pressure Elevation in Rat Eyes. Invest Ophthalmol Vis Sci 2020; 61:2. [PMID: 32031574 PMCID: PMC7325622 DOI: 10.1167/iovs.61.2.2] [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: 01/13/2023] Open
Abstract
Purpose The purpose of this study was to test the hypothesis that the superficial, intermediate, and deep retinal vascular plexus show different responses to intraocular pressure (IOP) elevation. Methods Anesthetized adult Long Evans rats (n = 14) were imaged using optical coherence tomography angiography (OCTA; Spectralis) at baseline (IOP 10 mm Hg) and in follow-up mode to examine the vasculature during IOP elevation (10 to 110 mm Hg, 10 mm Hg steps, each step 3 minutes). A 20° × 10° field was imaged. Vessel density within a 2D projection image was determined (%) for the superficial vascular complex (SVC), intermediate capillary plexus (ICP), and deep capillary plexus (DCP). Comparisons were made between layers using 2-way repeated measures ANOVA (layer versus IOP) following normalization to baseline (% relative to 10 mm Hg). Results The three vascular layers responded differently to IOP elevation. For IOPs between 40 and 60 mm Hg, DCP and ICP capillaries were significantly more resistant to IOP elevation than those in the SVC. When IOP was elevated above 70 mm Hg, all layers showed reduced vessel density. IOP induced change in SVC vessel density closely followed reductions in thickness of the inner retinal layers (nerve fiber, ganglion cell, and inner plexiform layer). This close relationship between reductions in tissue thickness and vessel density was less apparent for the ICP and DCP. Conclusions These data show that the intermediate and deep vascular plexus in the rat retina have a greater capacity for autoregulation against mild IOP elevation but are more affected at high IOP.
Collapse
|
34
|
Ferreira H, Martins J, Nunes A, Moreira PI, Castelo-Branco M, Ambrósio AF, Serranho P, Bernardes R. Characterization of the retinal changes of the 3×Tg-AD mouse model of Alzheimer’s disease. HEALTH AND TECHNOLOGY 2020. [DOI: 10.1007/s12553-020-00413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AbstractAlzheimer’s disease (AD) is a progressive neurodegenerative disorder whose diagnosis remains a notable challenge. The literature suggests that cerebral changes precede AD symptoms by over two decades, implying a significantly advanced stage of AD by the time it is usually diagnosed. In the study herein, texture analysis was applied to computed optical coherence tomography ocular fundus images to identify differences between a group of the transgenic mouse model of the Alzheimer’s disease (3×Tg-AD) and a group of wild-type mice, at the ages of one and two-months-old. A substantial difference between groups was found at both time-points across all neuroretina’s layers. Here, the inner nuclear layer stands out both in the level of statistically significant differences and on the extension of these differences which span through the imaged area. Also, the progression of AD is suggested to be spotted by texture analysis as demonstrated by the significant difference found in the inner plexiform and the outer nuclear layers from the age of one to the age of two-months-old. These findings demonstrate the potential of the use of the retina and texture analysis to the diagnosis of AD and monitor AD progression. Besides, the differences between groups found in this study suggest that the 3×Tg-AD model may be inappropriate to study early changes associated with the AD and other animal models should be tested following the same path and rationale. Moreover, these results also suggest that the human genes present in these transgenic mice may have an impact on the neurodevelopment of offspring which would justify the significant changes found at the age of one-month-old.
Collapse
|
35
|
Sayah DN, Zhou TE, Omri S, Mazzaferri J, Quiniou C, Wirth M, Côté F, Dabouz R, Desjarlais M, Costantino S, Chemtob S. Novel Anti-Interleukin-1β Therapy Preserves Retinal Integrity: A Longitudinal Investigation Using OCT Imaging and Automated Retinal Segmentation in Small Rodents. Front Pharmacol 2020; 11:296. [PMID: 32226385 PMCID: PMC7081735 DOI: 10.3389/fphar.2020.00296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Retinopathy of prematurity (ROP) is the leading cause of blindness in neonates. Inflammation, in particular interleukin-1β (IL-1β), is increased in early stages of the disorder, and contributes to inner and outer retinal vasoobliteration in the oxygen-induced retinopathy (OIR) model of ROP. A small peptide antagonist of IL-1 receptor, composed of the amino acid sequence, rytvela, has been shown to exert beneficial anti-inflammatory effects without compromising immunovigilance-related NF-κB in reproductive tissues. We conducted a longitudinal study to determine the efficacy of “rytvela” in preserving the integrity of the retina in OIR model, using optical coherence tomography (OCT) which provides high-resolution cross-sectional imaging of ocular structures in vivo. Sprague–Dawley rats subjected to OIR and treated or not with “rytvela” were compared to IL-1 receptor antagonist (Kineret). OCT imaging and custom automated segmentation algorithm used to measure retinal thickness (RT) were obtained at P14 and P30; gold-standard immunohistochemistry (IHC) was used to confirm retinal anatomical changes. OCT revealed significant retinal thinning in untreated animals by P30, confirmed by IHC; these changes were coherently associated with increased apoptosis. Both rytvela and Kineret subsided apoptosis and preserved RT. As anticipated, Kineret diminished both SAPK/JNK and NF-κB axes, whereas rytvela selectively abated the former which resulted in preserved monocyte phagocytic function. Altogether, OCT imaging with automated segmentation is a reliable non-invasive approach to study longitudinally retinal pathology in small animal models of retinopathy.
Collapse
Affiliation(s)
- Diane N Sayah
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Tianwei E Zhou
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Samy Omri
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | - Christiane Quiniou
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Maëlle Wirth
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Rabah Dabouz
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Michel Desjarlais
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Santiago Costantino
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Sylvain Chemtob
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
36
|
Yang JM, Park CS, Kim SH, Noh TW, Kim JH, Park S, Lee J, Park JR, Yoo D, Jung HH, Takase H, Shima DT, Schwaninger M, Lee S, Kim IK, Lee J, Ji YS, Jon S, Oh WY, Kim P, Uemura A, Ju YS, Kim I. Dll4 Suppresses Transcytosis for Arterial Blood-Retinal Barrier Homeostasis. Circ Res 2020; 126:767-783. [PMID: 32078435 DOI: 10.1161/circresaha.119.316476] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
RATIONALE Central nervous system has low vascular permeability by organizing tight junction (TJ) and limiting endothelial transcytosis. While TJ has long been considered to be responsible for vascular barrier in central nervous system, suppressed transcytosis in endothelial cells is now emerging as a complementary mechanism. Whether transcytosis regulation is independent of TJ and its dysregulation dominantly causes diseases associated with edema remain elusive. Dll4 signaling is important for various vascular contexts, but its role in the maintenance of vascular barrier in central nervous system remains unknown. OBJECTIVE To find a TJ-independent regulatory mechanism selective for transcytosis and identify its dysregulation as a cause of pathological leakage. METHODS AND RESULTS We studied transcytosis in the adult mouse retina with low vascular permeability and employed a hypertension-induced retinal edema model for its pathological implication. Both antibody-based and genetic inactivation of Dll4 or Notch1 induce hyperpermeability by increasing transcytosis without junctional destabilization in arterial endothelial cells, leading to nonhemorrhagic leakage predominantly in the superficial retinal layer. Endothelial Sox17 deletion represses Dll4 in retinal arteries, phenocopying Dll4 blocking-driven vascular leakage. Ang II (angiotensin II)-induced hypertension represses arterial Sox17 and Dll4, followed by transcytosis-driven retinal edema, which is rescued by a gain of Notch activity. Transcriptomic profiling of retinal endothelial cells suggests that Dll4 blocking activates SREBP1 (sterol regulatory element-binding protein 1)-mediated lipogenic transcription and enriches gene sets favorable for caveolae formation. Profiling also predicts the activation of VEGF (vascular endothelial growth factor) signaling by Dll4 blockade. Inhibition of SREBP1 or VEGF-VEGFR2 (VEGF receptor 2) signaling attenuates both Dll4 blockade-driven and hypertension-induced retinal leakage. CONCLUSIONS In the retina, Sox17-Dll4-SREBP1 signaling axis controls transcytosis independently of TJ in superficial arteries among heterogeneous regulations for the whole vessels. Uncontrolled transcytosis via dysregulated Dll4 underlies pathological leakage in hypertensive retina and could be a therapeutic target for treating hypertension-associated retinal edema.
Collapse
Affiliation(s)
- Jee Myung Yang
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Chan Soon Park
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Soo Hyun Kim
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Tae Wook Noh
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Ju-Hee Kim
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Seongyeol Park
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Jingu Lee
- Graduate School of Nanoscience and Technology (J.L., P.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,KAIST Institute for Health Science and Technology (J.L., J.R.P., W.-Y.O., P.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Jang Ryul Park
- KAIST Institute for Health Science and Technology (J.L., J.R.P., W.-Y.O., P.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,Mechanical Engineering (J.R.P., W.-Y.O.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Dohyun Yoo
- Biological Sciences (D.Y., S.J.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,KAIST Institute for the BioCentury (D.Y., S.J.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Hyun Ho Jung
- Ophthalmology, Chonnam National University Medical School and Hospital, Republic of Korea (H.H.J., Y.-S.J.)
| | - Hiroshi Takase
- Core Laboratory (H.T.), Nagoya City University Graduate School of Medical Sciences, Japan
| | - David T Shima
- Institute of Ophthalmology, University College London, United Kingdom (D.T.S.)
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany (M.S.)
| | - Seungjoo Lee
- Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea (S.L.)
| | - Il-Kug Kim
- Plastic and Reconstructive Surgery (I.-K.K.), Yeungnam University College of Medicine, Republic of Korea
| | - Junyeop Lee
- Ophthalmology (J.L.), Yeungnam University College of Medicine, Republic of Korea
| | - Yong-Sok Ji
- Ophthalmology, Chonnam National University Medical School and Hospital, Republic of Korea (H.H.J., Y.-S.J.)
