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











Base de datos
Intervalo de año de publicación
1.
Int Immunopharmacol ; 142(Pt A): 113041, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260309

RESUMEN

INTRODUCTION: Age-related macular degeneration (AMD) is a significant contributor to irreversible impairment in visual capability, particularly in its non-neovascular (dry) form. Ferroptosis, an emerging form of programmed necrosis, involves generating lipid peroxidation (LOS) through free iron and reactive oxygen species (ROS). Salidroside, a glycoside from Rhodiola rosea, known for anti-inflammatory and antioxidant properties. The research aim was exploring whether ferroptosis exists in dry AMD pathogenesis and elucidate salidroside's protective mechanisms against ferroptosis in AMD murine models and ARPE-19 cells. METHODS: ARPE-19 cells were treated with varying concentrations of ferrous ammonium citrate (FAC) and salidroside. In an in vivo model, C57BL/6 mice were administered intraperitoneal injections of salidroside for 7 consecutive days, followed by an intravitreal injection (IVT) of FAC. After 7 days, the eyeballs were harvested for subsequent analyses. Ferroptosis markers were assessed using western blotting, immunofluorescence staining, and flow cytometry. To further elucidate the modulatory role of Nrf2 in ferroptosis, ARPE-19 cells were transfected with si-Nrf2. RESULTS: In vitro, FAC-treated ARPE-19 cells exhibited reduced viability, decreased mitochondrial membrane potential (MMP), and accumulation of iron and lipid peroxidation (LOS) products. In vivo, FAC administration by IVT led to outer nuclear layer thinning and compromised tight junctions in RPE cells. The GPX4, Nrf2, and SLC7A11 expressions were downregulated both in vitro and in vivo. Salidroside upregulated Nrf2 and ameliorated these outcomes, but its effects were attenuated in ARPE-19 cells transfected with si-Nrf2. CONCLUSION: Our study establishes that FAC induces RPE cell ferroptosis within dry AMD, and salidroside exerts therapeutic effects by triggering Nrf2/SLC7A11/GPX4 signaling axis.

2.
J Neuroinflammation ; 20(1): 308, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38129891

RESUMEN

Pathological neovascularization is a pivotal biological process in wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP) and proliferative diabetic retinopathy (PDR), in which macrophages (Mφs) play a key role. Tip cell specialization is critical in angiogenesis; however, its interconnection with the surrounding immune environment remains unclear. Succinate is an intermediate in the tricarboxylic acid (TCA) cycle and was significantly elevated in patients with wet AMD by metabolomics. Advanced experiments revealed that SUCNR1 expression in Mφ and M2 polarization was detected in abnormal vessels of choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) models. Succinate-induced M2 polarization via SUCNR1, which facilitated vascular endothelial cell (EC) migration, invasion, and tubulation, thus promoting angiogenesis in pathological neovascularization. Furthermore, evidence indicated that succinate triggered the release of RBP4 from Mφs into the surroundings to regulate endothelial sprouting and pathological angiogenesis via VEGFR2, a marker of tip cell formation. In conclusion, our results suggest that succinate represents a novel class of vasculature-inducing factors that modulate Mφ polarization and the RBP4/VEGFR2 pathway to induce pathological angiogenic signaling through tip cell specialization.


Asunto(s)
Neovascularización Coroidal , Retinopatía de la Prematuridad , Recién Nacido , Humanos , Animales , Ácido Succínico/metabolismo , Ojo/metabolismo , Neovascularización Coroidal/metabolismo , Retinopatía de la Prematuridad/metabolismo , Macrófagos/metabolismo , Modelos Animales de Enfermedad , Proteínas Plasmáticas de Unión al Retinol/metabolismo
3.
Cell Biol Toxicol ; 39(6): 2615-2630, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36786954

RESUMEN

Dry eye disease (DED) is the most common disease affecting vision and quality of life. PM2.5 was a potential risk of DED. Herein, we conducted animal exposure and cell-based studies to evaluate the pathogenic effect of PM2.5 exposure on the ocular surface and DED etiological mechanisms. C57 mice were exposed to filtered air and PM2.5 aerosol. We assessed health conditions and inflammation of the ocular surface by corneal fluorescein staining and immunohistochemistry. In parallel, cultured human corneal epithelial cells (HCETs) were treated with PM2.5, followed by characterization of cell viability, intracellular ATP level, mitochondrial activities, and expression level of DED relevant mRNA and proteins. In mice, PM2.5 exposure induced severe superficial punctate keratopathy and inflammation in their cornea. In HCETs, cell proliferation and ROS generation followed dose-response and time-dependent manner; meanwhile, mitochondrial ROS (mtROS) level increased and mitochondrial membrane potential (MMP) level decreased. Inflammation cascade was triggered even after short-term exposure. The reduction of ATP production was alleviated with Nrf2 overexpression, NF-κB P65 knockdown, or ROS clearance. Nrf2 overexpression and P65 knockdown reduced inflammatory reaction through decreasing expression of P65 and increasing of Nrf2, respectively. They partly alleviated changes of ROS/mtROS/MMP. This research proved that PM2.5 would cause DED-related inflammation reaction on corneal epithelial cells and further explored its mechanism: ROS from mitochondrial dysfunctions of corneal epithelial cells after PM2.5 exposure partly inhibited the expression of anti-inflammatory protein Nrf2 led the activation of inflammatory protein P65 and its downstream molecules, which finally caused inflammation reaction.


Asunto(s)
Síndromes de Ojo Seco , Material Particulado , Humanos , Animales , Ratones , Material Particulado/toxicidad , Material Particulado/metabolismo , Especies Reactivas de Oxígeno , Factor 2 Relacionado con NF-E2 , Calidad de Vida , Síndromes de Ojo Seco/inducido químicamente , Síndromes de Ojo Seco/metabolismo , Síndromes de Ojo Seco/patología , Inflamación , Mitocondrias/metabolismo , Adenosina Trifosfato
4.
Biomed Res Int ; 2021: 6666506, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34761005

RESUMEN

Age-related macular degeneration (AMD) is a multifactor disease, which is primarily characterized by retinal pigment epithelium (RPE) cell loss. Since the retina is the most metabolically active tissue, RPE cells are exposed to consistent oxidative environment. So, oxidation-induced RPE cell death has long been considered a contributor to the onset of AMD. Here, we applied a retinal degeneration (RD) rat model induced by blue light-emitting diode (LED) and a cell model constructed by H2O2 stimulus to mimic the prooxidant environment of the retina. We detected that the expression of miR-27a was upregulated and the expression of FOXO1 was downregulated in both models. So, we furtherly investigated the role of miR-27a-FOXO1 axis in RPE in protesting against oxidants. Lentivirus-mediated RNA was injected intravitreally into rats to modulate the miR-27a-FOXO1 axis. Retinal function and histopathological changes were evaluated by electroretinography (ERG) analysis and hematoxylin and eosin (H&E) staining, respectively. Massive photoreceptor and RPE cell death were examined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). The damage to the retina was aggravated in the FOXO1 gene-knockdown and miR-27a-overexpression groups after exposure to LED but was alleviated in the FOXO1 gene-overexpression or miR-27a-knockdown groups. Dual luciferase assay was used to detect the binding site of miR-27a and FOXO1. Upregulated miR-27a inhibited the expression of FOXO1 by directly binding to the FOXO1 mRNA 3'UTR and decreased the autophagy activity of ARPE-19 cells, resulting in the accumulation of reactive oxygen species (ROS) and decrease of cell viability. The results suggest that miR-27a is a negative regulator of FOXO1. Also, our data emphasize the prominent role of miR-27a/FOXO1 axis in modulating ROS accumulation and cell death in RPE cell model under oxidative stress and influencing the retinal function in the LED-induced RD rat model.


Asunto(s)
MicroARNs/genética , Proteínas del Tejido Nervioso/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Autofagia/genética , Muerte Celular/genética , Supervivencia Celular/genética , China , Proteína Forkhead Box O1/metabolismo , Degeneración Macular/metabolismo , Degeneración Macular/fisiopatología , Masculino , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/fisiología , Neuronas/metabolismo , Oxidación-Reducción , Estrés Oxidativo/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Retina/patología , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/fisiología
5.
Drug Des Devel Ther ; 12: 4149-4161, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30584279

RESUMEN

PURPOSE: The purpose of this study was to evaluate the effect and mechanism of quercetin on TGF-ß1-induced retinal pigment epithelial (RPE) cell proliferation, migration, and extracellular matrix secretion. MATERIALS AND METHODS: Cell counting kit-8, transwell, wound-healing assays, and ELISA were used to assess viability, migration, and collagen I secretion, respectively. Western blot analysis and qPCR were employed to detect mRNA and protein expression levels, respectively. RESULTS: Quercetin suppressed TGF-ß1-induced cell proliferation, migration, and collagen I secretion. The results also showed that mRNA and protein expression of epithelial-mesenchymal transition (EMT)-related markers such as alpha-smooth muscle actin and N-cadherin was downregulated by quercetin in TGF-ß1-treated RPE cells; conversely, quercetin upregulated the expression of E-cadherin and tight junction protein 1 (ZO-1). In addition, quercetin could inhibit mRNA and protein expression of matrix metalloproteinases. Quercetin may reverse the progression of EMT via the Smad2/3 pathway. CONCLUSION: Our results demonstrate the protective effects of quercetin on RPE cell EMT, revealing a potential therapeutic agent for proliferative vitreoretinopathy treatment.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Quercetina/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Actinas/genética , Actinas/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Fosforilación , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
6.
Mol Med Rep ; 17(4): 5878-5886, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436636

RESUMEN

The aim of the present study was to investigate the effects of bradykinin (BK) on an epithelial-mesenchymal transition (EMT) model in retinal pigment epithelium (RPE) cells through exposure to transforming growth factor­ß1 (TGF­ß1). The aim was to improve the effect of BK on proliferative vitreoretinopathy (PVR) progression, and to find a novel method of clinical prevention and treatment for PVR. The morphology of ARPE­19 cells was observed using an inverted phase­contrast microscope. A Cell Counting Kit­8 was used to assess the effects of TGF­ß1 on the proliferation of ARPE­19 cells. Western blotting and immunofluorescence were used to detect the expression levels of the epithelial marker E­cadherin, mesenchymal markers α­smooth muscle actin (SMA) and vimentin, and phosphorylated (p) mothers against decapentaplegic homolog (Smad)3 and Smad7 of the TGF/Smad signaling pathway. Wound healing tests and Transwell assays were performed to detect cell migration ability. Reverse transcription­quantitative polymerase chain reaction (RT­qPCR) analysis was performed to detect the expression levels of pSmad3 and Smad7 in the TGF/Smad signaling pathway. The results revealed that the addition of 10 ng/ml TGF­ß1 resulted in the expression of factors associated with EMT in ARPE­19 cells. BK decreased the expression levels of the mesenchymal markers α­SMA and vimentin, and increased the expression of the epithelial marker E­cadherin. BK decreased cell migration in TGF­ß1­induced EMT. These effects were reversed by HOE­140, a specific BK 2 receptor antagonist. BK significantly downregulated the expression of pSmad3 and upregulated the expression of Smad7 in TGF­ß1­treated ARPE­19 cells, and the protective alterations produced by BK were inhibited by HOE­140. In conclusion, 10 ng/ml TGF­ß1 resulted in EMT in ARPE­19 cells and BK served a negative role in TGF­ß1­induced EMT. BK had effects in TGF­ß1­induced EMT by upregulating the expression of Smad7 and downregulating the expression of pSmad3 in TGF­ß/Smad signaling pathway, indicating that BK may be a novel and effective therapy for PVR.


Asunto(s)
Bradiquinina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio Pigmentado de la Retina/citología , Factor de Crecimiento Transformador beta1/farmacología , Antagonistas del Receptor de Bradiquinina B2/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Humanos , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vitreorretinopatía Proliferativa/etiología , Vitreorretinopatía Proliferativa/metabolismo , Vitreorretinopatía Proliferativa/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA