1
|
Agadagba SK, Yau SY, Liang Y, Dalton K, Thompson B. Bidirectional causality of physical exercise in retinal neuroprotection. Neural Regen Res 2025; 20:3400-3415. [PMID: 39688575 PMCID: PMC11974656 DOI: 10.4103/nrr.nrr-d-24-00942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
Collapse
Affiliation(s)
- Stephen K. Agadagba
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Suk-yu Yau
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Ying Liang
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Kristine Dalton
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Benjamin Thompson
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
2
|
Mohamed-Noriega K, Loya-Garcia D, Vera-Duarte GR, Morales-Wong F, Ortiz-Morales G, Navas A, Graue-Hernandez EO, Ramirez-Miranda A. Ocular Rosacea: An Updated Review. Cornea 2025; 44:525-537. [PMID: 39808113 PMCID: PMC11872267 DOI: 10.1097/ico.0000000000003785] [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: 10/04/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE Ocular rosacea is a chronic inflammatory disorder affecting the ocular surface, often associated with cutaneous rosacea. This review aims to explore its pathogenesis, treatment approaches, and future directions for management. METHODS A review of current literature on the pathophysiology, clinical features, and treatment strategies of ocular rosacea in adults and children (pediatric blepharokeratoconjunctivitis) was conducted. Emerging research on immune dysregulation, microbiome alterations, and potential therapeutic targets was analyzed. RESULTS Ocular rosacea involves dysregulation of the immune and neurovascular systems, with toll-like receptor activation and complement system involvement leading to chronic ocular surface inflammation. Alterations in the ocular microbiome have been implicated in disease progression. Treatment strategies emphasize a stepwise approach, incorporating ocular and skin hygiene, lifestyle modifications, and pharmacological interventions. Recent advancements in understanding the disease mechanisms have led to the exploration of targeted therapies, including biologics and small-molecule inhibitors. CONCLUSIONS Ocular rosacea remains challenging to diagnose and treat, particularly in children (pediatric blepharokeratoconjunctivitis), often leading to delayed intervention and poor outcomes. A multidisciplinary approach, including new therapeutic options, holds promise for improving patient care. Further research into the genetic and molecular basis of ocular rosacea may enable more personalized treatments.
Collapse
Affiliation(s)
- Karim Mohamed-Noriega
- Department of Ophthalmology, University Hospital and Faculty of Medicine, Autonomous University of Nuevo León (UANL), Monterrey, Nuevo Leon, Mexico; and
| | - Denise Loya-Garcia
- Instituto de Oftalmologia Fundacion Conde de Valenciana IAP, Mexico City, Mexico
| | | | - Fernando Morales-Wong
- Department of Ophthalmology, University Hospital and Faculty of Medicine, Autonomous University of Nuevo León (UANL), Monterrey, Nuevo Leon, Mexico; and
| | | | - Alejandro Navas
- Instituto de Oftalmologia Fundacion Conde de Valenciana IAP, Mexico City, Mexico
| | | | | |
Collapse
|
3
|
Yan K, Zhang Q, Liu Q, Han Y, Liu Z. Advances in adhesive hydrogels applied for ophthalmology: An overview focused on the treatment. Theranostics 2025; 15:915-942. [PMID: 39776812 PMCID: PMC11700875 DOI: 10.7150/thno.103266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Adhesive hydrogels, composed of hydrophilic polymers arranged in a three-dimensional network, have emerged as a pivotal innovation in ophthalmology due to their ability to securely adhere to ocular tissues while providing sustained therapeutic effects. The eye, with its delicate structure and specific needs, presents unique challenges for drug delivery and tissue regeneration. This review explores the transformative potential of adhesive hydrogels in addressing these challenges across a range of ocular conditions, including corneal injuries, cataracts, glaucoma, vitreoretinal disorders, and ocular trauma. By detailing the mechanisms of polymerization and adhesion, this paper highlights how these materials can be customized for specific ophthalmic applications, offering insights into their current use and future possibilities. The emphasis is placed on the clinical significance and future directions of adhesive hydrogels in advancing ophthalmic therapy, potentially revolutionizing the treatment of complex eye diseases.
Collapse
Affiliation(s)
- Ke Yan
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Qinghe Zhang
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Qiuping Liu
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Yi Han
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
| | - Zuguo Liu
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang Hunan 421001, China
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen Fujian 361005, China
| |
Collapse
|
4
|
Kaufmann M, Han Z. RPE melanin and its influence on the progression of AMD. Ageing Res Rev 2024; 99:102358. [PMID: 38830546 PMCID: PMC11260545 DOI: 10.1016/j.arr.2024.102358] [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: 02/29/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024]
Abstract
OBJECTIVE The aim of this review article is to summarize the latest findings and current understanding of the origin of melanin in the retinal pigment epithelium (RPE), its function within the RPE, its role in the pathogenesis of age-related macular degeneration (AMD), its effect on retinal development, and its potential therapeutic benefit in the treatment of AMD. METHODS A comprehensive search of peer-reviewed journals was conducted using various combinations of key terms such as "melanin," "retinal pigment epithelium" or "RPE," "age-related macular degeneration" or AMD," "lipofuscin," "oxidative stress," and "albinism." Databases searched include PubMed, Scopus, Science Direct, and Google Scholar. 147 papers published between the years of 1957 and 2023 were considered with an emphasis on recent findings. SUMMARY OF FINDINGS AMD is thought to result from chronic oxidative stress within the RPE that results in cellular dysfunction, metabolic dysregulation, inflammation, and lipofuscin accumulation. Melanin functions as a photoscreener, free radical scavenger, and metal cation binding reservoir within the RPE. RPE melanin does not regenerate, and it undergoes degradation over time in response to chronic light exposure and oxidative stress. RPE melanin is important for retinal development and RPE function, and in the aging eye, melanin loss is associated with increased lipid peroxidation, inflammation, and the accumulation of toxic oxidized cellular products. Therefore, melanin-based treatments may serve to preserve RPE and retinal function in AMD. CONCLUSIONS The pathogenesis of AMD is not fully understood, but RPE dysfunction and melanin loss in response to chronic oxidative stress and inflammation are thought to be primary drivers of the disease. Due to melanin's antioxidative effects, melanin-based nanotechnology represents a promising avenue for the treatment of AMD.
Collapse
Affiliation(s)
- Mary Kaufmann
- University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
5
|
Nolan ND, Cui X, Robbings BM, Demirkol A, Pandey K, Wu WH, Hu HF, Jenny LA, Lin CS, Hass DT, Du J, Hurley JB, Tsang SH. CRISPR editing of anti-anemia drug target rescues independent preclinical models of retinitis pigmentosa. Cell Rep Med 2024; 5:101459. [PMID: 38518771 PMCID: PMC11031380 DOI: 10.1016/j.xcrm.2024.101459] [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: 07/07/2023] [Revised: 12/21/2023] [Accepted: 02/14/2024] [Indexed: 03/24/2024]
Abstract
Retinitis pigmentosa (RP) is one of the most common forms of hereditary neurodegeneration. It is caused by one or more of at least 3,100 mutations in over 80 genes that are primarily expressed in rod photoreceptors. In RP, the primary rod-death phase is followed by cone death, regardless of the underlying gene mutation that drove the initial rod degeneration. Dampening the oxidation of glycolytic end products in rod mitochondria enhances cone survival in divergent etiological disease models independent of the underlying rod-specific gene mutations. Therapeutic editing of the prolyl hydroxylase domain-containing protein gene (PHD2, also known as Egln1) in rod photoreceptors led to the sustained survival of both diseased rods and cones in both preclinical autosomal-recessive and dominant RP models. Adeno-associated virus-mediated CRISPR-based therapeutic reprogramming of the aerobic glycolysis node may serve as a gene-agnostic treatment for patients with various forms of RP.
Collapse
Affiliation(s)
- Nicholas D Nolan
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Xuan Cui
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Brian M Robbings
- Department of Biochemistry, The University of Washington, Seattle, WA 98195, USA; Diabetes Institute, The University of Washington, Seattle, WA 98195, USA
| | - Aykut Demirkol
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA; Vocational School of Health Services, Uskudar University, 34672 Istanbul, Turkey
| | - Kriti Pandey
- Department of Biochemistry, The University of Washington, Seattle, WA 98195, USA
| | - Wen-Hsuan Wu
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Hannah F Hu
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Laura A Jenny
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Chyuan-Sheng Lin
- Herbert Irving Comprehensive Cancer Center, Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Departments of Ophthalmology, Pathology & Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniel T Hass
- Department of Biochemistry, The University of Washington, Seattle, WA 98195, USA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26501, USA
| | - James B Hurley
- Department of Biochemistry, The University of Washington, Seattle, WA 98195, USA.
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Institute of Human Nutrition, Columbia Stem Cell Initiative, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA; Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York-Presbyterian Hospital, New York, NY 10032, USA; Departments of Ophthalmology, Pathology & Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
6
|
Liu Y, Lin Y, Lin Y, Lin C, Lan G, Su Y, Hu F, Chang K, Chen V, Yeh Y, Chen T, Yu J. Injectable, Antioxidative, and Tissue-Adhesive Nanocomposite Hydrogel as a Potential Treatment for Inner Retina Injuries. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308635. [PMID: 38233151 PMCID: PMC10953571 DOI: 10.1002/advs.202308635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Reactive oxygen species (ROS) have been recognized as prevalent contributors to the development of inner retinal injuries including optic neuropathies such as glaucoma, non-arteritic anterior ischemic optic neuropathy, traumatic optic neuropathy, and Leber hereditary optic neuropathy, among others. This underscores the pivotal significance of oxidative stress in the damage inflicted upon retinal tissue. To combat ROS-related challenges, this study focuses on creating an injectable and tissue-adhesive hydrogel with tailored antioxidant properties for retinal applications. GelCA, a gelatin-modified hydrogel with photo-crosslinkable and injectable properties, is developed. To enhance its antioxidant capabilities, curcumin-loaded polydopamine nanoparticles (Cur@PDA NPs) are incorporated into the GelCA matrix, resulting in a multifunctional nanocomposite hydrogel referred to as Cur@PDA@GelCA. This hydrogel exhibits excellent biocompatibility in both in vitro and in vivo assessments, along with enhanced tissue adhesion facilitated by NPs in an in vivo model. Importantly, Cur@PDA@GelCA demonstrates the potential to mitigate oxidative stress when administered via intravitreal injection in retinal injury models such as the optic nerve crush model. These findings underscore its promise in advancing retinal tissue engineering and providing an innovative strategy for acute neuroprotection in the context of inner retinal injuries.
Collapse
Affiliation(s)
- Yi‐Chen Liu
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Yi‐Ke Lin
- Department of OphthalmologyCollege of MedicineNational Taiwan UniversityTaipei100233Taiwan
| | - Yu‐Ting Lin
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Che‐Wei Lin
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Guan‐Yu Lan
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Yu‐Chia Su
- Institute of Polymer Science and EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Fung‐Rong Hu
- Department of OphthalmologyCollege of MedicineNational Taiwan UniversityTaipei100233Taiwan
- Department of OphthalmologyNational Taiwan University HospitalTaipei100225Taiwan
| | - Kai‐Hsiang Chang
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Vincent Chen
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Yi‐Cheun Yeh
- Institute of Polymer Science and EngineeringNational Taiwan UniversityTaipei10617Taiwan
| | - Ta‐Ching Chen
- Department of OphthalmologyNational Taiwan University HospitalTaipei100225Taiwan
- Center of Frontier MedicineNational Taiwan University HospitalTaipei100225Taiwan
| | - Jiashing Yu
- Department of Chemical EngineeringNational Taiwan UniversityTaipei10617Taiwan
| |
Collapse
|
7
|
Li H, Zhang H, Chen L, Shen Y, Cao Y, Li X, Yao J. Indirubin alleviates retinal neurodegeneration through the regulation of PI3K/AKT signaling. J Biomed Res 2024; 38:256-268. [PMID: 38387889 PMCID: PMC11144936 DOI: 10.7555/jbr.37.20230078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 02/24/2024] Open
Abstract
Retinal neurodegenerative disease is a leading cause of blindness among the elderly in developed countries, including glaucoma, diabetic retinopathy, traumatic optic neuropathy and optic neuritis, etc. The current clinical treatment is not very effective. We investigated indirubin, one of the main bioactive components of the traditional Chinese medicine Danggui Longhui Pill, in the present study for its role in retinal neurodegeneration. Indirubin exhibited no detectable tissue toxicity in vivo or cytotoxicity in vitro. Moreover, indirubin improved visual function and ameliorated retinal neurodegeneration in mice after optic nerve crush injury in vivo. Furthermore, indirubin reduced the apoptosis of retinal ganglion cells induced by oxidative stress in vitro. In addition, indirubin significantly suppressed the increased production of intracellular reactive oxygen species and the decreased activity of superoxide dismutase induced by oxidative stress. Mechanically, indirubin played a neuroprotective role by regulating the PI3K/AKT/BAD/BCL-2 signaling. In conclusion, indirubin protected retinal ganglion cells from oxidative damage and alleviated retinal neurodegeneration induced by optic nerve crush injury. The present study provides a potential therapeutic medicine for retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Huan Li
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Huiying Zhang
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lushu Chen
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yaming Shen
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yuan Cao
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiumiao Li
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jin Yao
- Department of Ophthalmology, the Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Ophthalmology, the Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
8
|
Callan A, Jha S, Valdez L, Tsin A. Cellular and Molecular Mechanisms of Neuronal Degeneration in Early-Stage Diabetic Retinopathy. Curr Vasc Pharmacol 2024; 22:301-315. [PMID: 38693745 DOI: 10.2174/0115701611272737240426050930] [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: 07/18/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Studies on the early retinal changes in Diabetic Retinopathy (DR) have demonstrated that neurodegeneration precedes vascular abnormalities like microaneurysms or intraretinal hemorrhages. Therefore, there is a growing field of study to analyze the cellular and molecular pathways involved to allow for the development of novel therapeutics to prevent the onset or delay the progression of DR. Molecular Mechanisms: Oxidative stress and mitochondrial dysfunction contribute to neurodegeneration through pathways involving polyol, hexosamine, advanced glycation end products, and protein kinase C. Potential interventions targeting these pathways include aldose reductase inhibitors and protein kinase C inhibitors. Neurotrophic factor imbalances, notably brain-derived neurotrophic factor and nerve growth factor, also play a role in early neurodegeneration, and supplementation of these neurotrophic factors show promise in mitigating neurodegeneration. Cellular Mechanisms: Major cellular mechanisms of neurodegeneration include caspase-mediated apoptosis, glial cell reactivity, and glutamate excitotoxicity. Therefore, inhibitors of these pathways are potential therapeutic avenues. Vascular Component: The nitric oxide pathway, critical for neurovascular coupling, is disrupted in DR due to increased reactive oxygen species. Vascular Endothelial Growth Factor (VEGF), a long-known angiogenic factor, has demonstrated both damaging and neuroprotective effects, prompting a careful consideration of long-term anti-VEGF therapy. CONCLUSION Current DR treatments primarily address vascular symptoms but fall short of preventing or halting the disease. Insights into the mechanisms of retinal neurodegeneration in the setting of diabetes mellitus not only enhance our understanding of DR but also pave the way for future therapeutic interventions aimed at preventing disease progression and preserving vision.
Collapse
Affiliation(s)
- Andrew Callan
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, USA
| | - Sonal Jha
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, USA
| | - Laura Valdez
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, USA
| | - Andrew Tsin
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, USA
| |
Collapse
|
9
|
Cornwell A, Badiei A. The role of hydrogen sulfide in the retina. Exp Eye Res 2023; 234:109568. [PMID: 37460081 DOI: 10.1016/j.exer.2023.109568] [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: 01/19/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023]
Abstract
The discovery of the hydrogen sulfide (H2S) and the transsulfuration pathway (TSP) responsible for its synthesis in the mammalian retina has highlighted this molecule's wide range of physiological processes that influence cellular signaling, redox homeostasis, and cellular metabolism. The multi-level regulatory program that influences H2S levels in the retina depends on the relative expression and activity of TSP enzymes, which regulate the abundance of competitive substrates that support or abrogate H2S synthesis. In addition, and apart from TSP, intracellular H2S levels are regulated by mitochondrial sulfide oxidizing pathways. Retinal layers natively express differing levels of TSP enzymes, which highlight the differences in the metabolite and substrate requirement. Recent studies indicate that these systems are susceptible to pathophysiologies affecting the retina. Dysregulation at any level can upset the balance of redox and signaling processes and possibly upset oxidative stress, apoptotic signaling, ion channels, and immune response within this sensitive tissue. H2S donors are a potential therapeutic in such cases and have been demonstrated to bridge the gap, positively impacting the damaged retina. Here, we review the recent findings of H2S, how its multi-level regulation impacts the retina, and how its dysregulation is implicated in retinal pathologies.
Collapse
Affiliation(s)
- Alex Cornwell
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, 99775, AK, USA
| | - Alireza Badiei
- Department of Veterinary Medicine, University of Alaska Fairbanks, Fairbanks, 99775, AK, USA.
| |
Collapse
|
10
|
Yeh JL, Kuo CH, Shih PW, Hsu JH, I-Chen P, Huang YH. Xanthine derivative KMUP-1 ameliorates retinopathy. Biomed Pharmacother 2023; 165:115109. [PMID: 37406513 DOI: 10.1016/j.biopha.2023.115109] [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: 03/21/2023] [Revised: 06/17/2023] [Accepted: 06/28/2023] [Indexed: 07/07/2023] Open
Abstract
Retinal neovascularization (RNV) and cell apoptosis observed in retinopathy are the most common cause of vision loss worldwide. Increasing vascular endothelial growth factor (VEGF), which was driven by hypoxia or inflammation, would result in RNV. This study investigated the anti-inflammatory and anti-apoptotic xanthine-based derivative KMUP-1 on hypoxia-induced conditions in vitro and in vivo. In the oxygen-induced retinopathy animal model, KMUP-1 mitigated vaso-obliteration and neovascularization. In the cell model of hypoxic endothelium cultured at 1% O2, KMUP-1 inhibited endothelial migration and tube formation and had no cytotoxic effect on cell growth. Upregulation of pro-angiogenic factors, HIF-1α and VEGF, and pro-inflammatory cytokines, IL-1β and TNF-α, expression in the retinal-derived endothelial cells, RF/6 A cells, upon hypoxia stimulation, was suppressed by KMUP-1 treatment. RF/6 A cells treated with KMUP-1 showed a reduction of PI3K/Akt, ERK, and RhoA/ROCKs signaling pathways and induction of protective pathways such as eNOS and soluble guanylyl cyclase at 1% O2. Furthermore, KMUP-1 decreased the expression of VEGF, ICAM-1, TNF-α, and IL-1β and increased the BCL-2/BAX ratio in the oxygen-induced retinopathy mouse retina samples. In conclusion, the results of this study suggest that KMUP-1 has potential therapeutic value in retinopathy due to its triple effects on anti-angiogenesis, anti-inflammation, and anti-apoptosis in hypoxic endothelium.
Collapse
Affiliation(s)
- Jwu-Lai Yeh
- Department of Pharmacology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, 80424 Kaohsiung, Taiwan
| | - Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 70101, Taiwan
| | - Po-Wen Shih
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jong-Hau Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; Department of Pediatrics, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Peng I-Chen
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Hsun Huang
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
11
|
Khalili H, Kashkoli HH, Weyland DE, Pirkalkhoran S, Grabowska WR. Advanced Therapy Medicinal Products for Age-Related Macular Degeneration; Scaffold Fabrication and Delivery Methods. Pharmaceuticals (Basel) 2023; 16:620. [PMID: 37111377 PMCID: PMC10146656 DOI: 10.3390/ph16040620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Retinal degenerative diseases such as age-related macular degeneration (AMD) represent a leading cause of blindness, resulting in permanent damage to retinal cells that are essential for maintaining normal vision. Around 12% of people over the age of 65 have some form of retinal degenerative disease. Whilst antibody-based drugs have revolutionised treatment of neovascular AMD, they are only effective at an early stage and cannot prevent eventual progression or allow recovery of previously lost vision. Hence, there is a clear unmet need to find innovative treatment strategies to develop a long-term cure. The replacement of damaged retinal cells is thought to be the best therapeutic strategy for the treatment of patients with retinal degeneration. Advanced therapy medicinal products (ATMPs) are a group of innovative and complex biological products including cell therapy medicinal products, gene therapy medicinal products, and tissue engineered products. Development of ATMPs for the treatment of retinal degeneration diseases has become a fast-growing field of research because it offers the potential to replace damaged retinal cells for long-term treatment of AMD. While gene therapy has shown encouraging results, its effectiveness for treatment of retinal disease may be hampered by the body's response and problems associated with inflammation in the eye. In this mini-review, we focus on describing ATMP approaches including cell- and gene-based therapies for treatment of AMD along with their applications. We also aim to provide a brief overview of biological substitutes, also known as scaffolds, that can be used for delivery of cells to the target tissue and describe biomechanical properties required for optimal delivery. We describe different fabrication methods for preparing cell-scaffolds and explain how the use of artificial intelligence (AI) can aid with the process. We predict that combining AI with 3D bioprinting for 3D cell-scaffold fabrication could potentially revolutionise retinal tissue engineering and open up new opportunities for developing innovative platforms to deliver therapeutic agents to the target tissues.
Collapse
Affiliation(s)
- Hanieh Khalili
- School of Biomedical Science, University of West London, London W5 5RF, UK
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | | | | | - Sama Pirkalkhoran
- School of Biomedical Science, University of West London, London W5 5RF, UK
| | | |
Collapse
|
12
|
Sanches ES, Boia R, Leitão RA, Madeira MH, Fontes-Ribeiro CA, Ambrósio AF, Fernandes R, Silva AP. Attention-Deficit/Hyperactivity Disorder Animal Model Presents Retinal Alterations and Methylphenidate Has a Differential Effect in ADHD versus Control Conditions. Antioxidants (Basel) 2023; 12:antiox12040937. [PMID: 37107312 PMCID: PMC10135983 DOI: 10.3390/antiox12040937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is one of the most prevalent neurodevelopmental disorders. Interestingly, children with ADHD seem to experience more ophthalmologic abnormalities, and the impact of methylphenidate (MPH) use on retinal physiology remains unclear. Thus, we aimed to unravel the retina's structural, functional, and cellular alterations and the impact of MPH in ADHD versus the control conditions. For that, spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY) were used as animal models of ADHD and the controls, respectively. Animals were divided into four experimental groups as follows: WKY vehicle (Veh; tap water), WKY MPH (1.5 mg/kg/day), SHR Veh, SHR MPH. Individual administration was performed by gavage between P28-P55. Retinal physiology and structure were evaluated at P56 followed by tissue collection and analysis. The ADHD animal model presents the retinal structural, functional, and neuronal deficits, as well as the microglial reactivity, astrogliosis, blood-retinal barrier (BRB) hyperpermeability and a pro-inflammatory status. In this model, MPH had a beneficial effect on reducing microgliosis, BRB dysfunction, and inflammatory response, but did not correct the neuronal and functional alterations in the retina. Curiously, in the control animals, MPH showed an opposite effect since it impaired the retinal function, neuronal cells, and BRB integrity, and also promoted both microglia reactivity and upregulation of pro-inflammatory mediators. This study unveils the retinal alterations in ADHD and the opposite effects induced by MPH in the retina of ADHD and the control animal models.
Collapse
Affiliation(s)
- Eliane S Sanches
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Ricardo A Leitão
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Maria H Madeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Carlos A Fontes-Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
13
|
Grebstad Tune B, Melheim M, Åsegg-Atneosen M, Dotinga B, Saugstad OD, Solberg R, Baumbusch LO. Long Non-Coding RNAs in Hypoxia and Oxidative Stress: Novel Insights Investigating a Piglet Model of Perinatal Asphyxia. BIOLOGY 2023; 12:biology12040549. [PMID: 37106749 PMCID: PMC10135607 DOI: 10.3390/biology12040549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Birth asphyxia is the leading cause of death and disability in young children worldwide. Long non-coding RNAs (lncRNAs) may provide novel targets and intervention strategies due to their regulatory potential, as demonstrated in various diseases and conditions. We investigated cardinal lncRNAs involved in oxidative stress, hypoxia, apoptosis, and DNA damage using a piglet model of perinatal asphyxia. A total of 42 newborn piglets were randomized into 4 study arms: (1) hypoxia–normoxic reoxygenation, (2) hypoxia–3 min of hyperoxic reoxygenation, (3) hypoxia–30 min of hyperoxic reoxygenation, and (4) sham-operated controls. The expression of lncRNAs BDNF-AS, H19, MALAT1, ANRIL, TUG1, and PANDA, together with the related target genes VEGFA, BDNF, TP53, HIF1α, and TNFα, was assessed in the cortex, the hippocampus, the white matter, and the cerebellum using qPCR and Droplet Digital PCR. Exposure to hypoxia–reoxygenation significantly altered the transcription levels of BDNF-AS, H19, MALAT1, and ANRIL. BDNF-AS levels were significantly enhanced after both hypoxia and subsequent hyperoxic reoxygenation, 8% and 100% O2, respectively. Our observations suggest an emerging role for lncRNAs as part of the molecular response to hypoxia-induced damages during perinatal asphyxia. A better understanding of the regulatory properties of BDNF-AS and other lncRNAs may reveal novel targets and intervention strategies in the future.
Collapse
Affiliation(s)
- Benedicte Grebstad Tune
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Health, Nutrition and Management, Oslo Metropolitan University, 0130 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Maria Melheim
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
| | | | - Baukje Dotinga
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pediatrics, Division of Neonatology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Ola Didrik Saugstad
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Rønnaug Solberg
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pediatrics, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Lars Oliver Baumbusch
- Department of Pediatric Research, Division of Paediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Faculty of Health, Welfare and Organization, Østfold University College, 1757 Halden, Norway
| |
Collapse
|
14
|
Sarmah DT, Gujjar S, Mathapati S, Bairagi N, Chatterjee S. Identification of critical autophagy-related proteins in diabetic retinopathy: A multi-dimensional computational study. Gene 2023; 866:147339. [PMID: 36882123 DOI: 10.1016/j.gene.2023.147339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
Diabetic retinopathy (DR) is a common consequence of diabetes mellitus and a primary cause of visual impairment in middle-aged and elderly individuals. DR is susceptible to cellular degradation facilitated by autophagy. In this study, we have employed a multi-layer relatedness (MLR) approach to uncover novel autophagy-related proteins involved in DR. The objective of MLR is to determine the relatedness of autophagic and DR proteins by incorporating both expression and prior-knowledge-based similarities. We constructed a prior knowledge-based network and identified the topologically significant novel disease-related candidate autophagic proteins (CAPs). Then, we evaluated their significance in a gene co-expression and a differentially-expressed gene (DEG) network. Finally, we investigated the proximity of CAPs to the known disease-related proteins. Leveraging this methodology, we identified three crucial autophagy-related proteins, TP53, HSAP90AA1, and PIK3R1, which can influence the DR interactome in various layers of heterogeneity of clinical manifestations. They are strongly related to multiple detrimental characteristics of DR, such as pericyte loss, angiogenesis, apoptosis, and endothelial cell migration, and hence may be used to prevent or delay the progression and development of DR. We evaluated one of the identified targets, TP53, in a cell-based model and found that its inhibition resulted in reduced angiogenesis in high glucose condition required to control DR.
Collapse
Affiliation(s)
- Dipanka Tanu Sarmah
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Sunil Gujjar
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Santosh Mathapati
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Nandadulal Bairagi
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata 700032, India
| | - Samrat Chatterjee
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India.
| |
Collapse
|
15
|
Bianchetti G, Clementi ME, Sampaolese B, Serantoni C, Abeltino A, De Spirito M, Sasson S, Maulucci G. Metabolic Imaging and Molecular Biology Reveal the Interplay between Lipid Metabolism and DHA-Induced Modulation of Redox Homeostasis in RPE Cells. Antioxidants (Basel) 2023; 12:339. [PMID: 36829896 PMCID: PMC9952658 DOI: 10.3390/antiox12020339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetes-induced oxidative stress induces the development of vascular complications, which are significant causes of morbidity and mortality in diabetic patients. Among these, diabetic retinopathy (DR) is often caused by functional changes in the blood-retinal barrier (BRB) due to harmful oxidative stress events in lipids, proteins, and DNA. Docosahexaenoic acid (DHA) has a potential therapeutic effect against hyperglycemia-induced oxidative damage and apoptotic pathways in the main constituents of BRB, retinal pigment epithelium cells (ARPE-19). Effective antioxidant response elicited by DHA is driven by the activation of the Nrf2/Nqo1 signaling cascade, which leads to the formation of NADH, a reductive agent found in the cytoplasm. Nrf2 also induces the expression of genes encoding enzymes involved in lipid metabolism. This study, therefore, aims at investigating the modulation of lipid metabolism induced by high-glucose (HG) on ARPE-19 cells through the integration of metabolic imaging and molecular biology to provide a comprehensive functional and molecular characterization of the mechanisms activated in the disease, as well the therapeutic role of DHA. This study shows that HG augments RPE metabolic processes by enhancing lipid metabolism, from fatty acid uptake and turnover to lipid biosynthesis and β-oxidation. DHA exerts its beneficial effect by ameliorating lipid metabolism and reducing the increased ROS production under HG conditions. This investigation may provide novel insight for formulating novel treatments for DR by targeting lipid metabolism pathways.
Collapse
Affiliation(s)
- Giada Bianchetti
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Maria Elisabetta Clementi
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Largo Francesco Vito, 1, 00168 Rome, Italy
| | - Beatrice Sampaolese
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Largo Francesco Vito, 1, 00168 Rome, Italy
| | - Cassandra Serantoni
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Alessio Abeltino
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Marco De Spirito
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Shlomo Sasson
- Faculty of Medicine, Institute for Drug Research, The Hebrew University, Jerusalem 911210, Israel
| | - Giuseppe Maulucci
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| |
Collapse
|
16
|
Ma X, Dai Y, Qiu T, Chen X, Xiao P, Li W. Effects of acute exposure to amisulbrom on retinal development in zebrafish (Danio rerio) embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:46248-46256. [PMID: 36715803 DOI: 10.1007/s11356-023-25584-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Amisulbrom is an oomycete-specific fungicide that was developed by Nissan Chemical Industries Limited. The exposure of developing zebrafish embryo to amisulbrom caused disorders in the visual phototransduction system. However, the potential toxic mechanisms of amisulbrom on retinal development remains unclear. The research purpose of this study was to evaluate the adverse effects of amisulbrom on retinal development in a model organism, the zebrafish. Zebrafish embryos were treated with 0, 0.0075, 0.075, or 0.75 μM amisulbrom from 3 h post-fertilization (hpf) to 72 hpf. Compared with the control group, amisulbrom-treated zebrafish embryos displayed phenotypic microphthalmia, dysregulation of gene transcription levels (alcama, prox1a, sox2, vsx1, rho, bluops, rdops, uvops, and grops) related to the retinal cell layer differentiation, and increased retinal apoptosis. In addition, the content of glutathione and malondialdehyde increased significantly after exposure to amisulbrom. Overall, our data demonstrate the toxicity of amisulbrom to eye development, which will help to assess the potential ecotoxicological impacts posed by amisulbrom to aquatic species.
Collapse
Affiliation(s)
- Xueying Ma
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Yizhe Dai
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Tiantong Qiu
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Xin Chen
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Peng Xiao
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Zhejiang Provincial Key Lab for Water Environment and Marine Biological Resources Protection, Institute for Eco-Environmental Research of Sanyang Wetland, College of Life and Environmental Science, Wenzhou University, 325035, Wenzhou, People's Republic of China
| | - Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China.
| |
Collapse
|
17
|
Shu DY, Chaudhary S, Cho KS, Lennikov A, Miller WP, Thorn DC, Yang M, McKay TB. Role of Oxidative Stress in Ocular Diseases: A Balancing Act. Metabolites 2023; 13:187. [PMID: 36837806 PMCID: PMC9960073 DOI: 10.3390/metabo13020187] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Redox homeostasis is a delicate balancing act of maintaining appropriate levels of antioxidant defense mechanisms and reactive oxidizing oxygen and nitrogen species. Any disruption of this balance leads to oxidative stress, which is a key pathogenic factor in several ocular diseases. In this review, we present the current evidence for oxidative stress and mitochondrial dysfunction in conditions affecting both the anterior segment (e.g., dry eye disease, keratoconus, cataract) and posterior segment (age-related macular degeneration, proliferative vitreoretinopathy, diabetic retinopathy, glaucoma) of the human eye. We posit that further development of therapeutic interventions to promote pro-regenerative responses and maintenance of the redox balance may delay or prevent the progression of these major ocular pathologies. Continued efforts in this field will not only yield a better understanding of the molecular mechanisms underlying the pathogenesis of ocular diseases but also enable the identification of novel druggable redox targets and antioxidant therapies.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Suman Chaudhary
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - William P. Miller
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David C. Thorn
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tina B. McKay
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
18
|
Shin CS, Veettil RA, Sakthivel TS, Adumbumkulath A, Lee R, Zaheer M, Kolanthai E, Seal S, Acharya G. Noninvasive Delivery of Self-Regenerating Cerium Oxide Nanoparticles to Modulate Oxidative Stress in the Retina. ACS APPLIED BIO MATERIALS 2022; 5:5816-5825. [PMID: 36441967 DOI: 10.1021/acsabm.2c00809] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Diseases affecting the retina, such as age-related macular degeneration (AMD), diabetic retinopathy, macular edema, and retinal vein occlusions, are currently treated by the intravitreal injection of drug formulations. These disease pathologies are driven by oxidative damage due to chronic high concentrations of reactive oxygen species (ROS) in the retina. Intravitreal injections often induce retinal detachment, intraocular hemorrhage, and endophthalmitis. Furthermore, the severe eye pain associated with these injections lead to patient noncompliance and treatment discontinuation. Hence, there is a critical need for the development of a noninvasive therapy that is effective for a prolonged period for treating retinal diseases. In this study, we developed a noninvasive cerium oxide nanoparticle (CNP) delivery wafer (Cerawafer) for the modulation of ROS in the retina. We fabricated Cerawafer loaded with CNP and determined its SOD-like enzyme-mimetic activity and ability to neutralize ROS generated in vitro. We demonstrated Cerawafer's ability to deliver CNP in a noninvasive fashion to the retina in healthy mouse eyes and the CNP retention in the retina for more than a week. Our studies have demonstrated the in vivo efficacy of the Cerawafer to modulate ROS and associated down-regulation of VEGF expression in the retinas of very-low-density lipoprotein receptor knockout (vldlr-/-) mouse model. The development of a Cerawafer nanotherapeutic will fulfill a hitherto unmet need. Currently, there is no such therapeutic available, and the development of a Cerawafer nanotherapeutic will be a major advancement in the treatment of retinal diseases.
Collapse
Affiliation(s)
- Crystal S Shin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Remya Ammassam Veettil
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Tamil S Sakthivel
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, Nanoscience Technology Center, University of Central Florida, Orlando, Florida 32816, United States
| | - Aparna Adumbumkulath
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States.,Department of Materials Science and Nanoengineering, Rice University, Houston, Texas 77030, United States
| | - Richard Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Mahira Zaheer
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, Nanoscience Technology Center, University of Central Florida, Orlando, Florida 32816, United States
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, Nanoscience Technology Center, University of Central Florida, Orlando, Florida 32816, United States.,College of Medicine, Biionix Cluster, University of Central Florida, Orlando, Florida 32827, United States
| | - Ghanashyam Acharya
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States.,Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, United States.,Department of Materials Science and Nanoengineering, Rice University, Houston, Texas 77030, United States
| |
Collapse
|
19
|
Allyn MM, Rincon-Benavides MA, Chandler HL, Higuita-Castro N, Palmer AF, Swindle-Reilly KE. Sustained release of heme-albumin as a potential novel therapeutic approach for age-related macular degeneration. Biomater Sci 2022; 10:7004-7014. [PMID: 36342429 DOI: 10.1039/d2bm00905f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Globally, age-related macular degeneration (AMD) is the third most common visual impairment. Most often attributed to cellular fatigue with aging, over expression of reactive oxygen species (ROS) causes ROS accumulation in the retina, leading to chronic inflammatory immune signaling, cellular and tissue damage, and eventual blindness. If left uncontrolled, the disease will progress from the dry form of AMD to more severe forms such as geographic atrophy or wet AMD, hallmarked by choroidal neovascularization. There is no cure for AMD and treatment options are limited. Treatment options for wet AMD require invasive ocular injections or implants, yet fail to address the disease progressing factors. To provide more complete treatment of AMD, the application of a novel anti-inflammatory heme-bound human serum albumin (heme-albumin) protein complex delivered by antioxidant ROS scavenging polydopamine (PDA) nanoparticles (NPs) for sustained treatment of AMD was investigated. Through the induction of heme oxygenase-1 (HO-1) by heme-albumin in retinal pigment epithelial (RPE) cells, anti-inflammatory protection may be provided through the generation of carbon monoxide (CO) and biliverdin during heme catabolism. Our results show that the novel protein complex has negligible cytotoxicity towards RPE cells (ARPE-19), reduces oxidative stress in both inflammatory and ROS in vitro models, and induces a statistically significant increase in HO-1 protein expression. When incorporated into PDA NPs, heme-albumin was sustainably released for up to 6 months, showing faster release at higher oxidative stress levels. Through its ability to react with ROS, heme-albumin loaded PDA NPs showed further reduction of oxidative stress with minimal cytotoxicity. Altogether, we demonstrate that heme-albumin loaded PDA NPs reduce oxidative stress in vitro and can provide sustained therapeutic delivery for AMD treatment.
Collapse
Affiliation(s)
- Megan M Allyn
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 151 W Woodruff Ave, Columbus, OH 43210, USA.
| | - Maria A Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, 140 W 19th Ave, Columbus, OH 43210, USA.,Biophysics Graduate Program, The Ohio State University, 484 W 12th Ave, Columbus, OH 43210, USA
| | - Heather L Chandler
- College of Optometry, The Ohio State University, 338 W 10th Ave, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, 140 W 19th Ave, Columbus, OH 43210, USA.,Department of Surgery, The Ohio State University, 370 W 9th Ave, Columbus, OH 43210, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 151 W Woodruff Ave, Columbus, OH 43210, USA.
| | - Katelyn E Swindle-Reilly
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, 151 W Woodruff Ave, Columbus, OH 43210, USA. .,Department of Biomedical Engineering, The Ohio State University, 140 W 19th Ave, Columbus, OH 43210, USA.,Department of Ophthalmology and Visual Sciences, The Ohio State University, 915 Olentangy River Rd, Columbus, OH 43212, USA
| |
Collapse
|
20
|
Titialii-Torres KF, Morris AC. Embryonic hyperglycemia perturbs the development of specific retinal cell types, including photoreceptors. J Cell Sci 2022; 135:jcs259187. [PMID: 34851372 PMCID: PMC8767273 DOI: 10.1242/jcs.259187] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/18/2021] [Indexed: 01/12/2023] Open
Abstract
Diabetes is linked to various long-term complications in adults, such as neuropathy, nephropathy and diabetic retinopathy. Diabetes poses additional risks for pregnant women, because glucose passes across the placenta, and excess maternal glucose can result in diabetic embryopathy. While many studies have examined the teratogenic effects of maternal diabetes on fetal heart development, little is known about the consequences of maternal hyperglycemia on the development of the embryonic retina. To address this question, we investigated retinal development in two models of embryonic hyperglycemia in zebrafish. Strikingly, we found that hyperglycemic larvae displayed a significant reduction in photoreceptors and horizontal cells, whereas other retinal neurons were not affected. We also observed reactive gliosis and abnormal optokinetic responses in hyperglycemic larvae. Further analysis revealed delayed retinal cell differentiation in hyperglycemic embryos that coincided with increased reactive oxygen species (ROS). Our results suggest that embryonic hyperglycemia causes abnormal retinal development via altered timing of cell differentiation and ROS production, which is accompanied by visual defects. Further studies using zebrafish models of hyperglycemia will allow us to understand the molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Kayla F. Titialii-Torres
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Ann C. Morris
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| |
Collapse
|
21
|
Wei X, Li D, Feng C, Mao H, Zhu J, Cui Y, Yang J, Gao H, Wang C. Effects of hydrogen peroxide and l-tryptophan on antioxidative potential, apoptosis, and mammalian target of rapamycin signaling in bovine intestinal epithelial cells. J Dairy Sci 2022; 105:10007-10019. [DOI: 10.3168/jds.2022-21869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022]
|
22
|
Wang J, Li M, Geng Z, Khattak S, Ji X, Wu D, Dang Y. Role of Oxidative Stress in Retinal Disease and the Early Intervention Strategies: A Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7836828. [PMID: 36275903 PMCID: PMC9586758 DOI: 10.1155/2022/7836828] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 02/05/2023]
Abstract
The retina, owing to its cellular anatomy and physical location, is susceptible to generating reactive oxygen species (ROS), which are associated with several major retinal diseases. When ROS exceeds the body's natural antioxidants, the retina is in a state of oxidative stress, which is recognized as the pathogenesis of retinal diseases. The early stage of the pathogenic process is an adaptive change in which oxidative stress and endogenous defense mechanisms occur. If no treatment is applied, the retinal diseases will progress to the pathological stage with neuronal and vascular dysfunction or damage and even blindness. This review summarizes the role of oxidative stress in several common retinal diseases, including retinitis pigmentosa, age-related macular degeneration, diabetic retinopathy, glaucoma, and retinopathy of prematurity. In addition, we discuss the early intervention strategies for these diseases. An outline is provided to identify potential intervention targets for further research. Early intervention for retinal diseases is necessary and urgent and may offer hope to improve patients' quality of life through functional vision.
Collapse
Affiliation(s)
- Jun Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Mengling Li
- College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ziyue Geng
- School of Clinical Medicine, Henan University, Kaifeng, Henan, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Xinying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Dongdong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yalong Dang
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
- Sanmenxia Central Hospital, Sanmenxia, Henan, China
| |
Collapse
|
23
|
Drinking hydrogen water improves photoreceptor structure and function in retinal degeneration 6 mice. Sci Rep 2022; 12:13610. [PMID: 35948585 PMCID: PMC9365798 DOI: 10.1038/s41598-022-17903-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Retinitis pigmentosa (RP) is a genetically heterogeneous group of inherited retinal disorders involving the progressive dysfunction of photoreceptors and the retinal pigment epithelium, for which there is currently no treatment. The rd6 mouse is a natural model of autosomal recessive retinal degeneration. Given the known contributions of oxidative stress caused by reactive oxygen species (ROS) and selective inhibition of potent ROS peroxynitrite and OH·by H2 gas we have previously demonstrated, we hypothesized that ingestion of H2 water may delay the progression of photoreceptor death in rd6 mice. H2 mice showed significantly higher retinal thickness as compared to controls on optical coherence tomography. Histopathological and morphometric analyses revealed higher thickness of the outer nuclear layer for H2 mice than controls, as well as higher counts of opsin red/green-positive cells. RNA sequencing (RNA-seq) analysis of differentially expressed genes in the H2 group versus control group revealed 1996 genes with significantly different expressions. Gene and pathway ontology analysis showed substantial upregulation of genes responsible for phototransduction in H2 mice. Our results show that drinking water high in H2 (1.2-1.6 ppm) had neuroprotective effects and inhibited photoreceptor death in mice, and suggest the potential of H2 for the treatment of RP.
Collapse
|
24
|
Lv S, Yang H, Jing P, Song H. α-tocopherol pretreatment alleviates cerebral ischemia-reperfusion injury in rats. CNS Neurosci Ther 2022; 28:964-970. [PMID: 35301808 PMCID: PMC9062554 DOI: 10.1111/cns.13814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/14/2022] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shitao Lv
- Department of Emergency, Yantaishan Hospital, Yantai, China
| | - Haiyan Yang
- Department of Emergency, Yantaishan Hospital, Yantai, China
| | | | - Haiying Song
- Department of Gynecology, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
25
|
Srivastava GK, Rodriguez-Crespo D, Fernandez-Bueno I, Pastor JC. Factors influencing mesenchymal stromal cells in in vitro cellular models to study retinal pigment epithelial cell rescue. Hum Cell 2022; 35:1005-1015. [PMID: 35511404 DOI: 10.1007/s13577-022-00705-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/17/2022] [Indexed: 11/29/2022]
Abstract
Mesenchymal stromal cells (MSC) stop or slow retinal pigment epithelium (RPE) and neuroretina (NR) degeneration by paracrine activity in oxidative stress-induced retinal degenerative diseases. However, it is mandatory to develop adequate in vitro models that allow testing new treatment strategies against oxidative stress before performing in vivo studies. The viable double- and triple-layered setups are composed of separate layers of NR, MSC, and RPE (NR-MSC-RPE, NR-RPE, MSC-RPE) partially mimic in vivo retinal conditions. In this study, the paracrine neuroprotective effect of each setup's microenvironment on hydrogen peroxide (H2O2)-stressed was compared with unstressed RPE cells. RPE cell proliferation viability was assessed on day 1, 3, and 6 using Alamar Blue® (10%), MTT (10%) and a cell viability/cytotoxicity assay kit followed by data analysis. The results showed that RPE cells, highly viable (> 90%) in mixed medium of DMEM and neurobasal A (1:1), lost 50% viability on exposure to 400 µM of H2O2 (P < 0.05). The unexposed groups differed significantly from exposed groups for RPE cell growth (RPE and [Formula: see text]RPE (P < 0.0001), NR-MSC-RPE, and NR-MSC-[Formula: see text]RPE (P < 0.05), NR-RPE and NR-[Formula: see text]RPE (P < 0.01), and MSC-RPE and MSC-[Formula: see text]RPE (P < 0.01). NR-[Formula: see text]RPE and NR-RPE supported RPE cell proliferation viability better than other setups (P < 0.01) and RPE cells proliferated 0.49-fold more in NR-MSC-[Formula: see text]RPE than NR-MSC-RPE. Thus, NR and MSC presence improved significantly each setup's microenvironment for cell rescue, nevertheless, each setup also showed limitations for its use as an in vitro study tool. Health of microenvironment of such setups depends on many factors including cell-secreted trophic factors.
Collapse
Affiliation(s)
- Girish K Srivastava
- Retina Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Paseo de Belén, 17, 47011, Valladolid, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla Y León, Valladolid, Spain.
| | - David Rodriguez-Crespo
- Retina Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Paseo de Belén, 17, 47011, Valladolid, Spain
| | - Ivan Fernandez-Bueno
- Retina Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Paseo de Belén, 17, 47011, Valladolid, Spain
| | - José Carlos Pastor
- Retina Group, Instituto Universitario de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Campus Miguel Delibes, Paseo de Belén, 17, 47011, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla Y León, Valladolid, Spain
| |
Collapse
|
26
|
Gajendran MK, Rohowetz LJ, Koulen P, Mehdizadeh A. Novel Machine-Learning Based Framework Using Electroretinography Data for the Detection of Early-Stage Glaucoma. Front Neurosci 2022; 16:869137. [PMID: 35600610 PMCID: PMC9115110 DOI: 10.3389/fnins.2022.869137] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/28/2022] [Indexed: 01/05/2023] Open
Abstract
PurposeEarly-stage glaucoma diagnosis has been a challenging problem in ophthalmology. The current state-of-the-art glaucoma diagnosis techniques do not completely leverage the functional measures' such as electroretinogram's immense potential; instead, focus is on structural measures like optical coherence tomography. The current study aims to take a foundational step toward the development of a novel and reliable predictive framework for early detection of glaucoma using machine-learning-based algorithm capable of leveraging medically relevant information that ERG signals contain.MethodsERG signals from 60 eyes of DBA/2 mice were grouped for binary classification based on age. The signals were also grouped based on intraocular pressure (IOP) for multiclass classification. Statistical and wavelet-based features were engineered and extracted. Important predictors (ERG tests and features) were determined, and the performance of five machine learning-based methods were evaluated.ResultsRandom forest (bagged trees) ensemble classifier provided the best performance in both binary and multiclass classification of ERG signals. An accuracy of 91.7 and 80% was achieved for binary and multiclass classification, respectively, suggesting that machine-learning-based models can detect subtle changes in ERG signals if trained using advanced features such as those based on wavelet analyses.ConclusionsThe present study describes a novel, machine-learning-based method to analyze ERG signals providing additional information that may be used to detect early-stage glaucoma. Based on promising performance metrics obtained using the proposed machine-learning-based framework leveraging an established ERG data set, we conclude that the novel framework allows for detection of functional deficits of early/various stages of glaucoma in mice.
Collapse
Affiliation(s)
- Mohan Kumar Gajendran
- Department of Civil and Mechanical Engineering, School of Computing and Engineering, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Landon J. Rohowetz
- Vision Research Center, Department of Ophthalmology, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Amirfarhang Mehdizadeh
- Department of Civil and Mechanical Engineering, School of Computing and Engineering, University of Missouri-Kansas City, Kansas City, MO, United States
- Vision Research Center, Department of Ophthalmology, University of Missouri-Kansas City, Kansas City, MO, United States
- *Correspondence: Amirfarhang Mehdizadeh
| |
Collapse
|
27
|
Astaxanthin ameliorates hyperglycemia induced inflammation via PI3K/Akt–NF–κB signaling in ARPE-19 cells and diabetic rat retina. Eur J Pharmacol 2022; 926:174979. [DOI: 10.1016/j.ejphar.2022.174979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 01/12/2023]
|
28
|
Santos FM, Mesquita J, Castro-de-Sousa JP, Ciordia S, Paradela A, Tomaz CT. Vitreous Humor Proteome: Targeting Oxidative Stress, Inflammation, and Neurodegeneration in Vitreoretinal Diseases. Antioxidants (Basel) 2022; 11:505. [PMID: 35326156 PMCID: PMC8944522 DOI: 10.3390/antiox11030505] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is defined as an unbalance between pro-oxidants and antioxidants, as evidenced by an increase in reactive oxygen and reactive nitrogen species production over time. It is important in the pathophysiology of retinal disorders such as diabetic retinopathy, age-related macular degeneration, retinal detachment, and proliferative vitreoretinopathy, which are the focus of this article. Although the human organism's defense mechanisms correct autoxidation caused by endogenous or exogenous factors, this may be insufficient, causing an imbalance in favor of excessive ROS production or a weakening of the endogenous antioxidant system, resulting in molecular and cellular damage. Furthermore, modern lifestyles and environmental factors contribute to increased chemical exposure and stress induction, resulting in oxidative stress. In this review, we discuss the current information about oxidative stress and the vitreous proteome with a special focus on vitreoretinal diseases. Additionally, we explore therapies using antioxidants in an attempt to rescue the body from oxidation, restore balance, and maximize healthy body function, as well as new investigational therapies that have shown significant therapeutic potential in preclinical studies and clinical trial outcomes, along with their goals and strategic approaches to combat oxidative stress.
Collapse
Affiliation(s)
- Fátima Milhano Santos
- CICS-UBI—Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal; or (J.P.C.-d.-S.)
- Unidad de Proteomica, Centro Nacional de Biotecnología, CSIC, Campus de Cantoblanco, 28049 Madrid, Spain; (S.C.); (A.P.)
- C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, 6200-501 Covilhã, Portugal
| | - Joana Mesquita
- CICS-UBI—Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal; or (J.P.C.-d.-S.)
| | - João Paulo Castro-de-Sousa
- CICS-UBI—Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal; or (J.P.C.-d.-S.)
- Department of Ophthalmology, Centro Hospitalar de Leiria, 2410-197 Leiria, Portugal
| | - Sergio Ciordia
- Unidad de Proteomica, Centro Nacional de Biotecnología, CSIC, Campus de Cantoblanco, 28049 Madrid, Spain; (S.C.); (A.P.)
| | - Alberto Paradela
- Unidad de Proteomica, Centro Nacional de Biotecnología, CSIC, Campus de Cantoblanco, 28049 Madrid, Spain; (S.C.); (A.P.)
| | - Cândida Teixeira Tomaz
- CICS-UBI—Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal; or (J.P.C.-d.-S.)
- C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, 6200-501 Covilhã, Portugal
- Chemistry Department, Faculty of Sciences, University of Beira Interior, 6201-001 Covilhã, Portugal
| |
Collapse
|
29
|
Stoica SI, Bleotu C, Ciobanu V, Ionescu AM, Albadi I, Onose G, Munteanu C. Considerations about Hypoxic Changes in Neuraxis Tissue Injuries and Recovery. Biomedicines 2022; 10:481. [PMID: 35203690 PMCID: PMC8962344 DOI: 10.3390/biomedicines10020481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/06/2022] [Accepted: 02/13/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxia represents the temporary or longer-term decrease or deprivation of oxygen in organs, tissues, and cells after oxygen supply drops or its excessive consumption. Hypoxia can be (para)-physiological-adaptive-or pathological. Thereby, the mechanisms of hypoxia have many implications, such as in adaptive processes of normal cells, but to the survival of neoplastic ones, too. Ischemia differs from hypoxia as it means a transient or permanent interruption or reduction of the blood supply in a given region or tissue and consequently a poor provision with oxygen and energetic substratum-inflammation and oxidative stress damages generating factors. Considering the implications of hypoxia on nerve tissue cells that go through different ischemic processes, in this paper, we will detail the molecular mechanisms by which such structures feel and adapt to hypoxia. We will present the hypoxic mechanisms and changes in the CNS. Also, we aimed to evaluate acute, subacute, and chronic central nervous hypoxic-ischemic changes, hoping to understand better and systematize some neuro-muscular recovery methods necessary to regain individual independence. To establish the link between CNS hypoxia, ischemic-lesional mechanisms, and neuro-motor and related recovery, we performed a systematic literature review following the" Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA") filtering method by interrogating five international medical renown databases, using, contextually, specific keywords combinations/"syntaxes", with supplementation of the afferent documentation through an amount of freely discovered, also contributive, bibliographic resources. As a result, 45 papers were eligible according to the PRISMA-inspired selection approach, thus covering information on both: intimate/molecular path-physiological specific mechanisms and, respectively, consequent clinical conditions. Such a systematic process is meant to help us construct an article structure skeleton giving a primary objective input about the assembly of the literature background to be approached, summarised, and synthesized. The afferent contextual search (by keywords combination/syntaxes) we have fulfilled considerably reduced the number of obtained articles. We consider this systematic literature review is warranted as hypoxia's mechanisms have opened new perspectives for understanding ischemic changes in the CNS neuraxis tissue/cells, starting at the intracellular level and continuing with experimental research to recover the consequent clinical-functional deficits better.
Collapse
Affiliation(s)
- Simona Isabelle Stoica
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
| | - Coralia Bleotu
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania;
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest (PUB), 060042 Bucharest, Romania;
| | - Anca Mirela Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
| | - Irina Albadi
- Teaching Emergency County Hospital “Sf. Apostol Andrei”, 900591 Constanta, Romania;
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
| | - Constantin Munteanu
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
- Department of Research, Romanian Association of Balneology, 022251 Bucharest, Romania
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| |
Collapse
|
30
|
Ho KL, Yong PH, Wang CW, Kuppusamy UR, Ngo CT, Massawe F, Ng ZX. Peperomia pellucida (L.) Kunth and eye diseases: A review on phytochemistry, pharmacology and toxicology. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:292-304. [PMID: 35153134 DOI: 10.1016/j.joim.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022]
Abstract
Peperomia pellucida (L.) Kunth is a medicinal plant used to manage inflammatory illnesses such as conjunctivitis, and gastrointestinal and respiratory tract disorders in tropical and subtropical regions. However, little is known about its pharmacological mechanism of action against eye diseases. This review aims to critically discuss the phytochemistry, pharmacology and toxicology of P. pellucida as well as its roles in the treatment of cataract, glaucoma and diabetic retinopathy. Recent developments in the uses of P. pellucida for healthcare and nutraceutical products by the pharmaceutical industry are also covered in this review. For this review, a literature search was performed with PubMed, ScienceDirect, SciFinder Scholar and Scopus databases, using relevant keywords. Among the various phytochemicals identified from P. pellucida, β-caryophyllene, carotol, dillapiole, ellagic acid, pellucidin A, phytol and vitexin exhibit strong pharmacological activities within the mitogen-activated protein kinase and nuclear factor-κB signalling pathways in inflammatory eye diseases. The antihypertensive, anti-inflammatory, antioxidant, antihyperglycemic and anti-angiogenic activities displayed by P. pellucida extracts in many in vitro, in vivo and clinical studies suggest its potential role in the management of inflammatory eye diseases. P. pellucida extract was non-toxic against normal cell lines but displayed mild toxicity in animal models. The growing public interest in P. pellucida has inspired the nutraceutical and pharmaceutical industries to process the plant into health products. Although the potential pharmacological mechanisms against eye diseases have been summarized, further studies of the interactions among constituent phytochemicals from P. pellucida within various signalling pathways shall support the use of the plant as an alternative therapeutic source.
Collapse
Affiliation(s)
- Keat Lam Ho
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Selangor, Malaysia
| | - Phaik Har Yong
- School of Bioscience, Faculty of Medicine, Bioscience and Nursing, MAHSA University, 42610 Selangor, Malaysia
| | - Chee Woon Wang
- Department of Biochemistry, Faculty of Medicine, Bioscience and Nursing, MAHSA University, 42610 Selangor, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chek Tung Ngo
- Optimax Sunway Eye Specialist Centre, Bandar Sunway, 46150 Selangor, Malaysia
| | - Festo Massawe
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Selangor, Malaysia
| | - Zhi Xiang Ng
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Selangor, Malaysia.
| |
Collapse
|
31
|
Jauregui-Lozano J, Hall H, Stanhope SC, Bakhle K, Marlin MM, Weake VM. The Clock:Cycle complex is a major transcriptional regulator of Drosophila photoreceptors that protects the eye from retinal degeneration and oxidative stress. PLoS Genet 2022; 18:e1010021. [PMID: 35100266 PMCID: PMC8830735 DOI: 10.1371/journal.pgen.1010021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/10/2022] [Accepted: 01/08/2022] [Indexed: 12/28/2022] Open
Abstract
The aging eye experiences physiological changes that include decreased visual function and increased risk of retinal degeneration. Although there are transcriptomic signatures in the aging retina that correlate with these physiological changes, the gene regulatory mechanisms that contribute to cellular homeostasis during aging remain to be determined. Here, we integrated ATAC-seq and RNA-seq data to identify 57 transcription factors that showed differential activity in aging Drosophila photoreceptors. These 57 age-regulated transcription factors include two circadian regulators, Clock and Cycle, that showed sustained increased activity during aging. When we disrupted the Clock:Cycle complex by expressing a dominant negative version of Clock (ClkDN) in adult photoreceptors, we observed changes in expression of 15-20% of genes including key components of the phototransduction machinery and many eye-specific transcription factors. Using ATAC-seq, we showed that expression of ClkDN in photoreceptors leads to changes in activity of 37 transcription factors and causes a progressive decrease in global levels of chromatin accessibility in photoreceptors. Supporting a key role for Clock-dependent transcription in the eye, expression of ClkDN in photoreceptors also induced light-dependent retinal degeneration and increased oxidative stress, independent of light exposure. Together, our data suggests that the circadian regulators Clock and Cycle act as neuroprotective factors in the aging eye by directing gene regulatory networks that maintain expression of the phototransduction machinery and counteract oxidative stress.
Collapse
Affiliation(s)
- Juan Jauregui-Lozano
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Hana Hall
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, United States of America
| | - Sarah C. Stanhope
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Kimaya Bakhle
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Makayla M. Marlin
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Vikki M. Weake
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
32
|
González Fleitas MF, Dorfman D, Rosenstein RE. A novel viewpoint in glaucoma therapeutics: enriched environment. Neural Regen Res 2021; 17:1431-1439. [PMID: 34916414 PMCID: PMC8771091 DOI: 10.4103/1673-5374.330594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glaucoma is one of the world's most frequent visual impairment causes and leads to selective damage to retinal ganglion cells and their axons. Despite glaucoma's most accepted risk factor is increased intraocular pressure (IOP), the mechanisms behind the disease have not been fully elucidated. To date, IOP lowering remains the gold standard; however, glaucoma patients may still lose vision regardless of effective IOP management. Therefore, the exclusive IOP control apparently is not enough to stop the disease progression, and developing new resources to protect the retina and optic nerve against glaucoma is a goal of vast clinical importance. Besides pharmacological treatments, environmental conditions have been shown to prevent neurodegeneration in the central nervous system. In this review, we discuss current concepts on key pathogenic mechanisms involved in glaucoma, the effect of enriched environment on these mechanisms in different experimental models, as well as recent evidence supporting the preventive and therapeutic effect of enriched environment exposure against experimental glaucomatous damage. Finally, we postulate that stimulating vision may become a non-invasive and rehabilitative therapy that could be eventually translated to the human disease, preventing glaucoma-induced terrible sequelae resulting in permanent visual disability.
Collapse
Affiliation(s)
- María F González Fleitas
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Damián Dorfman
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| | - Ruth E Rosenstein
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires, Argentina
| |
Collapse
|
33
|
Rossino MG, Amato R, Amadio M, Rosini M, Basagni F, Cammalleri M, Dal Monte M, Casini G. A Nature-Inspired Nrf2 Activator Protects Retinal Explants from Oxidative Stress and Neurodegeneration. Antioxidants (Basel) 2021; 10:1296. [PMID: 34439544 PMCID: PMC8389314 DOI: 10.3390/antiox10081296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 01/02/2023] Open
Abstract
Oxidative stress (OS) plays a key role in retinal dysfunctions and acts as a major trigger of inflammatory and neurodegenerative processes in several retinal diseases. To prevent OS-induced retinal damage, approaches based on the use of natural compounds are actively investigated. Recently, structural features from curcumin and diallyl sulfide have been combined in a nature-inspired hybrid (NIH1), which has been described to activate transcription nuclear factor erythroid-2-related factor-2 (Nrf2), the master regulator of the antioxidant response, in different cell lines. We tested the antioxidant properties of NIH1 in mouse retinal explants. NIH1 increased Nrf2 nuclear translocation, Nrf2 expression, and both antioxidant enzyme expression and protein levels after 24 h or six days of incubation. Possible toxic effects of NIH1 were excluded since it did not alter the expression of apoptotic or gliotic markers. In OS-treated retinal explants, NIH1 strengthened the antioxidant response inducing a massive and persistent expression of antioxidant enzymes up to six days of incubation. These effects resulted in prevention of the accumulation of reactive oxygen species, of apoptotic cell death, and of gliotic reactivity. Together, these data indicate that a strategy based on NIH1 to counteract OS could be effective for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Maria Grazia Rossino
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
| | - Rosario Amato
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
| | - Marialaura Amadio
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Filippo Basagni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Maurizio Cammalleri
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| | - Giovanni Casini
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (M.G.R.); (R.A.); (M.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
34
|
Rasoulinejad SA, Akbari A, Nasiri K. Interaction of miR-146a-5p with oxidative stress and inflammation in complications of type 2 diabetes mellitus in male rats: Anti-oxidant and anti-inflammatory protection strategies in type 2 diabetic retinopathy. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1078-1086. [PMID: 34804425 PMCID: PMC8591764 DOI: 10.22038/ijbms.2021.56958.12706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/12/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVES This study aimed to evaluate the role of miR-146a-5p in the pathogenesis of diabetic retinopathy and its interaction with oxidative stress and inflammation in the ocular tissue of rats with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Twenty adult male Sprague Dawley rats (220 ±20 g) were randomly assigned to control and diabetic groups. A high-fat diet was used for three months to induce T2DM which was confirmed by the HOMA-IR index. After that, the levels of glucose and insulin in serum, HOMA-IR as an indicator of insulin resistance, the ocular level of oxidative markers, TNF-α, IL-1β, MIPs, and MCP-1 along with ocular gene expression of NF-κB, Nrf2, and miR-146a-5p were evaluated. RESULTS The level of lipid peroxidation along with metabolic and inflammatory factors significantly increased and the antioxidant enzyme activity significantly decreased in diabetic rats (P<0.05). The ocular expression of NF-κB and TNF-α increased and Nrf2, HO-1, and miR-146a-5p expression decreased in diabetic rats (P<0.05). In addition, a negative correlation between miR-146a-5p expression with NF-κB and HOMA-IR and a positive correlation between miR-146a-5p with Nrf2 were observed. CONCLUSION It can be concluded that miR-146a-5p may regulate Nrf2 and NF-κB expression and inflammation and oxidative stress in the ocular tissue of diabetic rats.
Collapse
Affiliation(s)
- Seyed Ahmad Rasoulinejad
- Department of Ophthalmology, Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Khadijeh Nasiri
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Iran,Corresponding author: Khadijeh Nasiri. Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Iran. Babolsar, Iran.
| |
Collapse
|
35
|
Escandon P, Vasini B, Whelchel AE, Nicholas SE, Matlock HG, Ma JX, Karamichos D. The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp Eye Res 2021; 208:108617. [PMID: 34010603 PMCID: PMC8594540 DOI: 10.1016/j.exer.2021.108617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARβ/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARβ/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARβ/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.
Collapse
Affiliation(s)
- Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Amy E Whelchel
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Harold Hamm Oklahoma Diabetes Center, 1000 N Lincoln Blvd, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
36
|
Jurkute N, Shanmugarajah PD, Hadjivassiliou M, Higgs J, Vojcic M, Horrocks I, Nadjar Y, Touitou V, Lenaers G, Poh R, Acheson J, Robson AG, Raymond FL, Reilly MM, Yu-Wai-Man P, Moore AT, Webster AR, Arno G. Expanding the FDXR-Associated Disease Phenotype: Retinal Dystrophy Is a Recurrent Ocular Feature. Invest Ophthalmol Vis Sci 2021; 62:2. [PMID: 33938912 PMCID: PMC8107637 DOI: 10.1167/iovs.62.6.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/07/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose The purpose of this study was to report retinal dystrophy as a novel clinical feature and expand the ocular phenotype in patients harboring biallelic candidate FDXR variants. Methods Patients carrying biallelic candidate FDXR variants were identified by whole genome sequencing (WGS) as part of the National Institute for Health Research BioResource rare-disease and the UK's 100,000 Genomes Project (100KGP) with an additional case identified by exome sequencing. Retrospective clinical data were collected from the medical records. Haplotype reconstruction was performed in families harboring the same missense variant. Results Ten individuals from 8 unrelated families with biallelic candidate variants in FDXR were identified. In addition to bilateral optic atrophy and variable extra-ocular findings, 7 of 10 individuals manifested retinal dystrophy comprising dysfunction and degeneration of both rod and cone photoreceptors. Five of 10 subjects had sensorineural hearing loss. The previously unreported missense variant (c.1115C > A, p.(Pro372His)) was found in 5 of 8 (62.5%) study families. Haplotype reconstruction using WGS data demonstrated a likely ancestral haplotype. Conclusions FDXR-associated disease is a phenotypically heterogeneous disorder with retinal dystrophy being a major clinical feature observed in this cohort. In addition, we hypothesize that a number of factors are likely to drive the pathogenesis of optic atrophy, retinal degeneration, and perhaps the associated systemic manifestations.
Collapse
Affiliation(s)
- Neringa Jurkute
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Priya D. Shanmugarajah
- Academic Department of Neurosciences, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Marios Hadjivassiliou
- Academic Department of Neurosciences, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Jenny Higgs
- Liverpool Centre for Genomic Medicine, Liverpool Women's Hospital, Liverpool, United Kingdom
| | - Miodrag Vojcic
- Departments of Neurology and Ophthalmology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Iain Horrocks
- Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, Scotland
| | - Yann Nadjar
- Department of Neurology, Reference Center for Lysosomal Diseases, Neuro-Genetic and Metabolism Unit, Paris, France
| | - Valerie Touitou
- Sorbonne University, Paris, France
- Groupe Hospitalier La Pitié Salpêtrière-Charles Foix, DHU Vision Et Handicaps, Paris, France
| | - Guy Lenaers
- Université Angers, MitoLab team, UMR CNRS 6015 - INSERM U1083, Angers, France
| | - Roy Poh
- Department of Neurogenetics, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, London, United Kingdom
| | - James Acheson
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS trust, London, United Kingdom
| | - Anthony G. Robson
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - F. Lucy Raymond
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Mary M. Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Patrick Yu-Wai-Man
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
- John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Anthony T. Moore
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
| | - Andrew R. Webster
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Gavin Arno
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Great Ormond Street Hospital for Children, London, United Kingdom
| | - for the Genomics England Research Consortium
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Academic Department of Neurosciences, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Liverpool Centre for Genomic Medicine, Liverpool Women's Hospital, Liverpool, United Kingdom
- Departments of Neurology and Ophthalmology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
- Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, Scotland
- Department of Neurology, Reference Center for Lysosomal Diseases, Neuro-Genetic and Metabolism Unit, Paris, France
- Sorbonne University, Paris, France
- Groupe Hospitalier La Pitié Salpêtrière-Charles Foix, DHU Vision Et Handicaps, Paris, France
- Université Angers, MitoLab team, UMR CNRS 6015 - INSERM U1083, Angers, France
- Department of Neurogenetics, The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, London, United Kingdom
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS trust, London, United Kingdom
- NIHR BioResource - Rare Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
- John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
- Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
37
|
Alsabaani N. Inhibition of Protein Kinase R by C16 Protects the Retinal Ganglion Cells from Hypoxia-induced Oxidative Stress, Inflammation, and Apoptosis. Curr Eye Res 2021; 46:719-730. [PMID: 33026257 DOI: 10.1080/02713683.2020.1826980] [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: 08/20/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022]
Abstract
AIM/PURPOSE Individually, hypoxia and protein kinase R (PKR) induce retinal ganglion cells (RGCs) damage by aggravating reactive oxygen species (ROS), oxidative stress, inflammation, and apoptosis. However, it is still not established in hypoxia mediates such damaging effect by modulating PKR. This study investigated the expression and activation of PKR in hypoxic RGCs and tested if suppression of PKR by C16 is protective. MATERIALS AND METHODS Isolated RGCs were under normoxic or hypoxic conditions for 12 h. In some cases, hypoxic cells were pre-treated with C16, a PKR inhibitor, or n-acetyl cysteine (NAC) a glutathione (GSH) precursor for 1 h and then exposed to hypoxia for the next 12 h. RESULTS Hypoxia increased cell death, lactate dehydrogenase (LDH) levels, and levels of single-stranded DNA (ssDNA). It also increased levels of ROS, the activity of the nuclear factor-kappa beta (NF-κB), JNK, and p38 MAPK, expression of Bax, p53, and cleaved caspase-3, levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and cytoplasmic levels of cytochrome-c. It concomitantly suppressed levels of GSH and Bcl-2. All these events were associated with increased phosphorylation (activation) of PKR and its target eukaryotic initiation factor 2 (eIF2). Pre-incubating the cells with NAC completely prevented all these effects in hypoxic cells. Similar protective effects without affecting levels of ROS and GSH levels were also seen in hypoxic cells pre-treated with C16. CONCLUSION Hypoxia induces oxidative stress, inflammation, and apoptosis in the RGCs mainly by ROS induced activation of PKR, whereas scavenging ROS by NAC or suppressing PKR by C16 is a novel protective mechanism.
Collapse
Affiliation(s)
- Nasser Alsabaani
- Ophthalmology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
38
|
Anwar T, Qureshi H, Mahnashi MH, Kabir F, Parveen N, Ahmed D, Afzal U, Batool S, Awais M, Ahmed Alyami S, Ahmed Alhaider H. Bioherbicidal ability and weed management of allelopathic methyl esters from Lantana camara. Saudi J Biol Sci 2021; 28:4365-4374. [PMID: 34354421 PMCID: PMC8325027 DOI: 10.1016/j.sjbs.2021.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 10/25/2022] Open
Abstract
Allelochemicals are secondary metabolites which are not edible and can be used as growth regulators and bio-herbicides. The goal of current study was to assess allelopathic ability of Lantana camara (Sage-plant) flowers against weeds viz. Avena fatua (Wild oat), Euphorbia helioscopia (Sun-spurge), Chenopodium album (Goosefoot), Phalaris minor (Canary-grass), and Rumex dentatus (Knotweed). Bioassay analysis of three methanolic fractions of the Combiflash from L. camara was performed at 50%, 75% and 100% concentration using germination percentage parameters, inhibition of plumule and radicle size. The fraction II of Combiflash strongly suppressed all weeds with negligible effect on T. aestivum. Gas chromatography-mass spectroscopy was conducted for the fraction, and isolated compounds were used to perform bioassays. From fraction II GC-MS detected four methyl esters of allelopathic fatty acid viz. Methyl oleate, methyl palmitate, methyl stearate and methyl linoleate. The evaluation of physiological effects of the bioassay revealed substantial suppression of chlorophyll, antioxidant enzymes (superoxide, dismutase peroxidase) and protein material in all weeds by methyl palmitate. Bioassay activity and study of physiological parameters revealed that the effective bio-herbicidal compound in Lantana camara flowers is methyl palmitate. This is the first time that methyl palmitate (a fatty acid methyl ester) has been related to herbicidal activity in L. camara flowers. It is proposed that field studies based on hormesis research and the mechanism of action of this compound be carried out.
Collapse
Affiliation(s)
- Tauseef Anwar
- Department of Botany, The Islamia University of Bahawalpur, Baghdad-ul-Jadeed Campus, Bahawalpur-63100, Pakistan
| | - Huma Qureshi
- Institute of Biological Sciences, Gomal University, Dera Ismail Khan-29050, Pakistan
| | - Mater H Mahnashi
- Department of Pharmaceutical Chemistry, Pharmacy School, Najran University, Saudi Arabia
| | - Faryal Kabir
- University Institute of Biochemistry and Biotechnology PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Nusrat Parveen
- Department of Botany, Government College University, Faisalabad-38000, Punjab, Pakistan
| | - Dawood Ahmed
- Department of Medical Lab Technology, University of Haripur, Haripur, Pakistan
| | - Umara Afzal
- Department of Chemistry, Rawalpindi Women University, Satellite Town, Rawalpindi-46300, Pakistan
| | - Salma Batool
- Department of Biochemistry, University of Central Punjab, Lahore-54590, Pakistan
| | - Muhammad Awais
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Sialkot, Punjab, Pakistan
| | | | | |
Collapse
|
39
|
Duarte JN. Neuroinflammatory Mechanisms of Mitochondrial Dysfunction and Neurodegeneration in Glaucoma. J Ophthalmol 2021; 2021:4581909. [PMID: 33953963 PMCID: PMC8064803 DOI: 10.1155/2021/4581909] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/29/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The exact mechanism of retinal ganglion cell loss in the pathogenesis of glaucoma is yet to be understood. Mitochondrial damage-associated molecular patterns (DAMPs) resulting from mitochondrial dysfunction have been linked to Leber's hereditary optic neuropathy and autosomal dominant optic atrophy, as well as to brain neurodegenerative diseases. Recent evidence shows that, in conditions where mitochondria are damaged, a sustained inflammatory response and downstream pathological inflammation may ensue. Mitochondrial damage has been linked to the accumulation of age-related mitochondrial DNA mutations and mitochondrial dysfunction, possibly through aberrant reactive oxygen species production and defective mitophagy. The present review focuses on how mitochondrial dysfunction may overwhelm the ability of neurons and glial cells to adequately maintain homeostasis and how mitochondria-derived DAMPs trigger the immune system and induce neurodegeneration.
Collapse
Affiliation(s)
- Joao N. Duarte
- Neuroinflammation Unit, Biotech Research & Innovation Center, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Miller WP, Sunilkumar S, Dennis MD. The stress response protein REDD1 as a causal factor for oxidative stress in diabetic retinopathy. Free Radic Biol Med 2021; 165:127-136. [PMID: 33524531 PMCID: PMC7956244 DOI: 10.1016/j.freeradbiomed.2021.01.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Diabetic Retinopathy (DR) is a major cause of visual dysfunction, yet much remains unknown regarding the specific molecular events that contribute to diabetes-induced retinal pathophysiology. Herein, we review the impact of oxidative stress on DR, and explore evidence that supports a key role for the stress response protein regulated in development and DNA damage (REDD1) in the development of diabetes-induced oxidative stress and functional defects in vision. It is well established that REDD1 mediates the cellular response to a number of diverse stressors through repression of the central metabolic regulator known as mechanistic target of rapamycin complex 1 (mTORC1). A growing body of evidence also supports that REDD1 acts independent of mTORC1 to promote oxidative stress by both enhancing the production of reactive oxygen species and suppressing the antioxidant response. Collectively, there is strong preclinical data to support a key role for REDD1 in the development and progression of retinal complications caused by diabetes. Furthermore, early proof-of-concept clinical trials have found a degree of success in combating ischemic retinal disease through intravitreal delivery of an siRNA targeting the REDD1 mRNA. Overall, REDD1-associated signaling represents an intriguing target for novel clinical therapies that go beyond addressing the symptoms of diabetes by targeting the underlying molecular mechanisms that contribute to DR.
Collapse
Affiliation(s)
- William P Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA; Department of Ophthalmology, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
41
|
Chen J, Shao Y, Sasore T, Moiseyev G, Zhou K, Ma X, Du Y, Ma JX. Interphotoreceptor Retinol-Binding Protein Ameliorates Diabetes-Induced Retinal Dysfunction and Neurodegeneration Through Rhodopsin. Diabetes 2021; 70:788-799. [PMID: 33334874 PMCID: PMC7897347 DOI: 10.2337/db20-0609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
Patients with diabetes often experience visual defects before any retinal pathologies are detected. The molecular mechanism for the visual defects in early diabetes has not been elucidated. Our previous study reported that in early diabetic retinopathy (DR), rhodopsin levels were reduced due to impaired 11-cis-retinal regeneration. Interphotoreceptor retinol-binding protein (IRBP) is a visual cycle protein and important for 11-cis-retinal generation. IRBP levels are decreased in the vitreous and retina of DR patients and animal models. To determine the role of IRBP downregulation in the visual defects in early DR, we induced diabetes in transgenic mice overexpressing IRBP in the retina. IRBP overexpression prevented diabetes-induced decline of retinal function. Furthermore, IRBP overexpression also prevented decreases of rhodopsin levels and 11-cis-retinal generation in diabetic mice. Diabetic IRBP transgenic mice also showed ameliorated retinal oxidative stress, inflammation, apoptosis, and retinal degeneration compared with diabetic wild-type mice. These findings suggest that diabetes-induced IRBP downregulation impairs the regeneration of 11-cis-retinal and rhodopsin, leading to retinal dysfunction in early DR. Furthermore, increased 11-cis-retinal-free opsin constitutively activates the phototransduction pathway, leading to increased oxidative stress and retinal neurodegeneration. Therefore, restored IRBP expression in the diabetic retina may confer a protective effect against retinal degeneration in DR.
Collapse
Affiliation(s)
- Jianglei Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Yan Shao
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Temmy Sasore
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gennadiy Moiseyev
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Kelu Zhou
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Xiang Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Yanhong Du
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
42
|
Lu ZG, May A, Dinh B, Lin V, Su F, Tran C, Adivikolanu H, Ehlen R, Che B, Wang ZH, Shaw DH, Borooah S, Shaw PX. The interplay of oxidative stress and ARMS2-HTRA1 genetic risk in neovascular AMD. ACTA ACUST UNITED AC 2021; 5. [PMID: 34017939 PMCID: PMC8133762 DOI: 10.20517/2574-1209.2020.48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in adults over 60 years old globally. There are two forms of advanced AMD: “dry” and “wet”. Dry AMD is characterized by geographic atrophy of the retinal pigment epithelium and overlying photoreceptors in the macular region; whereas wet AMD is characterized by vascular penetrance from the choroid into the retina, known as choroidal neovascularization (CNV). Both phenotypes eventually lead to loss of central vision. The pathogenesis of AMD involves the interplay of genetic polymorphisms and environmental risk factors, many of which elevate retinal oxidative stress. Excess reactive oxygen species react with cellular macromolecules, forming oxidation-modified byproducts that elicit chronic inflammation and promote CNV. Additionally, genome-wide association studies have identified several genetic variants in the age-related maculopathy susceptibility 2/high-temperature requirement A serine peptidase 1 (ARMS2-HTRA1) locus associated with the progression of late-stage AMD, especially the wet subtype. In this review, we will focus on the interplay of oxidative stress and HTRA1 in drusen deposition, chronic inflammation, and chronic angiogenesis. We aim to present a multifactorial model of wet AMD progression, supporting HTRA1 as a novel therapeutic target upstream of vascular endothelial growth factor (VEGF), the conventional target in AMD therapeutics. By inhibiting HTRA1’s proteolytic activity, we can reduce pro-angiogenic signaling and prevent proteolytic breakdown of the blood-retina barrier. The anti-HTRA1 approach offers a promising alternative treatment option to wet AMD, complementary to anti-VEGF therapy.
Collapse
Affiliation(s)
- Zhi-Gang Lu
- Department of Neurology, First People's Hospital of Jingmen, Jingchu University of Technology, Jingmen 448000, Hubei, China.,Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam May
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian Dinh
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Lin
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fei Su
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Tran
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Harini Adivikolanu
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rachael Ehlen
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Briana Che
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhi-Hao Wang
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel H Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Westview High School, San Diego, CA 92131, USA
| | - Shyamanga Borooah
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter X Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
43
|
Sabry DA, El-Badry D. Altered retina and cornea of Clarias gariepinus (Siluriformes: Clariidae) under the effect of bright and dim lights. ZOOLOGIA 2020. [DOI: 10.3897/zoologia.37.e51603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The purpose of this study was to investigate the influence of constant bright light on the cornea and retina of Clarias gariepinus (Burchell, 1822) and to examine whether it can change after constant exposure to dim light. Twenty-one adult individuals of C. gariepinus were divided into three groups (n = 7). The first group was maintained under normal light (NL). The second group was exposed to the intense bright light (BL) (3020 Lux) of white light lamps for seven days. The third group was exposed to dim light for seven days (DL) following the previous exposure to intense bright light for seven days. The eyes of each fish group were removed and fixed. The following aspects of the eye were investigated: histopathological, immunohistochemical (GFAP and BAX) staining and biochemical study for lactic dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA) and glucose-6-phosphate-dehydrogenase (G6PDH). Also, isoenzyme electrophoresis of LDH, G6PDH and SOD were performed. The present study found that, seven-days BL exposure caused damage to both cornea and retina. However, after exposure to dim-light after bright light there was partial improvement in corneal and retinal structure and an increase in the assayed SOD and G6PDH levels, along with a reduction in MDA content and activity of LDH. These findings demonstrate a plasticity that may help C. gariepinus survive disturbances in the aquatic environment.
Collapse
|
44
|
Hvozda Arana AG, Lasagni Vitar RM, Reides CG, Lerner SF, Ferreira SM. Glaucoma causes redox imbalance in the primary visual cortex by modulating NADPH oxidase-4, iNOS, and Nrf2 pathway in a rat experimental model. Exp Eye Res 2020; 200:108225. [DOI: 10.1016/j.exer.2020.108225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
|
45
|
Antioxidant and Biological Properties of Mesenchymal Cells Used for Therapy in Retinitis Pigmentosa. Antioxidants (Basel) 2020; 9:antiox9100983. [PMID: 33066211 PMCID: PMC7602011 DOI: 10.3390/antiox9100983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Both tissue repair and regeneration are a priority in regenerative medicine. Retinitis pigmentosa (RP), a complex retinal disease characterized by the progressive loss of impaired photoreceptors, is currently lacking effective therapies: this represents one of the greatest challenges in the field of ophthalmological research. Although this inherited retinal dystrophy is still an incurable genetic disease, the oxidative damage is an important pathogenetic element that may represent a viable target of therapy. In this review, we summarize the current neuroscientific evidence regarding the effectiveness of cell therapies in RP, especially those based on mesenchymal cells, and we focus on their therapeutic action: limitation of both oxidative stress and apoptotic processes triggered by the disease and promotion of cell survival. Cell therapy could therefore represent a feasible therapeutic option in RP.
Collapse
|
46
|
Han EH, Lim MK, Lee S, Lee SH, Yun SM, Yu HJ, Ryu SH, Lim YH. Efficacy of Ethanolic Extract of Opuntia ficus-indica var. saboten Stems for Improving Cognitive Function in Elderly Subjects 55-85 Years of Age: A Randomized, Double-Blind, Placebo-Controlled Study. J Med Food 2020; 23:1146-1154. [PMID: 33006504 DOI: 10.1089/jmf.2019.4678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Many natural compounds have been reported to improve cognitive function in cell- and animal-based studies. In this clinical trial, we evaluated the efficacy of ethanolic extract of Opuntia ficus-indica var. saboten stem for improving cognitive function using a randomized, double-blind, placebo-controlled trial (n = 81) in aged people. After 12 weeks of administration of OFE (a mixture of ethanolic extract of O. ficus-indica var. Saboten stem and dextrin) or placebo, the effect on cognitive function was assessed. Overall, OFE did not show a significant difference from the placebo in terms of efficacy. However, the cognitive function significantly improved in the OFE group compared with the placebo group in the subgroup ≤70 years of age, which means that the effect of OFE administration exhibits an age-dependent effect. In addition, the safety of OFE was confirmed by analyzing blood test results, vital signs, and electrocardiograms. In conclusion, OFE administration in participants ≤70 years of age shows a positive effect on overall cognitive function. The trial was registered on CRIS (the Clinical Research Information Service), administered by the Korea Centers for Disease Control & Prevention (Registration Number: KCT0003766; URL: https://cris.nih.go.kr/cris/en/search/search_result_st01.jsp?seq=12957).
Collapse
Affiliation(s)
- Eun Hye Han
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul, Republic of Korea.,R&D Center, Koreaeundan Healthcare Co., Ansan-si, Gyeonggi-do, Republic of Korea
| | - Mi Kyung Lim
- R&D Center, Koreaeundan Co., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soyeon Lee
- R&D Center, Koreaeundan Healthcare Co., Ansan-si, Gyeonggi-do, Republic of Korea.,Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sang Ho Lee
- R&D Center, Koreaeundan Healthcare Co., Ansan-si, Gyeonggi-do, Republic of Korea
| | - Su Min Yun
- R&D Center, Koreaeundan Healthcare Co., Ansan-si, Gyeonggi-do, Republic of Korea
| | - Heui-Jong Yu
- R&D Center, SK Bioland Co, Ltd., Ansan-si, Gyeonggi-do, Republic of Korea
| | - Seung-Ho Ryu
- Department of Psychiatry, School of Medicine, Konkuk University, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul, Republic of Korea.,Department of Public Health Science (Brain Korea 21 PLUS program), Graduate School, Korea University, Seoul, Republic of Korea.,Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
47
|
Oxidative Stress and Vascular Dysfunction in the Retina: Therapeutic Strategies. Antioxidants (Basel) 2020; 9:antiox9080761. [PMID: 32824523 PMCID: PMC7465265 DOI: 10.3390/antiox9080761] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Many retinal diseases, such as diabetic retinopathy, glaucoma, and age-related macular (AMD) degeneration, are associated with elevated reactive oxygen species (ROS) levels. ROS are important intracellular signaling molecules that regulate numerous physiological actions, including vascular reactivity and neuron function. However, excessive ROS formation has been linked to vascular endothelial dysfunction, neuron degeneration, and inflammation in the retina. ROS can directly modify cellular molecules and impair their function. Moreover, ROS can stimulate the production of inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) causing inflammation and cell death. However, there are various compounds with direct or indirect antioxidant activity that have been used to reduce ROS accumulation in animal models and humans. In this review, we report on the physiological and pathophysiological role of ROS in the retina with a special focus on the vascular system. Moreover, we present therapeutic approaches for individual retinal diseases targeting retinal signaling pathways involving ROS.
Collapse
|
48
|
Ciavarella C, Buzzi M, Bergantin E, Di Marco S, Giannaccare G, Campos E, Bisti S, Versura P. Effects of Cord Blood Serum (CBS) on viability of retinal Müller glial cells under in vitro injury. PLoS One 2020; 15:e0234145. [PMID: 32497126 PMCID: PMC7272066 DOI: 10.1371/journal.pone.0234145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/19/2020] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress and inflammation determine retinal ganglion cell degeneration, leading to retinal impairment and vision loss. Müller glial cells regulate retinal repair under injury, through gliosis. Meanwhile, reactive gliosis can turn in pathological effects, contributing to neurodegeneration. In the present study, we tested whether Cord Blood Serum (CBS), rich of growth factors, might improve the viability of Müller cells under in vitro damage. BDNF, NGF, TGF-α, GDNF and EGF levels were measured in CBS samples by Human Magnetic Luminex Assay. CBS effects were evaluated on rat (rMC-1) and human (MIO-M1) Müller cells, under H2O2 and IL-1β damage. Cells grown with FBS or CBS both at 5% were exposed to stress and analyzed in terms of cell viability, GFAP, IL-6 and TNF-α expression. CBS was also administrated after treatment with K252a, inhibitor of the neurotrophin receptor Trk. Cell viability of rMC-1 and MIO-M1 resulted significantly improved when pretreated with CBS and exposed to H2O2 and IL-1β, in comparison to the standard culture with FBS. Accordingly, the gliosis marker GFAP resulted down-regulated following CBS priming. In parallel, we observed a lower expression of the inflammatory mediators in rMC-1 (TNF-α) and MIO-M1 (IL-6, TNF- α), especially in presence of inflammatory damage. Trk inhibition through K252a administration impaired the effects of CBS under stress conditions on MIO-M1 and rMC-1 viability, not significantly different from FBS condition. CBS is enriched with neurotrophins and its administration to rMC-1 and MIO-M1 attenuates the cytotoxic effects of H2O2 and IL-1β. Moreover, the decrease of the main markers of gliosis and inflammation suggests a promising use of CBS for neuroprotection aims. This study is a preliminary basis that prompts future investigations to deeply explore and confirm the CBS potential.
Collapse
Affiliation(s)
- Carmen Ciavarella
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Marina Buzzi
- Emilia Romagna Cord Blood Bank-Transfusion Service, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Elisa Bergantin
- Emilia Romagna Cord Blood Bank-Transfusion Service, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | | | - Giuseppe Giannaccare
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Emilio Campos
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
| | - Silvia Bisti
- Vision Lab, DISCAB, University of L’Aquila, L’Aquila, Italy
- Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Piera Versura
- Ophthalmology Unit, DIMES, Alma Mater Studiorum University of Bologna, S.Orsola-Malpighi Teaching Hospital, Bologna, Italy
- * E-mail:
| |
Collapse
|
49
|
B. Domènech E, Marfany G. The Relevance of Oxidative Stress in the Pathogenesis and Therapy of Retinal Dystrophies. Antioxidants (Basel) 2020; 9:E347. [PMID: 32340220 PMCID: PMC7222416 DOI: 10.3390/antiox9040347] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Retinal cell survival requires an equilibrium between oxygen, reactive oxygen species, and antioxidant molecules that counteract oxidative stress damage. Oxidative stress alters cell homeostasis and elicits a protective cell response, which is most relevant in photoreceptors and retinal ganglion cells, neurons with a high metabolic rate that are continuously subject to light/oxidative stress insults. We analyze how the alteration of cellular endogenous pathways for protection against oxidative stress leads to retinal dysfunction in prevalent (age-related macular degeneration, glaucoma) as well as in rare genetic visual disorders (Retinitis pigmentosa, Leber hereditary optic neuropathy). We also highlight some of the key molecular actors and discuss potential therapies using antioxidants agents, modulators of gene expression and inducers of cytoprotective signaling pathways to treat damaging oxidative stress effects and ameliorate severe phenotypic symptoms in multifactorial and rare retinal dystrophies.
Collapse
Affiliation(s)
- Elena B. Domènech
- Departament de Genètica, Microbiologia i Estadística, Avda. Diagonal 643, Universitat de Barcelona, 08028 Barcelona, Spain;
- CIBERER, ISCIII, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Marfany
- Departament de Genètica, Microbiologia i Estadística, Avda. Diagonal 643, Universitat de Barcelona, 08028 Barcelona, Spain;
- CIBERER, ISCIII, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine (IBUB, IBUB-IRSJD), Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
50
|
Crocetin Prevents RPE Cells from Oxidative Stress through Protection of Cellular Metabolic Function and Activation of ERK1/2. Int J Mol Sci 2020; 21:ijms21082949. [PMID: 32331354 PMCID: PMC7215651 DOI: 10.3390/ijms21082949] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause for visual impairment in aging populations with limited established therapeutic interventions available. Oxidative stress plays an essential role in the pathogenesis of AMD, damaging the retinal pigment epithelium (RPE), which is essential for the function and maintenance of the light-sensing photoreceptors. This study aimed to evaluate the effects of crocetin, one of the main components of Saffron, on an in vitro RPE model of tert-butyl hydroperoxide (TBHP) induced oxidative stress using ARPE19 cells. The effects of crocetin were assessed using lactate de-hydrogenase (LDH) and ATP assays, as well as immunocytochemistry for cell morphology, junctional integrity, and nuclear morphology. The mechanism of crocetin action was determined via assessment of energy production pathways, including mitochondrial respiration and glycolysis in real-time as well as investigation of extracellular signal-regulated kinase 1/2 (ERK1/2) activation and distribution. Our results show that crocetin pre-treatment protects ARPE19 cells from TBHP-induced LDH release, intracellular ATP depletion, nuclear condensation, and disturbance of junctional integrity and cytoskeleton. The protective effect of crocetin is mediated via the preservation of energy production pathways and activation of ERK1/2 in the first minutes of TBHP exposure to potentiate survival pathways. The combined data suggest that a natural antioxidant, such as crocetin, represents a promising candidate to prevent oxidative stress in RPE cells and might halt or delay disease progression in AMD.
Collapse
|