Coenzyme Q10 prevents human lens epithelial cells from light-induced apoptotic cell-death by reducing oxidative stress and stabilizing BAX/Bcl-2 ratio

2009 ◽  
Vol 87 ◽  
pp. 0-0
Author(s):  
M KERNT ◽  
C HIRNEISS ◽  
AS NEUBAUER ◽  
MW ULBIG ◽  
A KAMPIK
2014 ◽  
Vol 40 (8) ◽  
pp. 822-829 ◽  
Author(s):  
Meng Cai ◽  
Jing Li ◽  
Shaofen Lin ◽  
Xiaoyun Chen ◽  
Juan Huang ◽  
...  

2016 ◽  
Vol 2016 ◽  
pp. 1-17 ◽  
Author(s):  
Jiaojie Zhou ◽  
Ke Yao ◽  
Yidong Zhang ◽  
Guangdi Chen ◽  
Kairan Lai ◽  
...  

Oxidative stress plays an essential role in the development of age-related cataract. Thioredoxin binding protein-2 (TBP-2) is a negative regulator of thioredoxin (Trx), which deteriorates cellular antioxidant system. Our study focused on the autophagy-regulating effect of TBP-2 under oxidative stress in human lens epithelial cells (LECs). Human lens epithelial cells were used for cell culture and treatment. Lentiviral-based transfection system was used for overexpression of TBP-2. Cytotoxicity assay, western blot analysis, GFP/mCherry-fused LC3 plasmid, immunofluorescence, and transmission electronic microscopy were performed. The results showed that autophagic response of LECs with increased LC3-II, p62, and GFP/mCherry-LC3 puncta (P<0.01) was induced by oxidative stress. Overexpression of TBP-2 further strengthens this response and worsens the cell viability (P<0.01). Knockdown of TBP-2 attenuates the autophagic response and cell viability loss induced by oxidative stress. TBP-2 mainly regulates autophagy in the initiation stage, which is mTOR-independent and probably caused by the dephosphorylation of Akt under oxidative stress. These findings suggest a novel role of TBP-2 in human LECs under oxidative stress. Oxidative stress can cause cell injury and autophagy in LECs, and TBP-2 regulates this response. Hence, this study provides evidence regarding the role of TBP-2 in lens and the possible mechanism of cataract development.


2018 ◽  
Vol 50 (1) ◽  
pp. 246-260 ◽  
Author(s):  
Xin Liu ◽  
Chang Liu ◽  
Kun Shan ◽  
Shujie Zhang ◽  
Yi Lu ◽  
...  

Background/Aims: Age-related cataract (ARC) remains the leading cause of visual impairment among the elderly population. Long non-coding RNAs (lncRNAs) have emerged as potential regulators in many ocular diseases. However, the role of lncRNAs in nuclear ARC, a subtype of ARC, requires further elucidation. Methods: LncRNA sequencing was performed to identify differentially expressed lncRNAs between the capsules of transparent and nuclear ARC lenses. Expression validation was confirmed by qRT-PCR. MTT assay, Calcein-AM and propidium iodide double staining, Rhodamine 123 and Hoechst double staining, EdU and transwell assay were used to determine the role of H19 or miR-675 in the viability, apoptosis, proliferation and migration of primary cultured human lens epithelial cells (HLECs). Bioinformatics and luciferase reporter assays were used to identify the binding target of miR-675. Results: Sixty-three lncRNAs are differentially expressed between the capsules of transparent and nuclear ARC lenses. One top abundantly expressed lncRNA, H19, is significantly up-regulated in the nuclear ARC lens capsules and positively associated with nuclear ARC grade. H19 knockdown accelerates apoptosis development and reduces the proliferation and migration of HLECs upon oxidative stress. H19 is the precursor of miR-675, and a reduction of H19 inhibits miR-675 expression. miR-675 regulates CRYAA expression by targeting the binding site within the 3’UTR. Moreover, miR-675 increases the proliferation and migration while decreasing the apoptosis of HLECs upon oxidative stress. Conclusion: H19 regulates HLECs function through miR-675-mediated CRYAA expression. This finding would provide a novel insight into the pathogenesis of nuclear ARC.


PLoS ONE ◽  
2013 ◽  
Vol 8 (8) ◽  
pp. e72370 ◽  
Author(s):  
Shuang Ni ◽  
Yibo Yu ◽  
Yidong Zhang ◽  
Wei Wu ◽  
Kairan Lai ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document