1
|
Traumatic optic neuropathy: a review of current studies. Neurosurg Rev 2022; 45:1895-1913. [PMID: 35034261 DOI: 10.1007/s10143-021-01717-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/26/2021] [Accepted: 12/09/2021] [Indexed: 10/24/2022]
Abstract
Traumatic optic neuropathy (TON) is a serious complication of craniofacial trauma that directly or indirectly damages the optic nerve and can cause severe vision loss. The incidence of TON has been gradually increasing in recent years. Research on the protection and regeneration of the optic nerve after the onset of TON is still at the level of laboratory studies and which is insufficient to support clinical treatment of TON. And, due to without clear guidelines, there is much ambiguity regarding its diagnosis and management. Clinical interventions for TON include observation only, treatment with corticosteroids alone, or optic canal (OC) decompression (with or without steroids). There is controversy in clinical practice concerning which treatment is the best. A review of available studies shows that the visual acuity of patients with TON can be significantly improved after OC decompression surgery (especially endoscopic transnasal/transseptal optic canal decompression (ETOCD)) with or without the use of corticosteroids. And new findings of laboratory studies such as mitochondrial therapy, lipid change studies, and other studies in favor of TON therapy have also been identified. In this review, we discuss the evolving perspective of surgical treatment and experimental study.
Collapse
|
2
|
Feizi S, Alemzadeh-Ansari M, Karimian F, Esfandiari H. Use of erythropoietin in ophthalmology: a review. Surv Ophthalmol 2021; 67:427-439. [PMID: 34157346 DOI: 10.1016/j.survophthal.2021.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Erythropoietin (EPO) is a glycoprotein hormone that regulates hematopoiesis in the human body. The presence of EPO and its receptors in different tissues indicates that this hormone has extramedullary effects in other tissues, including the eye. We focus on the biological roles of this hormone in the development and normal physiologic functions of the eye. Furthermore, we explore the role of EPO in the management of different ocular diseases - including diabetic retinopathy, retinopathy of prematurity, inherited retinal degeneration, branch and central retinal vein occlusion, retinal detachment, traumatic optic neuropathy, optic neuritis, methanol optic neuropathy, nonarteritic anterior ischemic optic neuropathy, glaucoma, and scleral necrosis.
Collapse
Affiliation(s)
- Sepehr Feizi
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Farid Karimian
- Ophthalmic Research Center, Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Esfandiari
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
3
|
Wang J, Struebing FL, Geisert EE. Commonalities of optic nerve injury and glaucoma-induced neurodegeneration: Insights from transcriptome-wide studies. Exp Eye Res 2021; 207:108571. [PMID: 33844961 PMCID: PMC9890784 DOI: 10.1016/j.exer.2021.108571] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 02/03/2023]
Abstract
Glaucoma is a collection of diseases that lead to an irreversible vision loss due to damage of retinal ganglion cells (RGCs). Although the underlying events leading to RGC death are not fully understood, recent research efforts are beginning to define the genetic changes that play a critical role in the initiation and progression of glaucomatous injury and RGC death. Several genetic and experimental animal models have been developed to mimic glaucomatous neurodegeneration. These models differ in many respects but all result in the loss of RGCs. Assessing transcriptional changes across different models could provide a more complete perspective on the molecular drivers of RGC degeneration. For the past several decades, changes in the retinal transcriptome during neurodegeneration process were defined using microarray methods, RNA sequencing and now single cell RNA sequencing. It is understood that these methods have strengths and weaknesses due to technical differences and variations in the analytical tools used. In this review, we focus on the use of transcriptome-wide expression profiling of the changes occurring as RGCs are lost across different glaucoma models. Commonalities of optic nerve crush and glaucoma-induced neurodegeneration are identified and discussed.
Collapse
Affiliation(s)
- Jiaxing Wang
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Felix L. Struebing
- Center for Neuropathology and Prion Research, Ludwig Maximilian University of Munich, Germany,Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Eldon E. Geisert
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA,Corresponding author: (E.E. Geisert)
| |
Collapse
|
4
|
Erythropoietin Gene Therapy Delays Retinal Degeneration Resulting from Oxidative Stress in the Retinal Pigment Epithelium. Antioxidants (Basel) 2021; 10:antiox10060842. [PMID: 34070383 PMCID: PMC8229633 DOI: 10.3390/antiox10060842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023] Open
Abstract
Erythropoietin (EPO) plays an important role in erythropoiesis by its action in blocking apoptosis of progenitor cells and protects both photoreceptors and retinal ganglion cells from induced or inherited degeneration. A modified form of EPO, EPO-R76E has attenuated erythropoietic activity but is effective in inhibiting apoptosis, oxidative stress, and inflammation in several models of retinal degeneration. In this study, we used recombinant Adeno Associated Virus (AAV) to provide long-term sustained delivery of EPO-R76E and demonstrated its effects in a mouse model of dry-AMD in which retinal degeneration is induced by oxidative stress in the retinal pigment epithelial (RPE) cells. Experimental vector AAV-EPO-R76E and control vector AAV-GFP were packaged into serotype-1 (AAV1) to enable RPE selective expression. RPE oxidative stress-mediated retinal degeneration was induced by exon specific deletion of the protective enzyme MnSOD (encoded by Sod2) by cre/lox mechanism. Experimental mice received subretinal injection of AAV-EPO-R76E in the right eye and AAV-GFP in the left eye. Western blotting of RPE/choroid protein samples from AAV-EPO-R76E injected eyes showed RPE specific EPO expression. Retinal function was monitored by electroretinography (ERG). EPO-R76E over-expression in RPE delayed the retinal degeneration as measured by light microscopy in RPE specific Sod2 knockout mice. Delivery of EPO-R76E vector can be used as a tool to prevent retinal degeneration induced by RPE oxidative stress, which is implicated as a potential cause of Age-Related Macular Degeneration.
Collapse
|
5
|
Lee D, Kang H, Yoon KY, Chang YY, Song HB. A mouse model of retinal hypoperfusion injury induced by unilateral common carotid artery occlusion. Exp Eye Res 2020; 201:108275. [PMID: 32991884 DOI: 10.1016/j.exer.2020.108275] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Retina, one of the highest oxygen demanding tissues, is vulnerable to vascular insufficiencies, and various ocular vascular disorders can cause chronic retinal ischemia. To investigate the pathophysiology, rodent models developed by bilateral common carotid artery occlusion (BCCAO) have been utilized. However, mice lack posterior communicating arteries in the circle of Willis and cannot endure the brain ischemia induced by the bilateral occlusion. A mouse model to better reflect the localized ischemic stress in the retina without affecting the brain is still needed. Here, we established a mouse model of ischemic injury by permanent unilateral common carotid artery occlusion (UCCAO). Adult male mice were subjected to UCCAO, and changes in the ipsilateral retina were examined in comparison with the contralateral retina. Delayed perfusion was observed in the ipsilateral retina right after the occlusion and was not recovered later on. Common features of retinal ischemia were observed: hypoxia-inducible factor (HIF) stabilization; upregulation of hypoxia-responsive genes; altered levels of cytokines and chemokines. Activation of astrocytes and Müller cells in the inner retina was detected at day 2, and thinning of the inner retinal layer became significant at week 10. Together, our model can simulate retinal ischemia with morphological and molecular changes. It can be utilized to investigate pathophysiology of ischemic retinopathies.
Collapse
Affiliation(s)
- Deokho Lee
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heekyoung Kang
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Young Yoon
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yuan Yi Chang
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Beom Song
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea; Department of Ophthalmology, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Simó R, Stitt AW, Gardner TW. Neurodegeneration in diabetic retinopathy: does it really matter? Diabetologia 2018; 61:1902-1912. [PMID: 30030554 PMCID: PMC6096638 DOI: 10.1007/s00125-018-4692-1] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
The concept of diabetic retinopathy as a microvascular disease has evolved, in that it is now considered a more complex diabetic complication in which neurodegeneration plays a significant role. In this article we provide a critical overview of the role of microvascular abnormalities and neurodegeneration in the pathogenesis of diabetic retinopathy. A special emphasis is placed on the pathophysiology of the neurovascular unit (NVU), including the contributions of microvascular and neural elements. The potential mechanisms linking retinal neurodegeneration and early microvascular impairment, and the effects of neuroprotective drugs are summarised. Additionally, we discuss how the assessment of retinal neurodegeneration could be an important index of cognitive status, thus helping to identify individuals at risk of dementia, which will impact on current procedures for diabetes management. We conclude that glial, neural and microvascular dysfunction are interdependent and essential for the development of diabetic retinopathy. Despite this intricate relationship, retinal neurodegeneration is a critical endpoint and neuroprotection, itself, can be considered a therapeutic target, independently of its potential impact on microvascular disease. In addition, interventional studies targeting pathogenic pathways that impact the NVU are needed. Findings from these studies will be crucial, not only for increasing our understanding of diabetic retinopathy, but also to help to implement a timely and efficient personalised medicine approach for treating this diabetic complication.
Collapse
Affiliation(s)
- Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - Alan W Stitt
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, UK
| | - Thomas W Gardner
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Erythropoietin either Prevents or Exacerbates Retinal Damage from Eye Trauma Depending on Treatment Timing. Optom Vis Sci 2017; 94:20-32. [PMID: 27281679 DOI: 10.1097/opx.0000000000000898] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Erythropoietin (EPO) is a promising neuroprotective agent and is currently in Phase III clinical trials for the treatment of traumatic brain injury. The goal of this study was to determine if EPO is also protective in traumatic eye injury. METHODS The left eyes of anesthetized DBA/2J or Balb/c mice were exposed to a single 26 psi overpressure air-wave while the rest of the body was shielded. DBA/2J mice were given intraperitoneal injections of EPO or buffer and analyses were performed at 3 or 7 days post-blast. Balb/c mice were given intramuscular injections of rAAV.EpoR76E or rAAV.eGFP either pre- or post-blast and analyses were performed at 1 month post-blast. RESULTS EPO had a bimodal effect on cell death, glial reactivity, and oxidative stress. All measures were increased at 3 days post-blast and decreased at 7-days post-blast. Increased retinal ferritin and NADPH oxygenases were detected in retinas from EPO-treated mice. The gene therapy approach protected against axon degeneration, cell death, and oxidative stress when given after blast, but not before. CONCLUSIONS Systemic, exogenous EPO and EPO-R76E protects the retina after trauma even when initiation of treatment is delayed by up to 3 weeks. Systemic treatment with EPO or EPO-R76E beginning before or soon after trauma may exacerbate protective effects of EPO within the retina as a result of increased iron levels from erythropoiesis and, thus, increased oxidative stress within the retina. This is likely overcome with time as a result of an increase in levels of antioxidant enzymes. Either intraocular delivery of EPO or treatment with non-erythropoietic forms of EPO may be more efficacious.
Collapse
|
8
|
Agarwal R, Agarwal P. Rodent models of glaucoma and their applicability for drug discovery. Expert Opin Drug Discov 2017; 12:261-270. [DOI: 10.1080/17460441.2017.1281244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Renu Agarwal
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Puneet Agarwal
- Faculty of Medicine, International Medical University, IMU Clinical Campus, Seremban, Malaysia
| |
Collapse
|
9
|
Rex TS, Kasmala L, Bond WS, de Lucas Cerrillo AM, Wynn K, Lewin AS. Erythropoietin Slows Photoreceptor Cell Death in a Mouse Model of Autosomal Dominant Retinitis Pigmentosa. PLoS One 2016; 11:e0157411. [PMID: 27299810 PMCID: PMC4907422 DOI: 10.1371/journal.pone.0157411] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/27/2016] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To test the efficacy of systemic gene delivery of a mutant form of erythropoietin (EPO-R76E) that has attenuated erythropoietic activity, in a mouse model of autosomal dominant retinitis pigmentosa. METHODS Ten-day old mice carrying one copy of human rhodopsin with the P23H mutation and both copies of wild-type mouse rhodopsin (hP23H RHO+/-,mRHO+/+) were injected into the quadriceps with recombinant adeno-associated virus (rAAV) carrying either enhanced green fluorescent protein (eGFP) or EpoR76E. Visual function (electroretinogram) and retina structure (optical coherence tomography, histology, and immunohistochemistry) were assessed at 7 and 12 months of age. RESULTS The outer nuclear layer thickness decreased over time at a slower rate in rAAV.EpoR76E treated as compared to the rAAV.eGFP injected mice. There was a statistically significant preservation of the electroretinogram at 7, but not 12 months of age. CONCLUSIONS Systemic EPO-R76E slows death of the photoreceptors and vision loss in hP23H RHO+/-,mRHO+/+ mice. Treatment with EPO-R76E may widen the therapeutic window for retinal degeneration patients by increasing the number of viable cells. Future studies might investigate if co-treatment with EPO-R76E and gene replacement therapy is more effective than gene replacement therapy alone.
Collapse
Affiliation(s)
- Tonia S. Rex
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States of America
| | - Lorraine Kasmala
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States of America
| | - Wesley S. Bond
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States of America
| | - Ana M. de Lucas Cerrillo
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States of America
| | - Kristi Wynn
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States of America
| | - Alfred S. Lewin
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL 32608, United States of America
| |
Collapse
|
10
|
Russo R, Varano GP, Adornetto A, Nucci C, Corasaniti MT, Bagetta G, Morrone LA. Retinal ganglion cell death in glaucoma: Exploring the role of neuroinflammation. Eur J Pharmacol 2016; 787:134-42. [PMID: 27044433 DOI: 10.1016/j.ejphar.2016.03.064] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/10/2016] [Accepted: 03/31/2016] [Indexed: 01/06/2023]
Abstract
In clinical glaucoma, as well as in experimental models, the loss of retinal ganglion cells occurs by apoptosis. This final event is preceded by inflammatory responses involving the activation of innate and adaptive immunity, with retinal and optic nerve resident glial cells acting as major players. Here we review the current literature on the role of neuroinflammation in neurodegeneration, focusing on the inflammatory molecular mechanisms involved in the pathogenesis and progression of the optic neuropathy.
Collapse
Affiliation(s)
- Rossella Russo
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy.
| | - Giuseppe Pasquale Varano
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Annagrazia Adornetto
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome Italy
| | | | - Giacinto Bagetta
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| | - Luigi Antonio Morrone
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
11
|
Virus-mediated EpoR76E gene therapy preserves vision in a glaucoma model by modulating neuroinflammation and decreasing oxidative stress. J Neuroinflammation 2016; 13:39. [PMID: 26876380 PMCID: PMC4753658 DOI: 10.1186/s12974-016-0499-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 02/01/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glaucoma is a complex neurodegeneration and a leading cause of blindness worldwide. Current therapeutic strategies, which are all directed towards lowering the intraocular pressure (IOP), do not stop progression of the disease. We have demonstrated that recombinant adeno-associated virus (rAAV) gene delivery of a form of erythropoietin with attenuated erythropoietic activity (EpoR76E) can preserve retinal ganglion cells, their axons, and vision without decreasing IOP. The goal of this study was to determine if modulation of neuroinflammation or oxidative stress played a role in the neuroprotective activity of EPO.R76E. METHODS Five-month-old DBA/2J mice were treated with either rAAV.EpoR76E or a control vector and collected at 8 months of age. Neuroprotection was assessed by quantification of axon transport and visual evoked potentials. Microglia number and morphology and cytokine and chemokine levels were quantified. Message levels of oxidative stress-related proteins were assessed. RESULTS Axon transport and visual evoked potentials were preserved in rAAV.EpoR76E-treated mice. The number of microglia was decreased in retinas from 8-month-old rAAV.EpoR76E-treated mice, but proliferation was unaffected. The blood-retina barrier was also unaffected by treatment. Levels of some pro-inflammatory cytokines were decreased in retinas from rAAV.EpoR76E-treated mice including IL-1, IL-12, IL-13, IL-17, CCL4, and CCL5. TNFα messenger RNA (mRNA) was increased in retinas from 8-month-old mice compared to 3-month-old controls regardless of treatment. Expression of several antioxidant proteins was increased in retinas of rAAV.EpoR76E-treated 8-month-old mice. CONCLUSIONS Treatment with rAAV.EpoR76E preserves vision in the DBA/2J model of glaucoma at least in part by decreasing infiltration of peripheral immune cells, modulating microglial reactivity, and decreasing oxidative stress.
Collapse
|
12
|
Singman EL, Daphalapurkar N, White H, Nguyen TD, Panghat L, Chang J, McCulley T. Indirect traumatic optic neuropathy. Mil Med Res 2016; 3:2. [PMID: 26759722 PMCID: PMC4709956 DOI: 10.1186/s40779-016-0069-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023] Open
Abstract
Indirect traumatic optic neuropathy (ITON) refers to optic nerve injury resulting from impact remote to the optic nerve. The mechanism of injury is not understood, and there are no confirmed protocols for prevention, mitigation or treatment. Most data concerning this condition comes from case series of civilian patients suffering blunt injury, such as from sports- or motor vehicle-related concussion, rather than military-related ballistic or blast damage. Research in this field will likely require the development of robust databases to identify patients with ITON and follow related outcomes, in addition to both in-vivo animal and virtual human models to study the mechanisms of damage and potential therapies.
Collapse
Affiliation(s)
- Eric L Singman
- Wilmer Eye Institute at Johns Hopkins Hospital, Baltimore, Maryland USA
| | | | - Helen White
- Director of Informatics and Information Management, Vision Center of Excellence [VCE], Office of the Assistant Secretary of Defense for Health Affairs [ASD-HA], United States Department of Defense [DOD], Walter Reed National Military Medical Center, Bethesda, MD USA
| | - Thao D Nguyen
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD USA
| | - Lijo Panghat
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD USA
| | - Jessica Chang
- Wilmer Eye Institute at Johns Hopkins Hospital, Baltimore, Maryland USA
| | - Timothy McCulley
- Wilmer Eye Institute at Johns Hopkins Hospital, Baltimore, Maryland USA
| |
Collapse
|
13
|
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: 3.0] [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
|
14
|
de Lucas Cerrillo A, Bond WS, Rex TS. Safety and angiogenic effects of systemic gene delivery of a modified erythropoietin. Gene Ther 2015; 22:365-73. [PMID: 25716531 PMCID: PMC4425572 DOI: 10.1038/gt.2015.12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/18/2014] [Accepted: 01/15/2015] [Indexed: 01/10/2023]
Abstract
Erythropoietin (EPO) is critical for red blood cell production and is also an effective neuroprotective agent. However, it may contribute to pathological angiogenesis. Here we investigate the angiogenic potential of EPO and a mutant form with attenuated erythropoietic activity, EPO-R76E, on primary human retinal microvascular endothelial cells (HRMECs) and in the adult retina. Assays of death, proliferation and tube formation were performed on HRMECs exposed to EPO, EPO-R76E or media alone. Postnatal day-9 wild-type mice were injected intramuscularly with adeno-associated virus vectors expressing either enhanced green fluorescent protein or EpoR76E. At 3 months, levels of EPO-R76E in the eye were quantified, and the health of the retinal vasculature was assessed by fluorescein angiography and isolectin immunolabeling. Immunohistochemistry, histology and electroretinogram (ERG) assessments were performed as measures of retinal health. Neither EPO nor EPO-R76E induced proliferation or tube formation in HRMECs under the conditions used. EPO-R76E decreased HRMEC death in a dose-dependent manner. Long-term systemic gene delivery of EPO-R76E was safe in terms of retinal vasculature, histology and the ERG in vivo. Our results show that EPO-R76E can block HRMEC death, consistent with its role in erythropoiesis and neuroprotection. In addition, long-term gene delivery of EPO-R76E is safe in the adult retina.
Collapse
Affiliation(s)
- Ana de Lucas Cerrillo
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Wesley S. Bond
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Tonia S. Rex
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| |
Collapse
|
15
|
Luo W, Hu L, Wang F. The Protective Effect of Erythropoietin on the Retina. Ophthalmic Res 2015; 53:74-81. [DOI: 10.1159/000369885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/11/2014] [Indexed: 11/19/2022]
|
16
|
Bond WS, Rex TS. Evidence That Erythropoietin Modulates Neuroinflammation through Differential Action on Neurons, Astrocytes, and Microglia. Front Immunol 2014; 5:523. [PMID: 25374571 PMCID: PMC4205853 DOI: 10.3389/fimmu.2014.00523] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/06/2014] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a normal and healthy response to neuronal damage. However, excessive or chronic neuroinflammation exacerbates neurodegeneration after trauma and in progressive diseases such as Alzheimer’s, Parkinson’s, age-related macular degeneration, and glaucoma. Therefore, molecules that modulate neuroinflammation are candidates as neuroprotective agents. Erythropoietin (EPO) is a known neuroprotective agent that indirectly attenuates neuroinflammation, in part, by inhibiting neuronal apoptosis. In this review, we provide evidence that EPO also modulates neuroinflammation upstream of apoptosis by acting directly on glia. Further, the signaling induced by EPO may differ depending on cell type and context possibly as a result of activation of different receptors. While significant progress has been made in our understanding of EPO signaling, this review also identifies areas for future study in terms of the role of EPO in modulating neuroinflammation.
Collapse
Affiliation(s)
- Wesley S Bond
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| | - Tonia S Rex
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| |
Collapse
|
17
|
Cooke Bailey JN, Yaspan BL, Pasquale LR, Hauser MA, Kang JH, Loomis SJ, Brilliant M, Budenz DL, Christen WG, Fingert J, Gaasterland D, Gaasterland T, Kraft P, Lee RK, Lichter PR, Liu Y, McCarty CA, Moroi SE, Richards JE, Realini T, Schuman JS, Scott WK, Singh K, Sit AJ, Vollrath D, Wollstein G, Zack DJ, Zhang K, Pericak-Vance MA, Allingham RR, Weinreb RN, Haines JL, Wiggs JL. Hypothesis-independent pathway analysis implicates GABA and acetyl-CoA metabolism in primary open-angle glaucoma and normal-pressure glaucoma. Hum Genet 2014; 133:1319-30. [PMID: 25037249 PMCID: PMC4273559 DOI: 10.1007/s00439-014-1468-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 07/08/2014] [Indexed: 12/15/2022]
Abstract
Primary open-angle glaucoma (POAG) is a leading cause of blindness worldwide. Using genome-wide association single-nucleotide polymorphism data from the Glaucoma Genes and Environment study and National Eye Institute Glaucoma Human Genetics Collaboration comprising 3,108 cases and 3,430 controls, we assessed biologic pathways as annotated in the KEGG database for association with risk of POAG. After correction for genic overlap among pathways, we found 4 pathways, butanoate metabolism (hsa00650), hematopoietic cell lineage (hsa04640), lysine degradation (hsa00310) and basal transcription factors (hsa03022) related to POAG with permuted p < 0.001. In addition, the human leukocyte antigen (HLA) gene family was significantly associated with POAG (p < 0.001). In the POAG subset with normal-pressure glaucoma (NPG), the butanoate metabolism pathway was also significantly associated (p < 0.001) as well as the MAPK and Hedgehog signaling pathways (hsa04010 and hsa04340), glycosaminoglycan biosynthesis-heparan sulfate pathway (hsa00534) and the phenylalanine, tyrosine and tryptophan biosynthesis pathway (hsa0400). The butanoate metabolism pathway overall, and specifically the aspects of the pathway that contribute to GABA and acetyl-CoA metabolism, was the only pathway significantly associated with both POAG and NPG. Collectively these results implicate GABA and acetyl-CoA metabolism in glaucoma pathogenesis, and suggest new potential therapeutic targets.
Collapse
Affiliation(s)
| | - Brian L. Yaspan
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Louis R. Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A. Hauser
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jae H. Kang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie J. Loomis
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Murray Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Donald L. Budenz
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, USA
| | | | - John Fingert
- Department of Ophthalmology, College of Medicine, University of Iowa, Iowa City, IO, USA
- Department of A natomy/Cell Biology, College of Medicine, University of Iowa, Iowa City, IO, USA
| | | | - Terry Gaasterland
- Scripps Genome Center, University of California at San Diego, San Diego, CA, USA
| | - Peter Kraft
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Richard K. Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paul R. Lichter
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yutao Liu
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Sayoko E. Moroi
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Julia E. Richards
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Tony Realini
- Department of Ophthalmology, WVU Eye Institute, Morgantown, WV, USA
| | - Joel S. Schuman
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - William K. Scott
- Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kuldev Singh
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
| | - Arthur J. Sit
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | - Gadi Wollstein
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald J. Zack
- Wilmer Eye Institute, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Kang Zhang
- Department of Ophthalmology, Hamilton Eye Center, University of California, San Diego, CA, USA
| | | | - R. Rand Allingham
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
| | - Robert N. Weinreb
- Department of Ophthalmology, Hamilton Eye Center, University of California, San Diego, CA, USA
| | - Jonathan L. Haines
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| |
Collapse
|
18
|
Li HY, Ruan YW, Ren CR, Cui Q, So KF. Mechanisms of secondary degeneration after partial optic nerve transection. Neural Regen Res 2014; 9:565-74. [PMID: 25206855 PMCID: PMC4146235 DOI: 10.4103/1673-5374.130093] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2014] [Indexed: 01/23/2023] Open
Abstract
Secondary degeneration occurs commonly in the central nervous system after traumatic injuries and following acute and chronic diseases, including glaucoma. A constellation of mechanisms have been shown to be associated with secondary degeneration including apoptosis, necrosis, autophagy, oxidative stress, excitotoxicity, derangements in ionic homeostasis and calcium influx. Glial cells, such as microglia, astrocytes and oligodendrocytes, have also been demonstrated to take part in the process of secondary injury. Partial optic nerve transection is a useful model which was established about 13 years ago. The merit of this model compared with other optic nerve injury models used for glaucoma study, including complete optic nerve transection model and optic nerve crush model, is the possibility to separate primary degeneration from secondary degeneration in location. Therefore, it provides a good tool for the study of secondary degeneration. This review will focus on the research progress of the mechanisms of secondary degeneration using partial optic nerve transection model.
Collapse
Affiliation(s)
- Hong-Ying Li
- Department of Ophthalmology, the University of Hong Kong, Hong Kong Special Administrative Region, China ; State Key Laboratory of Brain and Cognitive Science, the University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yi-Wen Ruan
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China ; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province, China
| | - Chao-Ran Ren
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China ; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province, China
| | - Qi Cui
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China ; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Department of Ophthalmology, the University of Hong Kong, Hong Kong Special Administrative Region, China ; GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China ; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province, China ; State Key Laboratory of Brain and Cognitive Science, the University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
19
|
Bricker-Anthony C, Hines-Beard J, Rex TS. Molecular changes and vision loss in a mouse model of closed-globe blast trauma. Invest Ophthalmol Vis Sci 2014; 55:4853-62. [PMID: 24994864 PMCID: PMC4123895 DOI: 10.1167/iovs.14-14353] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/22/2014] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To characterize retinal changes and assess vision after an eye-directed air blast. METHODS Adult C57Bl/6 mice were exposed to a blast directed at one eye. Optical coherence tomography and histology were performed to assess retina and optic nerve integrity. Cell death, oxidative stress, and glial reactivity were examined by immunohistochemistry. Visual changes were measured by ERG recordings and the optokinetic reflex. RESULTS In the outer retina, eye blast caused retinal pigment epithelium vacuoles and rare retinal detachments followed by regional cell death. Labeling for nitrotyrosine and markers of pyroptosis (caspase-1) and necroptosis (receptor-interacting protein kinases-1, -3) increased, primarily in the inner retina, after blast. Caspase-1 labeling was restricted primarily to the starburst amacrine cells. A few degenerating axons were detected at 28 days post blast. Despite a lack of substantial cell death or decreased ERG, there was a deficit in visual acuity after blast. CONCLUSIONS Oxidative stress, neuroinflammation, and cell death became increasingly prevalent, over time post blast suggestive of an ongoing neurodegenerative response. Outer retinal changes either resolved or remained focal. In contrast, inner retinal changes were more robust and spread from focal regions to the entire retina over time post blast. Our model of eye blast trauma causes molecular changes and a decrease in visual acuity within the first month post blast despite a lack of overt eye injury. This subtle response matches the delayed presentation of visual deficits in some blast-exposed Veterans.
Collapse
Affiliation(s)
- Courtney Bricker-Anthony
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jessica Hines-Beard
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Tonia S Rex
- Vanderbilt Eye Institute, Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
20
|
Entezari M, Esmaeili M, Yaseri M. A pilot study of the effect of intravenous erythropoetin on improvement of visual function in patients with recent indirect traumatic optic neuropathy. Graefes Arch Clin Exp Ophthalmol 2014; 252:1309-13. [DOI: 10.1007/s00417-014-2691-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 05/29/2014] [Accepted: 06/03/2014] [Indexed: 12/19/2022] Open
|
21
|
Hines-Beard J, Desai S, Haag R, Esumi N, D'Surney L, Parker S, Richardson C, Rex TS. Identification of a therapeutic dose of continuously delivered erythropoietin in the eye using an inducible promoter system. Curr Gene Ther 2014; 13:275-81. [PMID: 23773177 DOI: 10.2174/15665232113139990024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 12/30/2022]
Abstract
Erythropoietin (EPO) can protect the retina from acute damage, but long-term systemic treatment induces polycythemia. Intraocular gene delivery of EPO is not protective despite producing high levels of EPO likely due to its bellshaped dose curve. The goal of this study was to identify a therapeutic dose of continuously produced EPO in the eye. We packaged a mutated form of EPO (EPOR76E) that has equivalent neuroprotective activity as wild-type EPO and attenuated erythropoietic activity into a recombinant adeno-associated viral vector under the control of the tetracycline inducible promoter. This vector was injected into the subretinal space of homozygous postnatal 5-7 day retinal degeneration slow mice, that express the tetracycline transactivators from a retinal pigment epithelium specific promoter. At weaning, mice received a single intraperitoneal injection of doxycycline and were then maintained on water with or without doxycycline until postnatal day 60. Intraocular EPO levels and outer nuclear layer thickness were quantified and correlated. Control eyes contained 6.1 ± 0.1 (SEM) mU/ml EPO. The eyes of mice that received an intraperitoneal injection of doxycycline contained 11.8 ± 2.0 (SEM) mU/ml EPO-R76E. Treatment with doxycycline water induced production of 35.9 ± 2.4 (SEM) mU/ml EPO-R76E in the eye. The outer nuclear layer was approximately 8 μm thicker in eyes of mice that received doxycycline water as compared to the control groups. Our data indicates that drug delivery systems should be optimized to deliver at least 36 mU/ml EPO into the eye since this dose was effective for the treatment of a progressive retinal degeneration.
Collapse
Affiliation(s)
- Jessica Hines-Beard
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, 11435 MRBIV, 2213 Garland Ave, Nashville, TN 37232-8808, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Ismailov RM. Geographic altitude, ocular diseases and injuries. MEDICAL HYPOTHESIS, DISCOVERY & INNOVATION OPHTHALMOLOGY JOURNAL 2014; 3:62-4. [PMID: 25738162 PMCID: PMC4346680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Yang X, Bi Y, Chen E, Feng D. Overexpression of Wnt3a facilitates the proliferation and neural differentiation of neural stem cells in vitro and after transplantation into an injured rat retina. J Neurosci Res 2013; 92:148-61. [PMID: 24254835 DOI: 10.1002/jnr.23314] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 09/12/2013] [Accepted: 09/20/2013] [Indexed: 01/07/2023]
Affiliation(s)
- Xi‐Tao Yang
- Department of NeurosurgeryNo. 3 People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai China
| | - Yong‐Yan Bi
- Department of NeurosurgeryNo. 3 People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai China
| | - Er‐Tao Chen
- Department of NeurosurgeryNo. 3 People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai China
| | - Dong‐Fu Feng
- Department of NeurosurgeryNo. 3 People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of MedicineShanghai China
| |
Collapse
|
24
|
|
25
|
Shi X, Yang J, Zhu H, Ye L, Feng M, Li J, Huang H, Tao Q, Ye D, Sun LHK, Sun BNC, Sun CRY, Han G, Liu Y, Yao M, Zhou P, Ju D. Pharmacokinetics and pharmacodynamics of recombinant human EPO-Fc fusion protein in vivo. PLoS One 2013; 8:e72673. [PMID: 23977338 PMCID: PMC3747110 DOI: 10.1371/journal.pone.0072673] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 07/11/2013] [Indexed: 01/01/2023] Open
Abstract
In this study, the in vivo pharmacokinetics and pharmacodynamics
of a novel recombinant human erythropoietin (rhEPO) Fc fusion protein, rhEPO-Fc,
were studied in both rodents and rhesus monkeys. Animal models of anemia induced
by irradiation, cyclophosphamide and partial renal ablation were used to
evaluate therapeutic effects of rhEPO-Fc. We have demonstrated that serum
half-life of rhEPO-Fc was 29.5 to 38.9 h at doses of 8, 25, 80 µg/kg in
rhesus monkeys and 35.5 to 43.5 h at doses of 16, 50, 160 µg/kg in rats.
In anemia animal models, rhEPO-Fc dose-dependently (7.5–30.0 µg/kg
in mice, 5.4–21.4 µg/kg in rats and 5.0–10.0 µg/kg in
rhesus monkeys) increased reticulocyte level, followed by an increase of RBC
count, hemoglobin and hematocrit levels. At reduced intervention frequency of
weekly treatments, rhEPO-Fc showed similar hematopoietic effects as compared
with rhEPO given three times a week. These results indicated that rhEPO-Fc could
potentially be used in treatment of anemia and warrants future clinical
trials.
Collapse
Affiliation(s)
- Xunlong Shi
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Jianjun Yang
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | - Haiyan Zhu
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Li Ye
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Meiqing Feng
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Jiyang Li
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Hai Huang
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Qun Tao
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | - Dan Ye
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | | | | | | | - Guizhen Han
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Yuanyuan Liu
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Minghui Yao
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Pei Zhou
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
26
|
Nitric oxide in cerebral vasospasm: theories, measurement, and treatment. Neurol Res Int 2013; 2013:972417. [PMID: 23878735 PMCID: PMC3708422 DOI: 10.1155/2013/972417] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 05/23/2013] [Accepted: 05/28/2013] [Indexed: 12/22/2022] Open
Abstract
In recent decades, a large body of research has focused on the role of nitric oxide (NO) in the development of cerebral vasospasm (CV) following subarachnoid hemorrhage (SAH). Literature searches were therefore conducted regarding the role of NO in cerebral vasospasm, specifically focusing on NO donors, reactive nitrogen species, and peroxynitrite in manifestation of vasospasm. Based off the assessment of available evidence, two competing theories are reviewed regarding the role of NO in vasospasm. One school of thought describes a deficiency in NO due to scavenging by hemoglobin in the cisternal space, leading to an NO signaling deficit and vasospastic collapse. A second hypothesis focuses on the dysfunction of nitric oxide synthase, an enzyme that synthesizes NO, and subsequent generation of reactive nitrogen species. Both theories have strong experimental evidence behind them and hold promise for translation into clinical practice. Furthermore, NO donors show definitive promise for preventing vasospasm at the angiographic and clinical level. However, NO augmentation may also cause systemic hypotension and worsen vasospasm due to oxidative distress. Recent evidence indicates that targeting NOS dysfunction, for example, through erythropoietin or statin administration, also shows promise at preventing vasospasm and neurotoxicity. Ultimately, the role of NO in neurovascular disease is complex. Neither of these theories is mutually exclusive, and both should be considered for future research directions and treatment strategies.
Collapse
|
27
|
Dhanushkodi A, Akano EO, Roguski EE, Xue Y, Rao SK, Matta SG, Rex TS, McDonald MP. A single intramuscular injection of rAAV-mediated mutant erythropoietin protects against MPTP-induced parkinsonism. GENES BRAIN AND BEHAVIOR 2012. [PMID: 23190369 DOI: 10.1111/gbb.12001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Erythropoietin (Epo) is neuroprotective in a number of preparations, but can lead to unacceptably high and even lethal hematocrit levels. Recent reports show that modified Epo variants confer neuroprotection in models of glaucoma and retinal degeneration without raising hematocrit. In this study, neuroprotective effects of two Epo variants (EpoR76E and EpoS71E) were assessed in a model of Parkinson's disease. The constructs were packaged in recombinant adeno-associated viral (rAAV) vectors and injected intramuscularly. After 3 weeks, mice received five daily injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and were killed 5 weeks later. The MPTP-lesioned mice pretreated with rAAV.eGFP (negative control) exhibited a 7- to 9-Hz tremor and slower latencies to move on a grid test (akinesia). Both of these symptomatic features were absent in mice pretreated with either modified Epo construct. The rAAV.eGFP-treated mice lesioned with MPTP exhibited a 41% reduction in tyrosine hydroxylase (TH)-positive neurons in the substantia nigra. The rAAV.EpoS71E construct did not protect nigral neurons, but neuronal loss in mice pretreated with rAAV.EpoR76E was only half that of rAAV.eGFP controls. Although dopamine levels were normal in all groups, 3,4-dihydroxyphenylacetic acid (DOPAC) was significantly reduced only in MPTP-lesioned mice pretreated with rAAV.eGFP, indicating reduced dopamine turnover. Analysis of TH-positive fibers in the striatum showed normalized density in MPTP-lesioned mice pretreated with rAAV.EpoS71E, suggesting that enhanced sprouting induced by EpoS71E may have been responsible for normal behavior and dopaminergic tone in these mice. These results show that systemically administered rAAV-generated non-erythropoietic Epo may protect against MPTP-induced parkinsonism by a combination of neuroprotection and enhanced axonal sprouting.
Collapse
Affiliation(s)
- A Dhanushkodi
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Templeton JP, Geisert EE. A practical approach to optic nerve crush in the mouse. Mol Vis 2012; 18:2147-52. [PMID: 22876142 PMCID: PMC3413441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 07/24/2012] [Indexed: 10/27/2022] Open
Abstract
Our goal is to provide an instructional resource to help others wishing to use the optic nerve crush (ONC) as an experimental procedure. The process is described beginning with the anesthesia, followed by positioning of the mouse, the surgery itself, and post-surgical care. We analyzed the effect of ONC on retinal blood flow, using optical coherence tomography doppler. This procedure produces a consistent loss of cells in the ganglion cell layer, using whole mounts of retina stained with TO-PRO-3. An instructional video is presented that demonstrates a simple surgical approach to effectively crush the optic nerve of the mouse.
Collapse
|