Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 17.066
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Sci Rep ; 14(1): 18752, 2024 08 13.
Article in English | MEDLINE | ID: mdl-39138242

ABSTRACT

Subretinal fibrosis is a major untreatable cause of poor outcomes in neovascular age-related macular degeneration. Mouse models of subretinal fibrosis all possess a degree of invasiveness and tissue damage not typical of fibrosis progression. This project characterises JR5558 mice as a model to study subretinal fibrosis. Fundus and optical coherence tomography (OCT) imaging was used to non-invasively track lesions. Lesion number and area were quantified with ImageJ. Retinal sections, wholemounts and Western blots were used to characterise alterations. Subretinal lesions expand between 4 and 8 weeks and become established in size and location around 12 weeks. Subretinal lesions were confirmed to be fibrotic, including various cell populations involved in fibrosis development. Müller cell processes extended from superficial retina into subretinal lesions at 8 weeks. Western blotting revealed increases in fibronectin (4 wk and 8 wk, p < 0.001), CTGF (20 wks, p < 0.001), MMP2 (12 wks and 20 wks p < 0.05), αSMA (12 wks and 20 wks p < 0.05) and GFAP (8 wk and 12 wk, p ≤ 0.01), consistent with our immunofluorescence results. Intravitreal injection of Aflibercept reduced subretinal lesion growth. Our study provides evidence JR5558 mice have subretinal fibrotic lesions that grow between 4 and 8 weeks and confirms this line to be a good model to study subretinal fibrosis development and assess treatment options.


Subject(s)
Disease Models, Animal , Fibrosis , Retina , Tomography, Optical Coherence , Animals , Mice , Tomography, Optical Coherence/methods , Retina/pathology , Retina/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Fibronectins/metabolism , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Macular Degeneration/pathology , Macular Degeneration/metabolism , Matrix Metalloproteinase 2/metabolism , Intravitreal Injections , Glial Fibrillary Acidic Protein/metabolism , Actins/metabolism , Mice, Inbred C57BL , Recombinant Fusion Proteins
13.
Cell Rep Methods ; 4(8): 100837, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39127043

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) play a crucial role in several physiological light responses. In this study, we generate an improved Opn4cre knockin allele (Opn4cre(DSO)), which faithfully reproduces endogenous Opn4 expression and improves compatibility with widely used reporters. We evaluated the efficacy and sensitivity of Opn4cre(DSO) for labeling in retina and brain and provide an in-depth comparison with the extensively utilized Opn4cre(Saha) line. Through this characterization, Opn4cre(DSO) demonstrated higher specificity in labeling ipRGCs with minimal recombination escape. Leveraging a combination of electrophysiological, molecular, and morphological analyses, we confirmed its sensitivity in detecting all ipRGC types (M1-M6) and defined their unique topographical distribution across the retina. In the brain, the Opn4cre(DSO) line labels ipRGC projections with minimal labeling of cell bodies. Overall, the Opn4cre(DSO) mouse line represents an improved tool for studying ipRGC function and distribution, offering a means to selectively target these cells to study light-regulated behaviors and physiology.


Subject(s)
Integrases , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/metabolism , Mice , Integrases/genetics , Integrases/metabolism , Rod Opsins/genetics , Rod Opsins/metabolism , Retina/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Brain/metabolism
14.
Exp Eye Res ; 246: 110015, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089568

ABSTRACT

Fragile X Syndrome (FXS), the most common inherited form of human intellectual disability, is a monogenic neurodevelopmental disorder caused by a loss-of-function mutation of the FMR1 gene. FMR1 is encoding the Fragile X Messenger Ribonucleo Protein (FMRP) an RNA-binding protein that regulates the translation of synaptic proteins. The absence of FMRP expression has many important consequences on synaptic plasticity and function, leading to the FXS clinical phenotype. Over the last decade, a visual neurosensorial phenotype had been described in the FXS patients as well as in the murine model (Fmr1-/ymice), characterized by retinal deficits associated to retinal perception alterations. However, although the transcriptomic profile in the absence of FMRP has been studied in the cerebral part of the central nervous system (CNS), there are no actual data for the retina which is an extension of the CNS. Herein, we investigate the transcriptomic profile of mRNA from whole retinas of Fmr1-/ymice. Interestingly, we found a specific signature of Fmrp absence on retinal mRNA expression with few common genes compared to other brain studies. Gene Ontology on these retinal specific genes demonstrated an enrichment in retinal development genes as well as in synaptic genes. These alterations could be linked to the reported retinal phenotype of the FXS condition. In conclusion, we describe for the first time, retinal-specific transcriptomic changes in the absence of FMRP.


