Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80.156
Filter
1.
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
2.
Sci Rep ; 14(1): 17909, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095380

ABSTRACT

The effect of diabetes mellitus (DM) on individual retinal layers remains incompletely understood. We evaluated the intra-retinal layer thickness alterations in 71 DM eyes with no diabetic retinopathy (DR), 90 with mild DR, and 63 with moderate DR without macular edema, using spectral-domain optical coherence tomography (SD-OCT) and the Iowa Reference Algorithm for automated retinal layer segmentation. The average thickness of 10 intra-retinal layers was then corrected for ocular magnification using axial length measurements, and pairwise comparisons were made using multivariable linear regression models adjusted for gender and race. In DM no DR eyes, significant thinning was evident in the ganglion cell layer (GCL; p < 0.001), inner nuclear layer (INL; p = 0.001), and retinal pigment epithelium (RPE; p = 0.014) compared to normal eyes. Additionally, mild DR eyes exhibited a thinner inner plexiform layer (IPL; p = 0.008) than DM no DR eyes. Conversely, moderate DR eyes displayed thickening in the INL, outer nuclear layer, IPL, and retinal nerve fiber layer (all p ≤ 0.002), with notably worse vision. These findings highlight distinctive patterns: early diabetic eyes experience thinning in specific retinal layers, while moderate DR eyes exhibit thickening of certain layers and slightly compromised visual acuity, despite the absence of macular edema. Understanding these structural changes is crucial for comprehending diabetic eye complications.


Subject(s)
Diabetic Retinopathy , Tomography, Optical Coherence , Tomography, Optical Coherence/methods , Humans , Male , Female , Diabetic Retinopathy/diagnostic imaging , Diabetic Retinopathy/pathology , Middle Aged , Aged , Retina/diagnostic imaging , Retina/pathology , Macular Edema/diagnostic imaging , Macular Edema/pathology , Macula Lutea/diagnostic imaging , Macula Lutea/pathology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/diagnostic imaging , Retinal Ganglion Cells/pathology
3.
PeerJ ; 12: e17786, 2024.
Article in English | MEDLINE | ID: mdl-39104365

ABSTRACT

Background: Chronic kidney disease (CKD) is a significant global health concern, emphasizing the necessity of early detection to facilitate prompt clinical intervention. Leveraging the unique ability of the retina to offer insights into systemic vascular health, it emerges as an interesting, non-invasive option for early CKD detection. Integrating this approach with existing invasive methods could provide a comprehensive understanding of patient health, enhancing diagnostic accuracy and treatment effectiveness. Objectives: The purpose of this review is to critically assess the potential of retinal imaging to serve as a diagnostic tool for CKD detection based on retinal vascular changes. The review tracks the evolution from conventional manual evaluations to the latest state-of-the-art in deep learning. Survey Methodology: A comprehensive examination of the literature was carried out, using targeted database searches and a three-step methodology for article evaluation: identification, screening, and inclusion based on Prisma guidelines. Priority was given to unique and new research concerning the detection of CKD with retinal imaging. A total of 70 publications from 457 that were initially discovered satisfied our inclusion criteria and were thus subjected to analysis. Out of the 70 studies included, 35 investigated the correlation between diabetic retinopathy and CKD, 23 centered on the detection of CKD via retinal imaging, and four attempted to automate the detection through the combination of artificial intelligence and retinal imaging. Results: Significant retinal features such as arteriolar narrowing, venular widening, specific retinopathy markers (like microaneurysms, hemorrhages, and exudates), and changes in arteriovenous ratio (AVR) have shown strong correlations with CKD progression. We also found that the combination of deep learning with retinal imaging for CKD detection could provide a very promising pathway. Accordingly, leveraging retinal imaging through this technique is expected to enhance the precision and prognostic capacity of the CKD detection system, offering a non-invasive diagnostic alternative that could transform patient care practices. Conclusion: In summary, retinal imaging holds high potential as a diagnostic tool for CKD because it is non-invasive, facilitates early detection through observable microvascular changes, offers predictive insights into renal health, and, when paired with deep learning algorithms, enhances the accuracy and effectiveness of CKD screening.


Subject(s)
Photography , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/diagnostic imaging , Renal Insufficiency, Chronic/diagnosis , Photography/methods , Deep Learning , Artificial Intelligence , Retina/diagnostic imaging , Retina/pathology , Diabetic Retinopathy/diagnostic imaging , Diabetic Retinopathy/diagnosis , Retinal Vessels/diagnostic imaging , Retinal Vessels/pathology , Early Diagnosis
4.
Sci Data ; 11(1): 836, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095400

ABSTRACT

The eyes provide insights into psychology, potentially offering a distinctive perspective for psychological health profiles. However, there exist a notable deficiency in datasets that simultaneously encompass eye features and psychological assessments. To address this gap, our study presents a dataset that included Fundus Photography, Psychological Assessment, Retina Characteristics, and Multimodal Imaging (FPRM). FPRM dataset comprise fundus images at different wavelengths (548 nm and 605 nm), image of oxygen saturation for the retina and 8 specific retinal vessels, videos of retinal blood flow and pupillary light reflex, along with 61 items of multimodal quantitative measurement from 384 participants. Additionally, it features psychological assessments across five dimensions (geriatric depression, generalized anxiety disorder, insomnia, activities of daily living, and deterioration), accompanied by fundus photographs and 6 items of retina characteristics from 1683 participants. FPRM dataset is the first to integrate multimodal ophthalmic data and psychological assessments, not only advancing the development of machine learning applications but also facilitating in-depth research into the relationship between eye health and psychological health profiles.


Subject(s)
Multimodal Imaging , Retina , Humans , Retina/diagnostic imaging , Activities of Daily Living , Depression/diagnostic imaging , Photography
5.
J Neuroinflammation ; 21(1): 190, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095775

ABSTRACT

Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6bSTOP/STOP RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44-/- and Cd44-/-Pde6bSTOP/STOP mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44-/- and Cd44-/-Pde6bSTOP/STOP retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.


Subject(s)
Ependymoglial Cells , Hyaluronan Receptors , Retinitis Pigmentosa , Signal Transduction , Animals , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Mice , Ependymoglial Cells/metabolism , Signal Transduction/physiology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/genetics , Mice, Knockout , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/metabolism , Cell Survival/physiology , Mice, Transgenic , Retina/metabolism , Retina/pathology
6.
Cell Death Dis ; 15(8): 577, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117618

ABSTRACT

Shp2, a critical SH2-domain-containing tyrosine phosphatase, is essential for cellular regulation and implicated in metabolic disruptions, obesity, diabetes, Noonan syndrome, LEOPARD syndrome, and cancers. This study focuses on Shp2 in rod photoreceptor cells, revealing its enrichment, particularly in rods. Deletion of Shp2 in rods leads to age-dependent photoreceptor degeneration. Shp2 targets occludin (OCLN), a tight junction protein, and its deletion reduces OCLN expression in the retina and retinal pigment epithelium (RPE). The isolation of actively translating mRNAs from rods lacking Shp2, followed by RNA sequencing, reveals alterations in cell cycle regulation. Additionally, altered retinal metabolism is observed in retinal cells lacking Shp2. Our studies indicate that Shp2 is crucial for maintaining the structure and function of photoreceptors.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Retinal Degeneration , Animals , Retinal Degeneration/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Mice , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Mice, Knockout , Aging/metabolism , Aging/genetics , Occludin/metabolism , Occludin/genetics , Mice, Inbred C57BL , Gene Deletion , Retina/metabolism , Retina/pathology
7.
Cell Death Dis ; 15(8): 576, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117629

ABSTRACT

Due to the large number of genes and mutations that result in inherited retinal degenerations (IRD), there has been a paucity of therapeutic options for these patients. There is a large unmet need for therapeutic approaches targeting shared pathophysiologic pathways in a mutation-independent manner. The Fas receptor is a major activator and regulator of retinal cell death and inflammation in a variety of ocular diseases. We previously reported the activation of Fas-mediated photoreceptor (PR) cell death in two different IRD mouse models, rd10 and P23H, and demonstrated the protective effect of genetic Fas inhibition. The purpose of this study was to examine the effects of pharmacologic inhibition of Fas in these two models by intravitreal injection with a small peptide inhibitor of the Fas receptor, ONL1204. A single intravitreal injection of ONL1204 was given to one eye of rd10 mice at P14. Two intravitreal injections of ONL1204 were given to the P23H mice, once at P14 and again at 2-months of age. The fellow eyes were injected with vehicle alone. Fas activation, rate of PR cell death, retinal function, and the activation of immune cells in the retina were evaluated. In both rd10 and P23H mice, ONL1204 treatment resulted in decreased number of TUNEL (+) PRs, decreased caspase 8 activity, enhanced photoreceptor cell counts, and improved visual function compared with vehicle treated fellow eyes. Treatment with ONL1204 also reduced immune cell activation in the retinas of both rd10 and P23H mice. The protective effect of pharmacologic inhibition of Fas by ONL1204 in two distinct mouse models of retinal degeneration suggests that targeting this common pathophysiologic mechanism of cell death and inflammation represents a potential therapeutic approach to preserve the retina in patients with IRD, regardless of the genetic underpinning.


Subject(s)
Disease Models, Animal , Retina , Retinal Degeneration , fas Receptor , Animals , Retinal Degeneration/pathology , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Mice , fas Receptor/metabolism , fas Receptor/genetics , Retina/pathology , Retina/metabolism , Retina/drug effects , Mice, Inbred C57BL , Intravitreal Injections , Apoptosis/drug effects
8.
Nat Commun ; 15(1): 6792, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39117640

ABSTRACT

The development of the retina is under tight temporal and spatial control. To gain insights into the molecular basis of this process, we generate a single-nuclei dual-omic atlas of the human developing retina with approximately 220,000 nuclei from 14 human embryos and fetuses aged between 8 and 23-weeks post-conception with matched macular and peripheral tissues. This atlas captures all major cell classes in the retina, along with a large proportion of progenitors and cell-type-specific precursors. Cell trajectory analysis reveals a transition from continuous progression in early progenitors to a hierarchical development during the later stages of cell type specification. Both known and unrecorded candidate transcription factors, along with gene regulatory networks that drive the transitions of various cell fates, are identified. Comparisons between the macular and peripheral retinae indicate a largely consistent yet distinct developmental pattern. This atlas offers unparalleled resolution into the transcriptional and chromatin accessibility landscapes during development, providing an invaluable resource for deeper insights into retinal development and associated diseases.


Subject(s)
Gene Expression Regulation, Developmental , Retina , Single-Cell Analysis , Humans , Retina/embryology , Retina/metabolism , Retina/cytology , Retina/growth & development , Gene Regulatory Networks , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Differentiation/genetics , Fetus , Cell Nucleus/metabolism , Cell Nucleus/genetics , Atlases as Topic
9.
Transl Vis Sci Technol ; 13(8): 5, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39093294

ABSTRACT

Purpose: The effect of carotid artery stenting in patients with unilateral carotid artery stenosis on the retina and choroid was evaluated using swept-source optical coherence tomography angiography (SS-OCTA). Methods: SS-OCTA examination was conducted before stenting and 4 days and 3 months after stenting. The retinal nerve fiber layer, ganglion cell-inner plexiform layer (GCIPL), inner nuclear layer, superficial vascular complex (SVC), deep vascular complex (DVC), choroidal vascular volume (CVV), and choroidal vascular index were measured. Repeated-measures analysis of variance was performed to assess the impact of carotid artery stenting on optical coherence tomography angiography (OCTA) metrics. Results: At baseline, 303 eyes from 160 patients (61.82 ± 9.98 years; 85.29% males) were enrolled. SVC and DVC densities and CVV were lower in ipsilateral eyes (stenosed side) compared to contralateral eyes (all P < 0.05). Four days after stenting, a significant increase was seen in SVC density in ipsilateral eyes (P < 0.05) while a significant increase was seen in CVV in ipsilateral eyes and contralateral eyes (both P < 0.05). Three months after stenting (63 patients with 114 eyes), a significant decrease was seen in the GCIPL thickness of ipsilateral and contralateral eyes (all P < 0.001). Conclusions: Short term after carotid artery stenting, ipsilateral eyes showed a rapid and significant increase in SVC density and CVV. Translational Relevance: Optical coherence tomography (OCT)/OCTA measurements may have the potential to detect retinal and choroidal changes after stenting. Future research on the long-term effect of stenting on the retina and choroid will be guided by these findings.


Subject(s)
Carotid Stenosis , Choroid , Stents , Tomography, Optical Coherence , Humans , Female , Male , Stents/adverse effects , Middle Aged , Choroid/diagnostic imaging , Choroid/blood supply , Choroid/pathology , Carotid Stenosis/surgery , Carotid Stenosis/diagnostic imaging , Carotid Stenosis/therapy , Aged , Retina/diagnostic imaging , Retina/pathology , Retina/surgery , Prospective Studies
10.
PLoS One ; 19(8): e0306794, 2024.
Article in English | MEDLINE | ID: mdl-39110715

ABSTRACT

BACKGROUND AND OBJECTIVES: To develop and test VMseg, a new image processing algorithm performing automatic segmentation of retinal non-perfusion in widefield OCT-Angiography images, in order to estimate the non-perfusion index in diabetic patients. METHODS: We included diabetic patients with severe non-proliferative or proliferative diabetic retinopathy. We acquired images using the PlexElite 9000 OCT-A device with a photomontage of 5 images of size 12 x 12 mm. We then developed VMseg, a Python algorithm for non-perfusion detection, which binarizes a variance map calculated through convolution and morphological operations. We used 70% of our data set (development set) to fine-tune the algorithm parameters (convolution and morphological parameters, binarization thresholds) and evaluated the algorithm performance on the remaining 30% (test set). The obtained automatic segmentations were compared to a ground truth corresponding to manual segmentation from a retina expert and the inference processing time was estimated. RESULTS: We included 51 eyes of 30 patients (27 severe non-proliferative, 24 proliferative diabetic retinopathy). Using the optimal parameters found on the development set to tune the algorithm, the mean dice for the test set was 0.683 (sd = 0.175). We found a higher dice coefficient for images with a higher area of retinal non-perfusion (rs = 0.722, p < 10-4). There was a strong correlation (rs = 0.877, p < 10-4) between VMseg estimated non-perfusion indexes and indexes estimated using the ground truth segmentation. The Bland-Altman plot revealed that 3 eyes (5.9%) were significantly under-segmented by VMseg. CONCLUSION: We developed VMseg, an automatic algorithm for retinal non-perfusion segmentation on 12 x 12 mm OCT-A widefield photomontages. This simple algorithm was fast at inference time, segmented images in full-resolution and for the OCT-A format, was accurate enough for automatic estimation of retinal non-perfusion index in diabetic patients with diabetic retinopathy.


Subject(s)
Algorithms , Diabetic Retinopathy , Tomography, Optical Coherence , Humans , Diabetic Retinopathy/diagnostic imaging , Tomography, Optical Coherence/methods , Female , Male , Middle Aged , Aged , Image Processing, Computer-Assisted/methods , Retinal Vessels/diagnostic imaging , Retina/diagnostic imaging , Retina/pathology , Angiography/methods , Fluorescein Angiography/methods
11.
Transl Vis Sci Technol ; 13(8): 8, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39102240

ABSTRACT

Purpose: This study aims to explore the metabolic signature of aging retina and identify the potential metabolic biomarkers for the diagnosis of retinal aging. Methods: Retinal samples were collected from both young (two months) and aging (14 months) mice to conduct an unbiased metabolic profiling. Liquid chromatography-tandem mass spectrometry analysis was conducted to screen for the metabolic biomarkers and altered signaling pathways associated with retinal aging. Results: We identified 166 metabolites differentially expressed between young and aged retinas using a threshold of orthogonal projection to latent structures discriminant analysis variable importance in projection >1 and P < 0.05. These metabolites were significantly enriched in several metabolic pathways, including purine metabolism, citrate cycle, phenylalanine, tyrosine and tryptophan biosynthesis, glycerophospholipid metabolism, and alanine, aspartate and glutamate metabolism. Among these significantly enriched pathways, glycerophospholipid metabolites emerged as promising candidates for retinal aging biomarkers. We assessed the potential of these metabolites as biomarkers through an analysis of their sensitivity and specificity, determined by the area under the receiver-operating characteristic (ROC) curves. Notably, the metabolites like PC (15:0/22:6), PC (17:0/14:1), LPC (P-16:0), PE (16:0/20:4), and PS (17:0/16:1) demonstrated superior performance in sensitivity, specificity, and accuracy in predicting retinal aging. Conclusions: This study sheds light on the molecular mechanisms underlying retinal aging by identifying distinct metabolic profiles and pathways. These findings provide a valuable foundation for developing future clinical applications in diagnosing, identifying, and treating age-related retinal degeneration. Translational Relevance: This study sheds light on novel metabolic profiles and biomarkers in aging retinas, potentially paving the way for targeted interventions in preventing, diagnosing, and treating age-related retinal degeneration and other retinal diseases.


Subject(s)
Aging , Biomarkers , Mice, Inbred C57BL , Retina , Tandem Mass Spectrometry , Animals , Aging/metabolism , Retina/metabolism , Mice , Biomarkers/metabolism , Chromatography, Liquid , ROC Curve , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods
12.
Invest Ophthalmol Vis Sci ; 65(10): 11, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39106057

ABSTRACT

Purpose: The ramp aftereffect, a visual phenomenon in which perception of light changes dynamically after exposure to sawtooth-modulated light, was first described in 1967. Despite decades of psychophysical research, location and mechanisms of its generation remain unknown. In this study, we investigated a potential retinal contribution to effect formation with specific emphasis on on-/off-pathway involvement. Methods: A 100 ms flash electroretinogram (ERG) was employed to probe the adaptive state of retinal neurons after presentation of stimuli that were homogenous in space but modulated in time following a sawtooth pattern (upward or downward ramps at 2 Hz). Additionally, a psychophysical nulling experiment was performed. Results: Psychophysics data confirmed previous findings that the ramp aftereffect opposes the adapting stimuli in ramp direction and is stronger after upward ramps. The ERG study revealed significant changes of activity in every response component in the low-frequency range (a-wave, b-wave, on-PhNR, d-wave and off-PhNR) and high-frequency range (oscillatory potentials) in amplitudes, peak times, or both. The changes are neither specific to the on- or off-response nor antagonistic between ramp directions. With downward ramp adaptation, effects were stronger. Neither amplitudes nor peak times were correlated with perception strength. Amplitudes and peak times were uncorrelated, and the effect diminished over time, ceasing almost completely with three seconds. Conclusions: Despite abundant effects on retinal responses, the pattern of adaptational effects was not specific to the sawtooth nature of adaptation. Although not ruling out retinal contributions the present findings favor post-retinal mechanisms as the primary locus of the ramp aftereffect.


Subject(s)
Adaptation, Ocular , Electroretinography , Photic Stimulation , Humans , Electroretinography/methods , Adaptation, Ocular/physiology , Adult , Male , Female , Young Adult , Retina/physiology , Psychophysics
13.
BMJ Open Ophthalmol ; 9(1)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39181547

ABSTRACT

BACKGROUND/AIMS: The emerging concept of retinal age, a biomarker derived from retinal images, holds promise in estimating biological age. The retinal age gap (RAG) represents the difference between retinal age and chronological age, which serves as an indicator of deviations from normal ageing. This scoping review aims to collate studies on retinal age to determine its potential clinical utility and to identify knowledge gaps for future research. METHODS: Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses checklist, eligible non-review, human studies were identified, selected and appraised. PubMed, Scopus, SciELO, PsycINFO, Google Scholar, Cochrane, CINAHL, Africa Wide EBSCO, MedRxiv and BioRxiv databases were searched to identify literature pertaining to retinal age, the RAG and their associations. No restrictions were imposed on publication date. RESULTS: Thirteen articles published between 2022 and 2023 were analysed, revealing four models capable of determining biological age from retinal images. Three models, 'Retinal Age', 'EyeAge' and a 'convolutional network-based model', achieved comparable mean absolute errors: 3.55, 3.30 and 3.97, respectively. A fourth model, 'RetiAGE', predicting the probability of being older than 65 years, also demonstrated strong predictive ability with respect to clinical outcomes. In the models identified, a higher predicted RAG demonstrated an association with negative occurrences, notably mortality and cardiovascular health outcomes. CONCLUSION: This review highlights the potential clinical application of retinal age and RAG, emphasising the need for further research to establish their generalisability for clinical use, particularly in neuropsychiatry. The identified models showcase promising accuracy in estimating biological age, suggesting its viability for evaluating health status.


Subject(s)
Aging , Retina , Humans , Retina/diagnostic imaging , Aging/physiology
14.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125629

ABSTRACT

Photoreceptor degeneration is a major cause of untreatable blindness worldwide and has recently been targeted by emerging technologies, including cell- and gene-based therapies. Cell types of neural lineage have shown promise for replacing either photoreceptors or retinal pigment epithelial cells following delivery to the subretinal space, while cells of bone marrow lineage have been tested for retinal trophic effects following delivery to the vitreous cavity. Here we explore an alternate approach in which cells from the immature neural retinal are delivered to the vitreous cavity with the goal of providing trophic support for degenerating photoreceptors. Rat and human retinal progenitor cells were transplanted to the vitreous of rats with a well-studied photoreceptor dystrophy, resulting in substantial anatomical preservation and functional rescue of vision. This work provides scientific proof-of-principle for a novel therapeutic approach to photoreceptor degeneration that is currently being evaluated in clinical trials.


Subject(s)
Retina , Retinal Degeneration , Stem Cell Transplantation , Animals , Rats , Retinal Degeneration/therapy , Retinal Degeneration/pathology , Stem Cell Transplantation/methods , Humans , Retina/pathology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/transplantation , Disease Models, Animal
15.
Cells ; 13(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39195260

ABSTRACT

Neurite outgrowth is a crucial process for organizing neuronal circuits in neuronal development and regeneration after injury. Regenerative failure in the adult mammalian central nervous system (CNS) is attributed to axonal growth inhibitors such as the Nogo protein that commonly binds to Nogo receptor-1 (NgR1). We previously reported that lateral olfactory tract usher substance (LOTUS) functions as an endogenous antagonist for NgR1 in forming neuronal circuits in the developing brain and improving axonal regeneration in the adult injured CNS. However, another molecular and cellular function of LOTUS remains unknown. In this study, we found that cultured retinal explant neurons extend their neurites on the LOTUS-coating substrate. This action was also observed in cultured retinal explant neurons derived from Ngr1-deficient mouse embryos, indicating that the promoting action of LOTUS on neurite outgrowth may be mediated by unidentified LOTUS-binding protein(s). We therefore screened the binding partner(s) of LOTUS by using a liquid chromatography-tandem mass spectrometry (LC-MS/MS). LC-MS/MS analysis and pull-down assay showed that LOTUS interacts with Teneurin-4 (Ten-4), a cell adhesion molecule. RNAi knockdown of Ten-4 inhibited neurite outgrowth on the LOTUS substrate in retinoic acid (RA)-treated Neuro2A cells. Furthermore, a soluble form of Ten-4 attenuates the promoting action on neurite outgrowth in cultured retinal explant neurons on the LOTUS substrate. These results suggest that LOTUS promotes neurite outgrowth by interacting with Ten-4. Our findings may provide a new molecular mechanism of LOTUS to contribute to neuronal circuit formation in development and to enhance axonal regeneration after CNS injury.


Subject(s)
Neuronal Outgrowth , Animals , Neuronal Outgrowth/drug effects , Mice , Neurites/metabolism , Neurites/drug effects , Protein Binding/drug effects , Nogo Receptor 1/metabolism , Humans , Neurons/metabolism , Neurons/drug effects , Nerve Tissue Proteins/metabolism , Retina/metabolism
16.
Cells ; 13(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39195278

ABSTRACT

Retinal progenitor cells (RPCs) are a multipotent and highly proliferative population that give rise to all retinal cell types during organogenesis. Defining their molecular signature is a key step towards identifying suitable approaches to treat visual impairments. Here, we performed RNA sequencing of whole eyes from Xenopus at three embryonic stages and used differential expression analysis to define the transcriptomic profiles of optic tissues containing proliferating and differentiating RPCs during retinogenesis. Gene Ontology and KEGG pathway analyses showed that genes associated with developmental pathways (including Wnt and Hedgehog signaling) were upregulated during the period of active RPC proliferation in early retinal development (Nieuwkoop Faber st. 24 and 27). Developing eyes had dynamic expression profiles and shifted to enrichment for metabolic processes and phototransduction during RPC progeny specification and differentiation (st. 35). Furthermore, conserved adult eye regeneration genes were also expressed during early retinal development, including sox2, pax6, nrl, and Notch signaling components. The eye transcriptomic profiles presented here span RPC proliferation to retinogenesis and include regrowth-competent stages. Thus, our dataset provides a rich resource to uncover molecular regulators of RPC activity and will allow future studies to address regulators of RPC proliferation during eye repair and regrowth.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Transcriptome , Xenopus laevis , Animals , Xenopus laevis/genetics , Xenopus laevis/embryology , Transcriptome/genetics , Eye/metabolism , Eye/embryology , Retina/metabolism , Retina/growth & development , Cell Differentiation/genetics , Cell Proliferation/genetics , Organogenesis/genetics , Stem Cells/metabolism , Stem Cells/cytology , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Signal Transduction/genetics
17.
Int J Mol Sci ; 25(16)2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39201444

ABSTRACT

Emerging evidence suggests that retinal neurodegeneration is an early event in the pathogenesis of diabetic retinopathy (DR), preceding the development of microvascular abnormalities. Here, we assessed the impact of neuroinflammation on the retina of diabetic-induced rats. For this aim we have used a two-photon microscope to image the photoreceptors (PRs) at different eccentricities in unstained retinas obtained from both control (N = 4) and pathological rats (N = 4). This technique provides high-resolution images where individual PRs can be identified. Within each image, every PR was located, and its transversal area was measured and used as an objective parameter of neuroinflammation. In control samples, the size of the PRs hardly changed with retinal eccentricity. On the opposite end, diabetic retinas presented larger PR transversal sections. The ratio of PRs suffering from neuroinflammation was not uniform across the retina. Moreover, the maximum anatomical resolving power (in cycles/deg) was also calculated. This presents a double-slope pattern (from the central retina towards the periphery) in both types of specimens, although the values for diabetic retinas were significantly lower across all retinal locations. The results show that chronic retinal inflammation due to diabetes leads to an increase in PR transversal size. These changes are not uniform and depend on the retinal location. Two-photon microscopy is a useful tool to accurately characterize and quantify PR inflammatory processes and retinal alterations.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Animals , Diabetic Retinopathy/diagnostic imaging , Diabetic Retinopathy/pathology , Rats , Diabetes Mellitus, Experimental/pathology , Male , Photoreceptor Cells, Vertebrate/pathology , Disease Models, Animal , Retina/pathology , Retina/diagnostic imaging , Microscopy, Fluorescence, Multiphoton/methods , Microscopy/methods
18.
Sci Rep ; 14(1): 19530, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174604

ABSTRACT

Macular edema is a known side effect of taxane-based anticancer drugs. We retrospectively investigated data from 11 centers between January 2016 and December 2021. Among 14,260 patients, 30 (0.21%) developed macular edema; from these, the number of cases associated with nab-paclitaxel was 16 (0.43%), significantly higher than the number of cases associated with paclitaxel or docetaxel (P < 0.01). Visual acuity (VA) and retinal choroidal change were examined in 27 patients, with a follow-up of at least 3 months. The patients' mean age was 67.2 years; 14 (51.3%) were male and four (14.8%) had unilateral onset. The mean interval between anticancer drug initiation and the first ophthalmology visit was 290.1 days. Among the 20 patients who discontinued anticancer drugs, VA and edema significantly improved 2 months after discontinuation (LogMAR VA: 0.50 vs. 0.28, central retinal thickness: 472.7 µm vs. 282.5 µm, both P < 0.01). No significant changes were observed in the central choroidal thickness. A correlation was found between duration of taxane treatment and VA immediately before discontinuation of anticancer drugs (ß = 0.00050; 95% confidence interval: 0.00036-0.00097; P < 0.05). Although taxane-induced macular edema is reversible, slower anticancer drug discontinuation worsened VA, highlighting the need for regular ophthalmologic evaluation during treatments.


Subject(s)
Macular Edema , Taxoids , Visual Acuity , Humans , Female , Macular Edema/chemically induced , Macular Edema/drug therapy , Male , Aged , Retrospective Studies , Japan/epidemiology , Middle Aged , Taxoids/adverse effects , Incidence , Prognosis , Visual Acuity/drug effects , Bridged-Ring Compounds/adverse effects , Aged, 80 and over , Antineoplastic Agents/adverse effects , Docetaxel/adverse effects , Paclitaxel/adverse effects , Retina/drug effects , Retina/pathology , Retina/diagnostic imaging
19.
Mol Brain ; 17(1): 58, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175067

ABSTRACT

Protocadherin 19 (Pcdh19) is a homophilic cell adhesion molecule and is involved in a variety of neuronal functions. Here, we tested whether Pcdh19 has a regulatory role in axon guidance using the developing Xenopus retinotectal system. We performed targeted microinjections of a translation blocking antisense morpholino oligonucleotide to knock down the expression of Pcdh19 selectively in the central nervous system. Knocking down Pcdh19 expression resulted in navigational errors of retinal ganglion cell (RGC) axons specifically at the optic chiasm. Instead of projecting to the contralateral optic tectum, RGC axons in the Pcdh19-depleted embryo misprojected ipsilaterally. Although incorrectly delivered into the ipsilateral brain hemisphere, these axons correctly reached the optic tectum. These data suggest that Pcdh19 has a critical role in preventing mixing of RGC axons originating from the opposite eyes at the optic chiasm, highlighting the importance of cell adhesion in bundling of RGC axons.


Subject(s)
Axon Guidance , Axons , Cadherins , Protocadherins , Retinal Ganglion Cells , Xenopus Proteins , Xenopus laevis , Animals , Cadherins/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Retinal Ganglion Cells/metabolism , Xenopus laevis/embryology , Axons/metabolism , Retina/metabolism , Retina/embryology , Visual Pathways , Gene Knockdown Techniques , Optic Chiasm/embryology , Optic Chiasm/metabolism , Superior Colliculi/embryology , Superior Colliculi/metabolism , Gene Expression Regulation, Developmental
20.
Commun Biol ; 7(1): 1027, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169121

ABSTRACT

The retina is light-sensitive neuronal tissue in the back of the eye. The phospholipid composition of the retina is unique and highly enriched in polyunsaturated fatty acids, including docosahexaenoic fatty acid (DHA). While it is generally accepted that a high DHA content is important for vision, surprisingly little is known about the mechanisms of DHA enrichment in the retina. Furthermore, the biological processes controlled by DHA in the eye remain poorly defined as well. Here, we combined genetic manipulations with lipidomic analysis in mice to demonstrate that acyl-CoA synthetase 6 (Acsl6) serves as a regulator of the unique composition of retinal membranes. Inactivation of Acsl6 reduced the levels of DHA-containing phospholipids, led to progressive loss of light-sensitive rod photoreceptor neurons, attenuated the light responses of these cells, and evoked distinct transcriptional response in the retina involving the Srebf1/2 (sterol regulatory element binding transcription factors 1/2) pathway. This study identifies one of the major enzymes responsible for DHA enrichment in the retinal membranes and introduces a model allowing an evaluation of rod functioning and pathology caused by impaired DHA incorporation/retention in the retina.


Subject(s)
Coenzyme A Ligases , Phospholipids , Retinal Rod Photoreceptor Cells , Animals , Retinal Rod Photoreceptor Cells/metabolism , Mice , Phospholipids/metabolism , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Retina/metabolism , Docosahexaenoic Acids/metabolism , Mice, Knockout , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL