Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Exp Eye Res ; 239: 109758, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38123011

ABSTRACT

Recombinant adeno-associated viral vectors (rAAV) are the safest and most effective gene delivery platform to drive the treatment of many inherited eye disorders in well-characterized animal models. The use in rAAV of ubiquitous promoters derived from viral sequences such as CMV/CBA (chicken ß-actin promoter with cytomegalovirus enhancer) can lead to unwanted side effects such as pro-inflammatory immune responses and retinal cytotoxicity, thus reducing therapy efficacy. Thus, an advance in gene therapy is the availability of small promoters, that potentiate and direct gene expression to the cell type of interest, with higher safety and efficacy. In this study, we used six human mini-promoters packaged in rAAV2 quadruple mutant (Y-F) to test for transduction of the rat retina after intravitreal injection. After four weeks, immunohistochemical analysis detected GFP-labeled cells in the ganglion cell layer (GCL) for all constructs tested. Among them, Ple25sh1, Ple25sh2 and Ple53 promoted a widespread reporter-transgene expression in the GCL, with an increased number of GFP-expressing retinal ganglion cells when compared with the CMV/CBA vector. Moreover, Ple53 provided the strongest levels of GFP fluorescence in both cell soma and axons of retinal ganglion cells (RGCs) without any detectable adverse effects in retina function. Remarkably, a nearly 50-fold reduction in the number of intravitreally injected vector particles containing Ple53 promoter, still attained levels of transgene expression similar to CMV/CBA. Thus, the tested MiniPs show great potential for protocols of retinal gene therapy in therapeutic applications for retinal degenerations, especially those involving RGC-related disorders such as glaucoma.


Subject(s)
Cytomegalovirus Infections , Retinal Ganglion Cells , Rats , Humans , Animals , Retinal Ganglion Cells/metabolism , Genetic Vectors , Retina/metabolism , Transgenes , Intravitreal Injections , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Dependovirus/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Transduction, Genetic
2.
ASN Neuro ; 14: 17590914221076698, 2022.
Article in English | MEDLINE | ID: mdl-35103506

ABSTRACT

The retina of vertebrates is responsible for capturing light through visual (cones and rods) and non-visual photoreceptors (intrinsically photosensitive retinal ganglion cells and horizontal cells) triggering a number of essential activities associated to image- and non-image forming functions (photic entrainment of daily rhythms, pupillary light reflexes, pineal melatonin inhibition, among others). Although the retina contains diverse types of neuronal based-photoreceptors cells, originally classified as ciliary- or rhabdomeric-like types, in recent years, it has been shown that the major glial cell type of the retina, the Müller glial cells (MC), express blue photopigments as Opn3 (encephalopsin) and Opn5 (neuropsin) and display light responses associated to intracellular Ca2 + mobilization. These findings strongly propose MC as novel retinal photodetectors (Rios et al., 2019). Herein, we further investigated the intrinsic light responses of primary cultures of MC from embryonic chicken retinas specially focused on Ca2 + mobilization by fluorescence imaging and the identity of the internal Ca2 + stores responsible for blue light responses. Results clearly demonstrated that light responses were specific to blue light of long time exposure, and that the main Ca2 + reservoir to trigger downstream responses came from intracellular stores localized in the endoplasmic reticulum These observations bring more complexity to the intrinsic photosensitivity of retinal cells, particularly with regard to the detection of light in the blue range of visible spectra, and add novel functions to glial cells cooperating with other photoreceptors to detect and integrate ambient light in the retinal circuit and participate in cell to cell communication.Summary statement:Non-neuronal cells in the vertebrate retina, Muller glial cells, express non-canonical photopigments and sense blue light causing calcium release from intracellular stores strongly suggesting a novel intrinsic photosensitivity and new regulatory events mediating light-driven processes with yet unknown physiological implications.


Subject(s)
Calcium , Ependymoglial Cells , Animals , Calcium/metabolism , Chick Embryo , Ependymoglial Cells/metabolism , Neuroglia/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism
3.
Invest Ophthalmol Vis Sci ; 63(2): 5, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35103748

ABSTRACT

Purpose: Based on our preview evidence that reduced nuclear content of the transcription factor Myc-associated protein X (MAX) is an early event associated with degeneration of retinal ganglion cells (RGCs), in the present study, our purpose was to test whether the overexpression of human MAX had a neuroprotective effect against RGC injury. Methods: Overexpression of either MAX or green fluorescent protein (GFP) in the retina was achieved by intravitreal injections of recombinant adenovirus-associated viruses (rAAVs). Lister Hooded rats were used in three models of RGC degeneration: (1) cultures of retinal explants for 30 hours ex vivo from the eyes of 14-day-old rats that had received intravitreal injections of rAAV2-MAX or the control vector rAAV2-GFP at birth; (2) an optic nerve crush model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were operated on; and (3) an ocular hypertension (OHT) glaucoma model, in which 1-month-old rats received intravitreal injection of either rAAV2-MAX or rAAV2-GFP and, 4 weeks later, were subject to cauterization of the limbal plexus. Cell death was estimated by detection of pyknotic nuclei and TUNEL technique and correlated with MAX immunocontent in an ex vivo model of retinal explants. MAX expression was detected by quantitative RT-PCR. In the OHT model, survival of RGCs was quantified by retrograde labeling with DiI or immunostaining for BRN3a at 14 days after in vivo injury. Functional integrity of RGCs was analyzed through pattern electroretinography, and damage to the optic nerve was examined in semithin sections. Results: In all three models of RGC insult, gene therapy by overexpression of MAX prevented RGC death. Also, ON degeneration and electrophysiologic deficits were prevented in the OHT model. Conclusions: Our experiments offer proof of concept for a novel neuroprotective gene therapy for glaucomatous neurodegeneration based on overexpression of MAX.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation , Genetic Therapy/methods , Glaucoma/complications , Nerve Regeneration/genetics , Neurodegenerative Diseases/therapy , Neuroprotection/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Cell Death , Disease Models, Animal , Female , Glaucoma/genetics , Glaucoma/pathology , Male , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
4.
Neurosci Lett ; 763: 136197, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34437989

ABSTRACT

An insult can trigger a protective response or even cell death depending on different factors that include the duration and magnitude of the event and the ability of the cell to activate protective intracellular signals, including inflammatory cytokines. Our previous work showed that the treatment of Lister Hooded rat retinal cell cultures with 50 ng/mL phorbol 12-myristate 13-acetate (PMA), a protein kinase C activator, increases the survival of retinal ganglion cells (RGCs) kept in culture for 48 h after axotomy. Here we aim to analyze how PMA modulates the levels of TNF-α and IL-1ß (both key inflammatory mediators) and the impact of this modulation on RGCs survival. We hypothesize that the increase in RGCs survival mediated by PMA treatment depends upon modulation of the levels of IL-1ß and TNF-α. The effect of PMA treatment was assayed on cell viability, caspase 3 activation, TNF-α and IL-1ß release and TNF receptor type I (TNFRI) and TNF receptor type II (TNFRII) levels. PMA treatment increases IL-1ß and TNF-α levels in 15 min in culture and increases the release of both cytokines after 30 min and 24 h, respectively. Both IL-1ß and TNF-α levels decrease after 48 h of PMA treatment. PMA treatment also induces an increase in TNFRII levels while decreasing TNFRI after 24 h. PMA also inhibited caspase-3 activation, and decreased ROS production and EthD-1/calcein ratio in retinal cell cultures leading to an increase in cell viability. The neutralization of IL-1ß (anti-IL1ß 0,1ng/mL), the neutralization of TNF-α (anti-TNF-α 0,1ng/mL) and the TNF-α inhibition using a recombinant soluble TNFRII abolished PMA effect on RGCs survival. These data suggest that PMA treatment induces IL1ß and TNF-α release and modulation of TNFRI/TNFRII expression promoting RGCs survival after axotomy.


Subject(s)
Protein Kinase C/metabolism , Retinal Ganglion Cells/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn , Axotomy/adverse effects , Cell Survival/drug effects , Cells, Cultured , Female , Interleukin-1beta/metabolism , Male , Primary Cell Culture , Rats , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Retinal Ganglion Cells/metabolism , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
5.
Cells Dev ; 166: 203677, 2021 06.
Article in English | MEDLINE | ID: mdl-33994352

ABSTRACT

Slit-Robo signaling has been implicated in regulating several steps of retinal ganglion cell axon guidance, with a central role assigned to Slit2. We report here the phenotypical characterization of a CRISPR-Cas9-generated zebrafish null mutant for this gene, along with a detailed analysis of its expression pattern by WM-FISH. All evident defects in the optic axons in slit2-/- mutants were detected outside the retina, coincident with the major sites of expression at the ventral forebrain, around the developing optic nerve and anterior to the optic chiasm/proximal tract. Anterograde axon tracing experiments in zygotic and maternal-zygotic mutants, as well as morphants, showed the occurrence of axon sorting defects, which appeared mild at the optic nerve level, but more severe in the optic chiasm and the proximal tract. A remarkable sorting defect was the usual splitting of one of the optic nerves in two branches that surrounded the contralateral nerve at the chiasm. Although all axons eventually crossed the midline, the retinotopic order appeared lost at the proximal optic tract, to eventually correct distally. Time-lapse analysis demonstrated the sporadic occurrence of axon misrouting at the chiasm level, which could be responsible for the sorting errors. Our results support previous evidence of a channeling role for Slit molecules in retinal ganglion cell axons at the optic nerve, in addition to a function in the segregation of axons coming from each nerve and from different retinal regions at the medio-ventral area of the forebrain.


Subject(s)
Axons/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Optic Chiasm/metabolism , Optic Nerve/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Models, Biological , Mutation/genetics , Retinal Ganglion Cells/metabolism , Visual Pathways , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
6.
Genes (Basel) ; 12(3)2021 03 11.
Article in English | MEDLINE | ID: mdl-33799827

ABSTRACT

Glaucoma is a multifactorial neurodegenerative disease, characterized by degeneration of the retinal ganglion cells (RGCs). There has been little progress in developing efficient strategies for neuroprotection in glaucoma. We profiled the retina transcriptome of Lister Hooded rats at 2 weeks after optic nerve crush (ONC) and analyzed the data from the genomic fabric paradigm (GFP) to bring additional insights into the molecular mechanisms of the retinal remodeling after induction of RGC degeneration. GFP considers three independent characteristics for the expression of each gene: level, variability, and correlation with each other gene. Thus, the 17,657 quantified genes in our study generated a total of 155,911,310 values to analyze. This represents 8830x more data per condition than a traditional transcriptomic analysis. ONC led to a 57% reduction in RGC numbers as detected by retrograde labeling with 1,1'-dioctadecyl-3,3,3,3'-tetramethylindocarbocyanine perchlorate (DiI). We observed a higher relative expression variability after ONC. Gene expression stability was used as a measure of transcription control and disclosed a robust reduction in the number of very stably expressed genes. Predicted protein-protein interaction (PPI) analysis with STRING revealed axon and neuron projection as mostly decreased processes, consistent with RGC degeneration. Conversely, immune response PPIs were found among upregulated genes. Enrichment analysis showed that complement cascade and Notch signaling pathway, as well as oxidative stress and kit receptor pathway were affected after ONC. To expand our studies of altered molecular pathways, we examined the pairwise coordination of gene expressions within each pathway and within the entire transcriptome using Pearson correlations. ONC increased the number of synergistically coordinated pairs of genes and the number of similar profiles mainly in complement cascade and Notch signaling pathway. This deep bioinformatic study provided novel insights beyond the regulation of individual gene expression and disclosed changes in the control of expression of complement cascade and Notch signaling functional pathways that may be relevant for both RGC degeneration and remodeling of the retinal tissue after ONC.


Subject(s)
Glaucoma , Optic Nerve Injuries , Optic Nerve , Retinal Ganglion Cells , Transcriptome , Animals , Female , Glaucoma/genetics , Glaucoma/metabolism , Glaucoma/pathology , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
7.
Neurochem Res ; 46(7): 1701-1716, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33792824

ABSTRACT

Interleukin-2 (IL-2) is a classical pro-inflammatory cytokine known to display neuroprotective roles in the central nervous system including the retina. In the present study, we investigate the molecular targets involved in the neurotrophic effect of IL-2 on retinal ganglion cells (RGC) after optic nerve axotomy. Analysis of retrograde labeling of RGC showed that common cell survival mediators, as Trk receptors, Src, PI3K, PKC, and intracellular calcium do not mediate the neurotrophic effect of IL-2 on RGC. No involvement of MAPK p38 was also observed. However, other MAPKs as MEK and JNK appear to be mediating this IL-2 effect. Our data also indicate that JAK2/3 are important intracellular proteins for the IL-2 effect. Interestingly, we demonstrate that the IL-2 effect depends on dopamine D1 receptors (D1R), the cAMP/PKA pathway, interleukin-10 (IL-10), and NF-κB, suggesting that RGC survival induced by IL-2 encompasses a molecular network of major complexity. In addition, treatment of retinal cells with recombinant IL-10 or 6-Cl-pb (D1R full agonist) was able to increase RGC survival similar to IL-2. Taken together, our results suggest that after optic nerve axotomy, the increase in RGC survival triggered by IL-2 is mediated by IL-10 and D1R along with the intracellular pathways of MAPKs, JAK/STAT, and cAMP/PKA.


Subject(s)
Cell Survival/drug effects , Interleukin-10/metabolism , Interleukin-2/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Dopamine D1/metabolism , Retinal Ganglion Cells/drug effects , Animals , Animals, Newborn , Axotomy , Cells, Cultured , Female , Male , NF-kappa B/metabolism , Nerve Growth Factors/pharmacology , Optic Nerve/surgery , Rats , Retinal Ganglion Cells/metabolism
8.
J Alzheimers Dis ; 82(s1): S5-S18, 2021.
Article in English | MEDLINE | ID: mdl-33749647

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. This neurodegenerative syndrome affects cognition, memory, behavior, and the visual system, particularly the retina. OBJECTIVE: This work aims to determine whether the 5xFAD mouse, a transgenic model of AD, displays changes in the function of retinal ganglion cells (RGCs) and if those alterations are correlated with changes in the expression of glutamate and gamma-aminobutyric acid (GABA) neurotransmitters. METHODS: In young (2-3-month-old) and adult (6-7-month-old) 5xFAD and WT mice, we have studied the physiological response, firing rate, and burst of RGCs to various types of visual stimuli using a multielectrode array system. RESULTS: The firing rate and burst response in 5xFAD RGCs showed hyperactivity at the early stage of AD in young mice, whereas hypoactivity was seen at the later stage of AD in adults. The physiological alterations observed in 5xFAD correlate well with an increase in the expression of glutamate in the ganglion cell layer in young and adults. GABA staining increased in the inner nuclear and plexiform layer, which was more pronounced in the adult than the young 5xFAD retina, altering the excitation/inhibition balance, which could explain the observed early hyperactivity and later hypoactivity in RGC physiology. CONCLUSION: These findings indicate functional changes may be caused by neurochemical alterations of the retina starting at an early stage of the AD disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Disease Models, Animal , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Retinal Ganglion Cells/metabolism , Age Factors , Alzheimer Disease/physiopathology , Animals , Female , Glutamic Acid/metabolism , Male , Mice , Mice, Transgenic , Photic Stimulation/methods , gamma-Aminobutyric Acid/metabolism
9.
Mol Neurobiol ; 58(8): 3653-3664, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33786741

ABSTRACT

Glaucoma is a blindness-causing disease that involves selective damage to retinal ganglion cells (RGCs) and their axons. A subset of RGCs expressing the photopigment melanopsin regulates non-image-forming visual system functions, such as pupillary light reflex and circadian rhythms. We analyzed the effect of melatonin on the non-image-forming visual system alterations induced by experimental glaucoma. For this purpose, male Wistar rats were weekly injected with vehicle or chondroitin sulfate into the eye anterior chamber. The non-image-forming visual system was analyzed in terms of (1) melanopsin-expressing RGC number, (2) anterograde transport from the retina to the olivary pretectal nucleus and the suprachiasmatic nuclei, (3) blue- and white light-induced pupillary light reflex, (4) light-induced c-Fos expression in the suprachiasmatic nuclei, (5) daily rhythm of locomotor activity, and (6) mitochondria in melanopsin-expressing RGC cells. Melatonin prevented the effect of experimental glaucoma on melanopsin-expressing RGC number, blue- and white light-induced pupil constriction, retina-olivary pretectal nucleus, and retina- suprachiasmatic nuclei communication, light-induced c-Fos expression in the suprachiasmatic nuclei, and alterations in the locomotor activity daily rhythm. In addition, melatonin prevented the effect of glaucoma on melanopsin-expressing RGC mitochondrial alterations. These results support that melatonin protected the non-image-forming visual system against glaucoma, probably through a mitochondrial protective mechanism.


Subject(s)
Antioxidants/administration & dosage , Glaucoma/prevention & control , Melatonin/administration & dosage , Retinal Ganglion Cells/drug effects , Vision, Ocular/drug effects , Animals , Glaucoma/chemically induced , Glaucoma/metabolism , Light/adverse effects , Male , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism , Vision, Ocular/physiology
10.
Int J Mol Sci ; 21(7)2020 Mar 28.
Article in English | MEDLINE | ID: mdl-32231131

ABSTRACT

Diabetes produces several changes in the body triggered by high glycemia. Some of these changes include altered metabolism, structural changes in blood vessels and chronic inflammation. The eye and particularly the retinal ganglion cells (RGCs) are not spared, and the changes eventually lead to cell loss and visual function impairment. Understanding the mechanisms resulting in RGC damage and loss from diabetic retinopathy is essential to find an effective treatment. This review focuses mainly on the signaling pathways and molecules involved in RGC loss and the potential therapeutic approaches for the prevention of this cell death. Throughout the manuscript it became evident that multiple factors of different kind are responsible for RGC damage. This shows that new therapeutic agents targeting several factors at the same time are needed. Alpha-1 antitrypsin as an anti-inflammatory agent may become a suitable option for the treatment of RGC loss because of its beneficial interaction with several signaling pathways involved in RGC injury and inflammation. In conclusion, alpha-1 antitrypsin may become a potential therapeutic agent for the treatment of RGC loss and processes behind diabetic retinopathy.


Subject(s)
Diabetes Mellitus/pathology , Diabetic Retinopathy/pathology , Retinal Ganglion Cells/pathology , Animals , Cell Death , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Molecular Targeted Therapy , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects
11.
J Neurochem ; 153(6): 671-673, 2020 06.
Article in English | MEDLINE | ID: mdl-32162696

ABSTRACT

The aim of the present report was to analyze the involvement of glutamate neurotoxicity in retinal ganglion cell loss and optic nerve damage induced by experimental optic neuritis. For this purpose, the authors used an optic neuritis model induced by immunisation with myelin oligodendrocyte glycoprotein (AON). The authors describe a correlation in the timing of retinal ganglion cell (RGC) loss with alterations in the optic nerve actin cytoskeleton dynamic, and visual dysfunction. In addition, they show that an intravitreal injection of glutamate mimics, and an NMDA receptor antagonist avoids the effect of pre-clinical AON on visual functions and RGC number, as well as on optic nerve actin cytoskeleton. Taken together, their results support that avoiding glutamate neurotoxicity could become a new therapeutic approach for optic neuritis treatment.


Subject(s)
Optic Neuritis/immunology , Optic Neuritis/metabolism , Receptors, N-Methyl-D-Aspartate/immunology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Humans , Myelin-Oligodendrocyte Glycoprotein/toxicity , Optic Neuritis/chemically induced , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism
12.
Gene Ther ; 27(1-2): 27-39, 2020 02.
Article in English | MEDLINE | ID: mdl-31243393

ABSTRACT

After an injury, axons in the central nervous system do not regenerate over large distances and permanently lose their connections to the brain. Two promising approaches to correct this condition are cell and gene therapies. In the present work, we evaluated the neuroprotective and neuroregenerative potential of pigment epithelium-derived factor (PEDF) gene therapy alone and combined with human mesenchymal stem cell (hMSC) therapy after optic nerve injury by analysis of retinal ganglion cell survival and axonal outgrowth. Overexpression of PEDF by intravitreal delivery of AAV2 vector significantly increased Tuj1-positive cells survival and modulated FGF-2, IL-1ß, Iba-1, and GFAP immunostaining in the ganglion cell layer (GCL) at 4 weeks after optic nerve crush, although it could not promote axonal outgrowth. The combination of AAV2.PEDF and hMSC therapy showed a higher number of Tuj1-positive cells and a pronounced axonal outgrowth than unimodal therapy after optic nerve crush. In summary, our results highlight a synergistic effect of combined gene and cell therapy relevant for future therapeutic interventions regarding optic nerve injury.


Subject(s)
Eye Proteins/pharmacology , Nerve Growth Factors/pharmacology , Optic Nerve Injuries/therapy , Retinal Ganglion Cells/drug effects , Serpins/pharmacology , Animals , Axons/physiology , Cell Line, Tumor , Cell Survival , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Eye Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Nerve Crush , Nerve Growth Factors/metabolism , Nerve Regeneration , Neuroprotection , Optic Nerve , Rats, Wistar , Retina , Retinal Ganglion Cells/metabolism , Serpins/metabolism
13.
Curr Eye Res ; 45(8): 955-964, 2020 08.
Article in English | MEDLINE | ID: mdl-31842625

ABSTRACT

PURPOSE: The current study was undertaken to investigate whether Brazilian green propolis (BGP) can increase the viability of retinal ganglion cells (RGCs) in ischemic mouse retina, and examined the possible mechanisms underlying this neuroprotection. MATERIALS AND METHODS: C57BL/6J mice were subjected to constant elevation of intraocular pressure for 60 min to establish retinal ischemia-reperfusion injury. Mice then received saline or BGP (200 mg/kg) intraperitoneally once daily until sacrifice. The expression of hypoxia-inducing factor (HIF)-1α and glial fibrillary acidic protein (GFAP) and the level of histone acetylation were assessed at 1, 3, and 7 days after injury. The expression of Bax, Bcl-2, p53, NF-κB, Nrf2, and HO-1 were also analyzed at 3 days after injury. The neuroprotective effect of BGP treatment on RGC survival was evaluated using Brn3a immunohistochemical staining. RESULTS: The expression of HIF-1α and GFAP was increased and the level of histone acetylation decreased in saline-treated ischemic retinas within 7 days. BGP treatment effectively attenuated the elevated expression of HIF-1α, GFAP, Bax, NF-κB and p53. The expression of Bcl-2, Nrf2, HO-1 and the level of histone acetylation increased by BGP treatment, resulting in a significant difference between BGP-treated and saline-treated retinas. Immunohistochemical staining for Brn3a also revealed that BGP treatment protected against RGC loss in ischemic retina. CONCLUSIONS: Our results suggest that BGP has a neuroprotective effect on RGCs through the upregulation of histone acetylation, downregulation of apoptotic stimuli, and suppression of NF-κB mediated inflammatory pathway in ischemic retina. These findings suggest that BGP is a potential neuroprotective agent against RGC loss under oxidative stress.


Subject(s)
Neuroprotective Agents/therapeutic use , Propolis/therapeutic use , Reperfusion Injury/drug therapy , Retinal Diseases/drug therapy , Retinal Ganglion Cells/drug effects , Acetylation , Animals , Brazil , Cell Survival , Glial Fibrillary Acidic Protein/metabolism , Histones/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Neuroprotective Agents/chemistry , Oxidative Stress , Propolis/chemistry , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Transcription Factor Brn-3A/metabolism , Up-Regulation
14.
Sci Rep ; 9(1): 16286, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31705136

ABSTRACT

Glaucoma is a neurodegenerative disorder characterized by the progressive functional impairment and degeneration of the retinal ganglion cells (RGCs) and their axons, and is the leading cause of irreversible blindness worldwide. Current management of glaucoma is based on reduction of high intraocular pressure (IOP), one of its most consistent risk factors, but the disease proceeds in almost half of the patients despite such treatments. Several experimental models of glaucoma have been developed in rodents, most of which present shortcomings such as high surgical invasiveness, slow learning curves, damage to the transparency of the optic media which prevents adequate functional assessment, and variable results. Here we describe a novel and simple method to induce ocular hypertension in pigmented rats, based on low-temperature cauterization of the whole circumference of the limbal vascular plexus, a major component of aqueous humor drainage and easily accessible for surgical procedures. This simple, low-cost and efficient method produced a reproducible subacute ocular hypertension with full clinical recovery, followed by a steady loss of retinal ganglion cells and optic axons, accompanied by functional changes detected both by electrophysiological and behavioral methods.


Subject(s)
Disease Models, Animal , Disease Susceptibility , Glaucoma/etiology , Glaucoma/metabolism , Animals , Biomarkers , Cell Death , Electroretinography , Fluorescent Antibody Technique , Glaucoma/diagnosis , Immunohistochemistry , Intraocular Pressure , Nerve Degeneration , Psychomotor Performance , Rats , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
15.
Invest Ophthalmol Vis Sci ; 60(8): 3002-3012, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31310657

ABSTRACT

Purpose: To investigate the impact of obstructive sleep apnea (OSA) on the contribution of inner and outer retinal photoreceptors to the pupillary light response (PLR). Methods: Ninety-three eyes from 27 patients with OSA and 25 healthy controls were tested. OSA severity was graded according to the apnea-hypopnea index. PLR was measured monocularly with an eye tracker in a Ganzfeld in response to 1-second blue (470 nm) and red (640 nm) flashes at -3, -2, -1, 0, 1, 2, and 2.4 log cd/m2. Peak pupil constriction amplitude, peak latency, and the postillumination pupil response were measured. The Cambridge Colour Test, standard automatic perimetry, spectral domain optical coherence tomography, polysomnography, and the Pittsburgh Sleep Quality Index were used. Results: OSA patients have a significantly decreased peak pupil constriction amplitude for blue stimuli at -3, -2, -1, 1 log cd/m2 and at all red flash luminances (P < 0.050), revealing reduction of outer retina contributions to PLR. OSA patients showed reduced peak latency for blue (-2, 0, 2, 2.4 log cd/m2) and red stimuli (-2, 0 log cd/m2; P < 0.040). No significant difference was found in the melanopsin-mediated PLR. Conclusions: This study is the first to evaluate the inner and outer retinal contributions to PLR in OSA patients. The results showed that the outer retinal photoreceptor contributions to PLR were affected in moderate and severe OSA patients. In contrast, the inner retina contributions to PLR are preserved.


Subject(s)
Pupil/physiology , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Sleep Apnea, Obstructive/physiopathology , Sleep/physiology , Adult , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Photic Stimulation , Polysomnography , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Ganglion Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Tomography, Optical Coherence
16.
Stem Cell Res Ther ; 10(1): 121, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30995945

ABSTRACT

BACKGROUND: Retina and/or optic nerve injury may cause irreversible blindness, due to degeneration of retinal ganglion cells. We and others have previously shown that the intravitreal injection of mesenchymal stem cells (MSCs) protects injured retinal ganglion cells and stimulates their regeneration after optic nerve injury, but the long-term effects of this therapy are still unknown. METHODS: We injected rat MSC (rMSC) intravitreally in adult (3-5 months) Lister Hooded rats of either sex after optic nerve crush. Retinal ganglion cell survival, axonal regeneration, and reconnection were analyzed 60 and 240 days after crush by immunohistochemistry for Tuj1, anterograde labeling with cholera-toxin B and by immunohistochemistry for nerve growth factor-induced gene A (NGFI-A, driven by light stimulation) in the superior colliculus after a cycle of light deprivation-stimulation. Visual behaviors (optokinetic reflex, looming response, and preference for dark) were analyzed 70 days after crush. RESULTS: rMSC treatment doubled the number of surviving retinal ganglion cells, preferentially of a larger subtype, and of axons regenerating up to 0.5 mm. Some axons regenerated to the lateral geniculate nucleus and superior colliculus. NGFI-A+ cells were doubled in rMSC-treated animals 60 days after crush, but equivalent to vehicle-injected animals 240 days after crush, suggesting that newly formed synapses degenerated. Animals did not recover visual behaviors. CONCLUSIONS: We conclude that rMSC-induced neuroprotection is sustained at longer time points. Although rMSCs promoted long-term neuroprotection and long-distance axon regeneration, the reconnection of retinal ganglion cells with their targets was transitory, indicating that they need additional stimuli to make stable reconnections.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Nerve Regeneration , Optic Nerve Injuries , Optic Nerve/physiology , Allografts , Animals , Early Growth Response Protein 1/metabolism , Female , Male , Mesenchymal Stem Cells/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Optic Nerve Injuries/therapy , Rats , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
17.
Life Sci ; 218: 233-240, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30605647

ABSTRACT

Oxidative stress triggers ocular neurodegenerative diseases, such as glaucoma or macular degeneration. The increase of reactive oxygen and nitrogen species in retinal ganglion cells (RGCs) causes damage to the structure and function of the axons that make up the optic nerve, leading to cell death arising from apoptosis, necrosis or autophagy in the RCGs. The use of antioxidants to prevent visual neurodegenerative pathologies is a novel and possibly valuable therapeutic strategy. To investigate in vitro and in vivo neuroprotective efficacy of melatonin (MEL) in RGCs, we used a model of oxidative glutamate (GLUT) toxicity in combination with l-butionin-S, R-sulfoximine (BSO), which induces cell death by apoptosis through cytotoxicity and oxidative stress mechanisms. Histological sectioning and immunohistochemical assays using the TUNEL technique were performed to determine the damage generated in affected cells and to observe the death process of RGCs. Whit BSO-GLUT the results revealed a progressive RGCs death without any significant evidence of a decreased retinal function after 9 days of treatment. In this way, we were able to develop a retinal degeneration model in vivo to carry out treatment with MEL and observed an increase in the survival percentage of RGCs, showing that BSO-GLUT could not exert an oxidant effect on cells to counteract the effect of MEL. These findings reveal that MEL has a neuroprotective and antiapoptotic effect as evidenced by the reduction of oxidative stress damage. MEL demonstrated in this model makes it a promising neuroprotective agent for the treatment of ocular neurodegenerative diseases when administered locally.


Subject(s)
Melatonin/pharmacology , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Retinal Degeneration/drug therapy , Retinal Ganglion Cells/drug effects , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Chick Embryo , Glutamic Acid/pharmacology , In Vitro Techniques , Oxidative Stress/drug effects , Rabbits , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
18.
Neuropharmacology ; 145(Pt A): 87-98, 2019 02.
Article in English | MEDLINE | ID: mdl-29233635

ABSTRACT

Optic neuritis (ON) is an inflammatory, demyelinating, neurodegenerative, and presently untreatable condition of the optic nerve which might induce blindness. We analyzed the effect of environmental enrichment (EE) on visual pathway damage provoked by experimental ON induced by a microinjection of bacterial lipopolysaccharide (LPS) into the optic nerve. For this purpose, LPS was microinjected into the optic nerve from male Wistar rats. After injection, one group of animals was submitted to EE, and another group remained in standard environment (SE) for 21 days. EE prevented the decrease in pupil light reflex (PLR), visual evoked potentials, retinal anterograde transport, phosphorylated neurofilament immunoreactivity, myelination (luxol fast blue staining), and axon (toluidine blue staining) and retinal ganglion cell (Brn3a-immunoreactivity) number. EE also prevented microglial/macrophage reactivity (Iba-1- and ED1-immunoreactivity), and astrocytosis (glial fibrillary acidic protein-immunostaining) induced by experimental ON. LPS-injected optic nerves displayed oxidative damage and increased inducible nitric oxide synthase, cyclooxygenase-2, and interleukin-1ß and TNFα mRNA levels which were prevented by EE. EE increased optic nerve brain-derived neurotrophic factor levels. When EE started at 4 (but not 7) days post-injection of LPS, a preservation of the PLR was observed at 21 days post-LPS, which was blocked by the daily administration of ANA-12 from day 4 to day 7 post-LPS. Moreover, EE from day 4 to day 7 post-LPS significantly preserved the PLR at 21 days post-injection. Taken together, our data suggest that EE preserved visual functions and reduced neuroinflammation of the optic nerve. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".


Subject(s)
Environment , Optic Neuritis/therapy , Animals , Axons/metabolism , Axons/pathology , Disease Models, Animal , Evoked Potentials, Visual , Housing, Animal , Male , Neuroglia/metabolism , Neuroglia/pathology , Optic Nerve/pathology , Optic Nerve/physiopathology , Optic Neuritis/pathology , Optic Neuritis/physiopathology , Random Allocation , Rats, Wistar , Reflex, Pupillary , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Visual Pathways/pathology , Visual Pathways/physiopathology
19.
Biochem Biophys Res Commun ; 500(4): 917-923, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29705702

ABSTRACT

Protein kinase C (PKC) is a family of serine/threonine kinases related to several phenomena as cell proliferation, differentiation and survival. Our previous data demonstrated that treatment of axotomized neonatal rat retinal cell cultures for 48 h with phorbol 12-myristate 13-acetate (PMA), a PKC activator, increases retinal ganglion cells (RGCs) survival. Moreover, this treatment decreases M1 receptors (M1R) and modulates BDNF levels. The aim of this work was to assess the possible involvement of neurotrophins BDNF and NGF in the modulation of M1R levels induced by PKC activation, and its involvement on RGCs survival. Our results show that PMA (50 ng/mL) treatment, via PKC delta activation, modulates NGF, BDNF and M1R levels. BDNF and NGF mediate the decrease of M1R levels induced by PMA treatment. M1R activation is essential to PMA neuroprotective effect on RGCs as telenzepine (M1R selective antagonist) abolished it. Based on our results we suggest that PKC delta activation modulates neurotrophins levels by a signaling pathway that involves M1R activation and ultimately leading to an increase in RGCs survival in vitro.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Muscarinic Agonists/pharmacology , Nerve Growth Factor/genetics , Protein Kinase C-delta/genetics , Receptor, Muscarinic M1/genetics , Retinal Ganglion Cells/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival/drug effects , Gene Expression Regulation , Muscarinic Antagonists/pharmacology , Nerve Growth Factor/metabolism , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacology , Primary Cell Culture , Protein Kinase C-delta/metabolism , Rats , Receptor, Muscarinic M1/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Signal Transduction
20.
J Chem Neuroanat ; 90: 57-69, 2018 07.
Article in English | MEDLINE | ID: mdl-29277705

ABSTRACT

The rock cavy (Kerodon rupestris) is a crepuscular Hystricomorpha rodent that has been used in comparative analysis of retinal targets, but its retinal organization remains to be investigated. In order to better characterize its visual system, the present study analyzed neurochemical features related to the topographic organization of catecholaminergic cells and ganglion cells, as well the distribution of calcium-binding proteins in the outer and inner retina. Retinal sections and/or wholemounts were processed using tyrosine hydroxylase (TH), GABA, calbindin, parvalbumin and calretinin immunohistochemistry or Nissl staining. Two types of TH-immunoreactive (TH-IR) cells were found which differ in soma size, dendritic arborization, intensity of TH immunoreactivity and stratification pattern in the inner plexiform layer. The topographic distribution of all TH-IR cells defines a visual streak along the horizontal meridian in the superior retina. The ganglion cells are also distributed in a visual streak and the visual acuity estimated considering their peak density is 4.13 cycles/degree. A subset of TH-IR cells express GABA or calbindin. Calretinin is abundant in most of retinal layers and coexists with calbindin in horizontal cells. Parvalbumin is less abundant and expressed by presumed amacrine cells in the INL and some ganglion cells in the GCL. The topographic distribution of TH-IR cells and ganglion cells in the rock cavy retina indicate a suitable adaptation for using a broad extension of its inferior visual field in aspects that involve resolution, adjustment to ambient light intensity and movement detection without specialized eye movements.


Subject(s)
Calcium-Binding Proteins/metabolism , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Rodentia/anatomy & histology , Animals , Catecholamines/metabolism , Female , Male
SELECTION OF CITATIONS
SEARCH DETAIL