Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 318
Filtrar
1.
Bioengineered ; 12(1): 3947-3956, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34281454

RESUMO

Diabetic retinopathy (DR) represents the most typical complication of type 2 diabetes mellitus and one of the most primary oculopathy causing blindness. However, the mechanism of DR remains unknown. RIPK1/RIPK3, as homologous serine/threonine kinases, are key elements in mediating necroptosis and may have functions in DR development. To clarify the relationship between DR and RIPK1/RIPK3, this study established a model of apoptosis using high-glucose induced RGCs, which were treated with 7.5, 19.5, and 35 mM D-glucose for 12, 24, and 48 h, respectively. Subsequently, the expression of RIPK1/RIPK3 was determined and the protective effect of necrostatin-1 on RGCs injury induced by high glucose was explored. The results demonstrated that the expression of RIPK1 and RIPK3 in the cells was increased markedly following 12 h treatment with 19.5 mM D-glucose. Additionally, following an addition of 100 µM necrostatin-1 in 19.5 mM D-glucose medium for RGCs treatment 12 h, the protein expression of RIPK1 and RIPK3 was decreased markedly, and the number of Nissl bodies in cells was increased substantially. The findings of the present study indicated that high glucose could induce the expression of RIPK1/RIPK3, and necrostatin-1 could effectively protect RGCs from D-glucose-induced cell necrosis.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucose/toxicidade , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Células Ganglionares da Retina/enzimologia , Animais , Separação Celular , Imidazóis/farmacologia , Indóis/farmacologia , Camundongos Endogâmicos C57BL , Células Ganglionares da Retina/efeitos dos fármacos
2.
Invest Ophthalmol Vis Sci ; 62(6): 13, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33978676

RESUMO

Purpose: The goal of this study was to determine whether JNK2 played a causative role in endothelin-mediated loss of RGCs in mice. Methods: JNK2-/- and wild type (C57BL/6) mice were intravitreally injected in one eye with 1 nmole of ET-1, whereas the contralateral eye was injected with the vehicle. At two time points (two hours and 24 hours) after the intravitreal injections, mice were euthanized, and phosphorylated c-Jun was assessed in retinal sections. In a separate set of experiments, JNK2-/- and wild type mice were intravitreally injected with either 1 nmole of ET-1 or its vehicle and euthanized seven days after injection. Retinal flat mounts were stained with antibodies to the RGC marker, Brn3a, and surviving RGCs were quantified. Axonal degeneration was assessed in paraphenylenediamine stained optic nerve sections. Results: Intravitreal ET-1 administration produced a significant increase in immunostaining for phospho c-Jun in wild type mice, which was appreciably lower in the JNK2 -/- mice. A significant (P < 0.05) 26% loss of RGCs was found in wild type mice, seven days after injection with ET-1. JNK2-/- mice showed a significant protection from RGC loss following ET-1 administration, compared to wild type mice injected with ET-1. A significant decrease in axonal counts and an increase in the collapsed axons was found in ET-1 injected wild type mice eyes. Conclusions: JNK2 appears to play a major role in ET-1 mediated loss of RGCs in mice. Neuroprotective effects in JNK2-/- mice following ET-1 administration occur mainly in the soma and not in the axons of RGCs.


Assuntos
Endotelina-1/toxicidade , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Degeneração Retiniana/induzido quimicamente , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Axônios/patologia , Biomarcadores/metabolismo , Sobrevivência Celular , Feminino , Imuno-Histoquímica , Injeções Intravítreas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nervo Óptico/patologia , Fosforilação , Degeneração Retiniana/enzimologia , Células Ganglionares da Retina/enzimologia , Fator de Transcrição Brn-3A/metabolismo
3.
Int J Mol Sci ; 22(4)2021 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-33562231

RESUMO

The neurodegenerative disease amyotrophic lateral sclerosis (ALS) affects the spinal cord, brain stem, and cerebral cortex. In this pathology, both neurons and glial cells are affected. However, few studies have analyzed retinal microglia in ALS models. In this study, we quantified the signs of microglial activation and the number of retinal ganglion cells (RGCs) in an SOD1G93A transgenic mouse model at 120 days (advanced stage of the disease) in retinal whole-mounts. For SOD1G93A animals (compared to the wild-type), we found, in microglial cells, (i) a significant increase in the area occupied by each microglial cell in the total area of the retina; (ii) a significant increase in the arbor area in the outer plexiform layer (OPL) inferior sector; (iii) the presence of cells with retracted processes; (iv) areas of cell groupings in some sectors; (v) no significant increase in the number of microglial cells; (vi) the expression of IFN-γ and IL-1ß; and (vii) the non-expression of IL-10 and arginase-I. For the RGCs, we found a decrease in their number. In conclusion, in the SOD1G93A model (at 120 days), retinal microglial activation occurred, taking a pro-inflammatory phenotype M1, which affected the OPL and inner retinal layers and could be related to RGC loss.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Microglia/patologia , Mutação , Células Ganglionares da Retina/patologia , Superóxido Dismutase-1/fisiologia , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/etiologia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Microglia/enzimologia , Células Ganglionares da Retina/enzimologia
5.
Curr Eye Res ; 46(5): 719-730, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33026257

RESUMO

AIM/PURPOSE: Individually, hypoxia and protein kinase R (PKR) induce retinal ganglion cells (RGCs) damage by aggravating reactive oxygen species (ROS), oxidative stress, inflammation, and apoptosis. However, it is still not established in hypoxia mediates such damaging effect by modulating PKR. This study investigated the expression and activation of PKR in hypoxic RGCs and tested if suppression of PKR by C16 is protective. MATERIALS AND METHODS: Isolated RGCs were under normoxic or hypoxic conditions for 12 h. In some cases, hypoxic cells were pre-treated with C16, a PKR inhibitor, or n-acetyl cysteine (NAC) a glutathione (GSH) precursor for 1 h and then exposed to hypoxia for the next 12 h. RESULTS: Hypoxia increased cell death, lactate dehydrogenase (LDH) levels, and levels of single-stranded DNA (ssDNA). It also increased levels of ROS, the activity of the nuclear factor-kappa beta (NF-κB), JNK, and p38 MAPK, expression of Bax, p53, and cleaved caspase-3, levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and cytoplasmic levels of cytochrome-c. It concomitantly suppressed levels of GSH and Bcl-2. All these events were associated with increased phosphorylation (activation) of PKR and its target eukaryotic initiation factor 2 (eIF2). Pre-incubating the cells with NAC completely prevented all these effects in hypoxic cells. Similar protective effects without affecting levels of ROS and GSH levels were also seen in hypoxic cells pre-treated with C16. CONCLUSION: Hypoxia induces oxidative stress, inflammation, and apoptosis in the RGCs mainly by ROS induced activation of PKR, whereas scavenging ROS by NAC or suppressing PKR by C16 is a novel protective mechanism.


Assuntos
Apoptose , Hipóxia/metabolismo , Indóis/farmacologia , Inflamação/metabolismo , Estresse Oxidativo , Células Ganglionares da Retina/enzimologia , Tiazóis/farmacologia , eIF-2 Quinase/antagonistas & inibidores , Animais , Western Blotting , Sobrevivência Celular/fisiologia , Células Cultivadas , Citocinas/metabolismo , DNA de Cadeia Simples/metabolismo , Ensaio de Imunoadsorção Enzimática , L-Lactato Desidrogenase/metabolismo , MAP Quinase Quinase 4/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/fisiologia , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Células Ganglionares da Retina/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Diabetologia ; 64(3): 693-706, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33319325

RESUMO

AIMS/HYPOTHESIS: Diabetic retinopathy is characterised by retinal neurodegeneration and retinal vascular abnormalities, affecting one third of diabetic patients with disease duration of more than 10 years. Accumulated evidence suggests that serine racemase (SR) and D-serine are correlated with the pathogenesis of diabetic retinopathy and the deletion of the Srr gene reverses neurovascular pathologies in diabetic mice. Since D-serine content is balanced by SR synthesis and D-amino acid oxidase (DAAO) degradation, we examined the roles of DAAO in diabetic retinopathy and further explored relevant therapy. METHODS: Rats were used as a model of diabetes by i.p. injection of streptozotocin at the age of 2 months and blood glucose was monitored with a glucometer. Quantitative real-time PCR was used to examine Dao mRNA and western blotting to examine targeted proteins in the retinas. Bisulphite sequencing was used to examine the methylation of Dao mRNA promoter in the retinas. Intravitreal injection of DAAO-expressing adenovirus (AAV8-DAAO) was conducted one week before streptozotocin administration. Brain specific homeobox/POU domain protein 3a (Brn3a) immunofluorescence was conducted to indicate retinal ganglion cells at 3 months after virus injection. The permeability of the blood-retinal barrier was examined by Evans blue leakage from retinal capillaries. Periodic acid-Schiff staining and haematoxylin counterstaining were used to indicate retinal vasculature, which was further examined with double immunostaining at 7 months after virus injection. RESULTS: At the age of 12 months, DAAO mRNA and protein levels in retinas from diabetic animals were reduced to 66.2% and 70.4% of those from normal (control) animals, respectively. The Dao proximal promoter contained higher levels of methylation in diabetic than in normal retinas. Consistent with the observation, DNA methyltransferase 1 was increased in diabetic retinas. Injection of DAAO-expressing virus completely prevented the loss of retinal ganglion cells and the disruption of blood-retinal barrier in diabetic rats. Diabetic retinas contained retinal ganglion cells at a density of 54 ± 4/mm2, which was restored to 68 ± 9/mm2 by DAAO overexpression, similar to the levels in normal retinas. The ratio between the number of endothelial cells and pericytes in diabetic retinas was 6.06 ± 1.93/mm2, which was reduced to 3.42 ± 0.55/mm2 by DAAO overexpression; the number of acellular capillaries in diabetic retinas was 10 ± 5/mm2, which was restored to 6 ± 2/mm2 by DAAO overexpression, similar to the levels in normal retinas. Injection of the DAAO-expressing virus increased the expression of occludin and reduced gliosis, which were examined to probe the mechanism by which the disrupted blood-retinal barrier in diabetic rats was rescued and retinal neurodegeneration was prevented. CONCLUSIONS/INTERPRETATION: Altogether, overexpression of DAAO before the onset of diabetes protects against neurovascular abnormalities in retinas from diabetic rats, which suggests a novel strategy for preventing diabetic retinopathy. Graphical abstract.


Assuntos
Barreira Hematorretiniana/enzimologia , D-Aminoácido Oxidase/biossíntese , Retinopatia Diabética/prevenção & controle , Células Ganglionares da Retina/enzimologia , Animais , Barreira Hematorretiniana/patologia , Permeabilidade Capilar , D-Aminoácido Oxidase/genética , Metilação de DNA , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/enzimologia , Retinopatia Diabética/enzimologia , Retinopatia Diabética/etiologia , Retinopatia Diabética/patologia , Indução Enzimática , Masculino , Degeneração Neural , Regiões Promotoras Genéticas , Ratos Sprague-Dawley , Células Ganglionares da Retina/patologia , Fator de Transcrição Brn-3A/genética , Fator de Transcrição Brn-3A/metabolismo
7.
Invest Ophthalmol Vis Sci ; 61(13): 13, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33156340

RESUMO

Purpose: Activation of proteolytic enzymes, calpains and caspases, have been observed in many models of retinal disease. We previously demonstrated calpain activation in monkey retinal explants cultured under hypoxia. However, cellular responses are often species-specific. The purpose of the present study was to determine whether calpains or caspase-3 was involved in retinal ganglion cell (RGC) damage caused by hypoxia/reoxygenation in human retinal explants. The explant model was improved by use of an oxygen-controlled chamber. Methods: Human and monkey retinal explants were cultured under hypoxic conditions in an oxygen-controlled chamber and then reoxygenated. Calpain inhibitor SNJ-1945 was maintained throughout the culture period. Immunohistochemistry and immunoblotting were performed for calpains 1 and 2, calpastatin, α-spectrin, calpain-specific α-spectrin breakdown product at 150 kDa (SBDP150), caspase-3, and apoptosis-inducing factor (AIF). Propidium iodide (PI) staining measured membrane disruption, and TUNEL staining detected DNA fragmentation. Results: Activation of calpains in nerve fibers and increases of PI-positive RGCs were observed in retinal explants incubated for 16-hour hypoxia/8-hour reoxygenation. Except for autolysis of calpain 2, SNJ-1945 ameliorated these changes. In longer incubations under 24-hour hypoxia/16-hour reoxygenation, TUNEL-positive cells appeared, although activated caspase-3 and truncated AIF were not observed. DNA fragmentation was inhibited by SNJ-1945. Conclusions: An improved human retinal explant model showed that calpains, not caspase-3, were involved in cell damage induced by hypoxia/reoxygenation. This finding could be relevant for patient treatment with a calpain inhibitor if calpain activation is documented in human retinal ischemic diseases.


Assuntos
Calpaína/metabolismo , Caspase 3/metabolismo , Citosol/enzimologia , Hipóxia/enzimologia , Doenças Retinianas/enzimologia , Células Ganglionares da Retina/enzimologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Calpaína/antagonistas & inibidores , Carbamatos/farmacologia , Células Cultivadas , Criança , Fragmentação do DNA/efeitos dos fármacos , Ativação Enzimática , Humanos , Hipóxia/patologia , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Macaca mulatta , Pessoa de Meia-Idade , Doenças Retinianas/patologia , Células Ganglionares da Retina/patologia
8.
Exp Eye Res ; 190: 107892, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31811822

RESUMO

Neuronal excitotoxicity caused by over activation of N -Methyl-D-Aspartate (NMDA) receptors is an important risk factor for the retinal ganglion cells (RGCs) death in glaucoma. D-serine played a role as a key co-agonist for NMDA receptor activity and neurotoxicity. Our previous studies have demonstrated that increased D-serine and serine racemase (SR) expression in the retina of the chronic intraocular hypertension (COH) model were detected. D-amino acid oxidase (DAAO) treatment significantly increased RGCs survival in the glaucomatous eyes. However, the molecular mechanism remains unclear. In the present study, we investigated the extracellular signal-regulated protein kinase1/2 (ERK1/2) signaling pathway involved in DAAO neuroprotective effects on RGC survival and explore the effect of inhibited ERK1/2 phosphorylation on RGC survival and Müller cell activation in a COH rat model. We found that ERK1/2 phosphorylation and p38 kinase (p38) phosphorylation increased in the COH model, while c-Jun N-terminal kinase (JNK) phosphorylation didn't change. DAAO treatment induced ERK-1/2 MAP kinase phosphorylation and its upstream regulator, p-MEK increased in the COH model. The increased p-ERK was mainly located in retinal Müller cells. In contrast, p-JNK and p-p38 protein expression was not significantly different under these conditions. Quantitative analysis of RGC survival by fluorescent labeling and TdT-mediated dUTP nick-end labeling (TUNEL) assays confirmed that p-ERK1/2 inhibition by PD98059 attenuates DAAO-mediated reductions in RGC apoptosis. Additionally, p-ERK1/2 inhibition induced elevated glial fibrillary acidic protein (GFAP) expression in Müller cells in the COH model. Together, these results suggest that the ERK1/2 signaling pathway is involved in DAAO's neuroprotective effects on RGC survival.