| | - Sangyong Jon
- Biological Sciences (D.Y., S.J.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,KAIST Institute for the BioCentury (D.Y., S.J.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Wang-Yuhl Oh
- KAIST Institute for Health Science and Technology (J.L., J.R.P., W.-Y.O., P.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,Mechanical Engineering (J.R.P., W.-Y.O.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Pilhan Kim
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,Graduate School of Nanoscience and Technology (J.L., P.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon.,Mechanical Engineering (J.R.P., W.-Y.O.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Akiyoshi Uemura
- Retinal Vascular Biology (A.U.), Nagoya City University Graduate School of Medical Sciences, Japan
| | - Young Seok Ju
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| | - Injune Kim
- From the Graduate School of Medical Science and Engineering (J.M.Y., C.S.P., S.H.K., T.W.N., J.-H.K., S.P., P.K., Y.S.J., I.K.), Korea Advanced Institute of Science and Technology (KAIST), Daejeon
| |
Collapse
|
37
|
Chan K, Hoon M, Pattnaik BR, Ver Hoeve JN, Wahlgren B, Gloe S, Williams J, Wetherbee B, Kiland JA, Vogel KR, Jansen E, Salomons G, Walters D, Roullet JB, Gibson K M, McLellan GJ. Vigabatrin-Induced Retinal Functional Alterations and Second-Order Neuron Plasticity in C57BL/6J Mice. Invest Ophthalmol Vis Sci 2020; 61:17. [PMID: 32053727 PMCID: PMC7326505 DOI: 10.1167/iovs.61.2.17] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Vigabatrin (VGB) is an effective antiepileptic that increases concentrations of inhibitory γ-aminobutyric acid (GABA) by inhibiting GABA transaminase. Reports of VGB-associated visual field loss limit its clinical usefulness, and retinal toxicity studies in laboratory animals have yielded conflicting results. Methods We examined the functional and morphologic effects of VGB in C57BL/6J mice that received either VGB or saline IP from 10 to 18 weeks of age. Retinal structure and function were assessed in vivo by optical coherence tomography (OCT), ERG, and optomotor response. After euthanasia, retinas were processed for immunohistochemistry, and retinal GABA, and VGB quantified by mass spectrometry. Results No significant differences in visual acuity or total retinal thickness were identified between groups by optomotor response or optical coherence tomography, respectively. After 4 weeks of VGB treatment, ERG b-wave amplitude was enhanced, and amplitudes of oscillatory potentials were reduced. Dramatic rod and cone bipolar and horizontal cell remodeling, with extension of dendrites into the outer nuclear layer, was observed in retinas of VGB-treated mice. VGB treatment resulted in a mean 3.3-fold increase in retinal GABA concentration relative to controls and retinal VGB concentrations that were 20-fold greater than brain. Conclusions No evidence of significant retinal thinning or ERG a- or b-wave deficits were apparent, although we describe significant alterations in ERG b-wave and oscillatory potentials and in retinal cell morphology in VGB-treated C57BL/6J mice. The dramatic concentration of VGB in retina relative to the target tissue (brain), with a corresponding increase in retinal GABA, offers insight into the pathophysiology of VGB-associated visual field loss.
Collapse
Affiliation(s)
- Kore Chan
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Mrinalini Hoon
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Bikash R. Pattnaik
- McPherson Eye Research Institute, Madison, Wisconsin, United States
- Pediatrics Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - James N. Ver Hoeve
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Brad Wahlgren
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Shawna Gloe
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Jeremy Williams
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Brenna Wetherbee
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Julie A. Kiland
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kara R. Vogel
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Erwin Jansen
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Gajja Salomons
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Dana Walters
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - Jean-Baptiste Roullet
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - K Michael Gibson
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - Gillian J. McLellan
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| |
Collapse
|
38
|
Mesentier-Louro LA, Shariati MA, Dalal R, Camargo A, Kumar V, Shamskhou EA, de Jesus Perez V, Liao YJ. Systemic hypoxia led to little retinal neuronal loss and dramatic optic nerve glial response. Exp Eye Res 2020; 193:107957. [PMID: 32032627 PMCID: PMC7673281 DOI: 10.1016/j.exer.2020.107957] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/19/2019] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
Vision loss is a devastating consequence of systemic hypoxia, but the cellular mechanisms are unclear. We investigated the impact of acute hypoxia in the retina and optic nerve. We induced systemic hypoxia (10% O2) in 6-8w mice for 48 h and performed in vivo imaging using optical coherence tomography (OCT) at baseline and after 48 h to analyze structural changes in the retina and optic nerve. We analyzed glial cellular and molecular changes by histology and immunofluorescence and the impact of pretreatment with 4-phenylbutyric acid (4-PBA) in oligodendroglia survival. After 48 h hypoxia, we found no change in ganglion cell complex thickness and no loss of retinal ganglion cells. Despite this, there was significantly increased expression of CCAAT-enhancer-binding protein homologous protein (CHOP), a marker of endoplasmic reticulum stress, in the retina and optic nerve. In addition, hypoxia induced obvious increase of GFAP expression in the anterior optic nerve, where it co-localized with CHOP, and significant loss of Olig2+ oligodendrocytes. Pretreatment with 4-PBA, which has been shown to reduce endoplasmic reticulum stress, rescued total Olig2+ oligodendrocytes and increased the pool of mature (CC-1+) but not of immature (PDGFRa+) oligodendrocytes. Consistent with a selective vulnerability of the retina and optic nerve in hypoxia, the most striking changes in the 48 h murine model of hypoxia were in glial cells in the optic nerve, including increased CHOP expression in the astrocytes and loss of oligodendrocytes. Our data support a model where glial dysfunction is among the earliest events in systemic hypoxia - suggesting that glia may be a novel target in treatment of hypoxia.
Collapse
Affiliation(s)
| | - Mohammed Ali Shariati
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Roopa Dalal
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Alexandra Camargo
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Varun Kumar
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Elya Ali Shamskhou
- Department of Pulmonary Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Vinicio de Jesus Perez
- Department of Pulmonary Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, CA, USA; Department of Neurology, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
39
|
Harper DJ, Augustin M, Lichtenegger A, Gesperger J, Himmel T, Muck M, Merkle CW, Eugui P, Kummer S, Woehrer A, Glösmann M, Baumann B. Retinal analysis of a mouse model of Alzheimer's disease with multicontrast optical coherence tomography. NEUROPHOTONICS 2020; 7:015006. [PMID: 32042855 PMCID: PMC6999077 DOI: 10.1117/1.nph.7.1.015006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/07/2020] [Indexed: 05/18/2023]
Abstract
Significance. Recent Alzheimer's disease (AD) patient studies have focused on retinal analysis, as the retina is the only part of the central nervous system that can be imaged noninvasively by optical methods. However, as this is a relatively new approach, the occurrence and role of retinal pathological features are still debated. Aim. The retina of an APP/PS1 mouse model was investigated using multicontrast optical coherence tomography (OCT) in order to provide a documentation of what was observed in both transgenic and wild-type mice. Approach. Both eyes of 24 APP/PS1 transgenic mice (age: 45 to 104 weeks) and 15 age-matched wild-type littermates were imaged by the custom-built OCT system. At the end of the experiment, retinas and brains were harvested from a subset of the mice (14 transgenic, 7 age-matched control) in order to compare the in vivo results to histological analysis and to quantify the cortical amyloid beta plaque load. Results. The system provided a combination of standard reflectivity data, polarization-sensitive data, and OCT angiograms. Qualitative and quantitative information from the resultant OCT images was extracted on retinal layer thickness and structure, presence of hyper-reflective foci, phase retardation abnormalities, and retinal vasculature. Conclusions. Although multicontrast OCT revealed abnormal structural properties and phase retardation signals in the retina of this APP/PS1 mouse model, the observations were very similar in transgenic and control mice.
Collapse
Affiliation(s)
- Danielle J. Harper
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
- Address all correspondence to Danielle J. Harper, E-mail:
| | - Marco Augustin
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Antonia Lichtenegger
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Johanna Gesperger
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
- General Hospital and Medical University of Vienna, Institute of Neurology, Vienna, Austria
| | - Tanja Himmel
- University of Veterinary Medicine, Institute of Pathology, Vienna, Austria
| | - Martina Muck
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Conrad W. Merkle
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Pablo Eugui
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Stefan Kummer
- University of Veterinary Medicine, Core Facility for Research and Technology, Vienna, Austria
| | - Adelheid Woehrer
- General Hospital and Medical University of Vienna, Institute of Neurology, Vienna, Austria
| | - Martin Glösmann
- University of Veterinary Medicine, Core Facility for Research and Technology, Vienna, Austria
| | - Bernhard Baumann
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| |
Collapse
|
40
|
Cruz-Herranz A, Dietrich M, Hilla AM, Yiu HH, Levin MH, Hecker C, Issberner A, Hallenberger A, Cordano C, Lehmann-Horn K, Balk LJ, Aktas O, Ingwersen J, von Gall C, Hartung HP, Zamvil SS, Fischer D, Albrecht P, Green AJ. Monitoring retinal changes with optical coherence tomography predicts neuronal loss in experimental autoimmune encephalomyelitis. J Neuroinflammation 2019; 16:203. [PMID: 31684959 PMCID: PMC6827223 DOI: 10.1186/s12974-019-1583-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background Retinal optical coherence tomography (OCT) is a clinical and research tool in multiple sclerosis, where it has shown significant retinal nerve fiber (RNFL) and ganglion cell (RGC) layer thinning, while postmortem studies have reported RGC loss. Although retinal pathology in experimental autoimmune encephalomyelitis (EAE) has been described, comparative OCT studies among EAE models are scarce. Furthermore, the best practices for the implementation of OCT in the EAE lab, especially with afoveate animals like rodents, remain undefined. We aimed to describe the dynamics of retinal injury in different mouse EAE models and outline the optimal experimental conditions, scan protocols, and analysis methods, comparing these to histology to confirm the pathological underpinnings. Methods Using spectral-domain OCT, we analyzed the test-retest and the inter-rater reliability of volume, peripapillary, and combined horizontal and vertical line scans. We then monitored the thickness of the retinal layers in different EAE models: in wild-type (WT) C57Bl/6J mice immunized with myelin oligodendrocyte glycoprotein peptide (MOG35–55) or with bovine myelin basic protein (MBP), in TCR2D2 mice immunized with MOG35–55, and in SJL/J mice immunized with myelin proteolipid lipoprotein (PLP139–151). Strain-matched control mice were sham-immunized. RGC density was counted on retinal flatmounts at the end of each experiment. Results Volume scans centered on the optic disc showed the best reliability. Retinal changes during EAE were localized in the inner retinal layers (IRLs, the combination of the RNFL and the ganglion cell plus the inner plexiform layers). In WT, MOG35–55 EAE, progressive thinning of IRL started rapidly after EAE onset, with 1/3 of total loss occurring during the initial 2 months. IRL thinning was associated with the degree of RGC loss and the severity of EAE. Sham-immunized SJL/J mice showed progressive IRL atrophy, which was accentuated in PLP-immunized mice. MOG35–55-immunized TCR2D2 mice showed severe EAE and retinal thinning. MBP immunization led to very mild disease without significant retinopathy. Conclusions Retinal neuroaxonal damage develops quickly during EAE. Changes in retinal thickness mirror neuronal loss and clinical severity. Monitoring of the IRL thickness after immunization against MOG35–55 in C57Bl/6J mice seems the most convenient model to study retinal neurodegeneration in EAE.
Collapse
Affiliation(s)
- Andrés Cruz-Herranz
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hao H Yiu
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Marc H Levin
- Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.,Department of Ophthalmology, Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Christian Cordano
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Klaus Lehmann-Horn
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lisanne J Balk
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Scott S Zamvil
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA.,Program in Immunology, University of California, San Francisco, San Francisco, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Ari J Green
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA. .,Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
41
|
Deshpande G, Bawankule P, Raje D, Chakraborty M, Gupta R. Structural evaluation of perimetrically normal and affected hemifields in open angle glaucoma. Indian J Ophthalmol 2019; 67:1657-1662. [PMID: 31546503 PMCID: PMC6786176 DOI: 10.4103/ijo.ijo_1755_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Purpose To study macular ganglion cell layer--inner plexiform layer complex (GCL + IPL) in relation to peripapillary retinal nerve fiber layer (RNFL) in glaucomatous eyes with superior or inferior hemifield defects (HD) and to study structural configuration in normal hemifield. Methods This was an observational cross-sectional study. Data from consecutive 45 superior HD (SHD) and 50 inferior HD (IHD) eyes were analyzed. Each patient underwent detailed ocular examination, standard automated perimetry, and spectral domain optical coherence tomography (SD-OCT). After adjusting for age, gender, and signal strength, area under receiver operating characteristic curve (AUC) was calculated to determine diagnostic ability of GCL + IPL and peripapillary RNFL. Apparently normal hemifield was compared with true normal hemifield. Data were analyzed with SPSS, analysis of variance, t-test, Chi-square test, and receiver operating curve. Results In the SHD glaucoma group, best parameters for discriminating normal eyes from glaucomatous eyes were inferotemporal GCL + IPL thickness (0.935) and inferior quadrant RNFL thickness (0.971). For IHD glaucoma, average GCL + IPL thickness (0.877) and average RNFL thickness (0.950) had best AUC values. When evaluating apparently normal hemifield in both groups, statistically significant difference was found in inferior GCL + IPL sector (0.865) and inferior quadrant RNFL (0.883) in IHD and superonasal GCL + IPL (0.725) and superior quadrant RNFL (0.842) in SHD groups. Conclusion SD-OCT may be a useful ancillary diagnostic tool for evaluation of early macular and circumpapillary structural changes in glaucomatous eyes with localized visual field defects. Apparently normal hemifields show structural damage and should be considered in management of glaucoma.
Collapse
Affiliation(s)
- Gunjan Deshpande
- Department of Glaucoma, Sarakshi Netralaya, Nagpur, Maharashtra, India
| | | | - Dhananjay Raje
- Department of Glaucoma, Sarakshi Netralaya, Nagpur, Maharashtra, India
| | | | - Richa Gupta
- Department of Glaucoma, Sarakshi Netralaya, Nagpur, Maharashtra, India
| |
Collapse
|
42
|
Soukup P, Maloca P, Altmann B, Festag M, Atzpodien EA, Pot S. Interspecies Variation of Outer Retina and Choriocapillaris Imaged With Optical Coherence Tomography. ACTA ACUST UNITED AC 2019; 60:3332-3342. [DOI: 10.1167/iovs.18-26257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Petr Soukup
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
- Ophthalmology Section, Equine Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Peter Maloca
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- OCTlab Research Laboratory, Department of Ophthalmology, University of Basel, Basel, Switzerland
- Moorfields Eye Hospital, London, United Kingdom
| | - Bernd Altmann
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Matthias Festag
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Elke-Astrid Atzpodien
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Simon Pot
- Ophthalmology Section, Equine Department, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Rincón Montes V, Gehlen J, Lück S, Mokwa W, Müller F, Walter P, Offenhäusser A. Toward a Bidirectional Communication Between Retinal Cells and a Prosthetic Device - A Proof of Concept. Front Neurosci 2019; 13:367. [PMID: 31114470 PMCID: PMC6502975 DOI: 10.3389/fnins.2019.00367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/01/2019] [Indexed: 01/15/2023] Open
Abstract
Background: Significant progress toward the recovery of useful vision in blind patients with severe degenerative retinal diseases caused by photoreceptor death has been achieved with the development of visual prostheses that stimulate the retina electrically. However, currently used prostheses do not provide feedback about the retinal activity before and upon stimulation and do not adjust to changes during the remodeling processes in the retina. Both features are desirable to improve the efficiency of the electrical stimulation (ES) therapy offered by these devices. Accordingly, devices that not only enable ES but at the same time provide information about the retinal activity are beneficial. Given the above, a bidirectional communication strategy, in which inner retinal cells are stimulated and the output neurons of the retina, the ganglion cells, are recorded using penetrating microelectrode arrays (MEAs) is proposed. Methods: Custom-made penetrating MEAs with four silicon-based shanks, each one with three or four iridium oxide electrodes specifically designed to match retinal dimensions were used to record the activity of light-adapted wildtype mice retinas and degenerated retinas from rd10 mice in vitro. In addition, responses to high potassium concentration and to light stimulation in wildtype retinas were examined. Furthermore, voltage-controlled ES was performed. Results: The spiking activity of retinal ganglion cells (RGCs) was recorded at different depths of penetration inside the retina. Physiological responses during an increase of the extracellular potassium concentration and phasic and tonic responses during light stimulation were captured. Moreover, pathologic rhythmic activity was recorded from degenerated retinas. Finally, ES of the inner retina and simultaneous recording of the activity of RGCs was accomplished. Conclusion: The access to different layers of the retina with penetrating electrodes while recording at the same time the spiking activity of RGCs broadens the use and the field of action of multi-shank and multi-site penetrating MEAs for retinal applications. It enables a bidirectional strategy to stimulate inner retinal cells electrically and to record from the spiking RGCs simultaneously (BiMEA). This opens the possibility of a feedback loop system to acknowledge the success of ES carried out by retinal prostheses.
Collapse
Affiliation(s)
- Viviana Rincón Montes
- Bioelectronics, Institute of Complex Systems-8, Forschungszentrum Jülich, Jülich, Germany
| | - Jana Gehlen
- Cellular Biophysics, Institute of Complex Systems-4, Forschungszentrum Jülich, Jülich, Germany
| | - Stefan Lück
- Department of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - Wilfried Mokwa
- Department of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - Frank Müller
- Cellular Biophysics, Institute of Complex Systems-4, Forschungszentrum Jülich, Jülich, Germany
| | - Peter Walter
- Department of Ophthalmology, RWTH Aachen University, Aachen, Germany
| | - Andreas Offenhäusser
- Bioelectronics, Institute of Complex Systems-8, Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
44
|
Orduña Ríos M, Noguez Imm R, Hernández Godínez NM, Bautista Cortes AM, López Escalante DD, Liedtke W, Martínez Torres A, Concha L, Thébault S. TRPV4 inhibition prevents increased water diffusion and blood-retina barrier breakdown in the retina of streptozotocin-induced diabetic mice. PLoS One 2019; 14:e0212158. [PMID: 31048895 PMCID: PMC6497373 DOI: 10.1371/journal.pone.0212158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 01/02/2023] Open
Abstract
A better understanding of the molecular and cellular mechanisms involved in retinal hydro-mineral homeostasis imbalance during diabetic macular edema (DME) is needed to gain insights into retinal (patho-)physiology that will help elaborate innovative therapies with lower health care costs. Transient receptor potential cation channel subfamily vanilloid member 4 (TRPV4) plays an intricate role in homeostatic processes that needs to be deciphered in normal and diabetic retina. Based on previous findings showing that TRPV4 antagonists resolve blood-retina barrier (BRB) breakdown in diabetic rats, we evaluated whether TRPV4 channel inhibition prevents and reverts retinal edema in streptozotocin(STZ)-induced diabetic mice. We assessed retinal edema using common metrics, including retinal morphology/thickness (histology) and BRB integrity (albumin-associated tracer), and also by quantifying water mobility through apparent diffusion coefficient (ADC) measures. ADC was measured by diffusion-weighted magnetic resonance imaging (DW-MRI), acquired ex vivo at 4 weeks after STZ injection in diabetes and control groups. DWI images were also used to assess retinal thickness. TRPV4 was genetically ablated or pharmacologically inhibited as follows: left eyes were used as vehicle control and right eyes were intravitreally injected with TRPV4-selective antagonist GSK2193874, 24 h before the end of the 4 weeks of diabetes. Histological data show that retinal thickness was similar in nondiabetic and diabetic wt groups but increased in diabetic Trpv4-/- mice. In contrast, DWI shows retinal thinning in diabetic wt mice that was absent in diabetic Trpv4-/- mice. Disorganized outer nuclear layer was observed in diabetic wt but not in diabetic Trpv4-/- retinas. We further demonstrate increased water diffusion, increased distances between photoreceptor nuclei, reduced nuclear area in all nuclear layers, and BRB hyperpermeability, in diabetic wt mice, effects that were absent in diabetic Trpv4-/- mice. Retinas of diabetic mice treated with PBS showed increased water diffusion that was not normalized by GSK2193874. ADC maps in nondiabetic Trpv4-/- mouse retinas showed restricted diffusion. Our data provide evidence that water diffusion is increased in diabetic mouse retinas and that TRPV4 function contributes to retinal hydro-mineral homeostasis and structure under control conditions, and to the development of BRB breakdown and increased water diffusion in the retina under diabetes conditions. A single intravitreous injection of TRPV4 antagonist is however not sufficient to revert these alterations in diabetic mouse retinas.
Collapse
Affiliation(s)
- Maricruz Orduña Ríos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Ramsés Noguez Imm
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Ana María Bautista Cortes
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Wolfgang Liedtke
- Department of Medicine and Neurobiology, Center for Translational Neuroscience, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Atáulfo Martínez Torres
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Luis Concha
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Stéphanie Thébault
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
- * E-mail:
| |
Collapse
|
45
|
Lian Q, Zhao M, Li T, Wu K, Zhu D, Shang B, Mei T, Li W, Lin Y, Mao F, Liu Y, Liu C, Lu L, Zhao L. In vivo detecting mouse persistent hyperplastic primary vitreous by Spectralis Optical Coherence Tomography. Exp Eye Res 2019; 181:271-276. [PMID: 30817926 DOI: 10.1016/j.exer.2019.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/15/2019] [Accepted: 02/20/2019] [Indexed: 10/27/2022]
Abstract
To identify imaging characteristics of mouse persistent hyperplastic primary vitreous (PHPV) by Spectralis Optical Coherence Tomography (OCT), as well as to assess and compare the sensitivity and precision of OCT with color photography (CP) and Fundus Fluorescein Angiography (FFA) imaging in detecting mouse PHPV. Notch4-/- C57BL/6J mice (224 eyes) aged from 3 months to 7 months were examined in this study. CP, FFA and OCT imaging were utilized to examine vitreous cavity and retina of mouse eyes. Horizontal and radial OCT scan volume was centered on the optic nerve head. Hematoxylin and eosin (H&E) staining was performed to validate PHPV. For color photography and FFA imaging, retrolental irregular fibrovascular membrane-like tissues were found in 33 eyes with/without blood vessels in vitreous cavity. Among them, 31 eyes were visualized with lateral and oblique linear hyperreflective opacities in vitreous cavity using Spectralis OCT. Position of PHPV in posterior segment of eyes was also measured via OCT. Mouse PHPV was validated by H&E staining. Typical hyperreflective opacities in vitreous cavity were detected in PHPV mouse using Spectralis OCT. Spectralis OCT imaging can effectively detect mouse PHPV as color photography and FFA.
Collapse
Affiliation(s)
- Qing Lian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Tianzhong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Keling Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Deliang Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bizhi Shang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Tingfang Mei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Fuxiang Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chujun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
46
|
Mori K, Kurihara T, Miyauchi M, Ishida A, Jiang X, Ikeda SI, Torii H, Tsubota K. Oral crocetin administration suppressed refractive shift and axial elongation in a murine model of lens-induced myopia. Sci Rep 2019; 9:295. [PMID: 30670743 PMCID: PMC6343000 DOI: 10.1038/s41598-018-36576-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 11/23/2018] [Indexed: 11/09/2022] Open
Abstract
Increased global incidence of myopia necessitates establishment of therapeutic approaches against its progression. To explore agents which may control myopia, we screened 207 types of natural compounds and chemical reagents based on an activity of a myopia suppressive factor, early growth response protein 1 (Egr-1) in vitro. Among the candidates, crocetin showed the highest and dose-dependent activation of Egr-1. For in vivo analysis, experimental myopia was induced in 3-week-old C57BL/6 J mice with −30 diopter (D) lenses for 3 weeks. Animals were fed with normal or mixed chow containing 0.003% (n = 19) and 0.03% (n = 7) of crocetin during myopia induction. Refraction and axial length were measured at 3-week-old and the 6-week-old with an infrared photorefractor and a SD-OCT system. Compared to controls (n = 14), crocetin administration showed a significant smaller change of refractive errors (−13.62 ± 8.14 vs +0.82 ± 5.81 D for 0.003%, p < 0.01, −2.00 ± 4.52 D for 0.03%, p < 0.01) and axial elongation (0.27 ± 0.03 vs 0.22 ± 0.04 mm for 0.003%, p < 0.01, 0.23 ± 0.05 mm for 0.03%, p < 0.05). These results suggest that a dietary factor crocetin may have a preventive effect against myopia progression.
Collapse
Affiliation(s)
- Kiwako Mori
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Maki Miyauchi
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ayako Ishida
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Xiaoyan Jiang
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shin-Ichi Ikeda
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidemasa Torii
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
47
|
Nishioka C, Liang HF, Barsamian B, Sun SW. Sequential phases of RGC axonal and somatic injury in EAE mice examined using DTI and OCT. Mult Scler Relat Disord 2018; 27:315-323. [PMID: 30469023 DOI: 10.1016/j.msard.2018.11.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Clinical imaging modalities including optical coherence tomography (OCT) and diffusion tensor imaging (DTI) are vital in Multiple Sclerosis (MS), but their relationships during the different phases of Retinal ganglion cell (RGC) degeneration are not clear. We hypothesize that initial injury in optic nerve causes axonal degeneration leading to RGC loss in retina, which can be characterized by a combination of DTI and OCT. Our objective was to examine the correlation between noninvasive and histological data to chronicle the degeneration profile of RGCs in the retina and optic nerve in a mouse model of MS. MATERIALS AND METHODS Experimental Autoimmune Encephalomyelitis (EAE) was induced in 11 C57Bl/6 mice, with 8 mice reserved as controls. OCT and DTI was conducted 2-8 weeks after induction of EAE. The thickness of the retinal ganglion cell complex (GCC) was measured using OCT and compared to DTI indices measured in optic nerves. End-stage histology was used to quantify axon/myelin loss in the optic nerve and retinal thinning/RGC loss in the retina. RESULTS Significant changes in DTI-derived Axial Diffusivity (AD, -17.2%) and Trace Diffusivity (TR, -18.3%) began after 2 weeks of EAE. Later significant reductions in Fractional Anisotropy (FA) and AD, with increases in Radial Diffusion (RD) were apparent after 4 and 8 weeks. OCT-derived measures of GCC thickness were reduced after 4 weeks, and reached significant reduction after 8 weeks. Among EAE mice, DTI (FA, AD and RD measures) and OCT measures were all significantly correlated after 4 and 8 weeks. Among histology measures, RGC density (-23%), RGC size (-27%), and the number of SMI31+ axons (-54%) were reduced significantly. DTI measures of FA and AD along with GCC thinning were the best independent predictors of axon loss. CONCLUSIONS DTI and OCT measures are tightly correlated during the chronic phase of axonal degeneration (4-8 weeks) in EAE mice. After 8 weeks of EAE, both OCT and DTI measures are strong predictors of axon loss in the Optic Nerve.
Collapse
Affiliation(s)
- Christopher Nishioka
- Basic Sciences, School of Medicine, Loma Linda University, CA, United States; Neuroscience Graduate Program, University of California, Riverside, United States
| | - Hsiao-Fang Liang
- Basic Sciences, School of Medicine, Loma Linda University, CA, United States; Pharmaceutical Science, School of Pharmacy, Loma Linda University, CA, United States
| | - Barsam Barsamian
- Basic Sciences, School of Medicine, Loma Linda University, CA, United States; Neuroscience Graduate Program, University of California, Riverside, United States
| | - Shu-Wei Sun
- Basic Sciences, School of Medicine, Loma Linda University, CA, United States; Neuroscience Graduate Program, University of California, Riverside, United States; Pharmaceutical Science, School of Pharmacy, Loma Linda University, CA, United States.
| |
Collapse
|
48
|
Butler MC, Sullivan JM. Ultrahigh Resolution Mouse Optical Coherence Tomography to Aid Intraocular Injection in Retinal Gene Therapy Research. J Vis Exp 2018. [PMID: 30451216 DOI: 10.3791/55894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
HR-SD-OCT is utilized to monitor the progression of photoreceptor degeneration in live mouse models, assess the delivery of therapeutic agents into the subretinal space, and to evaluate toxicity and efficacy in vivo. HR-SD-OCT uses near infrared light (800-880 nm) and has optics specifically designed for the unique optics of the mouse eye with sub-2-micron axial resolution. Transgenic mouse models of outer retinal (photoreceptor) degeneration and controls were imaged to assess the disease progression. Pulled glass microneedles were used to deliver sub retinal injections of adeno-associated virus (AAV) or nanoparticles (NP) via a trans-scleral and trans-choroidal approach. Careful positioning of the needle into the subretinal space was required prior to a calibrated pressure injection, which delivers fluid into the sub retinal space. Real time subretinal surgery was conducted on our retinal imaging system (RIS). HR-SD-OCT demonstrated progressive uniform retinal degeneration due to expression of a toxic mutant human mutant rhodopsin (P347S) (RHOP347S) transgene in mice. HR-SD-OCT allows rigorous quantification of all the retinal layers. Outer nuclear layer (ONL) thickness and photoreceptor outer segment length (OSL) measurements correlate with photoreceptor vitality, degeneration, or rescue. The RIS delivery system allows real-time visualization of subretinal injections in neonatal (~P10-14) or adult mice, and HR-SD-OCT immediately determines success of delivery and maps areal extent. HR-SD-OCT is a powerful tool that can evaluate the success of subretinal surgery in mice, in addition to measuring vitality of photoreceptors in vivo. HR-SD-OCT can also be used to identify uniform animal cohorts to evaluate the extent of retinal degeneration, toxicity, and therapeutic rescue in preclinical gene therapy research studies.
Collapse
Affiliation(s)
- Mark C Butler
- Research Service, VA Western New York Healthcare System; Department of Ophthalmology, (Ross Eye Institute), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY
| | - Jack M Sullivan
- Research Service, VA Western New York Healthcare System; Department of Ophthalmology, (Ross Eye Institute), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Pharmacology/Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Physiology/Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; The RNA Institute, University at Buffalo- SUNY; The SUNY Eye Institute;
| |
Collapse
|
49
|
Xie W, Zhao M, Tsai SH, Burkes WL, Potts LB, Xu W, Payne HR, Hein TW, Kuo L, Rosa RH. Correlation of spectral domain optical coherence tomography with histology and electron microscopy in the porcine retina. Exp Eye Res 2018; 177:181-190. [PMID: 30120928 DOI: 10.1016/j.exer.2018.08.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Spectral domain optical coherence tomography (SD-OCT) is used as a non-invasive tool for retinal morphological assessment in vivo. Information on the correlation of SD-OCT with retinal histology in the porcine retina, a model resembling the human retina, is limited. Herein, we correlated the hypo- and hyper-reflective bands on SD-OCT with histology of the lamellar architecture and cellular constituents of the porcine retina. SD-OCT images were acquired with the Heidelberg Spectralis HRA + OCT. Histological analysis was performed using epoxy resin embedded tissue and transmission electron microscopy. Photomicrographs from the histologic sections were linearly scaled to correct for tissue shrinkage and correlated with SD-OCT images. SD-OCT images correlated well with histomorphometric data. A hyper-reflective band in the mid-to-outer inner nuclear layer correlated with the presence of abundant mitochondria in horizontal cell processes and adjacent bipolar cells. A concentration of cone nuclei corresponded to a relative hypo-reflective band in the outer portion of the outer nuclear layer. The presence of 3 hyper-reflective bands in the outer retina corresponded to: 1) the external limiting membrane; 2) the cone and rod ellipsoid zones; and 3) the interdigitation zone of photoreceptor outer segments/retinal pigment epithelium (RPE) apical cell processes and the RPE. These correlative and normative SD-OCT data may be employed to characterize and assess the in vivo histologic changes in retinal vascular and degenerative diseases and the responses to novel therapeutic interventions in this large animal model.
Collapse
Affiliation(s)
- Wankun Xie
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA; Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA
| | - Min Zhao
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA; Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA
| | - Shu-Huai Tsai
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA
| | - William L Burkes
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA
| | - Luke B Potts
- Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA
| | - Wenjuan Xu
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA
| | - H Ross Payne
- Image Analysis Laboratory, Texas A&M University College of Veterinary Medicine, College Station, TX, USA
| | - Travis W Hein
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA; Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA
| | - Lih Kuo
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA; Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA
| | - Robert H Rosa
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, TX, USA; Department of Ophthalmology and Ophthalmic Vascular Research Program, Scott & White Eye Institute, Temple, TX, USA.
| |
Collapse
|
50
|
Huang F, Zhang L, Wang Q, Yang Y, Li Q, Wu Y, Chen J, Qu J, Zhou X. Dopamine D1 Receptors Contribute Critically to the Apomorphine-Induced Inhibition of Form-Deprivation Myopia in Mice. ACTA ACUST UNITED AC 2018; 59:2623-2634. [PMID: 29847669 DOI: 10.1167/iovs.17-22578] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Furong Huang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Lishuai Zhang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Qiongsi Wang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Yanan Yang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Qihang Li
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Yi Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jiangfan Chen
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| |
Collapse
|