Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 523
Filter
1.
Exp Eye Res ; 242: 109879, 2024 May.
Article in English | MEDLINE | ID: mdl-38570182

ABSTRACT

Because the selective estrogen receptor modulator tamoxifen was shown to be retina-protective in the light damage and rd10 models of retinal degeneration, the purpose of this study was to test whether tamoxifen is retina-protective in a model where retinal pigment epithelium (RPE) toxicity appears to be the primary insult: the sodium iodate (NaIO3) model. C57Bl/6J mice were given oral tamoxifen (in the diet) or the same diet lacking tamoxifen, then given an intraperitoneal injection of NaIO3 at 25 mg/kg. The mice were imaged a week later using optical coherence tomography (OCT). ImageJ with a custom macro was utilized to measure retinal thicknesses in OCT images. Electroretinography (ERG) was used to measure retinal function one week post-injection. After euthanasia, quantitative real-time PCR (qRT-PCR) was performed. Tamoxifen administration partially protected photoreceptors. There was less photoreceptor layer thinning in OCT images of tamoxifen-treated mice. qRT-PCR revealed, in the tamoxifen-treated group, less upregulation of antioxidant and complement factor 3 mRNAs, and less reduction in the rhodopsin and short-wave cone opsin mRNAs. Furthermore, ERG results demonstrated preservation of photoreceptor function for the tamoxifen-treated group. Cone function was better protected than rods. These results indicate that tamoxifen provided structural and functional protection to photoreceptors against NaIO3. RPE cells were not protected. These neuroprotective effects suggest that estrogen-receptor modulation may be retina-protective. The fact that cones are particularly protected is intriguing given their importance for human visual function and their survival until the late stages of retinitis pigmentosa. Further investigation of this protective pathway could lead to new photoreceptor-protective therapeutics.


Subject(s)
Disease Models, Animal , Electroretinography , Iodates , Mice, Inbred C57BL , Retinal Degeneration , Tamoxifen , Tomography, Optical Coherence , Animals , Iodates/toxicity , Mice , Tomography, Optical Coherence/methods , Tamoxifen/pharmacology , Retinal Degeneration/prevention & control , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Real-Time Polymerase Chain Reaction , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Rhodopsin/metabolism , Rhodopsin/genetics , Selective Estrogen Receptor Modulators/pharmacology , RNA, Messenger/genetics , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/metabolism , Rod Opsins/metabolism
2.
Biomed Pharmacother ; 174: 116538, 2024 May.
Article in English | MEDLINE | ID: mdl-38579401

ABSTRACT

Glaucoma is considered a neurodegenerative disease characterized by progressive visual field defects that may lead to blindness. Although controlling intraocular pressure (IOP) is the mainstay of glaucoma treatment, some glaucoma patients have unmet needs due to unclear pathogenic mechanisms. Recently, there has been growing evidence that neuroinflammation is a potential target for the development of novel antiglaucoma agents. In this study, we investigated the protective effects and cellular mechanisms of H7E, a novel small molecule inhibits HDAC8, using in vitro and in vivo glaucoma-like models. Importantly, H7E mitigated extracellular MMP-9 activity and MCP-1 levels in glutamate- or S100B-stimulated reactive Müller glia. In addition, H7E inhibited the upregulation of inflammation- and proliferation-related signaling pathways, particularly the ERK and JNK MAPK pathways. Under conditions of oxidative damage, H7E prevents retinal cell death and reduces extracellular glutamate released from stressed Müller glia. In a mouse model of NMDA-induced retinal degeneration, H7E alleviated functional and structural defects within the inner retina as assessed by electroretinography and optical coherence tomography. Our results demonstrated that the newly identified compound H7E protects against glaucoma damage by specifically targeting HDAC8 activity in the retina. This protective effect is attributed to the inhibition of Müller glial activation and the prevention of retinal cell death caused by oxidative stress.


