Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1842(9): 1794-805, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24997453

ABSTRACT

Cataract-induced by sodium selenite in suckling rats is one of the suitable animal models to study the basic mechanism of human cataract formation. The aim of this present investigation is to study the endoplasmic reticulum (ER) stress-mediated activation of unfolded protein response (UPR), overproduction of reactive oxygen species (ROS), and suppression of Nrf2/Keap1-dependent antioxidant protection through endoplasmic reticulum-associated degradation (ERAD) pathway and Keap1 promoter DNA demethylation in human lens epithelial cells (HLECs) treated with sodium selenite. Lenses enucleated from sodium selenite injected rats generated overproduction of ROS in lens epithelial cells and newly formed lens fiber cells resulting in massive lens epithelial cells death after 1-5days. All these lenses developed nuclear cataracts after 4-5days. Sodium selenite treated HLECs induced ER stress and activated the UPR leading to release of Ca(2+) from ER, ROS overproduction and finally HLECs death. Sodium selenite also activated the mRNA expressions of passive DNA demethylation pathway enzymes such as Dnmt1, Dnmt3a, and Dnmt3b, and active DNA demethylation pathway enzyme, Tet1 leading to DNA demethylation in the Keap1 promoter of HLECs. This demethylated Keap1 promoter results in overexpression of Keap1 mRNA and protein. Overexpression Keap1 protein suppresses the Nrf2 protein through ERAD leading to suppression of Nrf2/Keap1 dependent antioxidant protection in the HLECs treated with sodium selenite. As an outcome, the cellular redox status is altered towards lens oxidation and results in cataract formation.


Subject(s)
Apoptosis , Cataract/pathology , Epithelial Cells/pathology , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/pathology , NF-E2-Related Factor 2/genetics , Oxidative Stress , Sodium Selenite/pharmacology , Animals , Blotting, Western , Cataract/chemically induced , Cataract/metabolism , Cell Proliferation , Cells, Cultured , DNA Methylation/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum-Associated Degradation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , NF-E2-Related Factor 2/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Trace Elements/pharmacology , Unfolded Protein Response
2.
Free Radic Biol Med ; 72: 134-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24746615

ABSTRACT

Age-related cataracts are a leading cause of blindness. Previously, we have demonstrated the association of the unfolded protein response with various cataractogenic stressors. However, DNA methylation alterations leading to suppression of lenticular antioxidant protection remains unclear. Here, we report the methylglyoxal-mediated sequential events responsible for Keap1 promoter DNA demethylation in human lens epithelial cells, because Keap1 is a negative regulatory protein that regulates the Nrf2 antioxidant protein. Methylglyoxal induces endoplasmic reticulum stress and activates the unfolded protein response leading to overproduction of reactive oxygen species before human lens epithelial cell death. Methylglyoxal also suppresses Nrf2 and DNA methyltransferases but activates the DNA demethylation pathway enzyme TET1. Bisulfite genomic DNA sequencing confirms the methylglyoxal-mediated Keap1 promoter DNA demethylation leading to overexpression of Keap1 mRNA and protein. Similarly, bisulfite genomic DNA sequencing shows that human clear lenses (n = 15) slowly lose 5-methylcytosine in the Keap1 promoter throughout life, at a rate of 1% per year. By contrast, diabetic cataractous lenses (n = 21) lose an average of 90% of the 5-methylcytosine regardless of age. Overexpressed Keap1 protein is responsible for decreasing Nrf2 by proteasomal degradation, thereby suppressing Nrf2-dependent stress protection. This study demonstrates for the first time the associations of unfolded protein response activation, Nrf2-dependent antioxidant system failure, and loss of Keap1 promoter methylation because of altered active and passive DNA demethylation pathway enzymes in human lens epithelial cells by methylglyoxal. As an outcome, the cellular redox balance is altered toward lens oxidation and cataract formation.


Subject(s)
Cataract/metabolism , DNA Methylation/physiology , Endoplasmic Reticulum Stress/physiology , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/metabolism , Promoter Regions, Genetic , Pyruvaldehyde/metabolism , Aging/genetics , Aging/metabolism , Animals , Blotting, Western , Cataract/genetics , Cells, Cultured , DNA Methylation/drug effects , Diabetes Mellitus, Experimental , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lens, Crystalline/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyruvaldehyde/pharmacology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Unfolded Protein Response/drug effects
3.
Exp Eye Res ; 121: 26-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24525405

ABSTRACT

Recent epidemiological studies confirm the prevalence of cataract in epileptic patients. Similarly, the drugs used to treat epilepsy also show the connection with increased cataract formation. In this present study, we investigated the suppression of Nrf2/Keap1 dependent antioxidant protection through induction of endoplasmic (ER) stress and Keap1 promoter DNA demethylation in human lens epithelial cells (HLECs) treated with valproic acid (VPA), an antiepileptic drug. 20 mM VPA induces ER stress and activates the unfolded protein response (UPR) within 4 h by activating the ER stress sensor proteins, such as PERK, IRE1α, and ATF6 in HLECs. Consequently, the integrated ER stress signals, such as eIF2α, ATF4, BiP, and CHOP are altered accordingly to induce ER-Ca2+ release, reactive oxygen species (ROS) overproduction, and cell death in HLECs treated with VPA. VPA also suppresses the Nrf2, catalase, and glutathione reductase expressions with significant increases in Keap1 protein. Bisulphite genomic DNA sequencing reveals the promoter DNA demethylation in the Keap1 promoter, which results in the overexpression of Keap1 mRNA and protein in HLECs treated with 20 mM VPA. VPA also alters the expression profiles of passive DNA demethylation pathway enzymes such Dnmt1, Dnmt3a, Dnmt3b, and active DNA demethylation pathway enzyme, TET1 leading to DNA demethylation in the Keap1 promoter of HLECs. Overexpressed Keap1 decreases the Nrf2 level, thereby abolishing the Nrf2 dependent antioxidant protection. This might be responsible for lenticular proteins oxidation and cataract formation.


Subject(s)
Anticonvulsants/pharmacology , DNA Methylation , Endoplasmic Reticulum/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/drug effects , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Valproic Acid/pharmacology , Antioxidants/metabolism , Apoptosis/drug effects , Blotting, Western , Calcium/metabolism , Catalase/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glutathione Reductase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lens, Crystalline/cytology , NF-E2-Related Factor 2/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
4.
Chem Biol Interact ; 200(1): 1-10, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22964297

ABSTRACT

To determine whether high levels of homocysteine (Hcy) induce endoplasmic reticulum (ER) stress with suppression of the nuclear factor-erythroid-2-related factor 2 (Nrf2)-dependent antioxidant protection in lens epithelial cells (LECs). ER stress was acutely induced by exposure of LECs to 100 µM Hcy without FCS and also by exposure to 5 mM Hcy with 10% FCS. After exposure to Hcy, significant changes were found in P-PERK, P-eIF2α, XBP1, Nrf2, and Keap1 within 24 h. The production of reactive oxygen species (ROS) was increased after Hcy exposure. The downstream enzymes of Nrf2 like, catalase, and glutathione reductase, were significantly decreased. These results suggested that the Hcy-induced ER stress suppressed the Nrf2-dependent antioxidant protection and simultaneously generated ROS which resulted in further oxidation and death of LECs. The loss of Nrf2 is mainly due to proteosomal degradation and m-calpain activation by the increased levels of cytoplasmic Ca(++). The caspases also play a role in the degradation of Nrf2. Our findings demonstrated that high levels of Hcy induce ER stress, chronic UPR, alter the levels of UPR specific proteins, increase the production of ROS, degrade Nrf2 and block the Nrf2-dependent antioxidant defense protection in LECs. Thus, the upregulation of ROS might exceed the Nrf2 dependent antioxidant defense protection in the LECs and result in the highly oxidized lenses and resulted in ARCs.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Cataract/metabolism , Endoplasmic Reticulum Stress/drug effects , Homocysteine/pharmacology , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cataract/chemically induced , Cataract/genetics , Cataract/pathology , Cattle , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors , Unfolded Protein Response/drug effects
5.
Biochem Biophys Res Commun ; 423(3): 542-8, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22683333

ABSTRACT

Age-related cataracts (ARCs) are the major cause of visual impairments worldwide, and diabetic adults tend to have an earlier onset of ARCs. Although age is the strongest risk factor for cataracts, little is known how age plays a role in the development of ARCs. It is known that oxidative stress in the lens increases with age and more so in the lenses of diabetics. One of the central adaptive responses against the oxidative stresses is the activation of the nuclear transcriptional factor, NF-E2-related factor 2 (Nrf2), which then activates more than 20 different antioxidative enzymes. Kelch-like ECH associated protein 1 (Keap1) targets and binds to Nrf2 for proteosomal degradation. We hypothesized that hyperglycemia will lead to a dysfunction of the Nrf2-dependent antioxidative protection in the lens of diabetics. We studied the methylation status of the CpG islands in 15 clear and 21 diabetic cataractous lenses. Our results showed significant levels of demethylated DNA in the Keap1 promoter in the cataractous lenses from diabetic patients. In contrast, highly methylated DNA was found in the clear lens and tumorized human lens epithelial cell (HLEC) lines (SRA01/04). HLECs treated with a demethylation agent, 5-aza-2'deoxycytidine (5-Aza), had a 10-fold higher levels of Keap1 mRNA, 3-fold increased levels of Keap1 protein, produced higher levels of ROS, and increased cell death. Our results indicated that demethylation of the CpG islands in the Keap1 promoter will activate the expression of Keap1 protein, which then increases the targeting of Nrf2 for proteosomal degradation. Decreased Nrf2 activity represses the transcription of many antioxidant enzyme genes and alters the redox-balance towards lens oxidation. Thus, the failure of antioxidant protection due to demethylation of the CpG islands in the Keap1 promoter is linked to the diabetic cataracts and possibly ARCs.


Subject(s)
Cataract/genetics , DNA Methylation/physiology , Diabetes Complications/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/metabolism , Oxidative Stress/genetics , Promoter Regions, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cells, Cultured , CpG Islands , DNA Methylation/drug effects , Decitabine , Humans , Kelch-Like ECH-Associated Protein 1 , Middle Aged
6.
Chem Biol Interact ; 188(1): 237-45, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20643114

ABSTRACT

Oxidative stress plays a crucial role in the progression and development of diabetes and its complications due to chronic hyperglycemia. The present study was aimed to investigate the kidney tissue protective nature of d-pinitol, a cyclitol present in soybean, by assessing the key markers of hyperglycemia-mediated oxidative stress, proinflammatory cytokines and ultrastructural alterations in streptozotocin-induced diabetic rats. Oral administration of d-pinitol (50mg/kg body weight/day) for 30 days to diabetic group of rats showed a significant elevation in the level of total protein and significant decline in the levels of blood urea, serum uric acid, creatinine and advanced glycation endproducts (AGEs) and kidney proinflammatory cytokines such as TNF-alpha, IL-1beta, IL-6, NF-kappaB p65 subunit and nitrite. Further, d-pinitol administration elicited a significant attenuation in the activities of kidney enzymatic antioxidants such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione-S-transferase (GST) and glutathione reductase (GR) and the levels of kidney non-enzymatic antioxidants such as vitamin E, vitamin C and reduced glutathione (GSH) in the diabetic group of rats, with a concomitant decline in the levels of kidney lipid peroxides, hydroperoxides and protein carbonyls. The histological and ultrastructural observations on the kidney tissues also confirmed the renoprotective nature of d-pinitol. Thus the present study demonstrated the renoprotective nature of d-pinitol by attenuating the hyperglycemia-mediated proinflammatory cytokines and antioxidant competence in kidney tissues of streptozotocin-induced diabetic rats.


Subject(s)
Cytokines/metabolism , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/metabolism , Inositol/analogs & derivatives , Kidney/drug effects , Oxidative Stress , Animals , Inositol/pharmacology , Kidney/ultrastructure , Microscopy, Electron, Transmission , Rats , Streptozocin
7.
Free Radic Res ; 44(6): 668-78, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20370563

ABSTRACT

The present study was aimed to investigate the effect of D-pinitol on hyperglycaemia mediated oxidative stress by analysing the hepatic antioxidant competence, pro-inflammatory cytokines and ultrastructural changes in liver tissues of streptozotocin-induced diabetic rats. Oral administration of D-pinitol (50 mg/kg b.w.) resulted in significant (p < 0.05) attenuation in blood glucose, glycosylated haemoglobin and pro-inflammatory markers such as TNF-alpha, IL-1beta, IL-6, NF-kappaB p65 unit and NO and significant (p < 0.05) elevation in the plasma insulin level. In addition, D-pinitol instigated a significant escalation in the levels of hepatic tissue non-enzymatic antioxidants and the activities enzymatic antioxidants of diabetic rats with significant (p < 0.05) decrease in lipid peroxides and hydroperoxides formation, thus demonstrating the protective role of D-pinitol on the hepatic tissues from oxidative stress-induced liver damage. These biochemical observations were complemented by histological and ultrastructural examination of liver section. Thus, the present study demonstrates the hepatoprotective nature of D-pinitol by attenuating hyperglycaemia-mediated pro-inflammatory cytokines and oxidative stress.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Experimental/pathology , Inositol/analogs & derivatives , Liver/pathology , Oxidative Stress/drug effects , Animals , Cytokines/analysis , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Inositol/pharmacology , Liver/drug effects , Male , Microscopy, Electron, Transmission , Rats , Rats, Wistar , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...