1
|
Holden JM, Calkins DJ. Bilateral astrocyte reaction to unilateral insult in the optic projection to the brain. Proc Natl Acad Sci U S A 2025; 122:e2502602122. [PMID: 40163738 PMCID: PMC12002340 DOI: 10.1073/pnas.2502602122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Affiliation(s)
- Joseph M. Holden
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN37212
- Vanderbilt Vision Research Center, Vanderbilt University Medical Center, Nashville, TN37212
| | - David J. Calkins
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN37212
- Vanderbilt Vision Research Center, Vanderbilt University Medical Center, Nashville, TN37212
| |
Collapse
|
2
|
Zhang Y, Yang X, Deng X, Yang S, Li Q, Xie Z, Hong L, Cao M, Yi G, Fu M. Single-cell transcriptomics-based multidisease analysis revealing the molecular dynamics of retinal neurovascular units under inflammatory and hypoxic conditions. Exp Neurol 2023; 362:114345. [PMID: 36736650 DOI: 10.1016/j.expneurol.2023.114345] [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: 10/10/2022] [Revised: 12/27/2022] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
The retinal neurovascular unit (NVU) is paramount to maintaining the homeostasis of the retina and determines the progression of various diseases, including diabetic retinopathy (DR), glaucoma, and retinopathy of prematurity (ROP). Although some studies have investigated these diseases, a combined analysis of disease-wide etiology in the NUV at the single-cell level is lacking. Herein, we constructed an atlas of the NVU under inflammatory and hypoxic conditions by integrating single-cell transcriptome data from retinas from wild-type, AireKO, and NdpKO mice. Based on the heterogeneity of the NVU structure and transcriptome diversity under normal and pathological conditions, we discovered two subpopulations of Müller cells: Aqp4hi and Aqp4lo cells. Specifically, Aqp4lo cells expresses phototransduction genes and represent a special type of Müller cell distinct from Aqp4hi cells, classical Müller cells. AireKO mice exhibit experimental autoimmune uveitis (EAU) with severe damage to the NVU structure, mainly degeneration of Aqp4hi cells. NdpKO mice exhibited familial exudative vitreoretinopathy (FEVR), with damage to the endothelial barrier, endothelial cell tight junction destruction and basement membrane thickening, accompanied by the reactive secretion of proangiogenic factors by Aqp4hi cells. In both EAU and FEVR, Aqp4hi cells are a key factor leading to NVU damage, and the mechanism by which they are generated is regulated by different transcription factors. By studying the pattern of immune cell infiltration in AireKO mice, we constructed a regulatory loop of "inflammatory cells/NVU - monocytes - APCs - Ifng+ T cells", providing a new target for blocking the inflammatory cascade. Our elucidation of the cell-specific molecular changes, cell-cell interactions and transcriptional mechanisms of the retinal NVU provides new insights to support the development of multipurpose drugs to block or even reverse NVU damage.
Collapse
Affiliation(s)
- Yuxi Zhang
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Xiongyi Yang
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Xiaoqing Deng
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Siyu Yang
- Department of Ophthalmology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Qiumo Li
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhuohang Xie
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Libing Hong
- Zhujiang Hospital, Southern Medical University, Guangzhou, PR China; The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Mingzhe Cao
- Department of Ophthalmology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China.
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-Sen University, No. 26, Erheng Road, Yuancun, Tianhe, Guangzhou, Guangdong, PR China.
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
3
|
Zhou L, Lin D, Xu G, Wang X, Chen Z, Wang D, Fan H. Alteration of neurofilament heavy chain and its phosphoforms reveals early subcellular damage beyond the optic nerve head in glaucoma. Front Neurol 2023; 14:1091697. [PMID: 37034083 PMCID: PMC10073422 DOI: 10.3389/fneur.2023.1091697] [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: 11/07/2022] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Abstract
Background Retinal ganglion cells (RGCs) axon loss at the site of optic nerve head (ONH) is long believed as the common pathology in glaucoma since different types of glaucoma possessing different characteristic of intraocular pressure, and this damage was only detected at the later stage. Methods To address these disputes and detect early initiating events underlying RGCs, we firstly detected somatic or axonal change and compared their difference in acute and chronic phase of primary angle-closed glaucoma (PACG) patient using optical coherence tomography (OCT), then an axonal-enriched cytoskeletal protein neurofilament heavy chain and its phosphoforms (NF-H, pNF-H) were utilized to reveal spatio-temporal undetectable damage insulted by acute and chronic ocular hypertension (AOH, COH) in two well characterized glaucoma mice models. Results In clinic, we detected nonhomogeneous changes such as ONH and soma of RGCs presenting edema in acute phase but atrophy in chronic one by OCT. In AOH animal models, an increase expression of NF-H especially its phosphorylation modification was observed as early as 4 h before RGCs loss, which presented as somatic accumulation in the peripheral retina and at the sites of ONH. In contrast, in microbeads induced COH model, NF-H and pNF-H reduced significantly, these changes firstly occurred as NF-H or pNF-H disconnection at ONH and optic nerve after 2 weeks when the intraocular pressure reaching the peak; Meanwhile, we detected aqueous humor pNF-H elevation after AOH and slight reduction in the COH. Conclusion Together, our data supports that early alteration of NF-H and its phosphoforms would reveal undetectable subcellular damage consisting of peripheral somatic neurofilament compaction, impaired axonal transport and distal axonal disorganization of cytoskeleton beyond the ONH, and identifies two distinct axonal degeneration which were Wallerian combination with retrograde degeneration in acute PACG and retrograde degeneration in the chronic one.
Collapse
Affiliation(s)
- Lan Zhou
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongyue Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guihua Xu
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Xiaoyi Wang
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Zilin Chen
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Dingding Wang
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Huiya Fan
- Ophthalmological Center of Huizhou Central People's Hospital, Huizhou, Guangdong, China
- *Correspondence: Huiya Fan
| |
Collapse
|
4
|
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: 14] [Impact Index Per Article: 4.7] [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
|
5
|
Kodati B, McGrady NR, Jefferies HB, Stankowska DL, Krishnamoorthy RR. Oral administration of a dual ET A/ET B receptor antagonist promotes neuroprotection in a rodent model of glaucoma. Mol Vis 2022; 28:165-177. [PMID: 36274816 PMCID: PMC9491150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Glaucoma is a neurodegenerative disease associated with elevated intraocular pressure and characterized by optic nerve axonal degeneration, cupping of the optic disc, and loss of retinal ganglion cells (RGCs). The endothelin (ET) system of vasoactive peptides (ET-1, ET-2, ET-3) and their G-protein coupled receptors (ETA and ETB receptors) have been shown to contribute to the pathophysiology of glaucoma. The purpose of this study was to determine whether administration of the endothelin receptor antagonist macitentan was neuroprotective to RGCs and optic nerve axons when administered after the onset of intraocular pressure (IOP) elevation in ocular hypertensive rats. METHODS Male and female Brown Norway rats were subjected to the Morrison model of ocular hypertension by injection of hypertonic saline through the episcleral veins. Following IOP elevation, macitentan (5 mg/kg body wt) was administered orally 3 days per week, and rats with IOP elevation were maintained for 4 weeks. RGC function was determined by pattern electroretinography (PERG) at 2 and 4 weeks post-IOP elevation. Rats were euthanized by approved humane methods, and retinal flat mounts were generated and immunostained for the RGC-selective marker Brn3a. PPD-stained optic nerve sections were imaged by confocal microscopy. RGC and axon counts were conducted in a masked manner and compared between the treatment groups. RESULTS Significant protection against loss of RGCs and optic nerve axons was found following oral administration of macitentan in rats with elevated IOP. In addition, a protective trend for RGC function, as measured by pattern ERG analysis, was evident following macitentan treatment. CONCLUSIONS Macitentan treatment had a neuroprotective effect on RGCs and their axons, independent of its IOP-lowering effect, suggesting that macitentan may complement existing treatments to prevent neurodegeneration during ocular hypertension. The findings presented have implications for the use of macitentan as an oral formulation to promote neuroprotection in glaucoma patients.
Collapse
Affiliation(s)
- Bindu Kodati
- University of North Texas Health Science Center, Pharmacology and Neuroscience, North Texas Eye Research Institute, Fort Worth, TX
| | | | - Hayden B. Jefferies
- University of Texas Health Science Center, McGovern Medical School, Houston, TX
| | - Dorota L. Stankowska
- University of North Texas Health Science Center, Pharmacology and Neuroscience, North Texas Eye Research Institute, Fort Worth, TX
| | - Raghu R. Krishnamoorthy
- University of North Texas Health Science Center, Pharmacology and Neuroscience, North Texas Eye Research Institute, Fort Worth, TX
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Calkins DJ. Adaptive responses to neurodegenerative stress in glaucoma. Prog Retin Eye Res 2021; 84:100953. [PMID: 33640464 DOI: 10.1016/j.preteyeres.2021.100953] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/08/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
Glaucoma causes loss of vision through degeneration of the retinal ganglion cell (RGC) projection to the brain. The disease is characterized by sensitivity to intraocular pressure (IOP) conveyed at the optic nerve head, through which RGC axons pass unmyelinated to form the optic nerve. From this point, a pathogenic triumvirate comprising inflammatory, oxidative, and metabolic stress influence both proximal structures in the retina and distal structures in the optic projection. This review focuses on metabolic stress and how the optic projection may compensate through novel adaptive mechanisms to protect excitatory signaling to the brain. In the retina and proximal nerve head, the unmyelinated RGC axon segment is energy-inefficient, which leads to increased demand for adenosine-5'-triphosphate (ATP) at the risk of vulnerability to Ca2+-related metabolic and oxidative pressure. This vulnerability may underlie the bidirectional nature of progression. However, recent evidence highlights that the optic projection in glaucoma is not passive but rather demonstrates adaptive processes that may push back against neurodegeneration. In the retina, even as synaptic and dendritic pruning ensues, early progression involves enhanced excitability of RGCs. Enhancement involves depolarization of the resting membrane potential and increased response to light, independent of RGC morphological type. This response is axogenic, arising from increased levels and translocation of voltage-gated sodium channels (NaV) in the unmyelinated segment. During this same early period, large-scale networks of gap-junction coupled astrocytes redistribute metabolic resources to the optic projection stressed by elevated IOP to slow loss of axon function. This redistribution may reflect more local remodeling, as astrocyte processes respond to focal metabolic duress by boosting glycogen turnover in response to axonal activity in an effort to promote survival of the healthiest axons. Both enhanced excitability and metabolic redistribution are transient, indicating that the same adaptive mechanisms that apparently serve to slow progression ultimately may be too expensive for the system to sustain over longer periods.
Collapse
Affiliation(s)
- David J Calkins
- The Vanderbilt Eye Institute, Nashville, TN, USA; Vanderbilt Vision Research Center, Vanderbilt University Medical Center, 1161 21st Ave S, AA7100 Medical Center North Nashville, Tennessee, 37232, USA.
| |
Collapse
|
9
|
Van Hook MJ, Monaco C, Bierlein ER, Smith JC. Neuronal and Synaptic Plasticity in the Visual Thalamus in Mouse Models of Glaucoma. Front Cell Neurosci 2021; 14:626056. [PMID: 33584206 PMCID: PMC7873902 DOI: 10.3389/fncel.2020.626056] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/21/2020] [Indexed: 11/20/2022] Open
Abstract
Homeostatic plasticity plays important role in regulating synaptic and intrinsic neuronal function to stabilize output following perturbations to circuit activity. In glaucoma, a neurodegenerative disease of the visual system commonly associated with elevated intraocular pressure (IOP), the early disease is associated with altered synaptic inputs to retinal ganglion cells (RGCs), changes in RGC intrinsic excitability, and deficits in optic nerve transport and energy metabolism. These early functional changes can precede RGC degeneration and are likely to alter RGC outputs to their target structures in the brain and thereby trigger homeostatic changes in synaptic and neuronal properties in those brain regions. In this study, we sought to determine whether and how neuronal and synaptic function is altered in the dorsal lateral geniculate nucleus (dLGN), an important RGC projection target in the thalamus, and how functional changes related to IOP. We accomplished this using patch-clamp recordings from thalamocortical (TC) relay neurons in the dLGN in two established mouse models of glaucoma—the DBA/2J (D2) genetic mouse model and an inducible glaucoma model with intracameral microbead injections to elevate IOP. We found that the intrinsic excitability of TC neurons was enhanced in D2 mice and these functional changes were mirrored in recordings of TC neurons from microbead-injected mice. Notably, many neuronal properties were correlated with IOP in older D2 mice, when IOP rises. The frequency of miniature excitatory synaptic currents (mEPSCs) was reduced in 9-month-old D2 mice, and vGlut2 staining of RGC synaptic terminals was reduced in an IOP-dependent manner. These data suggest that glaucoma-associated changes to neuronal excitability and synaptic inputs in the dLGN might represent a combination of both stabilizing/homeostatic plasticity and pathological dysfunction.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Corrine Monaco
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Elizabeth R Bierlein
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jennie C Smith
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Simons ES, Smith MA, Dengler-Crish CM, Crish SD. Retinal ganglion cell loss and gliosis in the retinofugal projection following intravitreal exposure to amyloid-beta. Neurobiol Dis 2021; 147:105146. [PMID: 33122075 DOI: 10.1016/j.nbd.2020.105146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Pathological accumulations of amyloid-beta (Aβ) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aβ1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aβ exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aβ in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aβ eye injections driving increased Aβ levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aβ-mediated microglial activation has been shown to propagate Aβ pathology, we also investigated the role of the Aβ-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aβ-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aβ pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aβ-mediated inflammatory damage.
Collapse
Affiliation(s)
- E S Simons
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - M A Smith
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States; Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH 44302, United States
| | - C M Dengler-Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - S D Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States.
| |
Collapse
|
11
|
Ahmed OM, Waisbourd M, Spaeth GL, Katz LJ. Improvement in structure and visual function in patients with glaucoma: the possible key to better treatment? Surv Ophthalmol 2020; 66:644-652. [PMID: 33316283 DOI: 10.1016/j.survophthal.2020.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 11/26/2022]
Abstract
Glaucoma is characterized by retinal ganglion cell loss that can lead to permanent visual loss. Current clinical management practice assumes that glaucomatous visual loss is irreversible; however, there is increasing evidence that permanent vision loss and cell death are preceded by reversible functional and structural changes. We propose that these changes should be considered by glaucoma specialists when treating their patients. We discuss the neurobiological basis of this phenomenon and provide clinical evidence of reversibility in both structure and function. Specifically, we review the findings of visual field testing, contrast sensitivity, electroretinography, and imaging of the optic nerve and their correlation with functional changes. We then discuss the clinical value of these observations in helping guide approaches toward the diagnosis and treatment of patients with glaucoma.
Collapse
Affiliation(s)
- Osama M Ahmed
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Yale University School of Medicine, New Haven, CT, USA
| | - Michael Waisbourd
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Department of Ophthalmology, Tel Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - George L Spaeth
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA
| | - L Jay Katz
- Glaucoma Research Center, Wills Eye Hospital, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Baudouin C, Kolko M, Melik-Parsadaniantz S, Messmer EM. Inflammation in Glaucoma: From the back to the front of the eye, and beyond. Prog Retin Eye Res 2020; 83:100916. [PMID: 33075485 DOI: 10.1016/j.preteyeres.2020.100916] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 02/08/2023]
Abstract
The pathophysiology of glaucoma is complex, multifactorial and not completely understood. Elevated intraocular pressure (IOP) and/or impaired retinal blood flow may cause initial optic nerve damage. In addition, age-related oxidative stress in the retina concurrently with chronic mechanical and vascular stress is crucial for the initiation of retinal neurodegeneration. Oxidative stress is closely related to cell senescence, mitochondrial dysfunction, excitotoxicity, and neuroinflammation, which are involved in glaucoma progression. Accumulating evidence from animal glaucoma models and from human ocular samples suggests a dysfunction of the para-inflammation in the retinal ganglion cell layer and the optic nerve head. Moreover, quite similar mechanisms in the anterior chamber could explain the trabecular meshwork dysfunction and the elevated IOP in primary open-angle glaucoma. On the other hand, ocular surface disease due to topical interventions is the most prominent and visible consequence of inflammation in glaucoma, with a negative impact on filtering surgery failure, topical treatment efficacy, and possibly on inflammation in the anterior segment. Consequently, glaucoma appears as an outstanding eye disease where inflammatory changes may be present to various extents and consequences along the eye structure, from the ocular surface to the posterior segment, and the visual pathway. Here we reviewed the inflammatory processes in all ocular structures in glaucoma from the back to the front of the eye and beyond. Our approach was to explain how para-inflammation is necessary to maintain homoeostasis, and to describe abnormal inflammatory findings observed in glaucomatous patients or in animal glaucoma models, supporting the hypothesis of a dysregulation of the inflammatory balance toward a pro-inflammatory phenotype. Possible anti-inflammatory therapeutic approaches in glaucoma are also discussed.
Collapse
Affiliation(s)
- Christophe Baudouin
- Quinze-Vingts National Ophthalmology Hospital, INSERM-DGOS CIC 1423, IHU Foresight, Paris, France; Sorbonne Université, INSERM, CNRS, Institut de La Vision, Paris, France; Department of Ophthalmology, Ambroise Paré Hospital, APHP, Université de Versailles Saint-Quentin en Yvelines, Boulogne-Billancourt, France.
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| | | | | |
Collapse
|
13
|
Lambert WS, Pasini S, Collyer JW, Formichella CR, Ghose P, Carlson BJ, Calkins DJ. Of Mice and Monkeys: Neuroprotective Efficacy of the p38 Inhibitor BIRB 796 Depends on Model Duration in Experimental Glaucoma. Sci Rep 2020; 10:8535. [PMID: 32444682 PMCID: PMC7244559 DOI: 10.1038/s41598-020-65374-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/04/2020] [Indexed: 01/23/2023] Open
Abstract
Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). Early progression involves retinal ganglion cell (RGC) axon dysfunction that precedes frank degeneration. Previously we demonstrated that p38 MAPK inhibition abates axonal dysfunction and slows degeneration in the inducible microbead occlusion model of glaucoma in rat. Here, we assessed the neuroprotective effect of topical eye delivery of the p38 MAPK inhibitor BIRB 796 in three models of glaucoma (microbead occlusion in rat and squirrel monkey and the genetic DBA/2 J mouse model) with distinct durations of IOP elevation. While BIRB 796 did not influence IOP, treatment over four weeks in rats prevented degradation of anterograde axonal transport to the superior colliculus and degeneration in the optic nerve. Treatment over months in the chronic DBA/2 J model and in the squirrel monkey model reduced expression and activation of p38 downstream targets in the retina and brain but did not rescue RGC axon transport or degeneration, suggesting the efficacy of BIRB 796 in preventing associated degeneration of the RGC projection depends on the duration of the experimental model. These results emphasize the importance of evaluating potential therapeutic compounds for neuroprotection in multiple models using elongated treatment paradigms for an accurate assessment of efficacy.
Collapse
Affiliation(s)
- Wendi S Lambert
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - Silvia Pasini
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - John W Collyer
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - Cathryn R Formichella
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - Purnima Ghose
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - Brian J Carlson
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA
| | - David J Calkins
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, 37232-2337, USA.
| |
Collapse
|
14
|
Du R, Wang X, He S. BDNF improves axon transportation and rescues visual function in a rodent model of acute elevation of intraocular pressure. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1337-1346. [PMID: 32201927 DOI: 10.1007/s11427-019-1567-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
Optic neuropathies lead to blindness; the common pathology is the degeneration of axons of the retinal ganglion cells. In this study, we used a rat model of retinal ischemia-reperfusion and a one-time intravitreal brain-derived neurotrophic factor (BDNF) injection; then we examined axon transportation function, continuity, physical presence of axons in different part of the optic nerve, and the expression level of proteins involved in axon transportation. We found that in the disease model, axon transportation was the most severely affected, followed by axon continuity, then the number of axons in the distal and proximal optic nerve. BDNF treatment relieved all reductions and significantly restored function. The molecular changes were more minor, probably due to massive gliosis of the optic nerve, so interpretation of protein expression data should be done with some caution. The process in this acute model resembles a fast-forward of changes in the chronic model of glaucoma. Therefore, impairment in axon transportation appears to be a common early process underlying different optic neuropathies. This research on effective intervention can be used to develop interventions for all optic neuropathies targeting axon transportation.
Collapse
Affiliation(s)
- Rui Du
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shigang He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Bio-X Institute, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
15
|
Fischer RA, Risner ML, Roux AL, Wareham LK, Sappington RM. Impairment of Membrane Repolarization Accompanies Axon Transport Deficits in Glaucoma. Front Neurosci 2019; 13:1139. [PMID: 31736686 PMCID: PMC6838637 DOI: 10.3389/fnins.2019.01139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Glaucoma is a leading cause of blindness worldwide, resulting from degeneration of retinal ganglion cells (RGCs), which form the optic nerve. In glaucoma, axon transport deficits appear to precede structural degeneration of RGC axons. The period of time between the onset of axon transport deficits and the structural degeneration of RGC axons may represent a therapeutic window for the prevention of irreversible vision loss. However, it is unclear how deficits in axon transport relate to the electrophysiological capacity of RGCs to produce and maintain firing frequencies that encode visual stimuli. Here, we examined the electrophysiological signature of individual RGCs in glaucomatous retina with respect to axon transport facility. Utilizing the Microbead Occlusion Model of murine ocular hypertension, we performed electrophysiological recordings of RGCs with and without deficits in anterograde axon transport. We found that RGCs with deficits in axon transport have a reduced ability to maintain spiking frequency that arises from elongation of the repolarization phase of the action potential. This repolarization phenotype arises from reduced cation flux and K+ dyshomeostasis that accompanies pressure-induced decreases in Na/K-ATPase expression and activity. In vitro studies with purified RGCs indicate that elevated pressure induces early internalization of Na/K-ATPase that, when reversed, stabilizes cation flux and prevents K+ dyshomeostasis. Furthermore, pharmacological inhibition of the Na/K-ATPase is sufficient to replicate pressure-induced cation influx and repolarization phase phenotypes in healthy RGCs. These studies suggest that deficits in axon transport also likely reflect impaired electrophysiological function of RGCs. Our findings further identify a failure to maintain electrochemical gradients and cation dyshomeostasis as an early phenotype of glaucomatous pathology in RGCs that may have significant bearing on efforts to restore RGC health in diseased retina.
Collapse
Affiliation(s)
- Rachel A Fischer
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Michael L Risner
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Abigail L Roux
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lauren K Wareham
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rebecca M Sappington
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
16
|
Bhandari A, Smith JC, Zhang Y, Jensen AA, Reid L, Goeser T, Fan S, Ghate D, Van Hook MJ. Early-Stage Ocular Hypertension Alters Retinal Ganglion Cell Synaptic Transmission in the Visual Thalamus. Front Cell Neurosci 2019; 13:426. [PMID: 31607867 PMCID: PMC6761307 DOI: 10.3389/fncel.2019.00426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
Axonopathy is a hallmark of many neurodegenerative diseases including glaucoma, where elevated intraocular pressure (ocular hypertension, OHT) stresses retinal ganglion cell (RGC) axons as they exit the eye and form the optic nerve. OHT causes early changes in the optic nerve such as axon atrophy, transport inhibition, and gliosis. Importantly, many of these changes appear to occur prior to irreversible neuronal loss, making them promising points for early diagnosis of glaucoma. It is unknown whether OHT has similarly early effects on the function of RGC output to the brain. To test this possibility, we elevated eye pressure in mice by anterior chamber injection of polystyrene microbeads. Five weeks post-injection, bead-injected eyes showed a modest RGC loss in the peripheral retina, as evidenced by RBPMS antibody staining. Additionally, we observed reduced dendritic complexity and lower spontaneous spike rate of On-αRGCs, targeted for patch clamp recording and dye filling using a Opn4-Cre reporter mouse line. To determine the influence of OHT on retinal projections to the brain, we expressed Channelrhodopsin-2 (ChR2) in melanopsin-expressing RGCs by crossing the Opn4-Cre mouse line with a ChR2-reporter mouse line and recorded post-synaptic responses in thalamocortical relay neurons in the dorsal lateral geniculate nucleus (dLGN) of the thalamus evoked by stimulation with 460 nm light. The use of a Opn4-Cre reporter system allowed for expression of ChR2 in a narrow subset of RGCs responsible for image-forming vision in mice. Five weeks following OHT induction, paired pulse and high-frequency stimulus train experiments revealed that presynaptic vesicle release probability at retinogeniculate synapses was elevated. Additionally, miniature synaptic current frequency was slightly reduced in brain slices from OHT mice and proximal dendrites of post-synaptic dLGN relay neurons, assessed using a Sholl analysis, showed a reduced complexity. Strikingly, these changes occurred prior to major loss of RGCs labeled with the Opn4-Cre mouse, as indicated by immunofluorescence staining of ChR2-expressing retinal neurons. Thus, OHT leads to pre- and post-synaptic functional and structural changes at retinogeniculate synapses. Along with RGC dendritic remodeling and optic nerve transport changes, these retinogeniculate synaptic changes are among the earliest signs of glaucoma.
Collapse
Affiliation(s)
- Ashish Bhandari
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jennie C Smith
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yang Zhang
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States.,Creighton University School of Medicine, Omaha, NE, United States.,Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Aaron A Jensen
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lisa Reid
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Toni Goeser
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shan Fan
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Deepta Ghate
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| | - Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
17
|
Claes M, De Groef L, Moons L. Target-Derived Neurotrophic Factor Deprivation Puts Retinal Ganglion Cells on Death Row: Cold Hard Evidence and Caveats. Int J Mol Sci 2019; 20:E4314. [PMID: 31484425 PMCID: PMC6747494 DOI: 10.3390/ijms20174314] [Citation(s) in RCA: 14] [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: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma and other optic neuropathies are characterized by axonal transport deficits. Axonal cargo travels back and forth between the soma and the axon terminus, a mechanism ensuring homeostasis and the viability of a neuron. An example of vital molecules in the axonal cargo are neurotrophic factors (NTFs). Hindered retrograde transport can cause a scarcity of those factors in the retina, which in turn can tilt the fate of retinal ganglion cells (RGCs) towards apoptosis. This postulation is one of the most widely recognized theories to explain RGC death in the disease progression of glaucoma and is known as the NTF deprivation theory. For several decades, research has been focused on the use of NTFs as a novel neuroprotective glaucoma treatment. Until now, results in animal models have been promising, but translation to the clinic has been highly disappointing. Are we lacking important knowledge to lever NTF therapies towards the therapeutic armamentarium? Or did we get the wrong end of the stick regarding the NTF deprivation theory? In this review, we will tackle the existing evidence and caveats advocating for and against the target-derived NTF deprivation theory in glaucoma, whilst digging into associated therapy efforts.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
18
|
Faiq MA, Wollstein G, Schuman JS, Chan KC. Cholinergic nervous system and glaucoma: From basic science to clinical applications. Prog Retin Eye Res 2019; 72:100767. [PMID: 31242454 PMCID: PMC6739176 DOI: 10.1016/j.preteyeres.2019.06.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 02/08/2023]
Abstract
The cholinergic system has a crucial role to play in visual function. Although cholinergic drugs have been a focus of attention as glaucoma medications for reducing eye pressure, little is known about the potential modality for neuronal survival and/or enhancement in visual impairments. Citicoline, a naturally occurring compound and FDA approved dietary supplement, is a nootropic agent that is recently demonstrated to be effective in ameliorating ischemic stroke, traumatic brain injury, Parkinson's disease, Alzheimer's disease, cerebrovascular diseases, memory disorders and attention-deficit/hyperactivity disorder in both humans and animal models. The mechanisms of its action appear to be multifarious including (i) preservation of cardiolipin, sphingomyelin, and arachidonic acid contents of phosphatidylcholine and phosphatidylethanolamine, (ii) restoration of phosphatidylcholine, (iii) stimulation of glutathione synthesis, (iv) lowering glutamate concentrations and preventing glutamate excitotoxicity, (v) rescuing mitochondrial function thereby preventing oxidative damage and onset of neuronal apoptosis, (vi) synthesis of myelin leading to improvement in neuronal membrane integrity, (vii) improving acetylcholine synthesis and thereby reducing the effects of mental stress and (viii) preventing endothelial dysfunction. Such effects have vouched for citicoline as a neuroprotective, neurorestorative and neuroregenerative agent. Retinal ganglion cells are neurons with long myelinated axons which provide a strong rationale for citicoline use in visual pathway disorders. Since glaucoma is a form of neurodegeneration involving retinal ganglion cells, citicoline may help ameliorate glaucomatous damages in multiple facets. Additionally, trans-synaptic degeneration has been identified in humans and experimental models of glaucoma suggesting the cholinergic system as a new brain target for glaucoma management and therapy.
Collapse
Affiliation(s)
- Muneeb A Faiq
- Department of Ophthalmology, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, United States
| | - Gadi Wollstein
- Department of Ophthalmology, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, United States
| | - Joel S Schuman
- Department of Ophthalmology, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, United States
| | - Kevin C Chan
- Department of Ophthalmology, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, United States; Department of Radiology, New York University (NYU) School of Medicine, NYU Langone Health, New York, NY, United States; Center for Neural Science, Faculty of Arts and Science, New York University, New York, NY, United States.
| |
Collapse
|
19
|
Towards A Microbead Occlusion Model of Glaucoma for a Non-Human Primate. Sci Rep 2019; 9:11572. [PMID: 31399621 PMCID: PMC6689098 DOI: 10.1038/s41598-019-48054-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/27/2019] [Indexed: 12/16/2022] Open
Abstract
Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). The disease causes vision loss through the degeneration of retinal ganglion cell neurons and their axons in the optic nerve. Using an inducible model of glaucoma, we elevated IOP in the squirrel monkey (Saimiri boliviensis) using intracameral injection of 35 μm polystyrene microbeads and measured common pathogenic outcomes in the optic projection. A 42% elevation in IOP over 28 weeks reduced anterograde transport of fluorescently-labeled cholera toxin beta from retina to the lateral geniculate nucleus (60% decrease), and to the superior colliculus (49% decrease). Pressure also reduced survival of ganglion cellaxons in the optic nerve by 22%. The same elevation caused upregulation of proteins associated with glaucomatous neurodegeneration in the retina and optic nerve, including complement 1q, interleukin 6, and brain-derived neurotrophic factor. That axon degeneration in the nerve lagged deficits in anterograde transport is consistent with progression in rodent models, while the observed protein changes also occur in tissue from human glaucoma patients. Thus, microbead occlusion in a non-human primate with a visual system similar to our own represents an attractive model to investigate neurodegenerative mechanisms and therapeutic interventions for glaucoma.
Collapse
|
20
|
Singh PK, Kasetti RB, Zode GS, Goyal A, Juzych MS, Kumar A. Zika Virus Infects Trabecular Meshwork and Causes Trabeculitis and Glaucomatous Pathology in Mouse Eyes. mSphere 2019; 4:e00173-19. [PMID: 31068433 PMCID: PMC6506617 DOI: 10.1128/msphere.00173-19] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/25/2019] [Indexed: 12/28/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy leads to devastating fetal outcomes, including neurological (microcephaly) and ocular pathologies such as retinal lesions, optic nerve abnormalities, chorioretinal atrophy, and congenital glaucoma. Only clinical case reports have linked ZIKV infection to causing glaucoma, a major blinding eye disease. In the present study, we have investigated the role of ZIKV in glaucoma pathophysiology using in vitro and in vivo experimental models. We showed that human primary trabecular meshwork (Pr. TM) cells, as well as a human GTM3 cell line, were permissive to ZIKV infection. ZIKV induced the transcription of various genes expressing pattern recognition receptors (TLR2, TLR3, and RIG-I), cytokines/chemokines (TNF-α, IL-1β, CCL5, and CXCL10), interferons (IFN-α2, IFN-β1, and IFN-γ), and interferon-stimulated genes (ISG15 and OAS2) in Pr. TM cells. ZIKV infection in IFNAR1-/- and wild-type (WT) mouse eyes resulted in increased intraocular pressure (IOP) and the development of chorioretinal atrophy. Anterior chamber (AC) inoculation of ZIKV caused infectivity in iridocorneal angle and TM, leading to the death of TM cells in the mouse eyes. Moreover, anterior segment tissue of infected eyes exhibited increased expression of inflammatory mediators and interferons. Furthermore, ZIKV infection in IFNAR1-/- mice resulted in retinal ganglion cell (RGC) death and loss, coinciding with optic nerve infectivity and disruption of anterograde axonal transport. Because of similarity in glaucomatous pathologies in our study and other experimental glaucoma models, ZIKV infection can be used to study infectious triggers of glaucoma, currently an understudied area of investigation.IMPORTANCE Ocular complications due to ZIKV infection remains a major public health concern because of their ability to cause visual impairment or blindness. Most of the previous studies have shown ZIKV-induced ocular pathology in the posterior segment (i.e., retina) of the eye. However, some recent clinical reports from affected countries highlighted the importance of ZIKV in affecting the anterior segment of the eye and causing congenital glaucoma. Because glaucoma is the second leading cause of blindness worldwide, it is imperative to study ZIKV infection in causing glaucoma to identify potential targets for therapeutic intervention. In this study, we discovered that ZIKV permissively infects human TM cells and evokes inflammatory responses causing trabeculitis. Using a mouse model, we demonstrated that ZIKV infection resulted in higher IOP, increased RGC loss, and optic nerve abnormalities, the classical hallmarks of glaucoma. Collectively, our study provides new insights into ocular ZIKV infection resulting in glaucomatous pathology.
Collapse
Affiliation(s)
- Pawan Kumar Singh
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ramesh B Kasetti
- The North Texas Eye Research Institute and the Department of Pharmacology and Neurosciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Gulab S Zode
- The North Texas Eye Research Institute and the Department of Pharmacology and Neurosciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Anju Goyal
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mark S Juzych
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences/Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
21
|
Pöyhönen S, Er S, Domanskyi A, Airavaara M. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Front Physiol 2019; 10:486. [PMID: 31105589 PMCID: PMC6499070 DOI: 10.3389/fphys.2019.00486] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/08/2019] [Indexed: 12/28/2022] Open
Abstract
Astrocytes, oligodendrocytes, and microglia are abundant cell types found in the central nervous system and have been shown to play crucial roles in regulating both normal and disease states. An increasing amount of evidence points to the critical importance of glia in mediating neurodegeneration in Alzheimer’s and Parkinson’s diseases (AD, PD), and in ischemic stroke, where microglia are involved in initial tissue clearance, and astrocytes in the subsequent formation of a glial scar. The importance of these cells for neuronal survival has previously been studied in co-culture experiments and the search for neurotrophic factors (NTFs) initiated after finding that the addition of conditioned media from astrocyte cultures could support the survival of primary neurons in vitro. This led to the discovery of the potent dopamine neurotrophic factor, glial cell line-derived neurotrophic factor (GDNF). In this review, we focus on the relationship between glia and NTFs including neurotrophins, GDNF-family ligands, CNTF family, and CDNF/MANF-family proteins. We describe their expression in astrocytes, oligodendrocytes and their precursors (NG2-positive cells, OPCs), and microglia during development and in the adult brain. Furthermore, we review existing data on the glial phenotypes of NTF knockout mice and follow NTF expression patterns and their effects on glia in disease models such as AD, PD, stroke, and retinal degeneration.
Collapse
Affiliation(s)
- Suvi Pöyhönen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Safak Er
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Zhu Y, Pappas AC, Wang R, Seifert P, Sun D, Jakobs TC. Ultrastructural Morphology of the Optic Nerve Head in Aged and Glaucomatous Mice. Invest Ophthalmol Vis Sci 2019; 59:3984-3996. [PMID: 30098187 PMCID: PMC6082327 DOI: 10.1167/iovs.18-23885] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To study age- and intraocular pressure–induced changes in the glial lamina of the murine optic nerve on the ultrastructural level. Methods Naïve C57bl/6 mice at various ages spanning the time between early adulthood (3 months) and senescence (30 months) were used in this study. In addition, the intraocular pressure (IOP) was increased in a group of young mice by injection of microbeads into the anterior chamber. The unmyelinated segments of the optic nerve containing the glial lamina were prepared for transmission electron microscopy and imaged at high resolution. Results Axon packing density decreased slightly with age. Aging nerves contained higher numbers of enlarged and degenerating axons. Mean axonal diameter and in particular the variance of axonal diameter correlated well with age. Axonal mitochondria also showed age-dependent signs of pathology. The mean diameter of axonal mitochondria increased, and aged axons often contained profiles of mitochondria with very few or no cristae. Astrocytic mitochondria remained normal even in very old nerves. Changes to axons and axonal mitochondria in young glaucomatous nerves were comparable with those of 18- to 30-month-old naïve mice. In addition to axons and mitochondria, aged and glaucomatous nerves showed thickening of the blood vessel basement membranes and increased deposition of basement membrane collagen. Conclusions On the ultrastructural level, the effects of age and elevated IOP are quite similar. One month of elevated IOP seems to have as strongly detrimental effects on the nerve as at least 18 months of normal aging.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Ophthalmology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States
| | - Anthony C Pappas
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States
| | - Rui Wang
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States.,Department of Ophthalmology, The First Hospital of Xi'an, Xi'an, Shaanxi, China
| | - Philip Seifert
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States
| | - Daniel Sun
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston Massachusetts, United States
| |
Collapse
|
23
|
Geeraerts E, Claes M, Dekeyster E, Salinas-Navarro M, De Groef L, Van den Haute C, Scheyltjens I, Baekelandt V, Arckens L, Moons L. Optogenetic Stimulation of the Superior Colliculus Confers Retinal Neuroprotection in a Mouse Glaucoma Model. J Neurosci 2019; 39:2313-2325. [PMID: 30655352 PMCID: PMC6433760 DOI: 10.1523/jneurosci.0872-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 11/15/2018] [Accepted: 12/29/2018] [Indexed: 12/12/2022] Open
Abstract
Glaucoma is characterized by a progressive loss of retinal ganglion cells (RGCs) in the eye, which ultimately results in visual impairment or even blindness. Because current therapies often fail to halt disease progression, there is an unmet need for novel neuroprotective therapies to support RGC survival. Various research lines suggest that visual target centers in the brain support RGC functioning and survival. Here, we explored whether increasing neuronal activity in one of these projection areas could improve survival of RGCs in a mouse glaucoma model. Prolonged activation of an important murine RGC target area, the superior colliculus (SC), was established via a novel optogenetic stimulation paradigm. By leveraging the unique channel kinetics of the stabilized step function opsin (SSFO), protracted stimulation of the SC was achieved with only a brief light pulse. SSFO-mediated collicular stimulation was confirmed by immunohistochemistry for the immediate-early gene c-Fos and behavioral tracking, which both demonstrated consistent neuronal activity upon repeated stimulation. Finally, the neuroprotective potential of optogenetic collicular stimulation was investigated in mice of either sex subjected to a glaucoma model and a 63% reduction in RGC loss was found. This work describes a new paradigm for optogenetic collicular stimulation and a first demonstration that increasing target neuron activity can increase survival of the projecting neurons.SIGNIFICANCE STATEMENT Despite glaucoma being a leading cause of blindness and visual impairment worldwide, no curative therapies exist. This study describes a novel paradigm to reduce retinal ganglion cell (RGC) degeneration underlying glaucoma. Building on previous observations that RGC survival is supported by the target neurons to which they project and using an innovative optogenetic approach, we increased neuronal activity in the mouse superior colliculus, a main projection target of rodent RGCs. This proved to be efficient in reducing RGC loss in a glaucoma model. Our findings establish a new optogenetic paradigm for target stimulation and encourage further exploration of the molecular signaling pathways mediating retrograde neuroprotective communication.
Collapse
Affiliation(s)
- Emiel Geeraerts
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Eline Dekeyster
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Manuel Salinas-Navarro
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Chris Van den Haute
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
- Viral Vector Core Leuven, KU Leuven, 3000 Leuven, Belgium, and
| | - Isabelle Scheyltjens
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology; KU Leuven, 3000 Leuven, Belgium
| | - Veerle Baekelandt
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
| | - Lutgarde Arckens
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology; KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium,
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
24
|
Smith MA, Plyler ES, Dengler-Crish CM, Meier J, Crish SD. Nodes of Ranvier in Glaucoma. Neuroscience 2018; 390:104-118. [PMID: 30149050 DOI: 10.1016/j.neuroscience.2018.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/15/2018] [Accepted: 08/17/2018] [Indexed: 01/13/2023]
Abstract
Retinal ganglion cell axons of the DBA/2J mouse model of glaucoma, a model characterized by extensive neuroinflammation, preserve synaptic contacts with their subcortical targets for a time after onset of anterograde axonal transport deficits, axon terminal hypertrophy, and cytoskeletal alterations. Though retrograde axonal transport is still evident in these axons, it is unknown if they retain their ability to transmit visual information to the brain. Using a combination of in vivo multiunit electrophysiology, neuronal tract tracing, multichannel immunofluorescence, and transmission electron microscopy, we report that eye-brain signaling deficits precede transport loss and axonal degeneration in the DBA/2J retinal projection. These deficits are accompanied by node of Ranvier pathology - consisting of increased node length and redistribution of the voltage-gated sodium channel Nav1.6 that parallel changes seen early in multiple sclerosis (MS) axonopathy. Further, with age, axon caliber and neurofilament density increase without corresponding changes in myelin thickness. In contrast to these findings in DBA/2J mice, node pathologies were not observed in the induced microbead occlusion model of glaucoma - a model that lacks pre-existing inflammation. After one week of systemic treatment with fingolimod, an immunosuppressant therapy for relapsing-remitting MS, DBA/2J mice showed a substantial reduction in node pathology and mild effects on axon morphology. These data suggest that neurophysiological deficits in the DBA/2J may be due to defects in intact axons and targeting node pathology may be a promising intervention for some types of glaucoma.
Collapse
Affiliation(s)
- M A Smith
- Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - E S Plyler
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, United States
| | - C M Dengler-Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - J Meier
- Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - S D Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States.
| |
Collapse
|
25
|
Fischer RA, Zhang Y, Risner ML, Li D, Xu Y, Sappington RM. Impact of Graphene on the Efficacy of Neuron Culture Substrates. Adv Healthc Mater 2018; 7:e1701290. [PMID: 29943431 PMCID: PMC6105445 DOI: 10.1002/adhm.201701290] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/11/2018] [Indexed: 01/09/2023]
Abstract
How graphene influences the behavior of living cells or tissues remains a critical issue for its application in biomedical studies, despite the general acceptance that graphene is biocompatible. While direct contact between cells and graphene is not a requirement for all biomedical applications, it is often mandatory for biosensing. Therefore, it is important to clarify whether graphene impedes the ability of cells to interact with biological elements in their environment. Here, a systematic study is reported to determine whether applying graphene on top of matrix substrates masks interactions between these substrates and retinal ganglion cells (RGCs). Six different platforms are tested for primary RGC cultures with three platforms comprised of matrix substrates compatible with these neurons, and another three having a layer of graphene placed on top of the matrix substrates. The results demonstrate that graphene does not impede interactions between RGCs and underlying substrate matrix, such that their positive or negative effects on neuron viability and vitality are retained. However, direct contact between RGCs and graphene reduces the number, but increases basal activity, of functional cation channels. The data indicate that, when proper baselines are established, graphene is a promising biosensing material for in vitro applications in neuroscience.
Collapse
Affiliation(s)
- Rachel A. Fischer
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy and Department of Electrical, Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Yuchen Zhang
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy and Department of Electrical, Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Michael L. Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy and Department of Electrical, Engineering and Computer Science, Vanderbilt University, Nashville, TN 37235, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Department of Pharmacology, Vanderbilt University School of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | | | | | | |
Collapse
|
26
|
Fry LE, Fahy E, Chrysostomou V, Hui F, Tang J, van Wijngaarden P, Petrou S, Crowston JG. The coma in glaucoma: Retinal ganglion cell dysfunction and recovery. Prog Retin Eye Res 2018; 65:77-92. [DOI: 10.1016/j.preteyeres.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/18/2018] [Accepted: 04/03/2018] [Indexed: 01/07/2023]
|
27
|
Cooper ML, Collyer JW, Calkins DJ. Astrocyte remodeling without gliosis precedes optic nerve Axonopathy. Acta Neuropathol Commun 2018; 6:38. [PMID: 29747701 PMCID: PMC5946396 DOI: 10.1186/s40478-018-0542-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 04/26/2018] [Indexed: 11/10/2022] Open
Abstract
Astroyctes serve myriad functions but are especially critical in white matter tracts, where energy-demanding axons propagate action potentials great distances between neurons. Axonal dependence on astrocytes for even normal function accentuates the critical role astrocytes serve during disease. In glaucoma, the most common optic neuropathy, sensitivity to intraocular pressure (IOP) challenges RGC axons early, including degradation of anterograde transport to the superior colliculus (SC). Astrocyte remodeling presages overt axon degeneration in glaucoma and thus may present a therapeutic opportunity. Here we developed a novel metric to quantify organization of astrocyte processes in the optic nerve relative to axon degeneration in the DBA/2 J hereditary mouse model of glaucoma. In early progression, as axons expand prior to loss, astrocyte processes become more parallel with migration to the nerve’s edge without a change in overall coverage of the nerve. As axons degenerate, astrocyte parallelism diminishes with increased glial coverage and reinvasion of the nerve. In longitudinal sections through aged DBA/2 J nerve, increased astrocyte parallelism reflected elevated levels of the astrocyte gap-junction protein connexin 43 (Cx43). In the distal nerve, increased Cx43 also indicated with a higher level of intact anterograde transport from retina to SC. Our results suggest that progression of axonopathy in the optic nerve involves astrocyte remodeling in two phases. In an early phase, astrocyte processes organize in parallel, likely through gap-junction coupling, while a later phase involves deterioration of organization as glial coverage increases and axons are lost.
Collapse
|
28
|
Sharif NA. iDrugs and iDevices Discovery Research: Preclinical Assays, Techniques, and Animal Model Studies for Ocular Hypotensives and Neuroprotectants. J Ocul Pharmacol Ther 2018; 34:7-39. [PMID: 29323613 DOI: 10.1089/jop.2017.0125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Discovery ophthalmic research is centered around delineating the molecular and cellular basis of ocular diseases and finding and exploiting molecular and genetic pathways associated with them. From such studies it is possible to determine suitable intervention points to address the disease process and hopefully to discover therapeutics to treat them. An investigational new drug (IND) filing for a new small-molecule drug, peptide, antibody, genetic treatment, or a device with global health authorities requires a number of preclinical studies to provide necessary safety and efficacy data. Specific regulatory elements needed for such IND-enabling studies are beyond the scope of this article. However, to enhance the overall data packages for such entities and permit high-quality foundation-building publications for medical affairs, additional research and development studies are always desirable. This review aims to provide examples of some target localization/verification, ocular drug discovery processes, and mechanistic and portfolio-enhancing exploratory investigations for candidate drugs and devices for the treatment of ocular hypertension and glaucomatous optic neuropathy (neurodegeneration of retinal ganglion cells and their axons). Examples of compound screening assays, use of various technologies and techniques, deployment of animal models, and data obtained from such studies are also presented.
Collapse
Affiliation(s)
- Najam A Sharif
- 1 Global Alliances & External Research , Santen Incorporated, Emeryville, California.,2 Department of Pharmaceutical Sciences, Texas Southern University , Houston, Texas.,3 Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center , Fort Worth, Texas
| |
Collapse
|
29
|
Crish SD, Schofield BR. Anterograde Tract Tracing for Assaying Axonopathy and Transport Deficits in Glaucoma. Methods Mol Biol 2018; 1695:171-185. [PMID: 29190027 DOI: 10.1007/978-1-4939-7407-8_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Whether to stage degeneration or investigate early pathology in glaucoma, examination of axonal structure and function is essential. There are a wide variety of methods available to investigators using animal models of glaucoma, with varying utilities depending on the questions asked. Here, we describe the use of anterograde neuronal tract tracing using cholera toxin B (CTB) for the determination of axon transport integrity of the retinofugal projection. This method reveals the structure of the retinal axons as well as the functional integrity of anterograde transport systems.
Collapse
Affiliation(s)
- Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, RGE-135, 4209 St. Rt. 44, Rootstown, OH, 44272, USA.
| | - Brett R Schofield
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| |
Collapse
|
30
|
Wang S, Hu T, Wang Z, Li N, Zhou L, Liao L, Wang M, Liao L, Wang H, Zeng L, Fan C, Zhou H, Xiong K, Huang J, Chen D. Macroglia-derived thrombospondin 2 regulates alterations of presynaptic proteins of retinal neurons following elevated hydrostatic pressure. PLoS One 2017; 12:e0185388. [PMID: 28953973 PMCID: PMC5617560 DOI: 10.1371/journal.pone.0185388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/12/2017] [Indexed: 02/03/2023] Open
Abstract
Many studies on retinal injury and repair following elevated intraocular pressure suggest that the survival ratio of retinal neurons has been improved by various measures. However, the visual function recovery is far lower than expected. The homeostasis of retinal synapses in the visual signal pathway is the key structural basis for the delivery of visual signals. Our previous studies found that complicated changes in the synaptic structure between retinal neurons occurred much earlier than obvious degeneration of retinal ganglion cells in rat retinae. The lack of consideration of these earlier retinal synaptic changes in the rescue strategy may be partly responsible for the limited visual function recovery with the types of protective methods for retinal neurons used following elevated intraocular pressure. Thus, research on the modulatory mechanisms of the synaptic changes after elevated intraocular pressure injury may give new light to visual function rescue. In this study, we found that thrombospondin 2, an important regulator of synaptogenesis in central nervous system development, was distributed in retinal macroglia cells, and its receptor α2δ-1 was in retinal neurons. Cell cultures including mixed retinal macroglia cells/neuron cultures and retinal neuron cultures were exposed to elevated hydrostatic pressure for 2 h. The expression levels of glial fibrillary acidic protein (the marker of activated macroglia cells), thrombospondin 2, α2δ-1 and presynaptic proteins were increased following elevated hydrostatic pressure in mixed cultures, but the expression levels of postsynaptic proteins were not changed. SiRNA targeting thrombospondin 2 could decrease the upregulation of presynaptic proteins induced by the elevated hydrostatic pressure. However, in retinal neuron cultures, elevated hydrostatic pressure did not affect the expression of presynaptic or postsynaptic proteins. Rather, the retinal neuron cultures with added recombinant thrombospondin 2 protein upregulated the level of presynaptic proteins. Finally, gabapentin decreased the expression of presynaptic proteins in mixed cultures by blocking the interaction of thrombospondin 2 and α2δ-1. Taken together, these results indicate that activated macroglia cells may participate in alterations of presynaptic proteins of retinal neurons following elevated hydrostatic pressure, and macroglia-derived thrombospondin 2 may modulate these changes via binding to its neuronal receptor α2δ-1.
Collapse
Affiliation(s)
- Shuchao Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Tu Hu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Zhen Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Na Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Lihong Zhou
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Mi Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Libin Liao
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Hui Wang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Leping Zeng
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Chunling Fan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Hongkang Zhou
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Sciences, Changsha, Hunan, China
| |
Collapse
|
31
|
He S, Stankowska DL, Ellis DZ, Krishnamoorthy RR, Yorio T. Targets of Neuroprotection in Glaucoma. J Ocul Pharmacol Ther 2017; 34:85-106. [PMID: 28820649 PMCID: PMC5963639 DOI: 10.1089/jop.2017.0041] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/08/2017] [Indexed: 12/14/2022] Open
Abstract
Progressive neurodegeneration of the optic nerve and the loss of retinal ganglion cells is a hallmark of glaucoma, the leading cause of irreversible blindness worldwide, with primary open-angle glaucoma (POAG) being the most frequent form of glaucoma in the Western world. While some genetic mutations have been identified for some glaucomas, those associated with POAG are limited and for most POAG patients, the etiology is still unclear. Unfortunately, treatment of this neurodegenerative disease and other retinal degenerative diseases is lacking. For POAG, most of the treatments focus on reducing aqueous humor formation, enhancing uveoscleral or conventional outflow, or lowering intraocular pressure through surgical means. These efforts, in some cases, do not always lead to a prevention of vision loss and therefore other strategies are needed to reduce or reverse the progressive neurodegeneration. In this review, we will highlight some of the ocular pharmacological approaches that are being tested to reduce neurodegeneration and provide some form of neuroprotection.
Collapse
Affiliation(s)
- Shaoqing He
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Dorota L Stankowska
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Dorette Z Ellis
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Raghu R Krishnamoorthy
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Thomas Yorio
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| |
Collapse
|
32
|
Yang H, Reynaud J, Lockwood H, Williams G, Hardin C, Reyes L, Stowell C, Gardiner SK, Burgoyne CF. The connective tissue phenotype of glaucomatous cupping in the monkey eye - Clinical and research implications. Prog Retin Eye Res 2017; 59:1-52. [PMID: 28300644 PMCID: PMC5603293 DOI: 10.1016/j.preteyeres.2017.03.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/14/2017] [Accepted: 03/06/2017] [Indexed: 10/20/2022]
Abstract
In a series of previous publications we have proposed a framework for conceptualizing the optic nerve head (ONH) as a biomechanical structure. That framework proposes important roles for intraocular pressure (IOP), IOP-related stress and strain, cerebrospinal fluid pressure (CSFp), systemic and ocular determinants of blood flow, inflammation, auto-immunity, genetics, and other non-IOP related risk factors in the physiology of ONH aging and the pathophysiology of glaucomatous damage to the ONH. The present report summarizes 20 years of technique development and study results pertinent to the characterization of ONH connective tissue deformation and remodeling in the unilateral monkey experimental glaucoma (EG) model. In it we propose that the defining pathophysiology of a glaucomatous optic neuropathy involves deformation, remodeling, and mechanical failure of the ONH connective tissues. We view this as an active process, driven by astrocyte, microglial, fibroblast and oligodendrocyte mechanobiology. These cells, and the connective tissue phenomena they propagate, have primary and secondary effects on retinal ganglion cell (RGC) axon, laminar beam and retrolaminar capillary homeostasis that may initially be "protective" but eventually lead to RGC axonal injury, repair and/or cell death. The primary goal of this report is to summarize our 3D histomorphometric and optical coherence tomography (OCT)-based evidence for the early onset and progression of ONH connective tissue deformation and remodeling in monkey EG. A second goal is to explain the importance of including ONH connective tissue processes in characterizing the phenotype of a glaucomatous optic neuropathy in all species. A third goal is to summarize our current efforts to move from ONH morphology to the cell biology of connective tissue remodeling and axonal insult early in the disease. A final goal is to facilitate the translation of our findings and ideas into neuroprotective interventions that target these ONH phenomena for therapeutic effect.
Collapse
Affiliation(s)
- Hongli Yang
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Juan Reynaud
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Howard Lockwood
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Galen Williams
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Christy Hardin
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Luke Reyes
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Cheri Stowell
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Stuart K Gardiner
- Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Claude F Burgoyne
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States.
| |
Collapse
|
33
|
Echevarria FD, Formichella CR, Sappington RM. Interleukin-6 Deficiency Attenuates Retinal Ganglion Cell Axonopathy and Glaucoma-Related Vision Loss. Front Neurosci 2017; 11:318. [PMID: 28620279 PMCID: PMC5450377 DOI: 10.3389/fnins.2017.00318] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/19/2017] [Indexed: 11/13/2022] Open
Abstract
The pleotropic cytokine interleukin-6 (IL-6) is implicated in retinal ganglion cell (RGC) survival and degeneration, including that associated with glaucoma. IL-6 protects RGCs from pressure-induced apoptosis in vitro. However, it is unknown how IL-6 impacts glaucomatous degeneration in vivo. To study how IL-6 influences glaucomatous RGC axonopathy, accompanying glial reactivity, and resultant deficits in visual function, we performed neural tracing, histological, and neurobehavioral assessments in wildtype (B6;129SF2/J; WT) and IL-6 knock-out mice (B6;129S2-IL6tm1kopf/J; IL-6-/-) after 8 weeks of unilateral or bilateral microbead-induced glaucoma (microbead occlusion model). IOP increased by 20% following microbead injection in both genotypes (p < 0.05). However, deficits in wound healing at the site of corneal injection were noted. In WT mice, elevated IOP produced degenerating axon profiles and decreased axon density in the optic nerve by 15% (p < 0.01). In IL-6-/- mice, axon density in the optic nerve did not differ between microbead- and saline-injected mice (p > 0.05) and degenerating axon profiles were minimal. Preservation of RGC axons was reflected in visual function, where visual acuity decreased significantly in a time-dependent manner with microbead-induced IOP elevation in WT (p < 0.001), but not IL-6-/- mice (p > 0.05). Despite this preservation of RGC axons and visual acuity, both microbead-injected WT and IL-6-/- mice exhibited a 50% decrease in anterograde CTB transport to the superior colliculus, as compared to saline-injected controls (p < 0.01). Assessment of glial reactivity revealed no genotype- or IOP-dependent changes in retinal astrocytes. IOP elevation decreased microglia density and percent retinal area covered in WT mice (p < 0.05), while IL-6-/- mice exhibited only a decrease in density (p < 0.05). Together, our findings indicate that two defining features of RGC axonopathy—axon transport deficits and structural degeneration of axons—likely occur via independent mechanisms. Our data suggest that IL-6 is part of a mechanism that specifically leads to structural degeneration of axons. Furthermore, its absence is sufficient to prevent both structural degeneration of the optic nerve and vision loss. Overall, our work supports the proposition that functional deficits in axon transport represent a therapeutic window for RGC axonopathy and identify IL-6 signaling as a strong target for such a therapeutic.
Collapse
Affiliation(s)
| | - Cathryn R Formichella
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of MedicineNashville, TN, United States.,Vanderbilt Eye Institute, Vanderbilt University Medical CenterNashville, TN, United States
| | - Rebecca M Sappington
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of MedicineNashville, TN, United States.,Vanderbilt Eye Institute, Vanderbilt University Medical CenterNashville, TN, United States.,Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, United States
| |
Collapse
|
34
|
Lambert WS, Carlson BJ, Formichella CR, Sappington RM, Ahlem C, Calkins DJ. Oral Delivery of a Synthetic Sterol Reduces Axonopathy and Inflammation in a Rodent Model of Glaucoma. Front Neurosci 2017; 11:45. [PMID: 28223915 PMCID: PMC5293777 DOI: 10.3389/fnins.2017.00045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/20/2017] [Indexed: 12/12/2022] Open
Abstract
Glaucoma is a group of optic neuropathies associated with aging and sensitivity to intraocular pressure (IOP). The disease is the leading cause of irreversible blindness worldwide. Early progression in glaucoma involves dysfunction of retinal ganglion cell (RGC) axons, which comprise the optic nerve. Deficits in anterograde transport along RGC axons to central visual structures precede outright degeneration, and preventing these deficits is efficacious at abating subsequent progression. HE3286 is a synthetic sterol derivative that has shown therapeutic promise in models of inflammatory disease and neurodegenerative disease. We examined the efficacy of HE3286 oral delivery in preventing loss of anterograde transport in an inducible model of glaucoma (microbead occlusion). Adult rats received HE3286 (20 or 100 mg/kg) or vehicle daily via oral gavage for 4 weeks. Microbead occlusion elevated IOP ~30% in all treatment groups, and elevation was not affected by HE3286 treatment. In the vehicle group, elevated IOP reduced anterograde axonal transport to the superior colliculus, the most distal site in the optic projection, by 43% (p = 0.003); HE3286 (100 mg/kg) prevented this reduction (p = 0.025). HE3286 increased brain-derived neurotrophic factor (BDNF) in the optic nerve head and retina, while decreasing inflammatory and pathogenic proteins associated with elevated IOP compared to vehicle treatment. Treatment with HE3286 also increased nuclear localization of the transcription factor NFκB in collicular and retinal neurons, but decreased NFκB in glial nuclei in the optic nerve head. Thus, HE3286 may have a neuroprotective influence in glaucoma, as well as other chronic neurodegenerations.
Collapse
Affiliation(s)
- Wendi S Lambert
- Vanderbilt University Medical Center, The Vanderbilt Eye Institute Nashville, TN, USA
| | - Brian J Carlson
- Vanderbilt University Medical Center, The Vanderbilt Eye Institute Nashville, TN, USA
| | - Cathryn R Formichella
- Vanderbilt University Medical Center, The Vanderbilt Eye Institute Nashville, TN, USA
| | - Rebecca M Sappington
- Vanderbilt University Medical Center, The Vanderbilt Eye Institute Nashville, TN, USA
| | | | - David J Calkins
- Vanderbilt University Medical Center, The Vanderbilt Eye Institute Nashville, TN, USA
| |
Collapse
|
35
|
Calkins DJ, Pekny M, Cooper ML, Benowitz L. The challenge of regenerative therapies for the optic nerve in glaucoma. Exp Eye Res 2017; 157:28-33. [PMID: 28153739 DOI: 10.1016/j.exer.2017.01.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/12/2017] [Accepted: 01/26/2017] [Indexed: 11/15/2022]
Abstract
This review arose from a discussion of regenerative therapies to treat optic nerve degeneration in glaucoma at the 2015 Lasker/IRRF Initiative on Astrocytes and Glaucomatous Neurodegeneration. In addition to the authors, participants included Jonathan Crowston, Andrew Huberman, Elaine Johnson, Richard Lu, Hemai Phatnami, Rebecca Sappington, and Don Zack. Glaucoma is a neurodegenerative disease of the optic nerve, and is the leading cause of irreversible blindness worldwide. The disease progresses as sensitivity to intraocular pressure (IOP) is conveyed through the optic nerve head to distal retinal ganglion cell (RGC) projections. Because the nerve and retina are components of the central nervous system (CNS), their intrinsic regenerative capacity is limited. However, recent research in regenerative therapies has resulted in multiple breakthroughs that may unlock the optic nerve's regenerative potential. Increasing levels of Schwann-cell derived trophic factors and reducing potent cell-intrinsic suppressors of regeneration have resulted in axonal regeneration even beyond the optic chiasm. Despite this success, many challenges remain. RGC axons must be able to form new connections with their appropriate targets in central brain regions and these connections must be retinotopically correct. Furthermore, for new axons penetrating the optic projection, oligodendrocyte glia must provide myelination. Additionally, reactive gliosis and inflammation that increase the regenerative capacity must be outweigh pro-apoptotic processes to create an environment within which maximal regeneration can occur.
Collapse
Affiliation(s)
- David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37205, USA.
| | - Milos Pekny
- Department of Clinical Neuroscience, Sahlgrenska Academy at University of Gothenburg, 41345, Göteborg, Sweden
| | - Melissa L Cooper
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37205, USA
| | - Larry Benowitz
- Departments of Neurosurgery and Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
36
|
Breen KT, Anderson SR, Steele MR, Calkins DJ, Bosco A, Vetter ML. Loss of Fractalkine Signaling Exacerbates Axon Transport Dysfunction in a Chronic Model of Glaucoma. Front Neurosci 2016; 10:526. [PMID: 27932942 PMCID: PMC5123443 DOI: 10.3389/fnins.2016.00526] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 10/31/2016] [Indexed: 01/01/2023] Open
Abstract
Neurodegeneration in glaucoma results in decline and loss of retinal ganglion cells (RGCs), and is associated with activation of myeloid cells such as microglia and macrophages. The chemokine fractalkine (FKN or Cx3cl1) mediates communication from neurons to myeloid cells. Signaling through its receptor Cx3cr1 has been implicated in multiple neurodegenerative diseases, but the effects on neuronal pathology are variable. Since it is unknown how FKN-mediated crosstalk influences RGC degeneration in glaucoma, we assessed this in a chronic mouse model, DBA/2J. We analyzed a DBA/2J substrain deficient in Cx3cr1, and compared compartmentalized RGC degeneration and myeloid cell responses to those in standard DBA/2J mice. We found that loss of FKN signaling exacerbates axon transport dysfunction, an early event in neurodegeneration, with a significant increase in RGCs with somal accumulation of the axonal protein phosphorylated neurofilament, and reduced retinal expression of genes involved in axon transport, Kif1b, and Atp8a2. There was no change in the loss of Brn3-positive RGCs, and no difference in the extent of damage to the proximal optic nerve, suggesting that the loss of fractalkine signaling primarily affects axon transport. Since Cx3cr1 is specifically expressed in myeloid cells, we assessed changes in retinal microglial number and activation, changes in gene expression, and the extent of macrophage infiltration. We found that loss of fractalkine signaling led to innate immune changes within the retina, including increased infiltration of peripheral macrophages and upregulated nitric oxide synthase-2 (Nos-2) expression in myeloid cells, which contributes to the production of NO and can promote axon transport deficits. In contrast, resident retinal microglia appeared unchanged either in number, morphology, or expression of the myeloid activation marker ionized calcium binding adaptor molecule 1 (Iba1). There was also no significant increase in the proinflammatory gene interleukin 1 beta (Il1β). We conclude that loss of fractalkine signaling causes a selective worsening of axon transport dysfunction in RGCs, which is linked to enhanced Nos-2 expression in myeloid cells. Our findings suggest that distinct mechanisms may contribute to different aspects of RGC decline in glaucoma, with axonal transport selectively altered after loss of Cx3cr1 in microglia and/or macrophages.
Collapse
Affiliation(s)
- Kevin T Breen
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Sarah R Anderson
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Michael R Steele
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Nashville, TN, USA
| | - Alejandra Bosco
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Monica L Vetter
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| |
Collapse
|
37
|
Mysona BA, Zhao J, Bollinger KE. Role of BDNF/TrkB pathway in the visual system: Therapeutic implications for glaucoma. EXPERT REVIEW OF OPHTHALMOLOGY 2016; 12:69-81. [PMID: 28751923 DOI: 10.1080/17469899.2017.1259566] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Neuroprotective therapeutics are needed to treat glaucoma, an optic neuropathy that results in death of retinal ganglion cells (RGCs). AREAS COVERED The BDNF/TrkB pathway is important for RGC survival. Temporal and spatial alterations in the BDNF/TrkB pathway occur in development and in response to acute optic nerve injury and to glaucoma. In animal models, BDNF supplementation is successful at slowing RGC death after acute optic nerve injury and in glaucoma, however, the BDNF/TrkB signaling is not the only pathway supporting long term RGC survival. EXPERT COMMENTARY Much remains to be discovered about the interaction between retrograde, anterograde, and retinal BDNF/TrkB signaling pathways in both neurons and glia. An ideal therapeutic agent for glaucoma likely has several modes of action that target multiple mechanisms of neurodegeneration including the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- B A Mysona
- Augusta University Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute. Address: Augusta University Department of Cellular Biology and Anatomy, Health Sciences Campus, 1120 15th Street, Augusta, GA 30912, USA,
| | - J Zhao
- Medical College of Georgia, Department of Ophthalmology at Augusta University, James and Jean Culver Vision Discovery Institute. Address: Medical College of Georgia, Department of Ophthalmology at Augusta University, 1120 15th Street, Augusta, GA 30912, USA,
| | - K E Bollinger
- Medical College of Georgia, Department of Ophthalmology at Augusta University, Augusta University Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute. Address: Medical College of Georgia, Department of Ophthalmology at Augusta University, 1120 15th Street, Augusta, GA 30912, USA,
| |
Collapse
|
38
|
Wilson GN, Smith MA, Inman DM, Dengler-Crish CM, Crish SD. Early Cytoskeletal Protein Modifications Precede Overt Structural Degeneration in the DBA/2J Mouse Model of Glaucoma. Front Neurosci 2016; 10:494. [PMID: 27857681 PMCID: PMC5093131 DOI: 10.3389/fnins.2016.00494] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/14/2016] [Indexed: 12/12/2022] Open
Abstract
Axonal transport deficits precede structural loss in glaucoma and other neurodegenerations. Impairments in structural support, including modified cytoskeletal proteins, and microtubule-destabilizing elements, could be initiating factors in glaucoma pathogenesis. We investigated the time course of changes in protein levels and post-translational modifications in the DBA/2J mouse model of glaucoma. Using anterograde tract tracing of the retinal projection, we assessed major cytoskeletal and transported elements as a function of transport integrity in different stages of pathological progression. Using capillary-based electrophoresis, single- and multiplex immunosorbent assays, and immunofluorescence, we quantified hyperphosphorylated neurofilament-heavy chain, phosphorylated tau (ptau), calpain-mediated spectrin breakdown product (145/150 kDa), β–tubulin, and amyloid-β42 proteins based on age and transport outcome to the superior colliculus (SC; the main retinal target in mice). Phosphorylated neurofilament-heavy chain (pNF-H) was elevated within the optic nerve (ON) and SC of 8–10 month-old DBA/2J mice, but was not evident in the retina until 12–15 months, suggesting that cytoskeletal modifications first appear in the distal retinal projection. As expected, higher pNF-H levels in the SC and retina were correlated with axonal transport deficits. Elevations in hyperphosphorylated tau (ptau) occurred in ON and SC between 3 and 8 month of age while retinal ptau accumulations occurred at 12–15 months in DBA/2J mice. In vitro co-immunoprecipitation experiments suggested increased affinity of ptau for the retrograde motor complex protein dynactin. We observed a transport-related decrease of β-tubulin in ON of 10–12 month-old DBA/2J mice, suggesting destabilized microtubule array. Elevations in calpain-mediated spectrin breakdown product were seen in ON and SC at the earliest age examined, well before axonal transport loss is evident. Finally, transport-independent elevations of amyloid-β42, unlike pNF-H or ptau, occurred first in the retina of DBA/2J mice, and then progressed to SC. These data demonstrate distal-to-proximal progression of cytoskeletal modifications in the progression of glaucoma, with many of these changes occurring prior to complete loss of functional transport and axon degeneration. The earliest changes, such as elevated spectrin breakdown and amyloid-β levels, may make retinal ganglion cells susceptible to future stressors. As such, targeting modification of the axonal cytoskeleton in glaucoma may provide unique opportunities to slow disease progression.
Collapse
Affiliation(s)
- Gina N Wilson
- Department of Pharmaceutical Sciences, Northeast Ohio Medical UniversityRootstown, OH, USA; School of Biomedical Sciences, Kent State UniversityKent, OH, USA
| | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical UniversityRootstown, OH, USA; Integrated Pharmaceutical Medicine Program, Northeast Ohio Medical UniversityRootstown, OH, USA
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA
| | | | - Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA
| |
Collapse
|
39
|
Abstract
The clinical phenomenon of cupping has 2 principal pathophysiologic components in all optic neuropathies: prelaminar thinning and laminar deformation. We define prelaminar thinning to be the portion of cup enlargement that results from thinning of the prelaminar tissues due to physical compression and/or loss of retinal ganglion cell axons. We define laminar deformation or laminar cupping to be the portion of cup enlargement that results from permanent intraocular pressure (IOP)-induced deformation of the lamina cribrosa and peripapillary scleral connective tissues after damage and/or remodeling. We propose that the defining phenomenon of glaucomatous cupping is deformation and/or remodeling of the neural and connective tissues of the optic nerve head (ONH), which is governed by the distribution of IOP-related connective tissue stress and strain, regardless of the mechanism of insult or the level of IOP at which deformation and/or remodeling occurs. In other words, "glaucomatous cupping" is the term clinicians use to describe the clinical appearance and behavior the ONH assumes as its neural and connective tissues deform, remodel, or mechanically fail: 1) in a pattern and 2) by the several pathophysiologic processes governed by IOP-related connective tissue stress and strain. ONH biomechanics explains why a given ONH will demonstrate a certain form of "cupping" and at what level of IOP that might happen. Animal models are allowing us to tease apart the important components of cupping in IOP-related and non-IOP-related forms of optic neuropathy. A paradigm change in spectral domain optical coherence tomography ONH, retinal nerve fiber layer, and macular imaging should improve our ability to phenotype the ocular manifestations of many forms of damage to the visual system including glaucoma.
Collapse
|
40
|
Central visual pathways in glaucoma: evidence for distal mechanisms of neuronal self-repair. J Neuroophthalmol 2016; 35 Suppl 1:S29-37. [PMID: 26274834 DOI: 10.1097/wno.0000000000000291] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
As in other age-related neurodegenerative diseases, progression of neurodegeneration in glaucoma involves early axonopathy. In glaucoma, this is marked by degradation of active transport along retinal ganglion cell (RGC) axons projecting from the retina to the brain. In experimental systems, transport degradation occurs first in the most distal site in the RGC projection, the superior colliculus (SC) of the midbrain. Even as degradation progresses from one retinotopic sector to the next, important structures in the affected sectors persist, including synapses from RGC axon terminals onto SC neurons. This structural persistence is accompanied by focally increased brain-derived neurotrophic factor in hypertrophic SC astrocyte glia and defines a therapeutic window of opportunity. Thus, central brain structures in glaucoma may respond to disease-relevant stress by induction of mechanisms useful for maintaining retinal signals.
Collapse
|
41
|
Smith MA, Xia CZ, Dengler-Crish CM, Fening KM, Inman DM, Schofield BR, Crish SD. Persistence of intact retinal ganglion cell terminals after axonal transport loss in the DBA/2J mouse model of glaucoma. J Comp Neurol 2016; 524:3503-3517. [PMID: 27072596 DOI: 10.1002/cne.24012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/30/2016] [Accepted: 04/05/2016] [Indexed: 01/24/2023]
Abstract
Axonal transport defects are an early pathology occurring within the retinofugal projection of the DBA/2J mouse model of glaucoma. Retinal ganglion cell (RGC) axons and terminals are detectable after transport is affected, yet little is known about the condition of these structures. We examined the ultrastructure of the glaucomatous superior colliculus (SC) with three-dimensional serial block-face scanning electron microscopy to determine the distribution and morphology of retinal terminals in aged mice exhibiting varying levels of axonal transport integrity. After initial axonal transport failure, retinal terminal densities did not vary compared with either transport-intact or control tissue. Although retinal terminals lacked overt signs of neurodegeneration, transport-intact areas of glaucomatous SC exhibited larger retinal terminals and associated mitochondria. This likely indicates increased oxidative capacity and may be a compensatory response to the stressors that this projection is experiencing. Areas devoid of transported tracer label showed reduced mitochondrial volumes as well as decreased active zone number and surface area, suggesting that oxidative capacity and synapse strength are reduced as disease progresses but before degeneration of the synapse. Mitochondrial volume was a strong predictor of bouton size independent of pathology. These findings indicate that RGC axons retain connectivity after losing function early in the disease process, creating an important therapeutic opportunity for protection or restoration of vision in glaucoma. J. Comp. Neurol. 524:3503-3517, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, 44272.,Integrated Pharmaceutical Medicine Program, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Christina Z Xia
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | | | - Kelly M Fening
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Brett R Schofield
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, 44272
| | - Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, 44272.
| |
Collapse
|
42
|
Zheng Y, Zhang YM, Ni X. Urocortin 2 But Not Urocortin 3 Promotes the Synaptic Formation in Hipppocampal Neurons via Induction of NGF Production by Astrocytes. Endocrinology 2016; 157:1200-10. [PMID: 26713785 DOI: 10.1210/en.2015-1812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CRH family peptides play differential role during various physiological and pathophysiological responses, such as stress. Urocortins (UCNs) have been implicated to play complementary or contrasting actions for the effects of CRH during stress. It has been shown that activation of CRH receptor type 1 (CRHR1) results in decreased synapse formation in hippocampus. We therefore explored the effect of UCN2 and UCN3, the exclusive CRHR2 agonists, on synaptic formation in hippocampus. In hippocampal slices cultures, UCN2 but not UCN3 treatment increased the levels of presynaptic protein synapsinI and postsynaptic protein postsynaptic density 95 (PSD95), which was reversed by CRHR2 antagonist astressin 2B. In isolated hippocampal neurons, however, UCN2 decreased the numbers of synapsinI- and PSD95-labeled terminals/clusters via CRHR2. Treatment of hippocampal neurons with the media of UCN2-treated astrocytes led to an increase in synapsinI- and PSD95-labeled terminals. In neuron-astrocyte cocultures, UCN2 also enhanced the numbers and level of synapsinI- and PSD95-labeled terminals. These effects did not occur if glial cells were transfected with CRHR2 small interfering RNA. UCN2 but not UCN3 treatment induced nerve growth factor (NGF) production in astrocytes via CRHR2. The effects of the media of UCN2-treated glial cells on synapse formation in hippocampal neurons were prevented by administration of NGF receptor antagonists. Our data indicate that UCN2 promotes synapse formation in hippocampus via induction of NGF secretion from astrocytes. CRHR2 in glial cells mediates the stimulatory effects of CRH. Glia-neuron communication is critical for neuronal circuits remodeling and synaptic plasticity in response to neurohormones or neuromodulators.
Collapse
Affiliation(s)
- You Zheng
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Yan-Min Zhang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Xin Ni
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
43
|
Sappington RM, Sidorova T, Ward NJ, Chakravarthy R, Ho KW, Calkins DJ. Activation of transient receptor potential vanilloid-1 (TRPV1) influences how retinal ganglion cell neurons respond to pressure-related stress. Channels (Austin) 2016; 9:102-13. [PMID: 25713995 DOI: 10.1080/19336950.2015.1009272] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Our recent studies implicate the transient receptor potential vanilloid-1 (TRPV1) channel as a mediator of retinal ganglion cell (RGC) function and survival. With elevated pressure in the eye, TRPV1 increases in RGCs, supporting enhanced excitability, while Trpv1 -/- accelerates RGC degeneration in mice. Here we find TRPV1 localized in monkey and human RGCs, similar to rodents. Expression increases in RGCs exposed to acute changes in pressure. In retinal explants, contrary to our animal studies, both Trpv1 -/- and pharmacological antagonism of the channel prevented pressure-induced RGC apoptosis, as did chelation of extracellular Ca(2+). Finally, while TRPV1 and TRPV4 co-localize in some RGC bodies and form a protein complex in the retina, expression of their mRNA is inversely related with increasing ocular pressure. We propose that TRPV1 activation by pressure-related insult in the eye initiates changes in expression that contribute to a Ca(2+)-dependent adaptive response to maintain excitatory signaling in RGCs.
Collapse
Affiliation(s)
- Rebecca M Sappington
- a The Vanderbilt Eye Institute and Vanderbilt Brain Institute ; Vanderbilt University School of Medicine ; Nashville , TN USA
| | | | | | | | | | | |
Collapse
|
44
|
Cooper ML, Crish SD, Inman DM, Horner PJ, Calkins DJ. Early astrocyte redistribution in the optic nerve precedes axonopathy in the DBA/2J mouse model of glaucoma. Exp Eye Res 2015; 150:22-33. [PMID: 26646560 DOI: 10.1016/j.exer.2015.11.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/12/2015] [Accepted: 11/23/2015] [Indexed: 10/22/2022]
Abstract
Glaucoma challenges the survival of retinal ganglion cell axons in the optic nerve through processes dependent on both aging and ocular pressure. Relevant stressors likely include complex interplay between axons and astrocytes, both in the retina and optic nerve. In the DBA/2J mouse model of pigmentary glaucoma, early progression involves axonopathy characterized by loss of functional transport prior to outright degeneration. Here we describe novel features of early pathogenesis in the DBA/2J nerve. With age the cross-sectional area of the nerve increases; this is associated generally with diminished axon packing density and survival and increased glial coverage of the nerve. However, for nerves with the highest axon density, as the nerve expands mean cross-sectional axon area enlarges as well. This early expansion was marked by disorganized axoplasm and accumulation of hyperphosphorylated neurofilamants indicative of axonopathy. Axon expansion occurs without loss up to a critical threshold for size (about 0.45-0.50 μm(2)), above which additional expansion tightly correlates with frank loss of axons. As well, early axon expansion prior to degeneration is concurrent with decreased astrocyte ramification with redistribution of processes towards the nerve edge. As axons expand beyond the critical threshold for loss, glial area resumes an even distribution from the center to edge of the nerve. We also found that early axon expansion is accompanied by reduced numbers of mitochondria per unit area in the nerve. Finally, our data indicate that both IOP and nerve expansion are associated with axon enlargement and reduced axon density for aged nerves. Collectively, our data support the hypothesis that diminished bioenergetic resources in conjunction with early nerve and glial remodeling could be a primary inducer of progression of axon pathology in glaucoma.
Collapse
Affiliation(s)
- Melissa L Cooper
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37205, United States
| | - Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Philip J Horner
- Biotherapeutics & Regenerative Medicine Research Center, Houston Methodist, Houston, TX 77030, United States
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37205, United States.
| |
Collapse
|
45
|
Dodson KH, Echevarria FD, Li D, Sappington RM, Edd JF. Retina-on-a-chip: a microfluidic platform for point access signaling studies. Biomed Microdevices 2015; 17:114. [PMID: 26559199 PMCID: PMC4707151 DOI: 10.1007/s10544-015-0019-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We report on a microfluidic platform for culture of whole organs or tissue slices with the capability of point access reagent delivery to probe the transport of signaling events. Whole mice retina were maintained for multiple days with negative pressure applied to tightly but gently bind the bottom of the retina to a thin poly-(dimethylsiloxane) membrane, through which twelve 100 μm diameter through-holes served as fluidic access points. Staining with toluidine blue, transport of locally applied cholera toxin beta, and transient response to lipopolysaccharide in the retina demonstrated the capability of the microfluidic platform. The point access fluidic delivery capability could enable new assays in the study of various kinds of excised tissues, including retina.
Collapse
Affiliation(s)
- Kirsten H Dodson
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Franklin D Echevarria
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Deyu Li
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA.
| | - Rebecca M Sappington
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Jon F Edd
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA.
- Cancer Center and BioMEMS Resource Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
46
|
Dekeyster E, Geeraerts E, Buyens T, Van den Haute C, Baekelandt V, De Groef L, Salinas-Navarro M, Moons L. Tackling Glaucoma from within the Brain: An Unfortunate Interplay of BDNF and TrkB. PLoS One 2015; 10:e0142067. [PMID: 26560713 PMCID: PMC4641732 DOI: 10.1371/journal.pone.0142067] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/17/2015] [Indexed: 11/18/2022] Open
Abstract
According to the neurotrophin deprivation hypothesis, diminished retrograde delivery of neurotrophic support during an early stage of glaucoma pathogenesis is one of the main triggers that induce retinal ganglion cell (RGC) degeneration. Therefore, interfering with neurotrophic signaling seems an attractive strategy to achieve neuroprotection. Indeed, exogenous neurotrophin administration to the eye has been shown to reduce loss of RGCs in animal models of glaucoma; however, the neuroprotective effect was mostly insufficient for sustained RGC survival. We hypothesized that treatment at the level of neurotrophin-releasing brain areas might be beneficial, as signaling pathways activated by target-derived neurotrophins are suggested to differ from pathways that are initiated at the soma membrane. In our study, first, the spatiotemporal course of RGC degeneration was characterized in mice subjected to optic nerve crush (ONC) or laser induced ocular hypertension (OHT). Subsequently, the well-known neurotrophin brain-derived neurotrophic factor (BDNF) was chosen as the lead molecule, and the levels of BDNF and its high-affinity receptor, tropomyosin receptor kinase B (TrkB), were examined in the mouse retina and superior colliculus (SC) upon ONC and OHT. Both models differentially influenced BDNF and TrkB levels. Next, we aimed for RGC protection through viral vector-mediated upregulation of collicular BDNF, thought to boost the retrograde neurotrophin delivery. Although the previously reported temporary neuroprotective effect of intravitreally delivered recombinant BDNF was confirmed, viral vector-induced BDNF overexpression in the SC did not result in protection of the RGCs in the glaucoma models used. These findings most likely relate to decreased neurotrophin responsiveness upon vector-mediated BDNF overexpression. Our results highlight important insights concerning the complexity of neurotrophic factor treatments that should surely be considered in future neuroprotective strategies.
Collapse
Affiliation(s)
- Eline Dekeyster
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Emiel Geeraerts
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tom Buyens
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Chris Van den Haute
- Neurobiology and Gene Therapy Research Group, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Neurobiology and Gene Therapy Research Group, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Manuel Salinas-Navarro
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
47
|
Bond WS, Hines-Beard J, GoldenMerry YPL, Davis M, Farooque A, Sappington RM, Calkins DJ, Rex TS. Virus-mediated EpoR76E Therapy Slows Optic Nerve Axonopathy in Experimental Glaucoma. Mol Ther 2015; 24:230-239. [PMID: 26502777 DOI: 10.1038/mt.2015.198] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/13/2015] [Indexed: 12/19/2022] Open
Abstract
Glaucoma, a common cause of blindness, is currently treated by intraocular pressure (IOP)-lowering interventions. However, this approach is insufficient to completely prevent vision loss. Here, we evaluate an IOP-independent gene therapy strategy using a modified erythropoietin, EPO-R76E, which has reduced erythropoietic function. We used two models of glaucoma, the murine microbead occlusion model and the DBA/2J mouse. Systemic recombinant adeno-associated virus-mediated gene delivery of EpoR76E (rAAV.EpoR76E) was performed concurrent with elevation of IOP. Axon structure and active anterograde transport were preserved in both models. Vision, as determined by the flash visual evoked potential, was preserved in the DBA/2J. These results show that systemic EpoR76E gene therapy protects retinal ganglion cells from glaucomatous degeneration in two different models. This suggests that EPO targets a component of the neurodegenerative pathway that is common to both models. The efficacy of rAAV.EpoR76E delivered at onset of IOP elevation supports clinical relevance of this treatment.
Collapse
Affiliation(s)
- Wesley S Bond
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jessica Hines-Beard
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - YPaul L GoldenMerry
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mara Davis
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alma Farooque
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rebecca M Sappington
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tonia S Rex
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
48
|
Wilson GN, Inman DM, Dengler Crish CM, Denger-Crish CM, Smith MA, Crish SD. Early pro-inflammatory cytokine elevations in the DBA/2J mouse model of glaucoma. J Neuroinflammation 2015; 12:176. [PMID: 26376776 PMCID: PMC4574349 DOI: 10.1186/s12974-015-0399-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 09/11/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation-astrogliosis, microglial activation, and changes in cytokine signaling-is a prominent feature of neurodegenerative disorders. Glaucoma is a group of chronic neurodegenerative conditions that make up the leading cause of irreversible blindness worldwide. Neuroinflammation has been postulated to play a significant role in the pathogenesis and progression of glaucomatous neurodegeneration. Though much is known regarding inflammation in the eye in glaucoma, little is known about cytokine activity outside of the retina where pathologies develop early. METHODS We traced the primary visual projection from the eye to the superior colliculus (SC) in DBA/2J and DBA/2J.Gpnmb (+) (control) mice using the anterograde tracer cholera toxin-B (CTB) to assay axonal transport deficits. Forty-eight hours later, visual structures were microdissected from fresh tissue based on transport outcome. Using magnetic bead multiplexing assays, we measured levels of 20 cytokines in the retina, proximal and distal optic nerves, CTB-positive and negative SC subdivisions, cerebellum, and serum at different ages representing different stages of pathology. RESULTS Pro- and anti-inflammatory cytokine levels in mice often changed in the same direction based on strain, age, and tissue. Significant elevations in retinal pro-inflammatory cytokines were observed in young DBA/2J mice compared to controls, followed by an age-dependent decrease in the DBA/2J mice. Proximal optic nerve of young DBA/2J mice showed a 50 % or greater decrease in levels of certain cytokines compared to older DBA/2J cohorts and controls, while both proximal and distal optic nerve of DBA/2Js showed elevations in IL-1β at all ages compared to controls. Pro-inflammatory cytokine IL-6 levels varied in accordance with transport outcome in the SC: IL-6 was elevated 44-80 % in glaucomatous DBA/2J collicular regions deficient in anterograde transport from retinal ganglion cells (RGCs) compared to areas with intact transport. CONCLUSION Dysregulation of cytokine signaling in the RGC projection of DBA/2J mice was evident early in distal retinal targets, well before intraocular pressure elevation or axonal degeneration begins.
Collapse
Affiliation(s)
- Gina N Wilson
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
- Biomedical Sciences Graduate Program, Kent State University, 800 E. Summit Street, Kent, OH, 44240, USA.
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| | - Christine M Dengler Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| | | | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
- Integrated Pharmaceutical Medicine Graduate Program, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| | - Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| |
Collapse
|
49
|
Abstract
Glaucoma is increasingly recognized as a neurodegenerative disorder, characterized by the accelerated loss of retinal ganglion cells (RGCs) and their axons. Impaired axonal transport has been implicated as a pathogenic mechanism in a number of neurodegenerative diseases, including glaucoma. The long RGC axon, with its high metabolic demand and crucial role in conveying neurotrophic signals, relies heavily on intact axonal transport. In this mini review, we consider the evidence for transport disruption along RGCs in association with glaucoma and other intraocular pressure models. We give a brief overview of the axonal transport process and the methods by which it is assessed. Spatial and temporal patterns of axonal transport disruption are considered as well as the reversibility of these changes. Biomechanical, metabolic and cytoskeletal insults may underlie the development of axonal transport deficits, and there are multiple perspectives on the impact that transport disruption has on the RGC. Eliciting the role of impaired axonal transport in glaucoma pathogenesis may uncover novel therapeutic targets for protecting the optic nerve and preventing vision loss in glaucoma.
Collapse
Affiliation(s)
- Eamonn T Fahy
- a Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne , Melbourne , Victoria , Australia
| | - Vicki Chrysostomou
- a Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne , Melbourne , Victoria , Australia
| | - Jonathan G Crowston
- a Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne , Melbourne , Victoria , Australia
| |
Collapse
|
50
|
Vecino E, Rodriguez FD, Ruzafa N, Pereiro X, Sharma SC. Glia-neuron interactions in the mammalian retina. Prog Retin Eye Res 2015; 51:1-40. [PMID: 26113209 DOI: 10.1016/j.preteyeres.2015.06.003] [Citation(s) in RCA: 560] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/18/2015] [Accepted: 06/02/2015] [Indexed: 02/07/2023]
Abstract
The mammalian retina provides an excellent opportunity to study glia-neuron interactions and the interactions of glia with blood vessels. Three main types of glial cells are found in the mammalian retina that serve to maintain retinal homeostasis: astrocytes, Müller cells and resident microglia. Müller cells, astrocytes and microglia not only provide structural support but they are also involved in metabolism, the phagocytosis of neuronal debris, the release of certain transmitters and trophic factors and K(+) uptake. Astrocytes are mostly located in the nerve fibre layer and they accompany the blood vessels in the inner nuclear layer. Indeed, like Müller cells, astrocytic processes cover the blood vessels forming the retinal blood barrier and they fulfil a significant role in ion homeostasis. Among other activities, microglia can be stimulated to fulfil a macrophage function, as well as to interact with other glial cells and neurons by secreting growth factors. This review summarizes the main functional relationships between retinal glial cells and neurons, presenting a general picture of the retina recently modified based on experimental observations. The preferential involvement of the distinct glia cells in terms of the activity in the retina is discussed, for example, while Müller cells may serve as progenitors of retinal neurons, astrocytes and microglia are responsible for synaptic pruning. Since different types of glia participate together in certain activities in the retina, it is imperative to explore the order of redundancy and to explore the heterogeneity among these cells. Recent studies revealed the association of glia cell heterogeneity with specific functions. Finally, the neuroprotective effects of glia on photoreceptors and ganglion cells under normal and adverse conditions will also be explored.
Collapse
Affiliation(s)
- Elena Vecino
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - F David Rodriguez
- Department of Biochemistry and Molecular Biology, E-37007, University of Salamanca, Salamanca, Spain
| | - Noelia Ruzafa
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Xandra Pereiro
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Sansar C Sharma
- Department of Ophthalmology, Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA; IKERBASQUE, Basque Foundation for Science at Dept. Cell Biology and Histology, UPV/EHU, Spain
| |
Collapse
|