Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.076
Filter
1.
FASEB J ; 38(13): e23792, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38953555

ABSTRACT

Age-related macular degeneration (AMD) is a common cause of vision loss. The aggressive form of AMD is associated with ocular neovascularization and subretinal fibrosis, representing a responsive outcome against neovascularization mediated by epithelial-mesenchymal transition of retinal pigment epithelium (RPE) cells. A failure of the current treatment (anti-vascular endothelial growth factor therapy) has also been attributed to the progression of subretinal fibrosis. Hypoxia-inducible factors (HIFs) increase gene expressions to promote fibrosis and neovascularization. HIFs act as a central pathway in the pathogenesis of AMD. HIF inhibitors may suppress ocular neovascularization. Nonetheless, further investigation is required to unravel the aspects of subretinal fibrosis. In this study, we used RPE-specific HIFs or von Hippel-Lindau (VHL, a regulator of HIFs) conditional knockout (cKO) mice, along with pharmacological HIF inhibitors, to demonstrate the suppression of subretinal fibrosis. Fibrosis was suppressed by treatments of HIF inhibitors, and similar suppressive effects were detected in RPE-specific Hif1a/Hif2a- and Hif1a-cKO mice. Promotive effects were observed in RPE-specific Vhl-cKO mice, where fibrosis-mediated pathologic processes were evident. Marine products' extracts and their component taurine suppressed fibrosis as HIF inhibitors. Our study shows critical roles of HIFs in the progression of fibrosis, linking them to the potential development of therapeutics for AMD.


Subject(s)
Fibrosis , Mice, Knockout , Retinal Pigment Epithelium , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Mice , Fibrosis/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/drug therapy , Retina/metabolism , Retina/pathology , Epithelial-Mesenchymal Transition/drug effects , Mice, Inbred C57BL
2.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38928111

ABSTRACT

Age-related macular degeneration (AMD) is strictly linked to chronic oxidative stress, inflammation, loss of epithelial barrier integrity, and often with abnormal new blood vessel development. In this study, the retinal epithelial cell line ARPE-19 was treated with pro-inflammatory transforming growth factor-beta (TGF-ß) to investigate the activity of vitamin D (VD) and sulforaphane (SF) in abating the consequences of oxidative stress and inflammation. The administration of VD and SF lowered reactive oxygen species (ROS) levels, and abated the related expression of the pro-inflammatory cytokines interleukin-6 and interleukin-8 induced by TGF-ß. We evaluated mitochondrial respiration as a source of ROS production, and we discovered that the increased transcription of respiratory elements triggered by TGF-ß was prevented by VD and SF. In this model of inflamed epithelium, the treatment with VD and SF also reduced the secretion of VEGF, a key angiogenic factor, and restored the markers of epithelial integrity. Remarkably, all the observed biological effects were potentiated by the co-stimulation with the two compounds and were not mediated by VD receptor expression but rather by the ERK 1/2 pathway. Altogether, the results of this study reveal the powerful synergistic anti-inflammatory activity of SF and VD and lay the foundation for future clinical assessment of their efficacy in AMD.


Subject(s)
Isothiocyanates , Macular Degeneration , Oxidative Stress , Reactive Oxygen Species , Sulfoxides , Vitamin D , Humans , Macular Degeneration/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Isothiocyanates/pharmacology , Oxidative Stress/drug effects , Sulfoxides/pharmacology , Vitamin D/pharmacology , Reactive Oxygen Species/metabolism , Cell Line , Vascular Endothelial Growth Factor A/metabolism , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Transforming Growth Factor beta/metabolism , Biomarkers/metabolism , Interleukin-8/metabolism
3.
Sci Rep ; 14(1): 13920, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886213

ABSTRACT

Age-related macular degeneration (AMD) is one of the major causes of blindness in the elderly worldwide. Anti-vascular endothelial growth factor (VEGF) drugs have been widely used to treat the neovascular type of AMD (nAMD). However, VEGF acts not only as a pro-angiogenic factor but also as an anti-apoptotic factor in the eyes. In this study, we found that anti-VEGF drugs, including bevacizumab (Bev), ranibizumab (Ran), and aflibercept (Afl), induced epithelial-mesenchymal transition (EMT) in ARPE-19 cells in vitro, accompanied by the induction of CCN2, a potent pro-fibrotic factor. Similarly, intravitreal injection of Afl into mouse eyes resulted in EMT in the retinal pigmented epithelium (RPE). Co-treatment with CCN5, an anti-fibrotic factor that down-regulates CCN2 expression, significantly attenuated the adverse effects of the anti-VEGF drugs both in vitro and in vivo. Inhibition of the VEGF signaling pathway with antagonists of VEGF receptors, SU5416 and ZM323881, induced EMT and up-regulated CCN2 in ARPE-19 cells. Additionally, knock-down of CCN2 with siRNA abolished the adverse effects of the anti-VEGF drugs in ARPE-19 cells. Collectively, these results suggest that anti-VEGF drugs induce EMT in RPE through the induction of CCN2 and that co-treatment with CCN5 attenuates the adverse effects of anti-VEGF drugs in mouse eyes.


Subject(s)
Epithelial-Mesenchymal Transition , Retinal Pigment Epithelium , Vascular Endothelial Growth Factor A , Epithelial-Mesenchymal Transition/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Animals , Humans , Mice , Vascular Endothelial Growth Factor A/metabolism , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/drug therapy , Macular Degeneration/chemically induced , Cell Line , Bevacizumab/pharmacology , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/genetics , Angiogenesis Inhibitors/pharmacology , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Repressor Proteins , Receptors, Vascular Endothelial Growth Factor
4.
Cell Death Dis ; 15(6): 385, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824126

ABSTRACT

Drusen, the yellow deposits under the retina, are composed of lipids and proteins, and represent a hallmark of age-related macular degeneration (AMD). Lipid droplets are also reported in the retinal pigment epithelium (RPE) from AMD donor eyes. However, the mechanisms underlying these disease phenotypes remain elusive. Previously, we showed that Pgc-1α repression, combined with a high-fat diet (HFD), induce drastic AMD-like phenotypes in mice. We also reported increased PGC-1α acetylation and subsequent deactivation in the RPE derived from AMD donor eyes. Here, through a series of in vivo and in vitro experiments, we sought to investigate the molecular mechanisms by which PGC-1α repression could influence RPE and retinal function. We show that PGC-1α plays an important role in RPE and retinal lipid metabolism and function. In mice, repression of Pgc-1α alone induced RPE and retinal degeneration and drusen-like deposits. In vitro inhibition of PGC1A by CRISPR-Cas9 gene editing in human RPE (ARPE19- PGC1A KO) affected the expression of genes responsible for lipid metabolism, fatty acid ß-oxidation (FAO), fatty acid transport, low-density lipoprotein (LDL) uptake, cholesterol esterification, cholesterol biosynthesis, and cholesterol efflux. Moreover, inhibition of PGC1A in RPE cells caused lipid droplet accumulation and lipid peroxidation. ARPE19-PGC1A KO cells also showed reduced mitochondrial biosynthesis, impaired mitochondrial dynamics and activity, reduced antioxidant enzymes, decreased mitochondrial membrane potential, loss of cardiolipin, and increased susceptibility to oxidative stress. Our data demonstrate the crucial role of PGC-1α in regulating lipid metabolism. They provide new insights into the mechanisms involved in lipid and drusen accumulation in the RPE and retina during aging and AMD, which may pave the way for developing novel therapeutic strategies targeting PGC-1α.


Subject(s)
Lipid Droplets , Lipid Metabolism , Macular Degeneration , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Retinal Pigment Epithelium , Retinal Pigment Epithelium/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Humans , Mice , Lipid Droplets/metabolism , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Male , Oxidative Stress
5.
Transl Vis Sci Technol ; 13(6): 7, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38874975

ABSTRACT

Purpose: The subsidence of the outer plexiform layer (OPL) is an important imaging biomarker on optical coherence tomography (OCT) associated with early outer retinal atrophy and a risk factor for progression to geographic atrophy in patients with intermediate age-related macular degeneration (AMD). Deep neural networks (DNNs) for OCT can support automated detection and localization of this biomarker. Methods: The method predicts potential OPL subsidence locations on retinal OCTs. A detection module (DM) infers bounding boxes around subsidences with a likelihood score, and a classification module (CM) assesses subsidence presence at the B-scan level. Overlapping boxes between B-scans are combined and scored by the product of the DM and CM predictions. The volume-wise score is the maximum prediction across all B-scans. One development and one independent external data set were used with 140 and 26 patients with AMD, respectively. Results: The system detected more than 85% of OPL subsidences with less than one false-positive (FP)/scan. The average area under the curve was 0.94 ± 0.03 for volume-level detection. Similar or better performance was achieved on the independent external data set. Conclusions: DNN systems can efficiently perform automated retinal layer subsidence detection in retinal OCT images. In particular, the proposed DNN system detects OPL subsidence with high sensitivity and a very limited number of FP detections. Translational Relevance: DNNs enable objective identification of early signs associated with high risk of progression to the atrophic late stage of AMD, ideally suited for screening and assessing the efficacy of the interventions aiming to slow disease progression.


Subject(s)
Macular Degeneration , Neural Networks, Computer , Tomography, Optical Coherence , Humans , Tomography, Optical Coherence/methods , Aged , Female , Male , Macular Degeneration/diagnostic imaging , Macular Degeneration/diagnosis , Macular Degeneration/pathology , Geographic Atrophy/diagnostic imaging , Geographic Atrophy/diagnosis , Disease Progression , Retina/diagnostic imaging , Retina/pathology , Middle Aged , Aged, 80 and over
6.
FASEB J ; 38(11): e23720, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38837708

ABSTRACT

Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed "disease-in-a-dish" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.


Subject(s)
ATP-Binding Cassette Transporters , Cathepsin D , Lysosomes , Retinal Pigment Epithelium , Stargardt Disease , Cathepsin D/metabolism , Cathepsin D/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Stargardt Disease/metabolism , Stargardt Disease/pathology , Stargardt Disease/genetics , Animals , Humans , Mice , Lysosomes/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/genetics , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics
7.
Mol Neurodegener ; 19(1): 49, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890703

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in elderly people in the developed world, and the number of people affected is expected to almost double by 2040. The retina presents one of the highest metabolic demands in our bodies that is partially or fully fulfilled by mitochondria in the neuroretina and retinal pigment epithelium (RPE), respectively. Together with its post-mitotic status and constant photooxidative damage from incoming light, the retina requires a tightly-regulated housekeeping system that involves autophagy. The natural polyphenol Urolithin A (UA) has shown neuroprotective benefits in several models of aging and age-associated disorders, mostly attributed to its ability to induce mitophagy and mitochondrial biogenesis. Sodium iodate (SI) administration recapitulates the late stages of AMD, including geographic atrophy and photoreceptor cell death. METHODS: A combination of in vitro, ex vivo and in vivo models were used to test the neuroprotective potential of UA in the SI model. Functional assays (OCT, ERGs), cellular analysis (flow cytometry, qPCR) and fine confocal microscopy (immunohistochemistry, tandem selective autophagy reporters) helped address this question. RESULTS: UA alleviated neurodegeneration and preserved visual function in SI-treated mice. Simultaneously, we observed severe proteostasis defects upon SI damage induction, including autophagosome accumulation, that were resolved in animals that received UA. Treatment with UA restored autophagic flux and triggered PINK1/Parkin-dependent mitophagy, as previously reported in the literature. Autophagy blockage caused by SI was caused by severe lysosomal membrane permeabilization. While UA did not induce lysosomal biogenesis, it did restore upcycling of permeabilized lysosomes through lysophagy. Knockdown of the lysophagy adaptor SQSTM1/p62 abrogated viability rescue by UA in SI-treated cells, exacerbated lysosomal defects and inhibited lysophagy. CONCLUSIONS: Collectively, these data highlight a novel putative application of UA in the treatment of AMD whereby it bypasses lysosomal defects by promoting p62-dependent lysophagy to sustain proteostasis.


Subject(s)
Coumarins , Animals , Mice , Coumarins/pharmacology , Autophagy/drug effects , Autophagy/physiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Retina/metabolism , Retina/drug effects , Retina/pathology , Mitophagy/drug effects , Mitophagy/physiology , Sequestosome-1 Protein/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Humans , Disease Models, Animal , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Iodates/toxicity
8.
Sci Rep ; 14(1): 13990, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38886462

ABSTRACT

In this retrospective case series on neovascular age-related macular degeneration (nAMD), we aimed to improve Choroidal Neovascularization (CNV) visualization in Optical Coherence Tomography Angiography (OCTA) scans by addressing segmentation errors. Out of 198 eyes, 73 OCTA scans required manual segmentation correction. We compared uncorrected scans to those with minimal (2 corrections), moderate (10 corrections), and detailed (50 corrections) efforts targeting falsely segmented Bruch's Membrane (BM). Results showed that 55% of corrected OCTAs exhibited improved quality after manual correction. Notably, minimal correction (2 scans) already led to significant improvements, with additional corrections (10 or 50) not further enhancing expert grading. Reduced background noise and improved CNV identification were observed, with the most substantial improvement after two corrections compared to baseline uncorrected images. In conclusion, our approach of correcting segmentation errors effectively enhances image quality in OCTA scans of nAMD. This study demonstrates the efficacy of the method, with 55% of resegmented OCTA images exhibiting enhanced quality, leading to a notable increase in the proportion of high-quality images from 63 to 83%.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Tomography, Optical Coherence , Humans , Choroidal Neovascularization/diagnostic imaging , Choroidal Neovascularization/pathology , Tomography, Optical Coherence/methods , Female , Male , Retrospective Studies , Aged , Macular Degeneration/diagnostic imaging , Macular Degeneration/pathology , Macular Degeneration/complications , Aged, 80 and over , Image Processing, Computer-Assisted/methods , Middle Aged , Fluorescein Angiography/methods
9.
Sci Transl Med ; 16(750): eadi4125, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38838135

ABSTRACT

Chronic inflammation is a constitutive component of many age-related diseases, including age-related macular degeneration (AMD). Here, we identified interleukin-1 receptor-associated kinase M (IRAK-M) as a key immunoregulator in retinal pigment epithelium (RPE) that declines during the aging process. Rare genetic variants of IRAK3, which encodes IRAK-M, were associated with an increased likelihood of developing AMD. In human samples and mouse models, IRAK-M abundance in the RPE declined with advancing age or exposure to oxidative stress and was further reduced in AMD. Irak3-knockout mice exhibited an increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M led to a disruption in RPE cell homeostasis, characterized by compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of adeno-associated virus (AAV)-expressing human IRAK3 rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in Irak3-knockout mice. Our data show that replenishment of IRAK-M in the RPE may redress dysregulated pro-inflammatory processes in AMD, suggesting a potential treatment for retinal degeneration.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Mice, Knockout , Oxidative Stress , Retinal Degeneration , Retinal Pigment Epithelium , Animals , Humans , Male , Mice , Cellular Senescence , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
10.
Stem Cell Res ; 78: 103458, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870564

ABSTRACT

The Stargardt's Disease, Type 1 (STGD1) is associated with the loss of function mutations in ABCA4. This gene codes for a retina-specific, ATP-binding cassette (ABC) family transporter, involved in the transport of the key visual cycle intermediate, all-trans-retinaldehyde (atRAL), across the photoreceptor cell membranes. Here, we report the establishment of a patient-specific, iPSC line (LVPEIi008-A), that carries a homozygous nonsense mutation at (c.6088C > T) position, within exon 44 of ABCA4. The patient-specific skin fibroblasts were reprogrammed using episomal plasmids and the stably expanding iPSC line expressed the key stemness and pluripotency markers, maintained its chromosomal integrity and tested negative for mycoplasma.


Subject(s)
ATP-Binding Cassette Transporters , Codon, Nonsense , Exons , Induced Pluripotent Stem Cells , Stargardt Disease , Induced Pluripotent Stem Cells/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Stargardt Disease/pathology , Humans , Homozygote , Cell Line , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/metabolism
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167239, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38750770

ABSTRACT

Fibulin-3 (FBLN3, aka EFEMP1) is a secreted extracellular matrix (ECM) glycoprotein implicated in ocular diseases including glaucoma and age-related macular degeneration. Yet surprisingly, little is known about its native biology, expression patterns, and localization in the eye. To overcome these shortcomings, we conducted gene expression analysis and immunohistochemistry for FBLN3 in ocular tissues from mice, pigs, non-human primates, and humans. Moreover, we evaluated age-related changes in FBLN3 and FBLN3-related ECM remodeling enzymes/inhibitors in aging mice. We found that FBLN3 displayed distinct staining patterns consistent across the mouse retina, particularly in the ganglion cell layer and inner nuclear layer (INL). In contrast, human retinas exhibited a unique staining pattern, with enrichment of FBLN3 in the retinal pigment epithelium (RPE), INL, and outer nuclear layer (ONL) in the peripheral retina. This staining transitioned to the outer plexiform layer (OPL) in the central retina/macula, and was accompanied by reduced RPE immunoreactivity approaching the fovea. Surprisingly, we found significant age-related increases in FBLN3 expression and protein abundance in the mouse retina which was paralleled by reduced transcript levels of FBLN3-degrading enzymes (i.e., Mmp2 and Htra1). Our findings highlight important species-dependent, retinal region-specific, and age-related expression and localization patterns of FBLN3 which favor its accumulation during aging. These findings contribute to a better understanding of FBLN3's role in ocular pathology and provide valuable insights for future FBLN3 research.


Subject(s)
Aging , Extracellular Matrix Proteins , Animals , Humans , Mice , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Aging/metabolism , Aging/genetics , Retina/metabolism , Swine , Male , Mice, Inbred C57BL , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Female , Species Specificity , Aged
12.
Aging (Albany NY) ; 16(9): 8044-8069, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38742956

ABSTRACT

Age-related macular degeneration (AMD) is a condition causing progressive central vision loss. Growing evidence suggests a link between cellular senescence and AMD. However, the exact mechanism by which cellular senescence leads to AMD remains unclear. Employing machine learning, we established an AMD diagnostic model. Through unsupervised clustering, two distinct AMD subtypes were identified. GO, KEGG, and GSVA analyses explored the diverse biological functions associated with the two subtypes. By WGCNA, we constructed a coexpression network of differential genes between the subtypes, revealing the regulatory role of hub genes at the level of transcription factors and miRNAs. We identified 5 genes associated with inflammation for the construction of the AMD diagnostic model. Additionally, we observed that the level of cellular senescence and pathways related to programmed cell death (PCD), such as ferroptosis, necroptosis, and pyroptosis, exhibited higher expression levels in subtype B than A. Immune microenvironments also differed between the subtypes, indicating potentially distinct pathogenic mechanisms and therapeutic targets. In summary, by leveraging cellular senescence-associated gene expression, we developed an AMD diagnostic model. Furthermore, we identified two subtypes with varying expression patterns of senescence genes, revealing their differential roles in programmed cell death, disease progression, and immune microenvironments within AMD.


Subject(s)
Cellular Senescence , Computational Biology , Macular Degeneration , Cellular Senescence/genetics , Macular Degeneration/genetics , Macular Degeneration/diagnosis , Macular Degeneration/pathology , Humans , Gene Regulatory Networks , Gene Expression Profiling , Machine Learning , MicroRNAs/genetics , MicroRNAs/metabolism
13.
Sci Rep ; 14(1): 12145, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802406

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of blindness worldwide, with a complex pathophysiology and phenotypic diversity. Here, we apply Similarity Network Fusion (SNF) to cluster AMD patients into putative metabolomics-derived endotypes. Using a discovery cohort of 163 AMD patients from Boston, US, and a validation cohort of 214 patients from Coimbra, Portugal, we identified four distinct metabolomics-derived endotypes with varying retinal structural and functional characteristics, confirmed across both cohorts. Patients clustered into Endotype 1 exhibited a milder form of AMD and were characterized by low levels of amino acids in specific metabolic pathways. Meanwhile, patients clustered into both Endotype 3 and 4 were associated with more severe AMD and exhibited low levels of fatty acid metabolites and elevated levels of sphingomyelins and fatty acid metabolites, respectively. These preliminary findings indicate that metabolomics-derived endotyping may offer a refined strategy for categorizing AMD patients based on their specific pathophysiological underpinnings, rather than relying solely on traditional observational clinical indicators.


Subject(s)
Macular Degeneration , Metabolomics , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Metabolomics/methods , Male , Female , Aged , Aged, 80 and over , Cohort Studies , Portugal , Middle Aged , Metabolome
14.
Sci Rep ; 14(1): 11681, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778065

ABSTRACT

This multicentre retrospective study evaluated the 1-year outcomes and safety profile of faricimab in treatment-naïve patients with neovascular age-related macular degeneration (nAMD). Fifty-five patients (57 eyes) underwent loading therapy comprising three monthly faricimab injections. If dryness was achieved by the third month, subsequent treat-and-extend (TAE) follow-up continued at a minimum 8-week interval thereafter. If wet macula persisted at the third month, a fourth dose was administered, followed by the TAE regimen. After 1 year, improvements in visual acuity (0.44 ± 0.46 [baseline] to 0.34 ± 0.48; p < 0.01) and central foveal thickness (326 ± 149 [baseline] to 195 ± 82 µm; p < 0.0001) were significant. Dry macula, characterised by the absence of intraretinal or subretinal fluid, was achieved in 65% of cases. Treatment intervals varied, ranging from 8 to 16 weeks, with 44% of eyes extending to a 16-week interval, followed by 33% at 8 weeks, 16% at 12 weeks, 5% at 14 weeks, and 2% at 10 weeks. Notably, 50% of the polypoidal choroidal vasculopathy patients exhibited complete regression of polypoidal lesions between 12 and 15 months. Faricimab treatment in nAMD patients induced significant improvements in central vision and retinal morphology. Two cases of retinal pigment epithelial tears and one case of iritis were reported as ocular complications.


Subject(s)
Visual Acuity , Humans , Male , Female , Aged , Japan , Retrospective Studies , Aged, 80 and over , Visual Acuity/drug effects , Treatment Outcome , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/administration & dosage , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Intravitreal Injections , Middle Aged , Tomography, Optical Coherence
15.
Immunohorizons ; 8(5): 363-370, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38775688

ABSTRACT

Although the pathogenesis of choroidal neovascularization (CNV) is largely unknown in age-related macular degeneration (AMD), inflammasomes may contribute to CNV development and progression. To understand the role NLRP3 inflammasomes in CNV, we used Ccr2RFPCx3cr1GFP dual-reporter mice and immunostaining techniques to confirm localization of NLRP3 inflammasomes in the laser-induced CNV (LCNV) lesions. Confocal microscopy was used to image and quantify LCNV volumes. MCC950 was used as NLRP3 inhibitor. ELISA and quantitative RT-PCR were used to confirm the activation of NLRP3 by monitoring the expression of IL-1ß protein and mRNA in choroidal tissues from LCNV mice. In addition, NLRP3 (-/-) LCNV mice were used to investigate whether NLRP3 inflammasomes contribute to the development of LCNV lesions. We observed that red fluorescent protein (RFP)-positive monocyte-derived macrophages and GFP-positive microglia-derived macrophages, in addition to other cell types, were localized in LCNV lesions at day 7 post-laser injury. In addition, NLRP3 inflammasomes are associated with LCNV lesions. Inhibition of NLRP3 inflammasomes, using MCC950, caused an increased Ccr2RFP-positive macrophages, Cx3cr1GFP-positive microglia, and other cells, resulting in an increase in total lesion size. NLRP3 (-/-) LCNV mice showed significantly increased lesion size compared with age-matched controls. Inhibition of NLRP3 resulted in decreased IL-1ß mRNA and protein expression in the choroidal tissues, suggesting that increased lesion size may not be directly related to IL-1ß.


Subject(s)
Choroidal Neovascularization , Indenes , Inflammasomes , Interleukin-1beta , Microglia , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Mice , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Monocytes/metabolism , Mice, Knockout , Sulfones/pharmacology , Mice, Inbred C57BL , Furans/pharmacology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Macrophages/metabolism , Macrophages/immunology , Sulfonamides/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Carrier Proteins/metabolism , Carrier Proteins/genetics , Choroid/metabolism , Choroid/pathology , Disease Models, Animal , Lasers/adverse effects , Macular Degeneration/pathology , Macular Degeneration/metabolism , Macular Degeneration/genetics
16.
Hum Cell ; 37(4): 1056-1069, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744794

ABSTRACT

Epithelial mesenchymal transition (EMT) occurring in retinal pigment epithelial cells (RPE) is a crucial mechanism that contributes to the development of age-related macular degeneration (AMD), a pivotal factor leading to permanent vision impairment. Long non-coding RNAs (lncRNAs) have emerged as critical regulators orchestrating EMT in RPE cells. In this study, we explored the function of the lncRNA CYTOR (cytoskeleton regulator RNA) in EMT of RPE cells and its underlying mechanisms. Through weighted correlation network analysis, we identified CYTOR as an EMT-related lncRNA associated with AMD. Experimental validation revealed that CYTOR orchestrates TGF-ß1-induced EMT, as well as proliferation and migration of ARPE-19 cells. Further investigation demonstrated the involvement of CYTOR in regulating the WNT5A/NFAT1 pathway and NFAT1 intranuclear translocation in the ARPE-19 cell EMT model. Mechanistically, CHIP, EMSA and dual luciferase reporter assays confirmed NFAT1's direct binding to CYTOR's promoter, promoting transcription. Reciprocally, CYTOR overexpression promoted NFAT1 expression, while NFAT1 overexpression increased CYTOR transcription. These findings highlight a mutual promotion between CYTOR and NFAT1, forming a positive feedback loop that triggers the EMT phenotype in ARPE-19 cells. These discoveries provide valuable insights into the molecular mechanisms of EMT and its association with AMD, offering potential avenues for targeted therapies in EMT-related conditions, including AMD.


Subject(s)
Epithelial-Mesenchymal Transition , Feedback, Physiological , Macular Degeneration , NFATC Transcription Factors , RNA, Long Noncoding , Retinal Pigment Epithelium , Epithelial-Mesenchymal Transition/genetics , Humans , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , RNA, Long Noncoding/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/etiology , Gene Expression/genetics , Cell Proliferation/genetics , Cell Movement/genetics , Transforming Growth Factor beta1/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Epithelial Cells/metabolism , Cell Line , Cells, Cultured
17.
Neurobiol Aging ; 140: 41-59, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38723422

ABSTRACT

Aging is the main risk factor for age-related macular degeneration (AMD), a retinal neurodegenerative disease that leads to irreversible blindness, particularly in people over 60 years old. Retinal pigmented epithelium (RPE) atrophy is an AMD hallmark. Genome-wide chromatin accessibility, DNA methylation, and gene expression studies of AMD and control RPE demonstrate epigenomic/transcriptomic changes occur during AMD onset and progression. However, mechanisms by which molecular alterations of normal aging impair RPE function and contribute to AMD pathogenesis are unclear. Here, we specifically interrogate the RPE translatome with advanced age and across sexes in a novel RPE reporter mouse model. We find differential age- and sex- associated transcript expression with overrepresentation of pathways related to inflammation in the RPE. Concordant with impaired RPE function, the phenotypic changes in the aged translatome suggest that aged RPE becomes immunologically active, in both males and females, with some sex-specific signatures, which supports the need for sex representation for in vivo studies.


Subject(s)
Aging , Macular Degeneration , Retinal Pigment Epithelium , Sex Characteristics , Animals , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Female , Male , Aging/genetics , Aging/physiology , Aging/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/etiology , Transcriptome , Disease Models, Animal , Gene Expression , Inflammation , Mice , Mice, Inbred C57BL
18.
Front Immunol ; 15: 1366841, 2024.
Article in English | MEDLINE | ID: mdl-38711521

ABSTRACT

Introduction: Age-related macular degeneration (AMD) is a prevalent, chronic and progressive retinal degenerative disease characterized by an inflammatory response mediated by activated microglia accumulating in the retina. In this study, we demonstrate the therapeutically effects and the underlying mechanisms of microglial repopulation in the laser-induced choroidal neovascularization (CNV) model of exudative AMD. Methods: The CSF1R inhibitor PLX3397 was used to establish a treatment paradigm for microglial repopulation in the retina. Neovascular leakage and neovascular area were examined by fundus fluorescein angiography (FFA) and immunostaining of whole-mount RPE-choroid-sclera complexes in CNV mice receiving PLX3397. Altered cellular senescence was measured by beta-galactosidase (SA-ß-gal) activity and p16INK4a expression. The effect and mechanisms of repopulated microglia on leukocyte infiltration and the inflammatory response in CNV lesions were analyzed. Results: We showed that ten days of the CSF1R inhibitor PLX3397 treatment followed by 11 days of drug withdrawal was sufficient to stimulate rapid repopulation of the retina with new microglia. Microglial repopulation attenuated pathological choroid neovascularization and dampened cellular senescence in CNV lesions. Repopulating microglia exhibited lower levels of activation markers, enhanced phagocytic function and produced fewer cytokines involved in the immune response, thereby ameliorating leukocyte infiltration and attenuating the inflammatory response in CNV lesions. Discussion: The microglial repopulation described herein are therefore a promising strategy for restricting inflammation and choroidal neovascularization, which are important players in the pathophysiology of AMD.


Subject(s)
Aminopyridines , Choroidal Neovascularization , Disease Models, Animal , Microglia , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Microglia/metabolism , Microglia/drug effects , Mice , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Mice, Inbred C57BL , Macular Degeneration/pathology , Macular Degeneration/metabolism , Macular Degeneration/drug therapy , Inflammation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Pyrroles/pharmacology , Pyrroles/therapeutic use , Cellular Senescence/drug effects
19.
Nat Commun ; 15(1): 3780, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710714

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) have emerged as promising gene therapy vectors due to their proven efficacy and safety in clinical applications. In non-human primates (NHPs), rAAVs are administered via suprachoroidal injection at a higher dose. However, high doses of rAAVs tend to increase additional safety risks. Here, we present a novel AAV capsid (AAVv128), which exhibits significantly enhanced transduction efficiency for photoreceptors and retinal pigment epithelial (RPE) cells, along with a broader distribution across the layers of retinal tissues in different animal models (mice, rabbits, and NHPs) following intraocular injection. Notably, the suprachoroidal delivery of AAVv128-anti-VEGF vector completely suppresses the Grade IV lesions in a laser-induced choroidal neovascularization (CNV) NHP model for neovascular age-related macular degeneration (nAMD). Furthermore, cryo-EM analysis at 2.1 Å resolution reveals that the critical residues of AAVv128 exhibit a more robust advantage in AAV binding, the nuclear uptake and endosome escaping. Collectively, our findings highlight the potential of AAVv128 as a next generation ocular gene therapy vector, particularly using the suprachoroidal delivery route.


Subject(s)
Choroidal Neovascularization , Dependovirus , Genetic Therapy , Genetic Vectors , Retinal Pigment Epithelium , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Genetic Therapy/methods , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Choroidal Neovascularization/therapy , Choroidal Neovascularization/genetics , Rabbits , Humans , Gene Transfer Techniques , Macular Degeneration/therapy , Macular Degeneration/genetics , Macular Degeneration/pathology , Disease Models, Animal , Capsid Proteins/genetics , Capsid Proteins/metabolism , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Mice, Inbred C57BL , Retina/metabolism , Retina/virology , Male , HEK293 Cells
20.
J Clin Invest ; 134(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38690727

ABSTRACT

Careful regulation of the complement system is critical for enabling complement proteins to titrate immune defense while also preventing collateral tissue damage from poorly controlled inflammation. In the eye, this balance between complement activity and inhibition is crucial, as a low level of basal complement activity is necessary to support ocular immune privilege, a prerequisite for maintaining vision. Dysregulated complement activation contributes to parainflammation, a low level of inflammation triggered by cellular damage that functions to reestablish homeostasis, or outright inflammation that disrupts the visual axis. Complement dysregulation has been implicated in many ocular diseases, including glaucoma, diabetic retinopathy, and age-related macular degeneration (AMD). In the last two decades, complement activity has been the focus of intense investigation in AMD pathogenesis, leading to the development of novel therapeutics for the treatment of atrophic AMD. This Review outlines recent advances and challenges, highlighting therapeutic approaches that have advanced to clinical trials, as well as providing a general overview of the complement system in the posterior segment of the eye and selected ocular diseases.


Subject(s)
Complement Activation , Complement System Proteins , Macular Degeneration , Humans , Macular Degeneration/immunology , Macular Degeneration/pathology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Complement Activation/immunology , Animals , Eye/immunology , Eye/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...