1
|
de la Monte SM, Tong M, Hapel AJ. Concordant and Discordant Cerebrospinal Fluid and Plasma Cytokine and Chemokine Responses in Mild Cognitive Impairment and Early-Stage Alzheimer's Disease. Biomedicines 2023; 11:2394. [PMID: 37760836 PMCID: PMC10525668 DOI: 10.3390/biomedicines11092394] [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: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Neuroinflammation may be a pathogenic mediator and biomarker of neurodegeneration at the boundary between mild cognitive impairment (MCI) and early-stage Alzheimer's disease (AD). Whether neuroinflammatory processes are endogenous to the central nervous system (CNS) or originate from systemic (peripheral blood) sources could impact strategies for therapeutic intervention. To address this issue, we measured cytokine and chemokine immunoreactivities in simultaneously obtained lumbar puncture cerebrospinal fluid (CSF) and serum samples from 39 patients including 18 with MCI or early AD and 21 normal controls using a 27-plex XMAP bead-based enzyme-linked immunosorbent assay (ELISA). The MCI/AD combined group had significant (p < 0.05 or better) or statistically trend-wise (0.05 ≤ p ≤ 0.10) concordant increases in CSF and serum IL-4, IL-5, IL-9, IL-13, and TNF-α and reductions in GM-CSF, b-FGF, IL-6, IP-10, and MCP-1; CSF-only increases in IFN-y and IL-7 and reductions in VEGF and IL-12p70; serum-only increases in IL-1β, MIP-1α, and eotaxin and reductions in G-CSF, IL-2, IL-8 and IL-15; and discordant CSF-serum responses with reduced CSF and increased serum PDGF-bb, IL-17a, and RANTES. The results demonstrate simultaneously parallel mixed but modestly greater pro-inflammatory compared to anti-inflammatory or neuroprotective responses in CSF and serum. In addition, the findings show evidence that several cytokines and chemokines are selectively altered in MCI/AD CSF, likely corresponding to distinct neuroinflammatory responses unrelated to systemic pathologies. The aggregate results suggest that early management of MCI/AD neuroinflammation should include both anti-inflammatory and pro-neuroprotective strategies to help prevent disease progression.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology (Neuropathology), Neurology, and Neurosurgery, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Andrew J. Hapel
- Department of Genome Biology, John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| |
Collapse
|
2
|
Siyanaki MRH, Azab MA, Lucke-Wold B. Traumatic Optic Neuropathy: Update on Management. ENCYCLOPEDIA 2023; 3:88-101. [PMID: 36718432 PMCID: PMC9884099 DOI: 10.3390/encyclopedia3010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Traumatic optic neuropathy is one of the causes of visual loss caused by blunt or penetrating head trauma and is classified as both direct and indirect. Clinical history and examination findings usually allow for the diagnosis of traumatic optic neuropathy. There is still controversy surrounding the management of traumatic optic neuropathy; some physicians advocate observation alone, while others recommend steroid therapy, surgery, or both. In this entry, we tried to highlight traumatic optic neuropathy's main pathophysiologic mechanisms with the most available updated treatment. Recent research suggests future therapies that may be helpful in traumatic optic neuropathy cases.
Collapse
Affiliation(s)
| | - Mohammed A. Azab
- Department of Neurosurgery, University of Cairo University, Cairo 12613, Egypt
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
3
|
Hsu CL, Wen YT, Hsu TC, Chen CC, Lee LY, Chen WP, Tsai RK. Neuroprotective Effects of Erinacine A on an Experimental Model of Traumatic Optic Neuropathy. Int J Mol Sci 2023; 24:1504. [PMID: 36675019 PMCID: PMC9864134 DOI: 10.3390/ijms24021504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Erinacine A (EA), a natural neuroprotectant, is isolated from a Chinese herbal medicine, Hericium erinaceus. The aim of this study was to investigate the neuroprotective effects of EA in a rat model of traumatic optic neuropathy. The optic nerves (ONs) of adult male Wistar rats were crushed using a standardized method and divided into three experimental groups: phosphate-buffered saline (PBS control)-treated group, standard EA dose-treated group (2.64 mg/kg in 0.5 mL of PBS), and double EA dose-treated group (5.28 mg/kg in 0.5 mL of PBS). After ON crush, each group was fed orally every day for 14 days before being euthanized. The visual function, retinal ganglion cell (RGC) density, and RGC apoptosis were determined using flash visual-evoked potentials (fVEP) analysis, retrograde Fluoro-Gold labelling, and TdT-dUTP nick end-labelling (TUNEL) assay, respectively. Macrophage infiltration of ON was detected by immunostaining (immunohistochemistry) for ED1. The protein levels of phosphor-receptor-interacting serine/threonine-protein kinase1 (pRIP1), caspase 8 (Cas8), cleaved caspase 3 (cCas3), tumour necrosis factor (TNF)-α, tumour necrosis factor receptor1 (TNFR1), interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), nuclear factor erythroid 2-related factor 2 (Nrf2), haem oxygenase-1 (HO-1), and superoxide dismutase 1 (SOD1) were evaluated by Western blotting. When comparing the standard EA dose-treated group and the double EA dose-treated group with the PBS-treated group, fVEP analysis showed that the amplitudes of P1−N2 in the standard EA dose group and the double EA dose-treated group were 1.8 and 2.4-fold, respectively, higher than that in the PBS-treated group (p < 0.05). The density of RGC in the standard EA dose-treated group and the double EA dose-treated group were 2.3 and 3.7-fold, respectively, higher than that in the PBS-treated group (p < 0.05). The TUNEL assay showed that the standard EA dose-treated group and the double EA dose-treated group had significantly reduced numbers of apoptotic RGC by 10.0 and 15.6-fold, respectively, compared with the PBS-treated group (p < 0.05). The numbers of macrophages on ON were reduced by 1.8 and 2.2-fold in the standard EA dose-treated group and the double EA dose-treated group, respectively (p < 0.01). On the retinal samples, the levels of pRIP, Cas8, cCas3, TNF-α, TNFR1, IL-1β, and iNOS were decreased, whereas those of Nrf2, HO-1, and SOD1 were increased in both EA-treated groups compared to those in the PBS-treated group (p < 0.05). EA treatment has neuroprotective effects on an experimental model of traumatic optic neuropathy by suppressing apoptosis, neuroinflammation, and oxidative stress to protect the RGCs from death as well as preserving the visual function.
Collapse
Affiliation(s)
- Chiao-Ling Hsu
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Tzu-Chao Hsu
- Department of Medical Education, Medical Administration Office, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grap King Bio Ltd., Taoyuan 325002, Taiwan
| | - Li-Ya Lee
- Biotech Research Institute, Grap King Bio Ltd., Taoyuan 325002, Taiwan
| | - Wan-Ping Chen
- Biotech Research Institute, Grap King Bio Ltd., Taoyuan 325002, Taiwan
| | - Rong-Kung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 970, Taiwan
| |
Collapse
|
4
|
Lowering the Intraocular Pressure in Rats and Rabbits by Cordyceps cicadae Extract and Its Active Compounds. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030707. [PMID: 35163975 PMCID: PMC8837943 DOI: 10.3390/molecules27030707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 01/01/2023]
Abstract
Cordyceps cicadae (CC), an entomogenous fungus that has been reported to have therapeutic glaucoma, is a major cause of blindness worldwide and is characterized by progressive retinal ganglion cell (RGC) death, mostly due to elevated intraocular pressure (IOP). Here, an ethanolic extract of C. cicadae mycelium (CCME), a traditional medicinal mushroom, was studied for its potential in lowering IOP in rat and rabbit models. Data showed that CCME could significantly (60.5%) reduce the IOP induced by microbead occlusion after 56 days of oral administration. The apoptosis of retinal ganglion cells (RGCs) in rats decreased by 77.2%. CCME was also shown to lower the IOP of normal and dextrose-infusion-induced rabbits within 60 min after oral feeding. There were dose effects, and the effect was repeatable. The active ingredient, N6-(2-hydroxyethyl)-adenosine (HEA), was also shown to alleviate 29.6% IOP at 0.2 mg/kg body weight in this rabbit model. CCME was confirmed with only minor inhibition in the phosphorylated myosin light chain 2 (pMLC2) pathway.
Collapse
|
5
|
Kang EYC, Liu PK, Wen YT, Quinn PMJ, Levi SR, Wang NK, Tsai RK. Role of Oxidative Stress in Ocular Diseases Associated with Retinal Ganglion Cells Degeneration. Antioxidants (Basel) 2021; 10:1948. [PMID: 34943051 PMCID: PMC8750806 DOI: 10.3390/antiox10121948] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Ocular diseases associated with retinal ganglion cell (RGC) degeneration is the most common neurodegenerative disorder that causes irreversible blindness worldwide. It is characterized by visual field defects and progressive optic nerve atrophy. The underlying pathophysiology and mechanisms of RGC degeneration in several ocular diseases remain largely unknown. RGCs are a population of central nervous system neurons, with their soma located in the retina and long axons that extend through the optic nerve to form distal terminals and connections in the brain. Because of this unique cytoarchitecture and highly compartmentalized energy demand, RGCs are highly mitochondrial-dependent for adenosine triphosphate (ATP) production. Recently, oxidative stress and mitochondrial dysfunction have been found to be the principal mechanisms in RGC degeneration as well as in other neurodegenerative disorders. Here, we review the role of oxidative stress in several ocular diseases associated with RGC degenerations, including glaucoma, hereditary optic atrophy, inflammatory optic neuritis, ischemic optic neuropathy, traumatic optic neuropathy, and drug toxicity. We also review experimental approaches using cell and animal models for research on the underlying mechanisms of RGC degeneration. Lastly, we discuss the application of antioxidants as a potential future therapy for the ocular diseases associated with RGC degenerations.
Collapse
Affiliation(s)
- Eugene Yu-Chuan Kang
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Pei-Kang Liu
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 80424, Taiwan;
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80424, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97403, Taiwan;
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; (P.M.J.Q.); (S.R.L.)
| | - Sarah R. Levi
- Jonas Children’s Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; (P.M.J.Q.); (S.R.L.)
| | - Nan-Kai Wang
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97403, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 97403, Taiwan
| |
Collapse
|
6
|
Huang CT, Wen YT, Desai TD, Tsai RK. Intravitreal Injection of Long-Acting Pegylated Granulocyte Colony-Stimulating Factor Provides Neuroprotective Effects via Antioxidant Response in a Rat Model of Traumatic Optic Neuropathy. Antioxidants (Basel) 2021; 10:1934. [PMID: 34943037 PMCID: PMC8750325 DOI: 10.3390/antiox10121934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022] Open
Abstract
Traumatic optic neuropathy (TON) may cause severe visual loss following direct or indirect head trauma which may result in optic nerve injuries and therefore contribute to the subsequent loss of retinal ganglion cells by inflammatory mediators and reactive oxygen species (ROS). Granulocyte colony-stimulating factor (G-CSF) provides the anti-inflammatory and anti-oxidative actions but has a short half-life and also induces leukocytosis upon typical systemic administration. The purpose of the present study was to investigate the relationship between the anti-oxidative response and neuroprotective effects of long-acting pegylated human G-CSF (PEG-G-CSF) in a rat model of optic nerve crush (ONC). Adult male Wistar rats (150-180 g) were chosen to have a sham operation in one eye and have ONC in the other. PEG-G-CSF or phosphate-buffered saline (PBS control) was immediately administered after ONC by intravitreal injection (IVI). We found the IVI of PEG-G-CSF does not induce systemic leukocytosis, but increases survival of RGCs and preserves the visual function after ONC. TUNEL assays showed fewer apoptotic cells in the retina in the PEG-G-CSF-treated eyes. The number of sorely ED1-positive cells was attenuated at the lesion site in the PEG-G-CSF-treated eyes. Immunoblotting showed up-regulation of p-Akt1, Nrf2, Sirt3, and HO-1 in the ON of the PEG-G-CSF-treated eyes. Our results demonstrated that one IVI of long-acting PEG-G-CSF is neuroprotective in the rONC. PEG-G-CSF activates the p-Akt1/Nrf2/Sirt3 and the p-Akt1/Nrf2/HO-1 axes to provide the antioxidative action and further attenuated RGC apoptosis and neuroinflammation. This provides crucial preclinical information for the development of alternative therapy with IVI of PEG-G-CSF in TON.
Collapse
Affiliation(s)
- Chin-Te Huang
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Department of Ophthalmology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Tushar Dnyaneshwar Desai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
| | - Rong-Kung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-T.W.); (T.D.D.)
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 970, Taiwan
| |
Collapse
|
7
|
Yang M, Jian L, Fan W, Chen X, Zou H, Huang Y, Chen X, Zhou YG, Yuan R. Axon regeneration after optic nerve injury in rats can be improved via PirB knockdown in the retina. Cell Biosci 2021; 11:158. [PMID: 34380548 PMCID: PMC8359350 DOI: 10.1186/s13578-021-00670-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/25/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND In the central nervous system (CNS), three types of myelin-associated inhibitors (MAIs) exert major inhibitory effects on nerve regeneration: Nogo-A, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp). MAIs have two co-receptors, Nogo receptor (NgR) and paired immunoglobulin-like receptor B (PirB). Existing studies confirm that inhibiting NgR only exerted a modest disinhibitory effect in CNS. However, the inhibitory effects of PirB on nerve regeneration after binding to MAIs are controversial too. We aimed to further investigate the effect of PirB knockdown on the neuroprotection and axonal regeneration of retinal ganglion cells (RGCs) after optic nerve injury in rats. METHODS The differential expression of PirB in the retina was observed via immunofluorescence and western blotting after 1, 3, and 7 days of optic nerve injury (ONI). The retina was locally transfected with adeno-associated virus (AAV) PirB shRNA, then, the distribution of virus in tissues and cells was observed 21 days after AAV transfection to confirm the efficiency of PirB knockdown. Level of P-Stat3 and expressions of ciliary neurotrophic factor (CNTF) were detected via western blotting. RGCs were directly labeled with cholera toxin subunit B (CTB). The new axons of the optic nerve were specifically labeled with growth associated protein-43 (GAP43) via immunofluorescence. Flash visual evoked potential (FVEP) was used to detect the P1 and N1 latency, as well as N1-P1, P1-N2 amplitude to confirm visual function. RESULTS PirB expression in the retina was significantly increased after ONI. PirB knockdown was successful and significantly promoted P-Stat3 level and CNTF expression in the retina. PirB knockdown promoted the regeneration of optic nerve axons and improved the visual function indexes such as N1-P1 and P1-N2 amplitude. CONCLUSIONS PirB is one of the key molecules that inhibit the regeneration of the optic nerve, and inhibition of PirB has an excellent effect on promoting nerve regeneration, which allows the use of PirB as a target molecule to promote functional recovery after ONI.
Collapse
Affiliation(s)
- Mei Yang
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Lan Jian
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Wei Fan
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Xing Chen
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, 10 Changjiang Zhilu, Chongqing, 400042, People's Republic of China
| | - Huan Zou
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Yanming Huang
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Xiaofan Chen
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China
| | - Yuan-Guo Zhou
- The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University, 10 Changjiang Zhilu, Chongqing, 400042, People's Republic of China.
| | - Rongdi Yuan
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, 183 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
8
|
Wen YT, Huang CW, Liu CP, Chen CH, Tu CM, Hwang CS, Chen YH, Chen WR, Lin KL, Ho YC, Chen TC, Tsai RK. Inhibition of Retinal Ganglion Cell Loss By a Novel ROCK Inhibitor (E212) in Ischemic Optic Nerve Injury Via Antioxidative and Anti-Inflammatory Actions. Invest Ophthalmol Vis Sci 2021; 62:21. [PMID: 34015079 PMCID: PMC8142697 DOI: 10.1167/iovs.62.6.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose This study investigated the neuroprotective effects of administration of ROCK inhibitor E212 on ischemic optic neuropathy. Methods Rats received an intravitreal injection of either E212 or PBS immediately after optic nerve infarct. The oxidative stress in the retina was detected by performing superoxide dismutase activity and CellROX assays. The integrity of retinal pigment epithelium was determined by staining of zona occludens 1. The visual function, retinal ganglion cell (RGC) density, and RGC apoptosis were determined by using flash visual-evoked potential analysis, retrograde FluoroGold labeling, and TdT-dUTP nick end-labeling assay. Macrophage infiltration was detected by staining for ED1. The protein levels of TNF-α, p-CRMP, p-AKT1, p-STAT3, and CD206 were evaluated using Western blotting. Results Administration of E212 resulted in a 1.23-fold increase in the superoxide dismutase activity of the retina and 2.28-fold decrease in RGC-produced reactive oxygen species as compared to the levels observed upon treatment with PBS (P < 0.05). Moreover, E212 prevented the disruption of the blood-retinal barrier (BRB) in contrast to PBS. The P1-N2 amplitude and RGC density in the E212-treated group were 1.75- and 2.05-fold higher, respectively, than those in the PBS-treated group (P < 0.05). The numbers of apoptotic RGCs and macrophages were reduced by 2.93- and 2.54-fold, respectively, in the E212-treated group compared with those in the PBS-treated group (P < 0.05). The levels of p-AKT1, p-STAT3, and CD206 were increased, whereas those of p-PTEN, p-CRMP2, and TNF-α were decreased after treatment with E212 (P < 0.05). Conclusions Treatment with E212 suppresses oxidative stress, BRB disruption, and neuroinflammation to protect the visual function in ischemic optic neuropathy.
Collapse
Affiliation(s)
- Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Wen Huang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Peng Liu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hung Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chia-Mu Tu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chrong-Shiong Hwang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Yi-Hsun Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wan-Ru Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Keh-Liang Lin
- Department of Medical laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chieh Ho
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
| |
Collapse
|
9
|
Abri Aghdam K, Aghajani A, Ashraf Khorasani M, Soltan Sanjari M, Chaibakhsh S, Habibi A, Falavarjani KG. Intravitreal Injection Of The Granulocyte-Colony Stimulating Factor For The Treatment Of Non-Arteritic Anterior Ischemic Optic Neuropathy: A Pilot Study. Semin Ophthalmol 2021; 36:649-657. [PMID: 33689586 DOI: 10.1080/08820538.2021.1896749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose To investigate the efficacy of intravitreal injection of granulocyte colony-stimulating factor (G-CSF) for the treatment of non-arteritic anterior ischemic optic neuropathy (NAION).Methods: Patients with acute NAION were enrolled in this prospective interventional case series. They received an intravitreal injection of 60 micrograms in 0.1 ml of G-CSF within 2 weeks of the onset of the disease. Visual acuity, visual field, intraocular pressure (IOP), corneal endothelial cell density, and peripapillary retinal nerve fiber layer (RNFL) thickness were recorded before injections and 1 week, 1 month, 3 months, 6 months, and one year after the injections. Full-field electroretinography (ERG) was obtained at the baseline, 1 month, and 12 months post- injections.Results: Fourteen eyes of 14 patients entered the study. Best-corrected visual acuity (BCVA) significantly improved in the first month following injections (p = .007), decreased subsequently, and the final BCVA showed no significant improvement (p = .278) compared to the baseline measurements. A significant decrease in RNFL thickness was observed in all quadrants compared to the baseline measurements. Also, no improvement in the visual field parameters was observed. From the toxicity aspect, no significant changes in the corneal endothelial cell density, IOP, and ERG recordings were observed.Conclusion: Intravitreal injection of G-CSF seems to be safe. The effect may last for one month and then decline.
Collapse
Affiliation(s)
- Kaveh Abri Aghdam
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Aghajani
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Ashraf Khorasani
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Soltan Sanjari
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Chaibakhsh
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Habibi
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
10
|
Huang W, Lan Q, Jiang L, Yan W, Tang F, Shen C, Huang H, Zhong H, Lv J, Zeng S, Li M, Mo Z, Hu B, Liang N, Chen Q, Zhang M, Xu F, Cui L. Fasudil attenuates glial cell-mediated neuroinflammation via ERK1/2 and AKT signaling pathways after optic nerve crush. Mol Biol Rep 2020; 47:8963-8973. [PMID: 33161529 DOI: 10.1007/s11033-020-05953-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/28/2020] [Indexed: 11/28/2022]
Abstract
To investigate the functional role of fasudil in optic nerve crush (ONC), and further explore its possible molecular mechanism. After ONC injury, the rats were injected intraperitoneally either with fasudil or normal saline once a day until euthanized. RGCs survival was assessed by retrograde labeling with FluoroGold. Retinal glial cells activation and population changes (GFAP, iba-1) were measured by immunofluorescence. The expressions of cleaved caspase 3 and 9, p-ERK1/2 and p-AKT were detected by western blot. The levels of the pro-inflammatory cytokines were determined using real-time polymerase chain reaction. Fasudil treatment inhibited RGCs apoptosis and reduced RGCs loss demonstrated by the decreased apoptosis-associated proteins expression and the increased fluorogold labeling of RGCs after ONC, respectively. In addition, the ONC + fasudil group compared had a significantly lower expression of GFAP and iba1 compared with the ONC group. The levels of pro-inflammatory cytokines were significantly reduced in the ONC + fasudil group than in the ONC group. Furthermore, the phosphorylation levels of ERK1/2 and AKT (p-ERK1/2 and p-AKT) were obviously elevated by the fasudil treatment. Our study demonstrated that fasudil attenuated glial cell-mediated neuroinflammation by up-regulating the ERK1/2 and AKT signaling pathways in rats ONC models. We conclude that fasudil may be a novel treatment for traumatic optic neuropathy.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.,Guangxi Medical University, Nanning, 530021, China
| | - Qianqian Lan
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Li Jiang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Wenya Yan
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Fen Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Chaolan Shen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Hui Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Haibin Zhong
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Jian Lv
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Siming Zeng
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Min Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Zhongxiang Mo
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Bing Hu
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Ning Liang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Qi Chen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning, 530021, China
| | - Fan Xu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.
| | - Ling Cui
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China.
| |
Collapse
|
11
|
Wu Y, Zhan Z, Quan Y, Yang Y, Chen X, Liu L, Wu K, Yu M. SP1-mediated upregulation of LINGO-1 promotes degeneration of retinal ganglion cells in optic nerve injury. CNS Neurosci Ther 2020; 26:1010-1020. [PMID: 32562344 PMCID: PMC7539844 DOI: 10.1111/cns.13426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUNDS Insults to the axons in the optic nerve head are the primary cause of loss of retinal ganglion cells (RGCs) in traumatic, ischemic nerve injury or degenerative ocular diseases. The central nervous system-specific leucine-rich repeat protein, LINGO-1, negatively regulates axon regeneration and neuronal survival after injury. However, the upstream molecular mechanisms that regulate LINGO-1 signaling and contribute to LINGO-1-mediated death of RGCs are unclear. METHODS The expression of SP1 was profiled in optic nerve crush (ONC)-injured RGCs. LINGO-1 level was examined after SP1 overexpression by qRT-PCR. Luciferase assay was used to examine the binding of SP1 to the promoter regions of LINGO-1. Primary RGCs from rat retina were isolated by immunopanning and RGCs apoptosis were determined by Tunnel. SP1 and LINGO-1 expression was investigated using immunohistochemistry and Western bolting. Neuroprotection was assessed by RGC counts, RNFL thickness, and VEP tests after inhibition of SP1 shRNA. RESULTS We demonstrate that SP1 was upregulated in ONC-injured RGCs. SP1 was bound to the LINGO-1 promoter, which led to increased expression of LINGO-1. Treatment with recombinant Nogo-66 or LINGO-1 promoted apoptosis of RGCs cultured under serum-deprivation conditions, while silencing of SP1 promoted the survival of RGCs. SP1 and LINGO-1 colocalized and were upregulated in ONC-injured retinas. Silencing of SP1 in vivo reduced LINGO-1 expression and protected the structure of RGCs from ONC-induced injury, but there was no sign of recovery in VEP. CONCLUSIONS Our findings imply that SP1 regulates LINGO-1 expression in RGCs in the injured retina and provide insight into mechanisms underlying LINGO-1-mediated RGC death in optic nerve injury.
Collapse
Affiliation(s)
- Yali Wu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Zongyi Zhan
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yadan Quan
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yangfan Yang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Xiaotao Chen
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Liling Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Kaili Wu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Minbin Yu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
12
|
Huang T, Li H, Zhang S, Liu F, Wang D, Xu J. Nrn1 Overexpression Attenuates Retinal Ganglion Cell Apoptosis, Promotes Axonal Regeneration, and Improves Visual Function Following Optic Nerve Crush in Rats. J Mol Neurosci 2020; 71:66-79. [DOI: 10.1007/s12031-020-01627-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022]
|
13
|
Liu PK, Wen YT, Lin W, Kapupara K, Tai M, Tsai RK. Neuroprotective effects of low-dose G-CSF plus meloxicam in a rat model of anterior ischemic optic neuropathy. Sci Rep 2020; 10:10351. [PMID: 32587280 PMCID: PMC7316837 DOI: 10.1038/s41598-020-66977-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 05/19/2020] [Indexed: 11/29/2022] Open
Abstract
Non-arteritic anterior ischemic optic neuropathy (NAION) causes a sudden loss of vision and lacks effective treatment. Granulocyte colony-stimulating factor (G-CSF) provides neuroprotection against the experimental optic nerve injuries but also induce leukocytosis upon typical administration. We found synergetic neuroprotective effects of meloxicam and low dose G-CSF without leukocytosis in a rat model of anterior ischemic optic neuropathy (rAION). The WBC counts in the low-dose G-CSF-plus meloxicam-treated group were similar to the sham-operated group. Combination treatment of low-dose G-CSF plus meloxicam preserved RGCs survival and visual function, reduced RGC apoptosis and the macrophages infiltration, and promote more M2 phenotype of macrophage/microglial transition than the low-dose GCSF treatment or the meloxicam treatment. Moreover, the combination treatment induced higher serine/threonine kinase 1 (Akt1) expression. The combination treatment of low-dose G-CSF plus meloxicam lessened the leukocytotic side effect and provided neuroprotective effects via Akt1 activation in the rAION model. This approach provides crucial preclinical information for the development of alternative therapy in AION.
Collapse
Affiliation(s)
- Pei-Kang Liu
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, Yuan's General Hospital, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei Lin
- Department of Optometry, Da-Yeh University, Changhwa, Taiwan
| | - Kishan Kapupara
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Minghong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan. .,Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung, Taiwan. .,Graduate Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan. .,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
14
|
Liu X, Feng L, Shinde I, Cole JD, Troy JB, Saggere L. Correlation between retinal ganglion cell loss and nerve crush force-impulse established with instrumented tweezers in mice. Neurol Res 2020; 42:379-386. [PMID: 32100638 DOI: 10.1080/01616412.2020.1733322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objectives: Rodent models of optic nerve crush (ONC) have often been used to study degeneration and regeneration of retinal ganglion cells (RGCs) and their axons as well as the underlying molecular mechanisms. However, ONC results from different laboratories exhibit a range of RGC injury with varying degree of axonal damage. We developed instrumented tweezers to measure optic nerve (ON) crush forces in real time and studied the correlation between RGC axon loss and force-impulse, the product of force and duration, applied through the instrumented tweezers in mice.Methods: A pair of standard self-closing #N7 tweezers were instrumented with miniature foil strain gauges at optimal locations on both tweezers' arms. The instrumented tweezers were capable of recording the tip closure forces in the form of voltages, which were calibrated through load cells to corresponding tip closure forces over the operating range. Using the instrumented tweezers, the ONs of multiple mice were crushed with varied forces and durations and the axons in the immunostained sections of the crushed ONs were counted.Results: We found that the surviving axon density correlated with crush force, with longer duration and stronger crush forces producing consistently more axon damage.Discussion: The instrumented tweezers enable a simple technique for measurement of ONC forces in real-time for the first time. Using the instrumented tweezers, experimenters can quantify crush forces during ONC to produce consistent and predictable post-crush cell death. This should permit future studies a way to produce nerve damage more consistently than is available now.
Collapse
Affiliation(s)
- Xiaorong Liu
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Liang Feng
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Ishan Shinde
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - James D Cole
- Neuroscience Graduate Program, Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - John B Troy
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Laxman Saggere
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
15
|
Algae Oil Treatment Protects Retinal Ganglion Cells (RGCs) via ERK Signaling Pathway in Experimental Optic Nerve Ischemia. Mar Drugs 2020; 18:md18020083. [PMID: 32012745 PMCID: PMC7074556 DOI: 10.3390/md18020083] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/17/2022] Open
Abstract
Background: We investigated the therapeutic effects and related mechanisms of algae oil (ALG) to protect retinal ganglion cells (RGCs) in a rat model of anterior ischemic optic neuropathy (rAION). Methods: Rats were daily gavaged with ALG after rAION induction for seven days. The therapeutic effects of ALG on rAION were evaluated using flash visual evoked potentials (FVEPs), retrograde labeling of RGCs, TUNEL assay of the retina, and ED1 staining of optic nerves (ONs). The levels of inducible nitric oxide synthase (iNOS), IL-1β, TNF-α, Cl-caspase-3, ciliary neurotrophic factor (CNTF), and p-ERK were analyzed by using western blots. Results: Protection of visual function in FVEPs amplitude was noted, with a better preservation of the P1–N2 amplitude in the ALG-treated group (p = 0.032) than in the rAION group. The density of RGCs was 2.4-fold higher in the ALG-treated group compared to that in the rAION group (p < 0.0001). The number of ED1-positive cells in ONs was significantly reduced 4.1-fold in the ALG-treated group compared to those in the rAION group (p = 0.029). The number of apoptotic RGCs was 3.2-fold lower in number in the ALG-treated group (p = 0.001) than that in the rAION group. The ALG treatment inhibited ERK activation to reduce the levels of iNOS, IL-1β, TNF-α, and Cl-caspase-3 and to increase the level of CNTF in the rAION model. Conclusion: The treatment with ALG after rAION induction inhibits ERK activation to provide both anti-inflammatory and antiapoptotic effects in rAION.
Collapse
|
16
|
Wen YT, Zhang JR, Kapupara K, Tsai RK. mTORC2 activation protects retinal ganglion cells via Akt signaling after autophagy induction in traumatic optic nerve injury. Exp Mol Med 2019; 51:1-11. [PMID: 31409770 PMCID: PMC6802655 DOI: 10.1038/s12276-019-0298-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/01/2019] [Accepted: 05/06/2019] [Indexed: 02/01/2023] Open
Abstract
Traumatic optic neuropathy is an injury to the optic nerve that leads to vision loss. Autophagy is vital for cell survival and cell death in central nervous system injury, but the role of autophagy in traumatic optic nerve injury remains uncertain. Optic nerve crush is a robust model of traumatic optic nerve injury. p62 siRNA and rapamycin are autophagy inducers and have different neuroprotective effects in the central nervous system. In this study, p62 and rapamycin induced autophagy, but only p62 siRNA treatment provided a favorable protective effect in visual function and retinal ganglion cell (RGC) survival. Moreover, the number of macrophages at the optic nerve lesion site was lower in the p62-siRNA-treated group than in the other groups. p62 siRNA induced more M2 macrophage polarization than rapamycin did. Rapamycin inhibited both mTORC1 and mTORC2 activation, whereas p62 siRNA inhibited only mTORC1 activation and maintained mTORC2 and Akt activation. Inhibition of mTORC2-induced Akt activation resulted in blood–optic nerve barrier disruption. Combined treatment with rapamycin and the mTORC2 activator SC79 improved RGC survival. Overall, our findings suggest that mTORC2 activation after autophagy induction is necessary for the neuroprotection of RGCs in traumatic optic nerve injury and may lead to new clinical applications. Regulating molecular signaling pathways that control the degradation of cellular components—a process known as autophagy—could offer a new approach to treating optic nerve damage after traumatic injuries. There is currently no established treatment option for traumatic optic nerve injury. Rong-Kung Tsai and colleagues at Tzu Chi University in Hualien, Taiwan, explored the role of a protein complex called mTORC2 in autophagy during the repair of optic nerves in rats. They investigated mTORC2 activation by small RNA molecules that also activate autophagy, and by drugs that activate autophagy but inhibit mTORC2. The results indicate that autophagy associated with activation of mTORC2 protects damaged retinal neurons and promotes visual recovery. In addition to treating optic nerve injuries, drugs activating mTORC2 and autophagy might help treat nerve-related diseases of the eye, including glaucoma.
Collapse
Affiliation(s)
- Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jia-Rong Zhang
- Department of Ophthalmology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kishan Kapupara
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan. .,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
17
|
Gupta R, Saha P, Sen T, Sen N. An augmentation in histone dimethylation at lysine nine residues elicits vision impairment following traumatic brain injury. Free Radic Biol Med 2019; 134:630-643. [PMID: 30790655 PMCID: PMC6588499 DOI: 10.1016/j.freeradbiomed.2019.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/16/2019] [Accepted: 02/13/2019] [Indexed: 12/26/2022]
Abstract
Traumatic Brain Injury (TBI) affects more than 1.7 million Americans each year and about 30% of TBI-patients having visual impairments. The loss of retinal ganglion cells (RGC) in the retina and axonal degeneration in the optic nerve have been attributed to vision impairment following TBI; however, the molecular mechanism has not been elucidated. Here we have shown that an increase in histone di-methylation at lysine 9 residue (H3K9Me2), synthesized by the catalytic activity of a histone methyltransferase, G9a is responsible for RGC loss and axonal degeneration in the optic nerve following TBI. To elucidate the molecular mechanism, we found that an increase in H3K9Me2 results in the induction of oxidative stress both in the RGC and optic nerve by decreasing the mRNA level of antioxidants such as Superoxide dismutase (sod) and catalase through impairing the transcriptional activity of Nuclear factor E2-related factor 2 (Nrf2) via direct interaction. The induction of oxidative stress is associated with death in RGC and oligodendrocyte precursor cells (OPCs). The death in OPCs is correlated with a reduction in myelination, and the expression of myelin binding protein (MBP) in association with degeneration of neurofilaments in the optic nerve. This event allied to an impairment of the retrograde transport of axons and loss of nerve fiber layer in the optic nerve following TBI. An administration of G9a inhibitor, UNC0638 attenuates the induction of H3K9Me2 both in RGC and optic nerve and subsequently activates Nrf2 to reduce oxidative stress. This event was concomitant with the rescue in the loss of retinal thickness, attenuation in optic nerve degeneration and improvement in the retrograde transport of axons following TBI.
Collapse
Affiliation(s)
- Rajaneesh Gupta
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, 15213, USA
| | - Pampa Saha
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, 15213, USA
| | - Tanusree Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, 15213, USA
| | - Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, 15213, USA.
| |
Collapse
|
18
|
Min JY, Lv Y, Mao L, Gong YY, Gu Q, Wei F. A rodent model of anterior ischemic optic neuropathy (AION) based on laser photoactivation of verteporfin. BMC Ophthalmol 2018; 18:304. [PMID: 30466418 PMCID: PMC6251118 DOI: 10.1186/s12886-018-0937-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 10/12/2018] [Indexed: 01/29/2023] Open
Abstract
Background A rodent model of photodynamic AION resulting from intravenous verteporfin is presented. The analysis of the morphological function, the pathological changes and the potential mechanism of action were further investigated. Methods Photodynamic treatment was conducted on the optic nerve head (ONH) following administration of the photosensitizer. The fellow eye was considered as sham control. Fundus Fluorescein angiography (FFA), spectral domain optical coherence tomography (SD-OCT) and Flash-visual evoked potential (F-VEP) recordings were conducted at different time points. Immunohistochemistry was used to observe apoptotic cell death (TUNEL) and macrophage infiltration (ED-1/Iba-1). Retrograde labeling of retinal ganglion cells (RGCs) was used to evaluate the loss of RGCs. Results After laser treatment, SD-OCT indicated optic nerve edema, while FFA indicated late leakage of the ONH. F-VEPs were distinctly reduced compared to control eyes. The number of apoptotic RGCs peaked on day 14 (5.71 ± 0.76, p < 0.01). The infiltration of ED-1 and Iba-1 increased on the 3rd day following PDT, while it peaked on day 14 (67.5 ± 9.57 and 77.5 ± 12.58 respectively, p < 0.01). Following 3 weeks of AION, the densities of RGCs in the central retinas of the normal and AION eyes were 3075 ± 298/mm2 and 2078 ± 141/mm2 (p < 0.01), respectively. Conclusions Verteporfin photodynamic treatment on rodents ONH can lead to functional, histological, and pathological changes. This type of animal model of AION is easy to establish and stable. It can be used for studying the mechanism and neuroprotective medicine of AION injury.
Collapse
Affiliation(s)
- Jing-Yu Min
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Yanan Lv
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Lei Mao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Yuan-Yuan Gong
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China. .,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China. .,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, NO.100, Haining Road, Hongkou District, Shanghai, 200080, China
| |
Collapse
|
19
|
Yin J, Zhang H, Chen H, Lv Q, Jin X. Hypertonic Saline Alleviates Brain Edema After Traumatic Brain Injury via Downregulation of Aquaporin 4 in Rats. Med Sci Monit 2018; 24:1863-1870. [PMID: 29600800 PMCID: PMC5890824 DOI: 10.12659/msm.907212] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Hypertonic saline (HS) has been successfully used for treatment of various forms of brain edema. Decreased expression of aquaporin (AQP)4 and pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β have been linked to edema pathogenesis. This study examined the effect of 3% HS on brain edema in a rat model of traumatic brain injury (TBI). Material/Methods Sprague-Dawley rats were subjected to TBI induced by a controlled cortical impactor. The HS group was injected with 3% NaCl until the end of the study period. AQP4, TNF-α, IL-1β, and caspase-3 levels were measured by Western blotting, immunohistochemistry, enzyme-linked immunosorbent assay, and quantitative real-time PCR. Brain water content was also measured. Apoptotic cells in brain tissue were detected with terminal deoxynucleotidyl transferase dUTP nick-end labeling. Brain water content decreased following treatment with 3% HS relative to the TBI group. Results This was accompanied by decreases in AQP4, TNF-α, and IL-1β mRNA and protein levels. TBI resulted in increases in caspase-3 mRNA expression and the number of apoptotic cells; treatment with 3% HS suppressed apoptosis as compared to the TBI group. Conclusions Treatment with 3% HS ameliorated TBI-induced brain edema, possibly by suppressing brain edema, pro-inflammatory cytokine expression, and apoptosis.
Collapse
Affiliation(s)
- Jian Yin
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Haixiao Zhang
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Huai Chen
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Qingping Lv
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xuhong Jin
- Department of Neurosurgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
20
|
Lin SF, Chien JY, Kapupara K, Huang CYF, Huang SP. Oroxylin A promotes retinal ganglion cell survival in a rat optic nerve crush model. PLoS One 2017. [PMID: 28640893 PMCID: PMC5480866 DOI: 10.1371/journal.pone.0178584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose To investigate the effect of oroxylin A on the survival of retinal ganglion cells (RGC) and the activation of microglial cells in a rat optic nerve (ON) crush model. Methods Oroxylin A (15mg/Kg in 0.2ml phosphate-buffered saline) or phosphate-buffered saline (PBS control) was immediately administered after ON crush once by subcutaneous injection. Rats were euthanized at 2 weeks after the crush injury. The density of RGC was counted by retrograde labeling with FluoroGold and immunostaining of retina flat mounts for Brn3a. Electrophysiological visual function was assessed by flash visual evoked potentials (FVEP). TUNEL assay, immunoblotting analysis of glial fibrillary acidic protein (GFAP), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in the retinas, and immunohistochemistry of GFAP in the retinas and ED1 in the ON were evaluated. Results Two weeks after the insult, the oroxylin A-treated group had significantly higher FG labeled cells and Brn3a+ cells suggesting preserved RGC density in the central and mid-peripheral retinas compared with those of the PBS-treated group. FVEP measurements showed a significantly better preserved latency of the P1 wave in the ON-crushed, oroxylin A-treated rats than the ON-crushed, PBS treated rats. TUNEL assays showed fewer TUNEL positive cells in the ON-crushed, oroxylin A-treated rats. The number of ED1 positive cells was reduced at the lesion site of the optic nerve in the ON-crushed, oroxylin A-treated group. Increased GFAP expression in the retina was reduced greatly in ON-crushed, oroxylin A-treated group. Furthermore, administration of oroxylin A significantly attenuated ON crush insult-induced iNOS and COX-2 expression in the retinas. Conclusions These results demonstrated that oroxylin A hasss neuroprotective effects on RGC survival with preserved visual function and a decrease in microglial infiltration in the ONs after ON crush injury.
Collapse
Affiliation(s)
- Shu-Fang Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jia-Ying Chien
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
- Institute of systems neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Kishan Kapupara
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Chi-Ying F. Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (CYFH); (SPH)
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
- * E-mail: (CYFH); (SPH)
| |
Collapse
|
21
|
Falavarjani KG, Habibi A, Pourhabibi A, Hosseini SB, Modarres M, Parvaresh MM, Shahriari-Ahmadi A. Retinal Toxicity of Intravitreal Granulocyte Colony-Stimulating Factor in Rabbit Eyes. Curr Eye Res 2016; 42:636-639. [DOI: 10.1080/02713683.2016.1227447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Khalil Ghasemi Falavarjani
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Habibi
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Pourhabibi
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Modarres
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Parvaresh
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shahriari-Ahmadi
- Department of Internal Medicine, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Wen YT, Huang TL, Huang SP, Chang CH, Tsai RK. Early applications of granulocyte colony-stimulating factor (G-CSF) can stabilize the blood-optic-nerve barrier and ameliorate inflammation in a rat model of anterior ischemic optic neuropathy (rAION). Dis Model Mech 2016; 9:1193-1202. [PMID: 27538969 PMCID: PMC5087822 DOI: 10.1242/dmm.025999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/11/2016] [Indexed: 12/27/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) was reported to have a neuroprotective effect in a rat model of anterior ischemic optic neuropathy (rAION model). However, the therapeutic window and anti-inflammatory effects of G-CSF in a rAION model have yet to be elucidated. Thus, this study aimed to determine the therapeutic window of G-CSF and investigate the mechanisms of G-CSF via regulation of optic nerve (ON) inflammation in a rAION model. Rats were treated with G-CSF on day 0, 1, 2 or 7 post-rAION induction for 5 consecutive days, and a control group were treated with phosphate-buffered saline (PBS). Visual function was assessed by flash visual evoked potentials at 4 weeks post-rAION induction. The survival rate and apoptosis of retinal ganglion cells were determined by FluoroGold labeling and TUNEL assay, respectively. ON inflammation was evaluated by staining of ED1 and Iba1, and ON vascular permeability was determined by Evans Blue extravasation. The type of macrophage polarization was evaluated using quantitative real-time PCR (qRT-PCR). The protein levels of TNF-α and IL-1β were analyzed by western blotting. A therapeutic window during which G-CSF could rescue visual function and retinal ganglion cell survival was demonstrated at day 0 and day 1 post-infarct. Macrophage infiltration was reduced by 3.1- and 1.6-fold by G-CSF treatment starting on day 0 and 1 post-rAION induction, respectively, compared with the PBS-treated group (P<0.05). This was compatible with 3.3- and 1.7-fold reductions in ON vascular permeability after G-CSF treatment compared with PBS treatment (P<0.05). Microglial activation was increased by 3.8- and 3.2-fold in the early (beginning treatment at day 0 or 1) G-CSF-treated group compared with the PBS-treated group (P<0.05). Immediate (within 30 mins of infarct) treatment with G-CSF also induced M2 microglia/macrophage activation. The cytokine levels were lower in the group that received immediate G-CSF treatment compared to those in the later G-CSF treatment group (P<0.05). Early treatment with G-CSF stabilized the blood-ON barrier to reduce macrophage infiltration and induced M2 microglia/macrophage polarization to decrease the expressions of pro-inflammatory cytokines in this rAION model.
Collapse
Affiliation(s)
- Yao-Tseng Wen
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Tzu-Lun Huang
- Department of Ophthalmology, Far Eastern Memorial Hospital, Banciao District, New Taipei City, Taiwan Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sung-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Chung-Hsing Chang
- Department of Dermatology, China Medical University Hospital, Taichung, Taiwan Department of Dermatology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
23
|
Wang J, Chen S, Zhang X, Huang W, Jonas JB. Intravitreal triamcinolone acetonide, retinal microglia and retinal ganglion cell apoptosis in the optic nerve crush model. Acta Ophthalmol 2016; 94:e305-11. [PMID: 25708663 DOI: 10.1111/aos.12698] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 01/12/2015] [Indexed: 01/20/2023]
Abstract
PURPOSE To evaluate the effect of intravitreal triamcinolone acetonide (TA) on the activation of retinal microglia cells (RMGCs) and survival of retinal ganglion cells (RGCs) in an optic nerve crush (ONC) model. METHODS Adult female Sprague-Dawley rats underwent a standardized ONC and either received an intravitreal injection of TA (TA group) or of phosphate-buffered saline (PBS, PBS group) in the right eyes. At 1, 3, 7, 14 and 28 days after the ONC, the animals were killed. The retinas were examined by immunohistochemistry, light microscopy, Western blot or retrograde labelling of RGCs by fluorogold injected into the superior colliculi. RESULTS The TA group as compared to the PBS control group showed a significantly (p < 0.0001) lower density of activated RMGCs, at 14 days [4.2 ± 1.6 versus 9.3 ± 2.2 cells/high-power microscopic field (HPF)] and at 28 days (2.3 ± 1.1 versus 4.4 ± 1.5 cells/HPF), and with a significantly lower expression of inflammatory factors. Central density of RGCs as stained by haematoxylin-eosin or by fluorogold was significantly (all p < 0.05) more reduced in the PBS group than in the TA group at days 14 and 28 after baseline. The survival rate (cell density in the study eye as compared to cell density in the contralateral unaffected eye) was significantly higher in the TA group than in the PBS group on days 14 (58% versus 45%; p = 0.003) and 28 (52% versus 41%; p = 0.022). CONCLUSIONS Intravitreal TA as compared to intravitreal PBS was associated with a lower density of activated RMGCs and a higher density of surviving RGCs in an ONC model.
Collapse
Affiliation(s)
- Jiawei Wang
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-Sen University; Guangzhou China
| | - Shida Chen
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-Sen University; Guangzhou China
| | - Xiulan Zhang
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-Sen University; Guangzhou China
| | - Wenbin Huang
- State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-Sen University; Guangzhou China
| | - Jost B. Jonas
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University; Heidelberg Germany
| |
Collapse
|
24
|
Autocrine protective mechanisms of human granulocyte colony-stimulating factor (G-CSF) on retinal ganglion cells after optic nerve crush. Exp Eye Res 2016; 143:132-40. [DOI: 10.1016/j.exer.2015.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/02/2015] [Accepted: 10/07/2015] [Indexed: 01/07/2023]
|
25
|
Retinal Cell Degeneration in Animal Models. Int J Mol Sci 2016; 17:ijms17010110. [PMID: 26784179 PMCID: PMC4730351 DOI: 10.3390/ijms17010110] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/25/2015] [Accepted: 01/08/2016] [Indexed: 01/01/2023] Open
Abstract
The aim of this review is to provide an overview of various retinal cell degeneration models in animal induced by chemicals (N-methyl-d-aspartate- and CoCl2-induced), autoimmune (experimental autoimmune encephalomyelitis), mechanical stress (optic nerve crush-induced, light-induced) and ischemia (transient retinal ischemia-induced). The target regions, pathology and proposed mechanism of each model are described in a comparative fashion. Animal models of retinal cell degeneration provide insight into the underlying mechanisms of the disease, and will facilitate the development of novel effective therapeutic drugs to treat retinal cell damage.
Collapse
|
26
|
Chien JY, Sheu JH, Wen ZH, Tsai RK, Huang SP. Neuroprotective effect of 4-(Phenylsulfanyl)butan-2-one on optic nerve crush model in rats. Exp Eye Res 2015; 143:148-57. [PMID: 26472213 DOI: 10.1016/j.exer.2015.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 08/18/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022]
Abstract
This study is to investigate the effect of coral-related compound, 4-(phenylsulfanyl)butan-2-one (4-PSB-2) on optic nerves (ON) and retinal ganglion cells (RGC) in a rat model subjected to ON crush. The ONs of adult male Wistar rat (150-180 g) were crushed by a standardized method. The control eyes received a sham operation. 4-PSB-2 (5 mg/kg in 0.2 mL phosphate-buffered saline) or phosphate-buffered saline (PBS control) was immediately administered after ON crush once by subcutaneous injection. Rats were euthanized at 2 weeks after the crush injury. RGC density was counted by retrograde labeling with FluoroGold (FG) application to the superior colliculus, and visual function was assessed by flash visual evoked potentials (FVEP). TUNEL assay, immunoblotting analysis of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) in the retinas, and immunohistochemistry of ED1 in the ON were evaluated. Two weeks after the insult, the RGC densities in the central and mid-peripheral retinas in ON-crushed, 4-PSB-2-treated rats were significantly higher than that of the corresponding ON-crushed, PBS-treated rats FVEP measurements showed a significantly better preserved latency of the P1 wave in the ON-crushed, 4-PSB-2-treated rats than the ON-crushed, PBS treated rats. TUNEL assays showed fewer TUNEL positive cells in the ON-crushed, 4-PSB-2-treated rats. The number of ED1 positive cells was reduced at the lesion site of the optic nerve in the ON-crushed, 4-PSB-2-treated group. Furthermore, administration of 4-PSB-2 significantly attenuated ON crush insult-stimulated iNOS and COX2 expression in the retinas. These results demonstrated that 4-PSB-2 protects RGCs and helps preserve the visual function in the rat model of optic nerve crush. 4-PSB-2 may work by being anti-apoptotic and by attenuation of the inflammatory responses involving less ED1 positive cells infiltration in ON as well as suppression of iNOS/COX-2 signaling pathway in the retinas to rescue RGCs after ON crush injury.
Collapse
Affiliation(s)
- Jia-Ying Chien
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
27
|
Huang SP, Chien JY, Tsai RK. Ethambutol induces impaired autophagic flux and apoptosis in the rat retina. Dis Model Mech 2015; 8:977-87. [PMID: 26092127 PMCID: PMC4527287 DOI: 10.1242/dmm.019737] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/07/2015] [Indexed: 12/11/2022] Open
Abstract
Ethambutol (EMB), an effective first-line antituberculosis agent, can cause serious visual impairment or irreversible vision loss in a significant number of patients. However, the mechanism underlying this ocular cytotoxicity remains to be elucidated. In this study, we found that there were statistically significant dose- and time-dependent increases in the number of cytoplasmic vacuoles and the level of cell death in EMB-treated RGC-5 cells (retinal ganglion cells). The protein kinase C (PKC)δ inhibitor rottlerin markedly reduced the EMB-induced activation of caspase-3 and the subsequent apoptosis of RGC-5 cells. Western blot analysis revealed that the expression levels of class III PI3K, Beclin-1, p62 and LC3-II were upregulated, and LC3 immunostaining results showed activation of the early phase and inhibition of the late stage of autophagy in retinas of the EMB-intraperitoneal (IP)-injected rat model. We further demonstrated that exposure to EMB induces autophagosome accumulation, which results from the impaired autophagic flux that is mediated by a PKCδ-dependent pathway, inhibits the PI3K/Akt/mTOR signaling pathway and leads to apoptotic death in retina neuronal cells. These results indicate that autophagy dysregulation in retinal neuronal cells might play a substantial role in EMB-induced optic neuroretinopathy. Summary: This study provides the first evidence that EMB induces autophagosome accumulation, which results from the impaired autophagic flux that is mediated by a PKCδ-dependent pathway, and leads to apoptotic death in retina neuronal cells.
Collapse
Affiliation(s)
- Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97002, Taiwan
| | - Jia-Ying Chien
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97002, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien 97002, Taiwan Institute of Medical Sciences, Tzu Chi University, Hualien 97002, Taiwan
| |
Collapse
|
28
|
Protective effects of systemic treatment with methylprednisolone in a rodent model of non-arteritic anterior ischemic optic neuropathy (rAION). Exp Eye Res 2014; 131:69-76. [PMID: 25543054 DOI: 10.1016/j.exer.2014.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/14/2014] [Accepted: 12/23/2014] [Indexed: 11/22/2022]
Abstract
This study investigated the protective effects of the administration of steroids on optic nerves (ON) and retinal ganglion cells (RGCs) in a rodent model of non-arteritic anterior ischemic optic neuropathy (rAION). We induced rAION using rose bengal and argon laser irradiation in a photodynamic procedure on the optic discs of rats. The treated groups received methylprednisolone (MP) via peritoneal injection for 2 weeks. The control group received intraperitoneal injections of phosphate-buffered saline (PBS) post-rAION. At the 4th week post-infarct, MP treatments significantly rescued the RGCs (mm(2)) in the central retinas (1920 ± 210, p < 0.001) and mid-peripheral retinas (950 ± 240, respectively, p = 0.018) compared with those of the PBS-treated rats (central: 900 ± 210 and mid-peripheral: 440 ± 180). Functional assessment with flash visual-evoked potentials demonstrated that P1 latency (ms) was shortened in the MP group compared to the PBS group (108 ± 14 and 147 ± 9, respectively, p < 0.001). In addition, the P1 amplitude (uV) was enhanced in the MP group compared to the PBS group (55 ± 12 and 41 ± 13, respectively, p < 0.05). TUNEL assays showed a decrease in the number of apoptotic cells in the RGC layers of MP-treated retinas compared to the PBS-treated group (p < 0.05). ED1 positive cells (/HPF) were significantly decreased in the ONs of the MP group compared to the PBS group (p < 0.001). In conclusion, systemic administration of MP had neuroprotective effects on RGC survival and ON function in the rAION animal model.
Collapse
|
29
|
Huang SP, Tsai RK. Efficacy of granulocyte-colony stimulating factor treatment in a rat model of anterior ischemic optic neuropathy. Neural Regen Res 2014; 9:1502-5. [PMID: 25317164 PMCID: PMC4192964 DOI: 10.4103/1673-5374.139472] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2014] [Indexed: 01/12/2023] Open
Abstract
Non-arteritic anterior ischemic optic neuropathy (NA-AION) is the most common cause of acute ischemic damage to the optic nerve (ON), and the leading cause of seriously impaired vision in people over 55 years of age. It demonstrated that subcutaneous administration of Granulocyte colony-stimulating factor (G-CSF) reduces RGC death in an ON crush model in rats, and that the neuroprotective effects may involve both anti-apoptotic and anti-inflammatory processes. Our recent work shows that the protective actions of G-CSF in rAION models may involve both anti-apoptotic and anti-inflammatory processes. However, the exact rescuing mechanisms involved in the administration of G-CSF in rAION models need further investigation. In addition, further studies on the administration of G-CSF at different time intervals after the induction of rAION may be able to illustrate whether treatment given at a later time is still neuroprotective. Further, it is unknown whether treatment using G-CSF combined with other drugs will result in a synergistic effect in a rAION model. Inflammation induced by ischemia plays an essential role on the ON head in NA-AION, which can result in disc edema and compartment changes. Therefore, it is reasonable that adding an anti-inflammatory drug may enhance the therapeutic effects of G-CSF. An ongoing goal is to evaluate the novel sites of action of both G-CSF and other anti-inflammatory drugs, and to identify the functionally protective pathways to enhance RGC survival. These investigations may open up new therapeutic avenues for the treatment of ischemic optic neuropathy.
Collapse
Affiliation(s)
- Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97002, Taiwan, China
| | - Rong-Kung Tsai
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien 97002, Taiwan, China ; Institute of Medical Sciences, Tzu Chi University, Hualien 97002, Taiwan, China
| |
Collapse
|
30
|
Nicholson JD, Leiba H, Goldenberg-Cohen N. Translational Preclinical Research may Lead to Improved Medical Management of Non-Arteritic Anterior Ischemic Optic Neuropathy. Front Neurol 2014; 5:122. [PMID: 25071709 PMCID: PMC4092366 DOI: 10.3389/fneur.2014.00122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/25/2014] [Indexed: 12/23/2022] Open
Affiliation(s)
- James D Nicholson
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center , Petach Tikva , Israel ; Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Hana Leiba
- Department of Ophthalmology, Kaplan Medical Center , Rehovot , Israel ; Hebrew University Hadassah Medical Center , Jerusalem , Israel
| | - Nitza Goldenberg-Cohen
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center , Petach Tikva , Israel ; Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv , Israel ; Pediatric Ophthalmology Unit, Schneider Children's Medical Center of Israel , Petach Tikva , Israel
| |
Collapse
|
31
|
Huang TL, Huang SP, Chang CH, Lin KH, Sheu MM, Tsai RK. Protective effects of cerebrolysin in a rat model of optic nerve crush. Kaohsiung J Med Sci 2014; 30:331-6. [DOI: 10.1016/j.kjms.2014.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/21/2014] [Accepted: 01/10/2014] [Indexed: 11/16/2022] Open
|
32
|
Huang TL, Huang SP, Chang CH, Lin KH, Sheu MM, Tsai RK. Factors influencing the retrograde labeling of retinal ganglion cells with fluorogold in an animal optic nerve crush model. Ophthalmic Res 2014; 51:173-8. [PMID: 24662310 DOI: 10.1159/000357736] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/04/2013] [Indexed: 01/21/2023]
Abstract
PURPOSE To investigate whether different crush durations or a different fluorogold (FG) injection timing can affect the efficiency of FG retrograde labeling of retinal ganglion cells (RGCs) in the optic nerve (ON) crush model. METHODS We performed the ON crush in rats with a clip at different durations or a jewel forceps to compare the effects of different crush methods with FG staining. RGC density was compared between the FG injection 1 week before the sacrifice of the animals (group A) and the injection before the crush experiment (group B). Double staining with CD11b and FG in the retinal sections was conducted to investigate the relationship between the overcounting of RGCs and microglia. RESULTS The FG-stained particles were significantly decreased at the distal part of the crush site compared to the proximal site of the ON with a crush duration of over 30 s or when crushed with the jewel forceps. Two weeks after ON crush, the RGC count was higher both in the central and mid-peripheral retinas in group B. The percentage of CD11b-stained cells among the FG-stained cells in the RGC layer of retinas in group B was higher than that of group A (34% in group B vs. 4% in group A, p = 0.0001). Overcounting of RGC density in group B was due to additional microglia with FG engulfing. CONCLUSIONS Our results suggest that each laboratory should test its setting conditions to avoid factors influencing the RGC density measurement before conducting ON crush experiments.
Collapse
Affiliation(s)
- Tzu-Lun Huang
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Tzu Chi University, Hualien, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
33
|
Chang CH, Huang TL, Huang SP, Tsai RK. Neuroprotective effects of recombinant human granulocyte colony-stimulating factor (G-CSF) in a rat model of anterior ischemic optic neuropathy (rAION). Exp Eye Res 2013; 118:109-16. [PMID: 24316388 DOI: 10.1016/j.exer.2013.11.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/18/2013] [Accepted: 11/21/2013] [Indexed: 01/22/2023]
Abstract
The purpose of this study was to investigate the neuroprotective effects of recombinant human granulocyte colony stimulating factor (G-CSF), as administered in a rat model of anterior ischemic optic neuropathy (rAION). Using laser-induced photoactivation of intravenously administered Rose Bengal in the optic nerve head of 60 adult male Wistar rats, an anterior ischemic optic neuropathy (rAION) was inducted. Rats either immediately received G-CSF (subcutaneous injections) or phosphate buffered saline (PBS) for 5 consecutive days. Rats were euthanized at 4 weeks post infarct. Density of retinal ganglion cells (RGCs) was counted using retrograde labeling of Fluoro-gold. Visual function was assessed by flash visual-evoked potentials (FVEP) at 4 weeks. TUNEL assay in the retinal sections and immunohistochemical staining of ED1 (marker of macrophage/microglia) were investigated in the optic nerve (ON) specimens. The RGC densities in the central and mid-peripheral retinas in the G-CSF treated rats were significantly higher than those of the PBS-treated rats (survival rate was 71.4% vs. 33.2% in the central retina; 61.8% vs. 22.7% in the mid-peripheral retina, respectively; both p < 0.05). FVEP measurements showed a significantly better preserved latency and amplitude of the p1 wave in the G-CSF-treated rats than that of the PBS-treated rats (latency120 ± 11 ms vs. 142 ± 12 ms, p = 0.03; amplitude 50 ± 11 μv vs. 31 ± 13 μv, p = 0.04). TUNEL assays showed fewer apoptotic cells in the retinal ganglion cell layers of G-CSF treated rats [2.1 ± 1.0 cells/high power field (HPF) vs. 8.0 ± 1.5/HPF; p = 0.0001]. In addition, the number of ED1 positive cells was attenuated at the optic nerve sections of G-CSF-treated rats (16 ± 6/HPF vs. 35 ± 10/HPF; p = 0.016). In conclusion, administration of G-CSF is neuroprotective in the rat model of anterior ischemic optic neuropathy, as demonstrated both structurally by RGC density and functionally by FVEP. G-CSF may work via the dual actions of anti-apoptosis for RGC surviving as well as anti-inflammation in the optic nerves as evidenced by less infiltration of ED1-povitive cells.
Collapse
Affiliation(s)
- Chung-Hsing Chang
- Department of Dermatology, Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Lun Huang
- Department of Ophthalmology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Department of Ophthalmology and Visual Science, Tzu Chi University, Hualien, Taiwan
| | - Shun-Ping Huang
- Department of Ophthalmology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Rong-Kung Tsai
- Department of Ophthalmology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Department of Ophthalmology and Visual Science, Tzu Chi University, Hualien, Taiwan; Department of Ophthalmology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
34
|
Avraham-Lubin BCR, Goldenberg-Cohen N, Sadikov T, Askenasy N. VEGF induces neuroglial differentiation in bone marrow-derived stem cells and promotes microglia conversion following mobilization with GM-CSF. Stem Cell Rev Rep 2013; 8:1199-210. [PMID: 22810360 DOI: 10.1007/s12015-012-9396-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE Evaluation of potential tropic effects of vascular endothelial growth factor (VEGF) on the incorporation and differentiation of bone-marrow-derived stem cells (BMSCs) in a murine model of anterior ischemic optic neuropathy (AION). METHODS In the first approach, small-sized subset of BMCs were isolated from GFP donors mice by counterflow centrifugal elutriation and depleted of hematopoietic lineages (Fr25lin(-)). These cells were injected into a peripheral vein (1 × 10(6) in 0.2 ml) or inoculated intravitreally (2 × 10(5)) to syngeneic mice, with or without intravitreal injection of 5 μg/2μL VEGF, simultaneously with AION induction. In a second approach, hematopoietic cells were substituted by myelablative transplant of syngeseic GFP + bone marrow cells. After 3 months, progenitors were mobilized with granulocyte-macrophage colony-stimulating factor (GM-CSF) followed by VEGF inoculation into the vitreous body and AION induction . Engraftment and phenotype were examined by immunohistochemistry and FISH at 4 and 24 weeks post-transplantation, and VEGF receptors were determined by real time PCR. RESULTS VEGF had no quantitative effect on incorporation of elutriated cells in the injured retina, yet it induced early expression of neuroal markers in cells incorporated in the RGC layer and promoted durable gliosis, most prominent perivascular astrocytes. These effects were mediated by VEGF-R1/Flt-1, which is constitutively expresses in the elutriated fraction of stem cells. Mobilization with GM-CSF limited the differentiation of bone marrow progenitors to microglia, which was also fostered by VEGF. CONCLUSIONS VEGF signaling mediated by Flt-1 induces early neural and sustained astrocytic differentiation of stem cells elutriated from adult bone-marrow, with significant contribution to stabilization retinal architecture following ischemic injury.
Collapse
Affiliation(s)
- Bat-Chen R Avraham-Lubin
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Tel Aviv University, Petach Tikva, Israel
| | | | | | | |
Collapse
|
35
|
Wang Y, Brown DP, Duan Y, Kong W, Watson BD, Goldberg JL. A novel rodent model of posterior ischemic optic neuropathy. JAMA Ophthalmol 2013; 131:194-204. [PMID: 23544206 DOI: 10.1001/2013.jamaophthalmol.271] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVES To develop a reliable, reproducible rat model of posterior ischemic optic neuropathy (PION) and study the cellular responses in the optic nerve and retina. METHODS Posterior ischemic optic neuropathy was induced in adult rats by photochemically induced ischemia. Retinal and optic nerve vasculature was examined by fluorescein isothiocyanate–dextran extravasation. Tissue sectioning and immunohistochemistry were used to investigate the pathologic changes. Retinal ganglion cell survival at different times after PION induction, with or without neurotrophic application, was quantified by fluorogold retrograde labeling. RESULTS Optic nerve injury was confirmed after PION induction, including local vascular leakage, optic nerve edema, and cavernous degeneration. Immunostaining data revealed microglial activation and focal loss of astrocytes, with adjacent astrocytic hypertrophy. Up to 23%, 50%, and 70% retinal ganglion cell loss was observed at 1 week, 2 weeks, and 3 weeks, respectively, after injury compared with a sham control group. Experimental treatment by brain-derived neurotrophic factor and ciliary neurotrophic factor remarkably prevented retinal ganglion cell loss in PION rats. At 3 weeks after injury, more than 40% of retinal ganglion cells were saved by the application of neurotrophic factors. CONCLUSIONS Rat PION created by photochemically induced ischemia is a reproducible and reliable animal model for mimicking the key features of human PION. CLINICAL RELEVANCE The correspondence between the features of this rat PION model to those of human PION makes it an ideal model to study the pathophysiologic course of the disease, most of which remains to be elucidated. Furthermore, it provides an optimal model for testing therapeutic approaches for optic neuropathies.
Collapse
Affiliation(s)
- Yan Wang
- Department of Ophthalmology and Vision Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Cytokines are important mediators of host defense and immunity, and were first identified for their role in immunity to infections. It was then found that some of them are pathogenic mediators in inflammatory diseases and much of the emphasis is now on pro-inflammatory cytokines, also in consideration of the fact that TNF inhibitors became effective drugs in chronic inflammatory diseases. The recent studies on the tissue-protective activities of erythropoietin (EPO) led to the term "tissue-protective cytokine." We discuss here how tissue-protective actions might be common to other cytokines, particularly those of the 4-alpha helical structural superfamily.
Collapse
|
37
|
Pereira Lopes FR, Martin PKM, Frattini F, Biancalana A, Almeida FM, Tomaz MA, Melo PA, Borojevic R, Han SW, Martinez AMB. Double gene therapy with granulocyte colony-stimulating factor and vascular endothelial growth factor acts synergistically to improve nerve regeneration and functional outcome after sciatic nerve injury in mice. Neuroscience 2012; 230:184-97. [PMID: 23103791 DOI: 10.1016/j.neuroscience.2012.10.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 12/17/2022]
Abstract
Peripheral-nerve injuries are a common clinical problem and often result in long-term functional deficits. Reconstruction of peripheral-nerve defects is currently undertaken with nerve autografts. However, there is a limited availability of nerves that can be sacrificed and the functional recovery is never 100% satisfactory. We have previously shown that gene therapy with vascular endothelial growth factor (VEGF) significantly improved nerve regeneration, neuronal survival, and muscle activity. Our hypothesis is that granulocyte colony-stimulating factor (G-CSF) synergizes with VEGF to improve the functional outcome after sciatic nerve transection. The left sciatic nerves and the adjacent muscle groups of adult mice were exposed, and 50 or 100 μg (in 50 μl PBS) of VEGF and/or G-CSF genes was injected locally, just below the sciatic nerve, and transferred by electroporation. The sciatic nerves were transected and placed in an empty polycaprolactone (PCL) nerve guide, leaving a 3-mm gap to challenge nerve regeneration. After 6 weeks, the mice were perfused and the sciatic nerve, the dorsal root ganglion (DRG), the spinal cord and the gastrocnemius muscle were processed for light and transmission electron microscopy. Treated animals showed significant improvement in functional and histological analyses compared with the control group. However, the best results were obtained with the G-CSF+VEGF-treated animals: quantitative analysis of regenerated nerves showed a significant increase in the number of myelinated fibers and blood vessels, and the number of neurons in the DRG and motoneurons in the spinal cord was significantly higher. Motor function also showed that functional recovery occurred earlier in animals receiving G-CSF+VEGF-treatment. The gastrocnemius muscle showed an increase in weight and in the levels of creatine phosphokinase, suggesting an improvement of reinnervation and muscle activity. These results suggest that these two factors acted synergistically and optimized the nerve repair potential, improving regeneration after a transection lesion.
Collapse
Affiliation(s)
- F R Pereira Lopes
- Programa de Neurociência Básica e Clínica, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fang TC, Pang CY, Chiu SC, Ding DC, Tsai RK. Renoprotective effect of human umbilical cord-derived mesenchymal stem cells in immunodeficient mice suffering from acute kidney injury. PLoS One 2012; 7:e46504. [PMID: 23029541 PMCID: PMC3459926 DOI: 10.1371/journal.pone.0046504] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 09/03/2012] [Indexed: 11/19/2022] Open
Abstract
It is unknown whether human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) can improve the renal function of patients suffering from acute kidney injury. Moreover, before beginning clinical trials, it is necessary to investigate this renoprotective effect of hUC-MSCs in a xenogeneic model of acute kidney injury. However, no previous studies have examined the application of hUC-MSCs to immunodeficient mice suffering from acute kidney injury. The objectives of this study were to examine whether hUC-MSCs could improve renal function in nonobese diabetic-severe combined immune deficiency (NOD-SCID) mice suffering from acute kidney injury, and to investigate the mechanism(s) for hUC-MSCs to improve renal function in this xenogeneic model. Early (3 hr) and late (12 hr) administrations of hUC-MSCs (106 cells) were performed via the external jugular vein into NOD-SCID mice suffering from either folic acid (FA) (250 mg/kg body weight) or vehicle. The results showed that early administration of hUC-MSCs improved the renal function of NOD-SCID mice suffering from FA-induced acute kidney injury, as evidenced by decreased serum urea nitrogen and serum creatinine levels, as well as a reduced tubular injury score. The beneficial effects of hUC-MSCs were through reducing apoptosis and promoting proliferation of renal tubular cells. These benefits were independent of inflammatory cytokine effects and transdifferentiation. Furthermore, this study is the first one to show that the reduced apoptosis of renal tubular cells by hUC-MSCs in this xenogeneic model is mediated through the mitochondrial pathway, and through the increase of Akt phosphorylation.
Collapse
Affiliation(s)
- Te-Chao Fang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| | | | | | | | | |
Collapse
|
39
|
Chao PK, Lu KT, Lee YL, Chen JC, Wang HL, Yang YL, Cheng MY, Liao MF, Ro LS. Early systemic granulocyte-colony stimulating factor treatment attenuates neuropathic pain after peripheral nerve injury. PLoS One 2012; 7:e43680. [PMID: 22937076 PMCID: PMC3427178 DOI: 10.1371/journal.pone.0043680] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 07/24/2012] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that opioid treatment can reduce pro-inflammatory cytokine production and counteract various neuropathic pain syndromes. Granulocyte colony-stimulating factor (G-CSF) can promote immune cell differentiation by increasing leukocytes (mainly opioid-containing polymorphonuclear (PMN) cells), suggesting a potential beneficial role in treating chronic pain. This study shows the effectiveness of exogenous G-CSF treatment (200 µg/kg) for alleviating thermal hyperalgesia and mechanical allodynia in rats with chronic constriction injury (CCI), during post-operative days 1-25, compared to that of vehicle treatment. G-CSF also increases the recruitment of opioid-containing PMN cells into the injured nerve. After CCI, single administration of G-CSF on days 0, 1, and 2, but not on day 3, relieved thermal hyperalgesia, which indicated that its effect on neuropathic pain had a therapeutic window of 0-48 h after nerve injury. CCI led to an increase in the levels of interleukin-6 (IL-6) mRNA and tumor necrosis factor-α (TNF-α) protein in the dorsal root ganglia (DRG). These high levels of IL-6 mRNA and TNF-α were suppressed by a single administration of G-CSF 48-144 h and 72-144 h after CCI, respectively. Furthermore, G-CSF administered 72-144 h after CCI suppressed the CCI-induced upregulation of microglial activation in the ipsilateral spinal dorsal horn, which is essential for sensing neuropathic pain. Moreover, the opioid receptor antagonist naloxone methiodide (NLXM) reversed G-CSF-induced antinociception 3 days after CCI, suggesting that G-CSF alleviates hyperalgesia via opioid/opioid receptor interactions. These results suggest that an early single systemic injection of G-CSF alleviates neuropathic pain via activation of PMN cell-derived endogenous opioid secretion to activate opioid receptors in the injured nerve, downregulate IL-6 and TNF-α inflammatory cytokines, and attenuate microglial activation in the spinal dorsal horn. This indicates that G-CSF treatment can suppress early inflammation and prevent the subsequent development of neuropathic pain.
Collapse
Affiliation(s)
- Po-Kuan Chao
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Kwok-Tung Lu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Yun-Lin Lee
- Division of Neuromuscular Disorders, Department of Neurology, Chang Gung Memorial Hospital and University, Chang-Gung University, Tao-Yuan, Taiwan
| | - Jin-Chung Chen
- Department of Pharmacology, Chang-Gung University, Tao-Yuan, Taiwan
| | - Hung-Li Wang
- Department of Physiology, Chang-Gung University, Tao-Yuan, Taiwan
| | - Yi-Ling Yang
- Institute of Biotechnology, National Chia-Yi University, Chia-Yi, Taiwan
| | - Mei-Yun Cheng
- Division of Neuromuscular Disorders, Department of Neurology, Chang Gung Memorial Hospital and University, Chang-Gung University, Tao-Yuan, Taiwan
| | - Ming-Feng Liao
- Division of Neuromuscular Disorders, Department of Neurology, Chang Gung Memorial Hospital and University, Chang-Gung University, Tao-Yuan, Taiwan
| | - Long-Sun Ro
- Division of Neuromuscular Disorders, Department of Neurology, Chang Gung Memorial Hospital and University, Chang-Gung University, Tao-Yuan, Taiwan
- * E-mail:
| |
Collapse
|
40
|
Nakatani M, Shinohara Y, Takii M, Mori H, Asai N, Nishimura S, Furukawa-Hibi Y, Miyamoto Y, Nitta A. Periocular injection of in situ hydrogels containing Leu-Ile, an inducer for neurotrophic factors, promotes retinal ganglion cell survival after optic nerve injury. Exp Eye Res 2011; 93:873-9. [PMID: 22001716 DOI: 10.1016/j.exer.2011.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 09/08/2011] [Accepted: 09/30/2011] [Indexed: 12/13/2022]
Abstract
Intraocular administration of neurotrophic factors has been shown to delay irreversible degeneration of retinal ganglion cells (RGCs). It would be beneficial for the treatment of optic nerve (ON) injury if such neurotrophic factors could be delivered in a less-invasive manner. The dipeptide leucine-isoleucine (Leu-Ile) appears to induce the production of neurotrophic factors, including brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF), in the brain. We therefore administered Leu-Ile via periocular depot injection in rats and investigated the dipeptide's ability to induce BDNF and GDNF in the retina and to delay RGC loss in an ON injury model. Poloxamer-alginate hydrogels containing Leu-Ile were injected into the subconjunctival space of intact or ON-injured rats. BDNF and GDNF levels in the retina were determined by an enzyme immunoassay. Survival of RGCs was assessed in retinal flatmounts. Activation of extracellular signal-regulated kinases (ERK) and cAMP response element binding protein (CREB) in the retina was examined by Western blotting. At 2 h after injection of fluorescein isothiocyanate-conjugated Leu-Ile, the fluorescence intensities in the retina were 4.3-fold higher than those in the saline control. Treatment with Leu-Ile significantly increased the retinal levels of BDNF at 6 h and GDNF at 6-72 h after injection. Treatment with Leu-Ile significantly increased RGC survival to 14 days after ON injury and enhanced the activation of ERK at 72 h and CREB at 48 h after injection in the ON-injured retina. These results suggest that periocular delivery of Leu-Ile induces BDNF and GDNF production in the retina, which may eventually enhance RGC survival after ON injury.
Collapse
Affiliation(s)
- Masayoshi Nakatani
- Bioengineering Institute, Research & Development Division, Nidek Co., Ltd., 13-2 Hama-cho, Gamagori, Aichi 443-0036, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The concept of neuroprotective therapy for glaucoma is that damage to retinal ganglion cells (RGCs) may be prevented by intervening in neuronal death pathways. This review focuses on strategies for neuroprotection and summarizes preclinical studies that have investigated potential agents over the last 2 years. RECENT FINDINGS Part of the challenge of studies in neuroprotection has been the utilization of an animal model that resembles human glaucoma. Several models have been utilized including acute and chronic intraocular pressure elevation, the DBA/2J mouse, optic nerve axotomy and crush. NMDA inhibitors continued to be explored however with limited success in human trials. Memantine failed to demonstrate neuroprotection in phase III clinical trials. Although its mechanism of neuroprotection has not been fully elaborated, topical brimonidine has shown some neuroprotective benefits. Exogeneous neurotrophins delay, but do not prevent, RGC death. Bioenergetic neuroprotection that is enhancing the energy supply to RGC has been explored with benefits in animal models. Other strategies include TNF-α, modulation of the immune system and inflammation, and blocking apoptotic signals and stem cells. SUMMARY Animal models of glaucoma and neuroprotective strategies continue to be refined. Establishing consensus guidelines for the execution and design of translational research in neuroprotection may optimize the facilitation of neuroprotection research.
Collapse
|
42
|
Huang TL, Chang CH, Lin KH, Sheu MM, Tsai RK. Lack of protective effect of local administration of triamcinolone or systemic treatment with methylprednisolone against damages caused by optic nerve crush in rats. Exp Eye Res 2010; 92:112-9. [PMID: 21185832 DOI: 10.1016/j.exer.2010.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 12/08/2010] [Accepted: 12/13/2010] [Indexed: 12/20/2022]
Abstract
The purpose of the present study was to investigate the effects of administrations of triamcinolone acetonide and systemic methylprednisolone sodium succinate on optic nerves (ON) and retinal ganglion cells (RGC) in a rat model of optic nerve crush. The treated groups either received triamcinolone immediately in the form of two pieces of soaked-gelform surrounding retrobulbar optic nerves (0.5 mg/per gelform) or methylprednisolone via peritoneal injection, and control group received intra-peritoneal injection with phosphate-buffered saline (PBS) after crush experiments. RGC density was counted by retrograde labeling with Fluorogold, and visual function was assessed by flash visual-evoked potentials. Terminal transferase dUTP nick end-labeling (TUNEL) assays, Western blot analysis of serine/threonine kinase (p-Akt), extracellular signal-regulated kinases (p-ERK) and signal transducer and activator of transcription 3 (p-STAT3) and immunohistochemistry of ED1, marker of macrophage/microglia in the optic nerve were conducted. Two and four weeks after optic nerve crush experiments, neither triamcinolone nor methylprednisolone treatment rescued the RGC from death in the central and mid-peripheral retinas compared with those of the corresponding optic nerve-crushed and PBS-treated rats. Visual-evoked potentials measurements showed a prolonged latency of the P(1) wave in all treated groups (triamcinolone-treated: 123 ± 23 ms, methylprednisolone-treated: 133 ± 25 ms and PBS-treated: 151 ± 55 ms) after two weeks. TUNEL assays showed that there was no decrease in apoptotic cells in the RGC layers of both triamcinolone treated and methylprednisolone-treated retinas. Western blot analysis showed that p-AKT, p-ERK and p-Stat3 were not up-regulated in either retina of the triamcinolone or methylprednisolone treated rats. In addition, the number of ED1-positive cells was not attenuated at the lesion sites of the ON in either treatment group. Based upon these results, we conclude that neither retrobulbar administration of triamcinolone nor systemic administration of methylprednisolone has any neuroprotective effects in a rat model of optic nerve crush.
Collapse
Affiliation(s)
- Tzu Lun Huang
- Department of Ophthalmology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | | | | | | | | |
Collapse
|
43
|
Bu P, Basith B, Stubbs EB, Perlman JI. Granulocyte colony-stimulating factor facilitates recovery of retinal function following retinal ischemic injury. Exp Eye Res 2010; 91:104-6. [DOI: 10.1016/j.exer.2010.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 03/25/2010] [Accepted: 03/31/2010] [Indexed: 01/02/2023]
|
44
|
Abstracts of the 2010 Meeting of the International Neuro-Ophthalmology Society, Lyon, France. Neuroophthalmology 2010. [DOI: 10.3109/01658107.2010.485833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Blood pressure treatment in acute ischemic stroke: a review of studies and recommendations. Curr Opin Neurol 2010; 23:46-52. [PMID: 20038827 DOI: 10.1097/wco.0b013e3283355694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Elevated blood pressure (BP) is frequent in patients with acute ischemic stroke. Pathophysiological data support its usefulness to maintain adequate perfusion of the ischemic penumba. This review article aims to summarize the available evidence from clinical studies that examined the prognostic role of BP during the acute phase of ischemic stroke and intervention studies that assessed the efficacy of active BP alteration. RECENT FINDINGS We found 34 observational studies (33,470 patients), with results being inconsistent among the studies; most studies reported a negative association between increased levels of BP and clinical outcome, whereas a few studies showed clinical improvement with higher BP levels, clinical deterioration with decreased BP, or no association at all. Similarly, the conclusions drawn by the 18 intervention studies included in this review (1637 patients) were also heterogeneous. Very recent clinical data suggest a possible beneficial effect of early treatment with some antihypertensives on late clinical outcome. SUMMARY Observational and interventional studies of management of acute poststroke hypertension yield conflicting results. We discuss different explanations that may account for this and discuss the current guidelines and pathophysiological considerations for the management of acute poststroke hypertension.
Collapse
|
46
|
Anti-apoptotic effects of human granulocyte colony-stimulating factor (G-CSF) on retinal ganglion cells after optic nerve crush are PI3K/AKT-dependent. Exp Eye Res 2010; 90:537-45. [PMID: 20144610 DOI: 10.1016/j.exer.2010.01.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 10/31/2009] [Accepted: 01/08/2010] [Indexed: 11/21/2022]
Abstract
The purpose of present study is to dissect the role of PI3K/AKT signaling in the anti-apoptotic effects of human granulocyte colony-stimulating factor (G-CSF) on rat retinal ganglion cells (RGCs) after optic nerve (ON) crush. The ONs of seventy-two adult male Wistar rats were crushed by a standardized method. Control eyes received a sham operation. G-CSF or phosphate-buffered saline (PBS) was immediately administrated after the ON event for 5 days. Twelve rats were used to investigate the signaling pathways using western blot analysis. In other sixty rats, each eye also received intravitreal injections of PI3K/AKT inhibitor (LY294002) or PBS immediately after the experiments. Rats were euthanized at 1 or 2 weeks after the experiment. RGC density was counted by retrograde labeling with Fluorogold. Western blot analysis of p-AKT, TUNEL assays, and immunohistochemistry of the retinas were conducted. Two weeks after ON injury, RGC densities in the central and mid-peripheral retinas of ON-crushed, G-CSF treated rats were significantly higher than those of corresponding ON-crushed, G-CSF-treated and LY294002-injected rats (survival rates of 60% vs. 39% and 43% vs. 33%, respectively; p < 0.01). Decreased TUNEL staining and the up-regulations of p-AKT signaling in retinas of ON-crushed, G-CSF-treated rats were blocked by intravitreal injections of LY294002. The double staining showed that p-AKT expression co-localized with RGCs in the ON crushed, G-CSF treated retinas. In conclusion, the anti-apoptotic effects of G-CSF on RGCs are PI3K/AKT signaling dependent in the retinas to rescue RGCs after ON crush injury.
Collapse
|