Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 146(5): 747-766, 2023 11.
Article in English | MEDLINE | ID: mdl-37682293

ABSTRACT

Central serous chorioretinopathy (CSCR) belongs to the pachychoroid spectrum, a pathological phenotype of the choroidal vasculature, in which blood flow is under the choroidal nervous system (ChNS) regulation. The pathogenesis of CSCR is multifactorial, with the most recognised risk factor being intake of glucocorticoids, which activate both the gluco- and the mineralocorticoid (MR) receptors. As MR over-activation is pathogenic in the retina and choroid, it could mediate the pathogenic effects of glucocorticoids in CSCR. But the role of MR signalling in pachychoroid is unknown and whether it affects the ChNS has not been explored. Using anatomo-neurochemical characterisation of the ChNS in rodents and humans, we discovered that beside innervation of arteries, choroidal veins and choriocapillaris are also innervated, suggesting that the entire choroidal vasculature is under neural control. The numerous synapses together with calcitonin gene-related peptide (CGRP) vesicles juxtaposed to choroidal macrophages indicate a neuro-immune crosstalk. Using ultrastructural approaches, we show that transgenic mice overexpressing human MR, display a pachychoroid-like phenotype, with signs of choroidal neuropathy including myelin abnormalities, accumulation and enlargement of mitochondria and nerves vacuolization. Transcriptomic analysis of the RPE/choroid complex in the transgenic mice reveals regulation of corticoids target genes, known to intervene in nerve pathophysiology, such as Lcn2, rdas1/dexras1, S100a8 and S100a9, rabphilin 3a (Rph3a), secretogranin (Scg2) and Kinesin Family Member 5A (Kif5a). Genes belonging to pathways related to vasculature development, hypoxia, epithelial cell apoptosis, epithelial mesenchymal transition, and inflammation, support the pachychoroid phenotype and highlight downstream molecular targets. Hypotheses on the imaging phenotype of pachychoroid in humans are put forward in the light of these new data. Our results provide evidence that MR overactivation causes a choroidal neuropathy that could explain the pachychoroid phenotype found in transgenic mice overexpressing human MR. In patients with pachychoroid and CSCR in which systemic dysautonomia has been demonstrated, MR-induced choroidal neuropathy could be the missing link between corticoids and pachychoroid.


Subject(s)
Receptors, Mineralocorticoid , Tomography, Optical Coherence , Animals , Mice , Humans , Receptors, Mineralocorticoid/genetics , Tomography, Optical Coherence/methods , Choroid/blood supply , Choroid/pathology , Adrenal Cortex Hormones , Glucocorticoids , Nervous System , Mice, Transgenic , Retrospective Studies
2.
Int J Mol Sci ; 22(17)2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34502527

ABSTRACT

Glucocorticoids are amongst the most used drugs to treat retinal diseases of various origins. Yet, the transcriptional regulations induced by glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation in retinal pigment epithelium cells (RPE) that form the outer blood-retina barrier are unknown. Levels of endogenous corticoids, ligands for MR and GR, were measured in human ocular media. Human RPE cells derived from induced pluripotent stem cells (iRPE) were used to analyze the pan-transcriptional regulations induced by aldosterone-an MR-specific agonist, or cortisol or cortisol + RU486-a GR antagonist. The retinal phenotype of transgenic mice that overexpress the human MR (P1.hMR) was analyzed. In the human eye, the main ligand for GR and MR is cortisol. The iRPE cells express functional GR and MR. The subset of genes regulated by aldosterone and by cortisol + RU-486, and not by cortisol alone, mimics an imbalance toward MR activation. They are involved in extracellular matrix remodeling (CNN1, MGP, AMTN), epithelial-mesenchymal transition, RPE cell proliferation and migration (ITGB3, PLAUR and FOSL1) and immune balance (TNFSF18 and PTX3). The P1.hMR mice showed choroidal vasodilation, focal alteration of the RPE/choroid interface and migration of RPE cells together with RPE barrier function alteration, similar to human retinal diseases within the pachychoroid spectrum. RPE is a corticosteroid-sensitive epithelium. MR pathway activation in the RPE regulates genes involved in barrier function, extracellular matrix, neural regulation and epithelial differentiation, which could contribute to retinal pathology.


Subject(s)
Aldosterone/metabolism , Hydrocortisone/metabolism , Pluripotent Stem Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Retinal Diseases/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Epithelial-Mesenchymal Transition , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/pathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Retinal Diseases/genetics , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology
3.
Transl Res ; 229: 83-99, 2021 03.
Article in English | MEDLINE | ID: mdl-33080394

ABSTRACT

Sulfonylureas, widely used as hypoglycemic agents in adults with type 2 diabetes, have neuroprotective effects in preclinical models of central nervous system injury, and in children with neuropsychomotor impairments linked to neonatal diabetes secondary to ATP-sensitive potassium channel mutations. In the human and rodent retina, we show that the glibenclamide-activated channel sulfonylurea receptor 1 (SUR1) is expressed in the retina and enriched in the macula; we also show that it colocalizes with the potassium channel Kir6.2, and with the cation channel transporter TRPM4. Glibenclamide (glyburide), administered at doses that did not decrease the glycemia, or injected directly into the eye, protected the structure and the function of the retina in various models of retinal injury that recapitulate the pathogenic neurodegenerative events in the diabetic retina. The downregulation of SUR1 using a siRNA suppressed the neuroprotective effects of glibenclamide on excitotoxic stress-induced cell death. The glibenclamide effects include the transcriptional regulation of antioxidant and neuroprotective genes. Ocular glibenclamide could be repurposed for diabetic retinopathy.


Subject(s)
Glyburide/pharmacology , Neuroprotective Agents/pharmacology , Retinal Diseases/drug therapy , Retinal Neurons/drug effects , Administration, Oral , Animals , Chlorocebus aethiops , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Female , Glyburide/administration & dosage , Humans , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Macaca fascicularis , Male , Middle Aged , Neuroprotective Agents/administration & dosage , Potassium Channels, Inwardly Rectifying/metabolism , Rats, Inbred Lew , Rats, Wistar , Retinal Diseases/etiology , Retinal Diseases/pathology , Retinal Neurons/pathology , Sulfonylurea Receptors/metabolism , TRPM Cation Channels/metabolism
4.
Med Sci (Paris) ; 36(6-7): 594-599, 2020.
Article in French | MEDLINE | ID: mdl-32614310

ABSTRACT

The neuroretina is a functional unit of the central nervous system that converts a light signal into a nerve impulse. Of neuroectodermal origin, derived from the diencephalon, the neuroretina is a layered tissue composed of six types of neuronal cells (two types of photoreceptors: cones and rods, horizontal, bipolar, amacrine and ganglion cells) and three types of glial cells (Müller glial cells, astrocytes and microglial cells). The neuroretina lays on the retinal pigmentary epithelium, that together form the retina. The existence of the internal and external blood-retinal barriers and intra-retinal junctions reflects the fineness of regulation of the retinal exchanges with the circulation and within the retina itself. The central zone of the human retina, which is highly specialized for visual acuity, has anatomical specificities. Recent imaging methods make it possible now to enrich our knowledge of the anatomical and functional characteristics of the retina, which are still imperfectly described.


TITLE: Anatomie de la rétine. ABSTRACT: La neurorétine est une unité fonctionnelle du système nerveux central assurant la conversion d'un signal lumineux en un influx nerveux. D'origine neuroectodermique, dérivée du diencéphale, la neurorétine est un tissu stratifié, composé de six types de cellules neuronales (deux types de photorécepteurs : les cônes et les bâtonnets ; les cellules horizontales, bipolaires, amacrines et ganglionnaires) et de trois types de cellules gliales (les cellules gliales de Müller, les astrocytes et les cellules microgliales). La neurorétine repose sur l'épithélium pigmentaire rétinien, l'ensemble constituant la rétine. L'existence des barrières hémato-rétiniennes interne et externe et des jonctions intra-rétiniennes rend compte de la finesse de la régulation des échanges de la rétine avec la circulation et au sein de la rétine elle-même. La zone centrale de la rétine humaine, la macula, zone hautement spécialisée pour assurer l'acuité visuelle, présente des spécificités anatomiques. Les méthodes d'imagerie récentes permettent d'enrichir nos connaissances sur les caractéristiques anatomiques et fonctionnelles de la rétine, qui restent encore imparfaitement décrites.


Subject(s)
Retina/anatomy & histology , Animals , Choroid/blood supply , Choroid/cytology , Choroid/physiology , Humans , Neuroglia/cytology , Neuroglia/physiology , Retina/cytology , Retina/physiology , Retina/ultrastructure , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/physiology , Retinal Pigment Epithelium/blood supply , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/physiology , Retinal Vessels/cytology , Retinal Vessels/physiology
5.
Graefes Arch Clin Exp Ophthalmol ; 257(11): 2401-2427, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31529323

ABSTRACT

PURPOSE: This study was conducted in order to study Sostdc1 expression in rat and human developing and adult eyes. METHODS: Using the yeast signal sequence trap screening method, we identified the Sostdc1 cDNA encoding a protein secreted by the adult rat retinal pigment epithelium. We determined by in situ hybridization, RT-PCR, immunohistochemistry, and western blot analysis Sostdc1 gene and protein expression in developing and postnatal rat ocular tissue sections. We also investigated Sostdc1 immunohistolocalization in developing and adult human ocular tissues. RESULTS: We demonstrated a prominent Sostdc1 gene expression in the developing rat central nervous system (CNS) and eyes at early developmental stages from E10.5 days postconception (dpc) to E13 dpc. Specific Sostdc1 immunostaining was also detected in most adult cells of rat ocular tissue sections. We also identified the rat ocular embryonic compartments characterized by a specific Sostdc1 immunohistostaining and specific Pax6, Sox2, Otx2, and Vsx2 immunohistostaining from embryonic stages E10.5 to E13 dpc. Furthermore, we determined the localization of SOSTDC1 immunoreactivity in ocular tissue sections of developing and adult human eyes. Indeed, we detected SOSTDC1 immunostaining in developing and adult human retinal pigment epithelium (RPE) and neural retina (NR) as well as in several developing and adult human ocular compartments, including the walls of choroidal and scleral vessels. Of utmost importance, we observed a strong SOSTDC1 expression in a pathological ocular specimen of type 2 Peters' anomaly complicated by retinal neovascularization as well in the walls ofother pathological extra-ocular vessels.  CONCLUSION: As rat Sostdc1 and human SOSTDC1 are dual antagonists of the Wnt/ß-catenin and BMP signaling pathways, these results underscore the potential crucial roles of these pathways and their antagonists, such as Sostdc1 and SOSTDC1, in developing and adult mammalian normal eyes as well as in syndromic and nonsyndromic congenital eye diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Eye Diseases, Hereditary/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Retinal Pigment Epithelium/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Aged , Animals , Blotting, Western , Child, Preschool , Disease Models, Animal , Eye Diseases, Hereditary/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development
6.
Sci Rep ; 9(1): 13873, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554875

ABSTRACT

Age Related Macular Degeneration (AMD) is the first cause of social blindness in people aged over 65 leading to atrophy of retinal pigment epithelial cells (RPE), photoreceptors and choroids, eventually associated with choroidal neovascularization. Accumulation of undigested cellular debris within RPE cells or under the RPE (Drusen), oxidative stress and inflammatory mediators contribute to the RPE cell death. The major risk to develop AMD is the Y402H polymorphism of complement factor H (CFH). CFH interacting with oxidized phospholipids on the RPE membrane modulates the functions of these cells, but the exact role of CFH in RPE cell death and survival remain poorly understood. The aim of this study was to analyze the potential protective mechanism of CFH on RPE cells submitted to oxidative stress. Upon exposure to oxidized lipids 4-HNE (4-hydroxy-2-nonenal) derived from photoreceptors, both the human RPE cell line ARPE-19 and RPE cells derived from human induced pluripotent stem cells were protected from death only in the presence of the full length human recombinant CFH in the culture medium. This protective effect was independent from the membrane attack complex (MAC) formation. CFH maintained RPE cells tight junctions' structure and regulated the caspase dependent apoptosis process. These results demonstrated the CFH anti-oxidative stress functions independently of its capacity to inhibit MAC formation.


Subject(s)
Complement Factor H/pharmacology , Complement Membrane Attack Complex/drug effects , Retinal Pigment Epithelium/drug effects , Aldehydes/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Death/drug effects , Cell Line , Complement Membrane Attack Complex/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Microscopy, Electron, Transmission , Oxidative Stress/drug effects , Real-Time Polymerase Chain Reaction , Recombinant Proteins , Retinal Pigment Epithelium/metabolism , Tight Junctions/drug effects
7.
Exp Eye Res ; 187: 107747, 2019 10.
Article in English | MEDLINE | ID: mdl-31394103

ABSTRACT

Preclinical and clinical evidences show that aldosterone and/or mineralocorticoid receptor (MR) over-activation by glucocorticoids can be deleterious to the retina and to the retinal pigment epithelium (RPE)-choroid complex. However, the exact molecular mechanisms driving these effects remain poorly understood and pathological consequences of chronic exposure of the retina and RPE/choroid to aldosterone have not been completely explored. We aimed to decipher the transcriptomic regulation in the RPE-choroid complex in rats in response to acute intraocular aldosterone injection and to explore the consequences of systemic chronic aldosterone exposure on the morphology and the gene regulation in RPE/choroid in mice. High dose of aldosterone (100 nM) was intravitreously injected in Lewis rat eyes in order to yield an aldosterone dose able to induce a molecular response at the apical side of the RPE-choroid complex. The posterior segment morphology was evaluated in vivo using optical coherence tomography (OCT) before and 24 h after aldosterone injection. Rat RPE-choroid complexes were used for RNA sequencing and analysis. Uninephrectomy/aldosterone/salt (NAS) model was created in wild-type C57BL/6 mice. After 6 weeks, histology of mouse posterior segments were observed ex vivo. Gene expression in the RPE-choroid complex was analyzed using quantitative PCR. Acute intravitreous injection of aldosterone induced posterior segment inflammation observed on OCT. RNA sequencing of rat RPE-choroid complexes revealed up-regulation of pathways involved in inflammation, oxidative stress and RNA procession, and down-regulation of genes involved in synaptic activity, muscle contraction, cytoskeleton, cell junction and transporters. Chronic aldosterone/salt exposure in NAS model induces retinal edema, choroidal vasodilation and RPE cell dysfunction and migration. Quantitative PCR showed deregulation of genes involved in inflammatory response, oxidative stress, particularly the NOX pathway, angiogenesis and cell contractility. Both rodent models share some common phenotypes and molecular regulations in the RPE-choroid complex that could contribute to pachychoroid epitheliopathy in humans. The difference in inflammatory status relies on different intraocular or systemic route of aldosterone administration and on the different doses of aldosterone exposed to the RPE-choroid complex.