Assuntos
D-Aminoácido Oxidase/farmacologia , Modelos Animais de Doenças , Glaucoma/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/farmacologia , Animais , Western Blotting , Células Ependimogliais/metabolismo , Flavonoides/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Glaucoma/enzimologia , Proteína Glial Fibrilar Ácida/metabolismo , Marcação In Situ das Extremidades Cortadas , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/enzimologia
9.
Invest Ophthalmol Vis Sci ; 60(14): 4619-4631, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31675426

RESUMO

Purpose: The purpose of this study was to extend the current understanding of endogenous lysine-specific demethylase 1 (LSD1) expression spatially and temporally in the retina. Toward that end, we determined the localization and levels of LSD1 and its substrates H3K4me1 and H3K4me2 (H3K4me1/2) within the murine eye. Methods: Immunofluorescent microscopy for LSD1, H3K4me1, and H3K4me2 was conducted on murine formalin-fixed paraffin-embedded eye sections across development in addition to Western immunoblotting to assess localization and protein levels. Results: Retinal LSD1 protein levels were highest at postnatal day 7 (P7), whereas its substrates H3K4me1 and H3K4me2 had equally high levels at P2 and P14. Concentrations of all three proteins gradually decreased over developmental time until reaching a basement level of ∼60% of maximum at P36. LSD1 and H3K4me1/2 were expressed uniformly in all retinal progenitor cells. By P36, there was variability in LSD1 expression in the ganglion cell layer, uniform expression in the inner nuclear layer, and dichotomous expression between photoreceptors in the outer nuclear layer. This contrasted with H3K4me1/2 expression, which remained uniform. Additionally, LSD1 was widely expressed in the lens, cornea, and retinal pigment epithelium. Conclusions: Consistent with its known role in neuronal differentiation, LSD1 is highly and uniformly expressed throughout all retinal progenitor cells. Variability in LSD1 expression, particularly in photoreceptors, may be indicative of their unique transcriptomes and epigenetic patterns of rods and cones. Murine rod nuclei exhibit LSD1 expression in a ring or shell, rather than throughout the nucleus, consistent with their unique inverted chromatin organization. LSD1 has substantial expression throughout adulthood, especially in cone nuclei. By providing insight into endogenous LSD1 expression, our current findings could directly inform future studies to determine the exact role of Lsd1 in the development and maintenance of specific structures and cell types within the eye.


Assuntos
Histona Desmetilases/metabolismo , Retina/enzimologia , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia de Fluorescência , Células Fotorreceptoras de Vertebrados/enzimologia , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/enzimologia , Células-Tronco/enzimologia
10.
Elife ; 82019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31621585

RESUMO

Elevated intraocular pressure (IOP) due to insufficient aqueous humor outflow through the trabecular meshwork and Schlemm's canal (SC) is the most important risk factor for glaucoma, a leading cause of blindness worldwide. We previously reported loss of function mutations in the receptor tyrosine kinase TEK or its ligand ANGPT1 cause primary congenital glaucoma in humans and mice due to failure of SC development. Here, we describe a novel approach to enhance canal formation in these animals by deleting a single allele of the gene encoding the phosphatase PTPRB during development. Compared to Tek haploinsufficient mice, which exhibit elevated IOP and loss of retinal ganglion cells, Tek+/-;Ptprb+/- mice have elevated TEK phosphorylation, which allows normal SC development and prevents ocular hypertension and RGC loss. These studies provide evidence that PTPRB is an important regulator of TEK signaling in the aqueous humor outflow pathway and identify a new therapeutic target for treatment of glaucoma.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Glaucoma/genética , Receptor TIE-2/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Células Ganglionares da Retina/enzimologia , Alelos , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Humor Aquoso/enzimologia , Contagem de Células , Modelos Animais de Doenças , Deleção de Genes , Glaucoma/enzimologia , Glaucoma/patologia , Heterozigoto , Humanos , Pressão Intraocular/fisiologia , Camundongos , Camundongos Knockout , Fosforilação , Receptor TIE-2/deficiência , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Células Ganglionares da Retina/patologia , Fatores de Risco , Transdução de Sinais , Malha Trabecular/enzimologia , Malha Trabecular/patologia
12.
Invest Ophthalmol Vis Sci ; 60(10): 3584-3594, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31425583

RESUMO

Purpose: Heterozygous mutations in the essential X-linked gene CASK associate with optic nerve hypoplasia (ONH) and other retinal disorders in girls. CASK+/- heterozygous knockout mice with mosaic CASK expression exhibit ONH with a loss of retinal ganglion cells (RGCs) but no changes in retinal morphology. It remains unclear if CASK deficiency selectively affects RGCs or also affects other retinal cells. Furthermore, it is not known if CASK expression in RGCs is critical for optic nerve (ON) development and maintenance. Methods: The visual behavior of CASK+/- mice was assessed and electroretinography (ERG) was performed. Using a mouse line with a floxed CASK gene that expresses approximately 40% CASK globally in all cells (hypomorph) under hemizygous and homozygous conditions, we investigated effects of CASK reduction on the retina and ON. CASK then was completely deleted from RGCs to examine its cell-autonomous role. Finally, for the first time to our knowledge, we describe a hemizygous CASK missense mutation in a boy with ONH. Results: CASK+/- heterozygous mutant mice display reduced visual contrast sensitivity, but ERG is indistinguishable from wildtype. CASK hypomorph mice exhibit ONH, but deletion of CASK from RGCs in this background does not exacerbate the condition. The boy with ONH harbors a missense mutation (p.Pro673Leu) that destabilizes CASK and weakens the crucial CASK-neurexin interaction. Conclusions: Our results demonstrate that mosaic or global reduction in CASK expression and/or function disproportionately affects RGCs. CASK expression in RGCs does not appear critical for cell survival, indicating a noncell autonomous role for CASK in the development of ON.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Guanilato Quinases/genética , Hipoplasia do Nervo Óptico/genética , Animais , Sobrevivência Celular , Pré-Escolar , Sensibilidades de Contraste/fisiologia , Eletrorretinografia , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Hipoplasia do Nervo Óptico/fisiopatologia , Retina/fisiopatologia , Células Ganglionares da Retina/enzimologia
13.
Mol Neurobiol ; 56(12): 8018-8034, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31161423

RESUMO

Excitotoxicity is known to modulate the nuclear accumulation, and thus activity state, of histone deacetylases (HDACs) in pyramidal neurons. In the retina, deregulation in activity and expression of different HDACs has been linked to pathological conditions such as retinitis pigmentosa, retinal ischemia, glaucoma, and acute optic nerve injury. Up to now, however, the effects of in vivo excitotoxicity on the different HDACs in retinal ganglion cells (RGCs) have not been thoroughly investigated. Here, we injected adult mice intravitreally with N-methyl-D-aspartate (NMDA) as a mean to trigger excitotoxicity-mediated RGC degeneration and we detected time-dependent loss of RGCs at 1 and 7 days after the insult. Further, we characterized the subcellular localization of HDACs belonging to class I (HDAC1, HDAC3), IIa (HDAC4, HDAC5, HDAC7, HDAC9), IIb (HDAC6, HDAC10), and IV (HDAC11) in RGCs. Our analyses revealed a differential pattern of HDACs nuclear distribution in RGCs following excitotoxicity. After 1 day, HDAC3, HDAC5, HDAC6, HDAC7, and HDAC11 showed altered subcellular localization in RGCs while 7 days after the excitotoxic insult, HDAC4 and HDAC9 were the only HDACs displaying changes in their subcellular distribution. Moreover, we found that in vivo selective inhibition of HDAC1/3 or HDAC4/5 via MS-275 (entinostat) or LMK-235, respectively, could prevent ongoing RGC degeneration. In conclusion, our results point towards a role of HDACs in RGC degeneration and identify HDAC1/3 and HDAC4/5 as potential therapeutic targets to treat degenerative retinal diseases.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Histona Desacetilases/metabolismo , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/enzimologia , Células Ganglionares da Retina/enzimologia , Animais , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Injeções Intravítreas/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/toxicidade , Degeneração Retiniana/tratamento farmacológico , Células Ganglionares da Retina/efeitos dos fármacos
14.
Invest Ophthalmol Vis Sci ; 59(11): 4453-4462, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30193318

RESUMO

Purpose: Ocular trauma is common in civilian and military populations. Among other injuries, closed globe blunt ocular trauma causes acute disruption of photoreceptor outer segments (commotio retinae) and retinal ganglion cell (RGC) death (traumatic optic neuropathy [TON]), both of which permanently impair vision. Caspase-2-dependent cell death is important and evidenced in models of RGC degeneration. We assessed the role of caspase-2 as a mediator of RGC and photoreceptor death in a rat blunt ocular trauma model. Methods: Bilateral ballistic closed globe blunt ocular trauma was induced in female Lister-hooded rats and caspase-2 cleavage and localization assessed by Western blotting and immunohistochemistry. Retinal caspase-2 was knocked down by intravitreal injection of caspase-2 small interfering RNA (siCASP2). In retinal sections, RGC survival was assessed by BRN3A-positive cell counts and photoreceptor survival by outer nuclear layer (ONL) thickness, respectively. Retinal function was assessed by electroretinography (ERG). Results: Raised levels of cleaved caspase-2 were detected in the retina at 5, 24, and 48 hours after injury and localized to RGC but not photoreceptors. Small interfering RNA-mediated caspase-2 knockdown neuroprotected RGC around but not in the center of the injury site. In addition, caspase-2 knockdown increased the amplitude of the ERG photopic negative response (PhNR) at 2 weeks after injury. However, siCASP2 was not protective for photoreceptors, suggesting that photoreceptor degeneration in this model is not mediated by caspase-2. Conclusions: Caspase-2 mediates death in a proportion of RGC but not photoreceptors at the site of blunt ocular trauma. Thus, intravitreally delivered siCASP2 is a possible therapeutic for the effective treatment of RGC death to prevent TON.


Assuntos
Morte Celular , Cisteína Endopeptidases/fisiologia , Traumatismos Oculares/patologia , Retina/lesões , Células Ganglionares da Retina/patologia , Ferimentos não Penetrantes/patologia , Animais , Western Blotting , Modelos Animais de Doenças , Eletrorretinografia , Traumatismos Oculares/enzimologia , Feminino , Inativação Gênica/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Injeções Intravítreas , Células Fotorreceptoras de Vertebrados/patologia , RNA Interferente Pequeno/genética , Ratos , Retina/fisiopatologia , Células Ganglionares da Retina/enzimologia , Ferimentos não Penetrantes/enzimologia
15.
Biomed Pharmacother ; 106: 923-929, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30119264

RESUMO

High glucose (HG)-induced apoptosis of retinal ganglion cells (RGCs) contributes to the pathogenesis of diabetic retinopathy, which is one of the most common and severe complications of diabetes mellitus. Accumulating evidence has documented that microRNAs (miRNAs) play an important role in the pathogenesis of diabetic retinopathy. However, the role of miRNAs in regulating HG-induced apoptosis of RGCs remains largely unknown. Various studies have suggested that miR-495 is an important regulator of cell apoptosis and survival. In this study, we aimed to investigate whether miR-495 is involved in regulating HG-induced apoptosis of RGCs and reveal its possible relevance in diabetic retinopathy. We found that miR-495 was significantly upregulated in HG-treated RGCs. Downregulation of miR-495 protected RGCs against HG-induced apoptosis, whereas overexpression of miR-495 had the opposite effect. Notably, Notch1 was identified as a target gene of miR-495, as miR-495 negatively regulated Notch1 expression and the Notch signaling pathway. Moreover, downregulation of miR-495 inhibited PTEN expression while promoting Akt activation. However, knockdown of Notch1 significantly abolished the protective effect of miR-495 inhibition against HG-induced apoptosis. Overall, our study suggests that downregulation of miR-495 alleviates HG-induced apoptosis of RGCs by targeting Notch1 to regulate PTEN/Akt signaling, which provides novel insights into understanding the pathogenesis of HG-induced apoptosis of RGCs.


Assuntos
Apoptose/efeitos dos fármacos , Glucose/toxicidade , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Células Cultivadas , Regulação da Expressão Gênica , MicroRNAs/genética , Ratos Sprague-Dawley , Receptor Notch1/genética , Células Ganglionares da Retina/enzimologia , Células Ganglionares da Retina/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
16.
J Comp Neurol ; 526(4): 742-766, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29218725

RESUMO

We report the retinal expression pattern of Ret, a receptor tyrosine kinase for the glial derived neurotrophic factor (GDNF) family ligands (GFLs), during development and in the adult mouse. Ret is initially expressed in retinal ganglion cells (RGCs), followed by horizontal cells (HCs) and amacrine cells (ACs), beginning with the early stages of postmitotic development. Ret expression persists in all three classes of neurons in the adult. Using RNA sequencing, immunostaining and random sparse recombination, we show that Ret is expressed in at least three distinct types of ACs, and ten types of RGCs. Using intersectional genetics, we describe the dendritic arbor morphologies of RGC types expressing Ret in combination with each of the three members of the POU4f/Brn3 family of transcription factors. Ret expression overlaps with Brn3a in 4 RGC types, with Brn3b in 5 RGC types, and with Brn3c in one RGC type, respectively. Ret+ RGCs project to the lateral geniculate nucleus (LGN), pretectal area (PTA) and superior colliculus (SC), and avoid the suprachiasmatic nucleus and accessory optic system. Brn3a+ Ret+ and Brn3c+ Ret+ RGCs project preferentially to contralateral retinorecipient areas, while Brn3b+ Ret+ RGCs shows minor ipsilateral projections to the olivary pretectal nucleus and the LGN. Our findings establish intersectional genetic approaches for the anatomic and developmental characterization of individual Ret+ RGC types. In addition, they provide necessary information for addressing the potential interplay between GDNF neurotrophic signaling and transcriptional regulation in RGC type specification.


Assuntos
Células Amácrinas/enzimologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células Ganglionares da Retina/enzimologia , Células Horizontais da Retina/enzimologia , Células Amácrinas/citologia , Animais , Dendritos/enzimologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos Transgênicos , Células Ganglionares da Retina/citologia , Células Horizontais da Retina/citologia , Fator de Transcrição Brn-3A/metabolismo , Vias Visuais/citologia , Vias Visuais/enzimologia , Vias Visuais/crescimento & desenvolvimento
17.
PLoS One ; 12(9): e0185390, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28938016

RESUMO

c-Jun, c-Jun N-terminal kinase(JNK) and endothelin B (ETB) receptor have been shown to contribute to the pathogenesis of glaucoma. Previously, we reported that an increase of c-Jun and CCAAT/enhancer binding protein ß (C/EBPß) immunohistostaining is associated with upregulation of the ETB receptor within the ganglion cell layer of rats with elevated intraocular pressure (IOP). In addition, both transcription factors regulate the expression of the ETB receptor in human non-pigmented ciliary epithelial cells (HNPE). The current study addressed the mechanisms by which ET-1 produced upregulation of ET receptors in primary rat retinal ganglion cells (RGCs) and HNPE cells. Treatment of ET-1 and ET-3 increased the immunocytochemical staining of c-Jun and C/EBPß in primary rat RGCs and co-localization of both transcription factors was observed. A marked increase in DNA binding activity of AP-1 and C/EBPß as well as elevated protein levels of c-Jun and c-Jun-N-terminal kinase (JNK) were detected following ET-1 treatment in HNPE cells. Overexpression of ETA or ETB receptor promoted the upregulation of c-Jun and also elevated its promoter activity. In addition, upregulation of C/EBPß augmented DNA binding and mRNA expression of c-Jun, and furthermore, the interaction of c-Jun and C/EBPß was confirmed using co-immunoprecipitation. Apoptosis of HNPE cells was identified following ET-1 treatment, and overexpression of the ETA or ETB receptor produced enhanced apoptosis. ET-1 mediated upregulation of c-Jun and C/EBPß and their interaction may represent a novel mechanism contributing to the regulation of endothelin receptor expression. Reciprocally, c-Jun was also found to regulate the ET receptors and C/EBPß appeared to play a regulatory role in promoting expression of c-Jun. Taken together, the data suggests that ET-1 triggers the upregulation of c-Jun through both ETA and ETB receptors, and conversely c-Jun also upregulates endothelin receptor expression, thereby generating a positive feed-forward loop of endothelin receptor activation and expression. This feed-forward regulation may contribute to RGC death and astrocyte proliferation following ET-1 treatment.


Assuntos
Células Epiteliais/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Receptores de Endotelina/metabolismo , Células Ganglionares da Retina/enzimologia , Animais , Apoptose/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Células Cultivadas , Cílios/enzimologia , Endotelina-1/metabolismo , Humanos , Ligação Proteica , Ratos Sprague-Dawley , Fator de Transcrição AP-1/metabolismo
18.
Cell Death Dis ; 8(6): e2847, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28569783

RESUMO

Damage-induced neuronal endopeptidase (DINE)/endothelin-converting enzyme-like 1 (ECEL1) is a membrane-bound metalloprotease that we identified as a nerve regeneration-associated molecule. The expression of DINE is upregulated in response to nerve injury in both the peripheral and central nervous systems, while its transcription is regulated by the activating transcription factor 3 (ATF3), a potent hub-transcription factor for nerve regeneration. Despite its unique hallmark of injury-induced upregulation, the physiological relevance of DINE in injured neurons has been unclear. In this study, we have demonstrated that the expression of DINE is upregulated in injured retinal ganglion cells (RGCs) in a coordinated manner with that of ATF3 after optic nerve injury, whereas DINE and ATF3 are not observed in any normal retinal cells. Recently, we have generated a mature DINE-deficient (KOTg) mouse, in which exogenous DINE is overexpressed specifically in embryonic motor neurons to avoid aberrant arborization of motor nerves and lethality after birth that occurs in the conventional DINE KO mouse. The DINE KOTg mice did not show any difference in retinal structure and the projection to brain from that of wild-type (wild type) mice under normal conditions. However, injured RGCs of DINE KOTg mice failed to regenerate even after the zymosan treatment, which is a well-known regeneration-promoting reagent. Furthermore, a DINE KOTg mouse crossed with a Atf3:BAC Tg mouse, in which green fluorescent protein (GFP) is visualized specifically in injured RGCs and optic nerves, has verified that DINE deficiency leads to regeneration failure. These findings suggest that injury-induced DINE is a crucial endopeptidase for injured RGCs to promote axonal regeneration after optic nerve injury. Thus, a DINE-mediated proteolytic mechanism would provide us with a new therapeutic strategy for nerve regeneration.


Assuntos
Fator 3 Ativador da Transcrição/genética , Metaloendopeptidases/genética , Regeneração Nervosa/genética , Traumatismos do Nervo Óptico/genética , Células Ganglionares da Retina/enzimologia , Fator 3 Ativador da Transcrição/metabolismo , Animais , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Metaloendopeptidases/deficiência , Camundongos , Camundongos Knockout , Fármacos Neuroprotetores/farmacologia , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/enzimologia , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/enzimologia , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Transdução de Sinais , Zimosan/farmacologia
19.
Cell Death Dis ; 8(4): e2752, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28425986

RESUMO

Glaucoma is a leading cause of irreversible blindness worldwide that is characterized by progressive retinal ganglion cell (RGC) death. However, RGC senescence as a phase before RGC death, and the mechanism of RGC senescence remains unclear. Here, we demonstrate that TANK-binding protein 1 (TBK1) is upregulated in acute IOP elevation-induced ischemic retinas mouse model. Moreover, pre-treatment with the TBK1 inhibitor BX-795 reduced p16INK4a (p16) expression and RGC senescence. Upregulation of TBK1 via plasmid transfection increased Akt phosphorylation at Ser473 and Bmi1 phosphorylation. The Akt inhibitor MK-2206 decreased the expression of p16 and Bmi1 serine phosphorylation. A Bmi1 Ser316 mutation also attenuated TBK1-induced p16 upregulation. Finally, silencing of TBK1 via shRNA knockdown reduced the expression of p16 as well as Akt and Bmi1 phosphorylation, reducing RGC senescence in vivo. These data suggest that acute IOP elevation-induced ischemia increases TBK1 expression, which then increases p16 expression through the Akt- Bmi1 phosphorylation pathway. This study therefore elucidates a novel mechanism whereby TBK1 regulates p16 expression and RGC senescence, suggesting a potential novel treatment strategy for minimizing RGC senescence in retinal ischemia and glaucoma.


Assuntos
Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Isquemia/enzimologia , Isquemia/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Células Ganglionares da Retina/enzimologia , Células Ganglionares da Retina/patologia , Regulação para Cima , Animais , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Pressão Intraocular/efeitos dos fármacos , Isquemia/fisiopatologia , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/farmacologia , RNA Interferente Pequeno/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tiofenos/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Diab Vasc Dis Res ; 14(3): 200-213, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28301218

RESUMO

Using a porcine model of diabetes mellitus and hypercholesterolaemia, we previously showed that diabetes mellitus and hypercholesterolaemia is associated with a chronic increase in blood-brain barrier permeability in the cerebral cortex, leading to selective binding of immunoglobulin G and deposition of amyloid-beta1-42 peptide in pyramidal neurons. Treatment with Darapladib (GlaxoSmithKline, SB480848), an inhibitor of lipoprotein-associated phospholipase-A2, alleviated these effects. Here, investigation of the effects of chronic diabetes mellitus and hypercholesterolaemia on the pig retina revealed a corresponding increased permeability of the blood-retina barrier coupled with a leak of plasma components into the retina, alterations in retinal architecture, selective IgG binding to neurons in the ganglion cell layer, thinning of retinal layers due to cell loss and increased glial fibrillary acidic protein expression in Müller cells, all of which were curtailed by treatment with Darapladib. These findings suggest that chronic diabetes mellitus and hypercholesterolaemia induces increased blood-retina barrier permeability that may be linked to altered expression of blood-retina barrier-associated tight junction proteins, claudin and occludin, leading to structural changes in the retina consistent with diabetic retinopathy. Additionally, results suggest that drugs with vascular anti-inflammatory properties, such as Darapladib, may have beneficial effects on eye diseases strongly linked to vascular abnormalities such as diabetic retinopathy and age-related macular degeneration.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/antagonistas & inibidores , Anti-Inflamatórios/farmacologia , Benzaldeídos/farmacologia , Barreira Hematorretiniana/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Retinopatia Diabética/prevenção & controle , Hipercolesterolemia/tratamento farmacológico , Oximas/farmacologia , Inibidores de Fosfolipase A2/farmacologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Barreira Hematorretiniana/enzimologia , Barreira Hematorretiniana/patologia , Barreira Hematorretiniana/fisiopatologia , Claudina-5/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/fisiopatologia , Retinopatia Diabética/enzimologia , Retinopatia Diabética/etiologia , Retinopatia Diabética/fisiopatologia , Gliose , Hipercolesterolemia/complicações , Hipercolesterolemia/enzimologia , Hipercolesterolemia/fisiopatologia , Imunoglobulina G/metabolismo , Masculino , Ocludina/metabolismo , Ligação Proteica , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/enzimologia , Sus scrofa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA