Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.310
Filter
1.
Vestn Oftalmol ; 140(4): 68-72, 2024.
Article in Russian | MEDLINE | ID: mdl-39254392

ABSTRACT

This article describes a clinical case of a female patient with choroidal nevus, who was previously diagnosed in another clinic with "subretinal neovascular membrane as a result of central serous chorioretinopathy" and subsequently underwent multiple intravitreal anti-VEGF injections. Based on the analysis of OCT angiography images, the macular changes in this case were interpreted as a polypoidal form of neovascularization in a patient with subfoveolar choroidal nevus.


Subject(s)
Angiogenesis Inhibitors , Choroid Neoplasms , Fluorescein Angiography , Intravitreal Injections , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A , Humans , Female , Choroid Neoplasms/diagnosis , Choroid Neoplasms/drug therapy , Fluorescein Angiography/methods , Tomography, Optical Coherence/methods , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Fundus Oculi , Choroid/blood supply , Choroid/diagnostic imaging , Choroid/pathology , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/etiology , Middle Aged , Nevus/diagnosis , Diagnosis, Differential , Treatment Outcome
2.
Retina ; 44(10): 1688-1695, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39287531

ABSTRACT

PURPOSE: To explore changes in reticular pseudodrusen (RPD) number and location after the development of macular neovascularization (MNV) in eyes with prior intermediate age-related macular degeneration, focusing on different retinal regions differently affected by MNV. METHODS: This retrospective longitudinal study included intermediate age-related macular degeneration eyes with RPD that developed MNV. Reticular pseudodrusen were assessed at baseline when MNV was diagnosed (MNV stage) and after anti-vascular endothelial growth factor treatment. Three regions of interest were considered: MNV area, subretinal fluid (SRF) area, and a marginal area of 1,000 µm around SRF (marginal zone). Reticular pseudodrusen counts were compared with age- and sex-matched control eyes with RPD that did not develop MNV. RESULTS: Reticular pseudodrusen number exhibited a significant decrease after MNV development in the MNV area (P = 0.048) and in the area with SRF (P = 0.078). A statistically significant decrease was also disclosed in the marginal area around SRF (P = 0.002), associated with larger SRF areas. Control eyes did not show any significant change in the RPD count. CONCLUSION: Reticular pseudodrusen reduction after MNV development suggests a complex interplay involving the MNV itself, the presence of SRF, and trophic changes. The results of this study highlight the role of MNV in retinal nutritional balance and provide intriguing results in the RPD life cycle.


Subject(s)
Angiogenesis Inhibitors , Fluorescein Angiography , Retinal Drusen , Retinal Neovascularization , Tomography, Optical Coherence , Humans , Retrospective Studies , Female , Retinal Drusen/diagnosis , Male , Aged , Tomography, Optical Coherence/methods , Fluorescein Angiography/methods , Aged, 80 and over , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Retinal Neovascularization/diagnosis , Retinal Neovascularization/etiology , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Intravitreal Injections , Visual Acuity , Follow-Up Studies , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/physiopathology , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/etiology , Wet Macular Degeneration/diagnosis , Wet Macular Degeneration/drug therapy , Fundus Oculi
3.
Invest Ophthalmol Vis Sci ; 65(11): 21, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39269368

ABSTRACT

Purpose: This study aims to investigate the relationship among STRA6, circadian rhythm, and choroidal neovascularization (CNV) formation, as well as the regulatory mechanism of STRA6 in CNV under circadian rhythm disturbances. Methods: C57BL/6J male mice (aged 6 weeks) were randomly divided into control and jet lag groups (using a time shift method every 4 days to disrupt the molecular clock's capacity to synchronize with a stable rhythm). A laser-induced CNV model was established in both the control and the jet lag group after 2 weeks of jet lag. The size of CNV lesions and vascular leakage were detected by morphological and imaging examination on the seventh day post laser. STRA6 was screened by full transcriptome sequencing. Bioinformatics analysis was conducted to assess the variation and association of STRA6 in the GSE29801 dataset. The effects of STRA6 were evaluated both in vivo and in vitro. The pathway mechanism was further elucidated and confirmed through immunofluorescence of paraffin sections and Western blotting. Results: The disturbance of circadian rhythm promotes the formation of CNV. Patients with age-related macular degeneration (AMD) exhibited higher levels of STRA6 expression compared to the control group, and STRA6 was enriched in pathways related to angiogenesis. In addition, CLOCK and BMAL1, which are initiators that drive the circadian cycle, had regulatory effects on STRA6. Knocking down STRA6 reversed the promotion of CNV formation caused by circadian rhythm disturbance in vivo, and it also affected the proliferation, migration, and VEGF secretion of RPE cells without circadian rhythm in vitro, as well as impacting endothelial cells. Through activation of the JAK2/STAT3/VEGFA signaling pathway in unsynchronized RPE cells, STRA6 promotes CNV formation. Conclusions: This study suggests that STRA6 reduces CNV production by inhibiting JAK2/STAT3 phosphorylation after circadian rhythm disturbance. The results suggest that STRA6 may be a new direction for the treatment of AMD.


Subject(s)
Choroidal Neovascularization , Circadian Rhythm , Disease Models, Animal , Mice, Inbred C57BL , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/genetics , Choroidal Neovascularization/physiopathology , Animals , Circadian Rhythm/physiology , Mice , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Gene Expression Regulation/physiology , Blotting, Western , Humans , Cell Proliferation/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
4.
Invest Ophthalmol Vis Sci ; 65(11): 32, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39302645

ABSTRACT

Purpose: To evaluate the response of type 1 and type 2 macular neovascularization (MNV) components under anti-vascular endothelial growth factor (VEGF) treatment in age-related macular degeneration (AMD) using projection-resolved optical coherence tomography angiography (PR-OCTA). Methods: This retrospective study included eyes with treatment-naïve exudative AMD treated with anti-VEGF injections under a pro re nata (PRN) protocol over 1 year. Two-dimensional MNV areas and three-dimensional MNV volumes were derived from macular PR-OCTA scans using an automated convolutional neural network. MNV was detected as flow signal within the outer retinal slab. Type 1 components and type 2 components were analyzed separately. Results: Of 17 enrolled eyes, 12 eyes were pure type 1 MNV and five eyes were type 2 MNV. In eyes with pure type 1, the total (sum of type 1 and type 2 components) MNV area and volume did not change from baseline to 6 months or 12 months (P > 0.05). In eyes with type 2 MNV, the total MNV area significantly decreased from the baseline to 6 months (P = 0.0074) and 12 months (P = 0.014). The total type 2 MNV volume also decreased from baseline visit to visits at 6 months and at 12 months, nearing statistical signifiicance (P = 0.061 and P = 0.074). In eyes with type 2 MNV, the type 1 component increased from 0.093 mm2 to 0.30 mm2 (P = 0.058), and the type 2 component decreased from 0.37 mm2 at 6 months to 0 at 12 months (P = 0.0087). Conclusions: Type 1 and type 2 MNV may have different response under PRN anti-VEGF treatment over 1 year.


Subject(s)
Angiogenesis Inhibitors , Choroidal Neovascularization , Fluorescein Angiography , Intravitreal Injections , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A , Humans , Tomography, Optical Coherence/methods , Retrospective Studies , Male , Female , Fluorescein Angiography/methods , Angiogenesis Inhibitors/therapeutic use , Aged , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/diagnostic imaging , Choroidal Neovascularization/diagnosis , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aged, 80 and over , Visual Acuity , Ranibizumab/therapeutic use , Ranibizumab/administration & dosage , Wet Macular Degeneration/drug therapy , Wet Macular Degeneration/diagnosis , Fundus Oculi , Follow-Up Studies , Undertreatment
5.
Ann Med ; 56(1): 2393273, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39189520

ABSTRACT

BACKGROUND: Polypoidal choroidal vasculopathy (PCV) is a hemorrhagic fundus disease that can lead to permanent vision loss. Predicting the treatment response to anti-VEGF monotherapy in PCV is consistently challenging. We aimed to conduct a prospective multicenter study to explore and identify the imaging biomarkers for predicting the anti-VEGF treatment response in PCV patients, establish predictive model, and undergo multicenter validation. METHODS: This prospective multicenter study utilized clinical characteristics and images of treatment naïve PCV patients from 15 ophthalmic centers nationwide to screen biomarkers, develop model, and validate its performance. Patients from Peking Union Medical College Hospital were randomly divided into a training set and an internal validation set. A nomogram was established by univariate, LASSO regression, and multivariate regression analysis. Patients from the other 14 centers served as an external test set. Area under the curve (AUC), sensitivity, specificity, and accuracy were calculated. Decision curve analysis (DCA) and clinical impact curve (CIC) were utilized to evaluate the practical utility in clinical decision-making. FINDINGS: The eye distribution for the training set, internal validation set, and external test set were 66, 31, and 71, respectively. The 'Good responder' exhibited a thinner subfoveal choroidal thickness (SFCT) (230.67 ± 61.96 vs. 314.42 ± 88.00 µm, p < 0.001), lower choroidal vascularity index (CVI) (0.31 ± 0.08 vs. 0.36 ± 0.05, p = 0.006), fewer choroidal vascular hyperpermeability (CVH) (31.0 vs. 62.2%, p = 0.012), and more intraretinal fluid (IRF) (58.6 vs. 29.7%, p = 0.018). SFCT (OR 0.990; 95% CI 0.981-0.999; p = 0.033) and CVI (OR 0.844; 95% CI 0.732-0.971; p = 0.018) were ultimately included as the optimal predictive biomarkers and presented in the form of a nomogram. The model demonstrated AUC of 0.837 (95% CI 0.738-0.936), 0.891 (95% CI 0.765-1.000), and 0.901 (95% CI 0.824-0.978) for predicting 'Good responder' in the training set, internal validation set, and external test set, respectively, with excellent sensitivity, specificity, and practical utility. INTERPRETATION: Thinner SFCT and lower CVI can serve as imaging biomarkers for predicting good treatment response to anti-VEGF monotherapy in PCV patients. The nomogram based on these biomarkers exhibited satisfactory performances.


