1
|
Pan WW, Wubben TJ, Zacks DN. Promising therapeutic targets for neuroprotection in retinal disease. Curr Opin Ophthalmol 2025; 36:247-252. [PMID: 39927457 DOI: 10.1097/icu.0000000000001123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
PURPOSE OF REVIEW Neurodegeneration is a common endpoint of various blinding retinal diseases. Yet, despite exciting advances in disease treatment, there continues to exist a critical need for the development of neuroprotective strategies to prevent retinal cell death. Here, we summarize the recent advances in neuroprotective strategies. RECENT FINDINGS From laboratory deciphering of the mechanisms involved in disease, many novel neuroprotective strategies have emerged and are currently under investigation for the treatment of various retinal and ocular diseases such as inherited retinal degeneration, retinal detachment, diabetic retinopathy, age-related macular degeneration, macular telangiectasia type 2, and glaucoma. These strategies include gene therapies, Fas inhibition, and targeting inflammatory, metabolic and reduction-oxidation abnormalities. Interestingly, investigation of several treatments across different diseases suggests shared neuroprotection mechanisms that can be targeted regardless of the particular disease. SUMMARY Retinal neuroprotection can improve treatment of different retinal diseases. Fortunately, the current landscape, with a plethora of novel neuroprotective therapies, portends a better future for patients.
Collapse
Affiliation(s)
- Warren W Pan
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
2
|
He XN, Jiang WD, Wu P, Liu Y, Ren HM, Jin XW, Kuang SY, Tang L, Li SW, Feng L, Zhou XQ. Aflatoxin B1 inhibited the development of primary myoblasts of grass carp (Ctenopharyngodon idella) by degrading extracellular matrix. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116332. [PMID: 38626608 DOI: 10.1016/j.ecoenv.2024.116332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/18/2024]
Abstract
According to the International Agency for Research on Cancer (IARC), aflatoxin B1 (AFB1) has been recognized as a major contaminant in food and animal feed and which is a common mycotoxin with high toxicity. Previous research has found that AFB1 inhibited zebrafish muscle development. However, the potential mechanism of AFB1 on fish muscle development is unknown, so it is necessary to conduct further investigation. In the present research, the primary myoblast of grass carp was used as a model, we treated myoblasts with AFB1 for 24 h. Our results found that 5 μM AFB1 significantly inhibited cell proliferation and migration (P < 0.05), and 10 μM AFB1 promoted lactate dehydrogenase (LDH) release (P < 0.05). Reactive oxygen species (ROS), protein carbonyl (PC) and malondialdehyde (MDA) levels were increased in 15, 5 and 10 μM AFB1 (P < 0.05), respectively. Catalase (CAT), glutathione peroxidase (GPx) and total superoxide dismutase (T-SOD) activities were decreased in 10, 10 and 15 μM AFB1 (P < 0.05), respectively. Furthermore, 15 μM AFB1 induced oxidative damage by Nrf2 pathway, also induced apoptosis in primary myoblast of grass carp. Meanwhile, 15 μM AFB1 decreased MyoD gene and protein expression (P < 0.05). Importantly, 15 μM AFB1 decreased the protein expression of collagen Ⅰ and fibronectin (P < 0.05), and increased the protein levels of urokinase plasminogen activator (uPA), matrix metalloproteinase 9 (MMP-9), matrix metalloproteinase 2 (MMP-2), and p38 mitogen-activated protein kinase (p38MAPK) (P < 0.05). As a result, our findings suggested that AFB1 damaged the cell morphology, induced oxidative damage and apoptosis, degraded ECM components, in turn inhibiting myoblast development by activating the p38MAPK/urokinase-type plasminogen activator (uPA)/matrix metalloproteinase (MMPs)/extracellular matrix (ECM) signaling pathway.
Collapse
Affiliation(s)
- Xiang-Ning He
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China.
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Provence, Sichuan 611130, China.
| |
Collapse
|
3
|
Davidescu M, Mezzasoma L, Fettucciari K, Pascucci L, Pariano M, Di Michele A, Bereshchenko O, Cagini C, Cellini B, Corazzi L, Bellezza I, Macchioni L. Cardiolipin-mediated temporal response to hydroquinone toxicity in human retinal pigmented epithelial cell line. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119554. [PMID: 37524263 DOI: 10.1016/j.bbamcr.2023.119554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
Hydroquinone, a potent toxic agent of cigarette smoke, damages retinal pigmented epithelial cells by triggering oxidative stress and mitochondrial dysfunction, two events causally related to the development and progression of retinal diseases. The inner mitochondrial membrane is enriched in cardiolipin, a phospholipid susceptible of oxidative modifications which determine cell-fate decision. Using ARPE-19 cell line as a model of retinal pigmented epithelium, we analyzed the potential involvement of cardiolipin in hydroquinone toxicity. Hydroquinone exposure caused an early concentration-dependent increase in mitochondrial reactive oxygen species, decrease in mitochondrial membrane potential, and rise in the rate of oxygen consumption not accompanied by changes in ATP levels. Despite mitochondrial impairment, cell viability was preserved. Hydroquinone induced cardiolipin translocation to the outer mitochondrial membrane, and an increase in the colocalization of the autophagosome adapter protein LC3 with mitochondria, indicating the induction of protective mitophagy. A prolonged hydroquinone treatment induced pyroptotic cell death by cardiolipin-mediated caspase-1 and gasdermin-D activation. Cardiolipin-specific antioxidants counteracted hydroquinone effects pointing out that cardiolipin can act as a mitochondrial "eat-me signal" or as a pyroptotic cell death trigger. Our results indicate that cardiolipin may act as a timer for the mitophagy to pyroptosis switch and propose cardiolipin-targeting compounds as promising approaches for the treatment of oxidative stress-related retinal diseases.
Collapse
Affiliation(s)
- Magdalena Davidescu
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Letizia Mezzasoma
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Katia Fettucciari
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo 4, 06126 Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Alessandro Di Michele
- Department of Physic and Geology, University of Perugia, Via Pascoli, Perugia 06123, Italy
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences, Humanities and Education, University of Perugia, Piazza Ermini 1, Perugia 06123, Italy
| | - Carlo Cagini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Lanfranco Corazzi
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Ilaria Bellezza
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy
| | - Lara Macchioni
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia 06132, Italy.
| |
Collapse
|
4
|
Nishiyama T, Tsujinaka H, Ueda T, Ogata N. Alteration in Melanin Content in Retinal Pigment Epithelial Cells upon Hydroquinone Exposure. Int J Mol Sci 2023; 24:16801. [PMID: 38069124 PMCID: PMC10706107 DOI: 10.3390/ijms242316801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Abnormal pigmentation or depigmentation of the retinal pigment epithelium (RPE) is a precursor to neovascular age-related macular degeneration (nAMD). In this study, we evaluated the effects of hydroquinone (HQ), the most potent reductant in cigarette smoke, on the melanin production in RPE cells. Induced pluripotent stem cell (iPS)-derived RPE and adult retinal pigment epithelial (ARPE-19) cells were cultured with HQ. Real-time reverse transcription polymerase chain reaction revealed that the expression of melanin-related genes decreased due to the addition of HQ for 1 day. Enzyme-linked immunosorbent immunoassay showed that the concentration of melanin significantly decreased due to the addition of HQ for 24 h. A suspension of RPE cells with HQ for 24 h was prepared, and the absorbance was measured. The absorbance decreased particularly under blue light, suggesting that blue light may reach the choroid and cause choroidal inflammation. Additionally, melanin levels significantly decreased due to the addition of HQ for 1 week. After blue light irradiation on the RPE with HQ for 1 week, the vascular endothelial growth factor in the medium was significantly higher in the HQ group than in the control group. HQ-induced changes in melanin production may be responsible for the uneven pigmentation of the RPE, and these changes may cause nAMD.
Collapse
Affiliation(s)
| | | | - Tetsuo Ueda
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8521, Japan
| | | |
Collapse
|
5
|
Liu M, Honjo M, Yamagishi R, Aihara M. Effects of Brimonidine, Omidenepag Isopropyl, and Ripasudil Ophthalmic Solutions to Protect against H 2O 2-Induced Oxidative Stress in Human Trabecular Meshwork Cells. Curr Eye Res 2023; 48:1014-1025. [PMID: 37466387 DOI: 10.1080/02713683.2023.2235892] [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: 02/22/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE We investigated whether hydrogen peroxide (H2O2)-induced oxidative stress causes human trabecular meshwork (HTM) cell dysfunction observed in open angle glaucoma (OAG) in vitro, and the effects of topical glaucoma medications on oxidative stress in HTM cells. METHODS We used commercially available ophthalmic solutions of brimonidine, omidenepag isopropyl, and ripasudil in the study. HTM cells were exposed to H2O2 for 1 h, with or without glaucoma medications. We assessed cell viability and senescence via WST-1 and senescence-associated-β-galactosidase (SA-β-Gal) activity assays. After exposure to H2O2 and glaucoma medications, we evaluated changes in markers of fibrosis and stress by using real-time quantitative polymerase chain reaction (qPCR) to measure the mRNA levels of collagen type I alpha 1 chain (COL1A1), fibronectin, alpha-smooth muscle actin (α-SMA), matrix metalloproteinase-2 (MMP-2), endoplasmic reticulum stress markers of C/EBP homologous protein (CHOP), 78-kDa glucose-regulated protein (GRP78), and splicing X-box binding protein-1 (sXBP-1). RESULTS HTM cell viability decreased and SA-β-Gal activity increased significantly after exposure to H2O2. Treatment with three ophthalmic solutions attenuated these changes. Real-time qPCR revealed that H2O2 upregulated the mRNA levels of COL1A1, fibronectin, α-SMA, CHOP, GRP78, and sXBP-1, whereas it downregulated MMP-2 mRNA expression significantly. Brimonidine suppressed the upregulation of stress markers CHOP and GRP78. Additionally, omidenepag isopropyl and ripasudil decreased the upregulation of COL1A1 and sXBP-1. Furthermore, ripasudil significantly suppressed fibrotic markers fibronectin and α-SMA, compared with the other two medications. CONCLUSION In vitro, H2O2 treatment of HTM cells induced characteristic changes of OAG, such as fibrosis changes and the upregulation of stress markers. These glaucomatous changes were attenuated by additional treatments with brimonidine, omidenepag isopropyl, and ripasudil ophthalmic solutions.
Collapse
Affiliation(s)
- Mengxuan Liu
- Department of Ophthalmology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Reiko Yamagishi
- Department of Ophthalmology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Ham Y, Mehta H, Kang-Mieler J, Mieler WF, Chang A. Novel Drug Delivery Methods and Approaches for the Treatment of Retinal Diseases. Asia Pac J Ophthalmol (Phila) 2023; 12:402-413. [PMID: 37523432 DOI: 10.1097/apo.0000000000000623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/30/2023] [Indexed: 08/02/2023] Open
Abstract
This review discusses emerging approaches to ocular drug delivery for retinal diseases. Intravitreal injections have proven to be an effective, safe, and commonly used drug delivery method. However, the optimal management of chronic retinal diseases requires frequent intravitreal injections over extended periods of time. Although this can be achieved in a clinical trial environment, it is difficult to replicate in routine clinical practice. In addition, frequent treatment increases the risk of complications, incurs more costs, and increases the treatment burden for patients and caregivers. Given the aging global population and diabetes pandemic, there is an urgent need for drug delivery methods that support more durable retinal therapy while maintaining the efficacy and safety of currently available intravitreal therapies. Several innovative drug delivery methods are currently being investigated. These include sustained-release implants and depots using prodrugs, microparticles, and hydrogels, surgically implanted reservoirs, gene therapy via submacular injections or suprachoroidal injections, as well as topical and systemic therapies.
Collapse
Affiliation(s)
- Yeji Ham
- Sydney Retina Clinic, Sydney, Australia
| | - Hemal Mehta
- Sydney Retina Clinic, Sydney, Australia
- Save Sight Registries, The University of Sydney, Sydney, Australia
- Strathfield Retina Clinic, Sydney, Australia
| | - Jennifer Kang-Mieler
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ
| | | | - Andrew Chang
- Sydney Retina Clinic, Sydney Eye Hospital, The University of Sydney, Sydney, Australia
| |
Collapse
|
7
|
Freeman WR, Bandello F, Souied E, Guymer RH, Garg SJ, Chen FK, Rich R, Holz FG, Patel SS, Kim K, López FJ, Guymer R, Korobelnik JF, Souied E, Holz F, Ziemssen F, Bandello F, Campos E, Grignolo/Eandi C, Midena E, Peiretti E, Staurenghi G, Viola F, Bailey C, Esposti SD, Jackson T, Menon G, Pagliarini S, Quhill F, Antoszyk A, Brooks L, Callanan D, Csaky K, Edwards A, Eichenbaum D, Freeman W, Garg S, Ghuman AT, Gonzalez V, Gupta S, Hamilton R, Khurana R, Kunimoto D, Kuppermann B, Lauer A, Lee SY, Maturi R, Patel S, Reddy R, Rich R, Rivellese M, Rose S, Segal Z, Wong R. Randomized Phase 2b Study of Brimonidine Drug Delivery System Generation 2 for Geographic Atrophy in Age-related Macular Degeneration. Ophthalmol Retina 2023:S2468-6530(23)00097-0. [PMID: 36906177 DOI: 10.1016/j.oret.2023.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
PURPOSE To evaluate the safety and efficacy of repeat injections of Brimonidine Drug Delivery System (Brimo DDS) Generation 2 (Gen 2) containing 400 μg brimonidine in patients with geographic atrophy (GA) secondary to age-related macular degeneration (AMD). DESIGN Phase 2b, randomized, multicenter, double-masked, sham-controlled, 30-month study (BEACON). PARTICIPANTS Patients diagnosed with GA secondary to AMD and multifocal lesions with total area >1.25 mm2 and ≤18 mm2 in the study eye. METHODS Enrolled patients were randomized to treatment with intravitreal injections of 400-μg Brimo DDS (n=154) or sham procedure (n=156) in the study eye every 3 months from day 1 through month 21. MAIN OUTCOME MEASURES The primary efficacy endpoint was change from baseline in the GA lesion area in the study eye, assessed with fundus autofluorescence imaging, at month 24. Safety measures included treatment-emergent adverse events (AEs). RESULTS The study was terminated early, at the time of the planned interim analysis, because of a slow GA progression rate (∼1.6 mm2/year) in the enrolled population. Least-squares mean (standard error) GA area change from baseline at month 24 (primary endpoint) was 3.24 (0.13) mm2 with Brimo DDS (n=84) versus 3.48 (0.13) mm2 with sham (n=91); the reduction in GA area change from baseline in the Brimo DDS group compared with the sham group was 0.25 mm2 (7%) (P = 0.150). At month 30, the GA area change from baseline was 4.09 (0.15) mm2 with Brimo DDS (n=49) versus 4.52 (0.15) mm2 with sham (n=46), a reduction of 0.43 mm2 (10%) with Brimo DDS compared with sham (P=0.033). Exploratory analysis showed numerically smaller loss over time in retinal sensitivity assessed with scotopic microperimetry with Brimo DDS compared with sham (P=0.053 at month 24). Treatment-related AEs were usually related to the injection procedure. No implant accumulation was observed. CONCLUSIONS Multiple intravitreal administrations of Brimo DDS (Gen 2) were well tolerated. The primary efficacy endpoint at 24 months was not met, but there was a numerical trend for reduction in GA progression at 24 months compared with sham treatment. The study was terminated early because of the lower-than-expected GA progression rate in the sham/control group.
Collapse
Affiliation(s)
- William R Freeman
- Jacobs Retina Center, University of California San Diego, La Jolla, California, USA.
| | - Francesco Bandello
- University Vita-Salute Scientific Institute, Hospital San Raffaele, Milan, Italy
| | - Eric Souied
- Centre Hospitalier Creteil, Service Universitaire d'Ophthalmologie, Creteil, France
| | - Robyn H Guymer
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, and Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, Australia
| | - Sunir J Garg
- Mid Atlantic Retina, the Retina Service of Wills Eye Hospital, Philadelphia, Pennsylvania, USA
| | - Fred K Chen
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, and Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, Australia; Centre for Ophthalmology and Visual Science (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, Western Australia, Australia
| | - Ryan Rich
- Retina Consultants of Southern Colorado PC, Colorado Springs, Colorado, USA
| | - Frank G Holz
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | | | - Kimmie Kim
- Allergan, an AbbVie company, Irvine, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Güven YZ, Akbalık T. A novel therapy in paclitaxel-related cystoid macular edema: Brimonidine. J Fr Ophtalmol 2023; 46:e154-e156. [PMID: 36828725 DOI: 10.1016/j.jfo.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 02/24/2023]
Affiliation(s)
- Y Z Güven
- İzmir Katip Çelebi University Atatürk Educating and Research Hospital, Department of Ophthalmology, İzmir, Turkey.
| | - T Akbalık
- İzmir Katip Çelebi University Atatürk Educating and Research Hospital, Department of Ophthalmology, İzmir, Turkey
| |
Collapse
|
9
|
Harju N. Regulation of oxidative stress and inflammatory responses in human retinal pigment epithelial cells. Acta Ophthalmol 2022; 100 Suppl 273:3-59. [DOI: 10.1111/aos.15275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Niina Harju
- School of Pharmacy University of Eastern Finland Kuopio Finland
| |
Collapse
|
10
|
Brimonidine Modulates the ROCK1 Signaling Effects on Adipogenic Differentiation in 2D and 3D 3T3-L1 Cells. Bioengineering (Basel) 2022; 9:bioengineering9070327. [PMID: 35877378 PMCID: PMC9311963 DOI: 10.3390/bioengineering9070327] [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: 06/05/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/19/2022] Open
Abstract
The additive effects of an α2-adrenergic agonist, brimonidine (BRI), on the pan-ROCK inhibitor (ROCK-i), ripasudil (Rip), and the ROCK2-I, KD025, on adipogenic differentiation (DIF+) were examined using two- or three-dimension (2D or 3D) cultures of 3T3-L1 cells. The following analyses were carried out: (1) lipid staining (2D and 3D), (2) real-time measurements of cellular metabolism (2D), (3) mRNA expression of DIF+ related genes and extracellular matrix molecules (ECMs) including collagen (Col)-1, -4, and -6, and fibronectin (Fn), and (4) the sizes and physical properties of the 3D spheroids. The findings indicate that DIF+ induced (1) a substantial enhancement in lipid staining and enhanced expression of the Pparγ and Fabp4 genes, (2) significantly larger and softer 3D spheroids, and (3) down-regulation of Col1 and Fn and up-regulation of Col4 and Col6 genes. Treatment with Rip alone caused a significant enhancement in adipogenesis of both the 2D and 3D cultured 3T3-L1 cells and in the physical properties of the 3D spheroids; these effects were substantially inhibited by BRI, and the effects induced by BRI or KD025 were not insignificant. These collective findings indicate that the addition of BRI inhibited the Rip-induced enhancement of DIF+ in 3T3-L1 cells, presumably by modulating ROCK1 signaling.
Collapse
|
11
|
Watanabe M, Sato T, Tsugeno Y, Higashide M, Furuhashi M, Umetsu A, Suzuki S, Ida Y, Hikage F, Ohguro H. An α2-Adrenergic Agonist, Brimonidine, Beneficially Affects the TGF-β2-Treated Cellular Properties in an In Vitro Culture Model. Bioengineering (Basel) 2022; 9:bioengineering9070310. [PMID: 35877361 PMCID: PMC9312232 DOI: 10.3390/bioengineering9070310] [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: 06/11/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
We report herein on the effects of brimonidine (BRI), an α2-adrenergic agonist, on two-dimensional (2D) and three-dimensional (3D) cell-cultured TGF-β2-untreated and -treated human trabecular meshwork (HTM) cells. In the presence of TGF-β2 (5 ng/mL), (1) the effects of BRI on (1) the 2D HTM monolayers’ barrier function were investigated as estimated using trans-endothelial electrical resistance (TEER) measurement and FITC dextran permeability; (2) real-time analyses of cellular metabolism using a Seahorse Bioanalyzer; (3) the largeness and hardness of 3D spheroids; and (4) the expression of genes that encode extracellular matrix (ECM) proteins, including collagens (COL) 1, 4, and 6; fibronectin (FN) and α-smooth muscle actin (α-SMA); ECM modulators, including a tissue inhibitor of matrix proteinase (TIMP) 1–4; matrix metalloproteinase (MMP) 2, 9, and 14; and several endoplasmic reticulum (ER) stress-related genes, including the X-box-binding protein 1 (XBP1), the spliced XBP1 (sXBP1), glucose-regulated protein (GRP)78, GRP94, and CCAAT-enhancer-binding protein homologous protein (CHOP). BRI markedly inhibited the TGF-β2-induced increase in the values of TEER of the 2D cell monolayer and the hardness of the 3D spheroids, although it had no effect on their sizes. BRI also cancelled the TGF-β2-induced reduction in mitochondrial maximal respiration but had no effect on the glycolytic capacity. In addition, the gene expression of these molecules was quite different between the 2D and 3D cultures of HTM cells. The present observations found in this study indicate that BRI may beneficially affect TGF-β2-induced changes in both cultures, 2D and 3D, of HTM cells, although their structural and functional properties that were altered varied significantly between both cultures of HTM cells.
Collapse
Affiliation(s)
- Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.S.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yuri Tsugeno
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (T.S.); (M.F.)
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Soma Suzuki
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Yosuke Ida
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Fumihito Hikage
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan; (M.W.); (Y.T.); (M.H.); (A.U.); (S.S.); (Y.I.); (F.H.)
- Correspondence: ; Tel.: +81-611-2111
| |
Collapse
|
12
|
Tamhane M, Luu KT, Attar M. Ocular Pharmacokinetics of Brimonidine Drug Delivery System in Monkeys and Translational Modeling for Selection of Dose and Frequency in Clinical Trials. J Pharmacol Exp Ther 2021; 378:207-214. [PMID: 34210753 DOI: 10.1124/jpet.120.000483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/28/2021] [Indexed: 01/07/2023] Open
Abstract
Brimonidine, a selective α 2-adrenoceptor agonist, displays putative retinal cyto- and neuroprotective activity in vitro and in vivo. An intravitreal sustained-release brimonidine implant, Brimonidine Posterior Segment Drug Delivery System (brimonidine DDS), allowing targeted drug delivery to the retina has been developed for potential clinical application. This study evaluates the in vivo posterior segment pharmacokinetics of brimonidine DDS implant in the monkey eye and applies translational pharmacokinetic modeling to predict tissue exposure in the human eye. Anesthetized cynomolgus monkeys received a single intravitreal injection of brimonidine DDS 400 µg implant before removal of study eyes at days 7, 30, 60, 92, 120, and 150 postimplant (three to four animals per time point) for assay of brimonidine in aqueous humor, vitreous, and retina samples. Brimonidine concentrations in the human eye were modeled using a linear, three-compartment model assuming bidirectional distribution to/from the aqueous humor and retina and elimination from the aqueous humor. Monkey tissue volumes were scaled up to human values; intercompartmental and elimination rate constants were assumed to be identical in the two species. Modeling and simulations were performed using NONMEM v. 7.3, R 3.5.1. Brimonidine exposure was highest in the monkey vitreous and retina; concentrations in the central (macula) and peripheral retina were maintained at high levels (>100 ng/g) for 3 to 4 months. Simulated brimonidine concentration-time profiles in human macula indicated that brimonidine DDS 400 µg implant would deliver effective drug concentrations (20.7‒82.2 ng/g, based on animal pharmacology) for approximately 3 months. Accordingly, administration of the 400 µg implant at 3-month intervals is recommended. SIGNIFICANCE STATEMENT: Brimonidine, an α 2-adrenoceptor agonist, is cyto- and neuroprotective in animal models of retinal/optic nerve injury. Brimonidine Posterior Segment Drug Delivery System (brimonidine DDS) is an intravitreal sustained-release implant with potential ophthalmological applications. This study explores the pharmacokinetics of brimonidine DDS 400 µg implant in the monkey eye and uses compartmental modeling to predict human ocular tissue exposure. Targeted retinal brimonidine delivery from vitreous was demonstrated in monkeys. Simulated tissue concentration-time profiles indicated persistence of pharmacologically effective brimonidine concentrations for ≈3 months in human retina.
Collapse
|
13
|
Rajagopalan L, Ghosn C, Tamhane M, Almazan A, Andrews-Jones L, Kulkarni A, Christie LA, Burke J, López FJ, Engles M. A nonhuman primate model of blue light-induced progressive outer retina degeneration showing brimonidine drug delivery system-mediated cyto- and neuroprotection. Exp Eye Res 2021; 209:108678. [PMID: 34153289 DOI: 10.1016/j.exer.2021.108678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/26/2022]
Abstract
Geographic atrophy (GA) is an advanced form of age-related macular degeneration (AMD) characterized by atrophy of the retinal pigment epithelium (RPE), loss of photoreceptors, and disruption of choriocapillaris. Excessive light exposure is toxic to the retina and is a known risk factor for AMD. We first investigated the effects of blue light-induced phototoxicity on RPE and photoreceptors in nonhuman primates (NHPs, a model of progressive retinal degeneration) and then evaluated the potential cyto- and neuroprotective effects of the brimonidine drug delivery system (Brimo DDS). In the first set of experiments related to model development, parafoveal lesions of varying severity were induced using blue light irradiation of the retina of cynomolgus monkeys to evaluate the level of phototoxicity in the RPE and photoreceptors. RPE damage was assessed using fundus autofluorescence imaging to quantify areas of hypofluorescence, while thinning of the outer nuclear layer (ONL, photoreceptor nuclei) was quantified using optical coherence tomography (OCT). Photoreceptor function was assessed using multifocal electroretinography (mfERG). RPE damage progressively increased across all lesion severities from 2 to 12 weeks, as did the extent of ONL thinning. Lesions of high severity continued to show reduction in mfERG amplitude, reaching a statistically significant maximum reduction at 12 weeks. Collectively, the first set of experiments showed that blue light irradiation of the NHP eye resulted in progressive retinal degeneration identified by damage to RPE, ONL thinning, and disrupted photoreceptor function - hallmarks of GA in humans. We then used the model to evaluate the cyto- and neuroprotective effects of Brimo DDS, administered as a therapeutic after allowing the lesions to develop for 5 weeks. Placebo DDS or Brimo DDS were administered intravitreally and a set of untreated animals were used as an additional control. In the placebo DDS group, hypofluorescence area continued to increase from baseline, indicating progressive RPE damage, while progression was significantly slowed in eyes receiving Brimo DDS. Likewise, ONL thinning continued to progress over time in eyes that received the placebo DDS, but was reduced in Brimo DDS-treated eyes. Pharmacologically relevant brimonidine concentrations were sustained in the retina for up to 26 weeks following Brimo DDS administration. In summary, Brimo DDS demonstrated cyto- and neuroprotective effects in a novel NHP GA model of progressive retinal degeneration.
Collapse
Affiliation(s)
| | - Corine Ghosn
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Mitalee Tamhane
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Alexandra Almazan
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | | | - Ashutosh Kulkarni
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Lori-Ann Christie
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - James Burke
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Francisco J López
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA
| | - Michael Engles
- Allergan (an AbbVie company), 2525 Dupont Drive, Irvine, CA, 92612, USA.
| |
Collapse
|
14
|
Bhattarai N, Korhonen E, Mysore Y, Kaarniranta K, Kauppinen A. Hydroquinone Induces NLRP3-Independent IL-18 Release from ARPE-19 Cells. Cells 2021; 10:cells10061405. [PMID: 34204067 PMCID: PMC8229790 DOI: 10.3390/cells10061405] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Age-related macular degeneration (AMD) is a retinal disease leading to impaired vision. Cigarette smoke increases the risk for developing AMD by causing increased reactive oxygen species (ROS) production and damage in the retinal pigment epithelium (RPE). We have previously shown that the cigarette tar component hydroquinone causes oxidative stress in human RPE cells. In the present study, we investigated the propensity of hydroquinone to induce the secretion of interleukin (IL)-1β and IL-18. The activation of these cytokines is usually regulated by the Nucleotide-binding domain, Leucine-rich repeat, and Pyrin domain 3 (NLRP3) inflammasome. ARPE-19 cells were exposed to hydroquinone, and cell viability was monitored using the lactate dehydrogenase (LDH) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide salt (MTT) assays. Enzyme-linked immunosorbent assays (ELISAs) were used to measure the levels of proinflammatory cytokines IL-1β and IL-18 as well as NLRP3, caspase-1, and poly (ADP-ribose) polymerase (PARP). Hydroquinone did not change IL-1β release but significantly increased the secretion of IL-18. Cytoplasmic NLRP3 levels increased after the hydroquinone treatment of IL-1α-primed RPE cells, but IL-18 was equally released from primed and nonprimed cells. Hydroquinone reduced the intracellular levels of PARP, which were restored by treatment with the ROS scavenger N-acetyl-cysteine (NAC). NAC concurrently reduced the NLRP3 levels but had no effect on IL-18 release. In contrast, the NADPH oxidase inhibitor ammonium pyrrolidinedithiocarbamate (APDC) reduced the release of IL-18 but had no effect on the NLRP3 levels. Collectively, hydroquinone caused DNA damage seen as reduced intracellular PARP levels and induced NLRP3-independent IL-18 secretion in human RPE cells.
Collapse
Affiliation(s)
- Niina Bhattarai
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (Y.M.)
- Correspondence: (N.B.); (A.K.); Tel.: +358-44-983-0424 (N.B.); +358-40-355-3216 (A.K.)
| | - Eveliina Korhonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (Y.M.)
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Yashavanthi Mysore
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (Y.M.)
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland;
- Department of Ophthalmology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (Y.M.)
- Correspondence: (N.B.); (A.K.); Tel.: +358-44-983-0424 (N.B.); +358-40-355-3216 (A.K.)
| |
Collapse
|
15
|
Wang L, Mao X. Role of Retinal Amyloid-β in Neurodegenerative Diseases: Overlapping Mechanisms and Emerging Clinical Applications. Int J Mol Sci 2021; 22:2360. [PMID: 33653000 PMCID: PMC7956232 DOI: 10.3390/ijms22052360] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/03/2023] Open
Abstract
Amyloid-β (Aβ) accumulations have been identified in the retina for neurodegeneration-associated disorders like Alzheimer's disease (AD), glaucoma, and age-related macular degeneration (AMD). Elevated retinal Aβ levels were associated with progressive retinal neurodegeneration, elevated cerebral Aβ accumulation, and increased disease severity with a decline in cognition and vision. Retinal Aβ accumulation and its pathological effects were demonstrated to occur prior to irreversible neurodegeneration, which highlights its potential in early disease detection and intervention. Using the retina as a model of the brain, recent studies have focused on characterizing retinal Aβ to determine its applicability for population-based screening of AD, which warrants a further understanding of how Aβ manifests between these disorders. While current treatments directly targeting Aβ accumulations have had limited results, continued exploration of Aβ-associated pathological pathways may yield new therapeutic targets for preserving cognition and vision. Here, we provide a review on the role of retinal Aβ manifestations in these distinct neurodegeneration-associated disorders. We also discuss the recent applications of retinal Aβ for AD screening and current clinical trial outcomes for Aβ-associated treatment approaches. Lastly, we explore potential future therapeutic targets based on overlapping mechanisms of pathophysiology in AD, glaucoma, and AMD.
Collapse
Affiliation(s)
- Liang Wang
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Kuppermann BD, Patel SS, Boyer DS, Augustin AJ, Freeman WR, Kerr KJ, Guo Q, Schneider S, López FJ. PHASE 2 STUDY OF THE SAFETY AND EFFICACY OF BRIMONIDINE DRUG DELIVERY SYSTEM (BRIMO DDS) GENERATION 1 IN PATIENTS WITH GEOGRAPHIC ATROPHY SECONDARY TO AGE-RELATED MACULAR DEGENERATION. Retina 2021; 41:144-155. [PMID: 32134802 DOI: 10.1097/iae.0000000000002789] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE To evaluate the safety and efficacy of Brimonidine Drug Delivery System (Brimo DDS), a biodegradable intravitreal implant, in the treatment of geographic atrophy (GA) secondary to age-related macular degeneration. METHODS Phase 2, randomized, multicenter, double-masked, 24-month study. Study eyes were treated (Day 1; Month 6 retreatment) with Brimo DDS 132 µg (n = 49), Brimo DDS 264 µg (n = 41), or sham procedure (n = 23). The primary timepoint for efficacy analysis was Month 12. RESULTS Mean GA area growth at Month 12 was 1.78 mm2, 1.59 mm2, and 2.19 mm2 in the Brimo DDS 132 µg, 264 µg, and sham groups, respectively. Geographic atrophy area growth was consistently smaller with Brimo DDS 132 and 264 µg than sham; between-group differences were significant (P ≤ 0.032) at Month 3. In patients with baseline lesion area ≥6 mm2 (two-thirds of patients), GA lesion area and effective radius growth was reduced with Brimo DDS 132 and 264 µg at Month 12 (P ≤ 0.050 vs. sham). Treatment-related adverse events were usually injection procedure-related. CONCLUSION Brimo DDS demonstrated a favorable safety profile and reduced GA lesion area growth at Month 3. Lesion growth at Month 12 was reduced in patients with baseline GA lesion area ≥6 mm2. The results support Phase 3 development.
Collapse
Affiliation(s)
- Baruch D Kuppermann
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, California
| | | | - David S Boyer
- Retina-Vitreous Associates Medical Group, Los Angeles, California
| | - Albert J Augustin
- Department of Ophthalmology, Staedtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - William R Freeman
- Jacobs Retina Center, University of California San Diego, San Diego, California; and
| | | | | | | | | |
Collapse
|
17
|
Therapeutic Options Under Development for Nonneovascular Age-Related Macular Degeneration and Geographic Atrophy. Drugs Aging 2020; 38:17-27. [PMID: 33355716 DOI: 10.1007/s40266-020-00822-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 01/05/2023]
Abstract
Age-related macular degeneration (AMD) is a chronic, multifactorial disease and a leading cause of irreversible blindness in the elderly population in the Western Hemisphere. Among the two major subtypes of AMD, the prevalence of the nonneovascular (dry) type is approximately 85-90% and the neovascular (wet) type is 10-15%. Healthy lifestyle and nutritional supplements of anti-oxidative micronutrients have been shown to delay the progression of dry AMD and lower the risk of development of wet AMD, and anti-vascular endothelial growth factor (anti-VEGF) injections have been shown to improve visual acuity for wet AMD patients. However, to date, there is no approved treatment for geographic atrophy (GA), a debilitating late stage of dry AMD. Thus, this represents a large unmet need in this patient population. This review focuses on the current management and treatment of nonneovascular AMD, the drugs and devices that have been under investigation for the treatment of GA, and the latest clinical trial results. A few therapeutic options have shown initial promising clinical trial results, but failed to show efficacy in larger trials, while others are awaiting future clinical trial results and long-term follow-up to evaluate safety and efficacy.
Collapse
|
18
|
Neal SE, Buehne KL, Besley NA, Yang P, Silinski P, Hong J, Ryde IT, Meyer JN, Jaffe GJ. Resveratrol Protects Against Hydroquinone-Induced Oxidative Threat in Retinal Pigment Epithelial Cells. Invest Ophthalmol Vis Sci 2020; 61:32. [PMID: 32334435 PMCID: PMC7401947 DOI: 10.1167/iovs.61.4.32] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose Oxidative stress in retinal pigment epithelial (RPE) cells is associated with age-related macular degeneration (AMD). Resveratrol exerts a range of protective biologic effects, but its mechanism(s) are not well understood. The aim of this study was to investigate how resveratrol could affect biologic pathways in oxidatively stressed RPE cells. Methods Cultured human RPE cells were treated with hydroquinone (HQ) in the presence or absence of resveratrol. Cell viability was determined with WST-1 reagent and trypan blue exclusion. Mitochondrial function was measured with the XFe24 Extracellular Flux Analyzer. Expression of heme oxygenase-1 (HO-1) and glutamate cysteine ligase catalytic subunit was evaluated by qPCR. Endoplasmic reticulum stress protein expression was measured by Western blot. Potential reactions between HQ and resveratrol were investigated using high-performance liquid chromatography mass spectrometry with resveratrol and additional oxidants for comparison. Results RPE cells treated with the combination of resveratrol and HQ had significantly increased cell viability and improved mitochondrial function when compared with HQ-treated cells alone. Resveratrol in combination with HQ significantly upregulated HO-1 mRNA expression above that of HQ-treated cells alone. Resveratrol in combination with HQ upregulated C/EBP homologous protein and spliced X-box binding protein 1. Additionally, new compounds were formed from resveratrol and HQ coincubation. Conclusions Resveratrol can ameliorate HQ-induced toxicity in RPE cells through improved mitochondrial bioenergetics, upregulated antioxidant genes, stimulated unfolded protein response, and direct oxidant interaction. This study provides insight into pathways through which resveratrol can protect RPE cells from oxidative damage, a factor thought to contribute to AMD pathogenesis.
Collapse
|
19
|
Bhattarai N, Korhonen E, Toppila M, Koskela A, Kaarniranta K, Mysore Y, Kauppinen A. Resvega Alleviates Hydroquinone-Induced Oxidative Stress in ARPE-19 Cells. Int J Mol Sci 2020; 21:ijms21062066. [PMID: 32192228 PMCID: PMC7139575 DOI: 10.3390/ijms21062066] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 12/18/2022] Open
Abstract
Retinal pigment epithelial (RPE) cells maintain homeostasis at the retina and they are under continuous oxidative stress. Cigarette smoke is a prominent environmental risk factor for age-related macular degeneration (AMD), which further increases the oxidant load in retinal tissues. In this study, we measured oxidative stress and inflammatory markers upon cigarette smoke-derived hydroquinone exposure on human ARPE-19 cells. In addition, we studied the effects of commercial Resvega product on hydroquinone-induced oxidative stress. Previously, it was observed that Resvega induces autophagy during impaired protein clearance in ARPE-19 cells, for which it has the potential to alleviate pro-inflammatory pathways. Cell viability was determined while using the lactate dehydrogenase (LDH) and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, and the cytokine levels were measured using the enzyme-linked immunosorbent assay (ELISA). Reactive oxygen species (ROS) production were measured using the 2',7'-dichlorofluorescin diacetate (H2DCFDA) probe. Hydroquinone compromised the cell viability and increased ROS production in ARPE-19 cells. Resvega significantly improved cell viability upon hydroquinone exposure and reduced the release of interleukin (IL)-8 and monocytic chemoattractant protein (MCP)-1 from RPE cells. Resvega, N-acetyl-cysteine (NAC) and aminopyrrolidine-2,4-dicarboxylic acid (APDC) alleviated hydroquinone-induced ROS production in RPE cells. Collectively, our results indicate that hydroquinone induces cytotoxicity and increases oxidative stress through NADPH oxidase activity in RPE cells, and resveratrol-containing Resvega products prevent those adverse effects.
Collapse
Affiliation(s)
- Niina Bhattarai
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (M.T.); (Y.M.)
- Correspondence: (N.B.); (A.K); Tel.: +358-44-9830424 (N.B.); +358-40-3553216 (A.K.)
| | - Eveliina Korhonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (M.T.); (Y.M.)
- Department of Clinical Chemistry, HUSLAB, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Maija Toppila
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (M.T.); (Y.M.)
| | - Ali Koskela
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.K.); (K.K.)
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.K.); (K.K.)
- Department of Ophthalmology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Yashavanthi Mysore
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (M.T.); (Y.M.)
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (E.K.); (M.T.); (Y.M.)
- Correspondence: (N.B.); (A.K); Tel.: +358-44-9830424 (N.B.); +358-40-3553216 (A.K.)
| |
Collapse
|
20
|
Cáceres-Del-Carpio J, Moustafa MT, Toledo-Corral J, Hamid MA, Atilano SR, Schneider K, Fukuhara PS, Costa RD, Norman JL, Malik D, Chwa M, Boyer DS, Limb GA, Kenney MC, Kuppermann BD. In vitro response and gene expression of human retinal Müller cells treated with different anti-VEGF drugs. Exp Eye Res 2020; 191:107903. [PMID: 31904361 PMCID: PMC7058176 DOI: 10.1016/j.exer.2019.107903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/18/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022]
Affiliation(s)
| | - M Tarek Moustafa
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | | | - Mohamed A Hamid
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Shari R Atilano
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Kevin Schneider
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Paula S Fukuhara
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | | | - J Lucas Norman
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Deepika Malik
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - David S Boyer
- Retina-Vitreous Associates Medical Group, Los Angeles, CA, USA
| | - G Astrid Limb
- Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK
| | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA.
| | - Baruch D Kuppermann
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, USA
| |
Collapse
|
21
|
Machado M, Silva GA, Bitoque DB, Ferreira J, Pinto LA, Morgado J, Ferreira Q. Self-Assembled Multilayer Films for Time-Controlled Ocular Drug Delivery. ACS APPLIED BIO MATERIALS 2019; 2:4173-4180. [PMID: 35021432 DOI: 10.1021/acsabm.9b00417] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The patient's compliance on the therapeutics to treat glaucoma is significantly low contributing for a fast evolution of the disease. This article presents an autonomous system with controlled release using an alpha2-adrenergic receptor agonist, brimonidine, usually used to treat glaucoma. More specifically, biocompatible and layer-by-layer drug delivery films containing monolayers with brimonidine encapsulated in polymer-β-cyclodextrin were prepared with the objective to obtain a system able to release precise amounts of drug at specific times. To delay the erosion-controlled drug release, we included nanosheets of graphene oxide and layers of a biodegradable polymer (poly-β-aminoester) between the drug-containing monolayers to obtain a time-controlled drug delivery system. An increase in the number of graphene oxide layers is proportional to the brimonidine release delay and its kinetic release can be tuned as a function of the number of layers. Two types of films with brimonidine encapsulated in β-cyclodextrin were analyzed. One of them composed of barrier layers with PBAE and another with two types of barrier layers, PBAE and graphene oxide. The results indicate that one graphene oxide bilayer can delay the brimonidine release for more than 24 h. In vitro assays confirmed that the films have a cell viability of 100%.
Collapse
Affiliation(s)
- Mónica Machado
- Instituto de Telecomunicações, Avenida Rovisco Pais, Lisbon 1049-001, Portugal
| | - Gabriela A Silva
- CEDOC Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, Lisboa 1169-056, Portugal.,NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal
| | - Diogo B Bitoque
- CEDOC Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, Lisboa 1169-056, Portugal
| | - Joana Ferreira
- Ophthalmology Department, Centro Hospitalar Universitário de Lisboa Central, Lisbon 1169-050, Portugal.,NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal
| | - Luís A Pinto
- Ophthalmology Department, Centro Hospitalar Universitário de Lisboa Norte, Lisbon 1649-035, Portugal.,Visual Sciences Study Centre, Faculty of Medicine, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Jorge Morgado
- Instituto de Telecomunicações, Avenida Rovisco Pais, Lisbon 1049-001, Portugal.,Bioengineering Department, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, Lisbon 1049-001, Portugal
| | - Quirina Ferreira
- Instituto de Telecomunicações, Avenida Rovisco Pais, Lisbon 1049-001, Portugal
| |
Collapse
|
22
|
Tsao SW, Gabriel R, Thaker K, Kuppermann BD, Kenney MC. Effects of Brimonidine on Retinal Pigment Epithelial Cells and Müller Cells Exposed to Amyloid-Beta 1-42 Peptide In Vitro. Ophthalmic Surg Lasers Imaging Retina 2019; 49:S23-S28. [PMID: 30339264 DOI: 10.3928/23258160-20180814-04] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVE To evaluate whether brimonidine can prevent cytotoxicity in human retinal pigment epithelial (RPE) and Müller (MIO) cells after exposure to amyloid-beta 1-42 (Aβ42). MATERIALS AND METHODS An in vitro model of geographic atrophy (GA), which is an end-stage complication of age-related macular degeneration (AMD), simulated with the application of Aβ42 in cell culture. RPE and MIO cells were pretreated with brimonidine for 6 hours, then exposed to 10μM Aβ42 for 24 hours. Several concentrations (one time [1×], two times [2×], and five times [5×]) of brimonidine were used to assess for a dose-related effect. Assays were immediately run following the treatment period. 2',7'-Dichlorofluorescein diacetate was used to assess reactive oxygen species production, the MTT assay was used to assess cell viability, and the JC-1 dye assay was used to assess mitochondrial membrane potential. The main outcome measures were reactive oxygen species (ROS) production, cell viability, and mitochondrial membrane potential (ΔΨm) of RPE and MIO cells following the treatment phase. RESULTS High-dose (5×) brimonidine was capable of reducing ROS production in RPE and MIO cells with exposure to Aβ42. The application of Aβ42 alone did not trigger a rise in ROS production. Brimonidine was unable to rescue cell viability and ΔΨm after exposure to Aβ42 in both cell cultures. Instead, high-dose (5×) brimonidine appeared to increase the toxicity to cell viability and ΔΨm in cultures exposed to Aβ42. However, this was not due to medication toxicity alone, because high-dose (5×) brimonidine without exposure to Aβ42 did not affect the cell viability in both cell types. CONCLUSION Brimonidine may have a role in preventing oxidative cellular injury in AMD. However, this role does not appear to translate into protection against some of the cytotoxic effects observed from this in vitro model of GA. In this cellular model of GA, brimonidine is able to reduce oxidative stress but is unable to rescue cell viability or prevent mitochondrial dysfunction. [Ophthalmic Surg Lasers Imaging Retina. 2018;49:S23-S28.].
Collapse
|
23
|
Moustafa MT, Ramirez C, Schneider K, Atilano SR, Limb GA, Kuppermann BD, Kenney MC. Protective Effects of Memantine on Hydroquinone-Treated Human Retinal Pigment Epithelium Cells and Human Retinal Müller Cells. J Ocul Pharmacol Ther 2017; 33:610-619. [PMID: 28961056 DOI: 10.1089/jop.2016.0129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Memantine (MEM) acts on the glutamatergic system by blocking N-methyl-d-aspartate (NMDA) glutamate receptors. The role that MEM plays in protecting retinal cells is unknown. Hydroquinone (HQ) is one of the cytotoxic components in cigarette smoke. In the present study, we tested whether pretreatment with MEM could protect against the cytotoxic effects of HQ on human retinal pigment epithelium cells (ARPE-19) and human retinal Müller cells (MIO-M1) in vitro. METHODS Cells were plated, pretreated for 6 h with 30 μM of MEM, and then exposed for 24 h to 200, 100, 50, and 25 μM of HQ while MEM was still present. Cell viability (CV), reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm), and lactate dehydrogenase (LDH) release assays were performed. RESULTS HQ-treated cells showed a dose-dependent decrease in CV and ΔΨm, but an increase in ROS production and LDH levels in both cell lines. MEM pretreatment reversed the CV in 50, 100, and 200 μM doses in ARPE-19 cells and at all HQ concentrations in MIO-M1 cells compared to HQ-treated cultures. ROS production was reversed in all HQ concentrations in both cell lines. ΔΨm was significantly increased after MEM pretreatment only in 50 μM HQ concentration for both cell lines. LDH levels were decreased at 50 and 25 μM HQ in ARPE-19 and MIO-M1 cells, respectively. CONCLUSION HQ-induced toxicity is concentration dependent in ARPE-19 and MIO-M1 cultures. MEM exerts protective effects against HQ-induced toxicity on human retinal pigment epithelial and Müller cells in vitro.
Collapse
Affiliation(s)
- Mohamed Tarek Moustafa
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
- 2 Ophthalmology Department, Minia University , Minia, Egypt
| | - Claudio Ramirez
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
| | - Kevin Schneider
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
| | - Shari R Atilano
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
| | - Gloria Astrid Limb
- 3 Division of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology , London, United Kingdom
| | - Baruch D Kuppermann
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
| | - Maria Cristina Kenney
- 1 Gavin Herbert Eye Institute, University of California, Irvine , Irvine, California
| |
Collapse
|
24
|
Li H, Chintalapudi SR, Jablonski MM. Current drug and molecular therapies for the treatment of atrophic age-related macular degeneration: phase I to phase III clinical development. Expert Opin Investig Drugs 2017; 26:1103-1114. [PMID: 28816076 DOI: 10.1080/13543784.2017.1369042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly. Atrophic AMD, including early, intermediate and geographic atrophy (GA), accounts for ~90% of all cases. It is a multifactorial degeneration characterized by chronic inflammation, oxidative stress and aging components. Although no FDA-approved treatment yet exists for the late stage of atrophic AMD, multiple pathological mechanisms are partially known and several promising therapies are in various stages of development. Areas covered: Underlying mechanisms that define atrophic AMD will help provide novel therapeutic targets that will address this largely unmet clinical need. The purpose of this paper is to review current promising drugs that are being evaluated in clinical trials. Because no pharmacological treatments are currently available for late stage of atrophic AMD, any new therapy would have extensive market potential. Expert opinion: The number of AMD patients is predicted to increase to ~30 million worldwide by 2020. In response to this enormous unmet clinical need, new promising therapies are being developed and evaluated in clinical trials. We propose that the assessment of novel interventions will also need to consider the genotypes of participants, as the benefit may be determined by polymorphisms in an individual's genetic background.
Collapse
Affiliation(s)
- Huiling Li
- a Department of Ophthalmology, The Second Xiangya Hospital , Central South University , Changsha , China
| | - Sumana R Chintalapudi
- b Department of Ophthalmology, The Hamilton Eye Institute , The University of Tennessee Health Science Center , Memphis , TN , USA.,c Department of Anatomy and Neurobiology , The University of Tennessee Health Science Center , Memphis , TN , USA
| | - Monica M Jablonski
- b Department of Ophthalmology, The Hamilton Eye Institute , The University of Tennessee Health Science Center , Memphis , TN , USA.,c Department of Anatomy and Neurobiology , The University of Tennessee Health Science Center , Memphis , TN , USA.,d Department of Pharmaceutical Sciences , The University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|