Subject(s)
Ependymoglial Cells , Glaucoma , Histone Deacetylase Inhibitors , Histone Deacetylases , Mice, Inbred C57BL , Oxidative Stress , Animals , Oxidative Stress/drug effects , Glaucoma/drug therapy , Glaucoma/metabolism , Glaucoma/pathology , Histone Deacetylase Inhibitors/pharmacology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Mice , Histone Deacetylases/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Disease Models, Animal , Neuroprotective Agents/pharmacology , Male , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/prevention & control
3.
Exp Eye Res ; 242: 109852, 2024 May.
Article in English | MEDLINE | ID: mdl-38460719

ABSTRACT

Oxidative stress plays a pivotal role in the pathogenesis of several neurodegenerative diseases. Retinal degeneration causes irreversible death of photoreceptor cells, ultimately leading to vision loss. Under oxidative stress, the synthesis of bioactive sphingolipid ceramide increases, triggering apoptosis in photoreceptor cells and leading to their death. This study investigates the effect of L-Cycloserine, a small molecule inhibitor of ceramide biosynthesis, on sphingolipid metabolism and the protection of photoreceptor-derived 661W cells from oxidative stress. The results demonstrate that treatment with L-Cycloserine, an inhibitor of Serine palmitoyl transferase (SPT), markedly decreases bioactive ceramide and associated sphingolipids in 661W cells. A nontoxic dose of L-Cycloserine can provide substantial protection of 661W cells against H2O2-induced oxidative stress by reversing the increase in ceramide level observed under oxidative stress conditions. Analysis of various antioxidant, apoptotic and sphingolipid pathway genes and proteins also confirms the ability of L-Cycloserine to modulate these pathways. Our findings elucidate the generation of sphingolipid mediators of cell death in retinal cells under oxidative stress and the potential of L-Cycloserine as a therapeutic candidate for targeting ceramide-induced degenerative diseases by inhibiting SPT. The promising therapeutic prospect identified in our findings lays the groundwork for further validation in in-vivo and preclinical models of retinal degeneration.


Subject(s)
Apoptosis , Ceramides , Cycloserine , Oxidative Stress , Sphingolipids , Oxidative Stress/drug effects , Cycloserine/pharmacology , Animals , Ceramides/metabolism , Ceramides/pharmacology , Mice , Sphingolipids/metabolism , Apoptosis/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/antagonists & inhibitors , Hydrogen Peroxide/toxicity , Hydrogen Peroxide/pharmacology , Cell Line , Retinal Degeneration/metabolism , Retinal Degeneration/prevention & control , Retinal Degeneration/pathology , Retinal Degeneration/drug therapy , Blotting, Western , Enzyme Inhibitors/pharmacology , Cell Survival/drug effects
4.
PLoS One ; 19(3): e0300045, 2024.
Article in English | MEDLINE | ID: mdl-38536853

ABSTRACT

Photoreceptor cell death can cause progressive and irreversible visual impairments. Still, effective therapies on retinal neuroprotection are not available. Hypoxia-inducible factors (HIFs) are transcriptional factors which strongly regulate angiogenesis, erythropoiesis, intracellular metabolism, and programed cell death under a hypoxic or an abnormal metabolic oxidative stress condition. Therefore, we aimed to unravel that inhibition of HIFs could prevent disease progression in photoreceptor cell death, as recent studies showed that HIFs might be pathologic factors in retinal diseases. Adult male balb/cAJcl (8 weeks old; BALB/c) were used to investigate preventive effects of a novel HIF inhibitor halofuginone (HF) on a murine model of light-induced retinopathy. After intraperitoneal injections of phosphate-buffered saline (PBS) or HF (0.4 mg/kg in PBS) for 5 days, male BALB/c mice were subjected to a dark-adaption to being exposed to a white LED light source at an intensity of 3,000 lux for 1 hour in order to induce light-induced retinal damage. After extensive light exposure, retinal damage was evaluated using electroretinography (ERG), optical coherence tomography (OCT), and TUNEL assay. Light-induced retinal dysfunction was suppressed by HF administration. The amplitudes of scotopic a-wave and b-wave as well as that of photopic b-wave were preserved in the HF-administered retina. Outer retinal thinning after extensive light exposure was suppressed by HF administration. Based on the TUNEL assay, cell death in the outer retina was seen after light exposure. However, its cell death was not detected in the HF-administered retina. Halofuginone was found to exert preventive effects on light-induced outer retinal cell death.


Subject(s)
Piperidines , Quinazolinones , Retinal Degeneration , Mice , Male , Animals , Retinal Degeneration/drug therapy , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Disease Models, Animal , Retina/pathology , Electroretinography
5.
Free Radic Biol Med ; 214: 42-53, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38309537

ABSTRACT

The degeneration of retinal pigment epithelium (RPE) plays an important role in the development of age-related macular degeneration (AMD). However, the underlying mechanism remains elusive. In this study, we identified that ZIP8, a metal-ion transporter, plays a crucial role in the degeneration of RPE cells mediated by ferroptosis. ZIP8 was found to be upregulated in patients with AMD through transcriptome analysis. Upregulated ZIP8 was also observed in both oxidative-stressed RPE cells and AMD mouse model. Importantly, knockdown of ZIP8 significantly inhibited ferroptosis in RPE cells induced by sodium iodate-induced oxidative stress. Blocking ZIP8 with specific antibodies reversed RPE degeneration and restored retinal function, improving visual loss in a mouse model of NaIO3-induced. Interestingly, the modification of the N-glycosylation sites N40, N72 and N88, but not N273, was essential for the intracellular iron accumulation mediated by ZIP8, which further led to increased lipid peroxidation and RPE death. These findings highlight the critical role of ZIP8 in RPE ferroptosis and provide a potential target for the treatment of diseases associated with retinal degeneration, including AMD.


Subject(s)
Ferroptosis , Macular Degeneration , Retinal Degeneration , Animals , Humans , Mice , Disease Models, Animal , Ferroptosis/genetics , Macular Degeneration/genetics , Retina , Retinal Degeneration/chemically induced , Retinal Degeneration/genetics , Retinal Degeneration/prevention & control , Retinal Pigments
6.
Invest Ophthalmol Vis Sci ; 65(1): 29, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38231527

ABSTRACT

Purpose: Retinal degeneration (RD) is a large cluster of retinopathies that is characterized by the progressive photoreceptor death and visual impairments. CX3CL1/CX3CR1 signaling has been documented to mediate the microglia activation and gliosis reaction during neurodegeneration. We intend to verify whether the CX3CL1/CX3CR1 signaling is involved in the RD pathology. Methods: A pharmacologically induced RD mice model was established. AZD8797, a CX3CR1 antagonist, was injected into the vitreous cavity of an RD model to modulate the neuroglia activation. Then, the experimental animals were subjected to functional, morphological, and behavioral analysis. Results: The CX3CL1/CX3CR1 signaling mediated neuroglia activation was implicated in the photoreceptor demise of an RD model. Intravitreal injection of AZD8797 preserved the retinal structure and enhanced the photoreceptor survival through inhibiting the CX3CL1/CX3CR1 expressions. Fundus photography showed that the distribution of retinal vessel was clear, and the severity of lesions was alleviated by AZD8797. In particular, these morphological benefits could be translated into remarkable functional improvements, as evidenced by the behavioral test and electroretinogram (mf-ERG) examination. A mechanism study showed that AZD8797 mitigated the microglia activation and migration in the degenerative retinas. The Müller cell hyper-reaction and secondary gliosis response were also suppressed by AZD8797. Conclusions: The neuroinflammation is implicated in the photoreceptor loss of RD pathology. Targeting the CX3CL1/CX3CR1 signaling may serve as an effective therapeutic strategy. Future refinements of these findings may cast light into the discovery of new medications for RD.


Subject(s)
Gliosis , Pyrimidines , Retinal Degeneration , Animals , Mice , Gliosis/drug therapy , Gliosis/prevention & control , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Thiazoles , Ependymoglial Cells
7.
Retina ; 44(1): 63-70, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37536462

ABSTRACT

PURPOSE: To evaluate prophylactic treatment (PTx) of lattice degeneration (LD) on retinal tear (RT) and rhegmatogenous retinal detachment (RRD) risk in fellow eyes of patients after primary RRD repair in the first eye. METHODS: This was a consecutive case series with cohort control involving patients with RRD repair from January 1, 2013, through December 31, 2017. Patients received PTx (PTx cohort) or no PTx (No-PTx cohort) in fellow eye with 5-year follow-up. Primary outcome measure was proportion with new fellow eye RT/RRD. Secondary outcomes included logarithm of minimum angle of resolution (logMAR) best-corrected visual acuity (BCVA) and status of myopia, posterior vitreous detachment, and pseudophakia. RESULTS: Four hundred ninety-eight patients were divided into 146 and 352 in PTx and No-PTx cohorts, respectively. PTx cohort developed significantly ( P < 0.05) fewer RT/RRD (17%) than No-PTx cohort (41%). PTx significantly ( P < 0.05) lowered RT/RRD irrespective of posterior vitreous detachment and myopia status. PTx patients undergoing phacoemulsification demonstrated significantly ( P < 0.05) less RT/RRD (22%) than No-PTx cohort (31%). There was no significant ( P = 0.96) final BCVA difference between PTx (median = 0 logMAR) and No-PTx (median = 0 logMAR) cohorts. CONCLUSION: PTx of asymptomatic fellow eye LD reduced RT/RRD risk.


Subject(s)
Cataract Extraction , Myopia , Retinal Degeneration , Retinal Detachment , Retinal Perforations , Vitreous Detachment , Humans , Retinal Detachment/prevention & control , Retinal Detachment/surgery , Retinal Detachment/complications , Vitreous Detachment/surgery , Vitreous Detachment/complications , Visual Acuity , Retina , Retinal Degeneration/prevention & control , Retinal Degeneration/surgery , Retinal Degeneration/complications , Retinal Perforations/surgery , Myopia/complications , Cataract Extraction/adverse effects , Retrospective Studies , Vitrectomy/adverse effects
8.
J Nanobiotechnology ; 21(1): 449, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38001463

ABSTRACT

Photoreceptor apoptosis is an important pathogenesis of retinal degeneration and a primary cause of vision loss with limited treatment methods. Mesenchymal stem/stromal cells-derived small extracellular vesicles (MSC-sEVs) have shown therapeutic value in various ocular disorders. Recent studies have revealed that hypoxic preconditioning can improve the effectiveness of MSC-sEVs in tissue regeneration. However, whether hypoxic preconditioned MSC-sEVs (Hyp-sEVs) exert superior effects on photoreceptor protection relative to normoxic conditioned MSC-sEVs (Nor-sEVs) remains unclear. Here, we reported that Hyp-sEVs further improved retinal structure, recovered retinal function, and suppressed photoreceptor apoptosis in N-methyl-N-nitrosourea (MNU)-induced mouse model compared with Nor-sEVs. Hyp-sEVs also exhibited enhanced anti-apoptotic roles in MNU-provoked 661 W cell injury in vitro. We then analyzed the protein profiles of Nor-sEVs and Hyp-sEVs by LC-MS/MS and found that growth-associated protein 43 (GAP43) was enriched in Hyp-sEVs. The knockdown of GAP43 abolished the retinal therapeutic effects of Hyp-sEVs. Mechanistically, hypoxic stimulation-induced hypoxia-inducible factor-1α (HIF-1α) activation was responsible for preventing tripartite motif-containing protein 25 (TRIM25)-mediated GAP43 ubiquitination and degradation, leading to the upregulation of GAP43 in Hyp-sEVs. Together, our findings uncover the efficacy and mechanism of Hyp-sEVs-based photoreceptor protection and highlight the potential of Hyp-sEVs as optimized therapeutics for retinal degeneration.


Subject(s)
Extracellular Vesicles , Retinal Degeneration , Mice , Animals , Retinal Degeneration/prevention & control , Retinal Degeneration/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Retina/metabolism , Extracellular Vesicles/metabolism , Hypoxia/metabolism
9.
FASEB J ; 37(11): e23250, 2023 11.
Article in English | MEDLINE | ID: mdl-37819682

ABSTRACT

Vision loss and blindness are frequently caused by photoreceptor degeneration, for example in age-related macular degeneration and retinitis pigmentosa. However, there is no effective medicine to treat these photoreceptor degeneration-related diseases. Cell senescence is a common phenotype in many diseases; however, few studies have reported whether it occurs in photoreceptor degeneration diseases. Herein, we identified that cell senescence is associated with photoreceptor degeneration induced by N-methyl-N-nitrosourea (MNU, a commonly used photoreceptor degeneration model), presented as increased senescence-associated ß-galactosidase activity, DNA damage, oxidative stress and inflammation-related cytokine Interleukin 6 (IL6), and upregulation of cyclin p21 or p16. These results suggested that visual function might be protected using anti-aging treatment. Furthermore, Hyperoside is reported to help prevent aging in various organs. In this study, we showed that Hyperoside, delivered intravitreally, alleviated photoreceptor cell senescence and ameliorated the functional and morphological degeneration of the retina in vivo and in vitro. Importantly, Hyperoside attenuated the MNU-induced injury and aging of photoreceptors via AMPK-ULK1 signaling inhibition. Taken together, our results demonstrated that Hyperoside can prevent MNU-induced photoreceptor degeneration by inhibiting cell senescence via the AMPK-ULK1 pathway.


Subject(s)
AMP-Activated Protein Kinases , Retinal Degeneration , Animals , AMP-Activated Protein Kinases/metabolism , Apoptosis , Cellular Senescence , Disease Models, Animal , Methylnitrosourea/toxicity , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinal Degeneration/chemically induced , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control
10.
Glia ; 71(11): 2511-2526, 2023 11.
Article in English | MEDLINE | ID: mdl-37533369

ABSTRACT

The expressions of ion channels by Müller glial cells (MGCs) may change in response to various retinal pathophysiological conditions. There remains a gap in our understanding of MGCs' responses to photoreceptor degeneration towards finding therapies. The study explores how an inhibition of store-operated Ca2+ entry (SOCE) and its major component, Orai1 channel, in MGCs protects photoreceptors from degeneration. The study revealed increased Orai1 expression in the MGCs of retinal degeneration 10 (rd10) mice. Enhanced expression of oxidative stress markers was confirmed as a crucial pathological mechanism in rd10 retina. Inducing oxidative stress in rat MGCs resulted in increasing SOCE and Ca2+ release-activated Ca2+ (CRAC) currents. SOCE inhibition by 2-Aminoethoxydiphenyl borate (2-APB) protected photoreceptors in degenerated retinas. Finally, molecular simulations proved the structural and dynamical features of 2-APB to the target structure Orai1. Our results provide new insights into the physiology of MGCs regarding retinal degeneration and shed a light on SOCE and Orai1 as new therapeutic targets.


Subject(s)
Calcium Channels , Retinal Degeneration , Rats , Mice , Animals , Calcium Channels/metabolism , Ependymoglial Cells/metabolism , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Calcium/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Calcium Signaling/physiology
11.
Ann N Y Acad Sci ; 1527(1): 60-74, 2023 09.
Article in English | MEDLINE | ID: mdl-37531162

ABSTRACT

With the increased use of artificial light and the prolonged use of optoelectronic products, light damage (LD) to the human retina has been identified as a global vision-threatening problem. While there is evidence of a significant correlation between light-induced retinal damage and age-related vision impairment in age-related macular degeneration, it is unclear how light-induced retinal degeneration manifests itself and whether there are agents capable of preventing the development of LD in the retina. This study investigated a mechanism by which blue light leads to photoreceptor death. By observing blue light exposure in retinal organoids and photoreceptor cells, we concluded that there could be significant apoptosis of the photoreceptors. We demonstrate that regenerating islet-derived 1 alpha (REG1A) prevents photoreceptors from undergoing this LD-induced apoptosis by increasing expression of the anti-apoptotic gene Bcl2 and downregulating expression of the pro-apoptotic gene Bax, resulting in reduced mitochondrial damage and improved aerobic capacity in photoreceptor cells. For the first time, REG1A has been shown to restore mitochondrial function and cell apoptosis after LD-induced damage, suggesting its potential application in the prevention and treatment of retinal vision loss.


Subject(s)
Retina , Retinal Degeneration , Humans , Retina/metabolism , Retinal Degeneration/prevention & control , Retinal Degeneration/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Apoptosis , Light , Lithostathine
12.
J Neuroimmune Pharmacol ; 18(3): 397-412, 2023 09.
Article in English | MEDLINE | ID: mdl-37326907

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.


Subject(s)
Retina , Retinal Degeneration , Humans , Retina/metabolism , Retina/pathology , Retinal Degeneration/prevention & control , Retinal Degeneration/drug therapy , Retinal Degeneration/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
13.
Int J Mol Sci ; 24(10)2023 May 13.
Article in English | MEDLINE | ID: mdl-37240082

ABSTRACT

An imbalance of homeostasis in the retina leads to neuron loss and this eventually results in a deterioration of vision. If the stress threshold is exceeded, different protective/survival mechanisms are activated. Numerous key molecular actors contribute to prevalent metabolically induced retinal diseases-the three major challenges are age-related alterations, diabetic retinopathy and glaucoma. These diseases have complex dysregulation of glucose-, lipid-, amino acid or purine metabolism. In this review, we summarize current knowledge on possible ways of preventing or circumventing retinal degeneration by available methods. We intend to provide a unified background, common prevention and treatment rationale for these disorders and identify the mechanisms through which these actions protect the retina. We suggest a role for herbal medicines, internal neuroprotective substances and synthetic drugs targeting four processes: parainflammation and/or glial cell activation, ischemia and related reactive oxygen species and vascular endothelial growth factor accumulation, apoptosis and/or autophagy of nerve cells and an elevation of ocular perfusion pressure and/or intraocular pressure. We conclude that in order to achieve substantial preventive or therapeutic effects, at least two of the mentioned pathways should be targeted synergistically. A repositioning of some drugs is considered to use them for the cure of the other related conditions.


Subject(s)
Diabetic Retinopathy , Glaucoma , Retinal Degeneration , Humans , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Retinal Degeneration/metabolism , Vascular Endothelial Growth Factor A/metabolism , Retina/metabolism , Diabetic Retinopathy/metabolism , Glaucoma/metabolism
14.
Sci Rep ; 13(1): 8380, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37225720

ABSTRACT

It is important to explore the effective approaches to prevent dry age-related macular degeneration (AMD). In this study, significantly decreased full-field electroretinograms wave amplitudes and disordered retina structures were detected in rat retinas of sodium iodate induced dry AMD model. Six a- and b-wave amplitudes and the antioxidant activities were significantly increased, and the outer nuclear layer thickness was significantly improved in the rat retinas treated with the combination of Lactobacillus fermentum NS9 (LF) and aronia anthocyanidin extract (AAE) compared with the model. The effects were much better than the treatment with AAE alone. The proteomics analysis showed the expressions of α-, ß- and γ-crystallins were increased by 3-8 folds in AAE treated alone and by 6-11 folds in AAE + LF treatment compared with the model, which was further confirmed by immuno-blotting analysis. Analysis of gut microbial composition indicated that higher abundance of the genus Parasutterella and species P. excrementihominis was found in the AAE + LF treatment compared with the other groups. The results indicated that the combined treatment of AAE + LF is a potential way to prevent the retina degeneration which is significantly better than the AAE treated alone.


Subject(s)
Geographic Atrophy , Limosilactobacillus fermentum , Photinia , Retinal Degeneration , Animals , Rats , Anthocyanins/pharmacology , Retinal Degeneration/chemically induced , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Retina , Plant Extracts/pharmacology
15.
J Neuroinflammation ; 20(1): 119, 2023 May 22.
Article in English | MEDLINE | ID: mdl-37217935

ABSTRACT

BACKGROUND: Chronic inflammation significantly contributes to photoreceptor death in blinding retinal diseases such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP). Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that act as key proinflammatory factors. We recently found the first-generation BET inhibitor JQ1 alleviated sodium iodate-induced retinal degeneration by suppressing cGAS-STING innate immunity. Here, we investigated the effects and mechanism of dBET6, a proteolysis­targeting chimera (PROTAC) small molecule that selectively degrades BET by the ubiquitin‒proteasome system, in light-induced retinal degeneration. METHODS: Mice were exposed to bright light to induce retinal degeneration, and the activation of cGAS-STING was determined by RNA-sequencing and molecular biology. Retinal function, morphology, photoreceptor viability and retinal inflammation were examined in the presence and absence of dBET6 treatment. RESULTS: Intraperitoneal injection of dBET6 led to the rapid degradation of BET protein in the retina without detectable toxicity. dBET6 improved retinal responsiveness and visual acuity after light damage (LD). dBET6 also repressed LD-induced retinal macrophages/microglia activation, Müller cell gliosis, photoreceptor death and retinal degeneration. Analysis of single-cell RNA-sequencing results revealed cGAS-STING components were expressed in retinal microglia. LD led to dramatic activation of the cGAS-STING pathway, whereas dBET6 suppressed LD-induced STING expression in reactive macrophages/microglia and the related inflammatory response. CONCLUSIONS: This study indicates targeted degradation of BET by dBET6 exerts neuroprotective effects by inhibiting cGAS-STING in reactive retinal macrophages/microglia, and is expected to become a new strategy for treatment of retinal degeneration.


Subject(s)
Retinal Degeneration , Mice , Animals , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Retinal Degeneration/metabolism , Inflammation/metabolism , Nucleotidyltransferases , RNA
16.
J Nanobiotechnology ; 21(1): 44, 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36747224

ABSTRACT

Photoreceptor degeneration is one of the major causes of progressive blindness which lacks of curative treatment. GW2580, a highly selective inhibitor of colony-stimulating factor 1 receptor, has the protective potential on neurons; however, little was known about the application of GW2580 on photoreceptor degeneration. In this study, BV-2 and 661W cells coculture system was constructed to investigate the interaction between microglia and photoreceptors. GW2580 was loaded into zeolitic imidazolate framework-90-rhodamine B (ZIF-90-RhB) to synthesize a novel kind of nanoparticles, namely, ZIF-90-RhB-GW2580, through a one-step self-assembly approach. A photoreceptor degeneration model was generated by intense light exposure in zebrafish and ZIF-90-RhB-GW2580 nanoparticles were delivered by the intraocular injection. The results showed that in vitro GW2580 treatment promoted phenotypic transformation in microglia and led to the blockade of photoreceptor apoptosis. Following the intraocular delivery of ZIF-90-RhB-GW2580 nanoparticles, the microglial proliferation and inflammatory response were significantly inhibited; moreover, the photoreceptors underwent alleviated injury with a recovery of retinal structure and visual function. In conclusion, the intraocular injection of ZIF-90-RhB-GW2580 at the early stage enables the precise delivery and sustained release of the GW2580, thus preventing the progression of photoreceptor degeneration.


Subject(s)
Nanoparticles , Retinal Degeneration , Zeolites , Animals , Zebrafish , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control
17.
Exp Eye Res ; 226: 109344, 2023 01.
Article in English | MEDLINE | ID: mdl-36509165

ABSTRACT

CLN2 neuronal ceroid lipofuscinosis is a rare hereditary neurodegenerative disorder characterized by deleterious sequence variants in TPP1 that result in reduced or abolished function of the lysosomal enzyme tripeptidyl peptidase 1 (TPP1). Children with this disorder experience progressive neurological decline and vision loss starting around 2-4 years of age. Ocular disease is characterized by progressive retinal degeneration and impaired retinal function culminating in total loss of vision. Similar retinal pathology occurs in a canine model of CLN2 disease with a null variant in TPP1. A study using the dog model was performed to evaluate the efficacy of ocular gene therapy to provide a continuous, long-term source of human TPP1 (hTPP1) to the retina, inhibit retinal degeneration and preserve retinal function. TPP1-/- dogs received an intravitreal injection of 1 x 1012 viral genomes of AAV2.CAG.hTPP1 in one eye and AAV2.CAG.GFP in the contralateral eye at 4 months of age. Ophthalmic exams, in vivo ocular imaging and electroretinography were repeated monthly to assess retinal structure and function. Retinal morphology, hTPP1 and GFP expression in the retina, optic nerve and lateral geniculate nucleus, and hTPP1 concentrations in the vitreous were evaluated after the dogs were euthanized at end stage neurological disease at approximately 10 months of age. Intravitreal administration of AAV2.CAG.hTPP1 resulted in stable, widespread expression of hTPP1 throughout the inner retina, prevented disease-related declines in retinal function and inhibited disease-related cell loss and storage body accumulation in the retina for at least 6 months. Uveitis occurred in eyes treated with the hTPP1 vector, but this did not prevent therapeutic efficacy. The severity of the uveitis was ameliorated with anti-inflammatory treatments. These results indicate that a single intravitreal injection of AAV2.CAG.hTPP1 is an effective treatment to inhibit ocular disease progression in canine CLN2 disease.


Subject(s)
Genetic Therapy , Neuronal Ceroid-Lipofuscinoses , Retinal Degeneration , Tripeptidyl-Peptidase 1 , Animals , Child , Dogs , Humans , Disease Models, Animal , Genetic Therapy/methods , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Neuronal Ceroid-Lipofuscinoses/pathology , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/prevention & control , Tripeptidyl-Peptidase 1/genetics , Intravitreal Injections
18.
Free Radic Biol Med ; 194: 209-219, 2023 01.
Article in English | MEDLINE | ID: mdl-36493984

ABSTRACT

Age-related macular degeneration is a common cause of blindless among the aged, which can mainly be attributed to oxidative stress and dysregulated autophagy in retinal pigment epithelium cells. Lactate was reported to act as a signaling molecule and exerted beneficial effect against oxidative stress. This study aims to investigate the protective effect of lactate against oxidative stress-induced retinal degeneration. Here, H2O2-induced oxidative stress cell model and sodium iodate-induced mice retinal degeneration model were established. It was found that H2O2 inhibited cell viability in ARPE-19 cells and sodium iodate induced deterioration of retinal pigment epithelium as well as apoptosis in retina. Pretreatment with lactate alleviated oxidative stress-induced cell death and retinal degeneration. Molecularly, lactate activated autophagy by up-regulating the ratio of LC3II/I, increased formation of LC3 puncta and autophagic vacuole. Further, lactate prevented H2O2-induced mitochondrial fission and maintained mitochondrial function by alleviating H2O2-induced mitochondrial membrane potential disruption and intracellular ROS generation. In contrast, application of 3-methyladenine, an inhibitor of autophagy, effectively weakened the protective effect of lactate against oxidative stress in vivo and in vitro. Taken together, all data in this study indicate that lactate protects against oxidative stress-induced retinal degeneration and preserves mitochondrial function by activating autophagy.


Subject(s)
Retinal Degeneration , Mice , Animals , Retinal Degeneration/chemically induced , Retinal Degeneration/drug therapy , Retinal Degeneration/prevention & control , Reactive Oxygen Species/metabolism , Lactic Acid/metabolism , Lactic Acid/pharmacology , Hydrogen Peroxide/metabolism , Oxidative Stress , Retinal Pigment Epithelium/metabolism , Autophagy
19.
J Ocul Pharmacol Ther ; 39(1): 80-85, 2023.
Article in English | MEDLINE | ID: mdl-36520599

ABSTRACT

Purpose: Age-related macular degeneration (AMD) is a leading cause of blindness in developed countries with little in the way of treatment that prevents progression to end-stage disease. Kaempferol (KF) is a plant-derived dietary flavonoid that has demonstrated as a strong antioxidant showing neuroprotection in stroke models. We hypothesize that KF has protective effects against retinal degeneration and may serve as a therapeutic agent against AMD. Methods: BALB/c albino mice were assigned to 1 of 2 groups: control-treated or KF-treated retinal light injury mice. Mice were exposed to 8,000 lux cool white fluorescent light for 2 h to induce light injury. Control or KF was injected intraperitoneally after light injury for 5 days. Scotopic electroretinography (ERG) was recorded before light injury and 7 days after light injury. The retinal morphology and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays were performed after light injury. Results: ERG a- and b-wave amplitudes were significantly reduced in the retinal light injury group compared with the nonretinal light injury group. Retinal light injury produced markedly thinning of the outer nuclear layer along with significant TUNEL-positive signals. In contrast KF treatments significantly attenuated reduction of ERG a- and b- wave amplitudes and the loss of the outer nuclear layer. Conclusions: KF protects retinal photoreceptors and preserves retinal function against retinal degeneration caused by light injury. These initial findings suggest that KF may represent a novel therapy for retinal degenerative conditions such as AMD.


Subject(s)
Macular Degeneration , Retinal Degeneration , Mice , Animals , Retinal Degeneration/drug therapy , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Kaempferols/pharmacology , Retina , Photoreceptor Cells, Vertebrate , Disease Models, Animal , Electroretinography , Macular Degeneration/complications , Apoptosis
20.
PLoS One ; 17(10): e0269437, 2022.
Article in English | MEDLINE | ID: mdl-36227868

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.


Subject(s)
Macular Degeneration , Retinal Degeneration , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Interferons/metabolism , Lipofuscin/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Mice , Nucleotidyltransferases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/drug therapy , Retinal Degeneration/etiology , Retinal Degeneration/prevention & control , Retinal Pigments/metabolism , Retinaldehyde/metabolism , Retinaldehyde/pharmacology , Retinoids/metabolism , Retinoids/pharmacology , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...