Subject(s)
Angiogenesis Inhibitors , Biomarkers , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A , Humans , Male , Female , Prospective Studies , Aged , Middle Aged , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Choroid/blood supply , Choroid/diagnostic imaging , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/diagnostic imaging , Treatment Outcome , Nomograms , Polyps/drug therapy , Polyps/diagnostic imaging , Polyps/diagnosis , Fluorescein Angiography/methods , Choroid Diseases/drug therapy , Choroid Diseases/diagnostic imaging , Choroid Diseases/diagnosis , Polypoidal Choroidal Vasculopathy
6.
Exp Eye Res ; 247: 110057, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179168

ABSTRACT

Vascular endothelial growth factor (VEGF) signaling is crucial for choroidal neovascularization (CNV), a major pathological feature of neovascular age-related macular degeneration (nAMD). Gene transcription of VEGF is mainly regulated by hypoxia-inducible factor 1-alpha (HIF-1α). The chromobox (CBX) family polycomb protein (Pc) subgroup includes CBX2, CBX4, CBX6, CBX7, and CBX8. CBX4 enhances hypoxia-induced VEGF expression and angiogenesis in hepatocellular carcinoma (HCC) cells by increasing HIF-1α's transcriptional activity. The objective of the study was to examine the functions of members of the CBX family Pc subgroup in choroidal vascular endothelial cells (CVECs) during CNV. CBX4 and CBX7 expression was up-regulated in hypoxic human choroidal vascular endothelial cells (HCVECs). In HCVECs, CBX7 facilitated HIF-1α transcription and expression, while CBX4 did not. In HCVECs, CBX7 stimulated HIF-1α's nuclear translocation and transcriptional activity, which in turn stimulated VEGF transcription and expression. The CBX7/HIF-1α/VEGF pathway promoted the migration, proliferation, and tube formation of HCVECs. The CBX7/HIF-1α/VEGF pathway was up-regulated in CVECs and in the mouse model with laser-induced CNV. Mouse CNV was lessened by the blockade of CBX7 through the down-regulation of HIF-1α/VEGF. In conclusion, CBX7 enhanced pro-angiogenic behaviors of hypoxic CVECs by up-regulating the HIF-1α/VEGF pathway, which contributing to the formation of mouse laser-induced CNV.


Subject(s)
Choroid , Choroidal Neovascularization , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit , Mice, Inbred C57BL , Polycomb Repressive Complex 1 , Vascular Endothelial Growth Factor A , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/genetics , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Humans , Choroid/blood supply , Choroid/metabolism , Signal Transduction/physiology , Cells, Cultured , Blotting, Western , Cell Proliferation/physiology , Endothelial Cells/metabolism , Gene Expression Regulation , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Cell Movement , Real-Time Polymerase Chain Reaction
7.
Sci Transl Med ; 16(760): eadi6626, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39141700

ABSTRACT

Neovascular age-related macular degeneration (nvAMD) is the leading cause of blindness in the elderly population. Although it is known that nvAMD is associated with focal inflammation, understanding of the precise immune components governing this process remains limited. Here, we identified natural killer (NK) cells as a prominent lymphocyte population infiltrating the perivascular space of choroidal neovascularization (CNV) lesions in patients with nvAMD and in mouse models. Olink proteomic analysis and single-cell RNA sequencing combined with knockout studies demonstrated the involvement of C-C chemokine receptor 5 (CCR5) in NK cell recruitment and extravasation at the CNV sites of mice. Depletion of NK cells or inhibition of activating receptor NK group 2, member D (NKG2D) inhibited the formation of neutrophil extracellular traps, increased vascular leakage, and exacerbated pathological angiogenesis, indicating that NK cells restrain pathogenesis in this mouse model. Age is the strongest risk factor for AMD, and we show that NK cells from aged human donors exhibited a less cytotoxic phenotype. NK cells from old mice exhibited compromised protective effects in the CNV mouse model. In addition, interleukin-2 complex-mediated expansion of NK cells improved CNV formation in mice. Collectively, our study highlights NK cells as a potential therapeutic target for patients with nvAMD.


Subject(s)
Extracellular Traps , Killer Cells, Natural , Macular Degeneration , Animals , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macular Degeneration/pathology , Humans , Extracellular Traps/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Neutrophils/immunology , Male , Aged , Female
8.
Sci Rep ; 14(1): 19278, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164449

ABSTRACT

To investigate quantitative associations between AI-assessed disease activity and optical coherence tomography angiography (OCTA)-derived parameters in patients with neovascular age-related macular degeneration (nAMD) undergoing anti-VEGF therapy. OCTA and SD-OCT images obtained from multicenter, randomized study data were evaluated. A deep learning algorithm (RetInSight) was used to detect and quantify macular fluid on SD-OCT. Mixed effects models were applied to evaluate correlations between fluid volumes, macular neovascularization (MNV)-type and OCTA-derived MNV parameters; lesion size (LS) and vessel area (NVA). 230 patients were included. A significant positive correlation was observed between SRF and NVA (estimate = 199.8 nl/mm2, p = 0.023), while a non-significant but negative correlation was found between SRF and LS (estimate = - 71.3 nl/mm2, p = 0.126). The presence of Type I and Type II MNV was associated with significantly less intraretinal fluid (IRF) compared to Type III MNV (estimate type I:- 52.1 nl, p = 0.019; estimate type II:- 51.7 nl, p = 0.021). A significant correlation was observed between pigment epithelial detachment (PED) and the interaction between NVA and LS (estimate:28.97 nl/mm2; p = 0.012). Residual IRF at week 12 significantly correlated to baseline NVA (estimate:38.1 nl/mm2; p = 0.015) and LS (estimate:- 22.6 nl/mm2; p = 0.012). Fluid in different compartments demonstrated disparate associations with MNV OCTA features. While IRF at baseline was most pronounced in type III MNV, residual IRF was driven by neovascular MNV characteristics. Greater NVA in proportion to LS was associated with higher amounts of SRF and PED. The correlation between these parameters may represent MNV maturation and can be used as a biomarker for resolution of disease activity. AI-based OCT analysis allows for a deeper understanding of neovascular disease in AMD and the potential to adjust therapeutic strategies to optimize outcomes through precision medicine.


Subject(s)
Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Female , Male , Aged , Macular Degeneration/diagnostic imaging , Macular Degeneration/pathology , Aged, 80 and over , Artificial Intelligence , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/therapeutic use , Fluorescein Angiography/methods , Choroidal Neovascularization/diagnostic imaging , Choroidal Neovascularization/pathology , Wet Macular Degeneration/diagnostic imaging , Wet Macular Degeneration/drug therapy , Deep Learning
9.
BMC Ophthalmol ; 24(1): 359, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169293

ABSTRACT

PURPOSE: To investigate the incidence of and risk factors for failure of detection of active fellow-eye neovascularization on optical coherence tomography(OCT) crosshair scans in patients with unilateral neovascular age-related macular degeneration(AMD). METHODS: In this retrospective study, patients who experienced the development of active neovascularization in the fellow eye during the follow-up period were included(n = 75). Cases in which the neovascularization in the fellow eye could be identified solely through crosshair scans were defined as the crosshair scan detection group(n = 63). Cases in which the aforementioned findings could not be identified through crosshair scans but could be identified through raster scans were defined as the raster scan detection group(n = 12). The factors were compared between the two groups. Risk factors related to undetected neovascularization on crosshair scans were additionally identified. RESULTS: Active fellow-eye neovascularization, was not detected on OCT crosshair scans in 12 cases(16.0%) but was identified on raster scans in all cases. There was a significant difference in the proportion of neovascularization types between the crosshair scan detection group and the raster scan detection group(P = 0.023). Among the 35 fellow-eye neovascularization cases in patients with type 3 macular neovascularization(MNV), 10(28.6%) were not detected on crosshair scans. Multivariate analysis revealed a significantly higher risk for undetectable fellow-eye neovascularization on crosshair scans in patients with type 3 MNV than in those with typical neovascular AMD(P = 0.037,ß = 9.600). CONCLUSIONS: Our findings suggest the need for routine OCT raster scans during fellow-eye examinations in patients with unilateral neovascular AMD, particularly when the first-affected eye is diagnosed with type 3 MNV.


Subject(s)
Tomography, Optical Coherence , Wet Macular Degeneration , Humans , Tomography, Optical Coherence/methods , Retrospective Studies , Male , Female , Aged , Wet Macular Degeneration/diagnosis , Aged, 80 and over , Visual Acuity , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/diagnostic imaging , Early Diagnosis , Fluorescein Angiography/methods , Risk Factors , Follow-Up Studies , Middle Aged
10.
ACS Appl Bio Mater ; 7(8): 5496-5505, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39107259

ABSTRACT

8-arm PEG (polyethylene-glycol) is a highly promising nanoplatform due to its small size (<10 nm), ease-of-conjugation (many functionalized variants are readily available with "click-like" properties), biocompatibility, and optical inactivity. This study evaluates 8-arm PEG uptake into cells (in vitro) and localization and clearance in vasculature (in vivo) for targeting of choroidal neovascularization in mice, an animal model of macular degeneration. 8-arm PEG nanoparticles were labeled with fluorescein isothiocyanate (FITC) and functionalized in the absence or presence of pentameric Ar-Gly-Asp (RGD; 4 RGD motifs and a PGC linker), one of the most common peptide motifs used for active targeting. In vitro studies show that RGD-conjugated 8-arm PEG nanoparticles exhibit enhanced cellular uptake relative to non-RGD-conjugated control NPs at 34% ± 9%. Laser-induced choroidal neovascularization (CNV) was performed in a mouse model to measure 8-arm PEG localization and clearance to model macular degeneration lesions in vivo. It was determined that both RGD-conjugated and non-RGD-conjugated (nRGD) 8-arm PEG particles localized to CNV lesions, with a half-life around 24 h. In vivo experiments showed that RGD-conjugated nanoparticles exhibited enhanced localization by 15-20% relative to without RGD controls. Exhibiting a high rate of localization and fast clearance relative to larger nanoparticles, targeted 8-arm PEG nanoparticles with a conjugated RGD-peptide could be a promising modality for macular degeneration diagnosis and therapy.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Nanoparticles , Particle Size , Polyethylene Glycols , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/pathology , Choroidal Neovascularization/metabolism , Animals , Polyethylene Glycols/chemistry , Mice , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Nanoparticles/chemistry , Disease Models, Animal , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Humans , Oligopeptides/chemistry , Mice, Inbred C57BL
11.
Opt Lett ; 49(15): 4314-4317, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090922

ABSTRACT

The results of depth-resolved multi-contrast in vivo mouse choroidal imaging using a polarization-diversity optical coherence tomography (PD-OCT) system are presented. A selectively chosen depth of focus that was fine-tuned with a sensorless adaptive optics technique and a simple segmentation based on the degree of polarization uniformity signal visualizes the detailed features of a mouse choroid from the OCT angiography images. A comprehensive image analysis of the choroid revealed the distinctive pathological characteristics of the laser-induced choroidal neovascularization mouse.


Subject(s)
Choroid , Choroidal Neovascularization , Tomography, Optical Coherence , Animals , Tomography, Optical Coherence/methods , Choroid/diagnostic imaging , Mice , Choroidal Neovascularization/diagnostic imaging
12.
Front Immunol ; 15: 1421175, 2024.
Article in English | MEDLINE | ID: mdl-39091492

ABSTRACT

Age-related macular degeneration (AMD), a prevalent and progressive degenerative disease of the macula, is the leading cause of blindness in elderly individuals in developed countries. The advanced stages include neovascular AMD (nAMD), characterized by choroidal neovascularization (CNV), leading to subretinal fibrosis and permanent vision loss. Despite the efficacy of anti-vascular endothelial growth factor (VEGF) therapy in stabilizing or improving vision in nAMD, the development of subretinal fibrosis following CNV remains a significant concern. In this review, we explore multifaceted aspects of subretinal fibrosis in nAMD, focusing on its clinical manifestations, risk factors, and underlying pathophysiology. We also outline the potential sources of myofibroblast precursors and inflammatory mechanisms underlying their recruitment and transdifferentiation. Special attention is given to the potential role of mast cells in CNV and subretinal fibrosis, with a focus on putative mast cell mediators, tryptase and granzyme B. We summarize our findings on the role of GzmB in CNV and speculate how GzmB may be involved in the pathological transition from CNV to subretinal fibrosis in nAMD. Finally, we discuss the advantages and drawbacks of animal models of subretinal fibrosis and pinpoint potential therapeutic targets for subretinal fibrosis.


Subject(s)
Fibrosis , Granzymes , Macular Degeneration , Humans , Animals , Macular Degeneration/pathology , Macular Degeneration/metabolism , Macular Degeneration/etiology , Granzymes/metabolism , Retina/pathology , Retina/metabolism , Retina/immunology , Mast Cells/immunology , Mast Cells/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/metabolism
13.
ACS Nano ; 18(33): 22194-22207, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39116033

ABSTRACT

Monotherapy, especially the use of antibodies targeting vascular endothelial growth factor (VEGF), has shown limitations in treating choroidal neovascularization (CNV) since reactive oxygen species (ROS) also exacerbate CNV formation. Herein, we developed a combination therapy based on a DNA origami platform targeting multiple components of ocular neovascularization. Our study demonstrated that ocular neovascularization was markedly suppressed by intravitreal injection of a rectangular DNA origami sheet modified with VEGF aptamers (Ap) conjugated to an anti-VEGF antibody (aV) via matrix metalloproteinase (MMP)-cleavable peptide linkers in a mouse model of CNV. Typically, the DNA origami-based therapeutic platform selectively accumulates in neovascularization lesions owing to the dual-targeting ability of the aV and Ap, followed by the cleavage of the peptide linker by MMPs to release the antibody. Together, the released antibody and Ap inhibited VEGF activity. Moreover, the residual bare DNA origami could effectively scavenge ROS, reducing oxidative stress at CNV sites and thus maximizing the synergistic effects of inhibiting neovascularization.


Subject(s)
Choroidal Neovascularization , DNA , Vascular Endothelial Growth Factor A , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Animals , Mice , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/chemistry , DNA/chemistry , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/chemistry , Antibodies/chemistry
14.
Cell Death Dis ; 15(8): 582, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122684

ABSTRACT

Age-related macular degeneration (AMD) causes severe blindness in the elderly due to choroidal neovascularization (CNV), which results from the dysfunction of the retinal pigment epithelium (RPE). While normal RPE depends exclusively on mitochondrial oxidative phosphorylation for energy production, the inflammatory conditions associated with metabolic reprogramming of the RPE play a pivotal role in CNV. Although mitochondrial pyruvate dehydrogenase kinase (PDK) is a central node of energy metabolism, its role in the development of CNV in neovascular AMD has not been investigated. In the present study, we used a laser-induced CNV mouse model to evaluate the effects of Pdk4 gene ablation and treatment with pan-PDK or specific PDK4 inhibitors on fluorescein angiography and CNV lesion area. Among PDK isoforms, only PDK4 was upregulated in the RPE of laser-induced CNV mice, and Pdk4 gene ablation attenuated CNV. Next, we evaluated mitochondrial changes mediated by PDK1-4 inhibition using siRNA or PDK inhibitors in inflammatory cytokine mixture (ICM)-treated primary human RPE (hRPE) cells. PDK4 silencing only in ICM-treated hRPE cells restored mitochondrial respiration and reduced inflammatory cytokine secretion. Likewise, GM10395, a specific PDK4 inhibitor, restored oxidative phosphorylation and decreased ICM-induced upregulation of inflammatory cytokine secretion. In a laser-induced CNV mouse model, GM10395 significantly alleviated CNV. Taken together, we demonstrate that specific PDK4 inhibition could be a therapeutic strategy for neovascular AMD by preventing mitochondrial metabolic reprogramming in the RPE under inflammatory conditions.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Macular Degeneration , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Retinal Pigment Epithelium , Animals , Humans , Mice , Macular Degeneration/metabolism , Macular Degeneration/pathology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/drug therapy , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Metabolic Reprogramming
15.
Transl Vis Sci Technol ; 13(8): 24, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39136959

ABSTRACT

Purpose: Intravitreal injection of anti-VEGF antibodies remains the primary therapy for exudative age-related macular degeneration (exAMD), although its efficacy is limited. Previous research has demonstrated that both a loss-of-function mutation of srr and the intravenous injection of a serine racemase inhibitor, L-aspartic acid ß-hydroxamate (L-ABH), significantly inhibit laser-induced choroidal neovascularization (CNV) in mice. Given that L-ABH is a small molecule, this study investigated the effects of L-ABH administered via eye drops on CNV, aiming to develop a noninvasive treatment strategy for exAMD. Methods: CNV models in mice and rhesus macaques were established through laser photocoagulation. Seven monkeys were randomly assigned to receive either saline solution or L-ABH eye drops. Intraperitoneal or intravenous injection of fluorescein characterized CNV in both mice and monkeys. Fluorescein fundus angiography was used to assess leakage, whereas optical coherence tomography measured retinal thickness in the monkeys. Results: L-ABH eye drops significantly reduced fluorescein leakage in laser-injured mice (P < 0.001 compared to saline). In laser-injured rhesus macaques, the average percent changes in leakage areas treated with L-ABH were 2.5% ± 25.8% (P = 0.004) and 1.5% ± 75.7% (P = 0.023 compared to saline solution) on day 14 and day 28, respectively. However, L-ABH eye drops did not significantly affect the number of grade IV laser spots or retinal thickness, whereas bevacizumab did. Conclusions: This study demonstrates the potential efficacy of an SRR inhibitor in two animal models of laser-induced CNV. Translational Relevance: This represents the first investigation into the effects of topical delivery of an SRR inhibitor on CNV.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Fluorescein Angiography , Macaca mulatta , Mice, Inbred C57BL , Tomography, Optical Coherence , Animals , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/pathology , Mice , Racemases and Epimerases/antagonists & inhibitors , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Laser Coagulation/adverse effects , Ophthalmic Solutions , Male , Choroid/drug effects , Choroid/pathology , Choroid/diagnostic imaging , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use
16.
Ann Afr Med ; 23(4): 723-726, 2024 Oct 01.
Article in French, English | MEDLINE | ID: mdl-39138925

ABSTRACT

Stargardt disease (SD) is a common inherited macular dystrophy. It exhibits a high degree of phenotypic and genotypic heterogeneity. Yellow-white flecks are often found in the posterior pole in the early stages of the disease with a reduction in central vision from foveal atrophy as it progresses. A characteristic dark choroid appearance is seen on fundus fluorescein angiography (FFA) in many cases, with occasional reports of choroidal neovascular membranes. We report a series of four Nigerian patients, with varied presentations diagnosed with SD in our facility. One patient had good vision, while the other three had variable degrees of reduced vision. All patients had macular atrophy and flecks, while three patients had a dark choroid appearance on FFA and one patient developed a choroidal neovascular membrane in one eye.


RésuméLa maladie de Stargardt (SD) est une dystrophie maculaire héréditaire courante. La maladie de Stargardt (SD) est une dystrophie maculaire héréditaire courante. Il présente un haut degré d'hétérogénéité phénotypique et génotypique. Il présente un haut degré d'hétérogénéité phénotypique et génotypique. Des taches jaune-blanc sont souvent trouvées dans le pôle postérieur aux premiers stades de la maladie avec une réduction de la vision centrale due à l'atrophie fovéale au fur et à mesure de sa progression. au fur et à mesure de sa progression. Une apparence choroïde foncée caractéristique est observée sur l'angiographie à la fluorescéine du fond d'œil (AFL) dans de nombreux cas, avec des rapports de membranes néovasculaires choroïdiennes. Nous rapportons une série de quatre patients Nigérians, avec des présentations variées diagnostiquées avec SD chez notre établissement. Un patient avait une bonne vision, tandis que les trois autres avaient des degrés variables de vision réduite. Tous les patients présentaient une atrophie maculaire et des taches, tandis que trois patients avaient une apparence choroïde foncée sur FFA et qu'un patient a développé une membrane néovasculaire choroïdienne dans un œil.


Subject(s)
Fluorescein Angiography , Macular Degeneration , Stargardt Disease , Visual Acuity , Humans , Macular Degeneration/genetics , Macular Degeneration/congenital , Macular Degeneration/diagnosis , Fluorescein Angiography/methods , Stargardt Disease/genetics , Male , Female , Adult , Nigeria , Tomography, Optical Coherence , Middle Aged , Choroidal Neovascularization/diagnosis , Fundus Oculi , Choroid/pathology , Choroid/diagnostic imaging , Adolescent
17.
Front Immunol ; 15: 1421012, 2024.
Article in English | MEDLINE | ID: mdl-38979414

ABSTRACT

Objective: This study revealed a core regulator and common upstream mechanisms for the multifaceted pathological processes of age-related macular degeneration (AMD) and provided proof-of-concept for this new therapeutic target. Methods: Comprehensive gene expression analysis was performed using RNA sequencing of eye cup from old mice as well as laser-induced choroidal neovascularization (CNV) mouse model. Through integrative analysis and protein-protein interaction (PPI) analysis, common pathways and key transcription factor was identified simultaneously engaged in age-related retinal degeneration and CNV, the two typical pathological process of AMD. Subsequently, the expression changes of Spi1, the key regulator, as well as the alternation of the downstream mechanisms were validated in both models through qRT-PCR, Elisa, flow cytometry and immunofluorescence. Further, we assessed the impact of Spi1 knockdown in vitro and in vivo using gene intervention vectors carried by adeno-associated virus or lentivirus to test its potential as a therapeutic target. Results: Compared to corresponding controls, we found 1,939 and 1,319 genes differentially expressed in eye cups of old and CNV mice respectively. The integrative analysis identified a total of 275 overlapping DEGs, of which 150 genes were co-upregulated. PPI analysis verified a central transcription factor, SPI1. The significant upregulation of Spi1 expression was then validated in both models, accompanied by macrophage polarization towards the M1 phenotype. Finally, SPI1 suppression significantly inhibited M1 polarization of BMDMs and attenuated neovascularization in CNV mice. Conclusion: This study demonstrates that SPI1 exerts a pivotal role in AMD by regulation of macrophage polarization and innate immune response, offering promise as an innovative target for treating AMD.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Macrophages , Macular Degeneration , Trans-Activators , Animals , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Macrophages/immunology , Macrophages/metabolism , Choroidal Neovascularization/immunology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Mice, Inbred C57BL , Macrophage Activation/genetics , Humans , Gene Expression Profiling , Male
18.
Theranostics ; 14(9): 3509-3525, 2024.
Article in English | MEDLINE | ID: mdl-38948065

ABSTRACT

Rationale: Current treatments for ocular angiogenesis primarily focus on blocking the activity of vascular endothelial growth factor (VEGF), but unfavorable side effects and unsatisfactory efficacy remain issues. The identification of novel targets for anti-angiogenic treatment is still needed. Methods: We investigated the role of tsRNA-1599 in ocular angiogenesis using endothelial cells, a streptozotocin (STZ)-induced diabetic model, a laser-induced choroidal neovascularization model, and an oxygen-induced retinopathy model. CCK-8 assays, EdU assays, transwell assays, and matrigel assays were performed to assess the role of tsRNA-1599 in endothelial cells. Retinal digestion assays, Isolectin B4 (IB4) staining, and choroidal sprouting assays were conducted to evaluate the role of tsRNA-1599 in ocular angiogenesis. Transcriptomic analysis, metabolic analysis, RNA pull-down assays, and mass spectrometry were utilized to elucidate the mechanism underlying angiogenic effects mediated by tsRNA-1599. Results: tsRNA-1599 expression was up-regulated in experimental ocular angiogenesis models and endothelial cells in response to angiogenic stress. Silencing of tsRNA-1599 suppressed angiogenic effects in endothelial cells in vitro and inhibited pathological ocular angiogenesis in vivo. Mechanistically, tsRNA-1599 exhibited little effect on VEGF signaling but could cause reduced glycolysis and NAD+/NADH production in endothelial cells by regulating the expression of HK2 gene through interacting with YBX1, thus affecting endothelial effects. Conclusions: Targeting glycolytic reprogramming of endothelial cells by a tRNA-derived small RNA represents an exploitable therapeutic approach for ocular neovascular diseases.


Subject(s)
Choroidal Neovascularization , Endothelial Cells , Glycolysis , Animals , Glycolysis/drug effects , Mice , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Humans , Y-Box-Binding Protein 1/metabolism , Y-Box-Binding Protein 1/genetics , Angiogenesis Inhibitors/pharmacology , Hexokinase/metabolism , Hexokinase/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL , Male , Disease Models, Animal , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/genetics , Human Umbilical Vein Endothelial Cells , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism
19.
Invest Ophthalmol Vis Sci ; 65(8): 12, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38967942

ABSTRACT

Purpose: Recruitment and activation of inflammatory cells, such as retinal microglia/macrophages, in the subretinal space contribute significantly to the pathogenesis of age-related macular degeneration (AMD). This study aims to explore the functional role of vascular endothelial growth factor (VEGF-A), placental growth factor (PlGF) and VEGF-A/PlGF heterodimer in immune homeostasis and activation during pathological laser-induced choroidal neovascularization (CNV). Methods: To investigate these roles, we utilized the PlGF-DE knockin (KI) mouse model, which is the full functional knockout (KO) of PlGF. In this model, mice express a variant of PlGF, named PlGF-DE, that is unable to bind and activate VEGFR-1 but can still form heterodimer with VEGF-A. Results: Our findings demonstrate that, although there is no difference in healthy conditions, PlGF-DE-KI mice exhibit decreased microglia reactivity and reduced recruitment of both microglia and monocyte-macrophages, compared to wild-type mice during laser-induced CNV. This impairment is associated with a reduction in VEGF receptor 1 (VEGFR-1) phosphorylation in the retinae of PlGF-DE-KI mice compared to C57Bl6/J mice. Corroborating these data, intravitreal delivery of PlGF or VEGF-A/PlGF heterodimer in PlGF-DE-KI mice rescued the immune cell response at the early phase of CNV compared to VEGF-A delivery. Conclusions: In summary, our study suggests that targeting PlGF and the VEGF-A/PlGF heterodimer, thereby preventing VEGFR-1 activation, could represent a potential therapeutic approach for the management of inflammatory processes in diseases such as AMD.


Subject(s)
Choroidal Neovascularization , Microglia , Placenta Growth Factor , Vascular Endothelial Growth Factor A , Animals , Mice , Choroidal Neovascularization/metabolism , Disease Models, Animal , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Placenta Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
20.
Biomed Phys Eng Express ; 10(5)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38955139

ABSTRACT

The prevalence of vision impairment is increasing at an alarming rate. The goal of the study was to create an automated method that uses optical coherence tomography (OCT) to classify retinal disorders into four categories: choroidal neovascularization, diabetic macular edema, drusen, and normal cases. This study proposed a new framework that combines machine learning and deep learning-based techniques. The utilized classifiers were support vector machine (SVM), K-nearest neighbor (K-NN), decision tree (DT), and ensemble model (EM). A feature extractor, the InceptionV3 convolutional neural network, was also employed. The performance of the models was evaluated against nine criteria using a dataset of 18000 OCT images. For the SVM, K-NN, DT, and EM classifiers, the analysis exhibited state-of-the-art performance, with classification accuracies of 99.43%, 99.54%, 97.98%, and 99.31%, respectively. A promising methodology has been introduced for the automatic identification and classification of retinal disorders, leading to reduced human error and saved time.


Subject(s)
Algorithms , Artificial Intelligence , Neural Networks, Computer , Retinal Diseases , Support Vector Machine , Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Retinal Diseases/diagnosis , Retinal Diseases/diagnostic imaging , Deep Learning , Retina/diagnostic imaging , Retina/pathology , Decision Trees , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/diagnostic imaging , Machine Learning , Choroidal Neovascularization/diagnostic imaging , Choroidal Neovascularization/diagnosis , Macular Edema/diagnostic imaging , Macular Edema/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL