Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Redox Biol ; 34: 101469, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32362442

RESUMEN

Iron has been implicated in the pathogenesis of retinal degenerative diseases, including ocular siderosis. However, the mechanisms of iron-induced retinal toxicity are incompletely understood. Previous work shows that intravitreal injection of Fe2+ leads to photoreceptor (PR) oxidative stress, resulting in PR death within 14 days, and cones are more susceptible than rods to iron-induced oxidative damage. In order to further investigate the mechanism of intravitreal iron-induced retinal toxicity and shed light on mechanisms of iron-induced retinopathy in other mouse models, Fe2+, Fe3+, or saline were injected into the vitreous of adult wild-type mice. Pre-treatment with Ferrostatin-1 was used to investigate whether iron-induced retinal toxicity resulted from ferroptosis. Color and autofluorescence in vivo retinal imaging and optical coherence tomography were performed on day 2 and day 7 post-injection. Eyes were collected for quantitative PCR and Western analysis on day 1 and for immunofluorescence on both day 2 and 7. In vivo imaging and immunofluorescence revealed that Fe2+, but not Fe3+, induced PR oxidative damage and autofluorescence on day 2, resulting in PR death and retinal pigment epithelial cell (RPE) autofluorescence on day 7. Quantitative PCR and Western analysis on day 1 indicated that both Fe2+ and Fe3+ induced iron accumulation in the retina. However, only Fe2+ elevated levels of oxidative stress markers and components of ferroptosis in the retina, and killed PRs. Ferrostatin-1 failed to protect the retina from Fe2+-induced oxidative damage. To investigate the mechanism of Fe2+-induced RPE autofluorescence, rd10 mutant mice aged 6 weeks, with almost total loss of PRs, were given intravitreal Fe2+ or Fe3+ injections: neither induced RPE autofluorescence. This result suggests Fe2+-induced RPE autofluorescence in wild-type mice resulted from phagocytosed, oxidized outer segments. Together these data suggest that intraretinal Fe2+ causes PR oxidative stress, leading to PR death and RPE autofluorescence.


Asunto(s)
Hierro , Degeneración Retiniana , Animales , Ratones , Retina , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/genética , Epitelio Pigmentado de la Retina , Sulfatos
2.
Oxid Med Cell Longev ; 2020: 1769871, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33414889

RESUMEN

[This corrects the article DOI: 10.1155/2017/6210694.].

3.
Invest Ophthalmol Vis Sci ; 60(13): 4378-4387, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31634395

RESUMEN

Purpose: Iron supplementation therapy is used for iron-deficiency anemia but has been associated with macular degeneration in a 43-year-old patient. Iron entry into the neurosensory retina (NSR) can be toxic. It is important to determine conditions under which serum iron might cross the blood retinal barrier (BRB) into the NSR. Herein, an established mouse model of systemic iron overload using high-dose intraperitoneal iron dextran (IP FeDex) was studied. In addition, because the NSR expresses the iron regulatory hormone hepcidin, which could limit iron influx into the NSR, we gave retina-specific hepcidin knockout (RS-HepcKO) mice IP FeDex to test this possibility. Methods: Wild-type (WT) and RS-HepcKO mice were given IP FeDex. In vivo retina imaging was performed. Blood and tissues were analyzed for iron levels. Quantitative PCR was used to measure levels of mRNAs encoding iron regulatory and photoreceptor-specific genes. Ferritin and albumin were localized in the retina by immunofluorescence. Results: IP FeDex in both WT and RS-HepcKO mice induced high levels of iron in the liver, serum, retinal vascular endothelial cells (rVECs), and RPE, but not the NSR. The BRB remained intact. Retinal degeneration did not occur. Conclusions: Following injection of high-dose IP FeDex, iron accumulated in the BRB, but not the NSR. Thus, the BRB can shield the NSR from iron delivered in this manner. This ability is not dependent on NSR hepcidin production.


Asunto(s)
Barrera Hematorretinal/metabolismo , Células Endoteliales/metabolismo , Sobrecarga de Hierro/metabolismo , Complejo Hierro-Dextran/administración & dosificación , Hierro/metabolismo , Vasos Retinianos/metabolismo , Albúminas/metabolismo , Animales , Modelos Animales de Enfermedad , Ferritinas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Hepcidinas/farmacología , Inyecciones Intraperitoneales , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transferrina/metabolismo
4.
Exp Eye Res ; 187: 107728, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31323276

RESUMEN

Retinal iron accumulation has been implicated in the pathogenesis of age-related macular degeneration (AMD) and other neurodegenerative diseases. The retina and the brain are protected from the systemic circulation by the blood retinal barrier (BRB) and blood brain barrier (BBB), respectively. Iron levels within the retina and brain need to be tightly regulated to prevent oxidative injury. The method of iron entry through the retina and brain vascular endothelial cells (r&bVECs), an essential component of the BRB and BBB, is not fully understood. However, localization of the cellular iron exporter, ferroportin (Fpn), to the abluminal membrane of these cells, leads to the hypothesis that Fpn may play an important role in the import of iron across the BRB and BBB. To test this hypothesis, a mouse model with deletion of Fpn within the VECs in both the retina and the brain was developed through tail vein injection of AAV9-Ple261(CLDN5)-icre to both experimental Fpnf/f, and control Fpn+/+ mice at P21. Mice were aged to 9 mo and changes in retinal and brain iron distribution were observed. In vivo fundus imaging and quantitative serum iron detection were used for model validation. Eyes and brains were collected for immunofluorescence. Deletion of Fpn from the retinal and brain VECs leads to ferritin-L accumulation, an indicator of elevated iron levels, in the retinal and brain VECs. This occurred despite lower serum iron levels in the experimental mice. This result suggests that Fpn normally transfers iron from retinal and brain VECs into the retina and brain. These results help to better define the method of retina and brain iron import and will increase understanding of neurodegenerative diseases involving iron accumulation.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Circulación Cerebrovascular/fisiología , Células Endoteliales/metabolismo , Ferritinas/metabolismo , Vasos Retinianos/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica , Barrera Hematorretinal , Claudina-5/genética , Dependovirus/genética , Técnica del Anticuerpo Fluorescente Indirecta , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Am J Pathol ; 189(9): 1814-1830, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31287995

RESUMEN

The liver secretes hepcidin (Hepc) into the bloodstream to reduce blood iron levels. Hepc accomplishes this by triggering degradation of the only known cellular iron exporter ferroportin in the gut, macrophages, and liver. We previously demonstrated that systemic Hepc knockout (HepcKO) mice, which have high serum iron, develop retinal iron overload and degeneration. However, it was unclear whether this is caused by high blood iron levels or, alternatively, retinal iron influx that would normally be regulated by retina-produced Hepc. To address this question, retinas of liver-specific and retina-specific HepcKO mice were studied. Liver-specific HepcKO mice had elevated blood and retinal pigment epithelium (RPE) iron levels and increased free (labile) iron levels in the retina, despite an intact blood-retinal barrier. This led to RPE hypertrophy associated with lipofuscin-laden lysosome accumulation. Photoreceptors also degenerated focally. In contrast, there was no change in retinal or RPE iron levels or degeneration in the retina-specific HepcKO mice. These data indicate that high blood iron levels can lead to retinal iron accumulation and degeneration. High blood iron levels can occur in patients with hereditary hemochromatosis or result from use of iron supplements or multiple blood transfusions. Our results suggest that high blood iron levels may cause or exacerbate retinal disease.


Asunto(s)
Hepcidinas/fisiología , Sobrecarga de Hierro/etiología , Hierro/metabolismo , Hígado/metabolismo , Retina/metabolismo , Degeneración Retiniana/etiología , Animales , Barrera Hematorretinal , Femenino , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
6.
Sci Rep ; 8(1): 15137, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30310136

RESUMEN

Oxidative stress in retinal pigment epithelium (RPE) is considered to be a major contributor to the development and progression of age-related macular degeneration (AMD). Previous investigations have shown that sodium tanshinone IIA sulfonate (STS) can alleviate oxidative stress in haemorrhagic shock-induced organ damage and cigarette smoke-induced chronic obstructive pulmonary disease in mice. However, whether STS has a protective effect in ARPE-19 cells under oxidative stress and its exact mechanisms have not yet been fully elucidated. In the present study, we utilized H2O2 to establish an oxidative stress environment. Our findings show that STS activated the PI3K/AKT/mTOR pathway to inhibit autophagy and diminished the expression of the autophagic proteins Beclin 1, ATG3, ATG7 and ATG9 in ARPE-19 cells under oxidative stress. Detection of the intrinsic apoptosis-related factors BAX, mitochondrial membrane potential (MMP), caspase-9, caspase-3 and BCL-2, as well as the extrinsic apoptosis-related factors c-FLIP, v-FLIP and caspase-8, confirmed that STS inhibited the intrinsic and extrinsic apoptotic pathways, and attenuated apoptosis in ARPE-19 cells under oxidative stress conditions. These findings shed new light on the protective effects of STS in ARPE-19 cells and its mechanisms under oxidative stress to provide novel and promising therapeutic strategies for AMD.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenantrenos/farmacología , Sustancias Protectoras/farmacología , Biomarcadores , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
7.
Pharmaceuticals (Basel) ; 11(4)2018 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-30360383

RESUMEN

Iron is essential for life, while excess iron can be toxic. Iron generates hydroxyl radical, which is the most reactive free radical, causing oxidative stress. Since iron is absorbed through the diet but not excreted from the body, it accumulates with age in tissues, including the retina, consequently leading to age-related toxicity. This accumulation is further promoted by inflammation. Hereditary diseases such as aceruloplasminemia, Friedreich's ataxia, pantothenate kinase-associated neurodegeneration, and posterior column ataxia with retinitis pigmentosa involve retinal degeneration associated with iron dysregulation. In addition to hereditary causes, dietary or parenteral iron supplementation has been recently reported to elevate iron levels in the retinal pigment epithelium (RPE) and promote retinal degeneration. Ocular siderosis from intraocular foreign bodies or subretinal hemorrhage can also lead to retinopathy. Evidence from mice and humans suggests that iron toxicity may contribute to age-related macular degeneration pathogenesis. Iron chelators can protect photoreceptors and RPE in various mouse models. The therapeutic potential for iron chelators is under investigation.

8.
Oxid Med Cell Longev ; 2017: 6210694, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29081889

RESUMEN

BACKGROUNDS: Age-related macular degeneration is closely related to lipid oxidation, while relationship between OX-LDL and choroidal neovascularization is unclear. Recently, cylindromatosis is proved to regulate angiogenesis. However, its role in CNV progression remained unclear. Salvianolic acid A is widely used in vascular diseases. We investigated the relationship between OX-LDL and CNV and explore antineovascularization mechanism of Sal A. METHODS: C57BL6/J mice were randomized into four groups and injected with PBS or OX-LDL, together with Sal A for one week. CNV was induced by laser; CNV severity was analyzed by fundus fluorescein angiography, H&E staining, and choroid flat mount after 1 week. In in vitro experiments, ARPE-19 and HUVECs were cultured with OX-LDL (with or without Sal A) for 48 hours. Angiogenic proteins, cell junction integrity, and tube formation were measured. CYLD siRNA and specific inhibitors were used to explore mechanisms of CYLD in promoting OX-LDL-induced CNV progression. RESULTS: OX-LDL promoted laser-induced CNV volume by increasing VEGF, PDGF, and CYLD levels. Sal A antagonized OX-LDL effects and restrained CNV progression by decreasing VEGF/PDGF/CYLD, increasing antiangiostatin levels, and promoting P62-CYLD-TRAF6 interaction. CONCLUSIONS: We demonstrated oxidation damage exacerbates CNV progression, and Sal A could be a clinical therapeutic reagent to exudative AMD.


Asunto(s)
Ácidos Cafeicos/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Lactatos/uso terapéutico , Lipoproteínas LDL/efectos adversos , Degeneración Macular/tratamiento farmacológico , Animales , Ácidos Cafeicos/farmacología , Neovascularización Coroidal/patología , Regulación hacia Abajo , Humanos , Lactatos/farmacología , Ratones , Ratones Endogámicos C57BL
9.
Discov Med ; 23(125): 129-147, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28371616

RESUMEN

BACKGROUND: Salvianolic acid A (Sal A), an active monomer of Salvia miltiorrhiza, is a phenolic carboxylic acid derivative. The present study was performed to investigate the underlying mechanism of the anti-inflammation effect of Sal A, especially focusing on mTOR-KEAP1-Nrf2 and P2X7R-PKR-NLRP3 signaling pathways. METHODS: SD mice were divided into four groups: PBS, oxidized-low density lipoprotein (ox-LDL, 3 mg/kg), and ox-LDL (3 mg/kg) + Sal A (5 mg/kg) and + Sal A (10 mg/ml) groups. In in vitro experiments, ARPE-19 cells were cultured with serum free medium (SFM) or ox-LDL (100 mg/L), with or without Sal A (5 µM/50 µM) for 24 hours. RESULTS: Sal A attenuated ox-LDL-induced lipidosis and apoptosis in the retinal pigment epithelium (RPE) layer. Ox-LDL elevated ROS level and induced RPE inflammation, which were inhibited by Sal A pretreatment. Sal A activated PI3K/AKT/mTOR signaling pathway, which further promoted the disassociation of Keap1-Nrf2 complex and the phosphorylation of Nrf2. PI3K and mTOR chemical inhibitors abolished Sal A-induced Nrf2 activation while it had no influence on nlrp3 expression. Sal A also inhibited RPE inflammation by inactivating the P2x7r-Pkr-Nlrp3 signaling pathway. CONCLUSIONS: The above results indicate that Sal A protects RPE from lipid oxidative damage and chronic inflammation through up-regulating Nrf2 and inactivating the P2x7r-Pkr-Nlrp3 signaling pathway.


Asunto(s)
Ácidos Cafeicos/uso terapéutico , Lactatos/uso terapéutico , Lipoproteínas LDL/metabolismo , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...