1
|
Arrigo A, Cremona O, Aragona E, Casoni F, Consalez G, Dogru RM, Hauck SM, Antropoli A, Bianco L, Parodi MB, Bandello F, Grosche A. Müller cells trophism and pathology as the next therapeutic targets for retinal diseases. Prog Retin Eye Res 2025; 106:101357. [PMID: 40254246 DOI: 10.1016/j.preteyeres.2025.101357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Müller cells are a crucial retinal cell type involved in multiple regulatory processes and functions that are essential for retinal health and functionality. Acting as structural and functional support for retinal neurons and photoreceptors, Müller cells produce growth factors, regulate ion and fluid homeostasis, and facilitate neuronal signaling. They play a pivotal role in retinal morphogenesis and cell differentiation, significantly contributing to macular development. Due to their radial morphology and unique cytoskeletal organization, Müller cells act as optical fibers, efficiently channeling photons directly to the photoreceptors. In response to retinal damage, Müller cells undergo specific gene expression and functional changes that serve as a first line of defense for neurons, but can also lead to unwarranted cell dysfunction, contributing to cell death and neurodegeneration. In some species, Müller cells can reactivate their developmental program, promoting retinal regeneration and plasticity-a remarkable ability that holds promising therapeutic potential if harnessed in mammals. The crucial and multifaceted roles of Müller cells-that we propose to collectively call "Müller cells trophism"-highlight the necessity of maintaining their functionality. Dysfunction of Müller cells, termed "Müller cells pathology," has been associated with a plethora of retinal diseases, including age-related macular degeneration, diabetic retinopathy, vitreomacular disorders, macular telangiectasia, and inherited retinal dystrophies. In this review, we outline how even subtle disruptions in Müller cells trophism can drive the pathological cascade of Müller cells pathology, emphasizing the need for targeted therapies to preserve retinal health and prevent disease progression.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Ottavio Cremona
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emanuela Aragona
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Casoni
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Consalez
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rüya Merve Dogru
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 80939, Germany
| | - Alessio Antropoli
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco Bandello
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
2
|
Liu G, Li H, Feng L, Li M, Gao P, Wang F. O-GlcNAcylation promotes astroglial-mesenchymal transition via the connexin43 pathway under high-glucose conditions. Exp Eye Res 2025; 251:110206. [PMID: 39672530 DOI: 10.1016/j.exer.2024.110206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/25/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
This study aimed to investigate the effects of O-linked N-acetylglucosamine modification (O-GlcNAcylation) on astroglial-mesenchymal transition through connexin43 (Cx43) pathway under high-glucose conditions. The primary rat astrocytes were cultured under normal and high-glucose conditions, and level of GFAP, α-SMA and Cx43 was investigated. To explore the influence of O-GlcNAcylation on astroglial-mesenchymal transition, Thiamet G treatment was employed to enhance O-GlcNAcylation, while Alloxan was used to decrease it. Cx43 knockdown was acquired through lentivirus constructs to explore its role in astrocyte transition. The levels of GFAP and α-SMA expressions were examined, while astrocyte proliferation was evaluated using the CCK-8 assay, and migration was assessed through wound healing assays. The results showed that primary rat astrocytes were identified by GFAP antibody staining. Under high-glucose conditions, the levels of GFAP, α-SMA, and Cx43 increased, as confirmed by Western blot and immunofluorescence. O-GlcNAcylation augmentation induced by Thiamet G treatment significantly increased the expression of GFAP, α-SMA, and Cx43 compared to both normal and high-glucose conditions. Conversely, the inhibition of O-GlcNAcylation reversed the high-glucose-induced increase in GFAP and α-SMA. Cx43 knockout led to the downregulation of GFAP and α-SMA compared to high-glucose and O-GlcNAcylation-augmented conditions. Additionally, levels of O-GlcNAcylation and VEGF were reduced in Cx43 knockout group. Consistently, CCK8 and wound healing assays demonstrated that Cx43 knockout could inhibit astrocyte proliferation and migration compared to the high-glucose and O-GlcNAcylation augmented groups. These findings demonstrate that astroglial-mesenchymal transition occurs under high-glucose conditions, and can be promoted by O-GlcNAcylation augmentation, but suppressed by Cx43 knockout. The study underscores the significant role of Cx43 in this transition and its potential involvement in diabetic complications.
Collapse
Affiliation(s)
- Guodong Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China; Tongji Eye Institute, Tongji University School of Medicine, Shanghai, PR China.
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China
| | - Le Feng
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China
| | - Peng Gao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai, 200072, PR China
| |
Collapse
|
3
|
Carrero L, Antequera D, Alcalde I, Megias D, Ordoñez-Gutierrez L, Gutierrez C, Merayo-Lloves J, Wandosell F, Municio C, Carro E. Altered Clock Gene Expression in Female APP/PS1 Mice and Aquaporin-Dependent Amyloid Accumulation in the Retina. Int J Mol Sci 2023; 24:15679. [PMID: 37958666 PMCID: PMC10648501 DOI: 10.3390/ijms242115679] [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: 09/28/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is a neurodegenerative disorder characterized by different pathological symptomatology, including disrupted circadian rhythm. The regulation of circadian rhythm depends on the light information that is projected from the retina to the suprachiasmatic nucleus in the hypothalamus. Studies of AD patients and AD transgenic mice have revealed AD retinal pathology, including amyloid-β (Aβ) accumulation that can directly interfere with the regulation of the circadian cycle. Although the cause of AD pathology is poorly understood, one of the main risk factors for AD is female gender. Here, we found that female APP/PS1 mice at 6- and 12-months old display severe circadian rhythm disturbances and retinal pathological hallmarks, including Aβ deposits in retinal layers. Since brain Aβ transport is facilitated by aquaporin (AQP)4, the expression of AQPs were also explored in APP/PS1 retina to investigate a potential correlation between retinal Aβ deposits and AQPs expression. Important reductions in AQP1, AQP4, and AQP5 were detected in the retinal tissue of these transgenic mice, mainly at 6-months of age. Taken together, our findings suggest that abnormal transport of Aβ, mediated by impaired AQPs expression, contributes to the retinal degeneration in the early stages of AD.
Collapse
Affiliation(s)
- Laura Carrero
- Neurobiology of Alzheimer’s Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.C.); (D.A.); (C.G.)
- PhD Program in Neuroscience, Autonoma de Madrid University, 28049 Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer’s Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.C.); (D.A.); (C.G.)
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, Fundación de Investigación Oftalmológica, 28012 Oviedo, Spain; (I.A.); (J.M.-L.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Diego Megias
- Advanced Optical Microscopy Unit, Unidades Centrales Científico-Técnicas, Instituto de Salud Carlos III, 28222 Madrid, Spain;
| | - Lara Ordoñez-Gutierrez
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.O.-G.); (F.W.)
| | - Cristina Gutierrez
- Neurobiology of Alzheimer’s Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.C.); (D.A.); (C.G.)
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, Fundación de Investigación Oftalmológica, 28012 Oviedo, Spain; (I.A.); (J.M.-L.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.O.-G.); (F.W.)
| | - Cristina Municio
- Neurobiology of Alzheimer’s Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.C.); (D.A.); (C.G.)
| | - Eva Carro
- Neurobiology of Alzheimer’s Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, 28029 Madrid, Spain; (L.C.); (D.A.); (C.G.)
| |
Collapse
|
4
|
Mohammed SR, Elmasry K, El-Gamal R, El-Shahat MA, Sherif RN. Alteration of Aquaporins 1 and 4 immunohistochemical and gene expression in the cerebellum of diabetic albino rat. Tissue Cell 2023; 82:102076. [PMID: 36989704 DOI: 10.1016/j.tice.2023.102076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/29/2023]
Abstract
Aquaporins (AQPs) are a family of transmembrane channel proteins. AQP1 and AQP4 are expressed in cerebellum amongst others. This study was designed to assess the effect of diabetes on AQP1 and AQP4 expression in cerebellum of rats. Diabetes was induced by a single intraperitoneal injection of Streptozotocin 45 mg/kg in 24 adult male Sprague Dawley rats. Six rats from control and diabetic groups were sacrificed at one, four, and eight weeks post diabetic confirmation. After eight weeks, measurement of malondialdehyde (MDA), reduced glutathione (GSH) concentrations, and cerebellar mRNA expression for AQP1 and AQP4 genes were performed. Immunohistochemical evaluation of AQP1, AQP4, and glial fibrillary acidic protein (GFAP) for cerebellar sections was performed for all groups. Diabetes caused degenerative changes in Purkinje cells with a significant increase in the cerebellar level of MDA and AQP1 immunoreactivity and a significant decrease in GSH level and AQP4 expression levels. However, the alteration in the AQP1 mRNA level was not statistically significant. GFAP immunoreactivity was increased in 8 W diabetic rats following its decrease in 1 W diabetic rats. Diabetes caused some alteration in the AQPs 1 and 4 expression in the cerebellum of diabetic rats which may contribute to diabetes-induced cerebellar complications.
Collapse
|
5
|
Padovani-Claudio DA, Ramos CJ, Capozzi ME, Penn JS. Elucidating glial responses to products of diabetes-associated systemic dyshomeostasis. Prog Retin Eye Res 2023; 94:101151. [PMID: 37028118 PMCID: PMC10683564 DOI: 10.1016/j.preteyeres.2022.101151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 04/08/2023]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. DR has non-proliferative stages, characterized in part by retinal neuroinflammation and ischemia, and proliferative stages, characterized by retinal angiogenesis. Several systemic factors, including poor glycemic control, hypertension, and hyperlipidemia, increase the risk of DR progression to vision-threatening stages. Identification of cellular or molecular targets in early DR events could allow more prompt interventions pre-empting DR progression to vision-threatening stages. Glia mediate homeostasis and repair. They contribute to immune surveillance and defense, cytokine and growth factor production and secretion, ion and neurotransmitter balance, neuroprotection, and, potentially, regeneration. Therefore, it is likely that glia orchestrate events throughout the development and progression of retinopathy. Understanding glial responses to products of diabetes-associated systemic dyshomeostasis may reveal novel insights into the pathophysiology of DR and guide the development of novel therapies for this potentially blinding condition. In this article, first, we review normal glial functions and their putative roles in the development of DR. We then describe glial transcriptome alterations in response to systemic circulating factors that are upregulated in patients with diabetes and diabetes-related comorbidities; namely glucose in hyperglycemia, angiotensin II in hypertension, and the free fatty acid palmitic acid in hyperlipidemia. Finally, we discuss potential benefits and challenges associated with studying glia as targets of DR therapeutic interventions. In vitro stimulation of glia with glucose, angiotensin II and palmitic acid suggests that: 1) astrocytes may be more responsive than other glia to these products of systemic dyshomeostasis; 2) the effects of hyperglycemia on glia are likely to be largely osmotic; 3) fatty acid accumulation may compound DR pathophysiology by promoting predominantly proinflammatory and proangiogenic transcriptional alterations of macro and microglia; and 4) cell-targeted therapies may offer safer and more effective avenues for DR treatment as they may circumvent the complication of pleiotropism in retinal cell responses. Although several molecules previously implicated in DR pathophysiology are validated in this review, some less explored molecules emerge as potential therapeutic targets. Whereas much is known regarding glial cell activation, future studies characterizing the role of glia in DR and how their activation is regulated and sustained (independently or as part of retinal cell networks) may help elucidate mechanisms of DR pathogenesis and identify novel drug targets for this blinding disease.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3321A Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, AA1324 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3307 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| |
Collapse
|
6
|
Rowe CJ, Delbridge-Perry M, Bonan NF, Cohen A, Bentley M, DeCicco-Skinner KL, Davidson T, Connaughton VP. Time dependent effects of prolonged hyperglycemia in zebrafish brain and retina. FRONTIERS IN OPHTHALMOLOGY 2022; 2:947571. [PMID: 38983568 PMCID: PMC11182107 DOI: 10.3389/fopht.2022.947571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/29/2022] [Indexed: 07/11/2024]
Abstract
Prolonged hyperglycemia causes long-term vision complications and an increased risk of cognitive deficits. High blood sugar also confers an osmotic load/stress to cells. We assessed behavioral and neurochemical changes in zebrafish brain and retina following prolonged hyperglycemia for 4-weeks or 8-weeks. At each time point, behavior was assessed using 3-chamber choice task and optomotor response; tissue was then collected and levels of inflammatory markers, tight junction proteins, and neurotransmitters determined using Western Blots. After 4-weeks, brain levels of v-rel reticuloendotheliosis viral oncogene homolog A (avian) (RelA; NF-kB subunit), IkB kinase (IKK), and glial fibrillary acidic protein (GFAP) were significantly elevated; differences in zonula occludens-1 (ZO-1), claudin-5, glutamic acid decarboxylase (GAD), and tyrosine hydroxylase (TH) were not significant. In retina, significant differences were observed only for TH (decreased), Rel A (increased), and GFAP (increased) levels. Glucose-specific differences in initial choice latency and discrimination ratios were also observed. After 8-weeks, RelA, GAD, and TH were significantly elevated in both tissues; IKK and GFAP levels were also elevated, though not significantly. ZO-1 and claudin-5 levels osmotically decreased in retina but displayed an increasing trend in glucose-treated brains. Differences in discrimination ratio were driven by osmotic load. OMRs increased in glucose-treated fish at both ages. In vivo analysis of retinal vasculature suggested thicker vessels after 4-weeks, but thinner vessels at 8-weeks. In vitro, glucose treatment reduced formation of nodes and meshes in 3B-11 endothelial cells, suggesting a reduced ability to form a vascular network. Overall, hyperglycemia triggered a strong inflammatory response causing initial trending changes in tight junction and neuronal markers. Most differences after 4-weeks of exposure were observed in glucose-treated fish suggesting effects on glucose metabolism independent of osmotic load. After 8-weeks, the inflammatory response remained and glucose-specific effects on neurotransmitter markers were observed. Osmotic differences impacted cognitive behavior and retinal protein levels; protein levels in brain displayed glucose-driven changes. Thus, we not only observed differential sensitivities of retina and brain to glucose-insult, but also different cellular responses, suggesting hyperglycemia causes complex effects at the cellular level and/or that zebrafish are able to compensate for the continued high blood glucose levels.
Collapse
Affiliation(s)
- Cassie J. Rowe
- Department of Biology, American University, Washington, DC, United States
- Center for Neuroscience and Behavior, American University, Washington, DC, United States
| | - Mikayla Delbridge-Perry
- Department of Biology, American University, Washington, DC, United States
- Department of Chemistry, American University, Washington, DC, United States
| | - Nicole F. Bonan
- Department of Biology, American University, Washington, DC, United States
| | - Annastelle Cohen
- Department of Biology, American University, Washington, DC, United States
| | - Meg Bentley
- Department of Biology, American University, Washington, DC, United States
| | - Kathleen L. DeCicco-Skinner
- Department of Biology, American University, Washington, DC, United States
- Center for Neuroscience and Behavior, American University, Washington, DC, United States
| | - Terry Davidson
- Center for Neuroscience and Behavior, American University, Washington, DC, United States
- Department of Neuroscience, and American University, Washington, DC, United States
| | - Victoria P. Connaughton
- Department of Biology, American University, Washington, DC, United States
- Center for Neuroscience and Behavior, American University, Washington, DC, United States
| |
Collapse
|
7
|
Zhao M, Zhao S, Tang M, Sun T, Zheng Z, Ma M. Aqueous Humor Biomarkers of Retinal Glial Cell Activation in Patients With or Without Age-Related Cataracts and With Different Stages of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 35262732 PMCID: PMC8934562 DOI: 10.1167/iovs.63.3.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Purpose To clarify the expression of biomarkers of retinal glial cell activation in the aqueous humor (AH) of patients with and without age-related cataracts (ARCs) at different stages of diabetic retinopathy (DR). Methods Patients were stratified by the presence of ARCs and then grouped by the presence of diabetes mellitus (DM), nonproliferative DR (NPDR), proliferative DR (PDR), and controls. Water channel aquaporin 1 (AQP1), water channel aquaporin 4 (AQP4), inwardly rectifying potassium channel 4.1 (Kir4.1), and glial fibrillary acidic protein (GFAP) were assayed in AH samples by ELISAs. Results We enrolled 82 patients. The AQP1 concentration was higher in AH from cataract control patients than in control patients without cataracts (P < 0.05). The APQ1 concentration was also higher in patients with DM, NPDR, and PDR than in controls (P < 0.05). The concentrations of AQP4 and GFAP were significantly increased in patients with NPDR and PDR (P < 0.05) but not in patients with DM. Kir4.1 concentration was significantly decreased in patients with NPDR and PDR (P < 0.05), but the decrease in patients with DM did not reach significance. There were no differences in AQP4, Kir4.1, and GFAP between patients with and without ARCs. Conclusions Increased AQP1 in AH may be a biomarker for ARCs in patients without diabetes and a biomarker for retinal glial cell activation in patients with diabetes without cataracts. AQP4, Kir4.1, and GFAP levels in AH suggested that retinal glial cell activation was affected by the progression of DR.
Collapse
Affiliation(s)
- Minjie Zhao
- Department of Ophthalmology, Yixing People's Hospital, Jiangsu University, Zhenjiang, China
| | - Shuzhi Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Min Tang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Tao Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Shanghai Eye Diseases Prevention & Treatment Center/Shanghai Eye Hospital, Shanghai, China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Mingming Ma
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
8
|
Bringmann A, Unterlauft JD, Barth T, Wiedemann R, Rehak M, Wiedemann P. Müller cells and astrocytes in tractional macular disorders. Prog Retin Eye Res 2021; 86:100977. [PMID: 34102317 DOI: 10.1016/j.preteyeres.2021.100977] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 02/04/2023]
Abstract
Tractional deformations of the fovea mainly arise from an anomalous posterior vitreous detachment and contraction of epiretinal membranes, and also occur in eyes with cystoid macular edema or high myopia. Traction to the fovea may cause partial- and full-thickness macular defects. Partial-thickness defects are foveal pseudocysts, macular pseudoholes, and tractional, degenerative, and outer lamellar holes. The morphology of the foveal defects can be partly explained by the shape of Müller cells and the location of tissue layer interfaces of low mechanical stability. Because Müller cells and astrocytes provide the structural scaffold of the fovea, they are active players in mediating tractional alterations of the fovea, in protecting the fovea from such alterations, and in the regeneration of the foveal structure. Tractional and degenerative lamellar holes are characterized by a disruption of the Müller cell cone in the foveola. After detachment or disruption of the cone, Müller cells of the foveal walls support the structural stability of the foveal center. After tractional elevation of the inner layers of the foveal walls, possibly resulting in foveoschisis, Müller cells transmit tractional forces from the inner to the outer retina leading to central photoreceptor layer defects and a detachment of the neuroretina from the retinal pigment epithelium. This mechanism plays a role in the widening of outer lameller and full-thickness macular holes, and contributes to visual impairment in eyes with macular disorders caused by conractile epiretinal membranes. Müller cells of the foveal walls may seal holes in the outer fovea and mediate the regeneration of the fovea after closure of full-thickness holes. The latter is mediated by the formation of temporary glial scars whereas persistent glial scars impede regular foveal regeneration. Further research is required to improve our understanding of the roles of glial cells in the pathogenesis and healing of tractional macular disorders.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany.
| | - Jan Darius Unterlauft
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany
| | - Thomas Barth
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany
| | - Renate Wiedemann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany
| | - Matus Rehak
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany
| | - Peter Wiedemann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, 04103, Leipzig, Germany
| |
Collapse
|
9
|
Jha KA, Gentry J, Del Mar NA, Reiner A, Sohl N, Gangaraju R. Adipose Tissue-Derived Mesenchymal Stem Cell Concentrated Conditioned Medium Alters the Expression Pattern of Glutamate Regulatory Proteins and Aquaporin-4 in the Retina after Mild Traumatic Brain Injury. J Neurotrauma 2021; 38:1702-1716. [PMID: 33183134 DOI: 10.1089/neu.2020.7309] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Concentrated conditioned media from adipose tissue-derived mesenchymal stem cells (ASC-CCM) show promise for retinal degenerative diseases. In this study, we hypothesized that ASC-CCM could rescue retinal damage and thereby improve visual function by acting through Müller glia in mild traumatic brain injury (mTBI). Adult C57Bl/6 mice were subjected to a 50-psi air pulse on the left side of the head, resulting in an mTBI. After blast injury, 1 μL (∼100 ng total protein) of human ASC-CCM was delivered intravitreally and followed up after 4 weeks for visual function assessed by electroretinogram and histopathological markers for Müller cell-related markers. Blast mice that received ASC-CCM, compared with blast mice that received saline, demonstrated a significant improvement in a- and b-wave response correlated with a 1.3-fold decrease in extracellular glutamate levels and a concomitant increase in glutamine synthetase (GS), as well as the glutamate transporter (GLAST) in Müller cells. Additionally, an increase in aquaporin-4 (AQP4) in Müller cells in blast mice received saline restored to normal levels in blast mice that received ASC-CCM. In vitro studies on rMC-1 Müller glia exposed to 100 ng/mL glutamate or RNA interference knockdown of GLAST expression mimicked the increased Müller cell glial fibrillary acidic protein (a marker of gliosis) seen with mTBI, and suggested that an increase in glutamate and/or a decrease in GLAST might contribute to the Müller cell activation in vivo. Taken together, our data suggest a novel neuroprotective role for ASC-CCM in the rescue of the visual deficits and pathologies of mTBI via restoration of Müller cell health.
Collapse
Affiliation(s)
- Kumar Abhiram Jha
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Jordy Gentry
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Nobel A Del Mar
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Anton Reiner
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Nicolas Sohl
- Cell Care Therapeutics, Inc., Monrovia, California, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
10
|
Barboni MTS, Vaillend C, Joachimsthaler A, Liber AMP, Khabou H, Roux MJ, Vacca O, Vignaud L, Dalkara D, Guillonneau X, Ventura DF, Rendon A, Kremers J. Rescue of Defective Electroretinographic Responses in Dp71-Null Mice With AAV-Mediated Reexpression of Dp71. Invest Ophthalmol Vis Sci 2020; 61:11. [PMID: 32049345 PMCID: PMC7326481 DOI: 10.1167/iovs.61.2.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To study the potential effect of a gene therapy, designed to rescue the expression of dystrophin Dp71 in the retinas of Dp71-null mice, on retinal physiology. Methods We recorded electroretinograms (ERGs) in Dp71-null and wild-type littermate mice. In dark-adapted eyes, responses to flashes of several strengths were measured. In addition, flash responses on a 25-candela/square meters background were measured. On- and Off-mediated responses to sawtooth stimuli and responses to photopic sine-wave modulation (3–30 Hz) were also recorded. After establishing the ERG phenotype, the ShH10-GFP adeno-associated virus (AAV), which has been previously shown to target specifically Müller glial cells (MGCs), was delivered intravitreously with or without (sham therapy) the Dp71 coding sequence under control of a CBA promoter. ERG recordings were repeated three months after treatment. Real-time quantitative PCR and Western blotting analyses were performed in order to quantify Dp71 expression in the retinas. Results Dp71-null mice displayed reduced b-waves in dark- and light-adapted flash ERGs and smaller response amplitudes to photopic rapid-on sawtooth modulation and to sine-wave stimuli. Three months after intravitreal injections of the ShH10-GFP-2A-Dp71 AAV vector, ERG responses were completely recovered in treated eyes of Dp71-null mice. The functional rescue was associated with an overexpression of Dp71 in treated retinas. Conclusions The present results show successful functional recovery accompanying the reexpression of Dp71. In addition, this experimental model sheds light on MGCs influencing ERG components, since previous reports showed that aquaporin 4 and Kir4.1 channels were mislocated in MGCs of Dp71-null mice, while their distribution could be normalized following intravitreal delivery of the same ShH10-GFP-2A-Dp71 vector.
Collapse
|
11
|
Lai IP, Huang WL, Yang CM, Yang CH, Ho TC, Hsieh YT. Renal Biomarkers for Treatment Effect of Ranibizumab for Diabetic Macular Edema. J Diabetes Res 2020; 2020:7239570. [PMID: 32908935 PMCID: PMC7450296 DOI: 10.1155/2020/7239570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS To investigate the correlations between renal biomarkers and the treatment outcomes of ranibizumab for diabetic macular edema (DME). METHODS This hospital-based study retrospectively enrolled 88 eyes from 67 patients who had received one-year intravitreal ranibizumab treatment for DME. Best-corrected visual acuity (BCVA) and optical coherence tomography (OCT) at baseline and during the follow-up period were recorded. BCVA and OCT characteristics at baseline and their changes after ranibizumab treatment were compared between different proteinuria and estimated glomerular filtration rate (eGFR) groups. RESULTS Of the 88 eyes studied, those with moderately increased proteinuria had a thicker central subfield foveal thickness (CFT) and a higher proportion of intraretinal cysts than those with no proteinuria (P = 0.012 and 0.045, respectively) at baseline. After one year of ranibizumab treatment, the reduction in CFT was greater in patients with severely increased proteinuria than those with normal to mildly increased proteinuria (P = 0.030). On the other hand, patients with an eGFR <30 tended to have poorer visual improvements than those with normal eGFR (P = 0.044). CONCLUSIONS After ranibizumab treatment for DME, patients with severe proteinuria tended to gain better anatomical improvement, while those with poor eGFR tended to have poorer visual improvement.
Collapse
Affiliation(s)
| | - Wei-Lun Huang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzyy-Chang Ho
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ting Hsieh
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Saadane A, Mast N, Trichonas G, Chakraborty D, Hammer S, Busik JV, Grant MB, Pikuleva IA. Retinal Vascular Abnormalities and Microglia Activation in Mice with Deficiency in Cytochrome P450 46A1-Mediated Cholesterol Removal. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:405-425. [PMID: 30448403 DOI: 10.1016/j.ajpath.2018.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
CYP46A1 is the cytochrome P450 enzyme that converts cholesterol to 24-hydroxycholesterol, a cholesterol elimination product and a potent liver X receptor (LXR) ligand. We conducted retinal characterizations of Cyp46a1-/- mice that had normal fasting blood glucose levels but up to a 1.8-fold increase in retinal cholesterol. The retina of Cyp46a1-/- mice exhibited venous beading and tortuosity, microglia/macrophage activation, and increased vascular permeability, features commonly associated with diabetic retinopathy. The expression of Lxrα and Lxrβ was increased in both the whole Cyp46a1-/- retina and retinal macroglia/macrophages. The LXR-target genes were affected as well, primarily in activated microglial cells and macrophages. In the latter, the LXR-transactivated genes (Abca1, Abcg1, Apod, Apoe, Mylip, and Arg2) were up-regulated; similarly, there was an up-regulation of the LXR-transrepressed genes (Ccl2, Ptgs2, Cxcl1, Il1b, Il6, Nos2, and Tnfa). For comparison, gene expression was investigated in bone marrow-derived macrophages from Cyp46a1-/- mice as well as retinal and bone marrow-derived macrophages from Cyp27a1-/- and Cyp27a1-/-Cyp46a1-/- mice. CYP46A1 expression was detected in retinal endothelial cells, and this expression was increased in the proinflammatory environment. Retinal Cyp46a1-/- phosphoproteome revealed altered phosphorylation of 30 different proteins, including tight junction protein zonula occludens 1 and aquaporin 4. Collectively, the data obtained establish metabolic and regulatory significance of CYP46A1 for the retina and suggest pharmacologic activation of CYP46A1 as a potential therapeutic approach to dyslipidemia-induced retinal damage.
Collapse
Affiliation(s)
- Aicha Saadane
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - George Trichonas
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | | | - Sandra Hammer
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Julia V Busik
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Maria B Grant
- Department of Ophthalmology, University of Alabama, Birmingham, Alabama
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
13
|
Abstract
The eye is susceptible to adverse toxic effects by direct application, inadvertent ocular contact, or systemic exposure to chemicals or their metabolites. Although the albino rat is a less than ideal model for ocular toxicity studies, it has gained popularity for specific applications and may be the first species in which the ocular toxicity of a systemically administered xenobiotic becomes evident. This chapter reviews the embryology, anatomy, and physiology of the eye and associated glands and describes common nonneoplastic and neoplastic lesions encountered in laboratory rats.
Collapse
|
14
|
Mechanisms of macular edema: Beyond the surface. Prog Retin Eye Res 2017; 63:20-68. [PMID: 29126927 DOI: 10.1016/j.preteyeres.2017.10.006] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
Macular edema consists of intra- or subretinal fluid accumulation in the macular region. It occurs during the course of numerous retinal disorders and can cause severe impairment of central vision. Major causes of macular edema include diabetes, branch and central retinal vein occlusion, choroidal neovascularization, posterior uveitis, postoperative inflammation and central serous chorioretinopathy. The healthy retina is maintained in a relatively dehydrated, transparent state compatible with optimal light transmission by multiple active and passive systems. Fluid accumulation results from an imbalance between processes governing fluid entry and exit, and is driven by Starling equation when inner or outer blood-retinal barriers are disrupted. The multiple and intricate mechanisms involved in retinal hydro-ionic homeostasis, their molecular and cellular basis, and how their deregulation lead to retinal edema, are addressed in this review. Analyzing the distribution of junction proteins and water channels in the human macula, several hypotheses are raised to explain why edema forms specifically in the macular region. "Pure" clinical phenotypes of macular edema, that result presumably from a single causative mechanism, are detailed. Finally, diabetic macular edema is investigated, as a complex multifactorial pathogenic example. This comprehensive review on the current understanding of macular edema and its mechanisms opens perspectives to identify new preventive and therapeutic strategies for this sight-threatening condition.
Collapse
|
15
|
Simone L, Gargano CD, Pisani F, Cibelli A, Mola MG, Frigeri A, Svelto M, Nicchia GP. Aquaporin-1 inhibition reduces metastatic formation in a mouse model of melanoma. J Cell Mol Med 2017; 22:904-912. [PMID: 29044946 PMCID: PMC5783831 DOI: 10.1111/jcmm.13378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Aquaporin-1 (AQP1) is a proangiogenic water channel protein promoting endothelial cell migration. We previously reported that AQP1 silencing by RNA interference reduces angiogenesis-dependent primary tumour growth in a mouse model of melanoma. In this study, we tested the hypothesis that AQP1 inhibition also affects animal survival and lung nodule formation. Melanoma was induced by injecting B16F10 cells into the back of C57BL6J mice. Intratumoural injection of AQP1 siRNA and CTRL siRNA was performed 10 days after tumour cell implantation. Lung nodule formation was analysed after the death of the mice. Western blot was used to quantify HIF-1α, caspase-3 (CASP3) and metalloproteinase-2 (MMP2) protein levels. We found that AQP1 knock-down (KD) strongly inhibited metastatic lung nodule formation. Moreover, AQP1 siRNA-treated mice showed a twofold survival advantage compared to mice receiving CTRL siRNAs. The reduced AQP1-dependent tumour angiogenesis caused a hypoxic condition, evaluated by HIF-1α significant increase, in turn causing an increased level of apoptosis in AQP1 KD tumours, assessed by CASP3 quantification and DNA fragmentation. Importantly, a decreased level of MMP2 after AQP1 KD indicated a decreased activity against extracellular matrix associated with reduced vascularization and metastatic formation. In conclusion, these findings highlight an additional role for AQP1 as an important determinant of tumour dissemination by facilitating tumour cell extravasation and metastatic formation. This study adds knowledge on the role played by AQP1 in tumour biology and supports the view of AQP1 as a potential drug target for cancer therapy.
Collapse
Affiliation(s)
- Laura Simone
- IRCCS Casa Sollievo della Sofferenza, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), San Giovanni Rotondo, FG, Italy.,Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Concetta Domenica Gargano
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Francesco Pisani
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Antonio Cibelli
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Maria Grazia Mola
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Antonio Frigeri
- School of Medicine, Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, BA, Italy.,Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, New York, Bronx, NY, USA
| | - Maria Svelto
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy.,Institute of Biomembranes and Bioenergetics, National Research Council, Bari, BA, Italy.,National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | - Grazia Paola Nicchia
- Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy.,Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University, New York, Bronx, NY, USA
| |
Collapse
|
16
|
Xia T, Rizzolo LJ. Effects of diabetic retinopathy on the barrier functions of the retinal pigment epithelium. Vision Res 2017; 139:72-81. [PMID: 28347688 DOI: 10.1016/j.visres.2017.02.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
Diabetic retinopathy is a debilitating microvascular complication of diabetes mellitus. A rich literature describes the breakdown of retinal endothelial cells and the inner blood-retinal barrier, but the effects of diabetes on the retinal pigment epithelium (RPE) has received much less attention. RPE lies between the choroid and neurosensory retina to form the outer blood-retinal barrier. RPE's specialized and dynamic barrier functions are crucial for maintaining retinal health. RPE barrier functions include a collection of interrelated structures and activities that regulate the transepithelial movement of solutes, including: diffusion through the paracellular spaces, facilitated diffusion through the cells, active transport, receptor-mediated and bulk phase transcytosis, and metabolic processing of solutes in transit. In the later stages of diabetic retinopathy, the tight junctions that regulate the paracellular space begin to disassemble, but there are earlier effects on the other aspects of RPE barrier function, particularly active transport and metabolic processing. With advanced understanding of RPE-specific barrier functions, and more in vivo-like culture models, the time is ripe for revisiting experiments in the literature to resolve controversies and extend our understanding of how diabetes affects the outer blood-retinal barrier.
Collapse
Affiliation(s)
- Tina Xia
- Departments of Surgery and Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208062, New Haven, CT 06520-8062, USA.
| | - Lawrence J Rizzolo
- Departments of Surgery and Ophthalmology and Visual Science, Yale University School of Medicine, PO Box 208062, New Haven, CT 06520-8062, USA.
| |
Collapse
|
17
|
Abstract
PURPOSE Recent studies have shown that aquaporins (AQPs) play an important role in proliferating tumor microvessels and angiogenesis. In this study, the authors investigated the expression of aquaporin-1 (AQP1) and aquaporin-3 (AQP3) in pterygial and normal conjunctival tissues. METHODS Fifteen patients with pterygium were enrolled in the study. Pterygium was excised, and a conjunctival rotational flap or autograft was inserted. Normal conjunctival tissue was obtained from the flap or graft. Western blot analysis was performed to assess the expression of AQP1 and AQP3 in pterygial and normal conjunctival tissues. Tissue localization of AQP1 and AQP3 was determined by immunohistochemical analysis. RESULTS AQP1 and AQP3 are localized in the epithelial and subepithelial regions in pterygial and normal conjunctival tissues. Protein expression of both AQP1 and AQP3 was elevated in pterygia when compared with conjunctival tissues. The significant increase in protein expression of AQP1 was 3-fold in pterygium over normal conjunctiva (P = 0.004) and 2-fold increase in AQP3 expression of pterygium was detected (P = 0.02) according to densitometric analysis. CONCLUSIONS Elevated protein expression of AQP1 and AQP3 was observed in pterygial tissues when compared with normal conjunctiva. The data suggest that the increased expression of AQP1 and AQP3 in pterygial tissues may be involved in the pathogenesis of pterygia, and therefore, AQP1 and AQP3 are potential therapeutic targets for preventing or delaying the progression of the disease.
Collapse
|
18
|
Gaddini L, Varano M, Matteucci A, Mallozzi C, Villa M, Pricci F, Malchiodi-Albedi F. Müller glia activation by VEGF-antagonizing drugs: An in vitro study on rat primary retinal cultures. Exp Eye Res 2015; 145:158-163. [PMID: 26607807 DOI: 10.1016/j.exer.2015.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/28/2015] [Accepted: 11/10/2015] [Indexed: 11/27/2022]
Abstract
The effects of the anti-Vascular Endothelial Growth Factor (VEGF) drugs ranibizumab and aflibercept were studied in Müller glia in primary mixed cultures from rat neonatal retina. Treatment with both agents induced activation of Müller glia, demonstrated by increased levels of Glial Fibrillary Acidic Protein. In addition, phosphorylated Extracellular-Regulated Kinase 1/2 (ERK 1/2) showed enhanced immunoreactivity in activated Müller glia. Treatment with aflibercept induced an increase in K(+) channel (Kir) 4.1 levels and both drugs upregulated Aquaporin 4 (AQP4) in activated Müller glia. The results show that VEGF-antagonizing drugs influence the homeostasis of Müller cells in primary retinal cultures, inducing an activated phenotype. Upregulation of Kir4.1 and AQP4 suggests that Müller glia activation following anti-VEGF drugs may not depict a detrimental gliotic reaction. Indeed, it could represent one of the mechanisms able to contribute to the therapeutic effects of these drugs, particularly in the presence of macular edema.
Collapse
Affiliation(s)
- Lucia Gaddini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy
| | - Monica Varano
- GB Bietti Eye Foundation IRCCS, Via Livenza, 3, Rome, 00198, Italy
| | - Andrea Matteucci
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy
| | - Cinzia Mallozzi
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy
| | - Marika Villa
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy
| | - Flavia Pricci
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy
| | - Fiorella Malchiodi-Albedi
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome, 00161, Italy.
| |
Collapse
|
19
|
de Senna PN, Xavier LL, Bagatini PB, Saur L, Galland F, Zanotto C, Bernardi C, Nardin P, Gonçalves CA, Achaval M. Physical training improves non-spatial memory, locomotor skills and the blood brain barrier in diabetic rats. Brain Res 2015; 1618:75-82. [PMID: 26032744 DOI: 10.1016/j.brainres.2015.05.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 10/23/2022]
Abstract
Type 1 diabetes mellitus (T1DM) progressively affects cognitive domains, increases blood-brain barrier (BBB) permeability and promotes neurovascular impairment in specific brain areas. Physical exercise, on the other hand, has beneficial effects on brain functions, improving learning and memory. This study investigated the effects of treadmill training on cognitive and motor behavior, and on the expression of proteins related to BBB integrity, such as claudin-5 and aquaporin-4 (AQP4) in the hippocampus and striatum in diabetic rats. For this study, 60 Wistar rats were divided into four groups (n=15 per group): non-trained control (NTC), trained control (TC), non-trained diabetic (NTD), trained diabetic (TD). After diabetic induction of 30 days by streptozotocin injection, the exercise groups were submitted to 5 weeks of running training. After that, all groups were assessed in a novel object-recognition task (NOR) and the rotarod test. Additionally, claudin-5 and AQP4 levels were measured using biochemical assays. The results showed that exercise enhanced NOR task performance and rotarod ability in the TC and TD animals. Diabetes produced a decrease in claudin-5 expression in the hippocampus and striatum and reduced AQP4 in the hippocampus. Exercise preserved the claudin-5 content in the striatum of TD rats, but not in the hippocampus. The reduction of AQP4 levels produced by diabetes was not reversed by exercise. We conclude that exercise improves short-term memory retention, enhances motor performance in diabetic rats and affects important structural components of the striatal BBB. The results obtained could enhance the knowledge regarding the neurochemical benefits of exercise in diabetes.
Collapse
Affiliation(s)
- Priscylla Nunes de Senna
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Biologia Celular e Tecidual, Departamento de Ciências Morfofisiológicas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Léder Leal Xavier
- Laboratório de Biologia Celular e Tecidual, Departamento de Ciências Morfofisiológicas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pamela Brambilla Bagatini
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lisiani Saur
- Laboratório de Biologia Celular e Tecidual, Departamento de Ciências Morfofisiológicas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fabiana Galland
- Departamento de Bioquímica, Instituto de Ciências Básica da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Caroline Zanotto
- Departamento de Bioquímica, Instituto de Ciências Básica da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Caren Bernardi
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Patrícia Nardin
- Departamento de Bioquímica, Instituto de Ciências Básica da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alberto Gonçalves
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básica da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Matilde Achaval
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
20
|
|
21
|
Shin ES, Sorenson CM, Sheibani N. Diabetes and retinal vascular dysfunction. J Ophthalmic Vis Res 2015; 9:362-73. [PMID: 25667739 PMCID: PMC4307665 DOI: 10.4103/2008-322x.143378] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetes predominantly affects the microvascular circulation of the retina resulting in a range of structural changes unique to this tissue. These changes ultimately lead to altered permeability, hyperproliferation of endothelial cells and edema, and abnormal vascularization of the retina with resulting loss of vision. Enhanced production of inflammatory mediators and oxidative stress are primary insults with significant contribution to the pathogenesis of diabetic retinopathy (DR). We have determined the identity of the retinal vascular cells affected by hyperglycemia, and have delineated the cell autonomous impact of high glucose on function of these cells. We discuss some of the high glucose specific changes in retinal vascular cells and their contribution to retinal vascular dysfunction. This knowledge provides novel insight into the molecular and cellular defects contributing to the development and progression of diabetic retinopathy, and will aid in the development of innovative, as well as target specific therapeutic approaches for prevention and treatment of DR.
Collapse
Affiliation(s)
- Eui Seok Shin
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA ; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA ; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
22
|
|
23
|
The role of inflammation in the pathogenesis of macular edema secondary to retinal vascular diseases. Mediators Inflamm 2014; 2014:432685. [PMID: 25152567 PMCID: PMC4134827 DOI: 10.1155/2014/432685] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/05/2014] [Accepted: 07/09/2014] [Indexed: 12/17/2022] Open
Abstract
Macular edema (ME) is a nonspecific sign of numerous retinal vascular diseases. This paper is an updated overview about the role of inflammatory processes in the genesis of both diabetic macular edema (DME) and ME secondary to retinal vein occlusion (RVO). We focus on the inflammatory mediators implicated, the effect of the different intravitreal therapies, the recruitment of leukocytes mediated by adhesion molecules, and the role of retinal Müller glial (RMG) cells.
Collapse
|
24
|
Schey KL, Wang Z, L Wenke J, Qi Y. Aquaporins in the eye: expression, function, and roles in ocular disease. Biochim Biophys Acta Gen Subj 2013; 1840:1513-23. [PMID: 24184915 DOI: 10.1016/j.bbagen.2013.10.037] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND All thirteen known mammalian aquaporins have been detected in the eye. Moreover, aquaporins have been identified as playing essential roles in ocular functions ranging from maintenance of lens and corneal transparency to production of aqueous humor to maintenance of cellular homeostasis and regulation of signal transduction in the retina. SCOPE OF REVIEW This review summarizes the expression and known functions of ocular aquaporins and discusses their known and potential roles in ocular diseases. MAJOR CONCLUSIONS Aquaporins play essential roles in all ocular tissues. Remarkably, not all aquaporin function as a water permeable channel and the functions of many aquaporins in ocular tissues remain unknown. Given their vital roles in maintaining ocular function and their roles in disease, aquaporins represent potential targets for future therapeutic development. GENERAL SIGNIFICANCE Since aquaporins play key roles in ocular physiology, an understanding of these functions is important to improving ocular health and treating diseases of the eye. It is likely that future therapies for ocular diseases will rely on modulation of aquaporin expression and/or function. This article is part of a Special Issue entitled Aquaporins.
Collapse
Affiliation(s)
- Kevin L Schey
- Department of Biochemistry, Vanderbilt School of Medicine, Vanderbilt University, Nashville, TN 37232, USA.
| | - Zhen Wang
- Department of Biochemistry, Vanderbilt School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Jamie L Wenke
- Department of Biochemistry, Vanderbilt School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Ying Qi
- Department of Biochemistry, Vanderbilt School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
25
|
Vogler S, Pannicke T, Hollborn M, Grosche A, Busch S, Hoffmann S, Wiedemann P, Reichenbach A, Hammes HP, Bringmann A. Müller cell reactivity in response to photoreceptor degeneration in rats with defective polycystin-2. PLoS One 2013; 8:e61631. [PMID: 23755094 PMCID: PMC3670868 DOI: 10.1371/journal.pone.0061631] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/12/2013] [Indexed: 12/18/2022] Open
Abstract
Background Retinal degeneration in transgenic rats that express a mutant cilia gene polycystin-2 (CMV-PKD2(1/703)HA) is characterized by initial photoreceptor degeneration and glial activation, followed by vasoregression and neuronal degeneration (Feng et al., 2009, PLoS One 4: e7328). It is unknown whether glial activation contributes to neurovascular degeneration after photoreceptor degeneration. We characterized the reactivity of Müller glial cells in retinas of rats that express defective polycystin-2. Methods Age-matched Sprague-Dawley rats served as control. Retinal slices were immunostained for intermediate filaments, the potassium channel Kir4.1, and aquaporins 1 and 4. The potassium conductance of isolated Müller cells was recorded by whole-cell patch clamping. The osmotic swelling characteristics of Müller cells were determined by superfusion of retinal slices with a hypoosmotic solution. Findings Müller cells in retinas of transgenic rats displayed upregulation of GFAP and nestin which was not observed in control cells. Whereas aquaporin-1 labeling of photoreceptor cells disappeared along with the degeneration of the cells, aquaporin-1 emerged in glial cells in the inner retina of transgenic rats. Aquaporin-4 was upregulated around degenerating photoreceptor cells. There was an age-dependent redistribution of Kir4.1 in retinas of transgenic rats, with a more even distribution along glial membranes and a downregulation of perivascular Kir4.1. Müller cells of transgenic rats displayed a slight decrease in their Kir conductance as compared to control. Müller cells in retinal tissues from transgenic rats swelled immediately under hypoosmotic stress; this was not observed in control cells. Osmotic swelling was induced by oxidative-nitrosative stress, mitochondrial dysfunction, and inflammatory lipid mediators. Interpretation Cellular swelling suggests that the rapid water transport through Müller cells in response to osmotic stress is altered as compared to control. The dislocation of Kir4.1 will disturb the retinal potassium and water homeostasis, and osmotic generation of free radicals and inflammatory lipids may contribute to neurovascular injury.
Collapse
Affiliation(s)
- Stefanie Vogler
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Margrit Hollborn
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Antje Grosche
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Stephanie Busch
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sigrid Hoffmann
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Wiedemann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
26
|
Ortak H, Cayli S, Ocaklı S, Söğüt E, Ekici F, Tas U, Demir S. Age-related changes of aquaporin expression patterns in the postnatal rat retina. Acta Histochem 2013; 115:382-8. [PMID: 23131425 DOI: 10.1016/j.acthis.2012.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 12/24/2022]
Abstract
Previous studies revealed that the rat retina contains numerous membrane-located water channels, the aquaporins (AQPs). Protein expression patterns of AQP1-4, 6 and 9 were examined by immunohistochemistry. In the present study, we investigated the immunolocalization of AQP1-4, 6 and 9 during postnatal development in the rat retina and examined the effect of age on the tissue distribution of these channels. AQP1, 3, 4, 6 and 9 showed gradually increased expression in rat retinas from postnatal week 1 to week 12, and decreased in the 40-week-old rat retinas. AQP2 expression was barely seen in the first week in rat retinas and displayed a significant increase from week 1 to week 4, however no significant alteration of AQP2 was observed after 4weeks of development. AQP1 and 4 immunoreactivities were present in the inner limiting membrane (ILM), the ganglion cell layer (GCL), inner nuclear layer (INL) and retinal pigment epithelium (RPE) in the 4-, 12- and 40-week-old rat retinas. The RPE, OLM and ILM showed a remarkable expression of AQP1-4, 6 and 9 in the 4, 12 and 40-week-old rat retinas. The reduced expression of AQPs in aged rat retinas may indicate the involvement of AQPs in the pathogenesis of age-related retinal diseases.
Collapse
Affiliation(s)
- Huseyin Ortak
- Department of Ophthalmology, Gaziosmanpasa University Faculty of Medicine, Tokat, Turkey.
| | | | | | | | | | | | | |
Collapse
|
27
|
Klaassen I, Van Noorden CJF, Schlingemann RO. Molecular basis of the inner blood-retinal barrier and its breakdown in diabetic macular edema and other pathological conditions. Prog Retin Eye Res 2013; 34:19-48. [PMID: 23416119 DOI: 10.1016/j.preteyeres.2013.02.001] [Citation(s) in RCA: 502] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/19/2012] [Accepted: 02/01/2013] [Indexed: 12/16/2022]
Abstract
Breakdown of the inner endothelial blood-retinal barrier (BRB), as occurs in diabetic retinopathy, age-related macular degeneration, retinal vein occlusions, uveitis and other chronic retinal diseases, results in vasogenic edema and neural tissue damage, causing loss of vision. The central mechanism of altered BRB function is a change in the permeability characteristics of retinal endothelial cells caused by elevated levels of growth factors, cytokines, advanced glycation end products, inflammation, hyperglycemia and loss of pericytes. Subsequently, paracellular but also transcellular transport across the retinal vascular wall increases via opening of endothelial intercellular junctions and qualitative and quantitative changes in endothelial caveolar transcellular transport, respectively. Functional changes in pericytes and astrocytes, as well as structural changes in the composition of the endothelial glycocalyx and the basal lamina around BRB endothelium further facilitate BRB leakage. As Starling's rules apply, active transcellular transport of plasma proteins by the BRB endothelial cells causing increased interstitial osmotic pressure is probably the main factor in the formation of macular edema. The understanding of the complex cellular and molecular processes involved in BRB leakage has grown rapidly in recent years. Although appropriate animal models for human conditions like diabetic macular edema are lacking, these insights have provided tools for rational design of drugs aimed at restoring the BRB as well as for design of effective transport of drugs across the BRB, to treat the chronic retinal diseases such as diabetic macular edema that affect the quality-of-life of millions of patients.
Collapse
Affiliation(s)
- Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
28
|
Johnson LE, Larsen M, Perez MT. Retinal adaptation to changing glycemic levels in a rat model of type 2 diabetes. PLoS One 2013; 8:e55456. [PMID: 23408985 PMCID: PMC3568153 DOI: 10.1371/journal.pone.0055456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 01/02/2013] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Glucose concentrations are elevated in retinal cells in undiagnosed and in undertreated diabetes. Studies of diabetic patients suggest that retinal function adapts, to some extent, to this increased supply of glucose. The aim of the present study was to examine such adaptation in a model of type 2 diabetes and assess how the retina responds to the subsequent institution of glycemic control. METHODS Electroretinography (ERG) was conducted on untreated Zucker diabetic fatty (ZDF) rats and congenic controls from 8-22 weeks of age and on ZDFs treated with daily insulin from 16-22 weeks of age. Retinal sections from various ages were prepared and compared histologically and by immunocytochemistry. PRINCIPAL FINDINGS/CONCLUSIONS Acute hyperglycemia did not have an effect on control rats while chronic hyperglycemia in the ZDF was associated with scotopic ERG amplitudes which were up to 20% higher than those of age-matched controls. This change followed the onset of hyperglycemia with a delay of over one month, supporting that habituation to hyperglycemia is a slow process. When glycemia was lowered, an immediate decrease in ZDF photoreceptoral activity was induced as seen by a reduction in a-wave amplitudes and maximum slopes of about 30%. A direct effect of insulin on the ERG was unlikely since the expression of phosphorylated Akt kinase was not affected by treatment. The electrophysiological differences between untreated ZDFs and controls preceded an activation of Müller cells in the ZDFs (up-regulation of glial fibrillary acidic protein), which was attenuated by insulin treatment. There were otherwise no signs of cell death or morphological alterations in any of the experimental groups. These data show that under chronic hyperglycemia, the ZDF retina became abnormally sensitive to variations in substrate supply. In diabetes, a similar inability to cope with intensive glucose lowering could render the retina susceptible to damage.
Collapse
Affiliation(s)
- Leif E Johnson
- Department of Ophthalmology, Glostrup Hospital, Glostrup, Denmark.
| | | | | |
Collapse
|
29
|
Beyond Polarity: Functional Membrane Domains in Astrocytes and Müller Cells. Neurochem Res 2012; 37:2513-23. [DOI: 10.1007/s11064-012-0824-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/01/2012] [Accepted: 06/08/2012] [Indexed: 10/28/2022]
|
30
|
Drechsler F, Köferl P, Hollborn M, Wiedemann P, Bringmann A, Kohen L, Rehak M. Effect of intravitreal anti-vascular endothelial growth factor treatment on the retinal gene expression in acute experimental central retinal vein occlusion. Ophthalmic Res 2011; 47:157-62. [PMID: 22116547 DOI: 10.1159/000330279] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 06/19/2011] [Indexed: 11/19/2022]
Abstract
PURPOSE To determine the effect of intravitreal bevacizumab and anti-vascular endothelial growth factor (VEGF) antibodies on the gene expression in the neural retina in a rat model of central retinal vein occlusion (CRVO). METHODS The CRVO was induced by laser photocoagulation of all retinal veins. The animals were divided into 3 groups (in each, n = 16): group CRVO only without any further treatment, group CRVO with bevacizumab, and group CRVO with anti-VEGF antibodies. The intravitreal injection of bevacizumab or anti-VEGF antibodies was performed 15 min after CRVO induction. The left eyes in all animals served as untreated controls. The expression of factors which influence the development of vascular edema (VEGF-A, pigment epithelium-derived factor, PEDF), of channels implicated in retinal osmohomeostasis (Kir4.1, AQP4, AQP1) and of the proinflammatory cytokines interleukin (IL)-1β and IL-6 was determined by using real-time RT-PCR after 1 and 3 days of CRVO. RESULTS CRVO induced a rapid transient upregulation of Vegfa after 1 day, and a delayed upregulation of Pedf after 3 days of CRVO. The expression levels of Kir4.1, Aqp4 and Aqp1 were strongly decreased, and the levels of Il1β and Il6 were strikingly increased after CRVO. Intravitreal bevacizumab and anti-VEGF antibodies fully prevented the upregulation of Vegfa after 1 day, and the upregulation of Pedf after 3 days of CRVO, and decreased the upregulation of Il1β after 1 day of CRVO. Anti-VEGF treatment had no effect on the expression levels of Kir4.1, Aqp4, Aqp1, and Il6. CONCLUSIONS It is suggested that the inhibitory effect on the upregulation of Vegfa and Il1β contributes to the edema-resolving effect of anti-VEGF treatment.
Collapse
|
31
|
Bringmann A, Wiedemann P. Müller glial cells in retinal disease. ACTA ACUST UNITED AC 2011; 227:1-19. [PMID: 21921569 DOI: 10.1159/000328979] [Citation(s) in RCA: 312] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 04/27/2011] [Indexed: 11/19/2022]
Abstract
Virtually all pathogenic stimuli activate Müller cells. Reactive Müller cells exert protective and toxic effects on photoreceptors and neurons. They contribute to oxidative stress and glutamate toxicity due to malfunctions of glutamate uptake and glutathione synthesis. Downregulation of potassium conductance disrupts transcellular potassium and water transport, resulting in neuronal hyperexcitability and edema. Protective effects of reactive Müller cells include upregulation of adenosine 5'-triphosphate (ATP)-degrading ectoenzymes, which enhances the extracellular availability of the neuroprotectant adenosine, abrogation of the osmotic release of ATP, which might protect retinal ganglion cells from apoptosis, and the release of antioxidants and neurotrophic factors. The dedifferentiation of reactive Müller cells to progenitor-like cells might have an impact on future therapeutic approaches. A better understanding of the gliotic mechanisms will be helpful in developing efficient therapeutic strategies aiming at increased protective and regenerative properties and decreased toxicity of reactive Müller cells.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
32
|
Hollborn M, Dukic-Stefanovic S, Pannicke T, Ulbricht E, Reichenbach A, Wiedemann P, Bringmann A, Kohen L. Expression of Aquaporins in the Retina of Diabetic Rats. Curr Eye Res 2011; 36:850-6. [DOI: 10.3109/02713683.2011.593108] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
|
34
|
Curtis TM, Hamilton R, Yong PH, McVicar CM, Berner A, Pringle R, Uchida K, Nagai R, Brockbank S, Stitt AW. Müller glial dysfunction during diabetic retinopathy in rats is linked to accumulation of advanced glycation end-products and advanced lipoxidation end-products. Diabetologia 2011; 54:690-8. [PMID: 21116609 DOI: 10.1007/s00125-010-1971-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 10/14/2010] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS The impact of AGEs and advanced lipoxidation end-products (ALEs) on neuronal and Müller glial dysfunction in the diabetic retina is not well understood. We therefore sought to identify dysfunction of the retinal Müller glia during diabetes and to determine whether inhibition of AGEs/ALEs can prevent it. METHODS Sprague-Dawley rats were divided into three groups: (1) non-diabetic; (2) untreated streptozotocin-induced diabetic; and (3) diabetic treated with the AGE/ALE inhibitor pyridoxamine for the duration of diabetes. Rats were killed and their retinas were evaluated for neuroglial pathology. RESULTS AGEs and ALEs accumulated at higher levels in diabetic retinas than in controls (p < 0.001). AGE/ALE immunoreactivity was significantly diminished by pyridoxamine treatment of diabetic rats. Diabetes was also associated with the up-regulation of the oxidative stress marker haemoxygenase-1 and the induction of glial fibrillary acidic protein production in Müller glia (p < 0.001). Pyridoxamine treatment of diabetic rats had a significant beneficial effect on both variables (p < 0.001). Diabetes also significantly altered the normal localisation of the potassium inwardly rectifying channel Kir4.1 and the water channel aquaporin 4 to the Müller glia end-feet interacting with retinal capillaries. These abnormalities were prevented by pyridoxamine treatment. CONCLUSIONS/INTERPRETATION While it is established that AGE/ALE formation in the retina during diabetes is linked to microvascular dysfunction, this study suggests that these pathogenic adducts also play a role in Müller glial dysfunction.
Collapse
Affiliation(s)
- T M Curtis
- Centre for Vision and Vascular Science, Queen's University Belfast, Royal Victoria Hospital, Belfast BT12 6BA, Northern Ireland, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Iandiev I, Dukic-Stefanovic S, Hollborn M, Pannicke T, Härtig W, Wiedemann P, Reichenbach A, Bringmann A, Kohen L. Immunolocalization of aquaporin-6 in the rat retina. Neurosci Lett 2011; 490:130-4. [DOI: 10.1016/j.neulet.2010.12.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/18/2010] [Accepted: 12/16/2010] [Indexed: 10/18/2022]
|
36
|
Fukuda M, Nakanishi Y, Fuse M, Yokoi N, Hamada Y, Fukagawa M, Negi A, Nakamura M. Altered expression of aquaporins 1 and 4 coincides with neurodegenerative events in retinas of spontaneously diabetic Torii rats. Exp Eye Res 2010; 90:17-25. [DOI: 10.1016/j.exer.2009.09.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 09/02/2009] [Accepted: 09/04/2009] [Indexed: 12/22/2022]
|
37
|
Qin Y, Xu G, Fan J, Witt RE, Da C. High-salt loading exacerbates increased retinal content of aquaporins AQP1 and AQP4 in rats with diabetic retinopathy. Exp Eye Res 2009; 89:741-7. [DOI: 10.1016/j.exer.2009.06.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 06/24/2009] [Accepted: 06/26/2009] [Indexed: 01/19/2023]
|
38
|
Qin Y, Fan J, Ye X, Xu G, Liu W, Da C. High salt loading alters the expression and localization of glial aquaporins in rat retina. Exp Eye Res 2009; 89:88-94. [PMID: 19268466 DOI: 10.1016/j.exer.2009.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 02/20/2009] [Accepted: 02/24/2009] [Indexed: 10/21/2022]
Abstract
In the neural retina, glial cells control the ionic concentrations in part by mediation of transmembrane water fluxes through aquaporin (AQP) water channels. The expression and immunolocalization of two water channels, AQP1 and AQP4, in the rat retina during experimental high salt loading were investigated in this study. Six-week-old Wistar rats were allowed free access to rat chow with 8% NaCl concentration. Of these rats, 6 were killed after 2, 6, 10 and 20 weeks. Twelve-week-old and 26-week-old Wistar rats with a normal diet (0.5% NaCl concentration) were used as controls. Retinal tissues were collected. Ultrathin sections stained with uranyl acetate and lead citrate were photographed using a transmission electron microscope (TEM). Retinal whole mounts and cryosections were immunostained with AQP1 and AQP4 antibodies to detect the immunolocalization changes by confocal microscopy. The AQP1 and AQP4 contents were evaluated by western blot analysis. In control tissues, no intracellular edema and mitochondria swelling were observed by TEM. The immunoreactive AQP4 was expressed by glial cells (Müller cells and astrocytes) predominantly in the inner retina, and AQP1 was expressed in the outer retina. In the retinas of high salt loading animals, obvious intracellular edema was observed by TEM in retinal ganglion cell (RGC) and mitochondria swelling was observed in glial cells. Strong expression of AQP1 was found in glial cells located in the innermost retinal layers, mainly in astrocytes. The superficial retinal vessels were surrounded by AQP4 in control retinas, but by both AQP4 and AQP1 in retina of high salt loading animals. A similar alteration in the localization of AQP1 has been described in the rat retina after transient ischemia and diabetes. Western blot results supported the conclusion that the AQP1 expression increased during high salt diet. Our findings indicate that high salt loading may induce neural retina edema, and that altered glial cell-mediated water transport via AQP channels in the retina may be one of the reasons for intracellular edema in the neural retina.
Collapse
Affiliation(s)
- Yaowu Qin
- EENT Hospital, Eye Institute, Fudan University, Fenyang Road 83, Shanghai, China
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
The aquaporins (AQPs) are integral membrane proteins whose main function is to transport water across cell membranes in response to osmotic gradients. At the ocular surface, AQP1 is expressed in corneal endothelium, AQP3 and AQP5 in corneal epithelium, and AQP3 in conjunctival epithelium. AQPs are also expressed in lens fiber cells (AQP0), lens epithelium (AQP1), ciliary epithelium (AQP1, AQP4) and retinal Müller cells (AQP4). Mutations in AQP0 produce congenital cataracts in humans. Analysis of knockout mice lacking individual AQPs suggests their involvement in maintenance of corneal and lens transparency, corneal epithelial repair, intraocular pressure (IOP) regulation, retinal signal transduction and retinal swelling following injury. The mouse phenotype findings implicate AQPs as potential drug targets for therapy of elevated IOP and ocular disorders involving the cornea, lens and retina. However, much research remains in defining cell-level mechanisms for the ocular AQP functions, in establishing the relevance to human eye disease of conclusions from knockout mice, and in developing AQP-modulating drugs.
Collapse
Affiliation(s)
- A S Verkman
- Department of Medicine and Physiology, University of California, San Francisco, 1246 Health Sciences East Tower, San Francisco, CA 94143-0521, USA.
| | | | | |
Collapse
|