Subject(s)
Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Retina , Transcriptome , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Animals , Mice , Retina/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mice, Inbred C57BL , Gene Expression Profiling , Mice, Knockout , Gene Expression Regulation/physiology , Male
15.
Commun Biol ; 7(1): 993, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143195

ABSTRACT

The pupillary light reflex (PLR) adapts the amount of light reaching the retina, protecting it and improving image formation. Two PLR mechanisms have been described in vertebrates. First, the pretectum receives retinal inputs and projects to the Edinger-Westphal nucleus (EWN), which targets the ciliary ganglion through the oculomotor nerve (nIII). Postganglionic fibers enter the eye-globe, traveling to the iris sphincter muscle. Additionally, some vertebrates exhibit an iris-intrinsic PLR mechanism mediated by sphincter muscle cells that express melanopsin inducing muscle contraction. Given the high degree of conservation of the lamprey visual system, we investigated the mechanisms underlying the PLR to shed light onto their evolutionary origins. Recently, a PLR mediated by melanopsin was demonstrated in lampreys, suggested to be brain mediated. Remarkably, we found that PLR is instead mediated by direct retino-iridal cholinergic projections. This retina-mediated PLR acts synergistically with an iris-intrinsic mechanism that, as in other vertebrates, is mediated by melanopsin and has contribution of gap junctions between muscle fibers. In contrast, we show that lampreys lack the brain-mediated PLR. Our results suggest that two eye-intrinsic PLR mechanisms were present in early vertebrate evolution, whereas the brain-mediated PLR has a more recent origin.


Subject(s)
Iris , Reflex, Pupillary , Retina , Animals , Reflex, Pupillary/physiology , Iris/physiology , Iris/metabolism , Retina/physiology , Retina/metabolism , Lampreys/physiology , Muscle Contraction/physiology , Rod Opsins/metabolism , Rod Opsins/genetics , Light , Vertebrates/physiology
16.
CRISPR J ; 7(4): 188-196, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39111828

ABSTRACT

Vascular endothelial growth factor receptor (VEGFR)-2 is a key switch for angiogenesis, which is observed in various human diseases. In this study, a novel system for advanced prime editing (PE), termed PE6h, is developed, consisting of dual lentiviral vectors: (1) a clustered regularly interspaced palindromic repeat-associated protein 9 (H840A) nickase fused with reverse transcriptase and an enhanced PE guide RNA and (2) a dominant negative (DN) MutL homolog 1 gene with nicking guide RNA. PE6h was used to edit VEGFR2 (c.18315T>A, 50.8%) to generate a premature stop codon (TAG from AAG), resulting in the production of DN-VEGFR2 (787 aa) in human retinal microvascular endothelial cells (HRECs). DN-VEGFR2 impeded VEGF-induced phosphorylation of VEGFR2, Akt, and extracellular signal-regulated kinase-1/2 and tube formation in PE6h-edited HRECs in vitro. Overall, our results highlight the potential of PE6h to inhibit angiogenesis in vivo.


Subject(s)
Angiogenesis , Endothelial Cells , Gene Editing , Vascular Endothelial Growth Factor Receptor-2 , Humans , Angiogenesis/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Endothelial Cells/metabolism , Gene Editing/methods , Genetic Vectors , Neovascularization, Pathologic/metabolism , Phosphorylation , Retina/metabolism , RNA, Guide, CRISPR-Cas Systems , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics
17.
Acta Neuropathol ; 148(1): 24, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160362

ABSTRACT

The retina is increasingly recognised as a potential source of biomarkers for neurodegenerative diseases. Hallmark protein aggregates in the retinal neuronal tissue could be imaged through light non-invasively. Post-mortem studies have already shown the presence of specific hallmark proteins in Alzheimer's disease, primary tauopathies, synucleinopathies and frontotemporal lobar degeneration. This study aims to assess proteinopathy in a post-mortem cohort with different neurodegenerative diseases and assess the presence of the primary pathology in the retina. Post-mortem eyes were collected in collaboration with the Netherlands Brain Bank from donors with Alzheimer's disease (n = 17), primary tauopathies (n = 8), synucleinopathies (n = 27), frontotemporal lobar degeneration (n = 8), mixed pathology (n = 11), other neurodegenerative diseases (n = 6), and cognitively normal controls (n = 25). Multiple cross sections of the retina and optic nerve tissue were immunostained using antibodies against pTau Ser202/Thr205 (AT8), amyloid-beta (4G8), alpha-synuclein (LB509), pTDP-43 Ser409/410 and p62-lck ligand (p62) and were assessed for the presence of aggregates and inclusions. pTau pathology was observed as a diffuse signal in Alzheimer's disease, primary tauopathies and controls with Alzheimer's disease neuropathological changes. Amyloid-beta was observed in the vessel wall and as cytoplasmic granular deposits in all groups. Alpha-synuclein pathology was observed as Lewy neurites in the retina in synucleinopathies associated with Lewy pathology and as oligodendroglial cytoplasmic inclusions in the optic nerve in multiple system atrophy. Anti-pTDP-43 generally showed typical neuronal cytoplasmic inclusion bodies in cases with frontotemporal lobar degeneration with TDP-43 and also in cases with later stages of limbic-associated TDP-43 encephalopathy. P62 showed inclusion bodies similar to those seen with anti-pTDP-43. Furthermore, pTau and alpha-synuclein pathology were significantly associated with increasing Braak stages for neurofibrillary tangles and Lewy bodies, respectively. Mixed pathology cases in this cohort consisted of cases (n = 6) with high Braak LB stages (> 4) and low or moderate AD pathology, high AD pathology (n = 1, Braak NFT 6, Thal phase 5) with moderate LB pathology, or a combination of low/moderate scores for different pathology scores in the brain (n = 4). There were no cases with advanced co-pathologies. In seven cases with Braak LB ≥ 4, LB pathology was observed in the retina, while tau pathology in the retina in the mixed pathology group (n = 11) could not be observed. From this study, we conclude that the retina reflects the presence of the major hallmark proteins associated with neurodegenerative diseases. Although low or moderate levels of copathology were found in the brains of most cases, the retina primarily manifested protein aggregates associated with the main neurodegenerative disease. These findings indicate that with appropriate retinal imaging techniques, retinal biomarkers have the potential to become highly accurate indicators for diagnosing the major neurodegenerative diseases of the brain.


Subject(s)
Neurodegenerative Diseases , Retina , tau Proteins , Humans , Aged , Female , Male , Retina/pathology , Retina/metabolism , Aged, 80 and over , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/metabolism , tau Proteins/metabolism , Middle Aged , alpha-Synuclein/metabolism , Autopsy , Tauopathies/pathology , Tauopathies/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , DNA-Binding Proteins/metabolism
18.
Gene ; 930: 148861, 2024 Dec 20.
Article in English | MEDLINE | ID: mdl-39153705

ABSTRACT

BACKGROUND: By identifying molecular biological markers linked to cuproptosis in diabetic retinopathy (DR), new pathobiological pathways and more accessible diagnostic markers can be developed. METHODS: The datasets related to DR were acquired from the Gene Expression Omnibus database, while genes associated with cuproptosis were sourced from previously published compilations. Consensus clustering was conducted to delineate distinct DR subclasses. Feature genes were identified utilizing weighted correlation network analysis (WGCNA). Additionally, two machine-learning algorithms were employed to refine the selection of feature genes. Finally, we conducted preliminary validation experiments to ascertain the involvement of cuproptosis in DR development and the transcriptional regulation of critical genes using both the streptozotocin-induced diabetic mouse model and the high glucose-induced BV2 model. RESULTS: In the STZ-induced diabetic mouse retinas, a decrease in the expression of cuproptosis signature proteins (FDX1, DLAT, and NDUFS8) suggested the occurrence of cuproptosis in DR. Subsequently, the expression of eight cuproptosis differential genes was validated through the STZ-induced diabetes and oxygen-induced retinopathy (OIR) models, with the key gene SLC31A1 showing upregulation in both models and dataset species. Further analyses, including weighted gene co-expression network analysis, GSVA, and immune infiltration analysis, indicated a close correlation between cuproptosis and microglia function. Additionally, validation in an in vitro model of microglia indicated the occurrence of cuproptosis in microglia under high glucose conditions, alongside abnormal expression of STAT1 with SLC31A1. CONCLUSION: Our findings suggest that STAT1/SLC31A1 may pave the way for a deeper comprehension of the mechanistic basis of DR and offer potential therapeutic avenues.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , STAT1 Transcription Factor , Animals , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Mice , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Male , Retina/metabolism , Retina/pathology , Mice, Inbred C57BL , Gene Expression Regulation , Gene Regulatory Networks , Humans
19.
Sci Adv ; 10(35): eadk4062, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39196935

ABSTRACT

The mammalian retina is considered an autonomous circuit, yet work dating back to Ramon y Cajal indicates that it receives inputs from the brain. How such inputs affect retinal processing has remained unknown. We confirmed brain-to-retina projections of histaminergic neurons from the mouse hypothalamus. Histamine application ex vivo altered the activity of various retinal ganglion cells (RGCs), including direction-selective RGCs that gained responses to high motion velocities. These results were reproduced in vivo with optic tract recordings where histaminergic retinopetal axons were activated chemogenetically. Such changes could improve vision of fast-moving objects (e.g., while running), which fits with the known increased activity of histaminergic neurons during arousal. An antihistamine drug reduced optomotor responses to high-speed moving stimuli in freely moving mice. In humans, the same antihistamine nonuniformly modulated visual sensitivity across the visual field, indicating an evolutionary conserved function of the histaminergic system. Our findings expose a previously unappreciated role for brain-to-retina projections in modulating retinal function.


Subject(s)
Histamine , Hypothalamus , Retina , Retinal Ganglion Cells , Animals , Histamine/pharmacology , Histamine/metabolism , Hypothalamus/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Mice , Retina/metabolism , Retina/physiology , Retina/drug effects , Retina/cytology , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Neurons/metabolism , Neurons/physiology , Neurons/drug effects , Humans , Mice, Inbred C57BL , Visual Pathways/drug effects , Visual Pathways/physiology
20.
Genesis ; 62(4): e23615, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39139090

ABSTRACT

Armadillo repeat-containing X-linked protein-1 (Armcx1) is a poorly characterized transmembrane protein that regulates mitochondrial transport in neurons. Its overexpression has been shown to induce neurite outgrowth in embryonic neurons and to promote retinal ganglion cell (RGC) survival and axonal regrowth in a mouse optic nerve crush model. In order to evaluate the functions of endogenous Armcx1 in vivo, we have created a conditional Armcx1 knockout mouse line in which the entire coding region of the Armcx1 gene is flanked by loxP sites. This Armcx1fl line was crossed with mouse strains in which Cre recombinase expression is driven by the promoters for ß-actin and Six3, in order to achieve deletion of Armcx1 globally and in retinal neurons, respectively. Having confirmed deletion of the gene, we proceeded to characterize the abundance and morphology of RGCs in Armcx1 knockout mice aged to 15 months. Under normal physiological conditions, no evidence of aberrant retinal or optic nerve development or RGC degeneration was observed in these mice. The Armcx1fl mouse should be valuable for future studies investigating mitochondrial morphology and transport in the absence of Armcx1 and in determining the susceptibility of Armcx1-deficient neurons to degeneration in the setting of additional heritable or environmental stressors.


Subject(s)
Armadillo Domain Proteins , Retinal Ganglion Cells , Animals , Mice , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Mice, Knockout , Optic Nerve/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL