1
|
Liu S, Ji Y, Li H, Ren L, Zhu J, Yang T, Li X, Yao J, Cao X, Yan B. EYE-503: A Novel Retinoic Acid Drug for Treating Retinal Neurodegeneration. Pharmaceuticals (Basel) 2023; 16:1033. [PMID: 37513944 PMCID: PMC10386480 DOI: 10.3390/ph16071033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Retinal neurodegeneration is a major cause of vision loss. Retinoic acid signaling is critical for the maintenance of retinal function, and its dysfunction can cause retinal neurodegeneration. However, the therapeutic effects of retinoic acid drugs on retinal neurodegeneration remain unclear. In this study, we designed a novel retinoic acid drug called EYE-503 and investigated its therapeutic effects of EYE-503 on retinal neurodegeneration. The optic nerve crush (ONC) model was selected for the retinal neurodegeneration study. H&E staining, TUNEL staining, immunofluorescence staining, and visual electrophysiology assays were performed to determine the role of EYE-503 in retinal neurodegeneration in vivo. The CCK-8 assay, EdU incorporation assay, PI staining, and flow cytometry assays were performed to investigate the effects of EYE-503 administration on retinal neurodegeneration in vitro. The potential mechanism of EYE-503 in retinal neurodegeneration was investigated by network pharmacology and Western blots. The results showed that EYE-503 administration had no detectable cytotoxicity and tissue toxicity. EYE-503 administration alleviated ONC-induced retinal injury and optic nerve injury in vivo. EYE-503 administration attenuated retinal ganglion cell apoptosis, inhibited reactive gliosis, and retarded the progression of retinal neurodegeneration. Mechanistically, EYE-503 regulated retinal neurodegeneration by targeting the JNK/p38 signaling pathway. This study suggests that EYE-503 is a promising therapeutic agent for retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Sha Liu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Yuke Ji
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Huan Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Ling Ren
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Junya Zhu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Tianjing Yang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210093, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210093, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Biao Yan
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200433, China
| |
Collapse
|
2
|
Kodati B, Zhang W, He S, Pham JH, Beall KJ, Swanger ZE, Krishnamoorthy VR, Harris PE, Hall T, Tran AV, Chaphalkar RM, Chavala SH, Stankowska DL, Krishnamoorthy RR. The endothelin receptor antagonist macitentan ameliorates endothelin-mediated vasoconstriction and promotes the survival of retinal ganglion cells in rats. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1185755. [PMID: 38464735 PMCID: PMC10921982 DOI: 10.3389/fopht.2023.1185755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Glaucoma is a chronic and progressive eye disease, commonly associated with elevated intraocular pressure (IOP) and characterized by optic nerve degeneration, cupping of the optic disc, and loss of retinal ganglion cells (RGCs). The pathological changes in glaucoma are triggered by multiple mechanisms and both mechanical effects and vascular factors are thought to contribute to the etiology of glaucoma. Various studies have shown that endothelin-1 (ET-1), a vasoactive peptide, acting through its G protein coupled receptors, ETA and ETB, plays a pathophysiologic role in glaucoma. However, the mechanisms by which ET-1 contribute to neurodegeneration remain to be completely understood. Our laboratory and others demonstrated that macitentan (MAC), a pan endothelin receptor antagonist, has neuroprotective effects in rodent models of IOP elevation. The current study aimed to determine if oral administration of a dual endothelin antagonist, macitentan, could promote neuroprotection in an acute model of intravitreal administration of ET-1. We demonstrate that vasoconstriction following the intravitreal administration of ET-1 was attenuated by dietary administration of the ETA/ETB dual receptor antagonist, macitentan (5 mg/kg body weight) in retired breeder Brown Norway rats. ET-1 intravitreal injection produced a 40% loss of RGCs, which was significantly lower in macitentan-treated rats. We also evaluated the expression levels of glial fibrillary acidic protein (GFAP) at 24 h and 7 days post intravitreal administration of ET-1 in Brown Norway rats as well as following ET-1 treatment in cultured human optic nerve head astrocytes. We observed that at the 24 h time point the expression levels of GFAP was upregulated (indicative of glial activation) following intravitreal ET-1 administration in both retina and optic nerve head regions. However, following macitentan administration for 7 days after intravitreal ET-1 administration, we observed an upregulation of GFAP expression, compared to untreated rats injected intravitreally with ET-1 alone. Macitentan treatment in ET-1 administered rats showed protection of RGC somas but was not able to preserve axonal integrity and functionality. The endothelin receptor antagonist, macitentan, has neuroprotective effects in the retinas of Brown Norway rats acting through different mechanisms, including enhancement of RGC survival and reduction of ET-1 mediated vasoconstriction.
Collapse
Affiliation(s)
- Bindu Kodati
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Wei Zhang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Shaoqing He
- Department of Pathology, Children’s Health at Dallas, Dallas, TX, United States
| | - Jennifer H. Pham
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Kallen J. Beall
- Department of General Surgery, Honor Health, Phoenix, AZ, United States
| | - Zoe E. Swanger
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, United States
| | | | - Payton E. Harris
- Department of Graduate Medical Education, Medical City, Fort Worth, TX, United States
| | - Trent Hall
- Williams College, Williamstown, MA, United States
| | - Ashley V. Tran
- School of Natural Sciences and Mathematics, The University of Texas at Dallas, Richardson, TX, United States
| | - Renuka M. Chaphalkar
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, CA, United States
| | - Sai H. Chavala
- Department of Surgery, Burnett School of Medicine at Texas Christian University (TCU), Fort Worth, TX, United States
| | - Dorota L. Stankowska
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Raghu R. Krishnamoorthy
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
3
|
Pai HL, Hsieh SMT, Su YS, Sue XY, Chang HH, Lin DPC. Short-Term Hyperuricemia Leads to Structural Retinal Changes That Can be Reversed by Serum Uric Acid Lowering Agents in Mice. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 36098977 PMCID: PMC9482320 DOI: 10.1167/iovs.63.10.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Metabolic disorders have been implicated in ocular diseases, such as age-related macular degeneration (AMD) and diabetic retinopathy (DR). Recently, hyperuricemia (HUA) has been proposed as another risk factor for AMD, although no cause-and-effect experimental data have been published. In this study, we investigated whether HUA would initiate AMD or related retinal damages in hyperuricemic mice. Methods HUA was induced in male ICR mice by dietary supplements of uric acid and oxonic acid potassium salt, with or without treatments by allopurinol or benzbromarone for various durations. Serum uric acid and angiotensin II concentrations were measured by enzyme-linked immunosorbent assay (ELISA) at regular intervals. The retinal damages were assessed by hematoxylin and eosin staining, immunostaining, and TUNEL assay. The cause-and-effect of HUA was compared among the study groups. Results The results showed that the total thickness of photoreceptor inner and outer segments, as well as the thickness of the photoreceptor outer segment alone, were reduced under HUA. Furthermore, HUA elevated serum angiotensin II, which indicated activation of the renin–angiotensin system (RAS), leading to higher matrix metalloproteinase-2 (MMP-2) expression, and glial activation in the ganglion cell layer. HUA also led to the reduction of retinal pigment epithelium gap junction protein connexin-43 and apoptosis. Uric acid lowering agents, allopurinol or benzbromarone, were effective in ameliorating the impairments. Conclusions HUA may pose as a causative factor of retinal injuries. The reduction of serum uric acid may reduce the detrimental effects caused by HUA.
Collapse
Affiliation(s)
- Hung-Liang Pai
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Sophie Meng-Tien Hsieh
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Yu-Shan Su
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Xin-Yuan Sue
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Han-Hsin Chang
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - David Pei-Cheng Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| |
Collapse
|
4
|
Peña JS, Vazquez M. Harnessing the Neuroprotective Behaviors of Müller Glia for Retinal Repair. FRONT BIOSCI-LANDMRK 2022; 27:169. [PMID: 35748245 PMCID: PMC9639582 DOI: 10.31083/j.fbl2706169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Progressive and irreversible vision loss in mature and aging adults creates a health and economic burden, worldwide. Despite the advancements of many contemporary therapies to restore vision, few approaches have considered the innate benefits of gliosis, the endogenous processes of retinal repair that precede vision loss. Retinal gliosis is fundamentally driven by Müller glia (MG) and is characterized by three primary cellular mechanisms: hypertrophy, proliferation, and migration. In early stages of gliosis, these processes have neuroprotective potential to halt the progression of disease and encourage synaptic activity among neurons. Later stages, however, can lead to glial scarring, which is a hallmark of disease progression and blindness. As a result, the neuroprotective abilities of MG have remained incompletely explored and poorly integrated into current treatment regimens. Bioengineering studies of the intrinsic behaviors of MG hold promise to exploit glial reparative ability, while repressing neuro-disruptive MG responses. In particular, recent in vitro systems have become primary models to analyze individual gliotic processes and provide a stepping stone for in vivo strategies. This review highlights recent studies of MG gliosis seeking to harness MG neuroprotective ability for regeneration using contemporary biotechnologies. We emphasize the importance of studying gliosis as a reparative mechanism, rather than disregarding it as an unfortunate clinical prognosis in diseased retina.
Collapse
Affiliation(s)
- Juan S. Peña
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| |
Collapse
|
5
|
Brown C, Agosta P, McKee C, Walker K, Mazzella M, Alamri A, Svinarich D, Chaudhry GR. Human primitive mesenchymal stem cell-derived retinal progenitor cells improved neuroprotection, neurogenesis, and vision in rd12 mouse model of retinitis pigmentosa. Stem Cell Res Ther 2022; 13:148. [PMID: 35395806 PMCID: PMC8994263 DOI: 10.1186/s13287-022-02828-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/20/2022] [Indexed: 01/05/2023] Open
Abstract
Background Currently, there is no treatment for retinal degenerative diseases (RDD) such as retinitis pigmentosa (RP). Stem cell-based therapies could provide promising opportunities to repair the damaged retina and restore vision. Thus far, primarily adult mesenchymal stem cells (MSCs) have been investigated in preclinical and clinical studies, and the results have not been convincing. We applied a new approach in which primitive (p) MSC-derived retinal progenitor cells (RPCs) were examined to treat retinal degeneration in an rd12 mouse model of RP. Methods Well-characterized pMSCs and RPCs labeled with PKH26 were intravitreally injected into rd12 mice. The vision and retinal function of transplanted animals were analyzed using electroretinography. Animals were killed 4 and 8 weeks after cell transplantation for histological, immunological, molecular, and transcriptomic analyses of the retina. Results Transplanted RPCs significantly improved vision and retinal thickness as well as function in rd12 mice. pMSCs and RPCs homed to distinct retinal layers. pMSCs homed to the retinal pigment epithelium, and RPCs migrated to the neural layers of the retina, where they improved the thickness of the respective layers and expressed cell-specific markers. RPCs induced anti-inflammatory and neuroprotective responses as well as upregulated the expression of genes involved in neurogenesis. The transcriptomic analysis showed that RPCs promoted neurogenesis and functional recovery of the retina through inhibition of BMP and activation of JAK/STAT and MAPK signaling pathways. Conclusions Our study demonstrated that RPCs countered inflammation, provided retinal protection, and promoted neurogenesis resulting in improved retinal structure and physiological function in rd12 mice. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02828-w.
Collapse
Affiliation(s)
- Christina Brown
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Patrina Agosta
- Ascension Providence Hospital, Southfield, MI, 48075, USA
| | - Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Matteo Mazzella
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Ali Alamri
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | | | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA.
| |
Collapse
|
6
|
Lin NH, Yang AW, Chang CH, Perng MD. Elevated GFAP isoform expression promotes protein aggregation and compromises astrocyte function. FASEB J 2021; 35:e21614. [PMID: 33908669 DOI: 10.1096/fj.202100087r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 01/22/2023]
Abstract
Alexander disease (AxD) caused by mutations in the coding region of GFAP is a neurodegenerative disease characterized by astrocyte dysfunction, GFAP aggregation, and Rosenthal fiber accumulation. Although how GFAP mutations cause disease is not fully understood, Rosenthal fibers could be induced by forced overexpression of human GFAP and this could be lethal in mice implicate that an increase in GFAP levels is central to AxD pathogenesis. Our recent studies demonstrated that intronic GFAP mutations cause disease by altering GFAP splicing, suggesting that an increase in GFAP isoform expression could lead to protein aggregation and astrocyte dysfunction that typify AxD. Here we test this hypothesis by establishing primary astrocyte cultures from transgenic mice overexpressing human GFAP. We found that GFAP-δ and GFAP-κ were disproportionately increased in transgenic astrocytes and both were enriched in Rosenthal fibers of human AxD brains. In vitro assembly studies showed that while the major isoform GFAP-α self-assembled into typical 10-nm filaments, minor isoforms including GFAP-δ, -κ, and -λ were assembly-compromised and aggregation prone. Lentiviral transduction showed that expression of these minor GFAP isoforms decreased filament solubility and increased GFAP stability, leading to the formation of Rosenthal fibers-like aggregates that also disrupted the endogenous intermediate filament networks. The aggregate-bearing astrocytes lost their normal morphology and glutamate buffering capacity, which had a toxic effect on neighboring neurons. In conclusion, our findings provide evidence that links elevated GFAP isoform expression with GFAP aggregation and impaired glutamate transport, and suggest a potential non-cell-autonomous mechanism underlying neurodegeneration through astrocyte dysfunction.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Ai-Wen Yang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Hsuan Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Ming-Der Perng
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan.,Department of Medical Science, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
7
|
Astrogliosis and episodic memory in late life: higher GFAP is related to worse memory and white matter microstructure in healthy aging and Alzheimer's disease. Neurobiol Aging 2021; 103:68-77. [PMID: 33845398 DOI: 10.1016/j.neurobiolaging.2021.02.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/05/2021] [Accepted: 02/18/2021] [Indexed: 12/22/2022]
Abstract
Astrocytes play a formative role in memory consolidation during physiological conditions; when dysregulated, astrocytes release glial fibrillary acidic protein (GFAP), which has been linked with negative memory outcomes in animal studies. We examined the association between blood GFAP, memory, and white matter (WM) integrity, accounting for blood markers of AD pathology (i.e., Aβ42) and neurodegeneration (i.e., total tau; neurofilament light chain) in 114 older adults (asymptomatic, n = 69; MCI/AD dementia, n = 45). Higher levels of GFAP were associated with lower memory scores (p < 0.0001), such that for 1 SD increase in mean GFAP values, the memory composite score decreased on average by 0.49 (Standard error = 0.071). These results remained significant after controlling for diagnostic status and AD-related blood biomarkers. Higher GFAP was also related to lower WM integrity in regions vulnerable to AD pathology; however, WM integrity did not account for the association between GFAP and memory. Study findings suggest that higher blood levels of a marker of astrogliosis may reflect impoverished memory functions and white matter health, independent of markers of amyloid or neurodegeneration.
Collapse
|
8
|
Grape seed proanthocyanidins protect retinal ganglion cells by inhibiting oxidative stress and mitochondrial alteration. Arch Pharm Res 2020; 43:1056-1066. [PMID: 33078305 DOI: 10.1007/s12272-020-01272-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
Grape seed proanthocyanidins (GSP) are known as condensed tannins and have been used as an anti-oxidant in various neurodegenerative diseases. In our study, GSP was used as a daily dietary supplement and the neuroprotective effects were evaluated on the retinal ganglion cells (RGCs) in the retinal tissues in glaucomatous DBA/2D (D2) mice. D2 mice and age-matched non-glaucomatous DBA/2J-Gpnmb+ (D2-Gpnmb+) mice were fed with GSP or a control diet for up to 6 months. The intraocular pressure (IOP), RGC survival, glial fibrillary acidic protein (GFAP), the levels of apoptotic proteins, and the expression of oxidative stress markers in retinal tissues were determined. In our study, the neuroprotective effects of GSP on retinal tissues were confirmed, as evidenced by (a) GSP inhibited the IOP elevation in D2 mice; (b) GSP enhanced RGC survival and mediated the apoptotic protein expression; (c) GSP suppressed GFAP expression; and (d) the oxidative stress and the levels of mitochondrial reactive oxygen species were regulated by GSP. Our findings indicate that GSP has promising potential to preserve retinal tissue functions via regulating oxidative stress and mitochondrial functions.
Collapse
|
9
|
Stojanovska V, McQuade RM, Miller S, Nurgali K. Effects of Oxaliplatin Treatment on the Myenteric Plexus Innervation and Glia in the Murine Distal Colon. J Histochem Cytochem 2018; 66:723-736. [PMID: 29741434 DOI: 10.1369/0022155418774755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oxaliplatin (platinum-based chemotherapeutic agent) is a first-line treatment of colorectal malignancies; its use associates with peripheral neuropathies and gastrointestinal side effects. These gastrointestinal dysfunctions might be due to toxic effects of oxaliplatin on the intestinal innervation and glia. Male Balb/c mice received intraperitoneal injections of sterile water or oxaliplatin (3 mg/kg/d) triweekly for 2 weeks. Colon tissues were collected for immunohistochemical assessment at day 14. The density of sensory, adrenergic, and cholinergic nerve fibers labeled with calcitonin gene-related peptide (CGRP), tyrosine hydroxylase (TH), and vesicular acetylcholine transporter (VAChT), respectively, was assessed within the myenteric plexus of the distal colon. The number and proportion of excitatory neurons immunoreactive (IR) against choline acetyltransferase (ChAT) were counted, and the density of glial subpopulations was determined by using antibodies specific for glial fibrillary acidic protein (GFAP) and s100β protein. Oxaliplatin treatment induced significant reduction of sensory and adrenergic innervations, as well as the total number and proportion of ChAT-IR neurons, and GFAP-IR glia, but increased s100β expression within the myenteric plexus of the distal colon. Treatment with oxaliplatin significantly alters nerve fibers and glial cells in the colonic myenteric plexus, which could contribute to long-term gastrointestinal side effects following chemotherapeutic treatment.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Rachel M McQuade
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia.,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Bollaerts I, Veys L, Geeraerts E, Andries L, De Groef L, Buyens T, Salinas-Navarro M, Moons L, Van Hove I. Complementary research models and methods to study axonal regeneration in the vertebrate retinofugal system. Brain Struct Funct 2017; 223:545-567. [DOI: 10.1007/s00429-017-1571-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/15/2017] [Indexed: 01/18/2023]
|
11
|
|
12
|
Zhou L, Wang H, Luo J, Xiong K, Zeng L, Chen D, Huang J. Regulatory effects of inhibiting the activation of glial cells on retinal synaptic plasticity. Neural Regen Res 2014; 9:385-393. [PMID: 25206825 PMCID: PMC4146193 DOI: 10.4103/1673-5374.128240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2014] [Indexed: 01/09/2023] Open
Abstract
Various retinal injuries induced by ocular hypertension have been shown to induce plastic changes in retinal synapses, but the potential regulatory mechanism of synaptic plasticity after retinal injury was still unclear. A rat model of acute ocular hypertension was established by injecting saline intravitreally for an hour, and elevating the intraocular pressure to 14.63 kPa (110 mmHg). Western blot assay and immunofluorescence results showed that synaptophysin expression had a distinct spatiotemporal change that increased in the inner plexiform layer within 1 day and spread across the outer plexiform layer after 3 days. Glial fibrillary acidic protein expression in retinae was greatly increased after 3 days, and reached a peak at 7 days, which was also consistent with the peak time of synaptophysin expression in the outer plexiform layer following the increased intraocular pressure. Fluorocitrate, a glial metabolic inhibitor, was intravitreally injected to inhibit glial cell activation following high intraocular pressure. This significantly inhibited the enhanced glial fibrillary acidic protein expression induced by high intraocular pressure injury. Synaptophysin expression also decreased in the inner plexiform layer within a day and the widened distribution in the outer plexiform layer had disappeared by 3 days. The results suggested that retinal glial cell activation might play an important role in the process of retinal synaptic plasticity induced by acute high intraocular pressure through affecting the expression and distribution of synaptic functional proteins, such as synaptophysin.
Collapse
Affiliation(s)
- Lihong Zhou
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jia Luo
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Leping Zeng
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jufang Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
13
|
Piret G, Perez MT, Prinz CN. Substrate porosity induces phenotypic alterations in retinal cells cultured on silicon nanowires. RSC Adv 2014. [DOI: 10.1039/c4ra04121f] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Limitations of silicon nanowire arrays produced using chemical etching for drug delivery.
Collapse
Affiliation(s)
- Gaëlle Piret
- Division of Solid State Physics
- Lund University
- SE-221 00 Lund, Sweden
- Neuronano Research Center
- Lund University
| | - Maria-Thereza Perez
- Department of Clinical Sciences
- Division of Ophthalmology
- Lund University
- SE-221 84 Lund, Sweden
- The Nanometer Structure Consortium
| | - Christelle N. Prinz
- Division of Solid State Physics
- Lund University
- SE-221 00 Lund, Sweden
- Neuronano Research Center
- Lund University
| |
Collapse
|