Subject(s)
Aldosterone/pharmacology , Choroid/drug effects , Eye Proteins/genetics , Gene Expression Regulation/physiology , Retinal Pigment Epithelium/drug effects , Acute Disease , Animals , Blood Pressure/drug effects , Cell Movement , Choroid/metabolism , Choroid/pathology , Choroid Diseases/chemically induced , Choroid Diseases/diagnosis , Chronic Disease , Disease Models, Animal , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Nephrectomy , Papilledema/chemically induced , Papilledema/diagnosis , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Sequence Analysis, RNA , Tomography, Optical Coherence
8.
Sci Adv ; 5(1): eaau9940, 2019 01.
Article in English | MEDLINE | ID: mdl-30662950

ABSTRACT

In retinal detachment (RD), photoreceptor death and permanent vision loss are caused by neurosensory retina separating from the retinal pigment epithelium because of subretinal fluid (SRF), and successful surgical reattachment is not predictive of total visual recovery. As retinal iron overload exacerbates cell death in retinal diseases, we assessed iron as a predictive marker and therapeutic target for RD. In the vitreous and SRF from patients with RD, we measured increased iron and transferrin (TF) saturation that is correlated with poor visual recovery. In ex vivo and in vivo RD models, iron induces immediate necrosis and delayed apoptosis. We demonstrate that TF decreases both apoptosis and necroptosis induced by RD, and using RNA sequencing, pathways mediating the neuroprotective effects of TF are identified. Since toxic iron accumulates in RD, we propose TF supplementation as an adjunctive therapy to surgery for improving the visual outcomes of patients with RD.


Subject(s)
Eye Diseases, Hereditary/metabolism , Iron/metabolism , Iron/toxicity , Neuroprotection , Retinal Detachment/metabolism , Transferrin/metabolism , Aged , Animals , Apoptosis/drug effects , Disease Models, Animal , Eye Diseases, Hereditary/surgery , Female , Humans , Iron/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Necrosis , Photoreceptor Cells, Vertebrate/metabolism , Rats , Rats, Long-Evans , Rats, Wistar , Retina/metabolism , Retinal Detachment/surgery , Retinal Pigment Epithelium/metabolism , Subretinal Fluid/metabolism , Transferrin/genetics
9.
Prog Retin Eye Res ; 63: 20-68, 2018 03.
Article in English | MEDLINE | ID: mdl-29126927

ABSTRACT

Macular edema consists of intra- or subretinal fluid accumulation in the macular region. It occurs during the course of numerous retinal disorders and can cause severe impairment of central vision. Major causes of macular edema include diabetes, branch and central retinal vein occlusion, choroidal neovascularization, posterior uveitis, postoperative inflammation and central serous chorioretinopathy. The healthy retina is maintained in a relatively dehydrated, transparent state compatible with optimal light transmission by multiple active and passive systems. Fluid accumulation results from an imbalance between processes governing fluid entry and exit, and is driven by Starling equation when inner or outer blood-retinal barriers are disrupted. The multiple and intricate mechanisms involved in retinal hydro-ionic homeostasis, their molecular and cellular basis, and how their deregulation lead to retinal edema, are addressed in this review. Analyzing the distribution of junction proteins and water channels in the human macula, several hypotheses are raised to explain why edema forms specifically in the macular region. "Pure" clinical phenotypes of macular edema, that result presumably from a single causative mechanism, are detailed. Finally, diabetic macular edema is investigated, as a complex multifactorial pathogenic example. This comprehensive review on the current understanding of macular edema and its mechanisms opens perspectives to identify new preventive and therapeutic strategies for this sight-threatening condition.


Subject(s)
Macular Edema/physiopathology , Blood-Retinal Barrier , Central Serous Chorioretinopathy/complications , Central Serous Chorioretinopathy/physiopathology , Choroidal Neovascularization/complications , Choroidal Neovascularization/physiopathology , Diabetic Retinopathy/complications , Diabetic Retinopathy/physiopathology , Fluorescein Angiography , Humans , Macular Edema/diagnosis , Macular Edema/prevention & control , Retinal Vein Occlusion/complications , Retinal Vein Occlusion/physiopathology , Retinal Vessels/physiopathology , Subretinal Fluid , Tomography, Optical Coherence
10.
Sci Rep ; 7(1): 8834, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821742

ABSTRACT

In diabetic retinopathy, the exact mechanisms leading to retinal capillary closure and to retinal barriers breakdown remain imperfectly understood. Rho-associated kinase (ROCK), an effector of the small GTPase Rho, involved in cytoskeleton dynamic regulation and cell polarity is activated by hyperglycemia. In one year-old Goto Kakizaki (GK) type 2 diabetic rats retina, ROCK-1 activation was assessed by its cellular distribution and by phosphorylation of its substrates, MYPT1 and MLC. In both GK rat and in human type 2 diabetic retinas, ROCK-1 is activated and associated with non-apoptotic membrane blebbing in retinal vessels and in retinal pigment epithelium (RPE) that respectively form the inner and the outer barriers. Activation of ROCK-1 induces focal vascular constrictions, endoluminal blebbing and subsequent retinal hypoxia. In RPE cells, actin cytoskeleton remodeling and membrane blebs in RPE cells contributes to outer barrier breakdown. Intraocular injection of fasudil, significantly reduces both retinal hypoxia and RPE barrier breakdown. Diabetes-induced cell blebbing may contribute to ischemic maculopathy and represent an intervention target.


Subject(s)
Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Retinal Pigment Epithelium/metabolism , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Aged , Animals , Biomarkers , Case-Control Studies , Cytoskeleton/metabolism , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Female , Fluorescent Antibody Technique , Humans , Hypoxia/metabolism , Immunohistochemistry , Male , Middle Aged , Rats , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Vessels/ultrastructure , rho-Associated Kinases/genetics
11.
J Cell Mol Med ; 21(12): 3453-3466, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28661040

ABSTRACT

Ageing and alteration of the functions of the retinal pigment epithelium (RPE) are at the origin of lost of vision seen in age-related macular degeneration (AMD). The RPE is known to be vulnerable to high-energy blue light. The white light-emitting diodes (LED) commercially available have relatively high content of blue light, a feature that suggest that they could be deleterious for this retinal cell layer. The aim of our study was to investigate the effects of "white LED" exposure on RPE. For this, commercially available white LEDs were used for exposure experiments on Wistar rats. Immunohistochemical stain on RPE flat mount, transmission electron microscopy and Western blot were used to exam the RPE. LED-induced RPE damage was evaluated by studying oxidative stress, stress response pathways and cell death pathways as well as the integrity of the outer blood-retinal barrier (BRB). We show that white LED light caused structural alterations leading to the disruption of the outer blood-retinal barrier. We observed an increase in oxidized molecules, disturbance of basal autophagy and cell death by necrosis. We conclude that white LEDs induced strong damages in rat RPE characterized by the breakdown of the BRB and the induction of necrotic cell death.


Subject(s)
Blood-Retinal Barrier/radiation effects , Eye Proteins/genetics , Light/adverse effects , Protein Kinase C/genetics , Retinal Pigment Epithelium/radiation effects , Animals , Autophagy/genetics , Autophagy/radiation effects , Blood-Retinal Barrier/metabolism , Eye Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Lighting/adverse effects , Male , Necrosis/etiology , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Oxidative Stress/radiation effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Wistar , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Tissue Culture Techniques
12.
Mol Brain ; 9(1): 64, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27267879

ABSTRACT

BACKGROUND: Amyloid precursor protein knockout mice (APP-KO) have impaired differentiation of amacrine and horizontal cells. APP is part of a gene family and its paralogue amyloid precursor-like protein 2 (APLP2) has both shared as well as distinct expression patterns to APP, including in the retina. Given the impact of APP in the retina we investigated how APLP2 expression affected the retina using APLP2 knockout mice (APLP2-KO). RESULTS: Using histology, morphometric analysis with noninvasive imaging technique and electron microscopy, we showed that APLP2-KO retina displayed abnormal formation of the outer synaptic layer, accompanied with greatly impaired photoreceptor ribbon synapses in adults. Moreover, APLP2-KO displayed a significant decease in ON-bipolar, rod bipolar and type 2 OFF-cone bipolar cells (36, 21 and 63 %, respectively). Reduction of the number of bipolar cells was accompanied with disrupted dendrites, reduced expression of metabotropic glutamate receptor 6 at the dendritic tips and alteration of axon terminals in the OFF laminae of the inner plexiform layer. In contrast, the APP-KO photoreceptor ribbon synapses and bipolar cells were intact. The APLP2-KO retina displayed numerous phenotypic similarities with the congenital stationary night blindness, a non-progressive retinal degeneration disease characterized by the loss of night vision. The pathological phenotypes in the APLP2-KO mouse correlated to altered transcription of genes involved in pre- and postsynatic structure/function, including CACNA1F, GRM6, TRMP1 and Gα0, and a normal scotopic a-wave electroretinogram amplitude, markedly reduced scotopic electroretinogram b-wave and modestly reduced photopic cone response. This confirmed the impaired function of the photoreceptor ribbon synapses and retinal bipolar cells, as is also observed in congenital stationary night blindness. Since congenital stationary night blindness present at birth, we extended our analysis to retinal differentiation and showed impaired differentiation of different bipolar cell subtypes and an altered temporal sequence of development from OFF to ON laminae in the inner plexiform layer. This was associated with the altered expression patterns of bipolar cell generation and differentiation factors, including MATH3, CHX10, VSX1 and OTX2. CONCLUSIONS: These findings demonstrate that APLP2 couples retina development and synaptic genes and present the first evidence that APLP2 expression may be linked to synaptic disease.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Eye Diseases, Hereditary/genetics , Gene Deletion , Genetic Diseases, X-Linked/genetics , Myopia/genetics , Night Blindness/genetics , Aging/pathology , Amacrine Cells/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Cell Differentiation , Complement System Proteins/metabolism , Dendrites/metabolism , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/physiopathology , Genetic Diseases, X-Linked/pathology , Genetic Diseases, X-Linked/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myopia/pathology , Myopia/physiopathology , Neurogenesis , Night Blindness/pathology , Night Blindness/physiopathology , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/ultrastructure , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/ultrastructure , Synaptic Transmission , Transcription Factors/metabolism , Transcription, Genetic
13.
Free Radic Biol Med ; 89: 1105-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26454080

ABSTRACT

Iron is essential for retinal function but contributes to oxidative stress-mediated degeneration. Iron retinal homeostasis is highly regulated and transferrin (Tf), a potent iron chelator, is endogenously secreted by retinal cells. In this study, therapeutic potential of a local Tf delivery was evaluated in animal models of retinal degeneration. After intravitreal injection, Tf spread rapidly within the retina and accumulated in photoreceptors and retinal pigment epithelium, before reaching the blood circulation. Tf injected in the vitreous prior and, to a lesser extent, after light-induced retinal degeneration, efficiently protected the retina histology and function. We found an association between Tf treatment and the modulation of iron homeostasis resulting in a decrease of iron content and oxidative stress marker. The immunomodulation function of Tf could be seen through a reduction in macrophage/microglial activation as well as modulated inflammation responses. In a mouse model of hemochromatosis, Tf had the capacity to clear abnormal iron accumulation from retinas. And in the slow P23H rat model of retinal degeneration, a sustained release of Tf in the vitreous via non-viral gene therapy efficently slowed-down the photoreceptors death and preserved their function. These results clearly demonstrate the synergistic neuroprotective roles of Tf against retinal degeneration and allow identify Tf as an innovative and not toxic therapy for retinal diseases associated with oxidative stress.


Subject(s)
Disease Models, Animal , Inflammation/prevention & control , Iron/toxicity , Oxidative Stress/drug effects , Retinal Degeneration/prevention & control , Transferrin/pharmacology , Animals , Cells, Cultured , Homeostasis/drug effects , Immunoenzyme Techniques , Inflammation/chemically induced , Male , Mice , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Free Radic Biol Med ; 84: 373-384, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25863264

ABSTRACT

Spectra of "white LEDs" are characterized by an intense emission in the blue region of the visible spectrum, absent in daylight spectra. This blue component and the high intensity of emission are the main sources of concern about the health risks of LEDs with respect to their toxicity to the eye and the retina. The aim of our study was to elucidate the role of blue light from LEDs in retinal damage. Commercially available white LEDs and four different blue LEDs (507, 473, 467, and 449nm) were used for exposure experiments on Wistar rats. Immunohistochemical stain, transmission electron microscopy, and Western blot were used to exam the retinas. We evaluated LED-induced retinal cell damage by studying oxidative stress, stress response pathways, and the identification of cell death pathways. LED light caused a state of suffering of the retina with oxidative damage and retinal injury. We observed a loss of photoreceptors and the activation of caspase-independent apoptosis, necroptosis, and necrosis. A wavelength dependence of the effects was observed. Phototoxicity of LEDs on the retina is characterized by a strong damage of photoreceptors and by the induction of necrosis.


Subject(s)
Apoptosis , Lighting/adverse effects , Oxidative Stress , Retina/radiation effects , Animals , Male , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/radiation effects , Rats, Wistar , Retina/metabolism , Retina/pathology , Retinal Degeneration/etiology , Retinal Degeneration/metabolism
15.
J Cell Mol Med ; 19(7): 1646-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25781645

ABSTRACT

Light-induced retinal degeneration is characterized by photoreceptor cell death. Many studies showed that photoreceptor demise is caspase-independent. In our laboratory we showed that leucocyte elastase inhibitor/LEI-derived DNase II (LEI/L-DNase II), a caspase-independent apoptotic pathway, is responsible for photoreceptor death. In this work, we investigated the activation of a pro-survival kinase, the protein kinase C (PKC) zeta. We show that light exposure induced PKC zeta activation. PKC zeta interacts with LEI/L-DNase II and controls its DNase activity by impairing its nuclear translocation. These results highlight the role of PKC zeta in retinal physiology and show that this kinase can control caspase-independent pathways.


Subject(s)
Endodeoxyribonucleases/metabolism , Light , Protein Kinase C/metabolism , Retinal Degeneration/enzymology , Amino Acid Sequence , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , HeLa Cells , Humans , Male , Molecular Sequence Data , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase Inhibitors/pharmacology , Rats , Retina/drug effects , Retina/enzymology , Retina/pathology , Retinal Degeneration/pathology , Serpins/metabolism
16.
Mol Vis ; 20: 908-20, 2014.
Article in English | MEDLINE | ID: mdl-24966662

ABSTRACT

PURPOSE: To evaluate whether anti-vascular endothelial growth factor (VEGF) neutralizing antibodies injected in the vitreous of rat eyes influence retinal microglia and macrophage activation. To dissociate the effect of anti-VEGF on microglia and macrophages subsequent to its antiangiogenic effect, we chose a model of acute intraocular inflammation. METHODS: Lewis rats were challenged with systemic lipopolysaccharide (LPS) injection and concomitantly received 5 µl of rat anti-VEGF-neutralizing antibody (1.5 mg/ml) in the vitreous. Rat immunoglobulin G (IgG) isotype was used as the control. The effect of anti-VEGF was evaluated at 24 and 48 h clinically (uveitis scores), biologically (cytokine multiplex analysis in ocular media), and histologically (inflammatory cell counts on eye sections). Microglia and macrophages were immunodetected with ionized calcium-binding adaptor molecule 1 (IBA1) staining and counted based on their differential shapes (round amoeboid or ramified dendritiform) on sections and flatmounted retinas using confocal imaging and automatic quantification. Activation of microglia was also evaluated with inducible nitric oxide synthase (iNOS) and IBA1 coimmunostaining. Coimmunolocalization of VEGF receptor 1 and 2 (VEGF-R1 and R2) with IBA1 was performed on eye sections with or without anti-VEGF treatment. RESULTS: Neutralizing rat anti-VEGF antibodies significantly decreased ocular VEGF levels but did not decrease the endotoxin-induced uveitis (EIU) clinical score or the number of infiltrating cells and cytokines in ocular media (interleukin [IL]-1ß, IL-6, tumor necrosis factor [TNF]-α, and monocyte chemoattractant protein [MCP]-1). Eyes treated with anti-VEGF showed a significantly decreased number of activated microglia and macrophages in the retina and the choroid and decreased iNOS-positive microglia. IBA1-positive cells expressed VEGF-R1 and R2 in the inflamed retina. CONCLUSIONS: Microglia and macrophages expressed VEGF receptors, and intravitreous anti-VEGF influenced the microglia and macrophage activation state. Taking into account that anti-VEGF drugs are repeatedly injected in the vitreous of patients with retinal diseases, part of their effects could result from unsuspected modulation of the microglia activation state. This should be further studied in other ocular pathogenic conditions and human pathology.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Macrophage Activation/drug effects , Microglia/pathology , Retina/pathology , Uveitis/drug therapy , Uveitis/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antibodies, Neutralizing/pharmacology , Calcium-Binding Proteins/metabolism , Cell Count , Disease Models, Animal , Humans , Lipopolysaccharides , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Neutralization Tests , Nitric Oxide Synthase Type II/metabolism , Rats , Receptors, Vascular Endothelial Growth Factor/metabolism , Retina/drug effects , Retina/enzymology , Vascular Endothelial Growth Factor A/metabolism
17.
PLoS One ; 8(11): e81600, 2013.
Article in English | MEDLINE | ID: mdl-24312324

ABSTRACT

AIMS/HYPOTHESIS: Diabetic macular edema represents the main cause of visual loss in diabetic retinopathy. Besides inner blood retinal barrier breakdown, the role of the outer blood retinal barrier breakdown has been poorly analyzed. We characterized the structural and molecular alterations of the outer blood retinal barrier during the time course of diabetes, focusing on PKCζ, a critical protein for tight junction assembly, known to be overactivated by hyperglycemia. METHODS: Studies were conducted on a type2 diabetes Goto-Kakizaki rat model. PKCζ level and subcellular localization were assessed by immunoblotting and immunohistochemistry. Cell death was detected by TUNEL assays. PKCζ level on specific layers was assessed by laser microdissection followed by Western blotting. The functional role of PKCζ was then evaluated in vivo, using intraocular administration of its specific inhibitor. RESULTS: PKCζ was localized in tight junction protein complexes of the retinal pigment epithelium and in photoreceptors inner segments. Strikingly, in outer segment PKCζ staining was restricted to cone photoreceptors. Short-term hyperglycemia induced activation and delocalization of PKCζ from both retinal pigment epithelium junctions and cone outer segment. Outer blood retinal barrier disruption and photoreceptor cone degeneration characterized long-term hyperglycemia. In vivo, reduction of PKCζ overactivation using a specific inhibitor, restored its tight-junction localization and not only improved the outer blood retinal barrier, but also reduced photoreceptor cell-death. CONCLUSIONS: In the retina, hyperglycemia induced overactivation of PKCζ is associated with outer blood retinal barrier breakdown and photoreceptor degeneration. In vivo, short-term inhibition of PKCζ restores the outer barrier structure and reduces photoreceptor cell death, identifying PKCζ as a potential target for early and underestimated diabetes-induced retinal pathology.


Subject(s)
Blood-Retinal Barrier/metabolism , Diabetic Retinopathy/metabolism , Protein Kinase C/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/pathology , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/pathology , Hyperglycemia/enzymology , Hyperglycemia/metabolism , Hyperglycemia/pathology , NF-kappa B/metabolism , Nerve Tissue Proteins , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism
18.
Bull Acad Natl Med ; 197(3): 661-74; discussion 674-5, 2013 Mar.
Article in French | MEDLINE | ID: mdl-25163348

ABSTRACT

Iron is necessary for cell metabolism, but excess iron can be toxic Iron can generate oxygen free radicals through the Fenton reaction. Iron accumulation has been observed in the retina of patients with age-related macular degeneration (AMD). We have shown its accumulation in photoreceptor segments in two animal models of genetic retinal degeneration (RCS rats and Rd10 mice). In these rodents, hTf, injected intraperitoneally or expressed by genetic modification, delayed photoreceptor degeneration. Our studies highlight the therapeutic potential of Tf in degenerative processes such as retinitis pigmentosa and AMD.


Subject(s)
Iron-Regulatory Proteins/metabolism , Iron/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Animals , Disease Models, Animal , Humans , Mice , Rats , Retinal Degeneration/genetics , Retinal Degeneration/pathology
19.
PLoS One ; 7(11): e49036, 2012.
Article in English | MEDLINE | ID: mdl-23152847

ABSTRACT

We have previously shown that the eye is a mineralocorticoid-sensitive organ and we now question the role of mineralocorticoid receptor (MR) in ocular inflammation. The endotoxin-induced uveitis (EIU), a rat model of human intraocular inflammation, was induced by systemic administration of lipopolysaccharide (LPS). Evaluations were made 6 and 24 hours after intraocular injection of aldosterone (simultaneous to LPS injection). Three hours after onset of EIU, the MR and the glucocorticoid metabolizing enzyme 11-beta hydroxysteroid dehydrogenase type 2 (11ß-HSD2) expression were down-regulated in iris/ciliary body and the corticosterone concentration was increased in aqueous humor, altering the normal MR/glucocorticoid receptor (GR) balance. At 24 hours, the GR expression was also decreased. In EIU, aldosterone reduced the intensity of clinical inflammation in a dose-dependent manner. The clinical benefit of aldosterone was abrogated in the presence of the MR antagonist (RU26752) and only partially with the GR antagonist (RU38486). Aldosterone reduced the release of inflammatory mediators (6 and 24 hours: TNF-α, IFN-γ, MIP-1α) in aqueous humor and the number of activated microglia/macrophages. Aldosterone partly prevented the uveitis-induced MR down-regulation. These results suggest that MR expression and activation in iris/ciliary body could protect the ocular structures against damages induced by EIU.


Subject(s)
Anti-Inflammatory Agents/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Uveitis/metabolism , Uveitis/pathology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Aldosterone/administration & dosage , Aldosterone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Chemokines/metabolism , Ciliary Body/enzymology , Ciliary Body/pathology , Down-Regulation/drug effects , Down-Regulation/genetics , Endotoxins , Female , Humans , Inflammation Mediators/metabolism , Intravitreal Injections , Iris/drug effects , Iris/enzymology , Iris/pathology , Lipopolysaccharides , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Rats, Inbred Lew , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Signal Transduction/drug effects , Spironolactone/administration & dosage , Spironolactone/pharmacology , Uveitis/chemically induced , Uveitis/drug therapy
20.
Angiogenesis ; 15(4): 609-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22869002

ABSTRACT

Inflammatory neovascularization, such as choroidal neovascularization (CNV), occur in the presence of Notch expressing macrophages. DLL4s anti-angiogenic effect on endothelial cells (EC) has been widely recognized, but its influence on Notch signaling on macrophages and its overall effect in inflammatory neovascularization is not well understood. We identified macrophages and ECs as the main Notch 1 and Notch 4 expressing cells in CNV. A soluble fraction spanning Ser28-Pro525 of the murine extracellular DLL4 domain (sDLL4/28-525) activated the Notch pathway, as it induces Notch target genes in macrophages and ECs and inhibited EC proliferation and vascular sprouting in aortic rings. In contrast, sDLL4/28-525 increased pro-angiogenic VEGF, and IL-1ß expression in macrophages responsible for increased vascular sprouting observed in aortic rings incubated in conditioned media from sDLL4/28-525 stimulated macrophages. In vivo, Dll4(+/-) mice developed significantly more CNV and sDLL4/28-525 injections inhibited CNV in Dll4(+/-) CD1 mice. Similarly, sDLL4/28-525 inhibited CNV in C57Bl6 and its effect was reversed by a γ-secretase inhibitor that blocks Notch signaling. The inhibition occurred despite increased VEGF, IL-1ß expression in infiltrating inflammatory macrophages in sDLL4/28-525 treated mice and might be due to direct inhibition of EC proliferation in laser-induced CNV as demonstrated by EdU labelling in vivo. In conclusion, Notch activation on macrophages and ECs leads to opposing effects in inflammatory neovascularization in situations such as CNV.


Subject(s)
Choroidal Neovascularization/prevention & control , Endothelium, Vascular/physiopathology , Intercellular Signaling Peptides and Proteins/physiology , Macrophages, Peritoneal/physiology , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Blotting, Western , Calcium-Binding Proteins , DNA Primers , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL