1
|
Lee EJ, Park DY, Han JC, Kee C. Earliest location of glaucomatous retinal nerve fibre layer damage is determined by intact baseline RNFL thickness profile. Br J Ophthalmol 2024:bjo-2024-325630. [PMID: 39542709 DOI: 10.1136/bjo-2024-325630] [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: 04/03/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND/AIMS To identify whether an intact retinal nerve fibre layer (RNFL) thickness profile could determine the location of the earliest RNFL defect in glaucoma. METHODS Retrospective longitudinal cohort study of patients with initial unilateral glaucoma who eventually developed new glaucoma in the fellow eye. Guided progression analysis (GPA) of serial optical coherence tomography (OCT) examinations was used to identify the angular locations of the earliest RNFL defect, peak and temporal edge of the baseline RNFL profile and major vessels based on the peripapillary OCT scan circle. RESULTS We identified 112 new RNFL defects in 109 fellow eyes of 109 consecutive patients. The locations of new defects were superotemporal (22 eyes), inferotemporal (87 eyes) and papillomacular bundle (3 eyes), respectively. Overall, the midpoint location of the earliest RNFL defect strongly coincided with the RNFL peak (p<0.001). Specifically, the location was mildly (4.3±12.0°) temporal to, rather than at the peak thickness, particularly in eyes with a focal thinning pattern of arcuate bundles (7.8±10.8°). The close topographical relationship was consistent regardless of the interindividual variability in the RNFL profile and vessels, as well as hemispheric locations, and after adjusting for potential factors including age, refractive error, baseline intraocular pressure, tilt ratio, tilt axis and glaucoma diagnosis. CONCLUSION The location of the earliest RNFL defect in glaucoma showed a close relationship with the intact RNFL profile within the same eye, regardless of variations in RNFL, hemispheric location and vessel distribution. In addition, the earliest defect was located mildly temporal to, rather than at, the peak RNFL thickness. The baseline RNFL profile may have a significant role in the regional vulnerability of glaucoma.
Collapse
Affiliation(s)
- Eun Jung Lee
- Samsung Medical Center, Department of Ophthalmology, Sungkyunkwan University School of Medicine, Seoul, Korea (the Republic of)
| | - Do Young Park
- Samsung Medical Center, Department of Ophthalmology, Sungkyunkwan University School of Medicine, Seoul, Korea (the Republic of)
| | - Jong Chul Han
- Samsung Medical Center, Department of Ophthalmology, Sungkyunkwan University School of Medicine, Seoul, Korea (the Republic of)
| | - Changwon Kee
- Kim's Eye Hospital, Seoul, Korea (the Republic of)
| |
Collapse
|
2
|
Quillen SE, Kimball EC, Ritter-Gordy KA, Du L, Yuan Z, Pease ME, Madhoun S, Nguyen TD, Johnson TV, Quigley HA, Pitha IF. The Mechanisms of Neuroprotection by Topical Rho Kinase Inhibition in Experimental Mouse Glaucoma and Optic Neuropathy. Invest Ophthalmol Vis Sci 2024; 65:43. [PMID: 39565302 PMCID: PMC11583991 DOI: 10.1167/iovs.65.13.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
Purpose The purpose of this study was to delineate the neuroprotective mechanisms of topical 2% ripasudil (Rip), a Rho kinase (ROCK) inhibitor. Methods In 340 mice, scheduled 2% Rip or balanced salt solution (BSS) saline drops were intermittently, unilaterally delivered. Intracameral microbead glaucoma (GL) injection increased intraocular pressure (IOP) from 1 day to 6 weeks (6W), whereas other mice underwent optic nerve (ON) crush. Retinal ganglion cell (RGC) loss was assessed using retinal wholemount anti-RNA Binding Protein with Multiple Splicing (RBPMS) labeling and ON axon counts. Axonal transport was quantified with β-amyloid precursor protein (APP) immunolocalization. Micro-Western (Wes) analysis quantified protein expression. Immunofluorescent expression of ROCK pathway molecules, quantitative astrocyte structural changes, and ON biomechanical strains (explanted eyes) were evaluated. ROCK activity assays were conducted in separate ON regions. Results At 6W GL, mean RGC axon loss was 6.6 ± 13.3% in Rip and 36.3 ± 30.9% in BSS (P = 0.04, n = 10/group). RGC soma loss after crush was lower with Rip (68.6 ± 8.2%) than BSS (80.5 ± 5.7%, P = 0.006, n = 10/group). After 6W GL, RGC soma loss was lower with Rip (34 ± 5.0%) than BSS (51 ± 8.1%, P = 0.03, n = 10/group). Axonal transport of APP within the unmyelinated ON (UON) was unaffected by Rip. Maximum principal mechanical strains increased similarly in Rip and BSS-treated mice. Retinal ROCK 1 and 2 activity was reduced by Rip in GL eyes. The pROCK2/ROCK2 protein ratio rose in the retina of BSS GL eyes, but not in Rip GL eyes. Conclusions Topical Rip reduced RGC loss in GL and ON crush, with suppression of ROCK signaling in the retina and ON. The neuroprotection mechanisms appear to involve effects on both RGC and astrocyte responses to IOP elevation.
Collapse
Affiliation(s)
- Sarah E Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Elizabeth C Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Kelsey A Ritter-Gordy
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Liya Du
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Zhuochen Yuan
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Mary E Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Salaheddine Madhoun
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Thao D Nguyen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Thomas V Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ian F Pitha
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
3
|
Waxman S, Schilpp H, Linton A, Jakobs TC, Sigal IA. Morphological comparison of astrocytes in the lamina cribrosa and glial lamina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.610493. [PMID: 39314351 PMCID: PMC11418941 DOI: 10.1101/2024.09.07.610493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Purpose Although the mechanisms underlying glaucomatous neurodegeneration are not yet well understood, cellular and small animal models suggest that LC astrocytes undergo early morphologic and functional changes, indicating their role as early responders to glaucomatous stress. These models, however, lack the LC found in larger animals and humans, leaving the in situ morphology of LC astrocytes and their role in glaucoma initiation underexplored. In this work, we aimed to characterize the morphology of LC astrocytes in situ and determine differences and similarities with astrocytes in the mouse glial lamina (GL), the analogous structure in a prominent glaucoma model. Methods Astrocytes in the LCs of twenty-two eyes from goats, sheep, and pigs were stochastically labeled via Multicolor DiOlistics and imaged in situ using confocal microscopy. 3D models of DiOlistically-labeled LC astrocytes and hGFAPpr-GFP mouse GL astrocytes were constructed to quantify morphological features related to astrocyte functions. LC and GL astrocyte cross-pore contacts, branching complexity, branch tortuosity, and cell and branch span were compared. Results LC astrocytes displayed distinct spatial relationships with collagen, greater branching complexity, and higher branch tortuosity compared to GL astrocytes. Despite substantial differences in their anatomical environments, LC and GL astrocytes had similar cell and branch spans. Conclusions Astrocyte morphology in the LC was characterized through Multicolor DiOlistic labeling. LC and GL astrocytes have both distinct and shared morphological features. Further research is needed to understand the potentially unique roles of LC astrocytes in glaucoma initiation and progression.
Collapse
Affiliation(s)
- Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Hannah Schilpp
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Ashley Linton
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Tatjana C. Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | - Ian A. Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
4
|
Cullen PF, Gammerdinger WJ, Sui SJH, Mazumder AG, Sun D. Transcriptional profiling of retinal astrocytes identifies a specific marker and points to functional specialization. Glia 2024; 72:1604-1628. [PMID: 38785355 PMCID: PMC11262981 DOI: 10.1002/glia.24571] [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: 01/03/2024] [Revised: 04/19/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Astrocyte heterogeneity is an increasingly prominent research topic, and studies in the brain have demonstrated substantial variation in astrocyte form and function, both between and within regions. In contrast, retinal astrocytes are not well understood and remain incompletely characterized. Along with optic nerve astrocytes, they are responsible for supporting retinal ganglion cell axons and an improved understanding of their role is required. We have used a combination of microdissection and Ribotag immunoprecipitation to isolate ribosome-associated mRNA from retinal astrocytes and investigate their transcriptome, which we also compared to astrocyte populations in the optic nerve. Astrocytes from these regions are transcriptionally distinct, and we identified retina-specific astrocyte genes and pathways. Moreover, although they share much of the "classical" gene expression patterns of astrocytes, we uncovered unexpected variation, including in genes related to core astrocyte functions. We additionally identified the transcription factor Pax8 as a highly specific marker of retinal astrocytes and demonstrated that these astrocytes populate not only the retinal surface, but also the prelaminar region at the optic nerve head. These findings are likely to contribute to a revised understanding of the role of astrocytes in the retina.
Collapse
Affiliation(s)
- Paul F Cullen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114 USA
| | - William J Gammerdinger
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Shannan J Ho Sui
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Arpan G Mazumder
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114 USA
| | - Daniel Sun
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
5
|
Pitha I, Du L, Nguyen TD, Quigley H. IOP and glaucoma damage: The essential role of optic nerve head and retinal mechanosensors. Prog Retin Eye Res 2024; 99:101232. [PMID: 38110030 PMCID: PMC10960268 DOI: 10.1016/j.preteyeres.2023.101232] [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: 08/17/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
There are many unanswered questions on the relation of intraocular pressure to glaucoma development and progression. IOP itself cannot be distilled to a single, unifying value, because IOP level varies over time, differs depending on ocular location, and can be affected by method of measurement. Ultimately, IOP level creates mechanical strain that affects axonal function at the optic nerve head which causes local extracellular matrix remodeling and retinal ganglion cell death - hallmarks of glaucoma and the cause of glaucomatous vision loss. Extracellular tissue strain at the ONH and lamina cribrosa is regionally variable and differs in magnitude and location between healthy and glaucomatous eyes. The ultimate targets of IOP-induced tissue strain in glaucoma are retinal ganglion cell axons at the optic nerve head and the cells that support axonal function (astrocytes, the neurovascular unit, microglia, and fibroblasts). These cells sense tissue strain through a series of signals that originate at the cell membrane and alter cytoskeletal organization, migration, differentiation, gene transcription, and proliferation. The proteins that translate mechanical stimuli into molecular signals act as band-pass filters - sensing some stimuli while ignoring others - and cellular responses to stimuli can differ based on cell type and differentiation state. Therefore, to fully understand the IOP signals that are relevant to glaucoma, it is necessary to understand the ultimate cellular targets of IOP-induced mechanical stimuli and their ability to sense, ignore, and translate these signals into cellular actions.
Collapse
Affiliation(s)
- Ian Pitha
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Glaucoma Center of Excellence, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liya Du
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thao D Nguyen
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Harry Quigley
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Glaucoma Center of Excellence, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
van Koeverden AK, Afiat BC, Nguyen CT, Bui BV, Lee PY. Understanding how ageing impacts ganglion cell susceptibility to injury in glaucoma. Clin Exp Optom 2024; 107:147-155. [PMID: 37980904 DOI: 10.1080/08164622.2023.2279734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023] Open
Abstract
Glaucoma is a leading cause of blindness worldwide, with a marked increase in prevalence with advancing age. Due to the multifactorial nature of glaucoma pathogenesis, dissecting how ageing impacts upon glaucoma risk requires analysis and synthesis of evidence from a vast literature. While there is a wealth of human clinical studies examining glaucoma pathogenesis and why older patients have increased risk, many aspects of the disease such as adaptations of retinal ganglion cells to stress, autophagy and the role of glial cells in glaucoma, require the use of animal models to study the complex cellular processes and interactions. Additionally, the accelerated nature of ageing in rodents facilitates the longitudinal study of changes that would not be feasible in human clinical studies. This review article examines evidence derived predominantly from rodent models on how the ageing process impacts upon various aspects of glaucoma pathology from the retinal ganglion cells themselves, to supporting cells and tissues such as glial cells, connective tissue and vasculature, in addition to oxidative stress and autophagy. An improved understanding of how ageing modifies these factors may lead to the development of different therapeutic strategies that target specific risk factors or processes involved in glaucoma.
Collapse
Affiliation(s)
- Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Brianna C Afiat
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Christine To 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
| | - Pei Ying Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Cameron EG, Nahmou M, Toth AB, Heo L, Tanasa B, Dalal R, Yan W, Nallagatla P, Xia X, Hay S, Knasel C, Stiles TL, Douglas C, Atkins M, Sun C, Ashouri M, Bian M, Chang KC, Russano K, Shah S, Woodworth MB, Galvao J, Nair RV, Kapiloff MS, Goldberg JL. A molecular switch for neuroprotective astrocyte reactivity. Nature 2024; 626:574-582. [PMID: 38086421 PMCID: PMC11384621 DOI: 10.1038/s41586-023-06935-3] [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: 09/21/2022] [Accepted: 12/05/2023] [Indexed: 01/27/2024]
Abstract
The intrinsic mechanisms that regulate neurotoxic versus neuroprotective astrocyte phenotypes and their effects on central nervous system degeneration and repair remain poorly understood. Here we show that injured white matter astrocytes differentiate into two distinct C3-positive and C3-negative reactive populations, previously simplified as neurotoxic (A1) and neuroprotective (A2)1,2, which can be further subdivided into unique subpopulations defined by proliferation and differential gene expression signatures. We find the balance of neurotoxic versus neuroprotective astrocytes is regulated by discrete pools of compartmented cyclic adenosine monophosphate derived from soluble adenylyl cyclase and show that proliferating neuroprotective astrocytes inhibit microglial activation and downstream neurotoxic astrocyte differentiation to promote retinal ganglion cell survival. Finally, we report a new, therapeutically tractable viral vector to specifically target optic nerve head astrocytes and show that raising nuclear or depleting cytoplasmic cyclic AMP in reactive astrocytes inhibits deleterious microglial or macrophage cell activation and promotes retinal ganglion cell survival after optic nerve injury. Thus, soluble adenylyl cyclase and compartmented, nuclear- and cytoplasmic-localized cyclic adenosine monophosphate in reactive astrocytes act as a molecular switch for neuroprotective astrocyte reactivity that can be targeted to inhibit microglial activation and neurotoxic astrocyte differentiation to therapeutic effect. These data expand on and define new reactive astrocyte subtypes and represent a step towards the development of gliotherapeutics for the treatment of glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Evan G Cameron
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Michael Nahmou
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Anna B Toth
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Lyong Heo
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Palo Alto, CA, USA
| | - Bogdan Tanasa
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Roopa Dalal
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wenjun Yan
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Pratima Nallagatla
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Palo Alto, CA, USA
| | - Xin Xia
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sarah Hay
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Cara Knasel
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Melissa Atkins
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Catalina Sun
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Masoumeh Ashouri
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Minjuan Bian
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kun-Che Chang
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kristina Russano
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sahil Shah
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
- University of California, San Diego, La Jolla, CA, USA
| | - Mollie B Woodworth
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Joana Galvao
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ramesh V Nair
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Palo Alto, CA, USA
| | - Michael S Kapiloff
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Medicine and Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
8
|
Fernández-Albarral JA, Ramírez AI, de Hoz R, Matamoros JA, Salobrar-García E, Elvira-Hurtado L, López-Cuenca I, Sánchez-Puebla L, Salazar JJ, Ramírez JM. Glaucoma: from pathogenic mechanisms to retinal glial cell response to damage. Front Cell Neurosci 2024; 18:1354569. [PMID: 38333055 PMCID: PMC10850296 DOI: 10.3389/fncel.2024.1354569] [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: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Glaucoma is a neurodegenerative disease of the retina characterized by the irreversible loss of retinal ganglion cells (RGCs) leading to visual loss. Degeneration of RGCs and loss of their axons, as well as damage and remodeling of the lamina cribrosa are the main events in the pathogenesis of glaucoma. Different molecular pathways are involved in RGC death, which are triggered and exacerbated as a consequence of a number of risk factors such as elevated intraocular pressure (IOP), age, ocular biomechanics, or low ocular perfusion pressure. Increased IOP is one of the most important risk factors associated with this pathology and the only one for which treatment is currently available, nevertheless, on many cases the progression of the disease continues, despite IOP control. Thus, the IOP elevation is not the only trigger of glaucomatous damage, showing the evidence that other factors can induce RGCs death in this pathology, would be involved in the advance of glaucomatous neurodegeneration. The underlying mechanisms driving the neurodegenerative process in glaucoma include ischemia/hypoxia, mitochondrial dysfunction, oxidative stress and neuroinflammation. In glaucoma, like as other neurodegenerative disorders, the immune system is involved and immunoregulation is conducted mainly by glial cells, microglia, astrocytes, and Müller cells. The increase in IOP produces the activation of glial cells in the retinal tissue. Chronic activation of glial cells in glaucoma may provoke a proinflammatory state at the retinal level inducing blood retinal barrier disruption and RGCs death. The modulation of the immune response in glaucoma as well as the activation of glial cells constitute an interesting new approach in the treatment of glaucoma.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lidia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Juan J. Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
9
|
Korneva A, Kimball EC, Johnson TV, Quillen SE, Pease ME, Quigley HA, Nguyen TD. Comparison of the Biomechanics of the Mouse Astrocytic Lamina Cribrosa Between Glaucoma and Optic Nerve Crush Models. Invest Ophthalmol Vis Sci 2023; 64:14. [PMID: 38088825 PMCID: PMC10720758 DOI: 10.1167/iovs.64.15.14] [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: 02/20/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Purpose The strain response of the mouse astrocytic lamina (AL) to an ex vivo mechanical test was compared between two protocols: eyes that underwent sustained intraocular pressure (IOP) increase and eyes after optic nerve crush. Methods Chronic IOP elevation was induced by microbead injection or the optic nerve was crushed in mice with widespread green fluorescence. After 3 days or 6 weeks, eyes were inflation tested by a published method of two-photon fluorescence to image the AL. Digital volume correlation was used to calculate strains. Optic nerve axon damage was also evaluated. Results In the central AL but not the peripheral AL, four strains were greater in eyes at the 3-day glaucoma time point than control (P from 0.029 to 0.049, n = 8 eyes per group). Also, at this time point, five strains were greater in the central AL compared to the peripheral AL (P from 0.041 to 0.00003). At the 6-week glaucoma time point, the strains averaged across the specimen, in the central AL, and the peripheral AL were indistinguishable from the respective controls. Strains were not significantly different between controls and eyes 3 days or 6 weeks after crush (n = 8 and 16). Conclusions We found alterations in the ex vivo mechanical behavior in eyes from mice with experimental glaucoma but not in those with crushed optic nerves. The results of this study demonstrate that significant axon injury does not directly affect mechanical behavior of the astrocytic lamina.
Collapse
Affiliation(s)
- Arina Korneva
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Elizabeth C. Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Thomas V. Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sarah E. Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Mary E. Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Harry A. Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Thao D. Nguyen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
10
|
Keuthan CJ, Schaub JA, Wei M, Fang W, Quillen S, Kimball E, Johnson TV, Ji H, Zack DJ, Quigley HA. Regional Gene Expression in the Retina, Optic Nerve Head, and Optic Nerve of Mice with Optic Nerve Crush and Experimental Glaucoma. Int J Mol Sci 2023; 24:13719. [PMID: 37762022 PMCID: PMC10531004 DOI: 10.3390/ijms241813719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
A major risk factor for glaucomatous optic neuropathy is the level of intraocular pressure (IOP), which can lead to retinal ganglion cell axon injury and cell death. The optic nerve has a rostral unmyelinated portion at the optic nerve head followed by a caudal myelinated region. The unmyelinated region is differentially susceptible to IOP-induced damage in rodent models and human glaucoma. While several studies have analyzed gene expression changes in the mouse optic nerve following optic nerve injury, few were designed to consider the regional gene expression differences that exist between these distinct areas. We performed bulk RNA-sequencing on the retina and separately micro-dissected unmyelinated and myelinated optic nerve regions from naïve C57BL/6 mice, mice after optic nerve crush, and mice with microbead-induced experimental glaucoma (total = 36). Gene expression patterns in the naïve unmyelinated optic nerve showed significant enrichment of the Wnt, Hippo, PI3K-Akt, and transforming growth factor β pathways, as well as extracellular matrix-receptor and cell membrane signaling pathways, compared to the myelinated optic nerve and retina. Gene expression changes induced by both injuries were more extensive in the myelinated optic nerve than the unmyelinated region, and greater after nerve crush than glaucoma. Changes present three and fourteen days after injury largely subsided by six weeks. Gene markers of reactive astrocytes did not consistently differ between injury states. Overall, the transcriptomic phenotype of the mouse unmyelinated optic nerve was significantly different from immediately adjacent tissues, likely dominated by expression in astrocytes, whose junctional complexes are inherently important in responding to IOP elevation.
Collapse
Affiliation(s)
- Casey J. Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| | - Julie A. Schaub
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| | - Meihan Wei
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Weixiang Fang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Sarah Quillen
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| | - Elizabeth Kimball
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| | - Thomas V. Johnson
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| | - Hongkai Ji
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
- Departments of Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Harry A. Quigley
- Department of Ophthalmology, Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (C.J.K.)
| |
Collapse
|
11
|
Cullen PF, Sun D. Astrocytes of the eye and optic nerve: heterogeneous populations with unique functions mediate axonal resilience and vulnerability to glaucoma. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1217137. [PMID: 37829657 PMCID: PMC10569075 DOI: 10.3389/fopht.2023.1217137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The role of glia, particularly astrocytes, in mediating the central nervous system's response to injury and neurodegenerative disease is an increasingly well studied topic. These cells perform myriad support functions under physiological conditions but undergo behavioral changes - collectively referred to as 'reactivity' - in response to the disruption of neuronal homeostasis from insults, including glaucoma. However, much remains unknown about how reactivity alters disease progression - both beneficially and detrimentally - and whether these changes can be therapeutically modulated to improve outcomes. Historically, the heterogeneity of astrocyte behavior has been insufficiently addressed under both physiological and pathological conditions, resulting in a fragmented and often contradictory understanding of their contributions to health and disease. Thanks to increased focus in recent years, we now know this heterogeneity encompasses both intrinsic variation in physiological function and insult-specific changes that vary between pathologies. Although previous studies demonstrate astrocytic alterations in glaucoma, both in human disease and animal models, generally these findings do not conclusively link astrocytes to causative roles in neuroprotection or degeneration, rather than a subsequent response. Efforts to bolster our understanding by drawing on knowledge of brain astrocytes has been constrained by the primacy in the literature of findings from peri-synaptic 'gray matter' astrocytes, whereas much early degeneration in glaucoma occurs in axonal regions populated by fibrous 'white matter' astrocytes. However, by focusing on findings from astrocytes of the anterior visual pathway - those of the retina, unmyelinated optic nerve head, and myelinated optic nerve regions - we aim to highlight aspects of their behavior that may contribute to axonal vulnerability and glaucoma progression, including roles in mitochondrial turnover and energy provisioning. Furthermore, we posit that astrocytes of the retina, optic nerve head and myelinated optic nerve, although sharing developmental origins and linked by a network of gap junctions, may be best understood as distinct populations residing in markedly different niches with accompanying functional specializations. A closer investigation of their behavioral repertoires may elucidate not only their role in glaucoma, but also mechanisms to induce protective behaviors that can impede the progressive axonal damage and retinal ganglion cell death that drive vision loss in this devastating condition.
Collapse
Affiliation(s)
- Paul F. Cullen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Daniel Sun
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Korneva A, Kimball EC, Quillen S, Jefferys JL, Nawathe M, Ling YTT, Nguyen TD, Quigley HA. Mechanical strain in the mouse astrocytic lamina increases after exposure to recombinant trypsin. Acta Biomater 2023; 163:312-325. [PMID: 35196555 PMCID: PMC9391529 DOI: 10.1016/j.actbio.2022.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/21/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
The responses of astrocytes in the optic nerve head (ONH) to mechanical and biochemical stimuli are important to understanding the degeneration of retinal ganglion cell axons in glaucoma. The ONH in glaucoma is vulnerable to stress produced by the intraocular pressure (IOP). Notably, after three days of elevated IOP in a mouse model, the junctions between the astrocytic processes and the peripapillary sclera were altered and the structural compliance of the ONH increased. In order to simulate this aspect of glaucomatous remodeling, explanted mouse eyes were treated with TrypLE, a recombinant trypsin enzyme. Treatment with TrypLE caused the periphery of the astrocytic lamina to contract radially by 0.044 ± 0.038. Transmission electron microscopy showed that TrypLE caused a separation of the end-feet of the astrocyte processes from the basement membrane at the junction with the sclera. Inflation testing after treatment with TrypLE caused an increased strain response in the astrocytic lamina compared to the strain response before treatment. The greatest increase was in the radial Green-Lagrange strain, Err = 0.028 ± 0.009, which increased by 340%. The alterations in the microstructure and in the strain response of the astrocytic lamina reported in mouse experimental glaucoma were partially reproduced by experimental treatment of mouse eyes with TrypLE. The results herein suggest that separation of junctions between the astrocyte processes and the sclera may be instrumental in increasing the structural compliance of the ONH after a period of elevated IOP. STATEMENT OF SIGNIFICANCE: Astrocytes of the optic nerve of the eye spread out from edge to edge across the optic nerve in a region referred to as the astrocytic lamina. In an experimental model of glaucoma caused by elevated eye-pressure, there is disruption of the connections between astrocytes and the edge of the astrocytic lamina. We caused a similar event in the lamina by incubating explanted mouse eyes with an enzyme. Disruption of the astrocyte connections to the edge of their tissue caused the tissue to stretch more when we increased the eye-pressure, compared to the control tissue. This work is the first on the tissue of the optic nerve to demonstrate the importance of cell connections in preventing the over-stretching of the astrocytic lamina.
Collapse
Affiliation(s)
- Arina Korneva
- Glaucoma Center of Excellence, Johns Hopkins Wilmer Eye Institute, United States; Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States.
| | - Elizabeth C Kimball
- Glaucoma Center of Excellence, Johns Hopkins Wilmer Eye Institute, United States.
| | - Sarah Quillen
- Glaucoma Center of Excellence, Johns Hopkins Wilmer Eye Institute, United States.
| | - Joan L Jefferys
- Glaucoma Center of Excellence, Johns Hopkins Wilmer Eye Institute, United States.
| | - Manasi Nawathe
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States.
| | - Yik Tung Tracy Ling
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States.
| | - Thao D Nguyen
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Materials Science, Johns Hopkins University, Baltimore, MD 21218, United States.
| | - Harry A Quigley
- Glaucoma Center of Excellence, Johns Hopkins Wilmer Eye Institute, United States; Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
13
|
Keuthan CJ, Schaub J, Wei M, Fang W, Quillen S, Kimball E, Johnson TV, Ji H, Zack DJ, Quigley HA. Regional Gene Expression in the Retina, Optic Nerve Head, and Optic Nerve of Mice with Experimental Glaucoma and Optic Nerve Crush. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.21.529410. [PMID: 36993314 PMCID: PMC10054954 DOI: 10.1101/2023.02.21.529410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A major risk factor for glaucomatous optic neuropathy is the level of intraocular pressure (IOP), which can lead to retinal ganglion cell axon injury and cell death. The optic nerve has a rostral unmyelinated portion at the optic nerve head followed by a caudal myelinated region. The unmyelinated region is differentially susceptible to IOP-induced damage in rodent models and in human glaucoma. While several studies have analyzed gene expression changes in the mouse optic nerve following optic nerve injury, few were designed to consider the regional gene expression differences that exist between these distinct areas. We performed bulk RNA-sequencing on the retina and on separately micro-dissected unmyelinated and myelinated optic nerve regions from naïve C57BL/6 mice, mice after optic nerve crush, and mice with microbead-induced experimental glaucoma (total = 36). Gene expression patterns in the naïve unmyelinated optic nerve showed significant enrichment of the Wnt, Hippo, PI3K-Akt, and transforming growth factor β pathways, as well as extracellular matrix-receptor and cell membrane signaling pathways, compared to the myelinated optic nerve and retina. Gene expression changes induced by both injuries were more extensive in the myelinated optic nerve than the unmyelinated region, and greater after nerve crush than glaucoma. Changes three and fourteen days after injury largely subsided by six weeks. Gene markers of reactive astrocytes did not consistently differ between injury states. Overall, the transcriptomic phenotype of the mouse unmyelinated optic nerve was significantly different from immediately adjacent tissues, likely dominated by expression in astrocytes, whose junctional complexes are inherently important in responding to IOP elevation.
Collapse
Affiliation(s)
- Casey J. Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Julie Schaub
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Meihan Wei
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Weixiang Fang
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Sarah Quillen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Elizabeth Kimball
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Thomas V. Johnson
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Donald J. Zack
- Departments of Ophthalmology, Wilmer Eye Institute, Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Harry A. Quigley
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
14
|
Amato R, Cammalleri M, Melecchi A, Bagnoli P, Porciatti V. Natural History of Glaucoma Progression in the DBA/2J Model: Early Contribution of Müller Cell Gliosis. Cells 2023; 12:cells12091272. [PMID: 37174673 PMCID: PMC10177096 DOI: 10.3390/cells12091272] [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: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Glaucoma is a chronic optic neuropathy characterized by progressive degeneration of retinal ganglion cells (RGCs). Elevated intraocular pressure (IOP) and the resulting mechanical stress are classically considered the main causes of RGC death. However, RGC degeneration and ensuing vision loss often occur independent of IOP, indicating a multifactorial nature of glaucoma, with the likely contribution of glial and vascular function. The aim of the present study was to provide a comprehensive evaluation of the time course of neuro-glial-vascular changes associated with glaucoma progression. We used DBA/2J mice in the age range of 2-15 months as a spontaneous model of glaucoma with progressive IOP elevation and RGC loss typical of human open-angle glaucoma. We found that the onset of RGC degeneration at 10 months of age coincided with that of IOP elevation and vascular changes such as decreased density, increased lacunarity and decreased tight-junction protein zonula occludens (ZO)-1, while hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF) were already significantly upregulated at 6 months of age together with the onset of Müller cell gliosis. Astrocytes, however, underwent significant gliosis at 10 months. These results indicate that Müller cell activation occurs well before IOP elevation, with probable inflammatory consequences, and represents an early event in the glaucomatous process. Early upregulation of HIF-1α and VEGF is likely to contribute to blood retinal barrier failure, facilitating RGC loss. The different time courses of neuro-glial-vascular changes during glaucoma progression provide further insight into the nature of the disease and suggest potential targets for the development of efficient therapeutic intervention aside from IOP lowering.
Collapse
Affiliation(s)
- Rosario Amato
- Department of Biology, University of Pisa, 56127 Pisa, Italy
| | | | | | - Paola Bagnoli
- Department of Biology, University of Pisa, 56127 Pisa, Italy
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
15
|
Sharif NA, Odani-Kawabata N, Lu F, Pinchuk L. FP and EP2 prostanoid receptor agonist drugs and aqueous humor outflow devices for treating ocular hypertension and glaucoma. Exp Eye Res 2023; 229:109415. [PMID: 36803996 DOI: 10.1016/j.exer.2023.109415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/21/2022] [Accepted: 02/08/2023] [Indexed: 02/21/2023]
Abstract
Prostaglandin (PG) receptors represent important druggable targets due to the many diverse actions of PGs in the body. From an ocular perspective, the discovery, development, and health agency approvals of prostaglandin F (FP) receptor agonists (FPAs) have revolutionized the medical treatment of ocular hypertension (OHT) and glaucoma. FPAs, such as latanoprost, travoprost, bimatoprost, and tafluprost, powerfully lower and control intraocular pressure (IOP), and became first-line therapeutics to treat this leading cause of blindness in the late 1990s to early 2000s. More recently, a latanoprost-nitric oxide (NO) donor conjugate, latanoprostene bunod, and a novel FP/EP3 receptor dual agonist, sepetaprost (ONO-9054 or DE-126), have also demonstrated robust IOP-reducing activity. Moreover, a selective non-PG prostanoid EP2 receptor agonist, omidenepag isopropyl (OMDI), was discovered, characterized, and has been approved in the United States, Japan and several other Asian countries for treating OHT/glaucoma. FPAs primarily enhance uveoscleral (UVSC) outflow of aqueous humor (AQH) to reduce IOP, but cause darkening of the iris and periorbital skin, uneven thickening and elongation of eyelashes, and deepening of the upper eyelid sulcus during chronic treatment. In contrast, OMDI lowers and controls IOP by activation of both the UVSC and trabecular meshwork outflow pathways, and it has a lower propensity to induce the aforementioned FPA-induced ocular side effects. Another means to address OHT is to physically promote the drainage of the AQH from the anterior chamber of the eye of patients with OHT/glaucoma. This has successfully been achieved by the recent approval and introduction of miniature devices into the anterior chamber by minimally invasive glaucoma surgeries. This review covers the three major aspects mentioned above to highlight the etiology of OHT/glaucoma, and the pharmacotherapeutics and devices that can be used to combat this blinding ocular disease.
Collapse
Affiliation(s)
- Najam A Sharif
- Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, USA; Singapore Eye Research Institute, Singapore; Eye-ACP Duke-National University of Singapore Medical School, Singapore; Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, TX, USA; Department of Pharmacy Sciences, Creighton University, Omaha, NE, USA; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA; Imperial College of Science and Technology, St. Mary's Campus, London, UK; Institute of Ophthalmology, University College London, London, UK.
| | | | - Fenghe Lu
- Product Development Division, Santen Inc., Emeryville, CA, USA
| | - Leonard Pinchuk
- Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, USA; Biomedical Engineering Department, University of Miami, Miami, FL, USA
| |
Collapse
|
16
|
St-Pierre MK, Carrier M, González Ibáñez F, Khakpour M, Wallman MJ, Parent M, Tremblay MÈ. Astrocytes display ultrastructural alterations and heterogeneity in the hippocampus of aged APP-PS1 mice and human post-mortem brain samples. J Neuroinflammation 2023; 20:73. [PMID: 36918925 PMCID: PMC10015698 DOI: 10.1186/s12974-023-02752-7] [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: 12/15/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
The past decade has witnessed increasing evidence for a crucial role played by glial cells, notably astrocytes, in Alzheimer's disease (AD). To provide novel insights into the roles of astrocytes in the pathophysiology of AD, we performed a quantitative ultrastructural characterization of their intracellular contents and parenchymal interactions in an aged mouse model of AD pathology, as aging is considered the main risk factor for developing AD. We compared 20-month-old APP-PS1 and age-matched C57BL/6J male mice, among the ventral hippocampus CA1 strata lacunosum-moleculare and radiatum, two hippocampal layers severely affected by AD pathology. Astrocytes in both layers interacted more with synaptic elements and displayed more ultrastructural markers of increased phagolysosomal activity in APP-PS1 versus C57BL6/J mice. In addition, we investigated the ultrastructural heterogeneity of astrocytes, describing in the two examined layers a dark astrocytic state that we characterized in terms of distribution, interactions with AD hallmarks, and intracellular contents. This electron-dense astrocytic state, termed dark astrocytes, was observed throughout the hippocampal parenchyma, closely associated with the vasculature, and possessed several ultrastructural markers of cellular stress. A case study exploring the hippocampal head of an aged human post-mortem brain sample also revealed the presence of a similar electron-dense, dark astrocytic state. Overall, our study provides the first ultrastructural quantitative analysis of astrocytes among the hippocampus in aged AD pathology, as well as a thorough characterization of a dark astrocytic state conserved from mouse to human.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Mohammadparsa Khakpour
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
| | - Marie-Josée Wallman
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Quebec City, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada.,CERVO Brain Research Center, Quebec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada. .,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada. .,Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada. .,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada. .,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada. .,Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
17
|
Ma D, Pasquale LR, Girard MJA, Leung CKS, Jia Y, Sarunic MV, Sappington RM, Chan KC. Reverse translation of artificial intelligence in glaucoma: Connecting basic science with clinical applications. FRONTIERS IN OPHTHALMOLOGY 2023; 2:1057896. [PMID: 36866233 PMCID: PMC9976697 DOI: 10.3389/fopht.2022.1057896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/05/2022] [Indexed: 04/16/2023]
Abstract
Artificial intelligence (AI) has been approved for biomedical research in diverse areas from bedside clinical studies to benchtop basic scientific research. For ophthalmic research, in particular glaucoma, AI applications are rapidly growing for potential clinical translation given the vast data available and the introduction of federated learning. Conversely, AI for basic science remains limited despite its useful power in providing mechanistic insight. In this perspective, we discuss recent progress, opportunities, and challenges in the application of AI in glaucoma for scientific discoveries. Specifically, we focus on the research paradigm of reverse translation, in which clinical data are first used for patient-centered hypothesis generation followed by transitioning into basic science studies for hypothesis validation. We elaborate on several distinctive areas of research opportunities for reverse translation of AI in glaucoma including disease risk and progression prediction, pathology characterization, and sub-phenotype identification. We conclude with current challenges and future opportunities for AI research in basic science for glaucoma such as inter-species diversity, AI model generalizability and explainability, as well as AI applications using advanced ocular imaging and genomic data.
Collapse
Affiliation(s)
- Da Ma
- School of Medicine, Wake Forest University, Winston-Salem, NC, United States
- Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, United States
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Louis R. Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Michaël J. A. Girard
- Ophthalmic Engineering & Innovation Laboratory (OEIL), Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Institute for Molecular and Clinical Ophthalmology, Basel, Switzerland
| | | | - Yali Jia
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Marinko V. Sarunic
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Rebecca M. Sappington
- School of Medicine, Wake Forest University, Winston-Salem, NC, United States
- Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, United States
| | - Kevin C. Chan
- Departments of Ophthalmology and Radiology, Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, United States
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, United States
| |
Collapse
|
18
|
Czerpak CA, Kashaf MS, Zimmerman BK, Quigley HA, Nguyen TD. The Strain Response to Intraocular Pressure Decrease in the Lamina Cribrosa of Patients with Glaucoma. Ophthalmol Glaucoma 2023; 6:11-22. [PMID: 35863747 PMCID: PMC9849479 DOI: 10.1016/j.ogla.2022.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To measure biomechanical strains in the lamina cribrosa (LC) of living human eyes with intraocular pressure (IOP) lowering. DESIGN Cohort study. PARTICIPANTS Patients with glaucoma underwent imaging before and after laser suturelysis after trabeculectomy surgery (29 image pairs; 26 persons). INTERVENTION Noninvasive imaging of the eye. MAIN OUTCOME MEASURES Strains in optic nerve head tissue and changes in depths of the anterior border of the LC. RESULTS Intraocular pressure decreases caused the LC to expand in thickness in the anterior-posterior strain (Ezz = 0.94 ± 1.2%; P = 0.00020) and contract in radius in the radial strain (Err = - 0.19 ± 0.33%; P = 0.0043). The mean LC depth did not significantly change with IOP lowering (1.33 ± 6.26 μm; P = 0.26). A larger IOP decrease produced a larger, more tensile Ezz (P < 0.0001), greater maximum principal strain (Emax; P < 0.0001), and greater maximum shear strain (Γmax; P < 0.0001). The average LC depth change was associated with the Γmax and radial-circumferential shear strain (Erθ; P < 0.02) but was not significantly related to tensile or compressive strains. An analysis by clock hour showed that in temporal clock hours 3 to 6, a more anterior LC movement was associated with a more positive Emax, and in clock hours 3, 5, and 6, it was associated with a more positive Γmax. At 10 o'clock, a more posterior LC movement was related to a more positive Emax (P < 0.004). Greater compliance (strain/ΔIOP) of Emax (P = 0.044), Γmax (P = 0.052), and Erθ (P = 0.018) was associated with a thinner retinal nerve fiber layer. Greater compliance of Emax (P = 0.041), Γmax (P = 0.021), Erθ (P = 0.024), and in-plane shear strain (Erz; P = 0.0069) was associated with more negative mean deviations. Greater compliance of Γmax (P = 0.055), Erθ (P = 0.040), and Erz (P = 0.015) was associated with lower visual field indices. CONCLUSIONS With IOP lowering, the LC moves either into or out of the eye but, on average, expands in thickness and contracts in radius. Shear strains are nearly as substantial as in-plane strains. Biomechanical strains are more compliant in eyes with greater glaucoma damage. This work was registered at ClinicalTrials.gov as NCT03267849.
Collapse
Affiliation(s)
- Cameron A Czerpak
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland.
| | - Michael Saheb Kashaf
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Brandon K Zimmerman
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland
| | - Harry A Quigley
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Thao D Nguyen
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland; Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
19
|
Madhoun S, Martins MTC, Korneva A, Johnson TV, Kimball E, Quillen S, Pease ME, Edwards M, Quigley H. Effects of experimental glaucoma in Lama1 nmf223 mutant mice. Exp Eye Res 2023; 226:109341. [PMID: 36476399 PMCID: PMC10204621 DOI: 10.1016/j.exer.2022.109341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
To identify changes in response to experimental intraocular pressure (IOP) elevation associated with the laminin α1 nmf223 mutation in mice. Laminin mutant (LM) mice (Lama1nmf223) and C57BL/6J (B6) mice in two age groups each (4-5 months and >1 year) underwent intracameral microbead injections to produce unilaterally elevated IOP. We assessed axonal transport block of immunofluorescently labeled amyloid precursor protein (APP) after 3 days and retinal ganglion cell (RGC) axon loss after 6 weeks. Light, electron and fluorescent microscopy was used to study baseline anatomic differences and effects of 3-day IOP elevation in younger LM mice. In younger mice of both LM and B6 strains, elevated IOP led to increased APP block in the retina, prelaminar optic nerve head (preONH), unmyelinated optic nerve (UON), and myelinated optic nerve (MON). APP blockade not significantly different between younger B6 and LM mouse strains. Older LM mice had greater APP accumulation in both control and glaucoma eyes compared to older B6, however, accumulation was not significantly greater in LM glaucoma eyes compared to LM controls. Axon loss at 6 weeks was 12.2% in younger LM and 18.7% in younger B6 mice (difference between strains, p = 0.22, Mann Whitney test). Untreated LM optic nerve area was lower compared to B6 (nerve area, p < 0.0001, t-test). Aberrant axon bundles, as well as defects, thickening and reduplication of pia mater, were seen in the optic nerves of younger LM mice. Axonal transport blockade significantly differed between old B6 and old LM mice in control and glaucoma eyes, and younger LM mice had abnormal axon paths and lower optic nerve area.
Collapse
Affiliation(s)
- Salaheddine Madhoun
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| | | | - Arina Korneva
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas V Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mary Ellen Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Malia Edwards
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Harry Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
20
|
Lysyl oxidase-like 1 deficiency alters ultrastructural and biomechanical properties of the peripapillary sclera in mice. Matrix Biol Plus 2022; 16:100120. [PMID: 36060791 PMCID: PMC9436796 DOI: 10.1016/j.mbplus.2022.100120] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022] Open
Abstract
Lysyl oxidate-like 1 knockout (Loxl1-/-) mice have decreased vision without elevated intraocular pressure. Loxl1-/- mice exhibit biometric changes of the anterior segment of the eye. Loxl1-/- mice have altered elastin and collagen structure in peripapillary sclera. Structural alternations of peripapillary sclera correlate with its increased stiffness in Loxl1-/- mice.
Lysyl oxidase-like 1 encoded by the LOXL1 gene is a member of the lysyl oxidase family of enzymes that are important in the maintenance of extracellular matrix (ECM)-rich tissue. LOXL1 is important for proper elastic fiber formation and mice lacking LOXL1 (Loxl1−/−) exhibit systemic elastic fiber disorders, such as pelvic organ prolapse, a phenotype associated with exfoliation syndrome (XFS) in humans. Patients with XFS have a significant risk of developing exfoliation glaucoma (XFG), a severe form of glaucoma, which is a neurodegenerative condition leading to irreversible blindness if not detected and treated in a timely fashion. Although Loxl1−/− mice have been used extensively to investigate mechanisms of pelvic organ prolapse, studies of eyes in those mice are limited and some showed inconsistent ocular phenotypes. In this study we demonstrate that Loxl1−/− mice have significant anterior segment biometric abnormalities which recapitulate some human XFS features. We then focused on the peripapillary sclera (PPS), a critical structure for maintaining optic nerve health. We discovered quantitative and qualitive changes in ultrastructure of PPS, such as reduced elastic fibers, enlarged collagen fibrils, and transformed collagen lamella organization detected by transmission electron microscopy (TEM). Importantly, these changes corelate with altered tissue biomechanics detected by Atomic Force Microscopy (AFM) of PPS in mice. Together, our results support a crucial role for LOXL1 in ocular tissue structure and biomechanics, and Loxl1−/− mice could be a valuable resource for understanding the role of scleral tissue biomechanics in ocular disease.
Collapse
|
21
|
Tang Y, Chen Y, Chen D. The heterogeneity of astrocytes in glaucoma. Front Neuroanat 2022; 16:995369. [PMID: 36466782 PMCID: PMC9714578 DOI: 10.3389/fnana.2022.995369] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/31/2022] [Indexed: 09/10/2023] Open
Abstract
Glaucoma is a leading cause of blindness with progressive degeneration of retinal ganglion cells. Aging and increased intraocular pressure (IOP) are major risk factors. Lowering IOP does not always stop the disease progression. Alternative ways of protecting the optic nerve are intensively studied in glaucoma. Astrocytes are macroglia residing in the retina, optic nerve head (ONH), and visual brain, which keep neuronal homeostasis, regulate neuronal activities and are part of the immune responses to the retina and brain insults. In this brief review, we discuss the activation and heterogeneity of astrocytes in the retina, optic nerve head, and visual brain of glaucoma patients and animal models. We also discuss some recent transgenic and gene knockout studies using glaucoma mouse models to clarify the role of astrocytes in the pathogenesis of glaucoma. Astrocytes are heterogeneous and play crucial roles in the pathogenesis of glaucoma, especially in the process of neuroinflammation and mitochondrial dysfunction. In astrocytes, overexpression of Stat3 or knockdown of IκKβ/p65, caspase-8, and mitochondrial uncoupling proteins (Ucp2) can reduce ganglion cell loss in glaucoma mouse models. Based on these studies, therapeutic strategies targeting the heterogeneity of reactive astrocytes by enhancing their beneficial reactivity or suppressing their detrimental reactivity are alternative options for glaucoma treatment in the future.
Collapse
Affiliation(s)
- Yunjing Tang
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongjiang Chen
- The School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Danian Chen
- Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Chaudhary P, Stowell C, Reynaud J, Gardiner SK, Yang H, Williams G, Williams I, Marsh-Armstrong N, Burgoyne CF. Optic Nerve Head Myelin-Related Protein, GFAP, and Iba1 Alterations in Non-Human Primates With Early to Moderate Experimental Glaucoma. Invest Ophthalmol Vis Sci 2022; 63:9. [PMID: 36239974 PMCID: PMC9586137 DOI: 10.1167/iovs.63.11.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Purpose The purpose of this study was to test if optic nerve head (ONH) myelin basic protein (MBP), 2′,3′-cyclic nucleotide 3′-phosphodiesterase (CNPase), glial fibrillary acidic protein (GFAP), and ionized calcium binding adaptor molecule 1 (Iba1) proteins are altered in non-human primate (NHP) early/moderate experimental glaucoma (EG). Methods Following paraformaldehyde perfusion, control and EG eye ONH tissues from four NHPs were paraffin embedded and serially (5 µm) vertically sectioned. Anti-MBP, CNPase, GFAP, Iba1, and nuclear dye-stained sections were imaged using sub-saturating light intensities. Whole-section images were segmented creating anatomically consistent laminar (L) and retrolaminar (RL) regions/sub-regions. EG versus control eye intensity/pixel-cluster density data within L and two RL regions (RL1 [1-250 µm]/RL2 [251-500 µm] from L) were compared using random effects models within the statistical program “R.” Results EG eye retinal nerve fiber loss ranged from 0% to 20%. EG eyes’ MBP and CNPase intensity were decreased within the RL1 (MBP = 31.4%, P < 0.001; CNPase =62.3%, P < 0.001) and RL2 (MBP = 19.6%, P < 0.001; CNPase = 56.1%, P = 0.0004) regions. EG eye GFAP intensity was decreased in the L (41.6%, P < 0.001) and RL regions (26.7% for RL1, and 28.4% for RL2, both P < 0.001). Iba1+ and NucBlue pixel-cluster density were increased in the laminar (28.2%, P = 0.03 and 16.6%, P = 0.008) and both RL regions (RL1 = 37.3%, P = 0.01 and 23.7%, P = 0.0002; RL2 = 53.7%, P = 0.002 and 33.2%, P < 0.001). Conclusions Retrolaminar myelin disruption occurs early in NHP EG and may be accompanied by laminar and retrolaminar decreases in astrocyte process labeling and increases in microglial/ macrophage density. The mechanistic and therapeutic implications of these findings warrant further study.
Collapse
Affiliation(s)
- Priya Chaudhary
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Cheri Stowell
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Juan Reynaud
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Stuart K Gardiner
- Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Hongli Yang
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Galen Williams
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Imee Williams
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | | | - Claude F Burgoyne
- Optic Nerve Head Research Laboratory, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States.,Discoveries in Sight, Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| |
Collapse
|
23
|
Guan C, Pease ME, Quillen S, Ling YTT, Li X, Kimball E, Johnson TV, Nguyen TD, Quigley HA. Quantitative Microstructural Analysis of Cellular and Tissue Remodeling in Human Glaucoma Optic Nerve Head. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 36269186 PMCID: PMC9617510 DOI: 10.1167/iovs.63.11.18] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/23/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose To measure quantitatively changes in lamina cribrosa (LC) cell and connective tissue structure in human glaucoma eyes. Methods We studied 27 glaucoma and 19 age-matched non-glaucoma postmortem eyes. In 25 eyes, LC cross-sections were examined by confocal and multiphoton microscopy to quantify structures identified by anti-glial fibrillary acidic protein (GFAP), phalloidin-labeled F-actin, nuclear 4',6-diamidino-2-phenylindole (DAPI), and by second harmonic generation imaging of LC beams. Additional light and transmission electron microscopy were performed in 21 eyes to confirm features of LC remodeling, including immunolabeling by anti-SOX9 and anti-collagen IV. All glaucoma eyes had detailed clinical histories of open-angle glaucoma status, and degree of axon loss was quantified in retrolaminar optic nerve cross-sections. Results Within LC pores, the proportionate area of both GFAP and F-actin processes was significantly lower in glaucoma eyes than in controls (P = 0.01). Nuclei were rounder (lower median aspect ratio) in glaucoma specimens (P = 0.02). In models assessing degree of glaucoma damage, F-actin process width was significantly wider in glaucoma eyes with more damage (P = 0.024), average LC beam width decreased with worse glaucoma damage (P = 0.042), and nuclear count per square millimeter rose with worse damage (P = 0.019). The greater cell count in LC pores represented 92.3% astrocytes by SOX9 labeling. The results are consistent with replacement of axons in LC pores by basement membrane labeled by anti-collagen IV and in-migrating astrocytes. Conclusions Alteration in LC structure in glaucoma involves migration of astrocytes into axonal bundles, change in astrocyte orientation and processes, production of basement membrane material, and thinning of connective tissue beams.
Collapse
Affiliation(s)
- Carolyn Guan
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Mary Ellen Pease
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Sarah Quillen
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Yik Tung Tracy Ling
- Departments of Mechanical Engineering and Materials Science & Engineering, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Ximin Li
- Department of Biostatistics, The Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Elizabeth Kimball
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Thomas V. Johnson
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Thao D. Nguyen
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
- Departments of Mechanical Engineering and Materials Science & Engineering, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Harry A. Quigley
- Wilmer Ophthalmological Institute, School of Medicine, The Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
24
|
Li Z, Peng F, Liu Z, Li S, Li L, Qian X. Mechanobiological responses of astrocytes in optic nerve head due to biaxial stretch. BMC Ophthalmol 2022; 22:368. [PMID: 36114477 PMCID: PMC9482189 DOI: 10.1186/s12886-022-02592-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Background Elevated intraocular pressure (IOP) is the main risk factor for glaucoma, which might cause the activation of astrocytes in optic nerve head. To determine the effect of mechanical stretch on the astrocytes, we investigated the changes in cell phenotype, proteins of interest and signaling pathways under biaxial stretch. Method The cultured astrocytes in rat optic nerve head were stretched biaxially by 10 and 17% for 24 h, respectively. Then, we detected the morphology, proliferation and apoptosis of the stretched cells, and performed proteomics analysis. Protein expression was analyzed by Isobaric tags for relative and absolute quantification (iTRAQ) mass spectrometry. Proteins of interest and signaling pathways were screened using Gene Ontology enrichment analysis and pathway enrichment analysis, and the results were verified by western blot and the gene-chip data from Gene Expression Omnibus (GEO) database. Result The results showed that rearrangement of the actin cytoskeleton in response to stimulation by mechanical stress and proliferation rate of astrocytes decreased under 10 and 17% stretch condition, while there was no significant difference on the apoptosis rate of astrocytes in both groups. In the iTRAQ quantitative experiment, there were 141 differential proteins in the 10% stretch group and 140 differential proteins in the 17% stretch group. These proteins include low-density lipoprotein receptor-related protein (LRP6), caspase recruitment domain family, member 10 (CARD10), thrombospondin 1 (THBS1) and tetraspanin (CD81). The western blot results of LRP6, THBS1 and CD81 were consistent with that of iTRAQ experiment. ANTXR2 and CARD10 were both differentially expressed in the mass spectrometry results and GEO database. We also screened out the signaling pathways associated with astrocyte activation, including Wnt/β–catenin pathway, NF-κB signaling pathway, PI3K-Akt signaling pathway, MAPK signaling pathway, Jak-STAT signaling pathway, ECM-receptor interaction, and transforming growth factor-β (TGF-β) signaling pathway. Conclusion Mechanical stimulation can induce changes in cell phenotype, some proteins and signaling pathways, which might be associated with astrocyte activation. These proteins and signaling pathways may help us have a better understanding on the activation of astrocytes and the role astrocyte activation played in glaucomatous optic neuropathy. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-022-02592-8.
Collapse
|
25
|
Kimball EC, Quillen S, Pease ME, Keuthan C, Nagalingam A, Zack DJ, Johnson TV, Quigley HA. Aquaporin 4 is not present in normal porcine and human lamina cribrosa. PLoS One 2022; 17:e0268541. [PMID: 35709078 PMCID: PMC9202842 DOI: 10.1371/journal.pone.0268541] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/03/2022] [Indexed: 02/02/2023] Open
Abstract
Aquaporin 4 is absent from astrocytes in the rodent optic nerve head, despite high expression in the retina and myelinated optic nerve. The purpose of this study was to quantify regional aquaporin channel expression in astrocytes of the porcine and human mouse optic nerve (ON). Ocular tissue sections were immunolabeled for aquaporins 1(AQP1), 4(AQP4), and 9(AQP9), myelin basic protein (MBP), glial fibrillary acidic protein (GFAP) and alpha-dystroglycan (αDG) for their presence in retina, lamina, myelin transition zone (MTZ, region just posterior to lamina) and myelinated ON (MON). Semi- quantification of AQP4 labeling & real-time quantitative PCR (qPCR) data were analyzed in retina and ON tissue. Porcine and control human eyes had abundant AQP4 in Müller cells, retinal astrocytes, and myelinated ON (MON), but minimal expression in the lamina cribrosa. AQP1 and AQP9 were present in retina, but not in the lamina. Immunolabeling of GFAP and αDG was similar in lamina, myelin transition zone (MTZ) and MON regions. Semi-quantitative AQP4 labeling was at background level in lamina, increasing in the MTZ, and highest in the MON (lamina vs MTZ, MON; p≤0.05, p≤0.01, respectively). Expression of AQP4 mRNA was minimal in lamina and substantial in MTZ and MON, while GFAP mRNA expression was uniform among the lamina, MTZ, and MON regions. Western blot assay showed AQP4 protein expression in the MON samples, but none was detected in the lamina tissue. The minimal presence of AQP4 in the lamina is a specific regional phenotype of astrocytes in the mammalian optic nerve head.
Collapse
Affiliation(s)
- Elizabeth C. Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mary E. Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Casey Keuthan
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Aru Nagalingam
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Donald J. Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Thomas V. Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Harry A. Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
26
|
Sharif NA. Degeneration of retina-brain components and connections in glaucoma: Disease causation and treatment options for eyesight preservation. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100037. [PMID: 36685768 PMCID: PMC9846481 DOI: 10.1016/j.crneur.2022.100037] [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: 03/15/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/25/2023] Open
Abstract
Eyesight is the most important of our sensory systems for optimal daily activities and overall survival. Patients who experience visual impairment due to elevated intraocular pressure (IOP) are often those afflicted with primary open-angle glaucoma (POAG) which slowly robs them of their vision unless treatment is administered soon after diagnosis. The hallmark features of POAG and other forms of glaucoma are damaged optic nerve, retinal ganglion cell (RGC) loss and atrophied RGC axons connecting to various brain regions associated with receipt of visual input from the eyes and eventual decoding and perception of images in the visual cortex. Even though increased IOP is the major risk factor for POAG, the disease is caused by many injurious chemicals and events that progress slowly within all components of the eye-brain visual axis. Lowering of IOP mitigates the damage to some extent with existing drugs, surgical and device implantation therapeutic interventions. However, since multifactorial degenerative processes occur during aging and with glaucomatous optic neuropathy, different forms of neuroprotective, nutraceutical and electroceutical regenerative and revitalizing agents and processes are being considered to combat these eye-brain disorders. These aspects form the basis of this short review article.
Collapse
Affiliation(s)
- Najam A. Sharif
- Duke-National University of Singapore Medical School, Singapore,Singapore Eye Research Institute (SERI), Singapore,Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA,Department of Pharmaceutical Sciences, Texas Southern University, Houston, TX, USA,Department of Surgery & Cancer, Imperial College of Science and Technology, St. Mary's Campus, London, UK,Department of Pharmacy Sciences, School of School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA.
| |
Collapse
|
27
|
Dias MS, Luo X, Ribas VT, Petrs-Silva H, Koch JC. The Role of Axonal Transport in Glaucoma. Int J Mol Sci 2022; 23:ijms23073935. [PMID: 35409291 PMCID: PMC8999615 DOI: 10.3390/ijms23073935] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a neurodegenerative disease that affects the retinal ganglion cells (RGCs) and leads to progressive vision loss. The first pathological signs can be seen at the optic nerve head (ONH), the structure where RGC axons leave the retina to compose the optic nerve. Besides damage of the axonal cytoskeleton, axonal transport deficits at the ONH have been described as an important feature of glaucoma. Axonal transport is essential for proper neuronal function, including transport of organelles, synaptic components, vesicles, and neurotrophic factors. Impairment of axonal transport has been related to several neurodegenerative conditions. Studies on axonal transport in glaucoma include analysis in different animal models and in humans, and indicate that its failure happens mainly in the ONH and early in disease progression, preceding axonal and somal degeneration. Thus, a better understanding of the role of axonal transport in glaucoma is not only pivotal to decipher disease mechanisms but could also enable early therapies that might prevent irreversible neuronal damage at an early time point. In this review we present the current evidence of axonal transport impairment in glaucomatous neurodegeneration and summarize the methods employed to evaluate transport in this disease.
Collapse
Affiliation(s)
- Mariana Santana Dias
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Xiaoyue Luo
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
| | - Vinicius Toledo Ribas
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Hilda Petrs-Silva
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
- Correspondence:
| |
Collapse
|
28
|
Holden JM, Al Hussein Al Awamlh S, Croteau LP, Boal AM, Rex TS, Risner ML, Calkins DJ, Wareham LK. Dysfunctional cGMP Signaling Leads to Age-Related Retinal Vascular Alterations and Astrocyte Remodeling in Mice. Int J Mol Sci 2022; 23:3066. [PMID: 35328488 PMCID: PMC8954518 DOI: 10.3390/ijms23063066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 12/17/2022] Open
Abstract
The nitric oxide-guanylyl cyclase-1-cyclic guanylate monophosphate (NO-GC-1-cGMP) pathway is integral to the control of vascular tone and morphology. Mice lacking the alpha catalytic domain of guanylate cyclase (GC1-/-) develop retinal ganglion cell (RGC) degeneration with age, with only modest fluctuations in intraocular pressure (IOP). Increasing the bioavailability of cGMP in GC1-/- mice prevents neurodegeneration independently of IOP, suggesting alternative mechanisms of retinal neurodegeneration. In continuation to these studies, we explored the hypothesis that dysfunctional cGMP signaling leads to changes in the neurovascular unit that may contribute to RGC degeneration. We assessed retinal vasculature and astrocyte morphology in young and aged GC1-/- and wild type mice. GC1-/- mice exhibit increased peripheral retinal vessel dilation and shorter retinal vessel branching with increasing age compared to Wt mice. Astrocyte cell morphology is aberrant, and glial fibrillary acidic protein (GFAP) density is increased in young and aged GC1-/- mice, with areas of dense astrocyte matting around blood vessels. Our results suggest that proper cGMP signaling is essential to retinal vessel morphology with increasing age. Vascular changed are preceded by alterations in astrocyte morphology which may together contribute to retinal neurodegeneration and loss of visual acuity observed in GC1-/- mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lauren K. Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.M.H.); (S.A.H.A.A.); (L.-P.C.); (A.M.B.); (T.S.R.); (M.L.R.); (D.J.C.)
| |
Collapse
|
29
|
Molecular regulation of neuroinflammation in glaucoma: Current knowledge and the ongoing search for new treatment targets. Prog Retin Eye Res 2022; 87:100998. [PMID: 34348167 PMCID: PMC8803988 DOI: 10.1016/j.preteyeres.2021.100998] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Neuroinflammation relying on the inflammatory responses of glial cells has emerged as an impactful component of the multifactorial etiology of neurodegeneration in glaucoma. It has become increasingly evident that despite early adaptive and reparative features of glial responses, prolonged reactivity of the resident glia, along with the peripheral immune cells, create widespread toxicity to retinal ganglion cell (RGC) axons, somas, and synapses. As much as the synchronized responses of astrocytes and microglia to glaucoma-related stress or neuron injury, their bi-directional interactions are critical to build and amplify neuroinflammation and to dictate the neurodegenerative outcome. Although distinct molecular programs regulate somatic and axonal degeneration in glaucoma, inhibition of neurodegenerative inflammation can provide a broadly beneficial treatment strategy to rescue RGC integrity and function. Since inflammatory toxicity and mitochondrial dysfunction are converging etiological paths that can boost each other and feed into a vicious cycle, anti-inflammatory treatments may also offer a multi-target potential. This review presents an overview of the current knowledge on neuroinflammation in glaucoma with particular emphasis on the cell-intrinsic and cell-extrinsic factors involved in the reciprocal regulation of glial responses, the interdependence between inflammatory and mitochondrial routes of neurodegeneration, and the research aspects inspiring for prospective immunomodulatory treatments. With the advent of powerful technologies, ongoing research on molecular and functional characteristics of glial responses is expected to accumulate more comprehensive and complementary information and to rapidly move the field forward to safe and effective modulation of the glial pro-inflammatory activities, while restoring or augmenting the glial immune-regulatory and neurosupport functions.
Collapse
|
30
|
Ikeshima-Kataoka H, Sugimoto C, Tsubokawa T. Integrin Signaling in the Central Nervous System in Animals and Human Brain Diseases. Int J Mol Sci 2022; 23:ijms23031435. [PMID: 35163359 PMCID: PMC8836133 DOI: 10.3390/ijms23031435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
The integrin family is involved in various biological functions, including cell proliferation, differentiation and migration, and also in the pathogenesis of disease. Integrins are multifunctional receptors that exist as heterodimers composed of α and β subunits and bind to various ligands, including extracellular matrix (ECM) proteins; they are found in many animals, not only vertebrates (e.g., mouse, rat, and teleost fish), but also invertebrates (e.g., planarian flatworm, fruit fly, nematodes, and cephalopods), which are used for research on genetics and social behaviors or as models for human diseases. In the present paper, we describe the results of a phylogenetic tree analysis of the integrin family among these species. We summarize integrin signaling in teleost fish, which serves as an excellent model for the study of regenerative systems and possesses the ability for replacing missing tissues, especially in the central nervous system, which has not been demonstrated in mammals. In addition, functions of astrocytes and reactive astrocytes, which contain neuroprotective subpopulations that act in concert with the ECM proteins tenascin C and osteopontin via integrin are also reviewed. Drug development research using integrin as a therapeutic target could result in breakthroughs for the treatment of neurodegenerative diseases and brain injury in mammals.
Collapse
Affiliation(s)
- Hiroko Ikeshima-Kataoka
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Correspondence:
| | - Chikatoshi Sugimoto
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
| | - Tatsuya Tsubokawa
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
| |
Collapse
|
31
|
Auler N, Tonner H, Pfeiffer N, Grus FH. Antibody and Protein Profiles in Glaucoma: Screening of Biomarkers and Identification of Signaling Pathways. BIOLOGY 2021; 10:biology10121296. [PMID: 34943212 PMCID: PMC8698915 DOI: 10.3390/biology10121296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Glaucoma is a chronic eye disease that is one of the leading causes of blindness worldwide. Currently, the only therapeutic option is to lower intraocular pressure. The onset of the disease is often delayed because patients do not notice visual impairment until very late, which is why glaucoma is also known as “the silent thief of sight”. Therefore, early detection and definition of specific markers, the so-called biomarkers, are immensely important. For the methodical implementation, high-throughput methods and omic-based methods came more and more into focus. Thus, interesting targets for possible biomarkers were already suggested by clinical research and basic research, respectively. This review article aims to join the findings of the two disciplines by collecting overlaps as well as differences in various clinical studies and to shed light on promising candidates concerning findings from basic research, facilitating conclusions on possible therapy options. Abstract Glaucoma represents a group of chronic neurodegenerative diseases, constituting the second leading cause of blindness worldwide. To date, chronically elevated intraocular pressure has been identified as the main risk factor and the only treatable symptom. However, there is increasing evidence in the recent literature that IOP-independent molecular mechanisms also play an important role in the progression of the disease. In recent years, it has become increasingly clear that glaucoma has an autoimmune component. The main focus nowadays is elucidating glaucoma pathogenesis, finding early diagnostic options and new therapeutic approaches. This review article summarizes the impact of different antibodies and proteins associated with glaucoma that can be detected for example by microarray and mass spectrometric analyzes, which (i) provide information about expression profiles and associated molecular signaling pathways, (ii) can possibly be used as a diagnostic tool in future and, (iii) can identify possible targets for therapeutic approaches.
Collapse
|
32
|
Sharif NA. Therapeutic Drugs and Devices for Tackling Ocular Hypertension and Glaucoma, and Need for Neuroprotection and Cytoprotective Therapies. Front Pharmacol 2021; 12:729249. [PMID: 34603044 PMCID: PMC8484316 DOI: 10.3389/fphar.2021.729249] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
Damage to the optic nerve and the death of associated retinal ganglion cells (RGCs) by elevated intraocular pressure (IOP), also known as glaucoma, is responsible for visual impairment and blindness in millions of people worldwide. The ocular hypertension (OHT) and the deleterious mechanical forces it exerts at the back of the eye, at the level of the optic nerve head/optic disc and lamina cribosa, is the only modifiable risk factor associated with glaucoma that can be treated. The elevated IOP occurs due to the inability of accumulated aqueous humor (AQH) to egress from the anterior chamber of the eye due to occlusion of the major outflow pathway, the trabecular meshwork (TM) and Schlemm’s canal (SC). Several different classes of pharmaceutical agents, surgical techniques and implantable devices have been developed to lower and control IOP. First-line drugs to promote AQH outflow via the uveoscleral outflow pathway include FP-receptor prostaglandin (PG) agonists (e.g., latanoprost, travoprost and tafluprost) and a novel non-PG EP2-receptor agonist (omidenepag isopropyl, Eybelis®). TM/SC outflow enhancing drugs are also effective ocular hypotensive agents (e.g., rho kinase inhibitors like ripasudil and netarsudil; and latanoprostene bunod, a conjugate of a nitric oxide donor and latanoprost). One of the most effective anterior chamber AQH microshunt devices is the Preserflo® microshunt which can lower IOP down to 10–13 mmHg. Other IOP-lowering drugs and devices on the horizon will be also discussed. Additionally, since elevated IOP is only one of many risk factors for development of glaucomatous optic neuropathy, a treatise of the role of inflammatory neurodegeneration of the optic nerve and retinal ganglion cells and appropriate neuroprotective strategies to mitigate this disease will also be reviewed and discussed.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| |
Collapse
|
33
|
Motz CT, Kabat V, Saxena T, Bellamkonda RV, Zhu C. Neuromechanobiology: An Expanding Field Driven by the Force of Greater Focus. Adv Healthc Mater 2021; 10:e2100102. [PMID: 34342167 PMCID: PMC8497434 DOI: 10.1002/adhm.202100102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The brain processes information by transmitting signals through highly connected and dynamic networks of neurons. Neurons use specific cellular structures, including axons, dendrites and synapses, and specific molecules, including cell adhesion molecules, ion channels and chemical receptors to form, maintain and communicate among cells in the networks. These cellular and molecular processes take place in environments rich of mechanical cues, thus offering ample opportunities for mechanical regulation of neural development and function. Recent studies have suggested the importance of mechanical cues and their potential regulatory roles in the development and maintenance of these neuronal structures. Also suggested are the importance of mechanical cues and their potential regulatory roles in the interaction and function of molecules mediating the interneuronal communications. In this review, the current understanding is integrated and promising future directions of neuromechanobiology are suggested at the cellular and molecular levels. Several neuronal processes where mechanics likely plays a role are examined and how forces affect ligand binding, conformational change, and signal induction of molecules key to these neuronal processes are indicated, especially at the synapse. The disease relevance of neuromechanobiology as well as therapies and engineering solutions to neurological disorders stemmed from this emergent field of study are also discussed.
Collapse
Affiliation(s)
- Cara T Motz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Victoria Kabat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, NC, 27709, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
34
|
Decorin-An Antagonist of TGF-β in Astrocytes of the Optic Nerve. Int J Mol Sci 2021; 22:ijms22147660. [PMID: 34299278 PMCID: PMC8306213 DOI: 10.3390/ijms22147660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 12/28/2022] Open
Abstract
During the pathogenesis of glaucoma, optic nerve (ON) axons become continuously damaged at the optic nerve head (ONH). This often is associated with reactive astrocytes and increased transforming growth factor (TGF-β) 2 levels. In this study we tested the hypothesis if the presence or absence of decorin (DCN), a small leucine-rich proteoglycan and a natural inhibitor of several members of the TGF family, would affect the expression of the TGF-βs and connective tissue growth factor (CTGF/CCN2) in human ONH astrocytes and murine ON astrocytes. We found that DCN is present in the mouse ON and is expressed by human ONH and murine ON astrocytes. DCN expression and synthesis was significantly reduced after 24 h treatment with 3 nM CTGF/CCN2, while treatment with 4 pM TGF-β2 only reduced expression of DCN significantly. Conversely, DCN treatment significantly reduced the expression of TGF-β1, TGF-β2 and CTGF/CCN2 vis-a-vis untreated controls. Furthermore, DCN treatment significantly reduced expression of fibronectin (FN) and collagen IV (COL IV). Notably, combined treatment with DCN and triciribine, a small molecule inhibitor of protein kinase B (AKT), attenuated effects of DCN on CTGF/CCN2, TGF-β1, and TGF-β2 mRNA expression. We conclude (1) that DCN is an important regulator of TGF-β and CTGF/CCN2 expression in astrocytes of the ON and ONH, (2) that DCN thereby regulates the expression of extracellular matrix (ECM) components and (3) that DCN executes its negative regulatory effects on TGF-β and CTGF/CCN2 via the pAKT/AKT signaling pathway in ON astrocytes.
Collapse
|
35
|
Multifactorial Pathogenic Processes of Retinal Ganglion Cell Degeneration in Glaucoma towards Multi-Target Strategies for Broader Treatment Effects. Cells 2021; 10:cells10061372. [PMID: 34199494 PMCID: PMC8228726 DOI: 10.3390/cells10061372] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/14/2021] [Accepted: 05/29/2021] [Indexed: 02/06/2023] Open
Abstract
Glaucoma is a chronic neurodegenerative disease characterized by apoptosis of retinal ganglion cell (RGC) somas, degeneration of axons, and loss of synapses at dendrites and axon terminals. Glaucomatous neurodegeneration encompasses multiple triggers, multiple cell types, and multiple molecular pathways through the etiological paths with biomechanical, vascular, metabolic, oxidative, and inflammatory components. As much as intrinsic responses of RGCs themselves, divergent responses and intricate interactions of the surrounding glia also play decisive roles for the cell fate. Seen from a broad perspective, multitarget treatment strategies have a compelling pathophysiological basis to more efficiently manipulate multiple pathogenic processes at multiple injury sites in such a multifactorial neurodegenerative disease. Despite distinct molecular programs for somatic and axonal degeneration, mitochondrial dysfunction and glia-driven neuroinflammation present interdependent processes with widespread impacts in the glaucomatous retina and optic nerve. Since dysfunctional mitochondria stimulate inflammatory responses and proinflammatory mediators impair mitochondria, mitochondrial restoration may be immunomodulatory, while anti-inflammatory treatments protect mitochondria. Manipulation of these converging routes may thus allow a unified treatment strategy to protect RGC axons, somas, and synapses. This review presents an overview of recent research advancements with emphasis on potential treatment targets to achieve the best treatment efficacy to preserve visual function in glaucoma.
Collapse
|
36
|
Lowry EA, Mansberger SL, Gardiner SK, Yang H, Sanchez F, Reynaud J, Demirel S, Burgoyne CF, Fortune B. Association of Optic Nerve Head Prelaminar Schisis With Glaucoma. Am J Ophthalmol 2021; 223:246-258. [PMID: 33166501 DOI: 10.1016/j.ajo.2020.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE To compare the frequency of observing optic nerve head (ONH) prelaminar schisis by optical coherence tomography (OCT) in glaucoma and glaucoma suspect (GL/S) eyes vs healthy control (HC) eyes and to assess its association with other markers of glaucoma severity. METHODS This cross-sectional study included 298 eyes of 150 GL/S patients and 88 eyes of 44 HCs. OCT scans were obtained, including 24 radial B-scans, each composed of 768 A-lines spanning 15°, centered on the ONH. Two reviewers masked to all other clinical, demographic, and ocular information independently graded the OCT scans for the presence of ONH prelaminar schisis on a 4-point scale of 0 (none) to 3 (severe). The probability of ONH schisis was compared between groups and against demographic and ocular factors, including structural and functional measures of glaucoma severity. RESULTS The frequency and severity of ONH prelaminar schisis were greater in GL/S than in HC (P = .009). Among the GL/S group, 165 eyes (55.4%) had no visible schisis (Grade 0), 71 (23.8%) had Grade 1, 46 (15.4%) had Grade 2 and 16 (5.4%) had Grade 3 schisis. Among HC eyes, 59 (67.0%) had Grade 0, 24 (27.3%) had Grade 1, 5 (5.7%) had Grade 2, none had Grade 3. ONH schisis was more common in eyes with thinner MRW and a deeper cup. CONCLUSIONS ONH prelaminar schisis may be a sign of glaucomatous deformation and reflect ongoing pathophysiological damage. ONH prelaminar schisis can impact OCT image segmentation and diagnostic parameters, resulting in substantial overestimation of the true rim tissue thickness and underestimation of cup depth.
Collapse
|
37
|
Kimball E, Schaub J, Quillen S, Keuthan C, Pease ME, Korneva A, Quigley H. The role of aquaporin-4 in optic nerve head astrocytes in experimental glaucoma. PLoS One 2021; 16:e0244123. [PMID: 33529207 PMCID: PMC7853498 DOI: 10.1371/journal.pone.0244123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023] Open
Abstract
PURPOSE To study aquaporin channel expression in astrocytes of the mouse optic nerve (ON) and the response to IOP elevation in mice lacking aquaporin 4 (AQP4 null). METHODS C57BL/6 (B6) and AQP4 null mice were exposed to bead-induced IOP elevation for 3 days (3D-IOP), 1 and 6 weeks. Mouse ocular tissue sections were immunolabeled against aquaporins 1(AQP1), 4(AQP4), and 9(AQP9). Ocular tissue was imaged to identify normal AQP distribution, ON changes, and axon loss after IOP elevation. Ultrastructure examination, cell proliferation, gene expression, and transport block were also analyzed. RESULTS B6 mice had abundant AQP4 expression in Müller cells, astrocytes of retina and myelinated ON (MON), but minimal AQP4in prelaminar and unmyelinated ON (UON). MON of AQP4 nulls had smaller ON area, smaller axon diameter, higher axon density, and larger proportionate axon area than B6 (all p≤0.05). Bead-injection led to comparable 3D-IOP elevation (p = 0.42) and axonal transport blockade in both strains. In B6, AQP4 distribution was unchanged after 3D-IOP. At baseline, AQP1 and AQP9 were present in retina, but not in UON and this was unaffected after IOP elevation in both strains. In 3D-IOP mice, ON astrocytes and microglia proliferated, more in B6 than AQP4 null. After 6 week IOP elevation, axon loss occurred equally in the two mouse types (24.6%, AQP4 null vs. 23.3%, B6). CONCLUSION Lack of AQP4 was neither protective nor detrimental to the effects of IOP elevation. The minimal presence of AQP4 in UON may be a vital aspect of the regionally specific phenotype of astrocytes in the mouse optic nerve head.
Collapse
Affiliation(s)
- Elizabeth Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Julie Schaub
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Casey Keuthan
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mary Ellen Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Arina Korneva
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Harry Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
38
|
Korneva A, Kimball EC, Jefferys JL, Quigley HA, Nguyen TD. Biomechanics of the optic nerve head and peripapillary sclera in a mouse model of glaucoma. J R Soc Interface 2020; 17:20200708. [PMID: 33323053 PMCID: PMC7811579 DOI: 10.1098/rsif.2020.0708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/17/2020] [Indexed: 01/09/2023] Open
Abstract
The deformation of the mouse astrocytic lamina (AL) and adjacent peripapillary sclera (PPS) was measured in response to elevated intraocular pressure. We subjected explanted mouse eyes to inflation testing, comparing control eyes to those 3 days and 6 weeks after induction of ocular hypertension (OHT) via ocular microbead injection. Laser scanning microscopy was used with second harmonic generation to image the collagenous PPS and two-photon fluorescence to image transgenic fluorescent astrocytes in the AL. Digital volume correlation was applied to calculate strains in the PPS and AL. The specimen-averaged strains were biaxial in the AL and PPS, with greater strain overall in the x- than y-direction in the AL and greater strain in the θ- than the r-direction in the PPS. Strains increased after 3-day OHT, with greater strain overall in the 3-day AL than control AL, and greater circumferential strain in the 3-day PPS than control PPS. In the 6-week OHT eyes, AL and PPS strains were similar overall to controls. This experimental glaucoma model demonstrated a dynamic change in the mechanical behaviour of the AL and PPS over time at the site of neuronal injury and remodelling in glaucoma.
Collapse
Affiliation(s)
- Arina Korneva
- Glaucoma Center of Excellence, Wilmer Eye Institute, John Hopkins University, Baltimore, MD, USA
| | - Elizabeth C. Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, John Hopkins University, Baltimore, MD, USA
| | - Joan L. Jefferys
- Glaucoma Center of Excellence, Wilmer Eye Institute, John Hopkins University, Baltimore, MD, USA
| | - Harry A. Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, John Hopkins University, Baltimore, MD, USA
| | - Thao D. Nguyen
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Ophthalmology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science, The Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
39
|
Ling YTT, Pease ME, Jefferys JL, Kimball EC, Quigley HA, Nguyen TD. Pressure-Induced Changes in Astrocyte GFAP, Actin, and Nuclear Morphology in Mouse Optic Nerve. Invest Ophthalmol Vis Sci 2020; 61:14. [PMID: 32910133 PMCID: PMC7488631 DOI: 10.1167/iovs.61.11.14] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/12/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose To conduct quantitative analysis of astrocytic glial fibrillary acidic protein (GFAP), actin and nuclei distribution in mouse optic nerve (ON) and investigate changes in the measured features after 3 days of ocular hypertension (OHT). Method Serial cross-sections of 3-day microbead-induced OHT and control ONs were fluorescently labelled and imaged using confocal microscope. Eighteen structural features were measured from the acquired images, including GFAP coverage, actin area fraction, process thickness, and aspect ratio of cell nucleus. The measured features were analyzed for variations with axial locations along ON and radial zones transverse to ON, as well as for the correlations with degree of intraocular pressure (IOP) change. Results The most significant changes in structural features after 3-day OHT occurred in the unmyelinated ON region (R1), and the changes were greater with greater IOP elevation. Although the GFAP, actin, axonal, and ON areas all increased in 3-day OHT ONs in R1 (P ≤ 0.004 for all), the area fraction of GFAP actually decreased (P = 0.02), the actin area fraction was stable and individual axon compartments were unchanged in size. Within R1, the number of nuclear clusters increased (P < 0.001), but the mean size of nuclear clusters was smaller (P = 0.02) and the clusters became rounder (P < 0.001). In all cross-sections of control ONs, astrocytic processes were thickest in the rim zone compared with the central and peripheral zones (P ≤ 0.002 for both), whereas the overall process width in R1 decreased after 3 days of OHT (P < 0.001). Conclusions The changes in structure elucidated IOP-generated alterations that underlie astrocyte mechanotranslational responses relevant to glaucoma.
Collapse
Affiliation(s)
- Yik Tung Tracy Ling
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States
| | - Mary E. Pease
- Wilmer Ophthalmological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Joan L. Jefferys
- Wilmer Ophthalmological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Elizabeth C. Kimball
- Wilmer Ophthalmological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Harry A. Quigley
- Wilmer Ophthalmological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Thao D. Nguyen
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|