Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biomed Sci ; 31(1): 48, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730462

ABSTRACT

Retinal degenerative diseases, including diabetic retinopathy (DR) and age-related macular degeneration (AMD), loom as threats to vision, causing detrimental effects on the structure and function of the retina. Central to understanding these diseases, is the compromised state of the blood-retinal barrier (BRB), an effective barrier that regulates the influx of immune and inflammatory components. Whether BRB breakdown initiates retinal distress, or is a consequence of disease progression, remains enigmatic. Nevertheless, it is an indication of retinal dysfunction and potential vision loss.The intricate intercellular dialogues among retinal cell populations remain unintelligible in the complex retinal milieu, under conditions of inflammation and oxidative stress. The retina, a specialized neural tissue, sustains a ceaseless demand for oxygen and nutrients from two vascular networks. The BRB orchestrates the exchange of molecules and fluids within this specialized region, comprising the inner BRB (iBRB) and the outer BRB (oBRB). Extracellular vesicles (EVs) are small membranous structures, and act as messengers facilitating intercellular communication in this milieu.EVs, both from retinal and peripheral immune cells, increase complexity to BRB dysfunction in DR and AMD. Laden with bioactive cargoes, these EVs can modulate the retinal microenvironment, influencing disease progression. Our review delves into the multifaceted role of EVs in retinal degenerative diseases, elucidating the molecular crosstalk they orchestrate, and their microRNA (miRNA) content. By shedding light on these nanoscale messengers, from their biogenesis, release, to interaction and uptake by target cells, we aim to deepen the comprehension of BRB dysfunction and explore their therapeutic potential, therefore increasing our understanding of DR and AMD pathophysiology.


Subject(s)
Blood-Retinal Barrier , Extracellular Vesicles , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/physiopathology , Extracellular Vesicles/metabolism , Humans , Diabetic Retinopathy/physiopathology , Diabetic Retinopathy/metabolism , Retinal Diseases/physiopathology , Retinal Diseases/metabolism , Macular Degeneration/physiopathology , Macular Degeneration/metabolism , Animals
2.
Exp Eye Res ; 235: 109627, 2023 10.
Article in English | MEDLINE | ID: mdl-37619829

ABSTRACT

The main purpose of this study is to analyze the effects of unilateral optic nerve crush in the gene expression of pro- and anti-inflammatory mediators, and gliosis markers in injured and contralateral retinas. Retinas from intact, unilaterally optic nerve injured or sham-operated C57BL/6J mice were analyzed 1, 3, 9 and 30 days after the surgery (n = 5/group and time point) and the relative expression of TGF-ß1, IL-1ß, TNF-α, Iba1, AQP4, GFAP, MHCII, and TSPO was analyzed in injured and contralateral using qPCR. The results indicated that compared with intact retinas, sham-operated animals showed an early (day 1) upregulation of IL-1ß, TNF-α and TSPO and a late (day 30) upregulation of TNF-α. In sham-contralateral retinas, TNF-α and TSPO mRNA expression were upregulated and day 30 while GFAP, Iba1, AQP4 and MHCII downregulated at day 9. Compared with sham-operated animals, in retinas affected by optic nerve crush GFAP and TSPO upregulated at day 1 and TNF-α, Iba1, AQP4 and MHCII at day 3. In the crushed-contralateral retinas, TGF-ß1, TNF-α, Iba1 and MHCII were upregulated at day 1. TSPO was upregulated up to day 30 whereas TGF-ß1 and Iba1 downregulated after day 9. In conclusion, both sham surgery and optic nerve crush changed the profile of inflammatory and gliosis markers in the injured and contralateral retinas, changes that were more pronounced for optic nerve crush when compared to sham.


Subject(s)
Optic Nerve Injuries , Transforming Growth Factor beta1 , Mice , Animals , Transforming Growth Factor beta1/pharmacology , Retinal Ganglion Cells/metabolism , Gliosis/metabolism , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Neuroinflammatory Diseases , Tumor Necrosis Factor-alpha/metabolism , Mice, Inbred C57BL , Retina/metabolism , Optic Nerve/metabolism , Nerve Crush/methods
3.
Int J Mol Sci ; 24(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36834893

ABSTRACT

Retinal organotypic cultures (ROCs) are used as an in vivo surrogate to study retinal ganglion cell (RGC) loss and neuroprotection. In vivo, the gold standard to study RGC degeneration and neuroprotection is optic nerve lesion. We propose here to compare the course of RGC death and glial activation between both models. The left optic nerve of C57BL/6 male mice was crushed, and retinas analyzed from 1 to 9 days after the injury. ROCs were analyzed at the same time points. As a control, intact retinas were used. Retinas were studied anatomically to assess RGC survival, microglial, and macroglial activation. Macroglial and microglial cells showed different morphological activation between models and were activated earlier in ROCs. Furthermore, microglial cell density in the ganglion cell layer was always lower in ROCs than in vivo. RGC loss after axotomy and in vitro followed the same trend up to 5 days. Thereafter, there was an abrupt decrease in viable RGCs in ROCs. However, RGC somas were still immuno-identified by several molecular markers. ROCs are useful for proof-of-concept studies on neuroprotection, but long-term experiments should be carried out in vivo. Importantly, the differential glial activation observed between models and the concomitant death of photoreceptors that occurs in vitro may alter the efficacy of RGC neuroprotective therapies when tested in in vivo models of optic nerve injury.


Subject(s)
Microphysiological Systems , Optic Nerve Injuries , Mice , Animals , Male , Mice, Inbred C57BL , Retina/metabolism , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolism , Axotomy , Cell Survival
4.
Glia ; 68(12): 2705-2724, 2020 12.
Article in English | MEDLINE | ID: mdl-32645245

ABSTRACT

Glaucoma is a degenerative disease that causes irreversible loss of vision and is characterized by retinal ganglion cell (RGC) loss. Others and we have demonstrated that chronic neuroinflammation mediated by reactive microglial cells plays a role in glaucomatous pathology. Exosomes are extracellular vesicles released by most cells, including microglia, that mediate intercellular communication. The role of microglial exosomes in glaucomatous degeneration remains unknown. Taking the prominent role of microglial exosomes in brain neurodegenerative diseases, we studied the contribution of microglial-derived exosomes to the inflammatory response in experimental glaucoma. Microglial cells were exposed to elevated hydrostatic pressure (EHP), to mimic elevated intraocular pressure, the main risk factor for glaucoma. Naïve microglia (BV-2 cells or retinal microglia) were exposed to exosomes derived from BV-2 cells under EHP conditions (BV-Exo-EHP) or cultured in control pressure (BV-Exo-Control). We found that BV-Exo-EHP increased the production of pro-inflammatory cytokines, promoted retinal microglia motility, phagocytic efficiency, and proliferation. Furthermore, the incubation of primary retinal neural cell cultures with BV-Exo-EHP increased cell death and the production of reactive oxygen species. Exosomes derived from retinal microglia (MG-Exo-Control or MG-Exo-EHP) were injected in the vitreous of C57BL/6J mice. MG-Exo-EHP sustained activation of retinal microglia, mediated cell death, and impacted RGC number. Herein, we show that exosomes derived from retinal microglia have an autocrine function and propagate the inflammatory signal in conditions of elevated pressure, contributing to retinal degeneration in glaucomatous conditions.


Subject(s)
Exosomes , Glaucoma , Animals , Inflammation , Mice , Mice, Inbred C57BL , Microglia , Retinal Ganglion Cells
5.
Exp Eye Res ; 199: 108195, 2020 10.
Article in English | MEDLINE | ID: mdl-32841650

ABSTRACT

Increasing evidence points to inflammation as a key factor in the pathogenesis of diabetic retinopathy (DR). Choroidal inflammatory changes in diabetes have been reported and in vivo choroidal thickness (CT) has been searched as a marker of retinopathy with contradictory results. We aimed to investigate the early stages in the retina and choroid in an animal model of Type 1 diabetes. Type 1 diabetes was induced in male Wistar rats via a single i.p. streptozotocin injection. At 8 weeks after disease onset, CT, choroidal vascular density, VEGF and VEGFR2 expression, microglial cell and pericyte distribution were evaluated. Diabetic rats showed no significant change in CT and choroidal vascular density. A widened pericyte-free gap between the retinal pigment epithelium and the choroid was observed in diabetic rats. The immunoreactivity of VEGFR2 was decreased in the retina of diabetic rats, despite no statistically significant difference in the immunoreactivity of VEGF. The density of microglial cells significantly increased in the choroid and retina of diabetic rats. Reactive microglial cells were found to be more abundant in the choroid of diabetic rats. Evidences of the interconnection between the superficial, intermediate, and deep plexuses of the retina were also observed. At early stages, Type 1 diabetes does not affect choroidal thickness and choroidal vascular density. Proliferation and reactivity of microglial cells occurs in the choroidal stroma and the retina. The expression of VEGFR2 decreases in the retina.


Subject(s)
Choroid/pathology , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/pathology , Retina/pathology , Animals , Cell Proliferation , Disease Progression , Fluorescein Angiography/methods , Fundus Oculi , Male , Rats , Rats, Wistar
6.
Int J Mol Sci ; 21(19)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33007835

ABSTRACT

Glaucoma is a progressive chronic retinal degenerative disease and a leading cause of global irreversible blindness, characterized by optic nerve damage and retinal ganglion cell (RGC) death. Elevated intraocular pressure (IOP) is a main risk factor of glaucoma. Neuroinflammation plays an important role in glaucoma. We have been demonstrating that elevated pressure triggers microglia reactivity that contribute to the loss of RGCs. Adenosine, acting on adenosine receptors, is a crucial modulator of microglia phenotype. Microglia express all adenosine receptors. Previously, we demonstrated that the activation of adenosine A3 receptor (A3R) affords protection to the retina, including RGCs, unveiling the possibility for a new strategy for glaucoma treatment. Since microglial cells express A3R, we now studied the ability of a selective A3R agonist (2-Cl-IB-MECA) in controlling microglia reactivity induced by elevated hydrostatic pressure (EHP), used to mimic elevated IOP. The activation of A3R reduced EHP-induced inducible nitric oxide synthase (iNOS) expression, microglia migration and phagocytosis in BV-2 cells. In retinal microglia, proliferation and phagocytosis elicited by EHP were also decreased by A3R activation. This work demonstrates that 2-Cl-IB-MECA, the selective agonist of A3R, is able to hinder microglia reactivity, suggesting that A3R agonists could afford protection against glaucomatous degeneration through the control of neuroinflammation.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Glaucoma/drug therapy , Receptor, Adenosine A3/genetics , Adenosine/genetics , Adenosine/pharmacology , Animals , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Microglia/drug effects , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Phagocytosis/drug effects , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
7.
Int J Mol Sci ; 21(7)2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32218163

ABSTRACT

The retinal ganglion cells (RGCs) are the output cells of the retina into the brain. In mammals, these cells are not able to regenerate their axons after optic nerve injury, leaving the patients with optic neuropathies with permanent visual loss. An effective RGCs-directed therapy could provide a beneficial effect to prevent the progression of the disease. Axonal injury leads to the functional loss of RGCs and subsequently induces neuronal death, and axonal regeneration would be essential to restore the neuronal connectivity, and to reestablish the function of the visual system. The manipulation of several intrinsic and extrinsic factors has been proposed in order to stimulate axonal regeneration and functional repairing of axonal connections in the visual pathway. However, there is a missing point in the process since, until now, there is no therapeutic strategy directed to promote axonal regeneration of RGCs as a therapeutic approach for optic neuropathies.


Subject(s)
Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/cytology , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Disease Progression , Humans , Retinal Ganglion Cells/drug effects
8.
Glia ; 67(5): 896-914, 2019 05.
Article in English | MEDLINE | ID: mdl-30667095

ABSTRACT

Glaucoma is a retinal degenerative disease characterized by the loss of retinal ganglion cells and damage of the optic nerve. Recently, we demonstrated that antagonists of adenosine A2A receptor (A2A R) control retinal inflammation and afford protection to rat retinal cells in glaucoma models. However, the precise contribution of microglia to retinal injury was not addressed, as well as the effect of A2A R blockade directly in microglia. Here we show that blocking microglial A2A R prevents microglial cell response to elevated pressure and it is sufficient to protect retinal cells from elevated pressure-induced death. The A2A R antagonist SCH 58261 or the knockdown of A2A R expression with siRNA in microglial cells prevented the increase in microglia response to elevated hydrostatic pressure. Furthermore, in retinal neural cell cultures, the A2A R antagonist decreased microglia proliferation, as well as the expression and release of pro-inflammatory mediators. Microglia ablation prevented neural cell death triggered by elevated pressure. The A2A R blockade recapitulated the effects of microglia depletion, suggesting that blocking A2A R in microglia is able to control neurodegeneration in glaucoma-like conditions. Importantly, in human organotypic retinal cultures, A2A R blockade prevented the increase in reactive oxygen species and the morphological alterations in microglia triggered by elevated pressure. These findings place microglia as the main contributors for retinal cell death during elevated pressure and identify microglial A2A R as a therapeutic target to control retinal neuroinflammation and prevent neural apoptosis elicited by elevated pressure.


Subject(s)
Inflammation/metabolism , Microglia/metabolism , Neurons/physiology , Oxidative Stress/physiology , Receptor, Adenosine A2A/metabolism , Retina/cytology , Adenosine A2 Receptor Antagonists/pharmacology , Adult , Aged , Animals , Animals, Newborn , Cell Death/drug effects , Cell Death/physiology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Inflammation/drug therapy , Male , Microglia/drug effects , Middle Aged , Neurons/drug effects , Organ Culture Techniques , Oxidative Stress/drug effects , Phagocytosis/drug effects , Pyrimidines/pharmacology , Rats , Rats, Wistar , Triazoles/pharmacology , Wounds and Injuries/drug therapy , Wounds and Injuries/metabolism
9.
Graefes Arch Clin Exp Ophthalmol ; 256(10): 1857-1865, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30039271

ABSTRACT

PURPOSE: The aim of this study was to evaluate subfoveal choroidal thickness (SFCT) as a marker of outcome in real-world treatment of diabetic macular edema (DME) and to correlate it with choroidal thicknesses (CT) collected around the fovea. METHODS: Prospective interventional case series included a total of 126 eyes from 126 patients with recently diagnosed DME treated with a 3-monthly loading dose of ranibizumab or aflibercept and PRN thereafter until 24 months (M). CT was manually measured in the central 3500 µm area, subfoveally (SFCT), at 1750 µm right and left from the center in the horizontal plane and at 1750 µm up and down from the center in the vertical plane, by OCT. Anatomic (10% decrease in central retinal thickness) and functional (gain ≥ 5 letters) responses were assessed using univariate and multivariate analyses. The areas under ROC curves were used to assess whether baseline SFCT was a predictor of outcome. RESULTS: CT significantly decreased in all follow-ups (3 months after the 3 injections' loading dose (3M), 6 months (6M), 12 months (12M), 18 months (18M), 24 months (24M)). SFCT and other CT parameters are correlated. SFCT decrease from baseline was related with treatment (p = 0.003 to p < 0.001) but not with anatomic (3M, p = 0.858; 6M p = 0.762) or functional response (3M, p = 0.746; 6M, p = 0.156). SFCT was not found to be predictive of anatomic (AUC = 0.575, p = 0.172) or functional (AUC = 0.515, p = 0.779) outcome. CONCLUSIONS: SFCT is a reliable marker of choroidal thickness. Baseline SFCT decreased with anti-VEGF treatment but did not predict DME outcome.


Subject(s)
Bevacizumab/administration & dosage , Choroid/pathology , Diabetic Retinopathy/drug therapy , Fluorescein Angiography/methods , Macular Edema/drug therapy , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Tomography, Optical Coherence/methods , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Diabetic Retinopathy/complications , Diabetic Retinopathy/diagnosis , Female , Fovea Centralis/pathology , Fundus Oculi , Humans , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/etiology , Male , Middle Aged , Prospective Studies , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity
10.
Ophthalmic Res ; 57(2): 77-86, 2017.
Article in English | MEDLINE | ID: mdl-27618367

ABSTRACT

Glaucoma, a leading cause of blindness worldwide, is a degenerative disease characterized by retinal ganglion cell (RGC) loss and optic nerve atrophy. Elevated intraocular pressure (IOP) is a main risk factor for onset and progression of the disease. Since increased IOP is the only modifiable risk factor, relevant models for glaucoma would comprise RGC and optic nerve damage triggered by ocular hypertension. Animal models of glaucoma have greatly contributed to the understanding of the molecular mechanisms of this pathology, and they have also facilitated the development of new pharmacological interventions. Although animal models of glaucoma have provided valuable information about the disease, there is still no ideal model for studying glaucoma due to its complexity. There is a recognized demand for in vitro models that can replace or reduce the need for animal experiments. Several in vitro models have emerged as a great opportunity in the field of glaucoma research, helping to clarify the mechanisms involved in disease progression. Several types of equipment have been developed to expose cells and tissue cultures to elevated pressures. Herein, we discuss the methodology used to increase pressure, the main findings, and the relevance of in vitro models for the study of the pathophysiology of glaucoma.


Subject(s)
Blindness/etiology , Glaucoma/complications , Intraocular Pressure , Animals , Disease Models, Animal , Glaucoma/physiopathology , Humans , Risk Factors
11.
Ophthalmic Res ; 58(3): 150-161, 2017.
Article in English | MEDLINE | ID: mdl-28793297

ABSTRACT

Calcium dobesilate (CaD) has been prescribed to some patients in the early stages of diabetic retinopathy to delay its progression. We previously reported that the treatment of diabetic animals (4 weeks of diabetes) with CaD, during the last 10 days of diabetes, prevents blood-retinal barrier breakdown. Here, we aimed to investigate whether later treatment of diabetic rats with CaD would reverse inflammatory processes in the retina. Diabetes was induced with streptozotocin, and 6 weeks after diabetes onset, CaD (100 mg/kg/day) was administered for 2 weeks. The treatment with CaD significantly increased glial fibrillary acidic protein (GFAP) levels in the retina of nondiabetic animals (138.6 ± 12.8% of control) and enhanced the diabetes-induced increase in GFAP levels (174.8 ± 5.6% of control). In addition, CaD prevented the increase in mRNA and protein expression of tumor necrosis factor and interleukin-1ß, as well as the formation of oxidized carbonyl residues and the increase in nitrotyrosine immunoreactivity, particularly in the ganglion cell layer of diabetic animals. We demonstrate that the treatment of diabetic animals with CaD can reverse the established proinflammatory processes in the retina. These beneficial effects appear to be attributed, at least partially, to the antioxidant properties of CaD.


Subject(s)
Calcium Dobesilate/pharmacology , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/complications , Diabetic Retinopathy/prevention & control , Inflammation/prevention & control , Oxidative Stress/drug effects , Retina/pathology , Animals , Apoptosis/drug effects , Blood-Retinal Barrier/drug effects , Diabetes Mellitus, Type 1/metabolism , Diabetic Retinopathy/diagnosis , Diabetic Retinopathy/metabolism , Hemostatics/pharmacology , Inflammation/metabolism , Inflammation/pathology , Male , Rats , Rats, Wistar , Retina/metabolism
12.
Nanomedicine ; 13(6): 2101-2113, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28428052

ABSTRACT

People affected with ocular diseases will significantly increase over the next decades, and, consequently, a substantial increase in health costs is expected. Diabetic retinopathy is the most common chronic complication of diabetes. The treatment of eye diseases affecting the posterior segment, such as diabetic retinopathy, is quite challenging due to the anatomy, physiology and biochemistry of the eye. Therefore, the development of new therapeutics for posterior eye diseases has been a major focus of pharmaceutical research in the area of vision sciences. Several nanosystems already offer efficient solutions for ophthalmological conditions, targeting internal eye tissues, as the retina, and many novel products are expected to appear hereafter. This review provides an insight on nanoparticle-based solutions for therapies directed to posterior segment of the eye diseases, particularly diabetic retinopathy, the present scenario, and the demands and expectations for the future.


Subject(s)
Diabetic Retinopathy/therapy , Nanomedicine , Nanoparticles/therapeutic use , Drug Delivery Systems , Humans
13.
Addict Biol ; 21(4): 755-65, 2016 07.
Article in English | MEDLINE | ID: mdl-25904345

ABSTRACT

Neuropeptide Y (NPY), which is widely expressed in the central nervous system is involved in several neuropathologies including addiction. Here we comprehensively and systematically review alterations on the central NPY system induced by several drugs. We report on the effects of psychostimulants [cocaine, amphetamine, methamphetamine, 3,4-methylenedioxymethamphetamine (MDMA) and nicotine], ethanol, and opioids on NPY protein levels and expression of different NPY receptors. Overall, expression and function of NPY and its receptors are changed under conditions of drug exposure, thus affecting several physiologic behaviors, such as feeding, stress and anxiety. Drugs of abuse differentially affect the components of the NPY system. For example methamphetamine and nicotine lead to a consistent increase in NPY mRNA and protein levels in different brain sites whereas ethanol and opioids decrease NPY mRNA and protein expression. Drug-induced alterations on the different NPY receptors show more complex regulation pattern. Manipulation of the NPY system can have opposing effects on reinforcing and addictive properties of drugs of abuse. NPY can produce pro-addictive effects (nicotine and heroin), but can also exert inhibitory effects on addictive behavior (AMPH, ethanol). Furthermore, NPY can act as a neuroprotective agent in chronically methamphetamine and MDMA-treated rodents. In conclusion, manipulation of the NPY system seems to be a potential target to counteract neural alterations, addiction-related behaviors and cognitive deficits induced by these drugs.


Subject(s)
Amphetamines/pharmacology , Cocaine/pharmacology , Ethanol/pharmacology , Neuropeptide Y/drug effects , Nicotine/pharmacology , Opiate Alkaloids/pharmacology , Animals , Brain/drug effects , Disease Models, Animal , Mice , Rats , Receptors, Neuropeptide Y/drug effects
14.
Ophthalmic Res ; 55(4): 212-8, 2016.
Article in English | MEDLINE | ID: mdl-26959995

ABSTRACT

Caffeine, the major component of coffee, is the most consumed psychostimulant in the world. Caffeine is an adenosine analog and acts as a nonselective adenosine receptor antagonist. The majority of the effects of caffeine are mainly mediated by the blockade of adenosine receptors, and the proved neuroprotective effects of caffeine in brain disorders have been mimicked by the blockade of adenosine A2A receptor (A2AR). A growing body of evidence demonstrates that microglia-mediated neuroinflammation plays a key role in the pathophysiology of brain and retinal diseases. Moreover, the control of microglia reactivity by blocking A2AR has been proposed to be the mechanism underlying the observed protective effects of caffeine. Hence, it is conceivable that caffeine and A2AR antagonists offer therapeutic value for the treatment of retinal diseases, mainly those involving microglia-mediated neuroinflammation.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Caffeine/therapeutic use , Central Nervous System Stimulants/therapeutic use , Neuroprotective Agents/therapeutic use , Retinal Diseases/drug therapy , Adenosine A2 Receptor Antagonists/pharmacology , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Humans , Microglia/drug effects , Neuroprotective Agents/pharmacology , Retina/drug effects
15.
J Neuroinflammation ; 12: 115, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26054642

ABSTRACT

BACKGROUND: Elevated intraocular pressure (IOP) is a major risk factor for glaucoma, a degenerative disease characterized by the loss of retinal ganglion cells (RGCs). There is clinical and experimental evidence that neuroinflammation is involved in the pathogenesis of glaucoma. Since the blockade of adenosine A2A receptor (A2AR) confers robust neuroprotection and controls microglia reactivity in the brain, we now investigated the ability of A2AR blockade to control the reactivity of microglia and neuroinflammation as well as RGC loss in retinal organotypic cultures exposed to elevated hydrostatic pressure (EHP) or lipopolysaccharide (LPS). METHODS: Retinal organotypic cultures were either incubated with LPS (3 µg/mL), to elicit a pro-inflammatory response, or exposed to EHP (+70 mmHg), to mimic increased IOP, for 4 or 24 h, in the presence or absence of the A2AR antagonist SCH 58261 (50 nM). A2AR expression, microglial reactivity and neuroinflammatory response were evaluated by immunohistochemistry, quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). RGC loss was assessed by immunohistochemistry. In order to investigate the contribution of pro-inflammatory mediators to RGC loss, the organotypic retinal cultures were incubated with rabbit anti-tumour necrosis factor (TNF) (2 µg/mL) and goat anti-interleukin-1ß (IL-1ß) (1 µg/mL) antibodies. RESULTS: We report that the A2AR antagonist (SCH 58261) prevented microglia reactivity, increase in pro-inflammatory mediators as well as RGC loss upon exposure to either LPS or EHP. Additionally, neutralization of TNF and IL-1ß prevented RGC loss induced by LPS or EHP. CONCLUSIONS: This work demonstrates that A2AR blockade confers neuroprotection to RGCs by controlling microglia-mediated retinal neuroinflammation and prompts the hypothesis that A2AR antagonists may be a novel therapeutic option to manage glaucomatous disorders.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Apoptosis/drug effects , Hydrostatic Pressure/adverse effects , Inflammation/complications , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/pathology , Animals , Apoptosis/physiology , Glaucoma/drug therapy , Inflammation/physiopathology , Lipopolysaccharides/pharmacology , Models, Animal , Nitric Oxide/metabolism , Organ Culture Techniques , Pyrimidines/pharmacology , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Triazoles/pharmacology
16.
Exp Eye Res ; 140: 65-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26297614

ABSTRACT

Death of retinal neural cells, namely retinal ganglion cells (RGCs), is a characteristic of several retinal neurodegenerative diseases. Although the role of adenosine A3 receptor (A3R) in neuroprotection is controversial, A3R activation has been reported to afford protection against several brain insults, with few studies in the retina. In vitro models (retinal neural and organotypic cultures) and animal models [ischemia-reperfusion (I-R) and partial optic nerve transection (pONT)] were used to study the neuroprotective properties of A3R activation against retinal neurodegeneration. The A3R selective agonist (2-Cl-IB-MECA, 1 µM) prevented apoptosis (TUNEL(+)-cells) induced by kainate and cyclothiazide (KA + CTZ) in retinal neural cultures (86.5 ± 7.4 and 37.2 ± 6.1 TUNEL(+)-cells/field, in KA + CTZ and KA + CTZ + 2-Cl-IB-MECA, respectively). In retinal organotypic cultures, 2-Cl-IB-MECA attenuated NMDA-induced cell death, assessed by TUNEL (17.3 ± 2.3 and 8.3 ± 1.2 TUNEL(+)-cells/mm(2) in NMDA and NMDA+2-Cl-IB-MECA, respectively) and PI incorporation (ratio DIV4/DIV2 3.3 ± 0.3 and 1.3 ± 0.1 in NMDA and NMDA+2-Cl-IB-MECA, respectively) assays. Intravitreal 2-Cl-IB-MECA administration afforded protection against I-R injury decreasing the number of TUNEL(+) cells by 72%, and increased RGC survival by 57%. Also, intravitreal administration of 2-Cl-IB-MECA inhibited apoptosis (from 449.4 ± 37.8 to 207.6 ± 48.9 annexin-V(+)-cells) and RGC loss (from 1.2 ± 0.6 to 8.1 ± 1.7 cells/mm) induced by pONT. This study demonstrates that 2-Cl-IB-MECA is neuroprotective to the retina, both in vitro and in vivo. Activation of A3R may have great potential in the management of retinal neurodegenerative diseases characterized by RGC death, as glaucoma and diabetic retinopathy, and ischemic diseases.


Subject(s)
Neuroprotection/physiology , Receptor, Adenosine A3/metabolism , Retinal Degeneration/prevention & control , Retinal Neurons/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Fluorescent Antibody Technique, Indirect , In Situ Nick-End Labeling , Intravitreal Injections , Male , N-Methylaspartate/toxicity , Optic Nerve Injuries/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Retina/drug effects , Retina/pathology , Retinal Degeneration/metabolism
17.
Ophthalmic Res ; 54(4): 204-11, 2015.
Article in English | MEDLINE | ID: mdl-26517861

ABSTRACT

Alzheimer's disease (AD) is the most common type of dementia worldwide; it is characterized by a progressive decline in cognitive functions and memory, resulting from synaptic and cell loss, and accompanied by a strong neuroinflammatory response. Besides the vast progress in the understanding of the pathophysiology of AD in the past decades, there is still no effective treatment. Moreover, the diagnosis occurs usually at an advanced stage of the disease, where the neurological damage has already occurred. The identification of biomarkers that would allow an early diagnosis of this disease is a major goal that would also help managing AD progression. Due to its cellular and physiological resemblances with the brain, the retina has long been regarded as a window to the brain. Several brain manifestations have been associated with retinal alterations. In AD patients, some structural and functional alterations in the retina can be associated with disease onset. However, only a few studies have focused on the alterations in retinal glial cells associated with AD. This review aims at giving an overview of the AD-associated retinal alterations, particularly in glial cells. The documented alterations in retinal glia will be discussed concerning their potential to predict the brain alterations occurring in AD.


Subject(s)
Alzheimer Disease/diagnosis , Neuroglia/pathology , Retinal Neurons/pathology , Retinitis/diagnosis , Biomarkers , Early Diagnosis , Humans
19.
Cell Biosci ; 14(1): 5, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38183095

ABSTRACT

BACKGROUND: Glaucoma, a progressive neurodegenerative disease, is a leading cause of irreversible vision loss worldwide. This study aims to elucidate the critical role of Müller glia (MG) in the context of retinal ganglion cell (RGC) death, particularly focusing on the influence of peripheral MG sensitivity to high pressure (HP). METHODS: Co-cultures of porcine RGCs with MG were isolated from both the central and peripheral regions of pig retinas and subjected to both normal and HP conditions. Mass spectrometry analysis of the MG-conditioned medium was conducted to identify the proteins released by MG under all conditions. RESULTS: Peripheral MG were found to secrete a higher quantity of neuroprotective factors, effectively promoting RGC survival under normal physiological conditions. However, under HP conditions, co-cultures with peripheral MG exhibited impaired RGC survival. Moreover, under HP conditions, peripheral MG significantly upregulated the secretion of proteins associated with apoptosis, oxidative stress, and inflammation. CONCLUSIONS: This study provides robust evidence suggesting the involvement of MG in RGC death in glaucoma, thus paving the way for future therapeutic investigations.

20.
J Biol Chem ; 287(6): 3860-72, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22041898

ABSTRACT

Ascorbate is an important antioxidant, which also displays important functions in neuronal tissues, including the retina. The retina is responsible for the initial steps of visual processing, which is further refined in cerebral high-order centers. The retina is also a prototypical model for studying physiologic aspects of cells that comprise the nervous system. Of major importance also is the cellular messenger nitric oxide (NO). Previous studies have demonstrated the significance of NO for both survival and proliferation of cultured embryonic retinal cells. Cultured retinal cells express a high-affinity ascorbate transporter, and the release of ascorbate is delicately regulated by ionotropic glutamate receptors. Therefore, we proposed whether there is interplay between the ascorbate transport system and NO signaling pathway in retinal cells. Here we show compelling evidence that ascorbate uptake is tightly controlled by NO and its downstream signaling pathway in culture. NO also modulates the expression of SVCT-2, an effect mediated by cGMP and PKG. Kinetic studies suggest that NO increases the transport capacity for ascorbate, but not the affinity of SVCT-2 for its substrate. Interestingly, NO utilizes the NF-κB pathway, in a PKG-dependent manner, to modulate both SVCT-2 expression and ascorbate uptake. These results demonstrate that NO exerts a fine-tuned control of the availability of ascorbate to cultured retinal cells and strongly reinforces ascorbate as an important bioactive molecule in neuronal tissues.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/physiology , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Retina/metabolism , Sodium-Coupled Vitamin C Transporters/metabolism , Animals , Ascorbic Acid/genetics , Ascorbic Acid/metabolism , Biological Transport, Active/physiology , Cell Proliferation , Chick Embryo , Chickens , Cyclic GMP-Dependent Protein Kinases/genetics , NF-kappa B/genetics , Nerve Tissue Proteins/genetics , Nitric Oxide/genetics , Retina/cytology , Retina/embryology , Signal Transduction/physiology , Sodium-Coupled Vitamin C Transporters/genetics
SELECTION OF CITATIONS
SEARCH DETAIL