1
|
Zhang X, Zhang W, Zhao L, Ma G, Huang Y, Geng Z, Jiang Q, Wen X, Lin Y, Meng Q, Zhang Z, Bi Y. Ocotillol Derivatives Mitigate Retinal Ischemia-Reperfusion Injury by Regulating the Keap1/Nrf2/ARE Signaling Pathway. J Med Chem 2024; 67:15268-15290. [PMID: 39145589 DOI: 10.1021/acs.jmedchem.4c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Retinal ischemia-reperfusion (RIR) injury can lead to various retinal diseases. Oxidative stress is considered an important pathological event in RIR injury. Here, we designed and synthesized 34 ocotillol derivatives, then examined their antioxidant and anti-inflammatory capacities; we found that compounds 7 (C24-R) and 8 (C24-S) were most active. To enhance their water solubility, sustained release, and biocompatibility, compounds 7 and 8 were encapsulated into liposomes for in vivo activity and mechanistic studies. In vivo studies indicated that compounds 7 and 8 protected normal retinal structure and physiological function after RIR injury, reversed damage to retinal ganglion cells, and the S-configuration exhibited significantly stronger activity compared with the R-configuration. Mechanistic studies showed that compound 8 exerted a therapeutic effect on RIR injury by activating the Keap1/Nrf2/ARE signaling pathway; compound 7 did not influence this pathway. We also demonstrated that differential isomerization at the C-24 position influenced protection against RIR injury.
Collapse
Affiliation(s)
- Xin Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Wen Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Laien Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Gongshan Ma
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Yanmei Huang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Zhiyuan Geng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Qian Jiang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Xiaomei Wen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Yuqi Lin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China
| |
Collapse
|
2
|
Qing KX, Lo ACY, Lu S, Zhou Y, Yang D, Yang D. Integrated bioinformatics analysis of retinal ischemia/reperfusion injury in rats with potential key genes. BMC Genomics 2024; 25:367. [PMID: 38622534 PMCID: PMC11017533 DOI: 10.1186/s12864-024-10288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/07/2024] [Indexed: 04/17/2024] Open
Abstract
The tissue damage caused by transient ischemic injury is an essential component of the pathogenesis of retinal ischemia, which mainly hinges on the degree and duration of interruption of the blood supply and the subsequent damage caused by tissue reperfusion. Some research indicated that the retinal injury induced by ischemia-reperfusion (I/R) was related to reperfusion time.In this study, we screened the differentially expressed circRNAs, lncRNAs, and mRNAs between the control and model group and at different reperfusion time (24h, 72h, and 7d) with the aid of whole transcriptome sequencing technology, and the trend changes in time-varying mRNA, lncRNA, circRNA were obtained by chronological analysis. Then, candidate circRNAs, lncRNAs, and mRNAs were obtained as the intersection of differentially expression genes and trend change genes. Importance scores of the genes selected the key genes whose expression changed with the increase of reperfusion time. Also, the characteristic differentially expressed genes specific to the reperfusion time were analyzed, key genes specific to reperfusion time were selected to show the change in biological process with the increase of reperfusion time.As a result, 316 candidate mRNAs, 137 candidate lncRNAs, and 31 candidate circRNAs were obtained by the intersection of differentially expressed mRNAs, lncRNAs, and circRNAs with trend mRNAs, trend lncRNAs and trend circRNAs, 5 key genes (Cd74, RT1-Da, RT1-CE5, RT1-Bb, RT1-DOa) were selected by importance scores of the genes. The result of GSEA showed that key genes were found to play vital roles in antigen processing and presentation, regulation of the actin cytoskeleton, and the ribosome. A network included 4 key genes (Cd74, RT1-Da, RT1-Bb, RT1-DOa), 34 miRNAs and 48 lncRNAs, and 81 regulatory relationship axes, and a network included 4 key genes (Cd74, RT1-Da, RT1-Bb, RT1-DOa), 9 miRNAs and 3 circRNAs (circRNA_10572, circRNA_03219, circRNA_11359) and 12 regulatory relationship axes were constructed, the subcellular location, transcription factors, signaling network, targeted drugs and relationship to eye diseases of key genes were predicted. 1370 characteristic differentially expressed mRNAs (spec_24h mRNA), 558 characteristic differentially expressed mRNAs (spec_72h mRNA), and 92 characteristic differentially expressed mRNAs (spec_7d mRNA) were found, and their key genes and regulation networks were analyzed.In summary, we screened the differentially expressed circRNAs, lncRNAs, and mRNAs between the control and model groups and at different reperfusion time (24h, 72h, and 7d). 5 key genes, Cd74, RT1-Da, RT1-CE5, RT1-Bb, RT1-DOa, were selected. Key genes specific to reperfusion time were selected to show the change in biological process with the increased reperfusion time. These results provided theoretical support and a reference basis for the clinical treatment.
Collapse
Affiliation(s)
- Kai-Xiong Qing
- Department of Cardiac & Vascular Surgery, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan Province, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Siduo Lu
- Department of Ophthalmology, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan Province, China
| | - You Zhou
- Department of Ophthalmology, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan Province, China
| | - Dan Yang
- Department of Ophthalmology, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan Province, China
| | - Di Yang
- Department of Ophthalmology, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
3
|
Ji KB, Wan W, Yang Y, He XJ, Xing YQ, Hu Z. Ameliorative effect of resveratrol on acute ocular hypertension induced retinal injury through the SIRT1/NF-κB pathway. Neurosci Lett 2024; 826:137712. [PMID: 38447888 DOI: 10.1016/j.neulet.2024.137712] [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: 11/28/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/08/2024]
Abstract
Glaucoma is a kind of neurodegenerative disorder characterized by irreversible loss of retinal ganglion cells (RGCs) and permanent visual impairment. It is reported that resveratrol (RES) is a promising drug for neurodegenerative diseases. However, the detailed molecular mechanisms underlying its protective potential have not yet been fully elucidated. The present study sought to investigate whether resveratrol could protect RGCs and retinal function triggered by acute ocular hypertension injury through the SIRT1/NF-κB pathway. An experimental glaucoma model was generated in C57BL/6J mice. Resveratrol was intraperitoneally injected for 5 days. Sirtinol was injected intravitreally on the day of retinal AOH injury. RGC survival was determined using immunostaining. TUNEL staining was conducted to evaluate retinal cell apoptosis. ERG was used to evaluate visual function. The proteins Brn3a, SIRT1, NF-κB, IL-6, Bax, Bcl2, and Cleaved Caspase3 were determined using western blot. The expression and localisation of SIRT1 and NF-κB in the retina were detected by immunofluorescence. Our data indicated that resveratrol treatment significantly increased Brn3a-labelled RGCs and reduced RGC apoptosis caused by AOH injury. Resveratrol administration also remarkably decreased NF-κB, IL-6, Bax, and Cleaved Caspase3 proteins and increased SIRT1 and Bcl2 proteins. Furthermore, resveratrol treatment obviously inhibited the reduction in ERG caused by AOH injury. Importantly, simultaneous administration of resveratrol and sirtinol abrogated the protective effect of resveratrol, decreased NF-κB protein expression, and increased SIRT1 protein levels. These results suggest that resveratrol administration significantly mitigates retinal AOH-induced RGCs loss and retinal dysfunction, and that this neuroprotective effect is partially regulated through the SIRT1/NF-κB pathway.
Collapse
Affiliation(s)
- Kai-Bao Ji
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wan
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Yang
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue-Jun He
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Qiao Xing
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Zhe Hu
- Deparment of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Qi SM, Zhang JT, Zhu HY, Wang Z, Li W. Review on potential effects of traditional Chinese medicine on glaucoma. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116063. [PMID: 36567037 DOI: 10.1016/j.jep.2022.116063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
ETHNIC PHARMACOLOGICAL RELEVANCE Glaucoma is the second most common blindness in the world, which seriously affects the life quality of patients. Traditional Chinese Medicines (TCM), are important plant materials, widely used for ocular disease all over the world. With the help of modern ophthalmic detection technology, TCM has gradually become an important content in the field of ophthalmology, characterized by more targets and lower toxicity. AIM OF THIS REVIEW This review presents an overview of the pathogenesis of glaucoma in both modern and traditional medicines, and summarizes the therapeutic effect of TCM on glaucoma including their formula, crude drugs and active components, and also the application of acupuncture. METHODS A collection and collation of relevant scientific articles from different scientific databases was performed regarding TCM and its application on glaucoma. The therapeutic effects of TCM were summarized and analyzed according to the existing experimental and clinical researches, while the GSE26299 database were employed to screen bioinformatics analysis of glaucoma based on the GEO database chip. RESULTS There were many positive signs showing that TCM could increase the survival rate of retinal ganglion cells, which may be related to its regulation of microcirculation, oxidative stress, and the immune system. Hence, TCM plays an active role in treating glaucoma. In addition, the bioinformatics analysis predicted that the pathogenesis of glaucoma might be related to p53, MAPK, NF-κB signal, as well as other pathways by KEGG analysis, and the results from bioinformatics analysis predicted that PIK3R6, FGF1, and TYRP1 etc. CONCLUSION: TCM exerts definite effects on preventing and treating ocular disease. It could alleviate and treat glaucoma in various ways. The differentiation syndrome should thus be taken as the basis to propose appropriate treatment options of TCM making their application on glaucoma more popular.
Collapse
Affiliation(s)
- Si-Min Qi
- College of Chinese Medicinal Materials, National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, 130118, China; College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, 130118, China.
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, 130118, China.
| | - Hong-Yan Zhu
- College of Chinese Medicinal Materials, National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, 130118, China; College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
5
|
Sun L. F-box and WD repeat domain-containing 7 (FBXW7) mediates the hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) signaling pathway to affect hypoxic-ischemic brain damage in neonatal rats. Bioengineered 2022; 13:560-572. [PMID: 34951343 PMCID: PMC8805906 DOI: 10.1080/21655979.2021.2011635] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/23/2021] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to determine whether F-box and WD repeat domain-containing 7 (FBXW7) can mediate the hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) signaling pathway to affect neonatal hypoxic-ischemic brain damage (HIBD) in neonatal rats. HIBD rats were treated with LV-shFBXW7. Cerebral infarct size was determined by 2,3,5-triphenyltetrazolium chloride (TTC) staining, while microvessel density (MVD) was evaluated by immunohistochemistry. Learning and memory were tested using the Morris water maze (MWM) test. FBXW7 and HIF-1α/VEGF signaling pathway proteins were measured by Western blotting. Brain microvascular endothelial cells (BMECs) were isolated to establish an oxygen-glucose deprivation (OGD) model to evaluate treatment with FBXW7 siRNA. Cell viability was detected using a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay, while cell migration was evaluated using a wound healing assay. The tube formation of BMECs was also assessed. The results demonstrated that HIBD rats exhibited increased protein expression of FBXW7, HIF-1α, and VEGF. HIBD rats also displayed increased cerebral infarct size, prolonged escape latency and a decreased number of platform crossings. However, HIBD rats treated with LV-shFBXW7 exhibited reversal of these changes. In vitro experiments showed that BMECs in the OGD group had significantly decreased cell viability, shorter vascular lumen length, and shorter migration distance than cells in the control group. Moreover, silencing FBXW7 promoted proliferation, tube formation and migration of BMECs. Taken together, silencing FBXW7 upregulates the HIF-1α/VEGF signaling pathway to promote the angiogenesis of neonatal HIBD rats after brain injury, reducing infarct volume and improving recovery of nerve function in HIBD rats.
Collapse
Affiliation(s)
- Ling Sun
- Neonatal Intensive Care Unit, Yantaishan Hospital, Yantai, China
| |
Collapse
|
6
|
Kim ML, Sung KR, Kwon J, Choi GW, Shin JA. Neuroprotective Effect of Statins in a Rat Model of Chronic Ocular Hypertension. Int J Mol Sci 2021; 22:12500. [PMID: 34830387 PMCID: PMC8621698 DOI: 10.3390/ijms222212500] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Glaucoma is an optic neuropathy in which the degeneration of retinal ganglion cells (RGCs) results in irreversible vison loss. Therefore, neuroprotection of RGCs from glaucomatous afflictions is crucial for glaucoma treatment. In this study, we aimed to investigate the beneficial effects of statins in the protection of RGCs using a rat model. Glaucomatous injury was induced in rats by chronic ocular hypertension (OHT) achieved after performing a circumlimbal suture. The rats were given either statins such as simvastatin and atorvastatin or a solvent weekly for 6 weeks. Retina sections underwent hematoxylin and eosin, Brn3a, or cleaved casepase-3 staining to evaluate RGC survival. In addition, modulation of glial activation was assessed. While the retinas without statin treatment exhibited increased RGC death due to chronic OHT, statins promoted the survival of RGCs and reduced apoptosis. Statins also suppressed chronic OHT-mediated glial activation in the retina. Our results demonstrate that statins exert neuroprotective effects in rat retinas exposed to chronic OHT, which may support the prospect of statins being a glaucoma treatment.
Collapse
Affiliation(s)
- Mi-Lyang Kim
- Biomedical Research Center, College of Medicine, University of Ulsan, Asan Medical Center, Seoul 05505, Korea; (M.-L.K.); (G.W.C.); (J.A.S.)
| | - Kyung Rim Sung
- Department of Ophthalmology, College of Medicine, University of Ulsan, Asan Medical Center, Seoul 05505, Korea;
| | - Junki Kwon
- Department of Ophthalmology, College of Medicine, University of Ulsan, Asan Medical Center, Seoul 05505, Korea;
| | - Go Woon Choi
- Biomedical Research Center, College of Medicine, University of Ulsan, Asan Medical Center, Seoul 05505, Korea; (M.-L.K.); (G.W.C.); (J.A.S.)
| | - Jin A Shin
- Biomedical Research Center, College of Medicine, University of Ulsan, Asan Medical Center, Seoul 05505, Korea; (M.-L.K.); (G.W.C.); (J.A.S.)
| |
Collapse
|
7
|
Fudalej E, Justyniarska M, Kasarełło K, Dziedziak J, Szaflik JP, Cudnoch-Jędrzejewska A. Neuroprotective Factors of the Retina and Their Role in Promoting Survival of Retinal Ganglion Cells: A Review. Ophthalmic Res 2021; 64:345-355. [PMID: 33454713 DOI: 10.1159/000514441] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 01/09/2021] [Indexed: 11/19/2022]
Abstract
Retinal ganglion cells (RGCs) play a crucial role in the visual pathway. As their axons form the optic nerve, apoptosis of these cells causes neurodegenerative vision loss. RGC death could be triggered by increased intraocular pressure, advanced glycation end products, or mitochondrial dysfunction. In this review, we summarize the role of some neuroprotective factors in RGC injury: ciliary neurotrophic factor (CNTF), nerve growth factor (NGF), brain-derived neurotrophic factor, vascular endothelial growth factor, pigment epithelium-derived factor, glial cell line-derived neurotrophic factor, and Norrin. Each, in their own unique way, prevents RGC damage caused by glaucoma, ocular hypertension, ischemic neuropathy, and even oxygen-induced retinopathy. These factors are produced mainly by neurons, leukocytes, glial cells, and epithelial cells. Neuroprotective factors act via various signaling pathways, including JAK/STAT, MAPK, TrkA, and TrkB, which promotes RGC survival. Many attempts have been made to develop therapeutic strategies using these factors. There are ongoing clinical trials with CNTF and NGF, but they have not yet been accepted for clinical use.
Collapse
Affiliation(s)
- Ewa Fudalej
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Justyniarska
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Kaja Kasarełło
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland,
| | - Jacek Dziedziak
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,Department of Ophthalmology, SPKSO Ophthalmic University Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Jacek P Szaflik
- Department of Ophthalmology, SPKSO Ophthalmic University Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Rao H, Jalali JA, Johnston TP, Koulen P. Emerging Roles of Dyslipidemia and Hyperglycemia in Diabetic Retinopathy: Molecular Mechanisms and Clinical Perspectives. Front Endocrinol (Lausanne) 2021; 12:620045. [PMID: 33828528 PMCID: PMC8020813 DOI: 10.3389/fendo.2021.620045] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetic retinopathy (DR) is a significant cause of vision loss and a research subject that is constantly being explored for new mechanisms of damage and potential therapeutic options. There are many mechanisms and pathways that provide numerous options for therapeutic interventions to halt disease progression. The purpose of the present literature review is to explore both basic science research and clinical research for proposed mechanisms of damage in diabetic retinopathy to understand the role of triglyceride and cholesterol dysmetabolism in DR progression. This review delineates mechanisms of damage secondary to triglyceride and cholesterol dysmetabolism vs. mechanisms secondary to diabetes to add clarity to the pathogenesis behind each proposed mechanism. We then analyze mechanisms utilized by both triglyceride and cholesterol dysmetabolism and diabetes to elucidate the synergistic, additive, and common mechanisms of damage in diabetic retinopathy. Gathering this research adds clarity to the role dyslipidemia has in DR and an evaluation of the current peer-reviewed basic science and clinical evidence provides a basis to discern new potential therapeutic targets.
Collapse
Affiliation(s)
- Hussain Rao
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Jonathan A. Jalali
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Thomas P. Johnston
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Peter Koulen
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
- *Correspondence: Peter Koulen,
| |
Collapse
|
9
|
Kovacs K, Vaczy A, Fekete K, Kovari P, Atlasz T, Reglodi D, Gabriel R, Gallyas F, Sumegi B. PARP Inhibitor Protects Against Chronic Hypoxia/Reoxygenation-Induced Retinal Injury by Regulation of MAPKs, HIF1α, Nrf2, and NFκB. Invest Ophthalmol Vis Sci 2019; 60:1478-1490. [PMID: 30973576 DOI: 10.1167/iovs.18-25936] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In the eye, chronic hypoxia/reoxygenation (H/R) contributes to the development of a number of ocular disorders. H/R induces the production of reactive oxygen species (ROS), leading to poly(ADP-ribose) polymerase-1 (PARP1) activation that promotes inflammation, cell death, and disease progression. Here, we analyzed the protective effects of the PARP1 inhibitor olaparib in H/R-induced retina injury and investigated the signaling mechanisms involved. Methods A rat retinal H/R model was used to detect histologic and biochemical changes in the retina. Results H/R induced reductions in the thickness of most retinal layers, which were prevented by olaparib. Furthermore, H/R caused increased levels of Akt and glycogen synthase kinase-3β phosphorylation, which were further increased by olaparib, contributing to retina protection. By contrast, H/R-induced c-Jun N-terminal kinase and p38 mitogen-activated protein kinases (MAPK) phosphorylation and activation were reduced by olaparib, via mitogen-activated protein kinase phosphatase 1 (MKP-1) expression. In addition, H/R-induced hypoxia-inducible factor 1α (HIF1α) levels were decreased by olaparib, which possibly contributed to reduced VEGF expression. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression was slightly increased by H/R and was further activated by olaparib. Nuclear factor-κB (NFκB) was also activated by H/R through phosphorylation (Ser536) and acetylation (Lys310) of the p65 subunit, although this was significantly reduced by olaparib. Conclusions Olaparib reduced H/R-induced degenerative changes in retinal morphology. The protective mechanisms of olaparib most probably involved Nrf2 activation and ROS reduction, as well as normalization of HIF1α and related VEGF expression. In addition, olaparib reduced inflammation by NFκB dephosphorylation/inactivation, possibly via the PARP1 inhibition-MKP-1 activation-p38 MAPK inhibition pathway. PARP inhibitors represent potential therapeutics in H/R-induced retinal disease.
Collapse
Affiliation(s)
- Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Alexandra Vaczy
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Petra Kovari
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Tamas Atlasz
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary.,Department of Sportbiology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Robert Gabriel
- Department of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
10
|
Baliño P, Gómez-Cadenas A, López-Malo D, Romero FJ, Muriach M. Is There A Role for Abscisic Acid, A Proven Anti-Inflammatory Agent, in the Treatment of Ischemic Retinopathies? Antioxidants (Basel) 2019; 8:E104. [PMID: 30999583 PMCID: PMC6523110 DOI: 10.3390/antiox8040104] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/03/2019] [Accepted: 04/13/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic retinopathies (IRs) are the main cause of severe visual impairment and sight loss, and are characterized by loss of blood vessels, accompanied by hypoxia, and neovascularization. Actual therapies, based on anti-vascular endothelial growth factor (VEGF) strategies, antioxidants or anti-inflammatory therapies are only partially effective or show some adverse side effects. Abscisic acid (ABA) is a phytohormone present in vegetables and fruits that can be naturally supplied by the dietary intake and has been previously studied for its benefits to human health. It has been demonstrated that ABA plays a key role in glucose metabolism, inflammation, memory and tumor growth. This review focuses on a novel and promising role of ABA as a potential modulator of angiogenesis, oxidative status and inflammatory processes in the retina, which are the most predominant characteristics of the IRs. Thus, this nutraceutical compound might shed some light in new therapeutic strategies focused in the prevention or amelioration of IRs-derived pathologies.
Collapse
Affiliation(s)
- Pablo Baliño
- Unitat predepartamental de Medicina, Universitat Jaume I, 12071 Castellón de la Plana, Spain.
| | - Aurelio Gómez-Cadenas
- Departament de Ciències Agràries i del Medi Natural, Universitat Jaume I, 12071 Castellón de la Plana, Spain.
| | - Daniel López-Malo
- Departamento de Ciencias Biomédicas, Universidad Europea de Valencia, 46010 Valencia, Spain.
| | - Francisco Javier Romero
- Departamento de Ciencias Biomédicas, Universidad Europea de Valencia, 46010 Valencia, Spain.
| | - María Muriach
- Universitat Jaume I, Unitat predepartamental de Medicina, Avda/Sos Baynat, S/N, 12071 Castellón de la Plana, Spain.
| |
Collapse
|
11
|
Sun W, Pan R, Song J, Sun H. The effects of simvastatin preconditioning on the expression of caspase-3 after myocardial ischemia reperfusion injury in rats. Exp Ther Med 2019; 17:2230-2234. [PMID: 30783483 PMCID: PMC6364223 DOI: 10.3892/etm.2019.7164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023] Open
Abstract
Effect of simvastatin on the expression of caspase-3 in myocardial ischemia reperfusion injury in rats was observed to explore the protective effect of caspase-3 through anti-apoptosis mechanism. A total of 48 healthy male SD rats weighing 160–240 g were selected and divided into 4 groups randomly, namely, the blank group, the sham operation group, the ischemia-reperfusion group and the simvastatin group, with 12 rats in each group. The model of SD rats was made by ligation. The loosen ligature made the reperfusion animal model, the occurrence of arrhythmia in the electrocardiogram of lead II in the experimental animal model was observed, and the area of myocardial infarction in the experimental animal models was detected. The number of apoptotic cells was detected by immunohistochemistry, and the expression of caspase-3 was detected by western blotting. The infarct area in the simvastatin group was significantly lower than the ischemia reperfusion group (P<0.05). The positive rate of the expression of caspase-3 and the positive rate of the expression of apoptotic cells in the ischemic reperfusion and simvastatin groups were significantly higher than that of the blank and sham operation groups, and the positive rate of the expression of caspase-3 and apoptotic cells in the simvastatin group was significantly lower than that of the ischemia-reperfusion group (P<0.05). The arrhythmia score of the simvastatin group was significantly lower than that of the ischemia-reperfusion group (P<0.05). Compared with the blank and sham operation groups, the expression of caspase-3 protein in the ischemia-reperfusion and simvastatin groups was significantly increased, and the expression of caspase-3 protein in the simvastatin group was significantly lower than that of the ischemia reperfusion group (P<0.05). Simvastatin has a protective effect on myocardial ischemia-reperfusion injury, which may be related to the reduction of caspase-3 expression and inhibition of apoptosis.
Collapse
Affiliation(s)
- Weixin Sun
- Department of Cardiology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, P.R. China
| | - Renyou Pan
- Department of Cardiology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, P.R. China
| | - Jun Song
- Department of Cardiology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, P.R. China
| | - Honglin Sun
- Department of Pharmacy, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224000, P.R. China
| |
Collapse
|
12
|
Wu ZY, Li H, Tang YJ. Effect of simvastatin on the SIRT2/NF-κB pathway in rats with acute pulmonary embolism. PHARMACEUTICAL BIOLOGY 2018; 56:511-518. [PMID: 31070532 PMCID: PMC6282435 DOI: 10.1080/13880209.2018.1508239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/07/2018] [Accepted: 06/26/2018] [Indexed: 06/09/2023]
Abstract
CONTEXT Statins have been widely used in acute pulmonary embolism (APE), while simvastatin has been well-established for the prevention of pulmonary hypertension, which was supposed to be an attractive recommendation for APE treatment. OBJECTIVE The current article studies the effect of simvastatin on the SIRT2/NF-κB pathway in rats with APE. MATERIALS AND METHODS Sprague-Dawley rats were divided into four groups (n = 24 per group): control group, rats were treated with saline once daily for 14 days before administration of saline (sham group) or a suspension of autologous emboli (APE group), or rats were treated with simvastatin (10 mg/kg) for 14 days before administration of autologous emboli (APE + simvastatin) group. The RVSP, mPAP and the arterial blood gas was analyzed. Besides, plasma inflammatory cytokines and MMPs levels, as well as the expression of SIRT2/NF-κB pathway were determined. RESULTS Compared with the control and sham groups, the levels of mPAP (31.06 ± 3.47 mmHg), RVSP (35.12 ± 6.02 mmHg), A-aDO2 (33.14 ± 6.16 mmHg) and MMP-9 (6.89 ± 0.84 ng/mL) activity were significantly elevated, but PaO2 (66.87 ± 7.85 mmHg) was highly decreased in rats from APE group at 24 h after APE. Meanwhile, the inflammatory changes were aggravated by the enhanced levels of TNF-α (138.85 ± 22.69 pg/mL), IL-1β (128.47 ± 22.14 pg/mL), IL-6 (103.16 ± 13.58 pg/mL) and IL-8 (179.28 ± 25.79 pg/mL), as well as increased NF-κB (5.29 ± 0.47 fold), but reduced SIRT2 (59 ± 6% reduction), and eNOS (61 ± 5% reduction) mRNA in APE rats. APE rats treated with simvastatin led to a significant opposite trend of the above indexes. CONCLUSIONS Simvastatin protects against APE-induced pulmonary artery pressure, hypoxemia and inflammatory changes probably due to the regulation of SIRT2/NF-κB signalling pathway, which suggest that simvastatin may have promising protective effects in patients with APE.
Collapse
Affiliation(s)
- Zhi-Yao Wu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), National Key Clinical Specialty, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), National Key Clinical Specialty, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Jun Tang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), National Key Clinical Specialty, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Carroll CB, Wyse RKH. Simvastatin as a Potential Disease-Modifying Therapy for Patients with Parkinson's Disease: Rationale for Clinical Trial, and Current Progress. JOURNAL OF PARKINSONS DISEASE 2018; 7:545-568. [PMID: 29036837 PMCID: PMC5676977 DOI: 10.3233/jpd-171203] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many now believe the holy grail for the next stage of therapeutic advance surrounds the development of disease-modifying approaches aimed at intercepting the year-on-year neurodegenerative decline experienced by most patients with Parkinson’s disease (PD). Based on recommendations of an international committee of experts who are currently bringing multiple, potentially disease-modifying, PD therapeutics into long-term neuroprotective PD trials, a clinical trial involving 198 patients is underway to determine whether Simvastatin provides protection against chronic neurodegeneration. Statins are widely used to reduce cardiovascular risk, and act as competitive inhibitors of HMG-CoA reductase. It is also known that statins serve as ligands for PPARα, a known arbiter for mitochondrial size and number. Statins possess multiple cholesterol-independent biochemical mechanisms of action, many of which offer neuroprotective potential (suppression of proinflammatory molecules & microglial activation, stimulation of endothelial nitric oxide synthase, inhibition of oxidative stress, attenuation of α-synuclein aggregation, modulation of adaptive immunity, and increased expression of neurotrophic factors). We describe the biochemical, physiological and pharmaceutical credentials that continue to underpin the rationale for taking Simvastatin into a disease-modifying trial in PD patients. While unrelated to the Simvastatin trial (because this conducted in patients who already have PD), we discuss conflicting epidemiological studies which variously suggest that statin use for cardiovascular prophylaxis may increase or decrease risk of developing PD. Finally, since so few disease-modifying PD trials have ever been launched (compared to those of symptomatic therapies), we discuss the rationale of the trial structure we have adopted, decisions made, and lessons learnt so far.
Collapse
Affiliation(s)
- Camille B Carroll
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | | |
Collapse
|
14
|
'Statins in retinal disease'. Eye (Lond) 2018; 32:981-991. [PMID: 29556012 DOI: 10.1038/s41433-018-0066-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/05/2018] [Accepted: 02/12/2018] [Indexed: 01/14/2023] Open
Abstract
Statins are known for their blood cholesterol-lowering effect and are widely used in patients with cardiovascular and metabolic diseases. Research over the past three decades shows that statins have diverse effects on different pathophysiological pathways involved in angiogenesis, inflammation, apoptosis, and anti-oxidation, leading to new therapeutic options. Recently, statins have attracted considerable attention for their immunomodulatory effect. Since immune reactivity has been implicated in a number of retinal diseases, such as uveitis, age-related macular degeneration (AMD) and diabetic retinopathy, there is now a growing body of evidence supporting the beneficial effects of statins in these retinopathies. This review evaluates the relationship between statins and the pathophysiological basis of these diseases, focusing on their potential role in treatment. A PubMed database search and literature review was conducted. Among AMD patients, there is inconsistent evidence regarding protection against development of early AMD or delaying disease progression; though they have been found to reduce the risk of developing choroidal neovascular membranes (CNV). In patients with retinal vein occlusion, there was no evidence to support a therapeutic benefit or a protective role with statins. In patients with diabetic retinopathy, statins demonstrate a reduction in disease progression and improved resolution of diabetic macular oedema (DMO). Among patients with uveitis, statins have a protective effect by reducing the likelihood of uveitis development.
Collapse
|
15
|
The Protective Effects of αB-Crystallin on Ischemia-Reperfusion Injury in the Rat Retina. J Ophthalmol 2017; 2017:7205408. [PMID: 29098085 PMCID: PMC5643040 DOI: 10.1155/2017/7205408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/13/2022] Open
Abstract
To investigate whether αB-crystallin protects against acute retinal ischemic reperfusion injury (I/R) and elucidate the potential antioxidant mechanisms. Retinal I/R injury was made by elevating the intraocular pressure (IOP) 110 mmHg for 60 min, and αB-crystallin (1 × 10−5 g/L) or vehicle solution was administered intravitreously immediately after I/R injury. The animal was sacrificed 24 h, 1 w, and 1 m after the I/R injury. The retina damage was detected by hematoxylin and eosin (HE) staining and electroretinography (ERG). The level of malondialdehyde (MDA), nitric oxide (NO), and the total superoxide dismutase (T-SOD) was determined. An immunohistochemical study was performed to detect the activation of inducible nitric oxide synthase (iNOS) and NF- (nuclear factor-) kappaB (NF-κB) p65. The decrease of retinal thickness and the number of retinal ganglion cells (RGCs) can be suppressed by αB-crystallin. And the amplitudes of a- and b-wave were remarkably greater without αB-crystallin. Similarly, αB-crystallin also significantly decreased the level of MDA and NO and enhanced the activities of T-SOD. The positive expression of iNOS and NF-kappaB p65 was obviously reduced while treated with αB-crystallin. αB-crystallin can inhibit the expression of NF-κB and its antioxidative effect to protect the retina from I/R injury.
Collapse
|
16
|
Ozaki T, Yamashita T, Tomita H, Sugano E, Ishiguro SI. The protection of rat retinal ganglion cells from ischemia/reperfusion injury by the inhibitory peptide of mitochondrial μ-calpain. Biochem Biophys Res Commun 2016; 478:1700-5. [PMID: 27596965 DOI: 10.1016/j.bbrc.2016.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Intracellular Ca(2+)-dependent cysteine proteases such as calpains have been suggested as critical factors in retinal ganglion cell (RGC) death. However, it is unknown whether mitochondrial calpains are involved in RGC death. The purpose of the present study was to determine whether the inhibition of mitochondrial μ-calpain activity protects against RGC death during ischemia/reperfusion (I/R) injury. This study used a well-established rat model of experimental acute glaucoma involving I/R injury. A specific peptide inhibitor of mitochondrial μ-calpain, Tat-μCL, was topically applied to rats via eye drops three times a day for 5 days after I/R. RGC death was determined by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. The truncation of apoptosis-inducing factor (AIF) was determined by western blot analyses. Retinal morphology was determined after staining with hematoxyline and eosin. In addition, the number of Fluoro Gold-labeled RGCs in flat-mounted retinas was used to determine the percentage of surviving RGCs after I/R injury. After 1 day of I/R, RGC death was observed in the ganglion cell layer. Treatment with Tat-μCL eye drops significantly prevented the death of RGCs and the truncation of AIF. After 5 days of I/R, RGC death decreased by approximately 40%. However, Tat-μCL significantly inhibited the decrease in the retinal sections and flat-mounted retinas. The results suggested that mitochondrial μ-calpain is associated with RGC death during I/R injury via truncation of AIF. In addition, the inhibition of mitochondrial μ-calpain activity by Tat-μCL had a neuroprotective effect against I/R-induced RGC death.
Collapse
Affiliation(s)
- Taku Ozaki
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan; Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan.
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Hiroshi Tomita
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan
| | - Eriko Sugano
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, Morioka, Japan
| | - Sei-Ichi Ishiguro
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
17
|
Hartsock MJ, Cho H, Wu L, Chen WJ, Gong J, Duh EJ. A Mouse Model of Retinal Ischemia-Reperfusion Injury Through Elevation of Intraocular Pressure. J Vis Exp 2016. [PMID: 27501124 DOI: 10.3791/54065] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Retinal ischemia-reperfusion (I/R) is a pathophysiological process contributing to cellular damage in multiple ocular conditions, including glaucoma, diabetic retinopathy, and retinal vascular occlusions. Rodent models of I/R injury are providing significant insights into mechanisms and treatment strategies for human I/R injury, especially with regard to neurodegenerative damage in the retinal neurovascular unit. Presented here is a protocol for inducing retinal I/R injury in mice through elevation of intraocular pressure (IOP). In this protocol, the ocular anterior chamber is cannulated with a needle, through which flows the drip of an elevated saline reservoir. Using this drip to raise IOP above systolic arterial blood pressure, a practitioner temporarily halts inner retinal blood flow (ischemia). When circulation is reinstated (reperfusion) by removal of the cannula, severe cellular damage ensues, resulting ultimately in retinal neurodegeneration. Recent studies demonstrate inflammation, vascular permeability, and capillary degeneration as additional elements of this model. Compared to alternative retinal I/R methodologies, such as retinal arterial ligation, retinal I/R injury by elevated IOP offers advantages in its anatomical specificity, experimental tractability, and technical accessibility, presenting itself as a valuable tool for examining neuronal pathogenesis and therapy in the retinal neurovascular unit.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Hongkwan Cho
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Lijuan Wu
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Wan-Ju Chen
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Junsong Gong
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Elia J Duh
- Department of Ophthalmology, School of Medicine, Johns Hopkins University;
| |
Collapse
|
18
|
Sohn EJ, Shin MJ, Eum WS, Kim DW, Yong JI, Ryu EJ, Park JH, Cho SB, Cha HJ, Kim SJ, Yeo HJ, Yeo EJ, Choi YJ, Im SK, Kweon HY, Kim DS, Yu YH, Cho SW, Park M, Park J, Cho YJ, Choi SY. Tat-NOL3 protects against hippocampal neuronal cell death induced by oxidative stress through the regulation of apoptotic pathways. Int J Mol Med 2016; 38:225-35. [PMID: 27221790 DOI: 10.3892/ijmm.2016.2596] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 04/28/2016] [Indexed: 11/05/2022] Open
Abstract
Oxidative stress-induced apoptosis is associated with neuronal cell death and ischemia. The NOL3 [nucleolar protein 3 (apoptosis repressor with CARD domain)] protein protects against oxidative stress-induced cell death. However, the protective mechanism responsible for this effect as well as the effects of NOL3 against oxidative stress in ischemia remain unclear. Thus, we examined the protective effects of NOL3 protein on hydrogen peroxide (H2O2)-induced oxidative stress and the mechanism responsible for these effects in hippocampal neuronal HT22 cells and in an animal model of forebrain ischemia using Tat-fused NOL3 protein (Tat-NOL3). Purified Tat-NOL3 protein transduced into the H2O2-exposed HT22 cells and inhibited the production of reactive oxygen species (ROS), DNA fragmentation and reduced mitochondrial membrane potential (ΔΨm). In addition, Tat-NOL3 prevented neuronal cell death through the regulation of apoptotic signaling pathways including Bax, Bcl-2, caspase-2, -3 and -8, PARP and p53. In addition, Tat-NOL3 protein transduced into the animal brains and significantly protected against neuronal cell death in the CA1 region of the hippocampus by regulating the activation of microglia and astrocytes. Taken together, these findings demonstrate that Tat-NOL3 protein protects against oxidative stress-induced neuronal cell death by regulating oxidative stress and by acting as an anti-apoptotic protein. Thus, we suggest that Tat-NOL3 represents a potential therapeutic agent for protection against ischemic brain injury.
Collapse
Affiliation(s)
- Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Gangwon-do 25457, Republic of Korea
| | - Ji In Yong
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Eun Ji Ryu
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Hyun Ju Cha
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Sang Jin Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Seung Kwon Im
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Hae Young Kweon
- Department of Agricultural Biology, National Academy of Agricultural Sciences, RDA, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-si, Chungnam 31538, Republic of Korea
| | - Yeon Hee Yu
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-si, Chungnam 31538, Republic of Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Meeyoung Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, College of Medicine, Hallym University, Chuncheon, Gangwon-do 24253, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| |
Collapse
|