Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 111
1.
Int Heart J ; 63(1): 113-121, 2022 Jan 29.
Article En | MEDLINE | ID: mdl-35034915

Sulfiredoxin1 (Srxn1), an endogenous antioxidant protein, is involved in cardiovascular diseases. In this study, we aimed to investigate the role of Srxn1 in VSMCs and its molecular mechanism. The murine vascular smooth muscle cells MOVAS were treated with different doses of platelet-derived growth factor-BB (PDGF-BB); then, Srxn1 expression was detected using reverse transcription-quantitative polymerase chain reaction and western blot analysis. MTT and wound healing assay were used to examine the effect of Srxn1 on MOVAS cell proliferation and migration. Reactive oxygen species (ROS) production, malondialdehyde (MDA) level, and superoxide dismutase (SOD) activity in MOVAS cells were detected using corresponding commercial kits. Moreover, the expression of proliferating cell nuclear antigen (PCNA), matrix metalloproteinase 2 (MMP-2), and nuclear factor erythroid-2-related factor 2 (Nrf2) /antioxidant response element (ARE) signaling-related proteins was detected using western blot analysis. In our study, PDGF-BB dose-dependently increased Srxn1 expression in MOVAS cells, and Srxn1 expression was increased with time dependence in PDGF-BB-treated MOVAS cells. The knockdown of Srxn1 increased PDGF-BB-induced the proliferation, migration, ROS production, MDA level, and the protein expression of PCNA and MMP-2, as well as decreased SOD activity and the expression of Nrf2/ARE signaling-related proteins in PDGF-BB-stimulated MOVAS cells. However, the overexpression of Srxn1 showed the opposite results to those of knockdown of Srxn1. Moreover, the inhibitory effects of Srxn1 overexpression on PDGF-BB induced proliferation, migration, ROS production, and MDA level and the promotion of Srxn1 overexpression on PDGF-BB induced SOD activity were partially reversed by the knockdown of Nrf2. Srxn1 inhibited PDGF-BB-induced proliferation, migration, and oxidative stress through activating Nrf2/ARE signaling.


Angiogenesis Inducing Agents/pharmacology , Becaplermin/pharmacology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Animals , Antioxidant Response Elements/physiology , Atherosclerosis/etiology , Atherosclerosis/pathology , Atherosclerosis/therapy , Cell Culture Techniques , Cell Movement , Cell Proliferation , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/physiology , Oxidative Stress , Signal Transduction
2.
Life Sci ; 291: 120111, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34732330

The Nrf2 transcription factor governs the expression of hundreds genes involved in cell defense against oxidative stress, the hallmark of numerous diseases such as neurodegenerative, cardiovascular, some viral pathologies, diabetes and others. The main route for Nrf2 activity regulation is via interactions with the Keap1 protein. Under the normoxia the Keap1 binds the Nrf2 and targets it to the proteasomal degradation, while the Keap1 is regenerated. Upon oxidative stress the interactions between Nrf2 and Keap1 are interrupted and the Nrf2 activates the transcription of the protective genes. Currently, the Nrf2 system activation is considered as a powerful cytoprotective strategy for treatment of different pathologies, which pathogenesis relies on oxidative stress including viral diseases of pivotal importance such as COVID-19. The implementation of this strategy is accomplished mainly through the inactivation of the Keap1 "guardian" function. Two approaches are now developing: the Keap1 modification via electrophilic agents, which leads to the Nrf2 release, and direct interruption of the Nrf2:Keap1 protein-protein interactions (PPI). Because of theirs chemical structure, the Nrf2 electrophilic inducers could non-specifically interact with others cellular proteins leading to undesired effects. Whereas the non-electrophilic inhibitors of the Nrf2:Keap1 PPI could be more specific, thereby widening the therapeutic window.


Antioxidant Response Elements/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Molecular Targeted Therapy/methods , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , Host-Pathogen Interactions/physiology , Humans , Ozone/therapeutic use , Protein Interaction Maps/drug effects , Signal Transduction , COVID-19 Drug Treatment
3.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article En | MEDLINE | ID: mdl-34768841

Ulcerative colitis (UC), which affects millions of people worldwide, is characterized by extensive colonic injury involving mucosal and submucosal layers of the colon. Nuclear factor E2-related factor 2 (Nrf2) plays a critical role in cellular protection against oxidant-induced stress. Antioxidant response element (ARE) is the binding site recognized by Nrf2 and leads to the expression of phase II detoxifying enzymes and antioxidant proteins. The Nrf2/ARE system is a key factor for preventing and resolving tissue injury and inflammation in disease conditions such as UC. Researchers have proposed that both Keap1-dependent and Keap1-independent cascades contribute positive effects on activation of the Nrf2/ARE pathway. In this review, we summarize the present knowledge on mechanisms controlling the activation process. We will further review nutritional compounds that can modulate activation of the Nrf2/ARE pathway and may be used as potential therapeutic application of UC. These comprehensive data will help us to better understand the Nrf2/ARE signaling pathway and promote its effective application in response to common diseases induced by oxidative stress and inflammation.


Antioxidant Response Elements/physiology , Colitis, Ulcerative/therapy , NF-E2-Related Factor 2/metabolism , Animals , Antioxidant Response Elements/genetics , Antioxidants/pharmacology , Colitis, Ulcerative/metabolism , Cytoprotection/drug effects , Humans , Inflammation/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/physiology , NF-E2-Related Factor 2/physiology , Oxidants/pharmacology , Oxidative Stress/physiology , Signal Transduction/physiology
4.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article En | MEDLINE | ID: mdl-34502501

Nrf2 is a basic region leucine-zipper transcription factor that plays a pivotal role in the coordinated gene expression of antioxidant and detoxifying enzymes, promoting cell survival in adverse environmental or defective metabolic conditions. After synthesis, Nrf2 is arrested in the cytoplasm by the Kelch-like ECH-associated protein 1 suppressor (Keap1) leading Nrf2 to ubiquitin-dependent degradation. One Nrf2 activation mechanism relies on disconnection from the Keap1 homodimer through the oxidation of cysteine at specific sites of Keap1. Free Nrf2 enters the nucleus, dimerizes with small musculoaponeurotic fibrosarcoma proteins (sMafs), and binds to the antioxidant response element (ARE) sequence of the target genes. Since oxidative stress, next to neuroinflammation and mitochondrial dysfunction, is one of the hallmarks of neurodegenerative pathologies, a molecular intervention into Nrf2/ARE signaling and the enhancement of the transcriptional activity of particular genes are targets for prevention or delaying the onset of age-related and inherited neurogenerative diseases. In this study, we review evidence for the Nrf2/ARE-driven pathway dysfunctions leading to various neurological pathologies, such as Alzheimer's, Parkinson's, and Huntington's diseases, as well as amyotrophic lateral sclerosis, and the beneficial role of natural and synthetic molecules that are able to interact with Nrf2 to enhance its protective efficacy.


Antioxidant Response Elements/physiology , NF-E2-Related Factor 2/metabolism , Neurodegenerative Diseases/metabolism , Alzheimer Disease/genetics , Antioxidant Response Elements/genetics , Antioxidants/metabolism , Humans , Huntington Disease/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/physiology , Neurodegenerative Diseases/physiopathology , Oxidative Stress/physiology , Signal Transduction/physiology
5.
In Vitro Cell Dev Biol Anim ; 57(7): 685-694, 2021 Aug.
Article En | MEDLINE | ID: mdl-34518994

The destruction of biological activity such as senescence and apoptosis caused by oxidative stress could play a pivotal role in the poor therapeutic efficiency of bone marrow mesenchymal stem cells (BMSCs) transplantation. Mitoquinone (MitoQ) has a highly effective mitochondrial antioxidant effect, and has been widely used in many oxidative damage models. This study aimed to investigate the protective effect of MitoQ on the oxidative stress-mediated senescence of canine BMSCs and the underlying mechanism. The senescence of BMSCs was determined by senescence-associated ß-galactosidase staining and quantitative real-time PCR. The expression of p-Nrf2 protein was detected by Western blotting. The results demonstrated that, as BMSCs were expanded in vitro, the senescent phenotype appeared. And the senescence of BMSCs may be caused by oxidative stress, manifested by increasing the level of ROS and decreasing the activity of antioxidant enzymes. Treatment of MitoQ down-regulated the mRNA levels of senescence-related and apoptosis-related genes, but up-regulated the mRNA levels of proliferation-related genes. Meanwhile, ROS generation and senescent activity were reduced in MitoQ-treated BMSCs. Further mechanism studies showed that MitoQ obviously promoted Nrf2 phosphorylation, and also facilitated the translocation of Nrf2 into the nucleus. Moreover, treatment of MitoQ increased the mRNA levels of downstream antioxidant genes and enhanced the activities of superoxide dismutase, catalase, and glutathione peroxidase. Thus, our study revealed that MitoQ, via the Nrf2/ARE signaling pathway, exerts an antioxidant effect as well as potentially delays OS-mediated senescence during BMSCs that were expanded in vitro, which may serve as a novel strategy to optimize the clinical application of BMSCs.


Antioxidant Response Elements/physiology , Mesenchymal Stem Cells/drug effects , NF-E2-Related Factor 2/metabolism , Organophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Animals , Antigens, CD/metabolism , Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Dogs , Enzymes/metabolism , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/metabolism , Oxidative Stress/physiology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Ubiquinone/pharmacology
6.
Exp Cell Res ; 405(2): 112689, 2021 08 15.
Article En | MEDLINE | ID: mdl-34107274

Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease globally. Elderly individuals are at a higher risk of developing NAFLD with severe clinical outcomes. Although NAFLD is closely related to liver aging, the role of hepatocyte senescence in the progression of NAFLD, especially in the development of fibrosis, is still unclear. The early stage of NAFLD is mainly characterized by lipid accumulation in hepatocytes, which could lead to severe oxidative stress, causing cellular senescence. In the present study, hepatocytes cultured in the presence of free fatty acids to induce lipid deposition were used as a hepatocyte senescence model in vitro. Senescent hepatocytes significantly increased the activation of co-cultured primary hepatic stellate cells (HSCs) and the expression of pro-fibrosis molecules. Moreover, the antioxidant regulator nuclear factor erythroid 2-related factor 2 (Nrf2) that was upregulated in senescent hepatocytes was found to be related to the activation of co-cultured HSCs. The Nrf2 agonist sulforaphane, which upregulated the transcriptional activity of the Nrf2-antioxidant response element (ARE) pathway, remarkably inhibited hepatocyte senescence and its activation effect on HSCs. However, the liver tissue obtained from non-alcoholic steatohepatitis (NASH) mice with Nrf2 knockdown showed decreased antioxidation and significant liver senescence and fibrosis. In conclusion, this study confirmed that lipid accumulation induces hepatocyte senescence, which leads to HSC activation and development of hepatic fibrosis. Increasing the activity of the Nrf2-ARE antioxidant pathway in senescent hepatocytes elicited the opposite effect, suggesting that targeting Nrf2 may prevent or delay the progression of aging-related liver fibrosis in NASH.


Antioxidant Response Elements/physiology , Hepatocytes/cytology , Lipid Metabolism/physiology , NF-E2-Related Factor 2/metabolism , Animals , Liver/metabolism , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects
7.
Brain Res Bull ; 173: 74-81, 2021 08.
Article En | MEDLINE | ID: mdl-33991607

Increasing evidence suggests that oxidative damage and neuroinflammation play a critical role in the pathogenesis of post-stroke depression (PSD). These pathologic processes are tightly regulated by the NF-E2-related factor 2/antioxidant response element (Nrf2/ARE) signaling pathway. The synthetic triterpenoid, 2-Cyano-3,12-dioxooleana-1,9-dien-28-imidazolide (CDDO-Im), is a potent Nrf2 activator. This study investigated whether CDDO-Im exhibited antidepressant-like activity and elucidated its protective mechanisms in a rat model of PSD, which was produced by middle cerebral artery occlusion (MCAO) followed by 28 days of chronic unpredictable mild stress (CUMS) in conjunction with solitary housing. The results demonstrated that CDDO-Im treatment markedly improved the depressive-like behaviors and reduced neuronal cell loss in the hippocampus, through decreasing the malondialdehyde (MDA) content (indicative of lipid peroxidation), superoxide dismutase (SOD), NF-kB activation, interleukin-6 (IL-6) and interleukin-1b (IL-1ß) in PSD rats. CDDO-Im treatment alleviated the oxidative stress and inflammatory response in PSD rats by promoting Nrf2 nuclear import and increasing the protein levels of Nrf2 downstream target genes, including heme oxygenase-1(HOMX1) and, quinone oxidoreductase-1(NQO1).These findings suggested that CDDO-Im treatment exhibited antidepressant-like effects and protected PSD rats from oxidative and inflammatory injury via the Nrf2/ARE pathway. Therefore, CDDO-Im treatment is worthy of further study.


Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Imidazoles/therapeutic use , Oleanolic Acid/analogs & derivatives , Signal Transduction/drug effects , Stroke/complications , Animals , Antidepressive Agents/pharmacology , Antioxidant Response Elements/physiology , Cell Death/drug effects , Depressive Disorder/etiology , Depressive Disorder/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Imidazoles/pharmacology , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/metabolism , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Rats
8.
Arch Toxicol ; 95(5): 1521-1533, 2021 05.
Article En | MEDLINE | ID: mdl-33554281

Nuclear factor erythroid 2-like 2 (Nrf2) is a transcription factor participating in response to cellular oxidative stress to maintain the redox balance. Generation of reactive oxygen species (ROS) and, in consequence, oxidative stress, are physiological as well as pathological processes which take place in almost all types of cells. Nrf2, in response to oxidative stress, activates expression and production of antioxidant enzymes to remove free radicals. However, the role of Nrf2 seems to be more sophisticated and its increased expression observed in cancer cells allows to draw a conclusion that its role is tissue-and condition-dependent. Interestingly, Nrf2 might also play a crucial role in response to environmental factors like mycotoxins. Thus, the aim of the study is to review the role of Nrf2 in cells exposed to most common mycotoxins to check if the Nrf2 signaling pathway serves as the main response element to mycotoxin-induced oxidative stress in human and animal cells and if it can be a target of detoxifying agents.


Mycotoxins/toxicity , NF-E2-Related Factor 2/metabolism , Animals , Antioxidant Response Elements/physiology , Antioxidants , Gene Expression Regulation , Humans , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , Response Elements , Signal Transduction
9.
Cell Biochem Biophys ; 79(2): 337-347, 2021 Jun.
Article En | MEDLINE | ID: mdl-33580396

Chelerythrine (CHE) is a natural benzophenanthridine alkaloid, which has shown its anti-fibrosis activity in kidney and liver, while the impact of CHE in pulmonary fibrosis is still unclear. This study is developed to explore the impact and mechanism of CHE in pulmonary fibrosis. Pulmonary fibrosis mouse models were established through intratracheal injection of bleomycin (BLM), after which the mice were intraperitoneally injected with CHE (0.375 or 0.75 mg/kg/d) every other day. The mice were sacrificed at the 28th day to collect blood serum, bronchoalveolar lavage fluid (BALF), and pulmonary tissues. Then, the severity of pulmonary fibrosis and the expression of nuclear factor erythroid 2 [NF-E2]-related factor 2 (Nrf2) in the pulmonary tissues were detected. Western blot analysis quantified the expressions of fibronectin and alpha-smooth muscle actin (α-SMA). The levels of 4-hydroxynonenal (4-HNE), glutathione (GSH), superoxide dismutase (SOD), TGF-ß and hydroxyproline (HP) in the BALF, and pulmonary tissues were measured. The expression levels of Nrf2 and its downstream genes, hemeoxygenase-1 (HO-1) and NAD (P) H: quinone oxidoreductase (NQO1) were examined. CHE at the concentration of 0.375 or 0.75 mg/kg/d could attenuate pulmonary fibrosis. CHE injection reduced the expression levels of fibronectin, α-SMA, and TGF-ß, upregulated the levels of SOD and GSH and decreased the levels of 4-HNE and HP. Also, CHE increased the expressions of Nrf2, HO-1, and NQO1. Treatment of Nrf2/antioxidant response element (ARE) inhibitor could block the Nrf2/ARE signaling pathway, thus perturbing the inhibition of CHE on BLM-stimulated pulmonary fibrosis in mice. CHE alleviates BLM-induced pulmonary fibrosis in mice through activating the Nrf2/ARE pathway to increase the activity of antioxidant enzymes.


Benzophenanthridines/pharmacology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Animals , Antioxidant Response Elements/physiology , Bronchoalveolar Lavage Fluid/chemistry , Collagen/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Up-Regulation/drug effects
10.
Int J Neurosci ; 131(7): 641-649, 2021 Jul.
Article En | MEDLINE | ID: mdl-32677581

INTRODUCTION: Neuroinflammation and oxidative stress are major mechanisms of post-stroke cognitive impairment (PSCI) neural injury and decreased spatial and memory capacity. Enriched environment (EE) is an effective method to improve cognitive dysfunction. However, the regulation by EE of neuroinflammation, oxidative stress and associated mechanisms in animal models remains unclear. MATERIALS AND METHODS: In this study, a rat PSCI model was established by middle cerebral artery occlusion (MCAO). Rats were randomly divided into the control group, standard environment (SE) group and EE group for 28 days. A Morris water-maze test was used to measure cognitive function at 7, 14 and 28 days after MCAO. Rats were sacrificed on the 28th day. Quantitative PCR, immunohistochemistry and ELISA were respectively used to detect mRNA expression of NF-E2-related factor 2 (Nrf2) and Nrf2 response genes, the expression of IL-1ß and levels of proinflammatory cytokines in the hippocampus. RESULTS: EE improved mNSS scores and cognitive ability in PSCI rats. EE increased mRNA expression of the Nrf2 and Nrf2 response genes, including heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO1). EE significantly decreased the level of malondialdehyde (MDA) and increased the levels of superoxide dismutase (SOD) and glutathione (GSH), in the hippocampus of PSCI rats. EE reduced the number of IL-1ß positive cells in the hippocampus, and IL-1ß levels in the hippocampus and serum. EE increased GFAP-positive astrocytes in the hippocampus, and BDNF levels in the hippocampus and serum. CONCLUSIONS: EE can improve cognitive function in PSCI rats by inhibiting neuroinflammation and oxidative stress.


Antioxidant Response Elements/physiology , Cognitive Dysfunction , Environment , Ischemic Stroke , NF-E2-Related Factor 2/metabolism , Neuroinflammatory Diseases , Oxidative Stress , Stroke Rehabilitation , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/therapy , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Ischemic Stroke/therapy , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/therapy , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
11.
Neurotox Res ; 38(4): 929-940, 2020 Dec.
Article En | MEDLINE | ID: mdl-32813208

Neuroprotective approaches comprising different mechanisms to counteract the noxious effects of excitotoxicity and oxidative stress need validation and detailed characterization. Although S-allylcysteine (SAC) is a natural compound exhibiting a broad spectrum of protective effects characterized by antioxidant, anti-inflammatory, and neuromodulatory actions, the mechanisms underlying its protective role on neuronal cell damage triggered by early excitotoxic insults remain elusive. In this study, we evaluated if the preconditioning or the post-treatment of isolated rat cortical slices with SAC (100 µM) can ameliorate the toxic effects induced by the excitotoxic metabolite quinolinic acid (QUIN, 100 µM), and whether this protective response involves the early display of specific antioxidant and neuroprotective signals. For this purpose, cell viability/mitochondrial reductive capacity, lipid peroxidation, levels of reduced and oxidized glutathione (GSH and GSSG, respectively), the rate of cell damage, the NF-E2-related factor 2/antioxidant response element (Nrf2/ARE) binding activity, heme oxygenase 1 (HO-1) regulation, extracellular signal-regulated kinase (ERK1/2) phosphorylation, and the levels of tumor necrosis factor-alpha (TNF-α) and the neurotrophin brain-derived neurotrophic factor (BDNF) were all estimated in tissue slices exposed to SAC and/or QUIN. The incubation of slices with QUIN augmented all toxic endpoints, whereas the addition of SAC prevented and/or recovered all toxic effects of QUIN, exhibiting better results when administered 60 min before the toxin and demonstrating protective and antioxidant properties. The early stimulation of Nrf2/ARE binding activity, the upregulation of HO-1, the ERK1/2 phosphorylation and the preservation of BDNF tissue levels by SAC demonstrate that this molecule displays a wide range of early protective signals by triggering orchestrated antioxidant responses and neuroprotective strategies. The relevance of the characterization of these mechanisms lies in the confirmation that the protective potential exerted by SAC begins at the early stages of excitotoxicity and neurodegeneration and supports the design of integral prophylactic/therapeutic strategies to reduce the deleterious effects observed in neurodegenerative disorders with inherent excitotoxic events.


Antioxidant Response Elements/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Cysteine/analogs & derivatives , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Animals , Antioxidant Response Elements/physiology , Cerebral Cortex/drug effects , Cysteine/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Oxidative Stress/physiology , Protein Binding/physiology , Rats , Rats, Wistar
12.
Exp Cell Res ; 393(2): 112100, 2020 08 15.
Article En | MEDLINE | ID: mdl-32442538

Oxidative stress is a key pathological factor for diabetic renal fibrosis by activating TGF-ß/Smad pathway in glomerular mesangial cells (GMCs) to promote the synthesis of extracellular matrix such as fibronectin (FN). Nuclear factor-E2-related factor (Nrf2)- anti-oxidant response element (ARE) anti-oxidative pathway has crucial renoprotective effects, and inhibiting ubiquitin-mediated degradation of Nrf2 delays diabetic renal fibrosis development. Ubiquitin-specific protease 9X (USP9X) has close relationship with oxidative stress and TGF-ß/Smad pathway, but whether it regulate diabetic renal fibrosis remains unclarified. Here, we found that advanced glycation-end products (AGEs) dose- and time-dependently reduced the protein expression and deubiquitinase activity of USP9X in GMCs. USP9X overexpression attenuated AGEs-induced upregulation of FN, TGF-ß1, and Collagen Ⅳ, three fibrosis-related marker proteins, in a deubiquitinase activity-dependent manner. While USP9X depletion with siRNAs further promoted the expressions of those proteins in AGEs-treated GMCs. Under AGEs treatment conditions, USP9X overexpression markedly increased the total and nuclear levels, ARE-binding ability, and transcriptional activity of Nrf2, upregulated the protein expressions of Nrf2 downstream genes HO-1 and NQO1, and eventually reduced the excessive production of ROS. Overexpression of the deubiquitinase catalytically inactive USP9X-C1556S mutant failed to exert such effects. Silencing Nrf2 abolished the renoprotective effects of USP9X. Further study showed that upon AGEs stimulation, Nrf2 transferred into the nucleus and the interaction between USP9X and Nrf2 was weakened. AGEs also increased Nrf2 ubiquitination level, and overexpression of USP9X, instead of USP9X-C1556S, significantly reduced the ubiquitination level of Nrf2. Taken together, USP9X reduced Nrf2 ubiquitination level and promoted Nrf2-ARE pathway activation to prevent the accumulation of extracellular matrix, eventually alleviated the pathological process of diabetic renal fibrosis.


Diabetes Mellitus, Experimental/prevention & control , Mesangial Cells/drug effects , Transforming Growth Factor beta1/metabolism , Ubiquitin Thiolesterase/pharmacology , Animals , Antioxidant Response Elements/drug effects , Antioxidant Response Elements/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glycation End Products, Advanced/pharmacology , Mesangial Cells/metabolism , Rats, Sprague-Dawley , Transforming Growth Factor beta1/drug effects , Ubiquitin-Specific Proteases/metabolism , Up-Regulation
13.
Toxicol Appl Pharmacol ; 395: 114978, 2020 05 15.
Article En | MEDLINE | ID: mdl-32234387

Parasympathetic nervous system dysfunction is common in patients with liver disease. We have previously shown that muscarinic acetylcholine receptors (mAchRs) play an important role in the regulation of hepatic fibrosis and that the receptor agonists and antagonists affect hepatocyte proliferation. However, little is known about the impact of the different mAchR subtypes and associated signaling pathways on liver injury. Here, we treated the human liver cell line HL7702 with 10 mmol/L carbon tetrachloride (CCL4) to induce hepatocyte damage. We found that CCL4 treatment increased the protein levels of group I mAchRs (M1, M3, M5) but reduced the expression of group II mAchRs (M2, M4) and activated the Nrf2/ARE and MAPK signaling pathways. Although overexpression of M1, M3, or M5 led to hepatocyte damage with an intact Nrf2/ARE pathway, overexpression of M2 or M4 increased, and siRNA-mediated knockdown of either M2 or M4 decreased the protein levels of Nrf2 and its downstream target genes. Moreover, CCL4 treatment increased serum ALT levels more significantly, but only induced slight changes in the expression of mAchRs, NQO1 and HO1, while reducing the expression of M2 and M4 in liver tissues of Nrf2-/- mice compared to wild type mice. Our findings suggest that group II mAchRs, M2 and M4, activate the Nrf2/ARE signaling pathway, which regulates the expression of M2 and M4, to protect the liver from CCL4-induced injury.


Antioxidant Response Elements/physiology , Liver Diseases/physiopathology , NF-E2-Related Factor 2/physiology , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M4/physiology , Receptors, Muscarinic/physiology , Signal Transduction/physiology , Animals , Carbon Tetrachloride/pharmacology , Cell Line , Chemical and Drug Induced Liver Injury/physiopathology , Gene Expression/drug effects , Gene Knockdown Techniques , Hepatocytes , Liver Diseases/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , RNA, Small Interfering/pharmacology , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M4/genetics , Receptors, Muscarinic/genetics , Signal Transduction/drug effects
14.
Chem Biol Interact ; 324: 109086, 2020 Jun 01.
Article En | MEDLINE | ID: mdl-32275923

Oxidative stress-induced apoptosis of retinal ganglion cells (RGCs) contributes to the development and progression of glaucoma. Sestrin2 (Sesn2), a stress-inducible protein, has a potent antioxidant capacity that can provide cytoprotection against various noxious stimuli. However, whether Sesn2 is involved in protecting RGCs from oxidative stress remains unexplored. The purpose of this study was to evaluate the role of Sesn2 in regulating hydrogen peroxide (H2O2)-induced oxidative stress of RGCs. Here, we showed that Sesn2 expression was induced in RGCs following H2O2 exposure. Sesn2 depletion markedly exacerbated H2O2-induced apoptosis and reactive oxygen species (ROS) generation in RGCs. Notably, upregulation of Sesn2 significantly decreased H2O2-induced apoptosis and ROS generation. Moreover, Sesn2 overexpression increased the nuclear translocation of nuclear factor erythroid-derived 2-like 2 (Nrf2), elevated Nrf2/antioxidant response element (ARE)-mediated transcriptional activity and upregulated the expression of Nrf2 target genes in H2O2-stimulated RGCs. Interestingly, we found that Sesn2 promoted Nrf2/ARE activation through downregulation of kelch-like ECH-associated protein 1 (Keap1). Restoration of Keap1 or inhibition of Nrf2 significantly reversed the Sesn2-mediated protective effect in H2O2-stimulated RGCs. In conclusion, these results elucidated that Sesn2 confers a protective effect in RGCs against H2O2-induced oxidative stress by reinforcing Nrf2/ARE activation via downregulation of Keap1. Our study suggests that the Sesn2/Keap1/Nrf2 axis may play an important role in retinal degeneration in glaucoma.


Apoptosis/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/physiology , Retinal Ganglion Cells/metabolism , Animals , Antioxidant Response Elements/physiology , Apoptosis/drug effects , Down-Regulation , Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Rats , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Up-Regulation
15.
Exp Neurol ; 328: 113285, 2020 06.
Article En | MEDLINE | ID: mdl-32165256

In recent years, a significant progress was made in understanding molecular mechanisms of long-term memory. Long-term memory formation requires strengthening of neuronal connections (LTP, long-term potentiation) associated with structural rearrangement of neurons. The key role in the synthesis of proteins essential for these rearrangements belong to mTOR (mammalian target of rapamycin) complexes and signaling pathways involved in mTOR regulation. Suppression of mTOR activity may impair synaptic plasticity and long-term memory, while mTOR activation inhibits autophagy, thereby potentiating amyloidosis and development of Alzheimer's disease (AD) accompanied by irreversible memory loss. Because of this, suppression/inhibition of mTOR might have unpredictable consequences on memory. The Nrf2/ARE signaling pathway affects almost all mitochondrial processes. The activation of this pathway improves memory and exhibits therapeutic effect in AD. In this review, we discuss the crosstalk between the Nrf2/ARE signaling and mTOR in the maintenance of synaptic plasticity. Nrf2 pathway can be activated by pharmacological agents and by changes in mitochondria functioning accompanying various neuronal dysfunctions.


Antioxidant Response Elements/physiology , Long-Term Potentiation/physiology , NF-E2-Related Factor 2/metabolism , Receptor Cross-Talk/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Memory, Long-Term/physiology
16.
Curr Diabetes Rev ; 16(8): 797-806, 2020.
Article En | MEDLINE | ID: mdl-32000646

Diabetes mellitus is associated with an increased risk of micro and macrovascular complications. During hyperglycemic conditions, endothelial cells and vascular smooth muscle cells are exquisitely sensitive to high glucose. This high glucose-induced sustained reactive oxygen species production leads to redox imbalance, which is associated with endothelial dysfunction and vascular wall remodeling. Nrf2, a redox-regulated transcription factor plays a key role in the antioxidant response element (ARE)-mediated expression of antioxidant genes. Although accumulating data indicate the molecular mechanisms underpinning the Nrf2 regulated redox balance, understanding the influence of the Nrf2/ARE axis during hyperglycemic condition on vascular cells is paramount. This review focuses on the context-dependent role of Nrf2/ARE signaling on vascular endothelial and smooth muscle cell function during hyperglycemic conditions. This review also highlights improving the Nrf2 system in vascular tissues, which could be a potential therapeutic strategy for vascular dysfunction.


Antioxidant Response Elements/genetics , Diabetes Mellitus, Type 2/genetics , Endothelial Cells/metabolism , Hyperglycemia/genetics , Myocytes, Smooth Muscle/metabolism , NF-E2-Related Factor 2/genetics , Animals , Antioxidant Response Elements/physiology , Antioxidants/metabolism , Diabetes Complications/genetics , Diabetes Complications/metabolism , Diabetes Complications/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Epigenesis, Genetic , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Hyperglycemia/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Oxidative Stress/genetics , Oxidative Stress/physiology , Phosphotransferases/genetics , Phosphotransferases/metabolism , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
17.
J Am Coll Surg ; 230(4): 562-571, 2020 04.
Article En | MEDLINE | ID: mdl-32032722

BACKGROUND: Mitochondrial oxidative stress plays a prominent role in the development of burn-induced cardiac dysfunction. AMP-activated kinase (AMPK), an energy sensor, has a central role in the pathogenesis of heart failure. However, its role in cardiac dysfunction after burn injury is unclear. Our hypothesis is that burn injury acts through the AMPK-sirtuin 1-PGC1α-nuclear factor erythroid 2-related factor 2 (NFE2L2)-ARE signaling pathway, leading to cardiac mitochondrial impairment, resulting in cardiac dysfunction. STUDY DESIGN: Male Sprague-Dawley rats underwent sham procedure or 60% total body surface area full-thickness burn. Echocardiograms were performed 24 hours post burn. Heart tissue was harvested at 24 hours post burn for biochemistry/molecular biologic analysis. AC16 cardiomyocytes were treated with either sham or burned rat serum (±AMPK inhibitor/AMPK activator/PGC1α activator) for evaluation of cardiomyocyte mitochondrial function by using seahorse in vitro. RESULTS: Burn injury-induced cardiac dysfunction was measured by echocardiogram. Burn injury suppressed cardiac AMPK, sirtuin 1, and PGC1 expression, leading to acetylation of cardiomyocyte proteins. In addition, burn injury caused NFE2L2 and NFE2L2 regulated antioxidants (heme oxygenase 1, NADH quinone oxidoreductase 1, glutamatecysteine ligase catalytic subunit, manganese superoxide dismutase, and glutathione peroxidase) to decrease, resulting in cardiac oxidative stress. In vitro, AMPK1 activator and PGC1α agonist treatment improved Ac16 cell mitochondrial dysfunction, and AMPK1 inhibitor treatment worsened Ac16 cellular damage. CONCLUSIONS: Burn-induced cardiac dysfunction and cardiac mitochondrial damage occur via the AMPK-sirtuin 1-PGC1α-NFE2L2-ARE signaling pathway. AMPK and PGC1α agonists might be promising therapeutic agents to reverse cardiac dysfunction after burn injury.


AMP-Activated Protein Kinases/physiology , Antioxidant Response Elements/physiology , Burns/complications , Heart Diseases/etiology , NF-E2-Related Factor 2/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Signal Transduction/physiology , Sirtuin 1/physiology , Animals , Male , Rats , Rats, Sprague-Dawley
18.
Biomed Pharmacother ; 123: 109732, 2020 Mar.
Article En | MEDLINE | ID: mdl-31945695

Hyperglycemia/oxidative stress has been implicated in the initiation and progression of diabetic complications while the components of Keap1/Nrf2/ARE signaling are being exploited as therapeutic targets for the treatment/management of these pathologies. Antioxidant agents like drugs, nutraceuticals and pure compounds that target the proteins of this pathway and their downstream genes hold the therapeutic strength to put the progression of this disease at bay. Here, we elucidate how the modulation of Keap1/Nrf2/ARE had been exploited for the treatment/management of end-stage diabetic kidney complication (diabetic nephropathy) by looking into (1) Nrf2 nuclear translocation and phosphorylation by some protein kinases at specific amino acid sequences and (2) Keap1 downregulation/Keap1-Nrf2 protein-protein inhibition (PPI) as potential therapeutic mechanisms exploited by Nrf2 activators for the modulation of diabetic nephropathy biomarkers (Collagen IV, Laminin, TGF-ß1 and Fibronectin) that ultimately lead to the amelioration of this disease progression. Furthermore, we brought to limelight the relationship between diabetic nephropathy and Keap1/Nrf2/ARE and finally elucidate how the modulation of this signaling pathway could be further explored to create novel therapeutic milestones.


Antioxidant Response Elements/physiology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Antioxidant Response Elements/genetics , Humans , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Oxidation-Reduction
19.
Nat Commun ; 10(1): 5043, 2019 11 06.
Article En | MEDLINE | ID: mdl-31695040

Abnormal cancer antioxidant capacity is considered as a potential mechanism of tumor malignancy. Modulation of oxidative stress status is emerging as an anti-cancer treatment. Our previous studies have found that Nestin-knockdown cells were more sensitive to oxidative stress in non-small cell lung cancer (NSCLC). However, the molecular mechanism by which Nestin protects cells from oxidative damage remains unclear. Here, we identify a feedback loop between Nestin and Nrf2 maintaining the redox homeostasis. Mechanistically, the ESGE motif of Nestin interacts with the Kelch domain of Keap1 and competes with Nrf2 for Keap1 binding, leading to Nrf2 escaping from Keap1-mediated degradation, subsequently promoting antioxidant enzyme generation. Interestingly, we also map that the antioxidant response elements (AREs) in the Nestin promoter are responsible for its induction via Nrf2. Taken together, our results indicate that the Nestin-Keap1-Nrf2 axis regulates cellular redox homeostasis and confers oxidative stress resistance in NSCLC.


Carcinoma, Non-Small-Cell Lung/metabolism , Homeostasis/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Nestin/metabolism , Nestin/pharmacology , A549 Cells , Animals , Antioxidant Response Elements/physiology , Antioxidants/metabolism , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nestin/genetics , Oxidative Stress , Ubiquitination/drug effects
20.
Med Sci Monit ; 25: 7966-7975, 2019 Oct 24.
Article En | MEDLINE | ID: mdl-31645538

BACKGROUND This study aimed to investigate the effects of dimethyl fumarate (DMF) on thoracic aortic atherosclerosis in the apolipoprotein E (apo-E)-deficient mouse model with streptozotocin (STZ)-induced hyperglycemia, and the signaling pathways involved. MATERIAL AND METHODS Eight-week-old ApoE-/- male mice (n=30) were randomly divided into three groups: the Control group (ApoE-/-) (n=10); the diabetic model (STZ) group (n=10); and the DMF-treated (25 mg/kg) diabetic model (DMF+STZ) group (n=10). The area of the thoracic aortic atherosclerosis was determined by histology. Reactive oxygen species (ROS) levels in mouse serum and homogenates of the thoracic aorta were determined by colorimetry. Levels of nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase subunit gp91phox were detected by immunological hybridization, and levels of heme oxygenase-1 (HO-1) were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS Compared with the Control group, in the STZ group, the area of aortic atherosclerosis was significantly increased, the levels of serum and aortic ROS, HO-1, nuclear factor-kappaB (NF-kappaB), intercellular adhesion molecule 1 (ICAM-1), and gp91phox were increased, and nuclear factor erythroid 2-related factor 2 (Nrf2), endothelial nitric oxide synthase (eNOS), and phosphorylated eNOS (p-eNOS) were significantly reduced. Compared with the STZ group, in the DMF+STZ group, the area of aortic atherosclerosis was significantly reduced, the levels of serum and aortic ROS, HO-1, NF-kappaB, ICAM-1, and gp91phox were significantly reduced, and Nrf2, eNOS, and p-eNOS were significantly increased. CONCLUSIONS In the apo-E-deficient mouse model with STZ-induced hyperglycemia, DMF reduced the development of atherosclerosis of the thoracic aorta through the nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf2/ARE) signaling pathway.


Atherosclerosis/drug therapy , Dimethyl Fumarate/pharmacology , Animals , Antioxidant Response Elements/physiology , Aorta/pathology , Apolipoproteins E/deficiency , China , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Heme Oxygenase-1/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/complications , Inflammation/pathology , Male , Mice , Mice, Knockout , NADPH Oxidases/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/deficiency , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Streptozocin/pharmacology
...