1
|
Conti F, Lazzara F, Thermos K, Zingale E, Spyridakos D, Romano GL, Di Martino S, Micale V, Kuchar M, Spadaro A, Pignatello R, Rossi S, D'Amico M, Maria Platania CB, Drago F, Bucolo C. Retinal pharmacodynamic and pharmacokinetic profile of cannabidiol in an in vivo model of retinal excitotoxicity. Eur J Pharmacol 2025; 991:177323. [PMID: 39892452 DOI: 10.1016/j.ejphar.2025.177323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/17/2024] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Cannabidiol (CBD) is one of the principal constituents of Cannabis Sativa with no psychoactive properties. CBD is a promising neuroprotective compound bearing anti-inflammatory and antioxidant properties. However, considering its low solubility, CBD delivery to the retina represents an unresolved issue. The first aim was to investigate the potential neuroprotective effects of CBD in an in vivo model of retinal excitotoxicity induced by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). Rats underwent intravitreal co-injection of AMPA (42 nmol) and CBD (10-4 M). The neuroprotective effect of CBD was investigated with histology and immunohistochemical evaluation of inflammatory and oxidative stress biomarkers. CBD reversed the AMPA-induced total retinal, inner nuclear layer and inner plexiform layer shrinkage and loss of amacrine cells. Moreover, CBD decreased the AMPA induced number of cleaved caspase-3, Iba-1 and nitrotyrosine (NT) positive cells. Based on this evidence, we developed a nanotechnological formulation of CBD to overcome critical issues related to its eye delivery. Particularly, nanostructured lipid carriers (NLC) loaded with CBD were prepared, optimized and characterized. Due to the optimal physicochemical characteristics, CBD-NLC3 has been selected and the in vitro release profile has been investigated. Additionally, CBD-NLC3 was topically administered to rats, and retinal CBD levels were determined. CBD-NLC3 formulation, after a single topical administration, efficiently delivered CBD in the retina (Cmax = 98 ± 25.9 ng/mg; Tmax = 60 min), showing a high translational value. In conclusion, these findings showed a good PD/PK profile of CBD warranting further pre-clinical and clinical evaluation of the new formulation for the treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Federica Conti
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Francesca Lazzara
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Kyriaki Thermos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece.
| | - Elide Zingale
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Catania, Italy; NANOMED-Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Catania, Italy
| | - Dimitris Spyridakos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | - Serena Di Martino
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Dept. Chemistry of Natural Compounds, University of Chemistry and Technologies, Prague, Czech Republic; Psychedelic Research Center, National Institute of Mental Health, Klecany, Czech Republic
| | - Angelo Spadaro
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Rosario Pignatello
- Laboratory of Drug Delivery Technology, Department of Drug and Health Sciences, University of Catania, Catania, Italy; NANOMED-Research Centre for Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, Catania, Italy
| | - Settimio Rossi
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Chiara Bianca Maria Platania
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy; Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy; Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy; Center for Research in Ocular Pharmacology-CERFO, University of Catania, Catania, Italy.
| |
Collapse
|
2
|
Tan Y, Yan Z, Yin J, Cao J, Xie B, Zhang F, Zhang W, Xiong W. Elucidating the role of genetically determined metabolites in Diabetic Retinopathy: insights from a mendelian randomization analysis. Acta Diabetol 2025; 62:193-203. [PMID: 39090426 DOI: 10.1007/s00592-024-02345-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024]
Abstract
AIMS Diabetic retinopathy (DR) results from complex genetic and metabolic interactions. Unraveling the links between blood metabolites and DR can advance risk prediction and therapy. METHODS Leveraging Mendelian Randomization (MR) and Linkage Disequilibrium Score Regression (LDSC), we analyzed 10,413 DR cases and 308,633 controls. Data was sourced from the Metabolomics GWAS server and the FinnGen project. RESULTS Our research conducted a comprehensive MR analysis across 486 serum metabolites to investigate their causal role in DR. After stringent selection and validation of instrumental variables, we focused on 480 metabolites for analysis. Our findings revealed 38 metabolites potentially causally associated with DR. Specifically, 4-androsten-3beta,17beta-diol disulfate 2 was identified as significantly associated with a reduced risk of DR (OR = 0.471, 95% CI = 0.324-0.684, p = 7.87 × 10- 5), even after rigorous adjustments for multiple testing. Sensitivity analyses further validated the robustness of this association, and linkage disequilibrium score regression analyses showed no significant genetic correlation between this metabolite and DR, suggesting a specific protective effect against DR. CONCLUSIONS Our study identifies 4-androsten-3beta,17beta-diol disulfate 2, a metabolite of androgens, as a significant protective factor against diabetic retinopathy, suggesting androgens as potential therapeutic targets.
Collapse
Affiliation(s)
- Yao Tan
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China
- Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha City, 410013, Hunan Province, China
| | - Zuyun Yan
- The Third Xiangya Hospital, Central South University, Changsha City, 410013, Hunan Province, China
| | - Jiayang Yin
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China
| | - Jiamin Cao
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China
| | - Bingyu Xie
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China
| | - Feng Zhang
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China
| | - Wenhua Zhang
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China.
| | - Wei Xiong
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha City, 410013, Hunan Province, China.
| |
Collapse
|
3
|
Zhang X, Huang Y, Xu N, Feng W, Qiao J, Liu M. Low serum dehydroepiandrosterone levels are associated with diabetic retinopathy in patients with type 2 diabetes mellitus. J Diabetes Investig 2023; 14:675-685. [PMID: 36811237 PMCID: PMC10119925 DOI: 10.1111/jdi.13997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
AIMS This cross-sectional study assessed the association of serum dehydroepiandrosterone levels with the risk of diabetic retinopathy in patients with type 2 diabetes mellitus in China. MATERIALS AND METHODS Patients with type 2 diabetes mellitus were included in a multivariate logistic regression analysis to assess the association of dehydroepiandrosterone with diabetic retinopathy after adjusting for confounding factors. A restricted cubic spline was also used to model the association of serum dehydroepiandrosterone level with the risk of diabetic retinopathy and to describe the overall dose-response correlation. Additionally, an interaction test was conducted in the multivariate logistic regression analysis to compare the effects of dehydroepiandrosterone on diabetic retinopathy among age, sex, obesity status, hypertension, dyslipidemia, and glycosylated hemoglobin level subgroups. RESULTS In total, 1,519 patients were included in the final analysis. Low serum dehydroepiandrosterone was significantly associated with diabetic retinopathy in patients with type 2 diabetes mellitus after adjustment for confounding factors (odds ratio [quartile 4 vs quartile 1]: 0.51; 95% confidence interval: 0.32-0.81; P = 0.012 for the trend). Additionally, the restricted cubic spline indicated that the odds of diabetic retinopathy decreased linearly as the dehydroepiandrosterone concentration increased (P-overall = 0.044; P-nonlinear = 0.364). Finally, the subgroup analyses showed that the dehydroepiandrosterone level stably affected diabetic retinopathy (all P for interaction >0.05). CONCLUSIONS Low serum dehydroepiandrosterone levels were significantly associated with diabetic retinopathy in patients with type 2 diabetes mellitus, suggesting that dehydroepiandrosterone contributes to the pathogenesis of diabetic retinopathy.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Yadi Huang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ning Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenli Feng
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingting Qiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Kokona D, Spyridakos D, Tzatzarakis M, Papadogkonaki S, Filidou E, Arvanitidis KI, Kolios G, Lamani M, Makriyannis A, Malamas MS, Thermos K. The endocannabinoid 2-arachidonoylglycerol and dual ABHD6/MAGL enzyme inhibitors display neuroprotective and anti-inflammatory actions in the in vivo retinal model of AMPA excitotoxicity. Neuropharmacology 2021; 185:108450. [PMID: 33450278 DOI: 10.1016/j.neuropharm.2021.108450] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/17/2020] [Accepted: 01/03/2021] [Indexed: 12/14/2022]
Abstract
The endocannabinoid system has been shown to be a putative therapeutic target for retinal disease. Here, we aimed to investigate the ability of the endocannabinoid 2-arachidonoylglycerol (2-AG) and novel inhibitors of its metabolic enzymes, α/β-hydrolase domain-containing 6 (ABHD6) and monoacylglycerol lipase (MAGL), a) to protect the retina against excitotoxicity and b) the mechanisms involved in the neuroprotection. Sprague-Dawley rats, wild type and Akt2-/- C57BL/6 mice were intravitreally administered with phosphate-buffered saline or (RS)-α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide (AMPA). 2-AG was intravitreally co-administered with AMPA in the absence and presence of AM251 or AM630 (cannabinoid 1 and 2 receptor antagonists, respectively) or Wortmannin [Phosphoinositide 3-Kinase (PI3K)/Akt inhibitor]. Inhibitors of ABHD6 and dual ABHD6/MAGL (AM12100 and AM11920, respectively) were co-administered with AMPA intravitreally in rats. Immunohistochemistry was performed using antibodies raised against retinal neuronal markers (bNOS), microglia (Iba1) and macroglia (GFAP). TUNEL assay and real-time PCR were also employed. The CB2 receptor was expressed in rat retina (approx. 62% of CB1 expression). 2-AG attenuated the AMPA-induced increase in TUNEL+ cells. 2-AG activation of both CB1 and CB2 receptors and the PI3K/Akt downstream signaling pathway, as substantiated by the use of Akt2-/- mice, afforded neuroprotection against AMPA excitotoxicity. AM12100 and AM11920 attenuated the AMPA-induced glia activation and produced a dose-dependent partial neuroprotection, with the dual inhibitor AM11920 being more efficacious. These results show that 2-AG has the pharmacological profile of a putative therapeutic for retinal diseases characterized by neurodegeneration and neuroinflammation, when administered exogenously or by the inhibition of its metabolic enzymes.
Collapse
Affiliation(s)
- Despina Kokona
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, 71003, Greece.
| | - Dimitris Spyridakos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, 71003, Greece.
| | - Manolis Tzatzarakis
- Department of Toxicology, School of Medicine, University of Crete, Heraklion, Crete, 71003, Greece.
| | - Sofia Papadogkonaki
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, 71003, Greece.
| | - Eirini Filidou
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece.
| | - Konstantinos I Arvanitidis
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece.
| | - George Kolios
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece.
| | - Manjunath Lamani
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Michael S Malamas
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, 02115, USA.
| | - Kyriaki Thermos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, 71003, Greece.
| |
Collapse
|
5
|
Strac DS, Konjevod M, Perkovic MN, Tudor L, Erjavec GN, Pivac N. Dehydroepiandrosterone (DHEA) and its Sulphate (DHEAS) in Alzheimer's Disease. Curr Alzheimer Res 2020; 17:141-157. [PMID: 32183671 DOI: 10.2174/1567205017666200317092310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/20/2020] [Accepted: 01/26/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Neurosteroids Dehydroepiandrosterone (DHEA) and Dehydroepiandrosterone Sulphate (DHEAS) are involved in many important brain functions, including neuronal plasticity and survival, cognition and behavior, demonstrating preventive and therapeutic potential in different neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease. OBJECTIVE The aim of the article was to provide a comprehensive overview of the literature on the involvement of DHEA and DHEAS in Alzheimer's disease. METHODS PubMed and MEDLINE databases were searched for relevant literature. The articles were selected considering their titles and abstracts. In the selected full texts, lists of references were searched manually for additional articles. RESULTS We performed a systematic review of the studies investigating the role of DHEA and DHEAS in various in vitro and animal models, as well as in patients with Alzheimer's disease, and provided a comprehensive discussion on their potential preventive and therapeutic applications. CONCLUSION Despite mixed results, the findings of various preclinical studies are generally supportive of the involvement of DHEA and DHEAS in the pathophysiology of Alzheimer's disease, showing some promise for potential benefits of these neurosteroids in the prevention and treatment. However, so far small clinical trials brought little evidence to support their therapy in AD. Therefore, large-scale human studies are needed to elucidate the specific effects of DHEA and DHEAS and their mechanisms of action, prior to their applications in clinical practice.
Collapse
Affiliation(s)
- Dubravka S Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Matea N Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Gordana N Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Nela Pivac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| |
Collapse
|
6
|
Ibán-Arias R, Lisa S, Poulaki S, Mastrodimou N, Charalampopoulos I, Gravanis A, Thermos K. Effect of topical administration of the microneurotrophin BNN27 in the diabetic rat retina. Graefes Arch Clin Exp Ophthalmol 2019; 257:2429-2436. [PMID: 31512044 DOI: 10.1007/s00417-019-04460-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/27/2019] [Accepted: 09/04/2019] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Diabetic retinopathy (DR) is a complex eye disease associated with diabetes mellitus. It is characterized by three pathophysiological components, namely microangiopathy, neurodegeneration, and inflammation. We recently reported that intraperitoneal administration of BNN27, a novel neurosteroidal microneurotrophin, reversed the diabetes-induced neurodegeneration and inflammation in rats treated with streptozotocin (STZ), by activating the NGF TrkA and p75 receptors. The aim of the present study was to investigate the efficacy of BNN27 to protect retinal neurons when applied topically as eye drops in the same model. METHODS The STZ rat model of DR was employed. BNN27 was administered as eye drops to diabetic Sprague-Dawley rats for 7 days, 4 weeks post-STZ (70 mg/kg) injection. Immunohistochemistry and western blot analyses were employed to examine the viability of retinal neurons in control, diabetic, and diabetic-treated animals and the involvement of the TrkA receptor and its downstream signaling ERK1/2 kinases, respectively. RESULTS BNN27 reversed the STZ-induced attenuation of the immunoreactive brain nitric oxide synthetase (bNOS)- and tyrosine hydroxylase (TH)-expressing amacrine cells and neurofilament (NFL)-expressing ganglion cell axons in a dose-dependent manner. In addition, BNN27 activated/phosphorylated the TrkA receptor and its downstream prosurvival signaling pathway, ERK1/2 kinases. CONCLUSIONS The results of this study provide solid evidence regarding the efficacy of BNN27 as a neuroprotectant to the diabetic retina when administered topically, and suggest that its pharmacodynamic and pharmacokinetic profiles render it a putative therapeutic for diabetic retinopathy.
Collapse
Affiliation(s)
- Ruth Ibán-Arias
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
- Department of Psychiatry, Laboratory of Molecular Biology and Genetics of Neurodegeneration, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Silvia Lisa
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
- Department of Cell Biology and Pathology, Instituto de Neurociencias de Castilla y León (INCyL) University of Salamanca & Institute of Biomedical Research, 37007, Salamanca, Spain
| | - Smaragda Poulaki
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
| | - Niki Mastrodimou
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
- Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas (FORTH), Heraklion, Crete, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece
- Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas (FORTH), Heraklion, Crete, Greece
| | - Kyriaki Thermos
- Department of Pharmacology, School of Medicine, University of Crete, 70013, Heraklion, Crete, Greece.
| |
Collapse
|
7
|
Luo ZW, Wang HT, Wang N, Sheng WW, Jin M, Lu Y, Bai YJ, Zou SQ, Pang YL, Xu H, Zhang X. Establishment of an adult zebrafish model of retinal neurodegeneration induced by NMDA. Int J Ophthalmol 2019; 12:1250-1261. [PMID: 31456914 PMCID: PMC6694058 DOI: 10.18240/ijo.2019.08.04] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
AIM To establish a model of retinal neurodegeneration induced by N-Methyl-D-aspartic acid (NMDA) in adult zebrafish. METHODS We compared the effects of three different NMDA delivery methods on retinal neurodegeneration in adult zebrafish: immersion (I.M.), intravitreal injection (I.V.), and intraperitoneal injection (I.P.), and examined retinal pathology and degeneration by hematoxylin and eosin and TUNEL staining in the treated zebrafish. Effects of the NMDA receptor antagonist MK-801 and the natural product resveratrol on NMDA-induced retinal neurodegeneration were also assessed. RESULTS The thickened inner retina was seen in histology with 100 µmol/L NMDA by I.M. administration. Significant apoptosis in the retinal ganglion cell layer and retinal thickness reduction occurred in 0.5 mol/L NMDA I.P. administration group.Seizure-like behavioral changes, but no retinal histological alteration occurred in 16 mg/kg NMDA I.P. administration group. Resveratrol and MK-801 prevented NMDA-induced retinal neurodegeneration in the zebrafish. CONCLUSION Among the three drug administration methods, I.V. injection of NMDA is the most suitable for establishment of an acute retinal damage model in zebrafish. I.M. with NMDA is likely the best for use as a chronic retinal damage model. I.P. treatment with NMDA causes brain damage. Resveratrol and MK801 may be a clinically valuable treatment for retinal neurodegeneration.
Collapse
Affiliation(s)
- Zhi-Wen Luo
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
- Queen Mary School of Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Han-Tsing Wang
- Institute of Life Science, Nanchang University, Nanchang 330031, Jiangxi Province, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang 330031, Jiangxi Province, China
| | - Ning Wang
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
- Queen Mary School of Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Wei-Wei Sheng
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
- Queen Mary School of Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Ming Jin
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
| | - Ye Lu
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
| | - Yi-Jiang Bai
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
- Queen Mary School of Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Su-Qi Zou
- Institute of Life Science, Nanchang University, Nanchang 330031, Jiangxi Province, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang 330031, Jiangxi Province, China
| | - Yu-Lian Pang
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
| | - Hong Xu
- Institute of Life Science, Nanchang University, Nanchang 330031, Jiangxi Province, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang 330031, Jiangxi Province, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University; Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, Jiangxi Province, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang 330031, Jiangxi Province, China
| |
Collapse
|
8
|
Tsoka P, Matsumoto H, Maidana DE, Kataoka K, Naoumidi I, Gravanis A, Vavvas DG, Tsilimbaris MK. Effects of BNN27, a novel C17-spiroepoxy steroid derivative, on experimental retinal detachment-induced photoreceptor cell death. Sci Rep 2018; 8:10661. [PMID: 30006508 PMCID: PMC6045604 DOI: 10.1038/s41598-018-28633-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 06/26/2018] [Indexed: 02/01/2023] Open
Abstract
Retinal detachment (RD) leads to photoreceptor cell death secondary to the physical separation of the retina from the underlying retinal pigment epithelium. Intensifying photoreceptor survival in the detached retina could be remarkably favorable for many retinopathies in which RD can be seen. BNN27, a blood-brain barrier (BBB)-permeable, C17-spiroepoxy derivative of dehydroepiandrosterone (DHEA) has shown promising neuroprotective activity through interaction with nerve growth factor receptors, TrkA and p75NTR. Here, we administered BNN27 systemically in a murine model of RD. TUNEL+ photoreceptors were significantly decreased 24 hours post injury after a single administration of 200 mg/kg BNN27. Furthermore, BNN27 increased inflammatory cell infiltration, as well as, two markers of gliosis 24 hours post RD. However, single or multiple doses of BNN27 were not able to protect the overall survival of photoreceptors 7 days post injury. Additionally, BNN27 did not induce the activation/phosphorylation of TrkAY490 in the detached retina although the mRNA levels of the receptor were increased in the photoreceptors post injury. Together, these findings, do not demonstrate neuroprotective activity of BNN27 in experimentally-induced RD. Further studies are needed in order to elucidate the paradox/contradiction of these results and the mechanism of action of BNN27 in this model of photoreceptor cell damage.
Collapse
Affiliation(s)
- Pavlina Tsoka
- Laboratory of Optics and Vision, University of Crete Medical School, Heraklion, Crete, Greece.,Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Matsumoto
- Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel E Maidana
- Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Keiko Kataoka
- Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Irene Naoumidi
- Laboratory of Optics and Vision, University of Crete Medical School, Heraklion, Crete, Greece
| | - Achille Gravanis
- Department of Pharmacology, University of Crete Medical School, Heraklion, Crete, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Demetrios G Vavvas
- Angiogenesis Laboratory, Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA.
| | - Miltiadis K Tsilimbaris
- Laboratory of Optics and Vision, University of Crete Medical School, Heraklion, Crete, Greece.
| |
Collapse
|
9
|
Schverer M, Lanfumey L, Baulieu EE, Froger N, Villey I. Neurosteroids: non-genomic pathways in neuroplasticity and involvement in neurological diseases. Pharmacol Ther 2018; 191:190-206. [PMID: 29953900 DOI: 10.1016/j.pharmthera.2018.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neurosteroids are neuroactive brain-born steroids. They can act through non-genomic and/or through genomic pathways. Genomic pathways are largely described for steroid hormones: the binding to nuclear receptors leads to transcription regulation. Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone have no corresponding nuclear receptor identified so far whereas some of their non-genomic targets have been identified. Neuroplasticity is the capacity that neuronal networks have to change their structure and function in response to biological and/or environmental signals; it is regulated by several mechanisms, including those that involve neurosteroids. In this review, after a description of their biosynthesis, the effects of Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone on their targets will be exposed. We then shall highlight that neurosteroids, by acting on these targets, can regulate neurogenesis, structural and functional plasticity. Finally, we will discuss the therapeutic potential of neurosteroids in the pathophysiology of neurological diseases in which alterations of neuroplasticity are associated with changes in neurosteroid levels.
Collapse
Affiliation(s)
- Marina Schverer
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France
| | - Laurence Lanfumey
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France.
| | - Etienne-Emile Baulieu
- MAPREG SAS, Le Kremlin-Bicêtre, France; Inserm UMR 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | | | | |
Collapse
|
10
|
Arbo BD, Ribeiro FS, Ribeiro MF. Astrocyte Neuroprotection and Dehydroepiandrosterone. VITAMINS AND HORMONES 2018; 108:175-203. [PMID: 30029726 DOI: 10.1016/bs.vh.2018.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dehydroepiandrosterone (DHEA) and its sulfate ester (DHEAS) are the most abundant steroid hormones in the systemic circulation of humans. Due to their abundance and reduced production during aging, these hormones have been suggested to play a role in many aspects of health and have been used as drugs for a multiple range of therapeutic actions, including hormonal replacement and the improvement of aging-related diseases. In addition, several studies have shown that DHEA and DHEAS are neuroprotective under different experimental conditions, including models of ischemia, traumatic brain injury, spinal cord injury, glutamate excitotoxicity, and neurodegenerative diseases. Since astrocytes are responsible for the maintenance of neural tissue homeostasis and the control of neuronal energy supply, changes in astrocytic function have been associated with neuronal damage and the progression of different pathologies. Therefore, the aim of this chapter is to discuss the neuroprotective effects of DHEA against different types of brain and spinal cord injuries and how the modulation of astrocytic function by DHEA could represent an interesting therapeutic approach for the treatment of these conditions.
Collapse
Affiliation(s)
- Bruno D Arbo
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - Felipe S Ribeiro
- Laboratório de Interação Neuro-Humoral, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Maria F Ribeiro
- Laboratório de Interação Neuro-Humoral, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
11
|
Ibán-Arias R, Lisa S, Mastrodimou N, Kokona D, Koulakis E, Iordanidou P, Kouvarakis A, Fothiadaki M, Papadogkonaki S, Sotiriou A, Katerinopoulos HE, Gravanis A, Charalampopoulos I, Thermos K. The Synthetic Microneurotrophin BNN27 Affects Retinal Function in Rats With Streptozotocin-Induced Diabetes. Diabetes 2018; 67:321-333. [PMID: 29208634 DOI: 10.2337/db17-0391] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 11/18/2017] [Indexed: 11/13/2022]
Abstract
BNN27, a C17-spiroepoxy derivative of DHEA, was shown to have antiapoptotic properties via mechanisms involving the nerve growth factor receptors (tropomyosin-related kinase A [TrkA]/neurotrophin receptor p75 [p75NTR]). In this study, we examined the effects of BNN27 on neural/glial cell function, apoptosis, and inflammation in the experimental rat streptozotocin (STZ) model of diabetic retinopathy (DR). The ability of BNN27 to activate the TrkA receptor and regulate p75NTR expression was investigated. BNN27 (2,10, and 50 mg/kg i.p. for 7 days) administration 4 weeks post-STZ injection (paradigm A) reversed the diabetes-induced glial activation and loss of function of amacrine cells (brain nitric oxide synthetase/tyrosine hydroxylase expression) and ganglion cell axons via a TrkA receptor (TrkAR)-dependent mechanism. BNN27 activated/phosphorylated the TrkAY490 residue in the absence but not the presence of TrkAR inhibitor and abolished the diabetes-induced increase in p75NTR expression. However, it had no effect on retinal cell death (TUNEL+ cells). A similar result was observed when BNN27 (10 mg/kg i.p.) was administered at the onset of diabetes, every other day for 4 weeks (paradigm B). However, BNN27 decreased the activation of caspase-3 in both paradigms. Finally, BNN27 reduced the proinflammatory (TNFα and IL-1β) and increased the anti-inflammatory (IL-10 and IL-4) cytokine levels. These findings suggest that BNN27 has the pharmacological profile of a therapeutic for DR, since it targets both the neurodegenerative and inflammatory components of the disease.
Collapse
MESH Headings
- Amacrine Cells/drug effects
- Amacrine Cells/immunology
- Amacrine Cells/metabolism
- Amacrine Cells/pathology
- Animals
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/therapeutic use
- Axons/drug effects
- Axons/immunology
- Axons/metabolism
- Axons/pathology
- Dehydroepiandrosterone/administration & dosage
- Dehydroepiandrosterone/therapeutic use
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Retinopathy/immunology
- Diabetic Retinopathy/metabolism
- Diabetic Retinopathy/pathology
- Diabetic Retinopathy/prevention & control
- Dose-Response Relationship, Drug
- Eye Proteins/agonists
- Eye Proteins/metabolism
- Female
- Ganglia, Sensory/drug effects
- Ganglia, Sensory/immunology
- Ganglia, Sensory/metabolism
- Ganglia, Sensory/pathology
- Male
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/metabolism
- Neuroglia/drug effects
- Neuroglia/immunology
- Neuroglia/metabolism
- Neuroglia/pathology
- Neuroprotective Agents/administration & dosage
- Neuroprotective Agents/therapeutic use
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Rats, Sprague-Dawley
- Receptor, Nerve Growth Factor/agonists
- Receptor, Nerve Growth Factor/metabolism
- Receptor, trkA/agonists
- Receptor, trkA/metabolism
- Retina/drug effects
- Retina/immunology
- Retina/pathology
- Retina/physiopathology
- Streptozocin
Collapse
Affiliation(s)
- Ruth Ibán-Arias
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Silvia Lisa
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Niki Mastrodimou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Despina Kokona
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Emmanuil Koulakis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Panagiota Iordanidou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Antonis Kouvarakis
- Laboratory of Environmental Chemical Processes, Department of Chemistry, University of Crete, Heraklion, Crete, Greece
| | - Myrto Fothiadaki
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Sofia Papadogkonaki
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Aggeliki Sotiriou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | | | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research & Technology-Hellas, University of Crete, Crete, Greece
| | | | - Kyriaki Thermos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
12
|
Bucolo C, Platania CBM, Drago F, Bonfiglio V, Reibaldi M, Avitabile T, Uva M. Novel Therapeutics in Glaucoma Management. Curr Neuropharmacol 2018; 16:978-992. [PMID: 28925883 PMCID: PMC6120119 DOI: 10.2174/1570159x15666170915142727] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 06/26/2017] [Accepted: 09/03/2017] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Glaucoma is a progressive optic neuropathy characterized by retinal ganglion cell death and alterations of visual field. Elevated intraocular pressure (IOP) is considered the main risk factor of glaucoma, even though other factors cannot be ruled out, such as epigenetic mechanisms. OBJECTIVE An overview of the ultimate promising experimental drugs to manage glaucoma has been provided. RESULTS In particular, we have focused on purinergic ligands, KATP channel activators, gases (nitric oxide, carbon monoxide and hydrogen sulfide), non-glucocorticoid steroidal compounds, neurotrophic factors, PI3K/Akt activators, citicoline, histone deacetylase inhibitors, cannabinoids, dopamine and serotonin receptors ligands, small interference RNA, and Rho kinase inhibitors. CONCLUSIONS The review has been also endowed of a brief chapter on last reports about potential neuroprotective benefits of anti-glaucoma drugs already present in the market.
Collapse
Affiliation(s)
- Claudio Bucolo
- Address correspondence to this author at the Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; Tel: +39 095 4781196;
| | | | | | | | | | | | | |
Collapse
|
13
|
Shi W, George SK, George B, Curry CV, Murzabdillaeva A, Alkan S, Amin HM. TrkA is a binding partner of NPM-ALK that promotes the survival of ALK + T-cell lymphoma. Mol Oncol 2017; 11:1189-1207. [PMID: 28557340 PMCID: PMC5579389 DOI: 10.1002/1878-0261.12088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 05/01/2017] [Accepted: 05/17/2017] [Indexed: 11/25/2022] Open
Abstract
Nucleophosmin‐anaplastic lymphoma kinase‐expressing (NPM‐ALK+) T‐cell lymphoma is an aggressive neoplasm that is more commonly seen in children and young adults. The pathogenesis of NPM‐ALK+ T‐cell lymphoma is not completely understood. Wild‐type ALK is a receptor tyrosine kinase that is physiologically expressed in neural tissues during early stages of human development, which suggests that ALK may interact with neurotrophic factors. The aberrant expression of NPM‐ALK results from a translocation between the ALK gene on chromosome 2p23 and the NPM gene on chromosome 5q35. The nerve growth factor (NGF) is the first neurotrophic factor attributed to non‐neural functions including cancer cell survival, proliferation, and metastasis. These functions are primarily mediated through the tropomyosin receptor kinase A (TrkA). The expression and role of NGF/TrkA in NPM‐ALK+ T‐cell lymphoma are not known. In this study, we tested the hypothesis that TrkA signaling is upregulated and sustains the survival of this lymphoma. Our data illustrate that TrkA and NGF are expressed in five NPM‐ALK+ T‐cell lymphoma cell lines and TrkA is expressed in 11 of 13 primary lymphoma tumors from patients. In addition, we found evidence to support that NPM‐ALK and TrkA functionally interact. A selective TrkA inhibitor induced apoptosis and decreased cell viability, proliferation, and colony formation of NPM‐ALK+ T‐cell lymphoma cell lines. These effects were associated with downregulation of cell survival regulatory proteins. Similar results were also observed using specific knockdown of TrkA in NPM‐ALK+ T‐cell lymphoma cells by siRNA. Importantly, the inhibition of TrkA signaling was associated with antitumor effects in vivo, because tumor xenografts in mice regressed and the mice exhibited improved survival. In conclusion, TrkA plays an important role in the pathogenesis of NPM‐ALK+ T‐cell lymphoma, and therefore, targeting TrkA signaling may represent a novel approach to eradicate this aggressive neoplasm.
Collapse
Affiliation(s)
- Wenyu Shi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Hematology, Affiliated Hospital of the University of Nantong, Jiangsu, China
| | - Suraj Konnath George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bhawana George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Choladda V Curry
- Department of Pathology and Immunology, Baylor College of Medicine & Texas Children's Hospital, Houston, TX, USA
| | - Albina Murzabdillaeva
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Serhan Alkan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
14
|
Garcia TB, Hollborn M, Bringmann A. Expression and signaling of NGF in the healthy and injured retina. Cytokine Growth Factor Rev 2017; 34:43-57. [PMID: 27964967 DOI: 10.1016/j.cytogfr.2016.11.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 01/02/2023]
Abstract
This review summarizes the present knowledge concerning the retinal localization of the nerve growth factor (NGF), its precursor proNGF, and the receptors TrkA and p75NTR in the developing and mature rodent retina. We further discuss the changes in the expression of NGF and the receptors in experimental models of retinal disorders and diseases like inherited retinitis pigmentosa, retinal detachment, glaucoma, and diabetic retinopathy. Since proNGF is now recognized as a bioactive signaling molecule which induces cell death through p75NTR activation, the role of proNGF in the induction of retinal cell loss under neurodegenerative conditions is also highlighted. In addition, we present the evidences for a potential therapeutic intervention with NGF for the treatment of retinal neurodegenerative diseases. Different strategies have been developed and experimentally tested in mice and rats in order to reduce cell loss and Müller cell gliosis, e.g., increasing the availability of endogenous NGF, administration of exogenous NGF, activation of TrkA, and inhibition of p75NTR. Here, we discuss the several lines of evidence supporting a protective effect of NGF on retinal cell loss, with specific emphasis on photoreceptor and retinal ganglion cell degeneration. A better understanding of the mechanisms underlying the effects of NGF and proNGF in the modulation of neurodegeneration and gliosis in the retina will help to develop efficient therapeutic strategies for various retinal diseases.
Collapse
Affiliation(s)
| | - Margrit Hollborn
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
15
|
Zhao L, Chen G, Li J, Fu Y, Mavlyutov TA, Yao A, Nickells RW, Gong S, Guo LW. An intraocular drug delivery system using targeted nanocarriers attenuates retinal ganglion cell degeneration. J Control Release 2017; 247:153-166. [PMID: 28063892 PMCID: PMC5323250 DOI: 10.1016/j.jconrel.2016.12.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/08/2016] [Accepted: 12/29/2016] [Indexed: 02/03/2023]
Abstract
Glaucoma is a common blinding disease characterized by loss of retinal ganglion cells (RGCs). To date, there is no clinically available treatment directly targeting RGCs. We aim to develop an RGC-targeted intraocular drug delivery system using unimolecular micelle nanoparticles (unimNPs) to prevent RGC loss. The unimNPs were formed by single/individual multi-arm star amphiphilic block copolymer poly(amidoamine)-polyvalerolactone-poly(ethylene glycol) (PAMAM-PVL-PEG). While the hydrophobic PAMAM-PVL core can encapsulate hydrophobic drugs, the hydrophilic PEG shell provides excellent water dispersity. We conjugated unimNPs with the cholera toxin B domain (CTB) for RGC-targeting and with Cy5.5 for unimNP-tracing. To exploit RGC-protective sigma-1 receptor (S1R), we loaded unimNPs with an endogenous S1R agonist dehydroepiandrosterone (DHEA) as an FDA-approved model drug. These unimNPs produced a steady DHEA release in vitro for over two months at pH7.4. We then co-injected (mice, intraocular) unimNPs with the glutamate analog N-methyl-d-aspartate (NMDA), which is excito-toxic and induces RGC death. The CTB-conjugated unimNPs (i.e., targeted NPs) accumulated at the RGC layer and effectively preserved RGCs at least for 14days, whereas the unimNPs without CTB (i.e., non-targeted NPs) showed neither accumulation at nor protection of NMDA-treated RGCs. Consistent with S1R functions, targeted NPs relative to non-targeted NPs showed markedly better inhibitory effects on apoptosis and oxidative/inflammatory stresses in the RGC layer. Hence, the DHEA-loaded, CTB-conjugated unimNPs represent an RGC/S1R dual-targeted nanoplatform that generates an efficacious template for further development of a sustainable intraocular drug delivery system to protect RGCs, which may be applicable to treatments directed at glaucomatous pathology.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Guojun Chen
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jun Li
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Department of Ophthalmology, The First Hospital of China Medical University, Shenyang 110001, China.; Department of Ophthalmology, The 3rd People's Hospital of Dalian, Dalian 116033, China
| | - Yingmei Fu
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai 200030, China
| | - Timur A Mavlyutov
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Annie Yao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Robert W Nickells
- Department of Ophthalmology and Vision Sciences, University of Wisconsin-Madison, 1300 University Ave, Madison, WI 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Shaoqin Gong
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Lian-Wang Guo
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
16
|
Glajch KE, Ferraiuolo L, Mueller KA, Stopford MJ, Prabhkar V, Gravanis A, Shaw PJ, Sadri-Vakili G. MicroNeurotrophins Improve Survival in Motor Neuron-Astrocyte Co-Cultures but Do Not Improve Disease Phenotypes in a Mutant SOD1 Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0164103. [PMID: 27716798 PMCID: PMC5055348 DOI: 10.1371/journal.pone.0164103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease caused by loss of motor neurons. ALS patients experience rapid deterioration in muscle function with an average lifespan of 3–5 years after diagnosis. Currently, the most effective therapeutic only extends lifespan by a few months, thus highlighting the need for new and improved therapies. Neurotrophic factors (NTFs) are important for neuronal development, maintenance, and survival. NTF treatment has previously shown efficacy in pre-clinical ALS models. However, clinical trials using NTFs produced no major improvements in ALS patients, due in part to the limited blood brain barrier (BBB) penetration. In this study we assessed the potential neuroprotective effects of a novel class of compounds known as MicroNeurotrophins (MNTs). MNTs are derivatives of Dehydroepiandrosterone (DHEA), an endogenous neurosteroid that can cross the BBB and bind to tyrosine kinase receptors mimicking the pro-survival effects of NTFs. Here we sought to determine whether MNTs were neuroprotective in two different models of ALS. Our results demonstrate that BNN27 (10 μM) attenuated loss of motor neurons co-cultured with astrocytes derived from human ALS patients with SOD1 mutations via the reduction of oxidative stress. Additionally, in the G93A SOD1 mouse, BNN27 (10 mg/kg) treatment attenuated motor behavioral impairment in the paw grip endurance and rotarod tasks at postnatal day 95 in female but not male mice. In contrast, BNN27 (10 mg/kg and 50 mg/kg) treatment did not alter any other behavioral outcome or neuropathological marker in male or female mice. Lastly, BNN27 was not detected in post-mortem brain or spinal cord tissue of treated mice due to the rapid metabolism of BNN27 by mouse hepatocytes relative to human hepatocytes. Together, these findings demonstrate that BNN27 treatment failed to yield significant neuroprotective effects in the G93A SOD1 model likely due to its rapid rate of metabolism in mice.
Collapse
Affiliation(s)
- Kelly E. Glajch
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Laura Ferraiuolo
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Kaly A. Mueller
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Matthew J. Stopford
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Varsha Prabhkar
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Institute of Molecular Biology & Biotechnology-FORTH, Heraklion 71003, Greece
| | - Pamela J. Shaw
- Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, United Kingdom
| | - Ghazaleh Sadri-Vakili
- NeuroEpigenetics Laboratory, MassGeneral Institute for Neurodegenerative Disease (MIND), Massachusetts General Hospital, Boston, MA, 02129–4404, United States of America
- * E-mail:
| |
Collapse
|
17
|
Arbo BD, Benetti F, Ribeiro MF. Astrocytes as a target for neuroprotection: Modulation by progesterone and dehydroepiandrosterone. Prog Neurobiol 2016; 144:27-47. [DOI: 10.1016/j.pneurobio.2016.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 01/14/2016] [Accepted: 03/14/2016] [Indexed: 01/19/2023]
|
18
|
Su TC, Lin SH, Lee PT, Yeh SH, Hsieh TH, Chou SY, Su TP, Hung JJ, Chang WC, Lee YC, Chuang JY. The sigma-1 receptor-zinc finger protein 179 pathway protects against hydrogen peroxide-induced cell injury. Neuropharmacology 2016; 105:1-9. [PMID: 26792191 PMCID: PMC5520630 DOI: 10.1016/j.neuropharm.2016.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/14/2015] [Accepted: 01/08/2016] [Indexed: 11/25/2022]
Abstract
The accumulation of reactive oxygen species (ROS) have implicated the pathogenesis of several human diseases including neurodegenerative disorders, stroke, and traumatic brain injury, hence protecting neurons against ROS is very important. In this study, we focused on sigma-1 receptor (Sig-1R), a chaperone at endoplasmic reticulum, and investigated its protective functions. Using hydrogen peroxide (H2O2)-induced ROS accumulation model, we verified that apoptosis-signaling pathways were elicited by H2O2 treatment. However, the Sig-1R agonists, dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS), reduced the activation of apoptotic pathways significantly. By performing protein-protein interaction assays and shRNA knockdown of Sig-1R, we identified the brain Zinc finger protein 179 (Znf179) as a downstream target of Sig-1R regulation. The neuroprotective effect of Znf179 overexpression was similar to that of DHEAS treatment, and likely mediated by affecting the levels of antioxidant enzymes. We also quantified the levels of peroxiredoxin 3 (Prx3) and superoxide dismutase 2 (SOD2) in the hippocampi of wild-type and Znf179 knockout mice, and found both enzymes to be reduced in the knockout versus the wild-type mice. In summary, these results reveal that Znf179 plays a novel role in neuroprotection, and Sig-1R agonists may be therapeutic candidates to prevent ROS-induced damage in neurodegenerative and neurotraumatic diseases.
Collapse
Affiliation(s)
- Tzu-Chieh Su
- Graduate Institute of Medical Science, Taipei Medical University, Taiwan
| | - Shu-Hui Lin
- Graduate Institute of Medical Science, Taipei Medical University, Taiwan; Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Pin-Tse Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Taiwan; Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, USA
| | - Shiu-Hwa Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Taiwan
| | - Tsung-Hsun Hsieh
- Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Szu-Yi Chou
- Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taiwan
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Intramural Research Program, National Institute on Drug Abuse, USA
| | - Jan-Jong Hung
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Science, Taipei Medical University, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan
| | - Yi-Chao Lee
- Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan.
| | - Jian-Ying Chuang
- Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taiwan.
| |
Collapse
|
19
|
Endogenous and Synthetic Cannabinoids as Therapeutics in Retinal Disease. Neural Plast 2016; 2016:8373020. [PMID: 26881135 PMCID: PMC4736800 DOI: 10.1155/2016/8373020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/16/2015] [Accepted: 12/01/2015] [Indexed: 11/25/2022] Open
Abstract
The functional significance of cannabinoids in ocular physiology and disease has been reported some decades ago. In the early 1970s, subjects who smoked Cannabis sativa developed lower intraocular pressure (IOP). This led to the isolation of phytocannabinoids from this plant and the study of their therapeutic effects in glaucoma. The main treatment of this disease to date involves the administration of drugs mediating either the decrease of aqueous humour synthesis or the increase of its outflow and thus reduces IOP. However, the reduction of IOP is not sufficient to prevent visual field loss. Retinal diseases, such as glaucoma and diabetic retinopathy, have been defined as neurodegenerative diseases and characterized by ischemia-induced excitotoxicity and loss of retinal neurons. Therefore, new therapeutic strategies must be applied in order to target retinal cell death, reduction of visual acuity, and blindness. The aim of the present review is to address the neuroprotective and therapeutic potential of cannabinoids in retinal disease.
Collapse
|
20
|
Kokona D, Thermos K. Synthetic and endogenous cannabinoids protect retinal neurons from AMPA excitotoxicity in vivo, via activation of CB1 receptors: Involvement of PI3K/Akt and MEK/ERK signaling pathways. Exp Eye Res 2015; 136:45-58. [DOI: 10.1016/j.exer.2015.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 11/29/2022]
|
21
|
|
22
|
Krøll J. Dehydroepiandrosterone, molecular chaperones and the epigenetics of primate longevity. Rejuvenation Res 2015; 18:341-346. [PMID: 25706901 DOI: 10.1089/rej.2014.1641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dehydroepiandrosterone (DHEA) and dehydroepiandrosterone-sulphate (DHEAS) are the most abundant circulating adrenal steroid hormones. The plasma level of DHEAS correlates with longevity in primates and varies during human development with a maximum in early adulthood and a marked decline during aging. DHEA promotes the expression of molecular chaperones which are housekeeping stress response proteins essential for the processes of folding, translocation, maintenance and repair of proteins, RNA and DNA, as well as for homeostasis, immune response and cancer resistance. The level of chaperone expression correlates with longevity and shows a decline during aging. DHEA-induced promotion of chaperone expression could contribute to the epigenetic evolution of primate longevity.
Collapse
Affiliation(s)
- Jens Krøll
- Hafnia Unit of Biogerontology , Godthåbsvej 111,3 , Frederiksberg, Denmark , DK 2000 , +45 38862220 ;
| |
Collapse
|
23
|
Hill M, Dušková M, Stárka L. Dehydroepiandrosterone, its metabolites and ion channels. J Steroid Biochem Mol Biol 2015; 145:293-314. [PMID: 24846830 DOI: 10.1016/j.jsbmb.2014.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/06/2014] [Accepted: 05/11/2014] [Indexed: 11/20/2022]
Abstract
This review is focused on the physiological and pathophysiological relevance of steroids influencing the activities of the central and peripheral nervous systems with regard to their concentrations in body fluids and tissues in various stages of human life like the fetal development or pregnancy. The data summarized in this review shows that DHEA and its unconjugated and sulfated metabolites are physiologically and pathophysiologically relevant in modulating numerous ion channels and participate in vital functions of the human organism. DHEA and its unconjugated and sulfated metabolites including 5α/β-reduced androstane steroids participate in various physiological and pathophysiological processes like the management of GnRH cyclic release, regulation of glandular and neurotransmitter secretions, maintenance of glucose homeostasis on one hand and insulin insensitivity on the other hand, control of skeletal muscle and smooth muscle activities including vasoregulation, promotion of tolerance to ischemia and other neuroprotective effects. In respect of prevalence of steroid sulfates over unconjugated steroids in the periphery and the opposite situation in the CNS, the sulfated androgens and androgen metabolites reach relevance in peripheral organs. The unconjugated androgens and estrogens are relevant in periphery and so much the more in the CNS due to higher concentrations of most unconjugated steroids in the CNS tissues than in circulation and peripheral organs. This article is part of a Special Issue entitled "Essential role of DHEA".
Collapse
Affiliation(s)
- M Hill
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - M Dušková
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - L Stárka
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| |
Collapse
|
24
|
Taylor MK, Stone M, Laurent HK, Rauh MJ, Granger DA. Neuroprotective-neurotrophic effect of endogenous dehydroepiandrosterone sulfate during intense stress exposure. Steroids 2014; 87:54-8. [PMID: 24887210 DOI: 10.1016/j.steroids.2014.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/15/2014] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Abstract
Recent reports demonstrate neurotrophic properties of dehydroepiandrosterone sulfate (DHEAS) in men at rest, as well as profound neurotrophic responses to stress in both men and women. Little is known of neuroprotective-neurotrophic effects of DHEAS during stress exposure, either in men or women. This translational study was designed to examine neuroprotective-neurotrophic effects of DHEAS throughout intense stress exposure in healthy men and women. The study took place within a stressful 12-day military survival course. Utilizing a longitudinal cross-sectional repeated measures design, One hundred sixteen healthy active-duty military personnel (80% male) were studied before, during, and 24h after the course. The dependent variable was the neurotrophin salivary nerve growth factor (sNGF). In terms of total hormone output, the effect of DHEAS on sNGF was mediated by testosterone. Unlike testosterone or cortisol, DHEAS reliably predicted sNGF at each time point, and change in DHEAS predicted change in sNGF across time points. Baseline DHEAS predicted total sNGF output across the stress trajectory. Consistent with preclinical as well as cross-sectional human research, this study demonstrates neuroprotective-neurotrophic effects of DHEAS in healthy men and women exposed to intense stress. Results are evaluated in relation to established criteria for causation.
Collapse
Affiliation(s)
- Marcus K Taylor
- Biobehavioral Sciences Lab, Warfighter Performance Department, Naval Health Research Center, San Diego, CA, United States; Institute for Interdisciplinary Salivary Bioscience Research, Arizona State University, Tempe, AZ, United States; School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States.
| | - Michael Stone
- Biobehavioral Sciences Lab, Warfighter Performance Department, Naval Health Research Center, San Diego, CA, United States; School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Heidemarie K Laurent
- Institute for Interdisciplinary Salivary Bioscience Research, Arizona State University, Tempe, AZ, United States; Department of Psychology, University of Oregon, Eugene, OR, United States
| | - Mitchell J Rauh
- Biobehavioral Sciences Lab, Warfighter Performance Department, Naval Health Research Center, San Diego, CA, United States; School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Douglas A Granger
- Institute for Interdisciplinary Salivary Bioscience Research, Arizona State University, Tempe, AZ, United States; Johns Hopkins School of Nursing and Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
25
|
Garcia TB, Pannicke T, Vogler S, Berk BA, Grosche A, Wiedemann P, Seeger J, Reichenbach A, Herculano AM, Bringmann A. Nerve growth factor inhibits osmotic swelling of rat retinal glial (Müller) and bipolar cells by inducing glial cytokine release. J Neurochem 2014; 131:303-13. [DOI: 10.1111/jnc.12822] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 01/02/2023]
Affiliation(s)
- Tarcyane Barata Garcia
- Paul Flechsig Institute of Brain Research; University of Leipzig; Leipzig Germany
- Institute of Biological Sciences; Federal University of Pará; Belém Brazil
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research; University of Leipzig; Leipzig Germany
| | - Stefanie Vogler
- Paul Flechsig Institute of Brain Research; University of Leipzig; Leipzig Germany
| | - Benjamin-Andreas Berk
- Paul Flechsig Institute of Brain Research; University of Leipzig; Leipzig Germany
- Institute of Veterinary Anatomy; University of Leipzig; Leipzig Germany
| | - Antje Grosche
- Institute of Human Genetics; University of Regensburg; Regensburg Germany
| | - Peter Wiedemann
- Department of Ophthalmology and Eye Hospital; University of Leipzig; Leipzig Germany
| | - Johannes Seeger
- Institute of Veterinary Anatomy; University of Leipzig; Leipzig Germany
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research; University of Leipzig; Leipzig Germany
| | | | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital; University of Leipzig; Leipzig Germany
| |
Collapse
|
26
|
Abstract
Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.
Collapse
|
27
|
Torres NI, Castilla V, Wachsman M. DHEA inhibits measles virus through a mechanism independent of its ability to modulate the Raf/MEK/ERK signaling pathway. Future Virol 2012. [DOI: 10.2217/fvl.12.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Despite the existence of an effective vaccine, measles infection is still frequent in many developing countries with reduced health infrastructure, and it is one of the major causes of child death globally. In the past decade numerous outbreaks have occurred in developed countries, giving a fresh impetus to antiviral research against measles virus. The aim of this study was to investigate the antiviral activity of the natural steroid hormone DHEA against measles virus and the role of the Raf/MEK/ERK signaling pathway in viral multiplication and DHEA’s antiviral activity. Materials & methods: The antiviral activity of DHEA and two ERK modulators, UO126 and anisomycin, was determined using a virus yield reduction assay. Furthermore, we studied DHEA’s virucidal activity and the viral multiplication step affected by the compound. The effect of virus infection on the Raf/MEK/ERK pathway and the activity of those compounds against measles virus spread and induced cytopathic effect were studied using western blot and indirect immunofluorescence. Results & conclusion: We found that DHEA and UO126 are active against measles virus and that they are able to diminish virus-induced cytopathic effects. Also, our study showed that early events in the viral multiplication cycle trigger ERK activation, suggesting that DHEA, a Raf/MEK/ERK modulator, may not exert its antiviral activity through the modulation of this pathway. Our results may provide a first step in the development of new antiviral agents against measles virus.
Collapse
Affiliation(s)
- Nicolás I Torres
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| | - Viviana Castilla
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| | - Mónica Wachsman
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| |
Collapse
|
28
|
Gravanis A, Calogeropoulou T, Panoutsakopoulou V, Thermos K, Neophytou C, Charalampopoulos I. Neurosteroids and Microneurotrophins Signal Through NGF Receptors to Induce Prosurvival Signaling in Neuronal Cells. Sci Signal 2012; 5:pt8. [DOI: 10.1126/scisignal.2003387] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|