Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Exp Toxicol ; 41: 9603271221076107, 2022.
Article in English | MEDLINE | ID: mdl-35264022

ABSTRACT

Diabetic retinopathy (DR) is a common diabetic complication that severely impacts the life quality of diabetic patients. Recently, cellular senescence in human retinal endothelial cells (HRECs) induced by high glucose has been linked to the pathogenesis of DR. Fluorometholone (FML) is a glucocorticoid drug applied in the treatment of inflammatory and allergic disorders of the eye. The objective of the present study is to investigate the protective function of FML on high glucose-induced cellular senescence in HRECs. The in vitro injury model was established by stimulating HRECs with 30 mm glucose. After evaluating the cytotoxicity of FML in HRECs, 0.05% and 0.1% FML were used as the optimal concentration in the entire experiment. It was found that the excessive released inflammatory factors including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8) in HRECs induced by high glucose were significantly suppressed by FML, accompanied by the inhibitory effects on the expression levels of vascular endothelial growth factor (VEGF) and tissue factor (TF). Declined telomerase activity and enhanced senescence-associated ß-galactosidase (SA-ß-gal) activity were found in high glucose-challenged HRECs, which were dramatically alleviated by FML, accompanied by the inactivation of the p53/p21 and retinoblastoma (Rb) signaling. Interestingly, FML ameliorated high glucose-induced dephosphorylation of Akt. Lastly, the protective effects of FML against high glucose-induced cellular senescence in HRECs were abolished by the co-treatment of the PI3K/Akt signaling inhibitor LY294002, suggesting the involvement of this pathway. Taken together, these data revealed that FML-inhibited high glucose-induced cellular senescence mediated by Akt in HERCs, suggesting a novel molecular mechanism of FML.


Subject(s)
Cell Proliferation/drug effects , Cellular Senescence/drug effects , Diabetic Retinopathy/prevention & control , Endothelial Cells/drug effects , Fluorometholone/pharmacology , Protective Agents/pharmacology , Retina/drug effects , Animals , Cells, Cultured/drug effects , Diabetes Mellitus, Experimental , Diabetic Retinopathy/physiopathology , Fluorometholone/administration & dosage , Humans , Protective Agents/administration & dosage
2.
Inflamm Res ; 70(10-12): 1141-1150, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34459932

ABSTRACT

UV-B stimulation can induce retinopathy, whose pathogenesis is currently unclear. UV-B mediated inflammation in retinal endothelial cells is reported to be involved in the pathogenesis of retinopathy. S14G-humanin (HNG) is a neuroprotective peptide that has recently been reported to exert significant anti-inflammatory effects and protective properties against cell death. The present study aims to investigate the protective effects of HNG against UV-B-challenged retinal endothelial cells and explore the underlying mechanism. UV-B radiation was used to induce an injury model in human retinal endothelial cells (HRECs). First, exposure to UV-B induced the expression of TXNIP. Additionally, we found that treatment with HNG inhibited the activation of the TXNIP/NLRP3 signaling pathway and mitigated the excessive release of IL-1ß and IL-18 in UV-B-challenged HRECs. UV-B increased the expression of the transcriptional factor endothelial growth response-1 (Egr-1). Interestingly, overexpression of Egr-1 increased the luciferase activity of the TXNIP promoter as well as the mRNA and protein expression of TXNIP. In contrast, the knockdown of Egr-1 reduced the expression of TXNIP under both the normal and UV-B exposure conditions. Importantly, treatment with HNG attenuated UV-B-induced expression of Egr-1. However, overexpression of Egr-1 abolished the inhibitory effects of HNG-induced activation of NLRP3 as well as the production of IL-1ß and IL-18. Taken together, our findings reveal that HNG protected retinal endothelial cells from UV-B-induced NLRP3 inflammation activation through inhibiting TXNIP mediated by Egr-1.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Peptides/pharmacology , Radiation-Protective Agents/pharmacology , Retina/cytology , Ultraviolet Rays , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/genetics , Inflammation/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
3.
FEBS Lett ; 589(19 Pt B): 2791-6, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26296312

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression by targeted repression of transcription and translation, and are involved in carcinogenesis. In this study, we demonstrated that the expression of miR-454 was up-regulated in uveal melanoma tissues compared to normal tissues. Ectopic expression of miR-454 resulted in significant promotion of cell proliferation, colony formation, invasion and induction of cell cycle in uveal melanoma cells. Furthermore, we identified PTEN as a direct target of miR-454. Our data revealed that ectopic expression of PTEN restored the effects of miR-454 on cell proliferation and invasion in uveal melanoma cells. These findings support an oncogene role of miR-454 in development of uveal melanoma.


Subject(s)
Melanoma/genetics , Melanoma/metabolism , MicroRNAs/genetics , Oncogenes , PTEN Phosphohydrolase/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/metabolism , Base Sequence , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Melanoma/pathology , Neoplasm Invasiveness , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/genetics , Uveal Neoplasms/pathology
4.
PLoS One ; 10(5): e0124428, 2015.
Article in English | MEDLINE | ID: mdl-25961751

ABSTRACT

MicroRNAs (miRNAs) are a group endogenous small non-coding RNAs that inhibit protein translation through binding to specific target mRNAs. Recent studies have demonstrated that miRNAs are implicated in the development of cancer. However, the role of miR-144 in uveal melanoma metastasis remains largely unknown. MiR-144 was downregulated in both uveal melanoma cells and tissues. Transfection of miR-144 mimic into uveal melanoma cells led to a decrease in cell growth and invasion. After identification of two putative miR-144 binding sites within the 3' UTR of the human c-Met mRNA, miR-144 was proved to inhibit the luciferase activity inMUM-2B cells with a luciferase reporter construct containing the binding sites. In addition, the expression of c-Met protein was inhibited by miR-144. Furthermore, c-Met-mediated cell proliferation and invasion were inhibited by restoration of miR-144 in uveal melanoma cells. In conclusion, miR-144 acts as a tumor suppressor in uveal melanoma, through inhibiting cell proliferation and migration. miR-144 might serve as a potential therapeutic target in uveal melanoma patients.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , Uveal Neoplasms/genetics , 3' Untranslated Regions , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Gene Expression , Humans , Melanoma/pathology , RNA Interference , Uveal Neoplasms/pathology
6.
Bioelectromagnetics ; 35(7): 512-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25196478

ABSTRACT

The aim of this study was to evaluate effects of intermediate frequency magnetic fields (IFMF) generated by a wireless power transmission (WPT) based on magnetic resonance from the perspective of cellular genotoxicity on cultured human lens epithelial cells (HLECs). We evaluated the effects of exposure to 90 kHz magnetic fields at 93.36 µT on cellular genotoxicity in vitro for 2 and 4 h. The magnetic flux density is approximately 3.5 times higher than the reference level recommended by the International Commission on Non-Ionizing Radiation Protection (ICNIRP) guidelines. For assessment of genotoxicity, we studied cellular proliferation, apoptosis and DNA damage by Cell Counting Kit-8 (CCK-8) assay, flow cytometry analysis, alkaline comet assay and phosphorylated histone H2AX (γH2AX) foci formation test. We did not detect any effect of a 90 kHz IFMF generated by WPT based on magnetic resonance on cell proliferation, apoptosis, comet assay, and γH2AX foci formation test. Our results indicated that exposure to 90 kHz IFMF generated by WPT based on magnetic resonance at 93.36 µT for 2 and 4 h does not cause detectable cellular genotoxicity.


Subject(s)
Epithelium/radiation effects , Lens, Crystalline/radiation effects , Magnetic Fields , Wireless Technology , Apoptosis/radiation effects , Cell Line , Cell Proliferation/radiation effects , Comet Assay , DNA Damage/radiation effects , Flow Cytometry , Histones/genetics , Histones/radiation effects , Humans , Microscopy, Fluorescence , Mutagenicity Tests , Phosphorylation/drug effects
7.
PLoS One ; 9(8): e104404, 2014.
Article in English | MEDLINE | ID: mdl-25116327

ABSTRACT

Human uveitis is a type of T cell-mediated autoimmune disease that often shows relapse-remitting courses affecting multiple biological processes. As a cytoplasmic process, autophagy has been seen as an adaptive response to cell death and survival, yet the link between autophagy and T cell-mediated autoimmunity is not certain. In this study, based on the differentially expressed genes (GSE19652) between the recurrent versus monophasic T cell lines, whose adoptive transfer to susceptible animals may result in respective recurrent or monophasic uveitis, we proposed grouping annotations on a subcellular layered interactome framework to analyze the specific bioprocesses that are linked to the recurrence of T cell autoimmunity. That is, the subcellular layered interactome was established by the Cytoscape and Cerebral plugin based on differential expression, global interactome, and subcellular localization information. Then, the layered interactomes were grouping annotated by the ClueGO plugin based on Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases. The analysis showed that significant bioprocesses with autophagy were orchestrated in the cytoplasmic layered interactome and that mTOR may have a regulatory role in it. Furthermore, by setting up recurrent and monophasic uveitis in Lewis rats, we confirmed by transmission electron microscopy that, in comparison to the monophasic disease, recurrent uveitis in vivo showed significantly increased autophagy activity and extended lymphocyte infiltration to the affected retina. In summary, our framework methodology is a useful tool to disclose specific bioprocesses and molecular targets that can be attributed to a certain disease. Our results indicated that targeted inhibition of autophagy pathways may perturb the recurrence of uveitis.


Subject(s)
Autoimmune Diseases/genetics , Autophagy/genetics , Gene Regulatory Networks , Molecular Sequence Annotation , T-Lymphocytes/metabolism , Uveitis/genetics , Animals , Autoimmune Diseases/immunology , Computational Biology , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Intracellular Space/genetics , Intracellular Space/metabolism , Rats , T-Lymphocytes/immunology , T-Lymphocytes/ultrastructure , Uveitis/immunology
8.
Biol Trace Elem Res ; 158(2): 219-23, 2014 May.
Article in English | MEDLINE | ID: mdl-24604151

ABSTRACT

The present study was designed to investigate the effect of vanadium in alloxan-induced diabetes and cataract in rats. Different doses of vanadium was administered once daily for 8 weeks to alloxan-induced diabetic rats. To know the mechanism of action of vanadium, lens malondialdehyde (MDA), protein carbonyl content, activity of superoxide dismutase (SOD), activities of aldose reductase (AR), and sorbitol levels were assayed, respectively. Supplementation of vanadium to alloxan-induced diabetic rats decreased the blood glucose levels due to hyperglycemia, inhibited the AR activity, and delayed cataract progression in a dose-dependent manner. The observed beneficial effects may be attributed to polyol pathway activation but not decreased oxidative stress. Overall, the results of this study demonstrate that vanadium could effectively reduce the alloxan-induced hyperglycemia and diabetic cataracts in rats.


Subject(s)
Cataract/complications , Cataract/prevention & control , Diabetes Mellitus, Experimental/complications , Hyperglycemia/drug therapy , Vanadium/pharmacology , Vanadium/therapeutic use , Alloxan/antagonists & inhibitors , Animals , Cataract/chemically induced , Cataract/pathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Female , Hyperglycemia/chemically induced , Hyperglycemia/complications , Hyperglycemia/pathology , Male , Oxidative Stress , Rats , Rats, Wistar , Vanadium/administration & dosage
9.
Invest Ophthalmol Vis Sci ; 51(12): 6381-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20574021

ABSTRACT

PURPOSE: Glutathione S-transferase (GST) polymorphisms have been considered risk factors for the development of senile cataract. However, the results are not consistent. In this study, the authors conducted a meta-analysis to assess the association between GSTM1 and GSTT1 null genotypes and the risk for senile cataract. METHODS: Published literature from PubMed, EMBASE, and other databases were retrieved. All studies evaluating the association between GSTM1/GSTT1 polymorphisms and senile cataract were included. Pooled odds ratio (OR) and 95% confidence interval (CI) were calculated using fixed- or random-effects model. RESULTS: Eleven studies on GSTM1 (1871 cases and 1267 controls) and five studies on GSTT1 (1180 cases, 706 controls) were included. Overall analysis showed that the association between GSTM1 null genotype and risk for senile cataract is not statistically significant (OR, 1.39; 95% CI, 0.99-1.94; P = 0.054) and that the association between GSTT1 null genotype and risk for senile cataract is not significant (OR, 1.09; 95% CI, 0.87-1.36; P = 0.454). Subgroup analysis showed that the association between GSTM1 null genotype and risk for senile cataract is statistically significant in Asians (OR, 1.66; 95% CI, 1.03-2.67; P = 0.039) but not in Caucasians (OR, 1.21; 95% CI, 0.74-1.96; P = 0.443). Similar results were observed for the association between GSTT1 null genotype and risk for senile cataract. CONCLUSIONS: The present meta-analysis suggested that GSTM1 and GSTT1 null genotypes are associated with increased risk for senile cataract in Asian populations but not in Caucasian populations. Given the limited sample size, the finding on GST polymorphisms merits further investigation.


Subject(s)
Cataract/genetics , Glutathione Transferase/genetics , Polymorphism, Genetic , Asian People/genetics , Genotype , Humans , Risk Factors , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...