1
|
Zaluski J, Bassetto M, Kiser PD, Tochtrop GP. Advances and therapeutic opportunities in visual cycle modulation. Prog Retin Eye Res 2025; 106:101360. [PMID: 40280538 DOI: 10.1016/j.preteyeres.2025.101360] [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: 02/14/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The visual cycle is a metabolic pathway that enables continuous vision by regenerating the 11-cis-retinal chromophore for photoreceptors opsins. Although integral to normal visual function, the flux of retinoids through this cycle can contribute to a range of retinal pathologies, including Stargardt disease, age-related macular degeneration, and diabetic retinopathy. In such conditions, intermediates and byproducts of the visual cycle, such as bisretinoid components of lipofuscin, can accumulate, concomitant with cellular damage and eventual photoreceptor loss. This has inspired efforts to modulate the visual cycle, aiming to slow or prevent the formation of these toxic intermediates and thus preserve retinal structure and function. Over the past two decades, multiple strategies to modulate the visual cycle have emerged. These include both intrinsic approaches, targeting key enzymes, retinoid-binding proteins, or receptors within the pigment epithelium or photoreceptors (e.g., RPE65, CRBP1, and rhodopsin inhibitors/antagonists) and extrinsic strategies that indirectly alter retinoid availability within the retina (e.g., RBP4 antagonists). Many of these agents have shown promise in animal models of visual cycle-associated retinal diseases, reducing pathological changes, and improving retinal survival. Several have advanced into clinical studies, although none are currently FDA-approved. Challenges remain in optimizing drug specificity and duration of action while minimizing side effects such as nyctalopia. In this review, we comprehensively examine current and emerging visual cycle modulators, discuss their medicinal chemistry, mechanisms of action, efficacy in preclinical and clinical studies, and highlight future opportunities for drug discovery aimed at safely and effectively preserving vision through modulation of this biochemical pathway.
Collapse
Affiliation(s)
- Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA; Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA.
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
2
|
Cui Y, Poudel S, Xu N, Zhou K, Cheng R, Liang W, Yuan T, Zhao L, Qin C, Stevens KG, Duerfeldt AS, Hu J, Xu Q, Ma JX. Sustained release of a novel non-fibrate PPARα agonist from microparticles for neuroprotection in murine models of age-related macular degeneration. J Control Release 2025; 380:910-926. [PMID: 39961437 DOI: 10.1016/j.jconrel.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Prior research has demonstrated the therapeutic potential of peroxisome proliferator-activated receptor α (PPARα) agonist fenofibrate on diabetic retinopathy. In the present study, a novel non-fibrate PPARα agonist, A190, was designed with higher potency and selectivity than fenofibrate in PPARα agonism. A190 was encapsulated in biodegradable microparticles (A190-MP) to ensure sustained drug release, with detection in the retina up to 6 months following a single intravitreal injection. A190-MP alleviated retinal dysfunction as shown by electroretinography in Vldlr-/- (wet-AMD model) and Abca4-/-/Rdh8-/- (dry-AMD model) mice. A190-MP also attenuated the decreases in cone photoreceptor density and outer nuclear layer thickness as demonstrated by optical coherence tomography and histology. Moreover, A190-MP reduced vascular leakage and neovascularization in Vldlr-/- mice, suggesting an anti-inflammatory and anti-angiogenic effect. A190-MP upregulated expression of PPARα, PGC1α, and TOMM20 in the retina of Vldlr-/- and Abca4-/-/Rdh8-/- mice. A190-MP also improved retinal mitochondrial function as shown by Seahorse analysis using retinal biopsy. In vitro, A190 attenuated oxidative stress and preserved cell viability in a photoreceptor-derived cell line exposed to 4-HNE and improved mitochondrial function, via a PPARα-dependent mechanism. These findings revealed sustained therapeutic effects of A190-MP in wet and dry AMD models, through improving mitochondrial function by activating PPARα.
Collapse
Affiliation(s)
- Yi Cui
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Sagun Poudel
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Nuo Xu
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America; Department of Ophthalmology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China
| | - Kelu Zhou
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Rui Cheng
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Wentao Liang
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Tian Yuan
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Long Zhao
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Chaolong Qin
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Katelyn G Stevens
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, United States of America
| | - Adam S Duerfeldt
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, United States of America
| | - Jianzhang Hu
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Qingguo Xu
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Department of Ophthalmology, Pediatrics, Biomedical Engineering, Center for Pharmaceutical Engineering, and Institute for Structural Biology, Drug Discovery & Development (ISB3D), Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States of America.
| |
Collapse
|
3
|
Liu X, Liu Z, Cui J, Tan C, Sun W, Lin Y. Clinical and Genetic Characteristics of 18 Patients from Southeast China with ABCA4-Associated Stargardt Disease. Int J Mol Sci 2025; 26:3354. [PMID: 40244202 PMCID: PMC11989659 DOI: 10.3390/ijms26073354] [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: 02/15/2025] [Revised: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Stargardt disease (STGD1), the most common retinal dystrophy caused by pathogenic variants of the biallelic ABCA4 gene, results in irreversible vision loss. This cross-sectional case series study analyzes 18 unrelated Stargardt disease (STGD1) patients from southeast China, examining clinical and genetic features. Ophthalmological assessments included BCVA, ophthalmoscopy, fundus photography, and autofluorescence, with ultra-widefield OCT angiography carried out on one patient. Genetic testing uses targeted exome sequencing for eye disease genes. The mean age of onset was 44.3 years for adult onset (6 patients) and 9.6 years for childhood/adolescent onset (12 patients). The mean logMAR visual acuity was 0.96 (right eye) and 0.91 (left eye). Eight novel ABCA4 variants were found, including two nonsense, two frameshift deletions, one copy number variant, one splice-site alternation, and two deep intronic variants. The genotypes are as follows: 77.8% (14/18) biallelic heterozygous, 16.7% (3/18) homozygous, and one patient with three variants. The study underscores STGD1's phenotypic and genotypic diversity, expands the ABCA4 mutation spectrum, and offers insights into therapeutic strategies.
Collapse
Affiliation(s)
- Xinyu Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; (X.L.); (Z.L.); (J.C.); (W.S.)
| | - Zehao Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; (X.L.); (Z.L.); (J.C.); (W.S.)
| | - Jinli Cui
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; (X.L.); (Z.L.); (J.C.); (W.S.)
| | - Chen Tan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
| | - Wenmin Sun
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; (X.L.); (Z.L.); (J.C.); (W.S.)
| | - Ying Lin
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; (X.L.); (Z.L.); (J.C.); (W.S.)
| |
Collapse
|
4
|
Lee SY, Yoon CK, Park UC, Park KH, Lee EK. CHOROIDAL HYPERREFLECTIVE FOCI AS BIOMARKERS OF SEVERITY IN STARGARDT DISEASE. Retina 2025; 45:774-784. [PMID: 39661867 DOI: 10.1097/iae.0000000000004367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
PURPOSE To investigate the clinical implications of choroidal hyperreflective foci (HF) as biomarkers of disease severity in Stargardt disease. METHODS One hundred and twenty-nine eyes from 66 patients with Stargardt disease were included. The primary outcome was the correlation between the number of foveal choroidal HF and indicators of disease severity. Secondary outcome included a comparison of choroidal HF between the pathologic and healthy borders of atrophy, and the 2-year longitudinal change of foveal choroidal HF. RESULTS Long disease duration, thin central macular thickness, large definitely decreased autofluorescence area, atrophic foveal retinal pigment epithelium, high Fishman stage, and low visual acuity were associated with a higher number of foveal choroidal HF. Foveal retinal pigment epithelium involvement (β = 0.522, P < 0.001) was the most strongly correlated factor in multivariate analysis. Genotype-phenotype relationships between the number of severe ABCA4 variants and choroidal HF could not be determined. The pathologic border had significantly more choroidal HF than the healthy border ( P < 0.001), and the number of foveal choroidal HF increased over 2 years ( P = 0.003). CONCLUSION Choroidal HF could serve as a biomarker of disease severity in Stargardt disease, correlating with various severity indicators and especially reflecting the presence of retinal pigment epithelium atrophy.
Collapse
Affiliation(s)
- Seo Yoon Lee
- Medical Program, Seoul National University College of Medicine, Seoul, Korea ; and
| | - Chang Ki Yoon
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Un Chul Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Eun Kyoung Lee
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
5
|
Lara-López A, Gonzalez-Imaz K, Rodríguez-Hidalgo M, Sarasola-Gastesi M, Escudero-Arrarás L, Milla-Navarro S, de la Villa P, Sagartzazu-Aizpurua M, Miranda JI, Aizpurua JM, de Munain AL, Vallejo-Illarramendi A, Ruiz-Ederra J. Topical Administration of Novel FKBP12 Ligand MP-004 Improves Retinal Function and Structure in Retinitis Pigmentosa Models. Invest Ophthalmol Vis Sci 2025; 66:56. [PMID: 40136284 PMCID: PMC11951062 DOI: 10.1167/iovs.66.3.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose This study evaluates the therapeutic potential of MP-004, a novel FKBP12 ligand, in the treatment of inherited retinal dystrophies (IRDs). MP-004 targets the FKBP12/RyR interaction, which is disrupted in several neurologic disorders with underlying oxidative stress. Methods The toxicity and efficacy of MP-004 were examined in vitro in 661W cells. Efficacy was evaluated in phototoxic and H2O2-induced damage using impedance assays, calcium imaging, and in situ PLA. In vivo, MP-004 efficacy was evaluated in the rd10 mouse model of retinitis pigmentosa (RP) by topical ocular instillation. Retinal function was assessed by electroretinography (ERG), visual acuity was measured using a water maze test, and retinal structure was analyzed morphometrically. Results MP-004 exhibited low toxicity (LD50: 1.22 mM) and effectively protected 661W cells from phototoxicity (EC50: 30.6 nM). Under oxidative stress conditions, MP-004 preserved the FKBP12.6/RyR2 interaction, restored cytosolic and endoplasmic reticulum calcium levels, and prevented cell death. In vivo, MP-004 significantly preserved retinal function in rd10 mice, with ERG wave amplitude increases of up to 50% in scotopic and 71% in photopic conditions, corresponding to rod and cone functions, respectively. Additionally, MP-004 improved visual acuity for low spatial frequency patterns and preserved retinal structure, with a 23% increase in outer nuclear layer thickness and preservation in the number of rods and cones and their segment length. Conclusions MP-004 shows promise as a therapeutic agent for RP, preserving retinal structure and function, likely through modulation of the FKBP12.6/RyR2 interaction. Further studies are needed to explore its pharmacokinetics and efficacy in other IRD models.
Collapse
Affiliation(s)
- Araceli Lara-López
- Miramoon Pharma, S.L., Donostia-San Sebastian, Spain
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Klaudia Gonzalez-Imaz
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | - María Rodríguez-Hidalgo
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
| | - Miren Sarasola-Gastesi
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- Department of Dermatology, Ophthalmology and ORL, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Leire Escudero-Arrarás
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
| | - Santiago Milla-Navarro
- Department of System Biology, University of Alcalá, Alcalá de Henares, Spain
- Visual Neurophysiology Group, Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pedro de la Villa
- Department of System Biology, University of Alcalá, Alcalá de Henares, Spain
- Visual Neurophysiology Group, Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Maialen Sagartzazu-Aizpurua
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - José Ignacio Miranda
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Jesús María Aizpurua
- Department of Organic Chemistry-I, Korta Research Center, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Adolfo López de Munain
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, OSAKIDETZA, Donostia-San Sebastián, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | - Javier Ruiz-Ederra
- Miramoon Pharma, S.L., Donostia-San Sebastian, Spain
- Groups of Sensorial Neurodegeneration and Neuromuscular Diseases, Neuroscience Area, Biogipuzkoa Health Research Institute (IIS Biodonostia), Donostia-San Sebastian, Spain
- Department of Dermatology, Ophthalmology and ORL, University of Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| |
Collapse
|
6
|
Yang B, Yang K, Xi R, Li S, Chen J, Wu Y. Inhibition of JNK signaling attenuates photoreceptor ferroptosis caused by all-trans-retinal. Free Radic Biol Med 2025; 227:179-189. [PMID: 39643129 DOI: 10.1016/j.freeradbiomed.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
The disruption of the visual cycle leads to the accumulation of all-trans-retinal (atRAL) in the retina, a hallmark of autosomal recessive Stargardt disease (STGD1) and dry age-related macular degeneration (AMD), both of which cause retinal degeneration. Although our previous studies have shown that atRAL induces ferroptosis and activates c-Jun N-terminal kinase (JNK) signaling in the retina, the relationship between JNK signaling and ferroptosis in atRAL-mediated photoreceptor damage remains unclear. Here, we reported that JNK activation by atRAL drove photoreceptor ferroptosis through ferritinophagy. In photoreceptor cells loaded with atRAL, activated JNK phosphorylated c-Jun, which facilitated its nuclear translocation and promoted the expression of the nuclear receptor coactivator 4 (NCOA4). Elevated NCOA4 induced ferritin degradation via lysosomal processing, a process known as ferritinophagy, thereby releasing a large amount of labile iron. Iron overload led to the generation of reactive oxygen species (ROS) and lipid peroxidation, ultimately culminating in ferroptosis. Treatment with the JNK inhibitor JNK-IN-8, as well as the knockout of Jnk1 and Jnk2 genes, significantly rescued atRAL-loaded photoreceptor cells from ferritinophagy-induced ferroptosis. Abca4-/-Rdh8-/- mice, which exhibit atRAL accumulation in the retina following light exposure, are commonly used to study the pathological processes of STGD1 and dry AMD. In these mice, light exposure activated the JNK/c-Jun/NCOA4 axis, resulting in ferritinophagy in the neural retina. Importantly, intraperitoneal administration of JNK-IN-8 significantly rescued retinal function and photoreceptors from ferritinophagy-induced ferroptosis and effectively mitigated retinal degeneration in light-exposed Abca4-/-Rdh8-/- mice. This study underscores the critical role of the JNK/c-Jun/NCOA4 axis in mediating atRAL-induced ferritinophagy, which drives ferroptosis and retinal atrophy, suggesting that targeting this pathway may offer a potential therapeutic approach for STGD1 and dry AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; 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, 361102, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; 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, 361102, China
| | - Ruitong Xi
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; 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, 361102, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361003, China; 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, 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China.
| |
Collapse
|
7
|
Giuffrida E, Platania CBM, Lazzara F, Conti F, Sotera L, Drago F, Herath D, Motterlini R, Foresti R, Bucolo C. α1D Adrenergic Receptor Antagonism Protects Against High Glucose-Induced Mitochondrial Dysfunction and Blood Retinal Barrier Breakdown in ARPE-19 Cells. Int J Mol Sci 2025; 26:967. [PMID: 39940746 PMCID: PMC11817144 DOI: 10.3390/ijms26030967] [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: 12/16/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Diabetic retinopathy (DR) is a microvascular complication of diabetes mellitus and a leading cause of blindness in the working-age population. Current pharmacological treatments counteract DR's later stages without targeting the earlier disease phases. Using computational approaches, our group previously identified the α1D and α2C adrenoceptors (α1DR and α2CR) as new putative drug targets for DR. Therefore, the aim of this work was to validate the role of these receptors in an in vitro model of DR, i.e., retinal pigmented epithelial cells (ARPE-19) challenged with high glucose (HG, 50 mM). We examined the effects of selective α1DR and α2CR agonists and antagonists on hyperglycemia-induced mitochondrial dysfunction and blood retinal barrier breakdown. Seahorse XFe was employed to assess the oxygen consumption rate and extracellular acidification rate. The integrity of the ARPE-19 barrier was evaluated through transepithelial electrical resistance measurements and a sodium fluorescein permeability test. α1DR pharmacological modulation through the α1DR antagonist BMY 7378 (0.1-1 µM, 24 h), but not α2CR, significantly attenuated HG-induced mitochondrial dysfunction. BMY 7378 (0.1-1 µM, 48 h) also prevented HG-mediated damage to retinal epithelial integrity. In contrast, the α1DR agonist phenylephrine (1-10 μM, 24 h) further reduced ARPE-19 mitochondrial activity compared to HG, indicating that α1D activation is directly implicated in DR-mediated mitochondrial dysfunction. In conclusion, the current in vitro study validated α1DR as a pharmacological target for DR.
Collapse
Affiliation(s)
- Erika Giuffrida
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Chiara Bianca Maria Platania
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95125 Catania, Italy
| | - Francesca Lazzara
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Federica Conti
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Ludovica Sotera
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95125 Catania, Italy
| | - Danushki Herath
- Faculty of Health, University Paris-Est Créteil, INSERM, IMRB, F-94010 Créteil, France
| | - Roberto Motterlini
- Faculty of Health, University Paris-Est Créteil, INSERM, IMRB, F-94010 Créteil, France
| | - Roberta Foresti
- Faculty of Health, University Paris-Est Créteil, INSERM, IMRB, F-94010 Créteil, France
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95125 Catania, Italy
- Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95125 Catania, Italy
| |
Collapse
|
8
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
9
|
Guarascio R, Cheetham ME. Light as a Mediator of Acute and Chronic Retina Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:247-251. [PMID: 39930204 DOI: 10.1007/978-3-031-76550-6_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
In age-related macula dystrophy (AMD) and some forms of inherited retinal dystrophies (IRDs), blindness is caused by the loss of photoreceptors and retinal pigment epithelium (RPE) cells. This process can be exacerbated by genetic and environmental risk factors, including exposure of the retina to bright light. Several light damage models have been developed and have proved to be powerful tools to study retinal degeneration. These models have enabled the investigation of common mechanisms of cell death and inflammation, as well as the identification of therapeutic targets and the assessment of potential new therapies against retinal degeneration. Here, we discuss the principal mechanisms of light-induced toxicity and highlight how this has been used in the development of therapeutic approaches to treat AMD and IRDs.
Collapse
|
10
|
Tao L, He D, Chen Y, Yang K, He B, Cai P, Cai B, Liao C, Liu Z, Li S, Chen J, Wu Y. Transferrin ameliorates retinal degeneration by mediating the dimerization of all-trans-retinal. J Biol Chem 2025; 301:108054. [PMID: 39653244 PMCID: PMC11742617 DOI: 10.1016/j.jbc.2024.108054] [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: 06/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024] Open
Abstract
High levels of all-trans-retinal (atRAL) in the retina is considered to be responsible for the development of autosomal recessive Stargardt's disease (STGD1) and dry age-related macular degeneration (dAMD). Two bisretinoids, all-trans-retinal dimer (atRAL-dimer) and N-retinyl-N-retinylidene ethanolamine (A2E), form from the dimerization of atRAL in the retina but they possess much lower toxicity and phototoxicity toward retinal pigment epithelium (RPE) cells than atRAL. Here, we introduced a novel function of transferrin (TRF) in mediating the conversion of atRAL into atRAL-dimer and A2E, which effectively protected the retina from damage by atRAL and prevented retinal function decline in mice, and rescued atRAL-loaded RPE cells. Moreover, TRF-mediated conversion of atRAL to atRAL-dimer and A2E required the help of bicarbonate ions (HCO3-). atRAL had the capacity to stimulate the expression of TRF in RPE and photoreceptor cells as well as RPE/choroid and neural retina of mice, reflecting that the elevation of TRF levels by atRAL is most likely to help defy level increase and cytotoxicity of atRAL through facilitating its dimerization and thereby serves as a mechanism of retinal self-protection. Our findings offer a promising avenue for the treatment of retinopathies characterized by disrupted clearance of atRAL.
Collapse
Affiliation(s)
- Lei Tao
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Danxue He
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Yuling Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Beiting He
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Peixin Cai
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China
| | - Binxiang Cai
- 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, China
| | - Chunyan Liao
- 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, China
| | - Zuguo Liu
- 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, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; 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, China; Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China.
| |
Collapse
|
11
|
Ryan EM, Norinskiy MA, Bracken AK, Lueders EE, Chen X, Fu Q, Anderson ET, Zhang S, Abbasov ME. Activity-Based Acylome Profiling with N-(Cyanomethyl)- N-(phenylsulfonyl)amides for Targeted Lysine Acylation and Post-Translational Control of Protein Function in Cells. J Am Chem Soc 2024; 146:27622-27643. [PMID: 39348182 PMCID: PMC11899832 DOI: 10.1021/jacs.4c09073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Lysine acylations are ubiquitous and structurally diverse post-translational modifications that vastly expand the functional heterogeneity of the human proteome. Hence, the targeted acylation of lysine residues has emerged as a strategic approach to exert biomimetic control over the protein function. However, existing strategies for targeted lysine acylation in cells often rely on genetic intervention, recruitment of endogenous acylation machinery, or nonspecific acylating agents and lack methods to quantify the magnitude of specific acylations on a global level. In this study, we develop activity-based acylome profiling (ABAP), a chemoproteomic strategy that exploits elaborate N-(cyanomethyl)-N-(phenylsulfonyl)amides and lysine-centric probes for site-specific introduction and proteome-wide mapping of posttranslational lysine acylations in human cells. Harnessing this framework, we quantify various artificial acylations and rediscover numerous endogenous lysine acylations. We validate site-specific acetylation of target lysines and establish a structure-activity relationship for N-(cyanomethyl)-N-(phenylsulfonyl)amides in proteins from diverse structural and functional classes. We identify paralog-selective chemical probes that acetylate conserved lysines within interferon-stimulated antiviral RNA-binding proteins, generating de novo proteoforms with obstructed RNA interactions. We further demonstrate that targeted acetylation of a key enzyme in retinoid metabolism engenders a proteoform with a conformational change in the protein structure, leading to a gain-of-function phenotype and reduced drug potency. These findings underscore the versatility of our strategy in biomimetic control over protein function through targeted delivery and global profiling of endogenous and artificial lysine acylations, potentially advancing therapeutic modalities and our understanding of biological processes orchestrated by these post-translational modifications.
Collapse
Affiliation(s)
- Elizabeth M Ryan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Michael A Norinskiy
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Amy K Bracken
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Emma E Lueders
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Xueer Chen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Qin Fu
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, New York 14853, United States
| | - Elizabeth T Anderson
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, New York 14853, United States
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, New York 14853, United States
| | - Mikail E Abbasov
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
12
|
Wang Y, Li T, Yu S, Gong Y, Zhang M, Wu Y, Liu W, Sun J, Chen J, Sun X. The central retinal thickness and its related genotype in ABCA4-related retinopathy. Eye (Lond) 2024; 38:2718-2723. [PMID: 38740961 PMCID: PMC11427577 DOI: 10.1038/s41433-024-03104-2] [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: 10/10/2023] [Revised: 03/14/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
PURPOSE To further explore the influence of genotype, including mutation type and structural domain, on the severity of macular atrophy, we measured the central retinal thickness (CRT) in patients with ABCA4-related retinopathy. METHODS A total of 66 patients were included in the cohort. This was a retrospective investigation. The patients were tested using whole exon sequencing and ophthalmic exams, including slip lamp exams, best-corrected visual acuity, optical coherence tomography, fundus photo, and fundus autofluorescence. RESULTS In this study, we discovered that mutations on nucleotide binding domains (NBD) lead to less CRT (45.00 ± 25.25μm, 95% CI: 31.54-58.46) had significantly less CRT than the others (89.75 ± 71.17μm, 95% CI: 30.25-149.25, p = 0.032), and could accelerate the rate of CRT decrease. CONCLUSIONS Our study provides new perspectives in the understanding of ABCA4-related retinopathy.
Collapse
Affiliation(s)
- Yimin Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tong Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Suqin Yu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yuanyuan Gong
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Min Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yidong Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Wenjia Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Jieqiong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Eye Disease, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| |
Collapse
|
13
|
Yang B, Yang K, Chen J, Wu Y. Crocin Protects the 661W Murine Photoreceptor Cell Line against the Toxic Effects of All- Trans-Retinal. Int J Mol Sci 2024; 25:10124. [PMID: 39337609 PMCID: PMC11432120 DOI: 10.3390/ijms251810124] [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: 07/30/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Age-related macular degeneration (AMD) is a common disease contributing to vision loss in the elderly. All-trans-retinal (atRAL) is a retinoid in the retina, and its abnormal accumulation exhibits toxicity to the retina and promotes oxidative stress-induced photoreceptor degeneration, which plays a crucial role in AMD progression. Crocin is a natural product extracted from saffron, which displays significant antioxidant and anti-inflammatory effects. The present study elucidates the protective effects of crocin on photoreceptor cell damage by atRAL and its potential mechanisms. The results revealed that crocin significantly attenuated cytotoxicity by repressing oxidative stress, mitochondrial injury, and DNA damage in atRAL-loaded photoreceptor cells. Moreover, crocin visibly inhibited DNA damage-induced apoptosis and gasdermin E (GSDME)-mediated pyroptosis in photoreceptor cells after exposure to atRAL. It was also observed that crocin distinctly prevented an increase in Fe2+ levels and lipid peroxidation caused by atRAL via suppressing the Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor-erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) signaling pathway, thereby ameliorating photoreceptor cell ferroptosis. In short, these findings provide new insights that crocin mitigates atRAL-induced toxicity to photoreceptor cells by inhibiting oxidative stress, apoptosis, pyroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Bo Yang
- 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 361102, China
| | - Kunhuan Yang
- 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 361102, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Yalin Wu
- 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 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| |
Collapse
|
14
|
Pazur E, Kalatanova A, Tasker NR, Vainionpää K, Leinonen H, Wipf P. Synthesis and Biological Analysis of Iso-dimethyltryptamines in a Model of Light-Induced Retinal Degeneration. ACS Med Chem Lett 2024; 15:1049-1056. [PMID: 39015263 PMCID: PMC11247652 DOI: 10.1021/acsmedchemlett.4c00130] [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: 03/21/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Iso-dimethyltryptamine (isoDMT) analogues with heterocyclic substitutions at the indole C(3) were prepared in a hydrogen autotransfer alkylation and tested in combination with natural and unnatural clavine alkaloids in a model of light-induced retinal degeneration for protection against retinal degeneration. On the basis of measurements with optical coherence tomography and electroretinography, three compounds showed better efficacy than the positive control bromocriptine at equivalent systemically administered doses. These studies provide further insights into the role of serotonin receptors and their potential therapeutic applications in ocular diseases.
Collapse
Affiliation(s)
- Ethan
J. Pazur
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Anna Kalatanova
- Yliopistonrinne
3, Canthia, School of Pharmacy, University
of Eastern Finland, 70211 Kuopio, Finland
| | - Nikhil R. Tasker
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Katri Vainionpää
- Yliopistonrinne
3, Canthia, School of Pharmacy, University
of Eastern Finland, 70211 Kuopio, Finland
| | - Henri Leinonen
- Yliopistonrinne
3, Canthia, School of Pharmacy, University
of Eastern Finland, 70211 Kuopio, Finland
| | - Peter Wipf
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Yliopistonrinne
3, Canthia, School of Pharmacy, University
of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
15
|
Chen C, Adler L, Milliken C, Rahman B, Kono M, Perry LP, Gonzalez-Fernandez F, Koutalos Y. The First Steps of the Visual Cycle in Human Rod and Cone Photoreceptors. Invest Ophthalmol Vis Sci 2024; 65:9. [PMID: 38958967 PMCID: PMC11223620 DOI: 10.1167/iovs.65.8.9] [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: 02/23/2024] [Accepted: 06/09/2024] [Indexed: 07/04/2024] Open
Abstract
Purpose Light detection destroys the visual pigment. Its regeneration, necessary for the recovery of light sensitivity, is accomplished through the visual cycle. Release of all-trans retinal by the light-activated visual pigment and its reduction to all-trans retinol comprise the first steps of the visual cycle. In this study, we determined the kinetics of all-trans retinol formation in human rod and cone photoreceptors. Methods Single living rod and cone photoreceptors were isolated from the retinas of human cadaver eyes (ages 21 to 90 years). Formation of all-trans retinol was measured by imaging its outer segment fluorescence (excitation, 360 nm; emission, >420 nm). The extent of conversion of released all-trans retinal to all-trans retinol was determined by measuring the fluorescence excited by 340 and 380 nm. Measurements were repeated with photoreceptors isolated from Macaca fascicularis retinas. Experiments were carried out at 37°C. Results We found that ∼80% to 90% of all-trans retinal released by the light-activated pigment is converted to all-trans retinol, with a rate constant of 0.24 to 0.55 min-1 in human rods and ∼1.8 min-1 in human cones. In M. fascicularis rods and cones, the rate constants were 0.38 ± 0.08 min-1 and 4.0 ± 1.1 min-1, respectively. These kinetics are several times faster than those measured in other vertebrates. Interphotoreceptor retinoid-binding protein facilitated the removal of all-trans retinol from human rods. Conclusions The first steps of the visual cycle in human photoreceptors are several times faster than in other vertebrates and in line with the rapid recovery of light sensitivity exhibited by the human visual system.
Collapse
Affiliation(s)
- Chunhe Chen
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Leopold Adler
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Cole Milliken
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bushra Rahman
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Masahiro Kono
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Lynn Poole Perry
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Federico Gonzalez-Fernandez
- Departments of Ophthalmology and Pathology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi, United States
| | - Yiannis Koutalos
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
16
|
Yang Y, Lin Y, Han Z, Wang B, Zheng W, Wei L. Ferroptosis: a novel mechanism of cell death in ophthalmic conditions. Front Immunol 2024; 15:1440309. [PMID: 38994366 PMCID: PMC11236620 DOI: 10.3389/fimmu.2024.1440309] [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: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Ferroptosis, a new type of programmed cell death proposed in recent years, is characterized mainly by reactive oxygen species and iron-mediated lipid peroxidation and differs from programmed cell death, such as apoptosis, necrosis, and autophagy. Ferroptosis is associated with a variety of physiological and pathophysiological processes. Recent studies have shown that ferroptosis can aggravate or reduce the occurrence and development of diseases by targeting metabolic pathways and signaling pathways in tumors, ischemic organ damage, and other degenerative diseases related to lipid peroxidation. Increasing evidence suggests that ferroptosis is closely linked to the onset and progression of various ophthalmic conditions, including corneal injury, glaucoma, age-related macular degeneration, diabetic retinopathy, retinal detachment, and retinoblastoma. Our review of the current research on ferroptosis in ophthalmic diseases reveals significant advancements in our understanding of the pathogenesis, aetiology, and treatment of these conditions.
Collapse
Affiliation(s)
- Yaqi Yang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yumeng Lin
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- Naniing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Bo Wang
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Wei Zheng
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Lijuan Wei
- Ophthalmology Department, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
17
|
Wei D, Qu C, Zhao N, Li S, Pu N, Song Z, Tao Y. The significance of precisely regulating heme oxygenase-1 expression: Another avenue for treating age-related ocular disease? Ageing Res Rev 2024; 97:102308. [PMID: 38615894 DOI: 10.1016/j.arr.2024.102308] [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: 01/20/2024] [Revised: 03/23/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Aging entails the deterioration of the body's organs, including overall damages at both the genetic and cellular levels. The prevalence of age-related ocular disease such as macular degeneration, dry eye diseases, glaucoma and cataracts is increasing as the world's population ages, imposing a considerable economic burden on individuals and society. The development of age-related ocular disease is predominantly triggered by oxidative stress and chronic inflammatory reaction. Heme oxygenase-1 (HO-1) is a crucial antioxidant that mediates the degradative process of endogenous iron protoporphyrin heme. It catalyzes the rate-limiting step of the heme degradation reaction, and releases the metabolites such as carbon monoxide (CO), ferrous, and biliverdin (BV). The potent scavenging activity of these metabolites can help to defend against peroxides, peroxynitrite, hydroxyl, and superoxide radicals. Other than directly decomposing endogenous oxidizing substances (hemoglobin), HO-1 is also a critical regulator of inflammatory cells and tissue damage, exerting its anti-inflammation activity through regulating complex inflammatory networks. Therefore, promoting HO-1 expression may act as a promising therapeutic strategy for the age-related ocular disease. However, emerging evidences suggest that the overexpression of HO-1 significantly contributes to ferroptosis due to its dual nature. Surplus HO-1 leads to excessive Fe2+ and reactive oxygen species, thereby causing lipid peroxidation and ferroptosis. In this review, we elucidate the role of HO-1 in countering age-related disease, and summarize recent pharmacological trials that targeting HO-1 for disease management. Further refinements of the knowledge would position HO-1 as a novel therapeutic target for age-related ocular disease.
Collapse
Affiliation(s)
- Dong Wei
- Department of ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China; College of Medicine, Zhengzhou University, China
| | - Chengkang Qu
- Department of ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Na Zhao
- College of Medicine, Zhengzhou University, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, China
| | - Ning Pu
- Department of ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China; College of Medicine, Zhengzhou University, China
| | - Zongming Song
- Department of ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China.
| | - Ye Tao
- Department of ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
18
|
Li D, Chang J, Wang Y, Du X, Xu J, Cui J, Zhang T, Chen Y. Hyperoside mitigates photoreceptor degeneration in part by targeting cGAS and suppressing DNA-induced microglial activation. Acta Neuropathol Commun 2024; 12:76. [PMID: 38755736 PMCID: PMC11097432 DOI: 10.1186/s40478-024-01793-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
Activated microglia play an important role in driving photoreceptor degeneration-associated neuroinflammation in the retina. Controlling pro-inflammatory activation of microglia holds promise for mitigating the progression of photoreceptor degeneration. Our previous study has demonstrated that pre-light damage treatment of hyperoside, a naturally occurring flavonol glycoside with antioxidant and anti-inflammatory activities, prevents photooxidative stress-induced photoreceptor degeneration and neuroinflammatory responses in the retina. However, the direct impact of hyperoside on microglia-mediated neuroinflammation during photoreceptor degeneration remains unknown. Upon verifying the anti-inflammatory effects of hyperoside in LPS-stimulated BV-2 cells, our results here further demonstrated that post-light damage hyperoside treatment mitigated the loss of photoreceptors and attenuated the functional decline of the retina. Meanwhile, post-light damage hyperoside treatment lowered neuroinflammatory responses and dampened microglial activation in the illuminated retinas. With respect to microglial activation, hyperoside mitigated the pro-inflammatory responses in DNA-stimulated BV-2 cells and lowered DNA-stimulated production of 2'3'-cGAMP in BV-2 cells. Moreover, hyperoside was shown to directly interact with cGAS and suppress the enzymatic activity of cGAS in a cell-free system. In conclusion, the current study suggests for the first time that the DNA sensor cGAS is a direct target of hyperoside. Hyperoside is effective at mitigating DNA-stimulated cGAS-mediated pro-inflammatory activation of microglia, which likely contributes to the therapeutic effects of hyperoside at curtailing neuroinflammation and alleviating neuroinflammation-instigated photoreceptor degeneration.
Collapse
Affiliation(s)
- Daijin Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jie Chang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaoye Du
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jing Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jingang Cui
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China.
- Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
19
|
Rozanowska M, Edge R, Land EJ, Navaratnam S, Sarna T, Truscott TG. Scavenging of Cation Radicals of the Visual Cycle Retinoids by Lutein, Zeaxanthin, Taurine, and Melanin. Int J Mol Sci 2023; 25:506. [PMID: 38203675 PMCID: PMC10779001 DOI: 10.3390/ijms25010506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
In the retina, retinoids involved in vision are under constant threat of oxidation, and their oxidation products exhibit deleterious properties. Using pulse radiolysis, this study determined that the bimolecular rate constants of scavenging cation radicals of retinoids by taurine are smaller than 2 × 107 M-1s-1 whereas lutein scavenges cation radicals of all three retinoids with the bimolecular rate constants approach the diffusion-controlled limits, while zeaxanthin is only 1.4-1.6-fold less effective. Despite that lutein exhibits greater scavenging rate constants of retinoid cation radicals than other antioxidants, the greater concentrations of ascorbate in the retina suggest that ascorbate may be the main protectant of all visual cycle retinoids from oxidative degradation, while α-tocopherol may play a substantial role in the protection of retinaldehyde but is relatively inefficient in the protection of retinol or retinyl palmitate. While the protection of retinoids by lutein and zeaxanthin appears inefficient in the retinal periphery, it can be quite substantial in the macula. Although the determined rate constants of scavenging the cation radicals of retinol and retinaldehyde by dopa-melanin are relatively small, the high concentration of melanin in the RPE melanosomes suggests they can be scavenged if they are in proximity to melanin-containing pigment granules.
Collapse
Affiliation(s)
- Malgorzata Rozanowska
- Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Cardiff CF10 3AX, UK
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Ruth Edge
- Dalton Cumbrian Facility, The University of Manchester, Westlakes Science Park, Moor Row, Cumbria CA24 3HA, UK;
| | - Edward J. Land
- The Paterson Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK;
| | - Suppiah Navaratnam
- Biomedical Sciences Research Institute, University of Salford, Manchester M5 4WT, UK;
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - T. George Truscott
- School of Chemical and Physical Sciences, Lennard-Jones Building, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
20
|
Różanowska MB. Lipofuscin, Its Origin, Properties, and Contribution to Retinal Fluorescence as a Potential Biomarker of Oxidative Damage to the Retina. Antioxidants (Basel) 2023; 12:2111. [PMID: 38136230 PMCID: PMC10740933 DOI: 10.3390/antiox12122111] [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: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Lipofuscin accumulates with age as intracellular fluorescent granules originating from incomplete lysosomal digestion of phagocytosed and autophagocytosed material. The purpose of this review is to provide an update on the current understanding of the role of oxidative stress and/or lysosomal dysfunction in lipofuscin accumulation and its consequences, particularly for retinal pigment epithelium (RPE). Next, the fluorescence of lipofuscin, spectral changes induced by oxidation, and its contribution to retinal fluorescence are discussed. This is followed by reviewing recent developments in fluorescence imaging of the retina and the current evidence on the prognostic value of retinal fluorescence for the progression of age-related macular degeneration (AMD), the major blinding disease affecting elderly people in developed countries. The evidence of lipofuscin oxidation in vivo and the evidence of increased oxidative damage in AMD retina ex vivo lead to the conclusion that imaging of spectral characteristics of lipofuscin fluorescence may serve as a useful biomarker of oxidative damage, which can be helpful in assessing the efficacy of potential antioxidant therapies in retinal degenerations associated with accumulation of lipofuscin and increased oxidative stress. Finally, amendments to currently used fluorescence imaging instruments are suggested to be more sensitive and specific for imaging spectral characteristics of lipofuscin fluorescence.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| |
Collapse
|
21
|
Engfer ZJ, Lewandowski D, Dong Z, Palczewska G, Zhang J, Kordecka K, Płaczkiewicz J, Panas D, Foik AT, Tabaka M, Palczewski K. Distinct mouse models of Stargardt disease display differences in pharmacological targeting of ceramides and inflammatory responses. Proc Natl Acad Sci U S A 2023; 120:e2314698120. [PMID: 38064509 PMCID: PMC10723050 DOI: 10.1073/pnas.2314698120] [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: 08/24/2023] [Accepted: 10/25/2023] [Indexed: 12/17/2023] Open
Abstract
Mutations in many visual cycle enzymes in photoreceptors and retinal pigment epithelium (RPE) cells can lead to the chronic accumulation of toxic retinoid byproducts, which poison photoreceptors and the underlying RPE if left unchecked. Without a functional ATP-binding cassette, sub-family A, member 4 (ABCA4), there is an elevation of all-trans-retinal and prolonged buildup of all-trans-retinal adducts, resulting in a retinal degenerative disease known as Stargardt-1 disease. Even in this monogenic disorder, there is significant heterogeneity in the time to onset of symptoms among patients. Using a combination of molecular techniques, we studied Abca4 knockout (simulating human noncoding disease variants) and Abca4 knock-in mice (simulating human misfolded, catalytically inactive protein variants), which serve as models for Stargardt-1 disease. We compared the two strains to ascertain whether they exhibit differential responses to agents that affect cytokine signaling and/or ceramide metabolism, as alterations in either of these pathways can exacerbate retinal degenerative phenotypes. We found different degrees of responsiveness to maraviroc, a known immunomodulatory CCR5 antagonist, and to the ceramide-lowering agent AdipoRon, an agonist of the ADIPOR1 and ADIPOR2 receptors. The two strains also display different degrees of transcriptional deviation from matched WT controls. Our phenotypic comparison of the two distinct Abca4 mutant-mouse models sheds light on potential therapeutic avenues previously unexplored in the treatment of Stargardt disease and provides a surrogate assay for assessing the effectiveness for genome editing.
Collapse
Affiliation(s)
- Zachary J. Engfer
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
- Department of Physiology and Biophysics, University of California, Irvine, CA92697
| | - Dominik Lewandowski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
| | - Zhiqian Dong
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
| | - Grazyna Palczewska
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
| | - Jianye Zhang
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
| | - Katarzyna Kordecka
- Ophthalmic Biology Group, International Centre for Translational Eye Research, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw01-224, Poland
| | - Jagoda Płaczkiewicz
- Ophthalmic Biology Group, International Centre for Translational Eye Research, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw01-224, Poland
| | - Damian Panas
- International Centre for Translational Eye Research, Warsaw01-224, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw01-224, Poland
| | - Andrzej T. Foik
- Ophthalmic Biology Group, International Centre for Translational Eye Research, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw01-224, Poland
| | - Marcin Tabaka
- International Centre for Translational Eye Research, Warsaw01-224, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw01-224, Poland
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA92697
- Department of Physiology and Biophysics, University of California, Irvine, CA92697
- Department of Chemistry, University of California, Irvine, CA92697
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA92697
| |
Collapse
|
22
|
Tao T, Xu N, Li J, Zhao M, Li X, Huang L. Conditional loss of Ube3d in the retinal pigment epithelium accelerates age-associated alterations in the retina of mice. J Pathol 2023; 261:442-454. [PMID: 37772657 DOI: 10.1002/path.6201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 07/07/2023] [Accepted: 08/11/2023] [Indexed: 09/30/2023]
Abstract
Several studies have suggested a correlation between the ubiquitin-proteasome system (UPS) and age-related macular degeneration (AMD), with its phenotypic severity ranging from mild visual impairment to blindness, but the mechanism for UPS dysfunction contributing to disease progression is unclear. In this study, we investigated the role of ubiquitin protein ligase E3D (UBE3D) in aging and degeneration in mouse retina. Conditional knockout of Ube3d in the retinal pigment epithelium (RPE) of mice led to progressive and irregular fundus lesions, attenuation of the retinal vascular system, and age-associated deterioration of rod and cone responses. Simultaneously, RPE-specific Ube3d knockout mice also presented morphological changes similar to the histopathological characteristics of human AMD, in which a defective UPS led to RPE abnormalities such as phagocytosis or degradation of metabolites, the interaction with photoreceptor outer segment, and the transport of nutrients or waste products with choroidal capillaries via Bruch's membrane. Moreover, conditional loss of Ube3d resulted in aberrant molecular characterizations associated with the autophagy-lysosomal pathway, oxidative stress damage, and cell-cycle regulation, which are implicated in AMD pathology. Thus, our findings strengthen and expand the impact of UPS dysfunction on retinal pathophysiology during aging, indicating that genetic Ube3d deficiency in the RPE could lead to the abnormal formation of pigment deposits and secondary fundus alterations. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tianchang Tao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Ningda Xu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
| | - Jiarui Li
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
| | - Xiaoxin Li
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
- Department of Ophthalmology, Xiamen Eye Center of Xiamen University, Xiamen, PR China
| | - Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing, PR China
- Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, PR China
- College of Optometry, Peking University Health Science Center, Beijing, PR China
| |
Collapse
|
23
|
Liénard MA. Sensory evolution: A dazzling hack to cope with bright light in owls and whales. Curr Biol 2023; 33:R1200-R1202. [PMID: 37989100 DOI: 10.1016/j.cub.2023.10.006] [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: 11/23/2023]
Abstract
When exposed to sudden changes in light intensity, rod-dominated retinas of animals with highly sensitive dim-vision risk critical damage. A new study finds that owls and deep-diving whales have evolved an identical photoprotection mechanism to delay toxic all-trans retinal release, a discovery with potential medical implications.
Collapse
Affiliation(s)
- Marjorie A Liénard
- Department of Biology, Lund University, Sölvegatan 35B, 22362 Lund, Sweden.
| |
Collapse
|
24
|
Castiglione GM, Chiu YLI, Gutierrez EDA, Van Nynatten A, Hauser FE, Preston M, Bhattacharyya N, Schott RK, Chang BSW. Convergent evolution of dim light vision in owls and deep-diving whales. Curr Biol 2023; 33:4733-4740.e4. [PMID: 37776863 DOI: 10.1016/j.cub.2023.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/21/2023] [Accepted: 09/05/2023] [Indexed: 10/02/2023]
Abstract
Animals with enhanced dim-light sensitivity are at higher risk of light-induced retinal degeneration when exposed to bright light conditions.1,2,3,4 This trade-off is mediated by the rod photoreceptor sensory protein, rhodopsin (RHO), and its toxic vitamin A chromophore by-product, all-trans retinal.5,6,7,8 Rod arrestin (Arr-1) binds to RHO and promotes sequestration of excess all-trans retinal,9,10 which has recently been suggested as a protective mechanism against photoreceptor cell death.2,11 We investigated Arr-1 evolution in animals at high risk of retinal damage due to periodic bright-light exposure of rod-dominated retinas. Here, we find the convergent evolution of enhanced Arr-1/RHO all-trans-retinal sequestration in owls and deep-diving whales. Statistical analyses reveal a parallel acceleration of Arr-1 evolutionary rates in these lineages, which is associated with the introduction of a rare Arr-1 mutation (Q69R) into the RHO-Arr-1 binding interface. Using in vitro assays, we find that this single mutation significantly enhances RHO-all-trans-retinal sequestration by ∼30%. This functional convergence across 300 million years of evolutionary divergence suggests that Arr-1 and RHO may play an underappreciated role in the photoprotection of the eye, with potentially vast clinical significance.
Collapse
Affiliation(s)
- Gianni M Castiglione
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA; Department of Ophthalmology & Visual Sciences, Vanderbilt University, Nashville, TN 37232, USA; Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; Evolutionary Studies, Vanderbilt University, Nashville, TN 37235, USA.
| | - Yan L I Chiu
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Eduardo de A Gutierrez
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Alexander Van Nynatten
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - Frances E Hauser
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - Matthew Preston
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Nihar Bhattacharyya
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Institute of Ophthalmology, University College London, London EC1V 2PD, UK
| | - Ryan K Schott
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; Department of Biology and Centre for Vision Research, York University, Toronto, ON M3J 1P3, Canada; Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - Belinda S W Chang
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, ON M5S 3B2, Canada.
| |
Collapse
|
25
|
Matsuyama M, Ortega JT, Fedorov Y, Scott-McKean J, Muller-Greven J, Buck M, Adams D, Jastrzebska B, Greenlee W, Matsuyama S. Development of novel cytoprotective small compounds inhibiting mitochondria-dependent cell death. iScience 2023; 26:107916. [PMID: 37841588 PMCID: PMC10568349 DOI: 10.1016/j.isci.2023.107916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/27/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
We identified cytoprotective small molecules (CSMs) by a cell-based high-throughput screening of Bax inhibitors. Through a medicinal chemistry program, M109S was developed, which is orally bioactive and penetrates the blood-brain/retina barriers. M109S protected retinal cells in ocular disease mouse models. M109S directly interacted with Bax and inhibited the conformational change and mitochondrial translocation of Bax. M109S inhibited ABT-737-induced apoptosis both in Bax-only and Bak-only mouse embryonic fibroblasts. M109S also inhibited apoptosis induced by staurosporine, etoposide, and obatoclax. M109S decreased maximal mitochondrial oxygen consumption rate and reactive oxygen species production, whereas it increased glycolysis. These effects on cellular metabolism may contribute to the cytoprotective activity of M109S. M109S is a novel small molecule protecting cells from mitochondria-dependent apoptosis both in vitro and in vivo. M109S has the potential to become a research tool for studying cell death mechanisms and to develop therapeutics targeting mitochondria-dependent cell death pathway.
Collapse
Affiliation(s)
- Mieko Matsuyama
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Joseph T. Ortega
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yuri Fedorov
- Department of Genetics and Genome Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jonah Scott-McKean
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Macromolecular Science and Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeannie Muller-Greven
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Matthias Buck
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Drew Adams
- Department of Genetics and Genome Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Shigemi Matsuyama
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
26
|
Plau J, Morgan CE, Fedorov Y, Banerjee S, Adams DJ, Blaner WS, Yu EW, Golczak M. Discovery of Nonretinoid Inhibitors of CRBP1: Structural and Dynamic Insights for Ligand-Binding Mechanisms. ACS Chem Biol 2023; 18:2309-2323. [PMID: 37713257 PMCID: PMC10591915 DOI: 10.1021/acschembio.3c00402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023]
Abstract
The dysregulation of retinoid metabolism has been linked to prevalent ocular diseases including age-related macular degeneration and Stargardt disease. Modulating retinoid metabolism through pharmacological approaches holds promise for the treatment of these eye diseases. Cellular retinol-binding protein 1 (CRBP1) is the primary transporter of all-trans-retinol (atROL) in the eye, and its inhibition has recently been shown to protect mouse retinas from light-induced retinal damage. In this report, we employed high-throughput screening to identify new chemical scaffolds for competitive, nonretinoid inhibitors of CRBP1. To understand the mechanisms of interaction between CRBP1 and these inhibitors, we solved high-resolution X-ray crystal structures of the protein in complex with six selected compounds. By combining protein crystallography with hydrogen/deuterium exchange mass spectrometry, we quantified the conformational changes in CRBP1 caused by different inhibitors and correlated their magnitude with apparent binding affinities. Furthermore, using molecular dynamic simulations, we provided evidence for the functional significance of the "closed" conformation of CRBP1 in retaining ligands within the binding pocket. Collectively, our study outlines the molecular foundations for understanding the mechanism of high-affinity interactions between small molecules and CRBPs, offering a framework for the rational design of improved inhibitors for this class of lipid-binding proteins.
Collapse
Affiliation(s)
- Jacqueline Plau
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Christopher E. Morgan
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department
of Chemistry, Thiel College, Greenville, Pennsylvania 16125, United States
| | - Yuriy Fedorov
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Surajit Banerjee
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14850, United States
- Northeastern
Collaborative Access Team, Argonne National
Laboratory, Argonne, Illinois 60439, United States
| | - Drew J. Adams
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - William S. Blaner
- Department
of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, United States
| | - Edward W. Yu
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Marcin Golczak
- Department
of Pharmacology, Small Molecule Drug Development Core Facility, Department of Genetics, and Cleveland Center
for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
27
|
Choi SW, Seo S, Hong HK, Yoon SJ, Kim M, Moon S, Lee JY, Lim J, Lee JB, Woo SJ. Therapeutic Extracellular Vesicles from Tonsil-Derived Mesenchymal Stem Cells for the Treatment of Retinal Degenerative Disease. Tissue Eng Regen Med 2023; 20:951-964. [PMID: 37440108 PMCID: PMC10519919 DOI: 10.1007/s13770-023-00555-8] [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: 04/11/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Retinal degenerative disease (RDD), one of the most common causes of blindness, is predominantly caused by the gradual death of retinal pigment epithelial cells (RPEs) and photoreceptors due to various causes. Cell-based therapies, such as stem cell implantation, have been developed for the treatment of RDD, but potential risks, including teratogenicity and immune reactions, have hampered their clinical application. Stem cell-derived extracellular vesicles (EVs) have recently emerged as a cell-free alternative therapeutic strategy; however, additional invasiveness and low yield of the stem cell extraction process is problematic. METHODS To overcome these limitations, we developed therapeutic EVs for the treatment of RDD which were extracted from tonsil-derived mesenchymal stem cells obtained from human tonsil tissue discarded as medical waste following tonsillectomy (T-MSC EVs). To verify the biocompatibility and cytoprotective effect of T-MSC EVs, we measured cell viability by co-culture with human RPE without or with toxic all-trans-retinal. To elucidate the cytoprotective mechanism of T-MSC EVs, we performed transcriptome sequencing using RNA extracted from RPEs. The in vivo protective effect of T-MSC EVs was evaluated using Pde6b gene knockout rats as an animal model of retinitis pigmentosa. RESULTS T-MSC EVs showed high biocompatibility and the human pigment epithelial cells were significantly protected in the presence of T-MSC EVs from the toxic effect of all-trans-retinal. In addition, T-MSC EVs showed a dose-dependent cell death-delaying effect in real-time quantification of cell death. Transcriptome sequencing analysis revealed that the efficient ability of T-MSC EVs to regulate intracellular oxidative stress may be one of the reasons explaining their excellent cytoprotective effect. Additionally, intravitreally injected T-MSC EVs had an inhibitory effect on the destruction of the outer nuclear layer in the Pde6b gene knockout rat. CONCLUSIONS Together, the results of this study indicate the preventive and therapeutic effects of T-MSC EVs during the initiation and development of retinal degeneration, which may be a beneficial alternative for the treatment of RDD.
Collapse
Affiliation(s)
- Seung Woo Choi
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea
| | - Sooin Seo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea
| | - Hye Kyoung Hong
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea
| | - So Jung Yoon
- Bundang CHA Biobank, CHA University College of Medicine, CHA University Bundang Medical Center, Seongnam, 13496, Korea
| | - Minah Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea
| | - Sunghyun Moon
- Department of Chemical Engineering, University of Seoul, 163 Seoul Siripdaero, Dongdaemun-Gu, Seoul, 02504, Korea
| | - Joo Yong Lee
- Department of Ophthalmology, Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | - Jaeseung Lim
- Cellatoz Therapeutics Lnc, Seongnam, 13487, Korea
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, 163 Seoul Siripdaero, Dongdaemun-Gu, Seoul, 02504, Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea.
| |
Collapse
|
28
|
Wang L, Shah SM, Mangwani-Mordani S, Gregori NZ. Updates on Emerging Interventions for Autosomal Recessive ABCA4-Associated Stargardt Disease. J Clin Med 2023; 12:6229. [PMID: 37834872 PMCID: PMC10573680 DOI: 10.3390/jcm12196229] [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: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Autosomal recessive Stargardt disease (STGD1) is an inherited retinal degenerative disease associated with a mutated ATP-binding cassette, subfamily A, member 4 (ABCA4) gene. STGD1 is the most common form of juvenile macular degeneration with onset in late childhood to early or middle adulthood and causes progressive, irreversible visual impairment and blindness. No effective treatment is currently available. In the present article, we review the most recent updates in clinical trials targeting the management of STGD1, including gene therapy, small molecule therapy, and stem cell therapy. In gene therapy, dual adeno-associated virus and non-viral vectors have been successful in delivering the human ABCA4 gene in preclinical studies. For pharmaceutical therapies ALK-001, deuterated vitamin A shows promise with preliminary data for phase 2 trial, demonstrating a decreased atrophy growth rate after two years. Stem cell therapy using human pluripotent stem cell-derived retinal pigment epithelium cells demonstrated long-term safety three years after implantation and visual acuity improvements in the first two years after initiation of therapy. Many other treatment options have ongoing investigations and clinical trials. While multiple potential interventions have shown promise in attenuating disease progression, further exploration is necessary to demonstrate treatment safety and efficacy.
Collapse
Affiliation(s)
- Liang Wang
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (L.W.); (S.M.S.)
| | - Serena M. Shah
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (L.W.); (S.M.S.)
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Simran Mangwani-Mordani
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Ninel Z. Gregori
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Miami Veterans Administration Medical Center, Miami, FL 33125, USA
| |
Collapse
|
29
|
Chang J, Wang Y, Xu J, Du X, Cui J, Zhang T, Chen Y. Ginsenoside Re Mitigates Photooxidative Stress-Mediated Photoreceptor Degeneration and Retinal Inflammation. J Neuroimmune Pharmacol 2023; 18:397-412. [PMID: 37326907 PMCID: PMC10577105 DOI: 10.1007/s11481-023-10073-y] [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: 03/03/2023] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
Loss of photoreceptors is the central pathology accountable for irreversible vision impairment in patients with photoreceptor degenerative disorders. Currently, mechanisms-based pharmacological therapies protecting photoreceptors from degenerative progression remain clinically unavailable. Photooxidative stress plays a pivotal role in initiating the degenerative cascade in photoreceptors. Meanwhile, photoreceptor degeneration interacts closely with neurotoxic inflammatory responses primarily mediated by aberrantly activated microglia in the retina. Thus, therapies with anti-oxidant and anti-inflammatory properties have been actively investigated for their pharmacological value in controlling photoreceptor degeneration. In the current study, we examined the pharmacological potentials of ginsenoside Re (Re), a naturally occurring antioxidant with anti-inflammatory activities, in photooxidative stress-mediated photoreceptor degeneration. Our results demonstrate that Re attenuates photooxidative stress and associated lipid peroxidation in the retina. Furthermore, Re treatment preserves the morphological and functional integrity of the retina, counteracts photooxidative stress-induced perturbation of the retinal gene expression profiles and mitigates photoreceptor degeneration-associated neuroinflammatory responses and microglia activation in the retina. Lastly, Re partially antagonizes the deleterious effects of photooxidative stress on müller cells, verifying its beneficial impact on retina homeostasis. In conclusion, the work here provides experimental evidence supporting novel pharmacological implications of Re in attenuating photooxidative stress-mediated photoreceptor degeneration and ensuing neuroinflammation.
Collapse
Affiliation(s)
- Jie Chang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jing Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaoye Du
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jingang Cui
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, China.
- Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
30
|
Choi EH, Suh S, Sears AE, Hołubowicz R, Kedhar SR, Browne AW, Palczewski K. Genome editing in the treatment of ocular diseases. Exp Mol Med 2023; 55:1678-1690. [PMID: 37524870 PMCID: PMC10474087 DOI: 10.1038/s12276-023-01057-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/14/2023] [Indexed: 08/02/2023] Open
Abstract
Genome-editing technologies have ushered in a new era in gene therapy, providing novel therapeutic strategies for a wide range of diseases, including both genetic and nongenetic ocular diseases. These technologies offer new hope for patients suffering from previously untreatable conditions. The unique anatomical and physiological features of the eye, including its immune-privileged status, size, and compartmentalized structure, provide an optimal environment for the application of these cutting-edge technologies. Moreover, the development of various delivery methods has facilitated the efficient and targeted administration of genome engineering tools designed to correct specific ocular tissues. Additionally, advancements in noninvasive ocular imaging techniques and electroretinography have enabled real-time monitoring of therapeutic efficacy and safety. Herein, we discuss the discovery and development of genome-editing technologies, their application to ocular diseases from the anterior segment to the posterior segment, current limitations encountered in translating these technologies into clinical practice, and ongoing research endeavors aimed at overcoming these challenges.
Collapse
Affiliation(s)
- Elliot H Choi
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Susie Suh
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Avery E Sears
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Rafał Hołubowicz
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Sanjay R Kedhar
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Andrew W Browne
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA.
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA.
- Department of Chemistry, University of California, Irvine, CA, USA.
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
31
|
Dobreva A, Camacho ET, Miranda M. Mathematical model for glutathione dynamics in the retina. Sci Rep 2023; 13:10996. [PMID: 37419948 PMCID: PMC10328985 DOI: 10.1038/s41598-023-37938-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/29/2023] [Indexed: 07/09/2023] Open
Abstract
The retina is highly susceptible to the generation of toxic reactive oxygen species (ROS) that disrupt the normal operations of retinal cells. The glutathione (GSH) antioxidant system plays an important role in mitigating ROS. To perform its protective functions, GSH depends on nicotinamide adenine dinucleotide phosphate (NADPH) produced through the pentose phosphate pathway. This work develops the first mathematical model for the GSH antioxidant system in the outer retina, capturing the most essential components for formation of ROS, GSH production, its oxidation in detoxifying ROS, and subsequent reduction by NADPH. We calibrate and validate the model using experimental measurements, at different postnatal days up to PN28, from control mice and from the rd1 mouse model for the disease retinitis pigmentosa (RP). Global sensitivity analysis is then applied to examine the model behavior and identify the pathways with the greatest impact in control compared to RP conditions. The findings underscore the importance of GSH and NADPH production in dealing with oxidative stress during retinal development, especially after peak rod degeneration occurs in RP, leading to increased oxygen tension. This suggests that stimulation of GSH and NADPH synthesis could be a potential intervention strategy in degenerative mouse retinas with RP.
Collapse
Affiliation(s)
- Atanaska Dobreva
- Department of Mathematics, Augusta University, Augusta, GA, 30912, USA.
| | - Erika Tatiana Camacho
- University of Texas at San Antonio, San Antonio, TX, 78249, USA
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - María Miranda
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115, Valencia, Spain
| |
Collapse
|
32
|
Moghadam Fard A, Mirshahi R, Naseripour M, Ghasemi Falavarjani K. Stem Cell Therapy in Stargardt Disease: A Systematic Review. J Ophthalmic Vis Res 2023; 18:318-327. [PMID: 37600916 PMCID: PMC10432931 DOI: 10.18502/jovr.v18i3.13780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/26/2023] [Indexed: 08/22/2023] Open
Abstract
This article aimed to review current literature on the safety and efficacy of stem cell therapy in Stargardt disease. A comprehensive literature search was performed, and two animal and eleven human clinical trials were retrieved. These studies utilized different kinds of stem cells, including human or mouse embryonic stem cells, mesenchymal stem cells, bone marrow mononuclear fraction, and autologous bone marrow-derived stem cells. In addition, different injection techniques including subretinal, intravitreal, and suprachoroidal space injections have been evaluated. Although stem cell therapy holds promise in improving visual function in patients with Stargardt disease, further investigation is needed to determine the long-term benefits, safety, and efficacy in determining the best delivery method and selecting the most appropriate stem cell type.
Collapse
Affiliation(s)
- Atousa Moghadam Fard
- Eye Research Center, The Five Senses Health Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Mirshahi
- Eye Research Center, The Five Senses Health Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masood Naseripour
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
33
|
Bassetto M, Zaluski J, Li B, Zhang J, Badiee M, Kiser PD, Tochtrop GP. Tuning the Metabolic Stability of Visual Cycle Modulators through Modification of an RPE65 Recognition Motif. J Med Chem 2023; 66:8140-8158. [PMID: 37279401 PMCID: PMC10824489 DOI: 10.1021/acs.jmedchem.3c00461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In the eye, the isomerization of all-trans-retinal to 11-cis-retinal is accomplished by a metabolic pathway termed the visual cycle that is critical for vision. RPE65 is the essential trans-cis isomerase of this pathway. Emixustat, a retinoid-mimetic RPE65 inhibitor, was developed as a therapeutic visual cycle modulator and used for the treatment of retinopathies. However, pharmacokinetic liabilities limit its further development including: (1) metabolic deamination of the γ-amino-α-aryl alcohol, which mediates targeted RPE65 inhibition, and (2) unwanted long-lasting RPE65 inhibition. We sought to address these issues by more broadly defining the structure-activity relationships of the RPE65 recognition motif via the synthesis of a family of novel derivatives, which were tested in vitro and in vivo for RPE65 inhibition. We identified a potent secondary amine derivative with resistance to deamination and preserved RPE65 inhibitory activity. Our data provide insights into activity-preserving modifications of the emixustat molecule that can be employed to tune its pharmacological properties.
Collapse
Affiliation(s)
- Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Research Service, VA Long Beach Healthcare System, Long Beach, California 90822, United States
| | - Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Bowen Li
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Jianye Zhang
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
| | - Mohsen Badiee
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, California 92697, United States
- Research Service, VA Long Beach Healthcare System, Long Beach, California 90822, United States
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
34
|
Lyu Y, Tschulakow AV, Wang K, Brash DE, Schraermeyer U. Chemiexcitation and melanin in photoreceptor disc turnover and prevention of macular degeneration. Proc Natl Acad Sci U S A 2023; 120:e2216935120. [PMID: 37155898 PMCID: PMC10194005 DOI: 10.1073/pnas.2216935120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Age-related macular degeneration, Stargardt disease, and their Abca4-/- mouse model are characterized by accelerated accumulation of the pigment lipofuscin, derived from photoreceptor disc turnover in the retinal pigment epithelium (RPE); lipofuscin accumulation and retinal degeneration both occur earlier in albino mice. Intravitreal injection of superoxide (O2•-) generators reverses lipofuscin accumulation and rescues retinal pathology, but neither the target nor mechanism is known. Here we show that RPE contains thin multi-lamellar membranes (TLMs) resembling photoreceptor discs, which associate with melanolipofuscin granules in pigmented mice but in albinos are 10-fold more abundant and reside in vacuoles. Genetically over-expressing tyrosinase in albinos generates melanosomes and decreases TLM-related lipofuscin. Intravitreal injection of generators of O2•- or nitric oxide (•NO) decreases TLM-related lipofuscin in melanolipofuscin granules of pigmented mice by ~50% in 2 d, but not in albinos. Prompted by evidence that O2•- plus •NO creates a dioxetane on melanin that excites its electrons to a high-energy state (termed "chemiexcitation"), we show that exciting electrons directly using a synthetic dioxetane reverses TLM-related lipofuscin even in albinos; quenching the excited-electron energy blocks this reversal. Melanin chemiexcitation assists in safe photoreceptor disc turnover.
Collapse
Affiliation(s)
- Yanan Lyu
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, University of Tuebingen, Tuebingen72076, Germany
| | - Alexander V. Tschulakow
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, University of Tuebingen, Tuebingen72076, Germany
- OcuTox GmbH, Preclinical Drug Assessment, Hechingen72379, Germany
| | - Kun Wang
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, University of Tuebingen, Tuebingen72076, Germany
| | - Douglas E. Brash
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT06520-8040
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT06520-8028
| | - Ulrich Schraermeyer
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, University of Tuebingen, Tuebingen72076, Germany
- OcuTox GmbH, Preclinical Drug Assessment, Hechingen72379, Germany
| |
Collapse
|
35
|
He D, Tao L, Cai B, Chen X, Wang Y, Li S, Liao C, Chen Y, Chen J, Liu Z, Wu Y. eIF2α incites photoreceptor cell and retina damage by all-trans-retinal. J Biol Chem 2023; 299:104686. [PMID: 37031820 DOI: 10.1016/j.jbc.2023.104686] [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: 11/09/2022] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/11/2023] Open
Abstract
Dry age-related macular degeneration (AMD) and recessive Stargardt's disease (STGD1) lead to irreversible blindness in humans. The accumulation of all-trans-retinal (atRAL) induced by chaos in visual cycle is closely associated with retinal atrophy in dry AMD and STGD1, but its critical downstream signaling molecules remain ambiguous. Here, we reported that activation of eukaryotic translation initiation factor 2α (eIF2α) by atRAL promoted retinal degeneration and photoreceptor loss through activating c-Jun N-terminal kinase (JNK) signaling-dependent apoptosis and gasdermin E (GSDME)-mediated pyroptosis. We determined that eIF2α activation by atRAL in photoreceptor cells resulted from endoplasmic reticulum (ER) homeostasis disruption caused at least in part by reactive oxygen species (ROS) production, and it activated JNK signaling independent of and dependent on activating transcription factor 4 (ATF4) and the ATF4/transcription factor C/EBP homologous protein (CHOP) axis. CHOP overexpression induced apoptosis of atRAL-loaded photoreceptor cells through activating JNK signaling rather than inhibiting the expression of anti-apoptotic gene Bcl2. JNK activation by eIF2α facilitated photoreceptor cell apoptosis caused by atRAL via caspase-3 activation and DNA damage. Additionally, we demonstrated that eIF2α was activated in neural retina of light-exposed Abca4-/-Rdh8-/- mice, a model that shows severe defects in atRAL clearance and displays primary features of human dry AMD and STGD1. Of note, inhibition of eIF2α activation by salubrinal effectively ameliorated retinal degeneration and photoreceptor apoptosis in Abca4-/-Rdh8-/- mice upon light exposure. The results of this study suggest that eIF2α is an important target to develop drug therapies for the treatment of dry AMD and STGD1.
Collapse
Affiliation(s)
- Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Tao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Binxiang Cai
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiangjun Chen
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Hangzhou, China
| | - Yan Wang
- Department of Ophthalmology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Shiying Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuling Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Shenzhen Research Institute of Xiamen University, Shenzhen, China; Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
36
|
Palczewska G, Wojtkowski M, Palczewski K. From mouse to human: Accessing the biochemistry of vision in vivo by two-photon excitation. Prog Retin Eye Res 2023; 93:101170. [PMID: 36787681 PMCID: PMC10463242 DOI: 10.1016/j.preteyeres.2023.101170] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The eye is an ideal organ for imaging by a multi-photon excitation approach, because ocular tissues such as the sclera, cornea, lens and neurosensory retina, are highly transparent to infrared (IR) light. The interface between the retina and the retinal pigment epithelium (RPE) is especially informative, because it reflects the health of the visual (retinoid) cycle and its changes in response to external stress, genetic manipulations, and drug treatments. Vitamin A-derived retinoids, like retinyl esters, are natural fluorophores that respond to multi-photon excitation with near IR light, bypassing the filter-like properties of the cornea, lens, and macular pigments. Also, during natural aging some retinoids form bisretinoids, like diretinoid-pyridiniumethanolamine (A2E), that are highly fluorescent. These bisretinoids appear to be elevated concurrently with aging. Vitamin A-derived retinoids and bisretinoidss are detected by two-photon ophthalmoscopy (2PO), using a new class of light sources with adjustable spatial, temporal, and spectral properties. Furthermore, the two-photon (2P) absorption of IR light by the visual pigments in rod and cone photoreceptors can initiate visual transduction by cis-trans isomerization of retinal, enabling parallel functional studies. Recently we overcame concerns about safety, data interpretation and complexity of the 2P-based instrumentation, the major roadblocks toward advancing this modality to the clinic. These imaging and retina-function assessment advancements have enabled us to conduct the first 2P studies with humans.
Collapse
Affiliation(s)
- Grazyna Palczewska
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA; International Center for Translational Eye Research, Polish Academy of Sciences, Warsaw, Poland; Polgenix, Inc., Department of Medical Devices, Cleveland, OH, USA; Department of Physical Chemistry of Biological Systems, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland.
| | - Maciej Wojtkowski
- International Center for Translational Eye Research, Polish Academy of Sciences, Warsaw, Poland; Department of Physical Chemistry of Biological Systems, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland; Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland.
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, CA, USA; Department of Physiology & Biophysics, School of Medicine, And Chemistry, Molecular Biology and Biochemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
37
|
Ortega JT, Parmar T, Jastrzebska B. Galanin receptor 3 - A new pharmacological target in retina degeneration. Pharmacol Res 2023; 188:106675. [PMID: 36693600 PMCID: PMC9918719 DOI: 10.1016/j.phrs.2023.106675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
The neuropeptide galanin receptor 3 (GALR3) is a class A G protein-coupled receptor (GPCR) broadly expressed in the nervous system, including the retina. GALR3 is involved in the modulation of immune and inflammatory responses. Tight control of these processes is critical for maintaining homeostasis in the retina and is required to sustain vision. Here, we investigated the role of GALR3 in retina pathologies triggered by bright light and P23H mutation in the rhodopsin (RHO) gene, associated with the activation of oxidative stress and inflammatory responses. We used a multiphase approach involving pharmacological inhibition of GALR3 with its antagonist SNAP-37889 and genetic depletion of GALR3 to modulate the GALR3 signaling. Our in vitro experiments in the retinal pigment epithelium-derived cells (ARPE19) susceptible to all-trans-retinal toxicity indicated that GALR3 could be involved in the cellular stress response to this phototoxic product. Indeed, blocking the GALR3 signaling in Abca4-/-/Rdh8-/- and wild-type Balb/cJ mice, sensitive to bright light-induced retina damage, protected retina health in these mice exposed to light. The retina morphology and function were substantially improved, and stress response processes were reduced in these mouse models compared to the controls. Furthermore, in P23H Rho knock-in mice, a model of retinitis pigmentosa (RP), both pharmacological inhibition and genetic ablation of GALR3 prolonged the survival of photoreceptors. These results indicate that GALR3 signaling contributes to acute light-induced and chronic RP-linked retinopathies. Together, this work provides the pharmacological knowledge base to evaluate GALR3 as a potential target for developing novel therapies to combat retinal degeneration.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Tanu Parmar
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA.
| |
Collapse
|
38
|
Ferroptosis: mechanisms and advances in ocular diseases. Mol Cell Biochem 2023:10.1007/s11010-022-04644-5. [PMID: 36617346 DOI: 10.1007/s11010-022-04644-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023]
Abstract
As an essential trace element in the body, iron is critical for the maintenance of organismal metabolism. Excessive iron facilitates reactive oxygen species generation and inflicts damage on cells and tissues. Ferroptosis, a newly identified iron-dependent type of programmed cell death, has been implicated in a broad set of metabolic disorders. Ferroptosis is mainly characterized by excess iron accumulation, elevated lipid peroxides and reactive oxygen species, and reduced levels of glutathione and glutathione peroxidase 4. The vast emerging literature on ferroptosis has shown that numerous diseases, such as cancers, neurodegeneration, and autoimmune diseases, are associated with ferroptosis. Meanwhile, recent studies have confirmed the relationship between ferroptosis and eye diseases including keratopathy, cataract, glaucoma, retinal ischemia-reperfusion injury, age-related macular degeneration, retinitis pigmentosa, diabetic retinopathy, and retinoblastoma, indicating the critical role of ferroptosis in ocular diseases. In this article, we introduce the primary signaling pathways of ferroptosis and review current advances in research on ocular diseases involving iron overload and ferroptosis. Furthermore, several unanswered questions in the area are raised. Addressing these unanswered questions promises to provide new insights into preventing, controlling, and treating not only ocular diseases but also a variety of other diseases in the near future.
Collapse
|
39
|
Chen C, Yang K, He D, Yang B, Tao L, Chen J, Wu Y. Induction of ferroptosis by HO-1 contributes to retinal degeneration in mice with defective clearance of all-trans-retinal. Free Radic Biol Med 2023; 194:245-254. [PMID: 36509314 DOI: 10.1016/j.freeradbiomed.2022.12.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The accumulation of all-trans-retinal (atRAL) in photoreceptors and the retinal pigment epithelium (RPE), which is induced by chaos in visual (retinoid) cycle, is closely associated with the pathogenesis of dry age-related macular degeneration (AMD) and autosomal recessive Stargardt's disease (STGD1). Although we have reported that the induction of ferroptosis by atRAL is an important cause of photoreceptor loss, but its mechanisms still remain unclear. In this study, we identified heme oxygenase-1 (HO-1) as an inducer of photoreceptor ferroptosis elicited by atRAL. In atRAL-loaded photoreceptor cells, inhibition of Kelch-like ECH-associated protein 1 (KEAP1) at least in part by reactive oxygen species (ROS) production evoked the release of nuclear factor-erythroid 2-related factor-2 (NRF2) from KEAP1, followed by the translocation of active NRF2 into the nucleus where it promoted the transcription of the Ho-1 gene, thereby leading to HO-1 overexpression in the cytosol. A significant elevation of Fe2+ levels in photoreceptor cells resulted from activation of HO-1 by atRAL, and it facilitated ROS overproduction and then triggered ferroptotic cell death through ROS-mediated lipid peroxidation. Both treatment with HO-1 repressor Zinc protoporphyrin IX (ZnPP) and knockout of Ho-1 gene clearly rescued photoreceptor cells against ferroptosis arising from atRAL overload. Light-exposed Abca4-/-Rdh8-/- mice rapidly display severe defects in atRAL clearance, and serve as an acute model of dry AMD and STGD1. HO-1 activation was distinctly observed in neural retina of Abca4-/-Rdh8-/- mice after exposure to light, and it was visibly relieved by intraperitoneally injected ferroptosis inhibitor ferrostatin-1. More notably, intraperitoneal administration of ZnPP effectively alleviated both photoreceptor degeneration and RPE atrophy in Abca4-/-Rdh8-/- mice in response to light exposure by repressing HO-1-mediated ferroptosis. Our study suggests that HO-1 is a key factor that regulates atRAL-induced ferroptosis in photoreceptors and the RPE, and its inhibition may hold promises for the therapy of dry AMD and STGD1.
Collapse
Affiliation(s)
- Chao Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Kunhuan Yang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Bo Yang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Lei Tao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518063, Guangdong, China; Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361001, Fujian, China.
| |
Collapse
|
40
|
Ren X, Léveillard T. Modulating antioxidant systems as a therapeutic approach to retinal degeneration. Redox Biol 2022; 57:102510. [PMID: 36274523 PMCID: PMC9596747 DOI: 10.1016/j.redox.2022.102510] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/21/2022] Open
Abstract
The human retina is facing a big challenge of reactive oxygen species (ROS) from endogenous and exogenous sources. Excessive ROS can cause damage to DNA, lipids, and proteins, triggering abnormal redox signaling, and ultimately lead to cell death. Thus, oxidative stress has been observed in inherited retinal diseases as a common hallmark. To counteract the detrimental effect of ROS, cells are equipped with various antioxidant defenses. In this review, we will focus on the antioxidant systems in the retina and how they can protect retina from oxidative stress. Both small antioxidants and antioxidant enzymes play a role in ROS removal. Particularly, the thioredoxin and glutaredoxin systems, as the major antioxidant systems in mammalian cells, exert functions in redox signaling regulation via modifying cysteines in proteins. In addition, the thioredoxin-like rod-derived cone viability factor (RdCVFL) and thioredoxin interacting protein (TXNIP) can modulate metabolism in photoreceptors and promote their survival. In conclusion, elevating the antioxidant capacity in retina is a promising therapy to curb the progress of inherited retinal degeneration.
Collapse
Affiliation(s)
- Xiaoyuan Ren
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden.
| | - Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
41
|
The novel visual cycle inhibitor (±)-RPE65-61 protects retinal photoreceptors from light-induced degeneration. PLoS One 2022; 17:e0269437. [PMID: 36227868 PMCID: PMC9560169 DOI: 10.1371/journal.pone.0269437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
The visual cycle refers to a series of biochemical reactions of retinoids in ocular tissues and supports the vision in vertebrates. The visual cycle regenerates visual pigments chromophore, 11-cis-retinal, and eliminates its toxic byproducts from the retina, supporting visual function and retinal neuron survival. Unfortunately, during the visual cycle, when 11-cis-retinal is being regenerated in the retina, toxic byproducts, such as all-trans-retinal and bis-retinoid is N-retinylidene-N-retinylethanolamine (A2E), are produced, which are proposed to contribute to the pathogenesis of the dry form of age-related macular degeneration (AMD). The primary biochemical defect in Stargardt disease (STGD1) is the accelerated synthesis of cytotoxic lipofuscin bisretinoids, such as A2E, in the retinal pigment epithelium (RPE) due to mutations in the ABCA4 gene. To prevent all-trans-retinal-and bisretinoid-mediated retinal degeneration, slowing down the retinoid flow by modulating the visual cycle with a small molecule has been proposed as a therapeutic strategy. The present study describes RPE65-61, a novel, non-retinoid compound, as an inhibitor of RPE65 (a key enzyme in the visual cycle), intended to modulate the excessive activity of the visual cycle to protect the retina from harm degenerative diseases. Our data demonstrated that (±)-RPE65-61 selectively inhibited retinoid isomerase activity of RPE65, with an IC50 of 80 nM. Furthermore, (±)-RPE65-61 inhibited RPE65 via an uncompetitive mechanism. Systemic administration of (±)-RPE65-61 in mice resulted in slower chromophore regeneration after light bleach, confirming in vivo target engagement and visual cycle modulation. Concomitant protection of the mouse retina from high-intensity light damage was also observed. Furthermore, RPE65-61 down-regulated the cyclic GMP-AMP synthase stimulator of interferon genes (cGAS-STING) pathway, decreased the inflammatory factor, and attenuated retinal apoptosis caused by light-induced retinal damage (LIRD), which led to the preservation of the retinal function. Taken together, (±)-RPE65-61 is a potent visual cycle modulator that may provide a neuroprotective therapeutic benefit for patients with STGD and AMD.
Collapse
|
42
|
Das A, Imanishi Y. Drug Discovery Strategies for Inherited Retinal Degenerations. BIOLOGY 2022; 11:1338. [PMID: 36138817 PMCID: PMC9495580 DOI: 10.3390/biology11091338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Inherited retinal degeneration is a group of blinding disorders afflicting more than 1 in 4000 worldwide. These disorders frequently cause the death of photoreceptor cells or retinal ganglion cells. In a subset of these disorders, photoreceptor cell death is a secondary consequence of retinal pigment epithelial cell dysfunction or degeneration. This manuscript reviews current efforts in identifying targets and developing small molecule-based therapies for these devastating neuronal degenerations, for which no cures exist. Photoreceptors and retinal ganglion cells are metabolically demanding owing to their unique structures and functional properties. Modulations of metabolic pathways, which are disrupted in most inherited retinal degenerations, serve as promising therapeutic strategies. In monogenic disorders, great insights were previously obtained regarding targets associated with the defective pathways, including phototransduction, visual cycle, and mitophagy. In addition to these target-based drug discoveries, we will discuss how phenotypic screening can be harnessed to discover beneficial molecules without prior knowledge of their mechanisms of action. Because of major anatomical and biological differences, it has frequently been challenging to model human inherited retinal degeneration conditions using small animals such as rodents. Recent advances in stem cell-based techniques are opening new avenues to obtain pure populations of human retinal ganglion cells and retinal organoids with photoreceptor cells. We will discuss concurrent ideas of utilizing stem-cell-based disease models for drug discovery and preclinical development.
Collapse
Affiliation(s)
- Arupratan Das
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yoshikazu Imanishi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
43
|
Kikuchi Y, Sugano E, Yuki S, Tabata K, Endo Y, Takita Y, Onoguchi R, Ozaki T, Fukuda T, Takai Y, Kurose T, Tanaka K, Honma Y, Perez E, Stock M, Fernández JR, Tamura M, Voronkov M, Stock JB, Tomita H. SIG-1451, a Novel, Non-Steroidal Anti-Inflammatory Compound, Attenuates Light-Induced Photoreceptor Degeneration by Affecting the Inflammatory Process. Int J Mol Sci 2022; 23:ijms23158802. [PMID: 35955937 PMCID: PMC9369167 DOI: 10.3390/ijms23158802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Age-related macular degeneration is a progressive retinal disease that is associated with factors such as oxidative stress and inflammation. In this study, we evaluated the protective effects of SIG-1451, a non-steroidal anti-inflammatory compound developed for treating atopic dermatitis and known to inhibit Toll-like receptor 4, in light-induced photoreceptor degeneration. SIG-1451 was intraperitoneally injected into rats once per day before exposure to 1000 lx light for 24 h; one day later, optical coherence tomography showed a decrease in retinal thickness, and electroretinogram (ERG) amplitude was also found to have decreased 3 d after light exposure. Moreover, SIG-1451 partially protected against this decrease in retinal thickness and increase in ERG amplitude. One day after light exposure, upregulation of inflammatory response-related genes was observed, and SIG-1451 was found to inhibit this upregulation. Iba-1, a microglial marker, was suppressed in SIG-1451-injected rats. To investigate the molecular mechanism underlying these effects, we used lipopolysaccharide (LPS)-stimulated rat immortalised Müller cells. The upregulation of C-C motif chemokine 2 by LPS stimulation was significantly inhibited by SIG-1451 treatment, and Western blot analysis revealed a decrease in phosphorylated I-κB levels. These results indicate that SIG-1451 indirectly protects photoreceptor cells by attenuating light damage progression, by affecting the inflammatory responses.
Collapse
Affiliation(s)
- Yuki Kikuchi
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Shiori Yuki
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Kitako Tabata
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Yuka Endo
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Yuya Takita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Reina Onoguchi
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Taku Ozaki
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Tomokazu Fukuda
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
| | - Yoshihiro Takai
- Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa 619-0216, Kyoto, Japan
| | - Takahiro Kurose
- Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa 619-0216, Kyoto, Japan
| | - Koichi Tanaka
- Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa 619-0216, Kyoto, Japan
| | - Yoichi Honma
- Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa 619-0216, Kyoto, Japan
| | - Eduardo Perez
- Signum Biosciences, 4999 Pearl East Circle, Boulder, CO 80301, USA
| | - Maxwell Stock
- Signum Biosciences, 4999 Pearl East Circle, Boulder, CO 80301, USA
| | | | - Masanori Tamura
- Signum Biosciences, 4999 Pearl East Circle, Boulder, CO 80301, USA
| | - Michael Voronkov
- Signum Biosciences, 4999 Pearl East Circle, Boulder, CO 80301, USA
| | - Jeffry B. Stock
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka 020-8551, Iwate, Japan
- Correspondence: ; Tel.: +81-19-621-6427
| |
Collapse
|
44
|
Różanowska MB, Czuba-Pełech B, Różanowski B. Is There an Optimal Combination of AREDS2 Antioxidants Zeaxanthin, Vitamin E and Vitamin C on Light-Induced Toxicity of Vitamin A Aldehyde to the Retina? Antioxidants (Basel) 2022; 11:antiox11061132. [PMID: 35740030 PMCID: PMC9220409 DOI: 10.3390/antiox11061132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/15/2022] [Accepted: 06/06/2022] [Indexed: 02/05/2023] Open
Abstract
Vitamins C and E and zeaxanthin are components of a supplement tested in a large clinical trial-Age-Related Eye Disease Study 2 (AREDS2)-and it has been demonstrated that they can inhibit the progression of age-related macular degeneration. The aim of this study was to determine the optimal combinations of these antioxidants to prevent the phototoxicity mediated by vitamin A aldehyde (ATR), which can accumulate in photoreceptor outer segments (POS) upon exposure to light. We used cultured retinal pigment epithelial cells ARPE-19 and liposomes containing unsaturated lipids and ATR as a model of POS. Cells and/or liposomes were enriched with lipophilic antioxidants, whereas ascorbate was added just before the exposure to light. Supplementing the cells and/or liposomes with single lipophilic antioxidants had only a minor effect on phototoxicity, but the protection substantially increased in the presence of both ways of supplementation. Combinations of zeaxanthin with α-tocopherol in liposomes and cells provided substantial protection, enhancing cell viability from ~26% in the absence of antioxidants to ~63% in the presence of 4 µM zeaxanthin and 80 µM α-tocopherol, and this protective effect was further increased to ~69% in the presence of 0.5 mM ascorbate. The protective effect of ascorbate disappeared at a concentration of 1 mM, whereas 2 mM of ascorbate exacerbated the phototoxicity. Zeaxanthin or α-tocopherol partly ameliorated the cytotoxic effects. Altogether, our results suggest that the optimal combination includes upper levels of zeaxanthin and α-tocopherol achievable by diet and/or supplementations, whereas ascorbate needs to be at a four-fold smaller concentration than that in the vitreous. The physiological relevance of the results is discussed.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK
- Cardiff Institute for Tissue Engineering and Repair (CITER), Cardiff University, Cardiff CF24 4HQ, Wales, UK
- Correspondence: ; Tel.: +44-292087-5057
| | - Barbara Czuba-Pełech
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland;
| | | |
Collapse
|
45
|
Kazmierczak de Camargo JP, Prezia GNDB, Shiokawa N, Sato MT, Rosati R, Beate Winter Boldt A. New Insights on the Regulatory Gene Network Disturbed in Central Areolar Choroidal Dystrophy-Beyond Classical Gene Candidates. Front Genet 2022; 13:886461. [PMID: 35656327 PMCID: PMC9152281 DOI: 10.3389/fgene.2022.886461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Central areolar choroidal dystrophy (CACD) is a rare hereditary disease that mainly affects the macula, resulting in progressive and usually profound visual loss. Being part of congenital retinal dystrophies, it may have an autosomal dominant or recessive inheritance and, until now, has no effective treatment. Given the shortage of genotypic information about the disease, this work systematically reviews the literature for CACD-causing genes. Three independent researchers selected 33 articles after carefully searching and filtering the Scielo, Pubmed, Lilacs, Web of Science, Scopus, and Embase databases. Mutations of six genes (PRPH2, GUCA1A, GUCY2D, CDHR1, ABCA4, and TTLL5) are implicated in the monogenic dominant inheritance of CACD. They are functionally related to photoreceptors (either in the phototransduction process, as in the case of GUCY2D, or the recovery of retinal photodegradation in photoreceptors for GUCA1A, or the formation and maintenance of specific structures within photoreceptors for PRPH2). The identified genetic variants do not explain all observed clinical features, calling for further whole-genome and functional studies for this disease. A network analysis with the CACD-related genes identified in the systematic review resulted in the identification of another 20 genes that may influence CACD onset and symptoms. Furthermore, an enrichment analysis allowed the identification of 13 transcription factors and 4 long noncoding RNAs interacting with the products of the previously mentioned genes. If mutated or dysregulated, they may be directly involved in CACD development and related disorders. More than half of the genes identified by bioinformatic tools do not appear in commercial gene panels, calling for more studies about their role in the maintenance of the retina and phototransduction process, as well as for a timely update of these gene panels.
Collapse
Affiliation(s)
| | - Giovanna Nazaré de Barros Prezia
- Post-Graduation Program in Biotechnology Applied to Child and Adolescent Health, Faculdades Pequeno Príncipe and Pelé Pequeno Príncipe Research Institute, Curitiba, Brazil
| | - Naoye Shiokawa
- Retina and Vitreo Consulting Eye Clinic, Curitiba, Brazil
| | - Mario Teruo Sato
- Retina and Vitreo Consulting Eye Clinic, Curitiba, Brazil.,Department of Ophthalmol/Otorhinolaryngology, Federal University of Paraná, Curitiba, Brazil
| | - Roberto Rosati
- Post-Graduation Program in Biotechnology Applied to Child and Adolescent Health, Faculdades Pequeno Príncipe and Pelé Pequeno Príncipe Research Institute, Curitiba, Brazil
| | | |
Collapse
|
46
|
Li M, Xu J, Wang Y, Du X, Zhang T, Chen Y. Astragaloside A Protects Against Photoreceptor Degeneration in Part Through Suppressing Oxidative Stress and DNA Damage-Induced Necroptosis and Inflammation in the Retina. J Inflamm Res 2022; 15:2995-3020. [PMID: 35645574 PMCID: PMC9130102 DOI: 10.2147/jir.s362401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Photoreceptors are specialized retinal neurons responsible for phototransduction. Loss of photoreceptors directly leads to irreversible vision impairment. Pharmacological therapies protecting against photoreceptor degeneration are clinically lacking. Oxidative stress and inflammation are common mechanisms playing important roles in the pathogenesis of photoreceptor degeneration. Astragaloside A (AS-A) is a naturally occurring antioxidant and anti-inflammatory agent with neuroprotective activities. However, the photoreceptor protective effects of AS-A remain unknown. The current study thus aims to illustrate the pharmacological potentials of AS-A in protecting against photoreceptor degeneration. Methods BALB/c and C57/BL6J mice were exposed to bright light and DNA alkylating agent methyl methanesulfonate (MMS) to develop oxidative stress and DNA damage-mediated photoreceptor degeneration, respectively. Microstructural, morphological and functional assessments were performed to directly evaluate the photoreceptor protective effects of AS-A. Ultrastructural and molecular changes in the retina were examined to better understand the pharmacological mechanisms of AS-A in protecting against photoreceptor degeneration. Results AS-A protected against bright light-induced photoreceptor impairment. Bright light-induced retinal oxidative stress and photoreceptor cell death were attenuated by AS-A treatment. AS-A treatment mitigated bright light-induced DNA damage, activation of poly (ADP-ribose) polymerase (PARP) and nuclear dislocation of high mobility group box 1 (HMGB1) in photoreceptors. AS-A broadly counteracted bright light-altered retinal gene expression profiles. In particular, AS-A decreased the retinal expression of genes involved in necroptosis and inflammatory responses. Bright light-induced microglial activation was also suppressed as a result of AS-A treatment. Furthermore, AS-A attenuated MMS-induced photoreceptor morphological impairment, elevated expression of pro-necroptotic and proinflammatory genes as well as microglial activation in the retina. Conclusion The work here demonstrates for the first time that AS-A protects against photoreceptor degeneration in part through mitigating oxidative stress and DNA damage-induced necroptosis and inflammatory responses in the retina.
Collapse
Affiliation(s)
- Mei Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China
| | - Jing Xu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China.,Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, People's Republic of China
| | - Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China
| | - Xiaoye Du
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China.,Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, People's Republic of China
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China.,Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, People's Republic of China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China.,Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200437, People's Republic of China.,Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200427, People's Republic of China
| |
Collapse
|
47
|
Vincent M, Simon L, Brabet P, Legrand P, Dorandeu C, Him JLK, Durand T, Crauste C, Begu S. Formulation and Evaluation of SNEDDS Loaded with Original Lipophenol for the Oral Route to Prevent Dry AMD and Stragardt’s Disease. Pharmaceutics 2022; 14:pharmaceutics14051029. [PMID: 35631617 PMCID: PMC9147958 DOI: 10.3390/pharmaceutics14051029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022] Open
Abstract
Dry age-related macular degeneration (Dry AMD) and Stargardt’s disease (STGD1) are common eye diseases, characterized by oxidative and carbonyl stress (COS)-inducing photoreceptor degeneration and vision loss. Previous studies have demonstrated the protective effect of photoreceptors after the intravenous administration of a new lipophenol drug, phloroglucinol-isopropyl-DHA (IP-DHA). In this study, we developed an oral formulation of IP-DHA (BCS Class IV) relying on a self-nanoemulsifying drug delivery system (SNEDDS). SNEDDS, composed of Phosal® 53 MCT, Labrasol®, and Transcutol HP® at a ratio of 25/60/15 (w/w/w), led to a homogeneous nanoemulsion (NE) with a mean size of 53.5 ± 4.5 nm. The loading of IP-DHA in SNEDDS (SNEDDS-IP-DHA) was successful, with a percentage of IP-DHA of 99.7% in nanoemulsions. The in vivo study of the therapeutic potency of SNEDDS-IP-DHA after oral administration on mice demonstrated photoreceptor protection after the induction of retinal degeneration with acute light stress (73–80%) or chronic light stress (52–69%). Thus, SNEDDS formulation proved to increase the solubility of IP-DHA, improving its stability in intestinal media and allowing its passage through the intestinal barrier after oral force-fed administration, while maintaining its biological activity. Therefore, SNEDDS-IP-DHA is a promising future preventive treatment for dry AMD and STGD1.
Collapse
Affiliation(s)
- Maxime Vincent
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France; (M.V.); (L.S.); (P.L.); (C.D.)
| | - Laurianne Simon
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France; (M.V.); (L.S.); (P.L.); (C.D.)
| | - Philippe Brabet
- Institut des Neurosciences de Montpellier, INSERM U1051, 34000 Montpellier, France
- Correspondence: (P.B.); (C.C.); (S.B.)
| | - Philippe Legrand
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France; (M.V.); (L.S.); (P.L.); (C.D.)
| | - Christophe Dorandeu
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France; (M.V.); (L.S.); (P.L.); (C.D.)
| | - Josephine Lai Kee Him
- Centre de Biochimie Structurale, CNRS UMR 5048, INSERM U1054, 34000 Montpellier, France;
| | - Thierry Durand
- IBMM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France;
| | - Céline Crauste
- IBMM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France;
- Correspondence: (P.B.); (C.C.); (S.B.)
| | - Sylvie Begu
- ICGM, Univ Montpellier, CNRS, ENSCM, 34000 Montpellier, France; (M.V.); (L.S.); (P.L.); (C.D.)
- Correspondence: (P.B.); (C.C.); (S.B.)
| |
Collapse
|
48
|
Matsuyama A, Kalargyrou AA, Smith AJ, Ali RR, Pearson RA. A comprehensive atlas of Aggrecan, Versican, Neurocan and Phosphacan expression across time in wildtype retina and in retinal degeneration. Sci Rep 2022; 12:7282. [PMID: 35508614 PMCID: PMC9068689 DOI: 10.1038/s41598-022-11204-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/07/2022] [Indexed: 11/08/2022] Open
Abstract
As photoreceptor cells die during retinal degeneration, the surrounding microenvironment undergoes significant changes that are increasingly recognized to play a prominent role in determining the efficacy of therapeutic interventions. Chondroitin Sulphate Proteoglycans (CSPGs) are a major component of the extracellular matrix that have been shown to inhibit neuronal regrowth and regeneration in the brain and spinal cord, but comparatively little is known about their expression in retinal degeneration. Here we provide a comprehensive atlas of the expression patterns of four individual CSPGs in three models of inherited retinal degeneration and wildtype mice. In wildtype mice, Aggrecan presented a biphasic expression, while Neurocan and Phosphacan expression declined dramatically with time and Versican expression remained broadly constant. In degeneration, Aggrecan expression increased markedly in Aipl1-/- and Pde6brd1/rd1, while Versican showed regional increases in the periphery of Rho-/- mice. Conversely, Neurocan and Phosphacan broadly decrease with time in all models. Our data reveal significant heterogeneity in the expression of individual CSPGs. Moreover, there are striking differences in the expression patterns of specific CSPGs in the diseased retina, compared with those reported following injury elsewhere in the CNS. Better understanding of the distinct distributions of individual CSPGs will contribute to creating more permissive microenvironments for neuro-regeneration and repair.
Collapse
Affiliation(s)
- A Matsuyama
- Ocular Cell and Gene therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK.
- University College London Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan.
| | - A A Kalargyrou
- Ocular Cell and Gene therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
- University College London Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - A J Smith
- Ocular Cell and Gene therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
- University College London Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - R R Ali
- Ocular Cell and Gene therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
- University College London Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - R A Pearson
- Ocular Cell and Gene therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK.
- University College London Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
49
|
Vats A, Xi Y, Feng B, Clinger OD, St Leger AJ, Liu X, Ghosh A, Dermond CD, Lathrop KL, Tochtrop GP, Picaud S, Chen Y. Non-retinoid chaperones improve rhodopsin homeostasis in a mouse model of retinitis pigmentosa. JCI Insight 2022; 7:153717. [PMID: 35472194 PMCID: PMC9220944 DOI: 10.1172/jci.insight.153717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Rhodopsin-associated (RHO-associated) retinitis pigmentosa (RP) is a progressive retinal disease that currently has no cure. RHO protein misfolding leads to disturbed proteostasis and the death of rod photoreceptors, resulting in decreased vision. We previously identified nonretinoid chaperones of RHO, including YC-001 and F5257-0462, by small-molecule high-throughput screening. Here, we profile the chaperone activities of these molecules toward the cell-surface level of 27 RP-causing human RHO mutants in NIH3T3 cells. Furthermore, using retinal explant culture, we show that YC-001 improves retinal proteostasis by supporting RHO homeostasis in RhoP23H/+ mouse retinae, which results in thicker outer nuclear layers (ONL), indicating delayed photoreceptor degeneration. Interestingly, YC-001 ameliorated retinal immune responses and reduced the number of microglia/macrophages in the RhoP23H/+ retinal explants. Similarly, F5257-0462 also protects photoreceptors in RhoP23H/+ retinal explants. In vivo, intravitreal injection of YC-001 or F5257-0462 microparticles in PBS shows that F5257-0462 has a higher efficacy in preserving photoreceptor function and delaying photoreceptor death in RhoP23H/+ mice. Collectively, we provide proof of principle that nonretinoid chaperones are promising drug candidates in treating RHO-associated RP.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Archisha Ghosh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Chase D Dermond
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Kira L Lathrop
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, United States of America
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
50
|
Cai B, Liao C, He D, Chen J, Han J, Lu J, Qin K, Liang W, Wu X, Liu Z, Wu Y. Gasdermin E mediates photoreceptor damage by all-trans-retinal in the mouse retina. J Biol Chem 2021; 298:101553. [PMID: 34973334 PMCID: PMC8800116 DOI: 10.1016/j.jbc.2021.101553] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 01/01/2023] Open
Abstract
The breakdown of all-trans-retinal (atRAL) clearance is closely associated with photoreceptor cell death in dry age-related macular degeneration (AMD) and autosomal recessive Stargardt's disease (STGD1), but its mechanisms remain elusive. Here, we demonstrate that activation of gasdermin E (GSDME) but not gasdermin D promotes atRAL-induced photoreceptor damage by activating pyroptosis and aggravating apoptosis through a mitochondria-mediated caspase-3-dependent signaling pathway. Activation of c-Jun N-terminal kinase was identified as one of the major causes of mitochondrial membrane rupture in atRAL-loaded photoreceptor cells, resulting in the release of cytochrome c from mitochondria to the cytosol, where it stimulated caspase-3 activation required for cleavage of GSDME. Aggregation of the N-terminal fragment of GSDME in the mitochondria revealed that GSDME was likely to penetrate mitochondrial membranes in photoreceptor cells after atRAL exposure. ABC (subfamily A, member 4) and all-trans-retinol dehydrogenase 8 are two key proteins responsible for clearing atRAL in the retina. Abca4−/−Rdh8−/− mice exhibit serious defects in atRAL clearance upon light exposure and serve as an acute model for dry AMD and STGD1. We found that N-terminal fragment of GSDME was distinctly localized in the photoreceptor outer nuclear layer of light-exposed Abca4−/−Rdh8−/− mice. Of note, degeneration and caspase-3 activation in photoreceptors were significantly alleviated in Abca4−/−Rdh8−/−Gsdme−/− mice after exposure to light. The results of this study indicate that GSDME is a common causative factor of photoreceptor pyroptosis and apoptosis arising from atRAL overload, suggesting that repressing GSDME may represent a potential treatment of photoreceptor atrophy in dry AMD and STGD1.
Collapse
Affiliation(s)
- Binxiang Cai
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen City, Fujian, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen City, Fujian, China
| | - Jiaying Lu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Kaiqi Qin
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Wenxu Liang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Xiaoling Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University; Xiamen Eye Center of Xiamen University, Xiamen City, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen City, Guangdong, China.
| |
Collapse
|