Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
DNA Cell Biol ; 42(11): 668-679, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37903234

RESUMO

Although the contribution of ferroptosis, an iron-dependent cell death, to ischemia reperfusion (IR)-induced retinal injury has been reported before, to optimize therapeutic strategy, there is still an urgent need to identify potential candidates involved in this process. Androgen Receptor-Associated Protein of 70 kDa (ARA70) is a cargo receptor for ferritinophagy, and its role in retinal ferroptosis has not been revealed yet. Herein, we explored the role of ARA70 in IR-associated retinal lesions by in vivo (C57BL/6 J mice with intraocular pressure of 90-100 mmHg) and in vitro (retinal ganglion cells (RGCs) stimulated with tert-butyl hydroperoxide (tBH)) experiments. It was found that IR upregulated ARA70 expression and accelerated lipid peroxidation in retinal tissues. We first confirmed that two ferroptosis inhibitors, deferiprone or ferrostatin-1 (Fer-1), suppressed ferritin degradation, restrained apoptosis and inflammation, and protected mouse retinas against IR stress. Next, primary mouse RGCs were treated with tBH to simulate IR environment in vitro. ARA70 expression was decreased at lower concentrations of tBH (5-20 µM), but increased at higher concentrations (40-80 µM). Interestingly, the expression of ferritin-related proteins (ferritin heavy chain, FTH; ferritin light chain, FTL) showed an opposite alteration. Knockdown of ARA70 protected RGCs from tBH-induced damage. It inhibited the delivery of ferritin to lysosomes for ferritinophagy and thus reducing cellular Fe2+ concentration. Besides, ARA70 knockdown suppressed autophagy and inflammation of tBH-treated RGCs. These findings provide novel insights into the pathogenesis of retinal IR, and may be helpful for treatment of retinal diseases.


Assuntos
Autofagia , Ferritinas , Ferroptose , Coativadores de Receptor Nuclear , Traumatismo por Reperfusão , Doenças Retinianas , Células Ganglionares da Retina , Animais , Camundongos , Ferritinas/metabolismo , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Coativadores de Receptor Nuclear/genética , Coativadores de Receptor Nuclear/fisiologia , Traumatismo por Reperfusão/metabolismo , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
2.
J Cell Physiol ; 234(6): 8873-8887, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30515784

RESUMO

Retinal neovascularization occurs in various ocular disorders including proliferative diabetic retinopathy and secondary neovascular glaucoma, resulting in blindness. This paper aims to investigate the effect of microRNA-141-3p (miR-141-3p) on retinal neovascularization and retinal ganglion cells (RGCs) in glaucoma mice through the Docking protein 5 (DOK5)-mediated mitogen-activated protein kinase (MAPK) signaling pathway. Chip retrieval and difference analysis were used for the potential mechanism of miR-141-3p on glaucoma. All modeled mice were transfected with different expression of mimic or inhibitor. The expressions of miR-141-3p, DOK5, and related genes and proteins of the MAPK signaling pathway were detected by Reverse transcription quantitative polymerase chain reaction and western blot analysis. Cell proliferation, lumen formation, and apoptosis in the retinal vascular epithelial cells and RGCs were detected using Matrigel angiogenesis and terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling assays. Moreover, a total of 63 and 294 differentially expressed genes were obtained in GSE2378 and GSE9944 chips, and 4 genes were within the intersection of the chips. In addition, the results showed that miR-141-3p was found to inhibit the DOK5 gene and activate the MAPK pathway. The number of RGCs, the expression of p38, extracellular-signal-regulated kinases (ERK), Jun N-terminal kinase (JNK), IGF-1, VEGF, HIF1-α, Bax, caspase-3, and the extent of p38, ERK, and JNK phosphorylated were decreased with miR-141-3p upregulation. Lastly, the results obtained showed that miR-141-3p inhibited the proliferation of retinal vascular epithelial cells and inhibited angiogenesis, as well as promoted apoptosis of RGCs. The study suggests that miR-141-3p inhibits retinal neovascularization in glaucoma mice by impeding the activation of the DOK5-mediated MAPK signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glaucoma/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , MicroRNAs/metabolismo , Células Ganglionares da Retina/metabolismo , Neovascularização Retiniana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/fisiologia , Proliferação de Células , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Glaucoma/patologia , Camundongos , MicroRNAs/genética
3.
Biochem Biophys Res Commun ; 463(4): 1071-6, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26102024

RESUMO

Autophagy plays an important role in the development of diabetic retinopathy (DR). Retinal pigment epithelial (RPE) cells are the main cells involved in DR, a process in which hyperglycemia plays a crucial role. This study was conducted to investigate the protective effect of autophagy against high glucose-induced inflammatory response in ARPE-19 cells and its underlying mechanism. In the present study we subjected ARPE-19 cells to high glucose stress and showed that ARPE-19 cells respond to high glucose with an increase in autophagy. 3-methyladenine (3-MA) inhibited occurrence of autophagy and it leaded to the accumulation of damaged-mitochondria-producing-ROS, and the activation of NLRP3 inflammasome, and subsequently, caused IL-1ß secretion.


Assuntos
Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Glucose/metabolismo , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico , Linhagem Celular , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo
4.
Graefes Arch Clin Exp Ophthalmol ; 253(6): 875-84, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25875044

RESUMO

BACKGROUND: The formation of scar-like fibrous tissue in age-related macular degeneration (AMD) is associated with hypoxia. Under hypoxia, retinal pigment epithelial (RPE) cells can secret more transforming growth factor-ß2 (TGF-ß2), which is determined to induce epithelial-mesenchymal transition (EMT) at certain concentrations. Whether hypoxia can induce EMT by stimulating RPE cell line secrets TGF-ß2 or not remains unknown. To gain a better understanding of the signaling mechanisms of fibrosis in AMD under hypoxic conditions, we investigated EMT in retinal pigment epithelial (RPE) cells and the effect of TGF-ß2 and Snail in this process. METHODS: Human RPE cell line (ARPE-19) was incubated with 5 % O2 for different periods of time. The expression of N-cadherin, α-smooth muscle actin (α-SMA), TGF-ß2 , and Snail were determined by Western blot and real-time PCR. Cell proliferation was assessed by CCK8 kit. RNA interference was used for multi-gene silencing of TGF-ß2 and Snail genes. RESULTS: N-cadherin was decreased and mesenchymal cell marker α-SMA was increased after the ARPE-19 cell line was incubated with 5 % O2. Meanwhile, the proliferation capability of the cell line was increased. TGF-ß2 and Snail expression were increased in a time-dependent manner under hypoxia. After multi-silencing TGF-ß2 and Snail genes, N-cadherin was increased and α-SMA was reduced. Meanwhile, the proliferation of the cell line was suppressed. CONCLUSIONS: Under hypoxic conditions, RPE cells undergo EMT. Endogenic TGF-ß2 and Snail are involved in this process. Furthermore, knockdown of both TGF-ß2 and Snail inhibited EMT to a greater extent than knockdown of either gene individually.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Inativação Gênica/fisiologia , Hipóxia/metabolismo , Epitélio Pigmentado da Retina/citologia , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta2/genética , Actinas/genética , Actinas/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Western Blotting , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Proliferação de Células , Humanos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Epitélio Pigmentado da Retina/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Transfecção , Fator de Crescimento Transformador beta2/metabolismo
5.
Graefes Arch Clin Exp Ophthalmol ; 253(1): 17-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24824367

RESUMO

PURPOSE: There is increasing evidence that suggests stromal cell-derived factor-1 (SDF-1) can induce a protective response against ischemic injury in various organs. In this study, we examined the expression of SDF-1 in a rat model of retinal ischemia-reperfusion (IR) injury. Further, we explored the effect of estrogen 17ß-estradiol (E2), and the role of estrogen receptor (ER) in regulating SDF-1 expression. METHODS: Retinal IR injury was established in Sprague-Dawley rats by elevating the intraocular pressure to 110 mmHg for 60 mins. Relative expression levels of SDF-1 mRNA and protein in the retina at 6 h, 12 h, and 24 h after reperfusion were determined by RT-PCR and western blot respectively. To investigate the influence of estrogen and ER on SDF-1 expression, E2 was administered intraperitoneally 30 mins before induction of ischemia, and the estrogen receptor antagonist ICI 182-780 was administered 1 h before E2 injection. RESULTS: SDF-1 expression in IR-injured retina is upregulated at 6 h, 12 h, and 24 h after injury, with maximum expression at 12 h. As expected, pretreatment of retinal IR rats with E2 enhanced the upregulation in SDF-1 expression after injury, through activation of the estrogen receptor. We proved this hypothesis by demonstrating that pretreatment of retinal IR rats with ICI 182-780 led to a partial decrease in E2-induced SDF-1 expression. CONCLUSIONS: Our findings suggest that 17ß-estradiol offers protection against retinal ischemic injury by inducing an upregulation in SDF-1 expression through activation of the estrogen receptor.


Assuntos
Quimiocina CXCL12/genética , Estradiol/farmacologia , Estrogênios/farmacologia , Regulação da Expressão Gênica/fisiologia , Receptores de Estrogênio/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Retina/efeitos dos fármacos , Animais , Western Blotting , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Estradiol/análogos & derivados , Antagonistas do Receptor de Estrogênio/farmacologia , Fulvestranto , Masculino , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Retina/metabolismo , Regulação para Cima
6.
Mol Vis ; 19: 1656-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23901249

RESUMO

PURPOSE: Oxidative stress on retinal pigment epithelial (RPE) cells is thought to play a crucial role in the development and progression of age-related macular degeneration. Astaxanthin (AST) is a carotenoid that shows significant antioxidant properties. This study was designed to investigate the protective effect of AST on ARPE-19 cells against oxidative stress and the possible underlying mechanism. METHODS: ARPE-19 cells exposed to different doses of H2O2 were incubated with various concentrations of AST and cell viability subsequently detected with the (4-[3-[4-iodophenyl]-2-4(4-nitrophenyl)-2H-5- tetrazolio-1,3-benzene disulfonate]; WST-1) assay. The apoptosis rate and intracellular levels of reactive oxygen species (ROS) were measured with flow cytometry. NAD(P)H quinine oxidoreductase 1 (NQO1), hemeoxygenase-1 (HO-1), glutamate-cysteine ligase modifier subunit (GCLM), and glutamate-cysteine ligase catalytic subunit (GCLC) expression were examined with real-time PCR and western blotting. The nuclear localization of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) protein and the expression levels of cleaved caspase-3 and protein kinase B proteins were evaluated with western blotting. RESULTS: AST clearly reduced H2O2-induced cell viability loss, cell apoptosis, and intracellular generation of ROS. Furthermore, treatment with AST activated the Nrf2-ARE pathway by inducing Nrf2 nuclear localization. Consequently, Phase II enzymes NQO1, HO-1, GCLM, and GCLC mRNA and proteins were increased. AST inhibited expression of H2O2-induced cleaved caspase-3 protein. Activation of the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway was involved in the protective effect of AST on the ARPE-19 cells. CONCLUSIONS: AST protected ARPE-19 cells against H2O2-induced oxidative stress via Nrf2-mediated upregulation of the expression of Phase II enzymes involving the PI3K/Akt pathway.


Assuntos
Células Epiteliais/citologia , Desintoxicação Metabólica Fase II , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/citologia , Regulação para Cima/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio/toxicidade , NAD(P)H Desidrogenase (Quinona) , Fosfatidilinositol 3-Quinases/metabolismo , Substâncias Protetoras/química , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Xantofilas/química , Xantofilas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...