Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.292
Filter
1.
Transl Vis Sci Technol ; 13(7): 16, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39042048

ABSTRACT

Purpose: The purpose of this study was to investigate the ocular morphological characteristics of Col4a3-/- mice as a model of Alport syndrome (AS) and the potential pathogenesis. Methods: The expression of collagen IV at 8, 12, and 21 weeks of age was evaluated by immunohistochemistry in wild-type (WT) and Col4a3-/- mice. Hematoxylin and eosin (H&E) staining and thickness measurements were performed to assess the thickness of anterior lens capsule and retina. Ultrastructure analysis of corneal epithelial basement membrane, anterior lens capsule, internal limiting membrane (ILM), and retinal pigment epithelium (RPE) basement membrane was performed using transmission electron microscopy. Finally, Müller cell activation was evaluated by glial fibrillary acidic protein (GFAP) expression. Results: Collagen IV was downregulated in the corneal epithelial basement membrane and ILM of Col4a3-/- mice. The hemidesmosomes of Col4a3-/- mice corneal epithelium became flat and less electron-dense than those of the WT group. Compared with those of the WT mice, the anterior lens capsules of Col4a3-/- mice were thinner. Abnormal structure was detected at the ILM Col4a3-/- mice, and the basal folds of the RPE basement membrane in Col4a3-/- mice were thicker and shorter. The retinas of Col4a3-/- mice were thinner than those of WT mice, especially within 1000 µm away from the optic nerve. GFAP expression enhanced in each age group of Col4a3-/- mice. Conclusions: Our results suggested that Col4a3-/- mice exhibit ocular anomalies similar to patients with AS. Additionally, Müller cells may be involved in AS retinal anomalies. Translational Relevance: This animal model could provide an opportunity to understand the underlying mechanisms of AS ocular disorders and to investigate potential new treatments.


Subject(s)
Basement Membrane , Collagen Type IV , Disease Models, Animal , Mice, Knockout , Nephritis, Hereditary , Animals , Nephritis, Hereditary/pathology , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Collagen Type IV/deficiency , Mice , Basement Membrane/metabolism , Basement Membrane/pathology , Basement Membrane/ultrastructure , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Microscopy, Electron, Transmission , Mice, Inbred C57BL , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Lens Capsule, Crystalline/ultrastructure , Epithelium, Corneal/pathology , Epithelium, Corneal/ultrastructure , Epithelium, Corneal/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Retina/pathology , Retina/metabolism , Retina/ultrastructure , Autoantigens/genetics , Autoantigens/metabolism , Ependymoglial Cells/pathology , Ependymoglial Cells/metabolism , Ependymoglial Cells/ultrastructure , Immunohistochemistry , Male
2.
Medicine (Baltimore) ; 103(29): e38853, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39029076

ABSTRACT

RATIONALE: Autosomal recessive bestrophinopathy (ARB) is a subtype of bestrophinopathy caused by biallelic mutations of the BEST1 gene, which affect the retinal pigment epithelium (RPE). Studying RPE abnormalities through imaging is essential for understanding ARB. This case series involved the use of multimodal imaging techniques, namely autofluorescence (AF) imaging at 488 nm [short-wavelength AF] and 785 nm [near-infrared AF (NIR-AF)] and polarization-sensitive optical coherence tomography (PS-OCT), to investigate RPE changes in 2 siblings with ARB. PATIENT CONCERNS: Two Japanese siblings (Case 1: male, followed for 20-23 years; Case 2: female, followed for 13-17 years) carried compound heterozygous mutations of the BEST1 gene. DIAGNOSIS: Both siblings were diagnosed with ARB. INTERVENTIONS AND OUTCOMES: Multimodal imaging techniques were used to evaluate RPE changes. Both siblings had funduscopic changes similar to those seen in the vitelliruptive stage of Best vitelliform macular dystrophy during the follow-up period. NIR-AF imaging showed hypo-AF of the entire macular lesion in both cases, and this hypo-AF remained stable over time. PS-OCT confirmed reduced RPE melanin content in these hypo-AF areas. Additionally, hyper-NIR-AF dots were observed within hypo-NIR-AF areas. Concomitant identification of focally thickened RPE melanin on PS-OCT imaging and hyper-AF on short-wavelength AF imaging at the sites containing hyper-NIR-AF dots indicated that the hyper-NIR-AF dots had originated from either stacked RPE cells or RPE dysmorphia. LESSONS: We confirmed RPE abnormalities in ARB, including diffuse RPE melanin damage in the macula alongside evidence of RPE activity-related changes. This case series demonstrates that multimodal imaging, particularly NIR-AF and PS-OCT, provides detailed insights into RPE alterations in ARB.


Subject(s)
Bestrophins , Eye Diseases, Hereditary , Multimodal Imaging , Retinal Diseases , Retinal Pigment Epithelium , Tomography, Optical Coherence , Humans , Multimodal Imaging/methods , Male , Female , Tomography, Optical Coherence/methods , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/diagnostic imaging , Retinal Diseases/genetics , Retinal Diseases/diagnostic imaging , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/pathology , Bestrophins/genetics , Young Adult , Optical Imaging/methods , Adolescent , Siblings
3.
Cells ; 13(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39056772

ABSTRACT

The chromatin-associated protein WDR5 has been widely studied due to its role in histone modification and its potential as a pharmacological target for the treatment of cancer. In humans, the protein with highest sequence homology to WDR5 is encoded by the retrogene WDR5B, which remains unexplored. Here, we used CRISPR-Cas9 genome editing to generate WDR5B knockout and WDR5B-FLAG knock-in cell lines for further characterization. In contrast to WDR5, WDR5B exhibits low expression in pluripotent cells and is upregulated upon neural differentiation. Loss or shRNA depletion of WDR5B impairs cell growth and increases the fraction of non-viable cells in proliferating retinal pigment epithelial (RPE) cultures. CUT&RUN chromatin profiling in RPE and neural progenitors indicates minimal WDR5B enrichment at established WDR5 binding sites. These results suggest that WDR5 and WDR5B exhibit several divergent biological properties despite sharing a high degree of sequence homology.


Subject(s)
Cell Proliferation , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , CRISPR-Cas Systems/genetics , Epithelial Cells/metabolism , Cell Differentiation , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cell Line , Animals , Gene Editing
4.
Cells ; 13(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39056810

ABSTRACT

Age-related ocular diseases such as age-related macular degeneration, glaucoma, and diabetic retinopathy are major causes of irreversible vision impairment in the elderly. Conventional treatments focus on symptom relief and disease slowdown, often involving surgery, but fall short of providing a cure, leading to substantial vision loss. Regenerative medicine, particularly mesenchymal stem cells (MSCs), holds promise for ocular disease treatment. This study investigates the synergistic potential of combining placenta-derived MSCs (PD-MSCs) with Achyranthis radix extract (ARE) from Achyranthes japonica to enhance therapeutic outcomes. In a 24-h treatment, ARE significantly increased the proliferative capacity of PD-MSCs and delayed their senescence (* p < 0.05). ARE also enhanced antioxidant capabilities and increased the expression of regeneration-associated genes in an in vitro injured model using chemical damages on human retinal pigment epithelial cell line (ARPE-19) (* p < 0.05). These results suggest that ARE-primed PD-MSC have the capability to enhance the activation of genes associated with regeneration in the injured eye via increasing antioxidant properties. Taken together, these findings support the conclusion that ARE-primed PD-MSC may serve as an enhanced source for stem cell-based therapy in ocular diseases.


Subject(s)
Antioxidants , Mesenchymal Stem Cells , Placenta , Plant Extracts , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Antioxidants/pharmacology , Plant Extracts/pharmacology , Female , Placenta/metabolism , Placenta/drug effects , Pregnancy , Achyranthes/chemistry , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , Cell Proliferation/drug effects , Cell Line
5.
Transl Vis Sci Technol ; 13(7): 7, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980261

ABSTRACT

Purpose: Lipid nanoparticles (LNPs) show promise in their ability to introduce mRNA to drive protein expression in specific cell types of the mammalian eye. Here, we examined the ability of mRNA encapsulated in LNPs with two distinct formulations to drive gene expression in mouse and human retina and other ocular tissues. Methods: We introduced mRNA-carrying LNPs into two biological systems. Intravitreal injections were tested to deliver LNPs into the mouse eye. Human retinal pigment epithelium (RPE) and retinal explants were used to assess mRNA expression in human tissue. We analyzed specificity of expression using histology, immunofluorescence, and imaging. Results: In mice, mRNAs encoding GFP and ciliary neurotrophic factor (CNTF) were specifically expressed by Müller glia and RPE. Acute inflammatory changes measured by microglia distribution (Iba-1) or interleukin-6 (IL-6) expression were not observed 6 hours post-injection. Human RPE also expressed high levels of GFP. Human retinal explants expressed GFP in cells with apical and basal processes consistent with Müller glia and in perivascular cells consistent with macrophages. Conclusions: We demonstrated the ability to reliably transfect subpopulations of retinal cells in mouse eye tissues in vivo and in human ocular tissues. Of significance, intravitreal injections were sufficient to transfect the RPE in mice. To our knowledge, we demonstrate delivery of mRNA using LNPs in human ocular tissues for the first time. Translational Relevance: Ocular gene-replacement therapies using non-viral vector methods are a promising alternative to adeno-associated virus (AAV) vectors. Our studies show that mRNA LNP delivery can be used to transfect retinal cells in both mouse and human tissues without inducing significant inflammation. This methodology could be used to transfect retinal cell lines, tissue explants, mice, or potentially as gene-replacement therapy in a clinical setting in the future.


Subject(s)
Intravitreal Injections , Nanoparticles , RNA, Messenger , Retinal Pigment Epithelium , Animals , Humans , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Mice , Retinal Pigment Epithelium/metabolism , Nanoparticles/chemistry , Mice, Inbred C57BL , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/administration & dosage , Retina/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ependymoglial Cells/metabolism , Gene Transfer Techniques , Liposomes
6.
Braz J Biol ; 84: e282840, 2024.
Article in English | MEDLINE | ID: mdl-38985071

ABSTRACT

Bisphenol A (BPA) may adversely affect human health by inducing oxidative stress and irreversible damage to cells. Bioactive compounds found in some functional foods, individually or in combination, can attenuate the negative effects of BPA exposure; an example is the multi-supplement containing guarana (Gua), selenium (Se), and L-carnitine (LC) -GSC- which has already demonstrated antioxidant, genoprotective, and immunomodulatory activities. This study aimed to determine the effect of GSC and its constituents on oxidative and genotoxic alterations triggered by BPA exposure in the retinal epithelial cell line. The cells exposed to BPA (0.001, 0.01, 0.1, 1, 3, and 10 µM) to determine the lowest concentration required to induce cyto-genotoxicity. ARPE-19 cells were then concomitantly exposed to the selected BPA concentration, GSC, and its components (Gua, 1.07 mg/mL; Se, 0.178 µg/mL; and LC, 1.43 mg/mL). Flow cytometry, biochemical assays, qRT-PCR, genotoxicity, apoptosis, and cellular proliferation. Based on our results, 10 µM of BPA could induce cyto-genotoxic and oxidative alterations. BPA did not alter the Bcl-2/BAX expression ratio but induced Casp3 and Casp8 overexpression, suggesting that apoptosis was induced mainly via the extrinsic pathway. GSC partially reversed the alterations triggered by BPA in ARPE-19 cells. However, Se had unexpected negative effects on ARPE-19 cells. The multi-supplement GSC may attenuate changes in oxidative and genotoxic markers related to exposure of ARPE-19 cells to BPA. our results revealed that the antioxidant, anti-apoptotic, and genoprotective properties of GSC were not universally shared by its individual, once Se did not exhibit any positive impact.


Subject(s)
Apoptosis , Benzhydryl Compounds , Carnitine , Oxidative Stress , Phenols , Retinal Pigment Epithelium , Selenium , Phenols/toxicity , Benzhydryl Compounds/toxicity , Humans , Selenium/pharmacology , Carnitine/pharmacology , Retinal Pigment Epithelium/drug effects , Oxidative Stress/drug effects , Apoptosis/drug effects , Cell Line , Paullinia/chemistry , DNA Damage/drug effects , Antioxidants/pharmacology , Epithelial Cells/drug effects , Flow Cytometry , Dietary Supplements
7.
J Biomed Opt ; 29(Suppl 2): S22707, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38962492

ABSTRACT

Significance: Adaptive optics fluorescence lifetime ophthalmoscopy (AOFLIO) provides a label-free approach to observe functional and molecular changes at cellular scale in vivo. Adding multispectral capabilities improves interpretation of lifetime fluctuations due to individual fluorophores in the retinal pigment epithelium (RPE). Aim: To quantify the cellular-scale changes in autofluorescence with age and eccentricity due to variations in lipofuscin, melanin, and melanolipofuscin in RPE using multispectral AOFLIO. Approach: AOFLIO was performed on six subjects at seven eccentricities. Four imaging channels ( λ ex / λ em ) were used: 473/SSC, 473/LSC, 532/LSC, and 765/NIR. Cells were segmented and the timing signals of each pixel in a cell were combined into a single histogram, which were then used to compute the lifetime and phasor parameters. An ANOVA was performed to investigate eccentricity and spectral effects on each parameter. Results: A repeatability analysis revealed < 11.8 % change in lifetime parameters in repeat visits for 532/LSC. The 765/NIR and 532/LSC had eccentricity and age effects similar to previous reports. The 473/LSC had a change in eccentricity with mean lifetime and a phasor component. Both the 473/LSC and 473/SSC had changes in eccentricity in the short lifetime component and its relative contribution. The 473/SSC had no trend in eccentricity in phasor. The comparison across the four channels showed differences in lifetime and phasor parameters. Conclusions: Multispectral AOFLIO can provide a more comprehensive picture of changes with age and eccentricity. These results indicate that cell segmentation has the potential to allow investigations in low-photon scenarios such as in older or diseased subjects with the co-capture of an NIR channel (such as 765/NIR) with the desired spectral channel. This work represents the first multispectral, cellular-scale fluorescence lifetime comparison in vivo in the human RPE and may be a useful method for tracking diseases.


Subject(s)
Ophthalmoscopy , Retinal Pigment Epithelium , Humans , Ophthalmoscopy/methods , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/chemistry , Adult , Male , Female , Aging/physiology , Middle Aged , Aged , Young Adult , Optical Imaging/methods , Lipofuscin/metabolism , Lipofuscin/analysis , Lipofuscin/chemistry , Feasibility Studies
8.
Int Ophthalmol ; 44(1): 314, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965086

ABSTRACT

BACKGROUND: Oxidative stress-induced retinal pigment epithelium (RPE) cell damage is a major factor in age-related macular degeneration (AMD). Vitamin D3 (VD3) is a powerful antioxidant and it has been suggested to have anti-aging properties and potential for treating AMD. This study aimed to investigate the effect of VD3 on RPE cell oxidative apoptosis of RPE cells in order to provide experimental evidence for the treatment of AMD. METHODS: Human retinal pigment epithelial cell 19 (ARPE-19) cells were divided into four groups: blank group (untreated), model group (incubated in medium with 400 µmol/L H2O2 for 1 h), VD3 group (incubated in medium with 100 µmol/L VD3 for 24 h), and treatment group (incubated in medium with 400 µmol/L H2O2 for 1 h and 100 µmol/L VD3 for 24 h). Cell viability, cell senescence, ROS content, expression levels of vitamin D specific receptors, Akt, Sirt1, NAMPT, and JNK mRNA expression levels, SOD activity, and MDA, GSH, and GPX levels were measured. RESULTS: We first established an ARPE-19 cell stress model with H2O2. Our control experiment showed that VD3 treatment had no significant effect on ARPE-19 cell viability within 6-48 h. Treating the stressed ARPE-19 cells with VD3 showed mixed results; caspase-3 expression was decreased, Bcl-2 expression was increased, MDA level of ARPE-19 cells was decreased, GSH-PX, GPX and SOD levels were increased, the relative mRNA expression levels of Akt, Sirt1, NAMPT were increased (P < 0.05), and the relative mRNA expression level of JNK was decreased (P < 0.05). CONCLUSION: VD3 can potentially slow the development of AMD.


Subject(s)
Apoptosis , Cell Survival , Oxidative Stress , Retinal Pigment Epithelium , Humans , Oxidative Stress/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Cell Survival/drug effects , Apoptosis/drug effects , Macular Degeneration/metabolism , Vitamins/pharmacology , Vitamin D/pharmacology , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism , Cells, Cultured , Sirtuin 1/metabolism , Sirtuin 1/genetics , Cellular Senescence/drug effects , Cell Line , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/toxicity
9.
Sci Rep ; 14(1): 16498, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020017

ABSTRACT

Epithelial-Mesenchymal Transition (EMT) of retinal pigment epithelial (RPE) cells is recognized as pivotal in various retinal diseases. Previous studies have suggested a reciprocal regulation between reactive oxygen species (ROS) and EMT, though the involvement of peroxidized lipids or the effects of reducing them has remained unclear. The present study disclosed that EMT of ARPE-19 cells induced by TGF-ß2 and TNF-α involves increased lipid peroxidation, and Ferrostatin-1 (Fer-1), a lipophilic antioxidative agent, successfully inhibited the increase in lipid peroxidation. Fer-1 suppressed the formation of EMT-associated fibrotic deposits, while EMT induction or Fer-1 treatment did not influence the cell viability or proliferation. Functionally, Fer-1 impeded EMT-driven cell migration and reduction in transepithelial electrical resistance. It demonstrated regulatory prowess by downregulating the mesenchymal marker fibronectin, upregulating the epithelial marker ZO-1, and inhibiting the EMT-associated transcriptional factor ZEB1. Additionally, VEGF, a major pathogenic cytokine in various retinal diseases, is also upregulated during EMT, and Fer-1 significantly mitigated the effect. The present study disclosed the involvement of lipid peroxidation in EMT of RPE cells, and suggests the suppression of lipid peroxidation may be a potential therapeutic target in retinal diseases in which EMT is implicated.


Subject(s)
Epithelial-Mesenchymal Transition , Lipid Peroxidation , Retinal Pigment Epithelium , Epithelial-Mesenchymal Transition/drug effects , Humans , Retinal Pigment Epithelium/metabolism , Cell Line , Cell Movement/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Transforming Growth Factor beta2/metabolism , Epithelial Cells/metabolism , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Proliferation , Zonula Occludens-1 Protein/metabolism , Fibronectins/metabolism
10.
BMJ Open Ophthalmol ; 9(1)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38981710

ABSTRACT

Lesions of incomplete retinal pigment epithelium and outer retinal atrophy (iRORA) are associated with disease progression in age-related macular degeneration. However, the corresponding functional impact of these precursor lesions is unknown.We present a cross-sectional study of four patients employing clinical-grade MAIA (stimulus size: 0.43°, ~125 µm) and adaptive optics scanning light ophthalmoscope (AOSLO, stimulus size 0.07°, ~20 µm) based microperimetry (MP) to assess the specific impact of iRORA lesions on retinal sensitivity.AOSLO imaging showed overall reduced photoreceptor reflectivity and patches of hyporeflective regions at drusen with interspersed hyper-reflective foci in iRORA regions. MAIA-MP yielded an average retinal sensitivity loss of -7.3±3.1 dB at iRORA lesions compared with the in-eye control. With AOSLO-MP, the corresponding sensitivity loss was 20.1±4.8 dB.We demonstrated that iRORA lesions are associated with a severe impairment in retinal sensitivity. Larger cohort studies will be necessary to validate our findings.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Tomography, Optical Coherence , Visual Field Tests , Humans , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/diagnostic imaging , Cross-Sectional Studies , Macular Degeneration/pathology , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Female , Male , Aged , Tomography, Optical Coherence/methods , Visual Field Tests/methods , Visual Acuity/physiology , Aged, 80 and over , Visual Fields/physiology , Ophthalmoscopy/methods , Atrophy/pathology
11.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000171

ABSTRACT

Recurrent computed tomography (CT) examination has become a common diagnostic procedure for several diseases and injuries. Though each singular CT scan exposes individuals at low doses of low linear energy transfer (LET) radiation, the cumulative dose received from recurrent CT scans poses an increasing concern for potential health risks. Here, we evaluated the biological effects of recurrent CT scans on the DNA damage response (DDR) in human fibroblasts and retinal pigment epithelial cells maintained in culture for five months and subjected to four CT scans, one every four weeks. DDR kinetics and eventual accumulation of persistent-radiation-induced foci (P-RIF) were assessed by combined immunofluorescence for γH2AX and 53BP1, i.e., γH2AX/53BP1 foci. We found that CT scan repetitions significantly increased both the number and size of γH2AX/53BP1 foci. In particular, after the third CT scan, we observed the appearance of giant foci that might result from the overlapping of individual small foci and that do not associate with irreversible growth arrest, as shown by DNA replication in the foci-carrying cells. Whether these giant foci represent coalescence of unrepaired DNA damage as reported following single exposition to high doses of high LET radiation is still unclear. However, morphologically, these giant foci resemble the recently described compartmentalization of damaged DNA that should facilitate the repair of DNA double-strand breaks but also increase the risk of chromosomal translocations. Overall, these results indicate that for a correct evaluation of the damage following recurrent CT examinations, it is necessary to consider the size and composition of the foci in addition to their number.


Subject(s)
DNA Damage , Fibroblasts , Histones , Tomography, X-Ray Computed , Tumor Suppressor p53-Binding Protein 1 , Humans , Tumor Suppressor p53-Binding Protein 1/metabolism , Tomography, X-Ray Computed/methods , Histones/metabolism , Fibroblasts/radiation effects , Fibroblasts/metabolism , Dose-Response Relationship, Radiation , Retinal Pigment Epithelium/radiation effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/cytology , Cell Line , DNA Repair , Linear Energy Transfer
12.
Medicine (Baltimore) ; 103(28): e38908, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996139

ABSTRACT

The retinal pigment epithelium (RPE) is a regularly arranged monolayer of cells in the outermost layer of the retina. It is crucial for transporting nutrients and metabolic substances in the retina and maintaining the retinal barrier. RPE dysfunction causes diseases related to vision loss. Thus, understanding the mechanisms involved in normal RPE function is vital. Adenosine monophosphate-activated protein kinase (AMPK) is an RPE energy sensor regulating various signaling and metabolic pathways to maintain cellular energetic homeostasis. AMPK activation is involved in multiple signaling pathways regulated by autophagy in the RPE, thereby protecting the cells from oxidative stress and slowing RPE degeneration. In this review, we attempt to broaden the understanding of the pathogenesis of RPE dysfunction by focusing on the role and mechanism of AMPK regulation of autophagy in the RPE. The correlation between RPE cellular homeostasis and role of AMPK was determined by analyzing the structure and mechanism of AMPK and its signaling pathway in autophagy. The protective effect of AMPK-regulated autophagy on the RPE for gaining insights into the regulatory pathways of RPE dysfunction has been discussed.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Homeostasis , Retinal Pigment Epithelium , Signal Transduction , Autophagy/physiology , Retinal Pigment Epithelium/metabolism , Humans , Homeostasis/physiology , AMP-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Oxidative Stress/physiology
13.
Sci Rep ; 14(1): 16129, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997397

ABSTRACT

The choroid, a vascularized tissue situated between the retina and the sclera, plays a crucial role in maintaining ocular homeostasis. Despite its significance, research on choroidal abnormalities and the establishment of effective in vitro models have been limited. In this study, we developed an in vitro choroid model through the co-culture of human induced pluripotent stem cells (hiPSC)-derived endothelial cells (ECs) and mouse choroidal fibroblasts (msCFs) with hiPSC-derived retinal pigment epithelial (RPE) cells via a permeable membrane. This model, inclusive of ECs, CFs, and RPE cells, exhibited similarities with in vivo choroidal vessels, as confirmed through immunohistochemistry of extracellular matrix markers and vascular-related markers, as well as choroid angiogenesis sprouting assay analysis. The effectiveness of our in vitro model was demonstrated in assessing vascular changes induced by drugs targeting vasoregulation. Our model offers a valuable tool for gaining insights into the pathological mechanisms underlying choroid development and the progression of choroidal vascular diseases.


Subject(s)
Choroid , Coculture Techniques , Endothelial Cells , Induced Pluripotent Stem Cells , Retinal Pigment Epithelium , Choroid/blood supply , Choroid/metabolism , Animals , Humans , Mice , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Endothelial Cells/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , Fibroblasts/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Cells, Cultured
14.
Transl Vis Sci Technol ; 13(7): 2, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949633

ABSTRACT

Purpose: We sought to evaluate the efficacy of growth differentiation factor (GDF)-15 treatment for suppressing epithelial-mesenchymal transition (EMT) and alleviating transforming growth factor ß2 (TGFß2)-induced lens opacity. Methods: To test whether GDF-15 is a molecule that prevents EMT, we pretreated the culture with GDF-15 in neural progenitor cells, retinal pigment epithelial cells, and lens epithelial cells and then treated with factors that promote EMT, GDF-11, and TGFß2, respectively. To further investigate the efficacy of GDF-15 on alleviating lens opacity, we used mouse lens explant culture to mimic secondary cataracts. We pretreated the lens culture with GDF-15 and then added TGFß2 to develop lens opacity (n = 3 for each group). Western blot and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to measure EMT protein and gene expression, respectively. Results: In cell culture, GDF-15 pretreatment significantly attenuated EMT marker expression in cultured cells induced by treatment with GDF-11 or TGFß2. In the lens explant culture, GDF-15 pretreatment also reduced mouse lens opacity induced by exposure to TGFß2. Conclusions: Our results indicate that GDF-15 could alleviate TGFß2-induced EMT and is a potential therapeutic agent to slow or prevent posterior capsular opacification (PCO) progression after cataract surgery. Translational Relevance: Cataracts are the leading cause of blindness worldwide, with the only current treatment involving surgical removal of the lens and replacement with an artificial lens. However, PCO, also known as secondary cataract, is a common complication after cataract surgery. The development of an adjuvant that slows the progression of PCO will be beneficial to the field of anterior complications.


Subject(s)
Cataract , Epithelial-Mesenchymal Transition , Growth Differentiation Factor 15 , Lens, Crystalline , Transforming Growth Factor beta2 , Animals , Epithelial-Mesenchymal Transition/drug effects , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Cataract/pathology , Cataract/metabolism , Cataract/prevention & control , Mice , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/drug effects , Mice, Inbred C57BL , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Blotting, Western , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/metabolism
15.
Proc Natl Acad Sci U S A ; 121(30): e2402560121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39018199

ABSTRACT

The key role of a thyroid hormone receptor in determining the maturation and diversity of cone photoreceptors reflects a profound influence of endocrine signaling on the cells that mediate color vision. However, the route by which hormone reaches cones remains enigmatic as cones reside in the retinal photoreceptor layer, shielded by the blood-retina barrier. Using genetic approaches, we report that cone differentiation is regulated by a membrane transporter for thyroid hormone, MCT8 (SLC16A2), in the retinal pigment epithelium (RPE), which forms the outer blood-retina barrier. Mct8-deficient mice display hypothyroid-like cone gene expression and compromised electroretinogram responses. Mammalian color vision is typically facilitated by cone types that detect medium-long (M) and short (S) wavelengths of light but Mct8-deficient mice have a partial shift of M to S cone identity, resembling the phenotype of thyroid hormone receptor deficiency. RPE-specific ablation of Mct8 results in similar shifts in cone identity and hypothyroid-like gene expression whereas reexpression of MCT8 in the RPE in Mct8-deficient mice partly restores M cone identity, consistent with paracrine-like control of thyroid hormone signaling by the RPE. Our findings suggest that in addition to transport of essential solutes and homeostatic support for photoreceptors, the RPE regulates the thyroid hormone signal that promotes cone-mediated vision.


Subject(s)
Cell Differentiation , Mice, Knockout , Monocarboxylic Acid Transporters , Retinal Cone Photoreceptor Cells , Retinal Pigment Epithelium , Symporters , Animals , Retinal Cone Photoreceptor Cells/metabolism , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Symporters/metabolism , Symporters/genetics , Retinal Pigment Epithelium/metabolism , Mice , Thyroid Hormones/metabolism , Electroretinography
16.
Cell Commun Signal ; 22(1): 359, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992691

ABSTRACT

PURPOSE: Bietti crystalline dystrophy (BCD) is an inherited retinal degeneration disease caused by mutations in the CYP4V2 gene. Currently, there is no clinical therapy approach available for BCD patients. Previous research has suggested that polyunsaturated fatty acids (PUFAs) may play a significant role in the development of BCD, implicating the involvement of ferroptosis in disease pathogenesis. In this work, we aimed to investigate the interplay between ferroptosis and BCD and to detect potential therapeutic strategies for the disease. METHODS: Genetic-edited RPE cell line was first established in this study by CRISPR-Cas9 technology. Cyp4v3 (the homologous gene of human CYP4V2) knock out (KO) mice have also been used. Lipid profiling and transcriptome analysis of retinal pigment epithelium (RPE) cells from Cyp4v3 KO mice have been conducted. Ferroptosis phenotypes have been first investigated in BCD models in vitro and in vivo, including lipid peroxidation, mitochondrial changes, elevated levels of reactive oxygen species (ROS), and altered gene expression. Additionally, an iron chelator, deferiprone (DFP), has been tested in vitro and in vivo to determine its efficacy in suppressing ferroptosis and restoring the BCD phenotype. RESULTS: Cyp4v3 KO mice exhibited progressive retinal degeneration and lipid accumulation, similar to the BCD phenotype, which was exacerbated by a high-fat diet (HFD). Increased levels of PUFAs, such as EPA (C22:5) and AA (C20:4), were observed in the RPE of Cyp4v3 KO mice. Transcriptome analysis of RPE in Cyp4v3 KO mice revealed changes in genes involved in iron homeostasis, particularly an upregulation of NCOA4, which was confirmed by immunofluorescence. Ferroptosis-related characteristics, including mitochondrial defects, lipid peroxidation, ROS accumulation, and upregulation of related genes, were detected in the RPE both in vitro and in vivo. Abnormal accumulation of ferrous iron was also detected. DFP, an iron chelator administration suppressed ferroptosis phenotype in CYP4V2 mutated RPE. Oral administration of DFP also restored the retinal function and morphology in Cyp4v3 KO mice. CONCLUSION: This study represented the first evidence of the substantial role of ferroptosis in the development of BCD. PUFAs resulting from CYP4V2 mutation may serve as substrates for ferroptosis, potentially working in conjunction with NCOA4-regulated iron accumulation, ultimately leading to RPE degeneration. DFP administration, which chelates iron, has demonstrated its ability to reverse BCD phenotype both in vitro and in vivo, suggesting a promising therapeutic approach in the future.


Subject(s)
Corneal Dystrophies, Hereditary , Ferroptosis , Mice, Knockout , Retinal Pigment Epithelium , Animals , Ferroptosis/genetics , Ferroptosis/drug effects , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/pathology , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/drug therapy , Humans , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/drug effects , Mice , Reactive Oxygen Species/metabolism , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Diseases/metabolism , Retinal Diseases/drug therapy , Cytochrome P450 Family 4/genetics , Mice, Inbred C57BL , Cell Line , Lipid Peroxidation/drug effects
17.
Cells ; 13(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38994953

ABSTRACT

The retinal pigment epithelium (RPE) is an essential component of the retina that plays multiple roles required to support visual function. These include light onset- and circadian rhythm-dependent tasks, such as daily phagocytosis of photoreceptor outer segments. Mitochondria provide energy to the highly specialized and energy-dependent RPE. In this study, we examined the positioning of mitochondria and how this is influenced by the onset of light. We identified a population of mitochondria that are tethered to the basal plasma membrane pre- and post-light onset. Following light onset, mitochondria redistributed apically and interacted with melanosomes and phagosomes. In a choroideremia mouse model that has regions of the RPE with disrupted or lost infolding of the plasma membrane, the positionings of only the non-tethered mitochondria were affected. This provides evidence that the tethering of mitochondria to the plasma membrane plays an important role that is maintained under these disease conditions. Our work shows that there are subpopulations of RPE mitochondria based on their positioning after light onset. It is likely they play distinct roles in the RPE that are needed to fulfil the changing cellular demands throughout the day.


Subject(s)
Cell Membrane , Light , Mitochondria , Retinal Pigment Epithelium , Retinal Pigment Epithelium/metabolism , Animals , Mitochondria/metabolism , Mice , Cell Membrane/metabolism , Mice, Inbred C57BL , Melanosomes/metabolism , Circadian Rhythm/physiology , Phagosomes/metabolism
18.
Sci Rep ; 14(1): 16322, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009704

ABSTRACT

Age-related macular degeneration (AMD) is one of the leading causes of blindness. AMD is currently incurable; the best solution is to prevent its occurrence. To develop drugs for AMD, it is crucial to have a model system that mimics the symptoms and mechanisms in patients. It is most important to develop safer and more effective anti-AMD drug. In this study, the dose of A2E and the intensity of blue light were evaluated to establish an appropriate atrophic in vitro model of AMD and anti-AMD effect and therapeutic mechanism of Codonopsis lanceolata. The experimental groups included a control group an AMD group treated with A2E and blue light, a lutein group treated with 25 µM lutein after AMD induction, and three groups treated with different doses of C. lanceolata (10, 20, and 50 µg/mL) after AMD induction. Intrinsic apoptotic pathway (Bcl-2 family), anti-oxidative system (Keap1/Nrf2/HO-1 antioxidant response element), and anti-carbonyl effect (4-hydroxynonenal [4-HNE]) were evaluated using immunofluorescence, MTT, TUNEL, FACS, and western blotting analyses. A2E accumulation in the cytoplasm of ARPE-19 cells depending on the dose of A2E. Cell viability of ARPE-19 cells according to the dose of A2E and/or blue light intensity. The population of apoptotic or necrotic cells increased based on the A2E dose and blue light intensity. Codonopsis lanceolata dose-dependently prevented cell death which was induced by A2E and blue light. The antiapoptotic effect of that was caused by activating Keap1/Nrf2/HO-1 pathway, suppressing 4-HNE, and modulating Bcl-2 family proteins like increase of antiapoptotic proteins such as Bcl-2 and Bcl-XL and decrease of proapoptotic protein such as Bim. Based on these findings, 30 µM A2E and 20 mW/cm2 blue light on adult retinal pigment epithelium-19 cells was an appropriate condition for AMD model and C. lanceolata shows promise as an anti-AMD agent.


Subject(s)
Apoptosis , Codonopsis , Macular Degeneration , NF-E2-Related Factor 2 , Oxidative Stress , Codonopsis/chemistry , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Oxidative Stress/drug effects , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Antioxidants/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Cell Line , Aldehydes/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Light/adverse effects , Proto-Oncogene Proteins c-bcl-2/metabolism
19.
Retina ; 44(8): 1394-1402, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39047130

ABSTRACT

PURPOSE: The aim of this study was to explore the potential benefits of retinal pigment epithelium replacement therapy in patients with Bietti crystalline dystrophy (BCD) by assessing the disease pathology with the distinctive relationship between fundus autofluorescence (FAF) abnormality and visual field defect. METHODS: Sixteen eyes from 16 patients with BCD and 16 eyes from 16 patients with RHO-associated retinitis pigmentosa were included. Fundus autofluorescence, optical coherence tomography, and Goldmann perimetry results were retrospectively reviewed and assessed using image analyses. RESULTS: In patients with BCD, the FAF abnormality area was not correlated with the overall visual field defect area and median overall visual field defect area (57.5%) was smaller than FAF abnormality area (98.5%). By contrast, the ellipsoid zone width was significantly correlated with the central visual field area (r = 0.806, P < 0.001). In patients with RHO-associated retinitis pigmentosa, the FAF abnormality area and ellipsoid zone width were significantly correlated with the overall visual field defect area (r = 0.833, P < 0.001) and central visual field area (r = 0.887, P < 0.001), respectively. CONCLUSION: The FAF abnormality shown in patients with BCD involves retinal pigment epithelium degeneration without complete loss of photoreceptors or visual function. These results suggest that patients with BCD are good candidates for retinal pigment epithelium replacement therapy for preservation of residual visual function.


Subject(s)
Corneal Dystrophies, Hereditary , Fluorescein Angiography , Fundus Oculi , Retinal Pigment Epithelium , Tomography, Optical Coherence , Visual Acuity , Visual Field Tests , Visual Fields , Humans , Visual Fields/physiology , Female , Male , Retrospective Studies , Middle Aged , Tomography, Optical Coherence/methods , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/physiopathology , Fluorescein Angiography/methods , Adult , Retinal Pigment Epithelium/pathology , Aged , Visual Acuity/physiology , Retinal Diseases/diagnosis , Retinal Diseases/physiopathology , Vision Disorders/physiopathology , Vision Disorders/diagnosis , Optical Imaging , Retinitis Pigmentosa/physiopathology , Retinitis Pigmentosa/diagnosis , Young Adult
20.
Retina ; 44(8): 1351-1359, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39047196

ABSTRACT

PURPOSE: In this study, differences in retinal feature visualization of high-resolution optical coherence tomography (OCT) devices were investigated with different axial resolutions in quantifications of retinal pigment epithelium and photoreceptors (PRs) in intermediate age-related macular degeneration. METHODS: Patients were imaged with standard SPECTRALIS HRA + OCT and the investigational High-Res OCT device (both by Heidelberg Engineering, Heidelberg, Germany). Drusen, retinal pigment epithelium, and PR layers were segmented using validated artificial intelligence-based algorithms followed by manual corrections. Thickness and drusen maps were computed for all patients. Loss and thickness measurements were compared between devices, drusen versus nondrusen areas, and early treatment diabetic retinopathy study subfields using mixed-effects models. RESULTS: Thirty-three eyes from 28 patients with intermediate age-related macular degeneration were included. Normalized PR integrity loss was significantly higher with 4.6% for standard OCT compared with 2.5% for High-Res OCT. The central and parafoveal PR integrity loss was larger than the perifoveal loss (P < 0.05). Photoreceptor thickness was increased on High-Res OCT and in nondrusen regions (P < 0.001). Retinal pigment epithelium appeared thicker on standard OCT and above drusen (P < 0.01). CONCLUSION: Our study shows that High-Res OCT is able to identify the condition of investigated layers in intermediate age-related macular degeneration with higher precision. This improved in vivo imaging technology might promote our understanding of the pathophysiology and progression of age-related macular degeneration.


Subject(s)
Retinal Pigment Epithelium , Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/diagnostic imaging , Female , Male , Aged , Aged, 80 and over , Photoreceptor Cells, Vertebrate/pathology , Visual Acuity/physiology , Retinal Drusen/diagnosis , Retinal Drusen/diagnostic imaging , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL