Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 389
1.
Mediators Inflamm ; 2022: 1610293, 2022.
Article En | MEDLINE | ID: mdl-36091668

Purpose: To study the effects of a six-week endurance training protocol and calcitonin gene-related peptide (CGRP) inhibition on the nuclear factor erythroid 2-related factor 2 (Nrf2) and protein kinase B (PKB) or AKT expression in the hippocampal tissue of male Wistar rats. Main Methods. Building on a controlled experimental design with a posttest, 28 healthy Wistar male rats were randomly assigned to four groups (n = 7 per group), including control, control+CGRP inhibition, endurance training, and endurance training+CGRP inhibition groups. The training groups were trained for six weeks. Rats in the CGRP inhibition group received CGRP receptor antagonist daily (0.25 mg/kg) via intravenous (IV) injection. The Nrf2 and AKT (PKB) expression was measured using the real-time PCR technique. Results: In the endurance training group, Nrf2 expression in the hippocampal tissue was increased significantly more than in other groups (P < 0.05). There was also a significant increase in the AKT expression in the endurance training group compared to the control group (P = 0.048) and in the endurance training+CGRP inhibition compared to the control group (P = 0.012). In addition, there was no significant relationship between AKT (PKB) and Nrf2 (r = -0.27, n = 28, P = 0.16). Conclusion: Endurance training alone has been able to increase Nrf2 and AKT (PKB) mRNA levels in the hippocampal tissue, considering that endurance training had no significant effect on AKT and Nrf2 expression after adding to CGRP inhibition.


Calcitonin Gene-Related Peptide , Hippocampus , NF-E2-Related Factor 2 , Physical Conditioning, Animal , Proto-Oncogene Proteins c-akt , Animals , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/metabolism , Endurance Training/methods , Hippocampus/metabolism , Male , Models, Animal , NF-E2-Related Factor 2/biosynthesis , Physical Conditioning, Animal/physiology , Proto-Oncogene Proteins c-akt/biosynthesis , Rats , Rats, Wistar
2.
BMC Cancer ; 22(1): 289, 2022 Mar 18.
Article En | MEDLINE | ID: mdl-35300626

BACKGROUND: Pheochromocytomas (PCC) and paragangliomas (PGL) are catecholamine-producing neuroendocrine tumors. According to the World Health Organization Classification 2017, all PCC/PGL are considered to have malignant potential. There is growing evidence that PCC/PGL represent a metabolic disease that leads to aerobic glycolysis. Cellular energy metabolism involves both transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and succinate dehydrogenase (SDH) subtypes, but the association of these substances with PCC/PGL is largely unknown. METHODS: We investigated SDHB gene mutation and protein expressions for SDHB and Nrf2 in surgical specimens from 29 PCC/PGL. We also assessed preoperative maximum standard glucose uptake (SUVmax) on [18F]fluorodeoxy-glucose positron emission tomography and mRNA levels for Nrf2. RESULTS: Among 5 PCC/PGL with a PASS Score ≥ 4 or with a moderately to poorly differentiated type in the GAPP Score, 4 were metastatic and found to be SDHB mutants with homogeneous deletion of SDHB protein. SDHB mutants showed a higher expression of Nrf2 protein and a higher preoperative SUVmax than non-SDHB mutants with a PASS < 4 or a well-differentiated GAPP type. Furthermore, protein expression of Nrf2 was positively associated with preoperative SUVmax. The Nrf2 mRNA level positively correlated with malignant phenotype, higher expression for Nrf2 protein and SDHB gene mutant, but negatively correlated with expression for SDHB protein. There was also a positive correlation between Nrf2 mRNA level and SUVmax. CONCLUSION: These results suggest that activation of Nrf2 and elevated metabolism play roles in PCC/PGL with malignant potential that have SDHB gene mutation and SDHB deficiency.


Adrenal Gland Neoplasms/genetics , Glucose/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Paraganglioma/genetics , Pheochromocytoma/genetics , Succinate Dehydrogenase/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adult , Aged , Female , Germ-Line Mutation , Humans , Male , Middle Aged , NF-E2-Related Factor 2/genetics , Paraganglioma/metabolism , Paraganglioma/pathology , Phenotype , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , RNA, Messenger/analysis , Retrospective Studies , Succinate Dehydrogenase/deficiency
3.
Toxicol Appl Pharmacol ; 435: 115852, 2022 01 15.
Article En | MEDLINE | ID: mdl-34973290

Imatinib Mesylate (IMA) has been widely used to treat with chronic myeloid leukemia (CML). However, cardiotoxicity associated with IMA is included among the therapeutic strategies. The present study was aimed to discover whether ferroptosis, a programmed iron-dependent cell death, is involved in IMA-induced cardiotoxicity. In vivo, mouse model was established after treated with 25 mg/kg, 50 mg/kg and 100 mg/kg IMA. Serum CK, LDH, AST activities were determined. Cardiac tissues were examined by H&E and Oil Red O staining. MDA was measured to assess production of lipid peroxide. Tissue iron and GSH content were measured. In vitro, cell viability, mitochondria membrane potential, generation of reactive oxygen species (ROS) and cellular iron levels were performed to explore the mechanism of IMA. The in vivo results revealed that IMA treatment significantly increased serum CK, LDH and AST. H&E staining showed that IMA caused cardiac structural injuries. The dose-dependent decrease of GSH and increase of tissue iron and MDA were observed in IMA-treated groups. Oil Red O staining suggested obvious cardiac lipid accumulation after treated with IMA. In H9c2 cardiomyocytes, IMA significantly inhibited cell proliferation in a dose-dependent manner. Mitochondria membrane potential assay showed that IMA destroyed the mitochondrial function. Additionally, IMA increased the cellular ROS and iron levels. Furthermore, IMA down-regulated the expression of Nrf2 and up-regulated the expression of P53 and TfR. These results provided compelling evidence that ferroptosis participates in IMA-induced cardiotoxicity. Ferroptosis could be regarded as a target to protect against cardiotoxicity in IMA-exposed patients.


Antineoplastic Agents/toxicity , Cardiotoxicity , Ferroptosis/drug effects , Imatinib Mesylate/toxicity , NF-E2-Related Factor 2/biosynthesis , Protein Kinase Inhibitors/toxicity , Signal Transduction/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glutathione/metabolism , Iron/metabolism , Lipid Metabolism/drug effects , Lipid Peroxides/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism
4.
J Neuroinflammation ; 18(1): 226, 2021 Oct 13.
Article En | MEDLINE | ID: mdl-34645465

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) is a severe anoxic brain injury that leads to premature mortality or long-term disabilities in infants. Neuroinflammation is a vital contributor to the pathogenic cascade post-HIE and a mediator to secondary neuronal death. As a plasma membrane G-protein-coupled receptor, GPR39, exhibits anti-inflammatory activity in several diseases. This study aimed to explore the neuroprotective function of GPR39 through inhibition of inflammation post-hypoxic-ischemic (HI) injury and to elaborate the contribution of sirtuin 1(SIRT1)/peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α)/nuclear factor, erythroid 2 like 2(Nrf2) in G-protein-coupled receptor 39 (GPR39)-mediated protection. METHODS: A total of 206 10-day-old Sprague Dawley rat pups were subjected to HIE or sham surgery. TC-G 1008 was administered intranasally at 1 h, 25 h, 49 h, and 73 h post-HIE induction. SIRT1 inhibitor EX527, GPR39 CRISPR, and PGC-1α CRISPR were administered to elucidate the underlying mechanisms. Brain infarct area, short-term and long-term neurobehavioral tests, Nissl staining, western blot, and immunofluorescence staining were performed post-HIE. RESULTS: The expression of GPR39 and pathway-related proteins, SIRT1, PGC-1α and Nrf2 were increased in a time-dependent manner, peaking at 24 h or 48-h post-HIE. Intranasal administration of TC-G 1008 reduced the percent infarcted area and improved short-term and long-term neurological deficits. Moreover, TC-G 1008 treatment significantly increased the expression of SIRT1, PGC-1α and Nrf2, but downregulated the expressions of IL-6, IL-1ß, and TNF-α. GPR39 CRISPR EX527 and PGC-1α CRISPR abolished GPR39's neuroprotective effects post-HIE. CONCLUSIONS: TC-G 1008 attenuated neuroinflammation in part via the SIRT1/PGC-1α/Nrf2 pathway in a neonatal rat model of HIE. TC-G 1008 may be a novel therapeutic target for treatment post-neonatal HIE injury.


Hypoxia-Ischemia, Brain/metabolism , NF-E2-Related Factor 2/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/biosynthesis , Sirtuin 1/biosynthesis , Sulfonamides/pharmacology , Animals , Animals, Newborn , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/therapeutic use
5.
J Neuroinflammation ; 18(1): 249, 2021 Oct 30.
Article En | MEDLINE | ID: mdl-34717678

BACKGROUND: Many neurological diseases involve neuroinflammation, during which overproduction of cytokines by immune cells, especially microglia, can aggregate neuronal death. Ferroptosis is a recently discovered cell metabolism-related form of cell death and RSL3 is a well-known inducer of cell ferroptosis. Here, we aimed to investigate the effects of RSL3 in neuroinflammation and sensitivity of different type of microglia and macrophage to ferroptosis. METHODS: Here, we used quantitative RT-PCR analysis and ELISA analysis to analyze the production of proinflammatory cytokine production of microglia and macrophages after lipopolysaccharides (LPS) stimulation. We used CCK8, LDH, and flow cytometry analysis to evaluate the sensitivity of different microglia and macrophages to RSL3-induced ferroptosis. Western blot was used to test the activation of inflammatory signaling pathway and knockdown efficiency. SiRNA-mediated interference was conducted to knockdown GPX4 or Nrf2 in BV2 microglia. Intraperitoneal injection of LPS was performed to evaluate systemic inflammation and neuroinflammation severity in in vivo conditions. RESULTS: We found that ferroptosis inducer RSL3 inhibited lipopolysaccharides (LPS)-induced inflammation of microglia and peritoneal macrophages (PMs) in a cell ferroptosis-independent manner, whereas cell ferroptosis-conditioned medium significantly triggered inflammation of microglia and PMs. Different type of microglia and macrophages showed varied sensitivity to RSL3-induced ferroptosis. Mechanistically, RSL3 induced Nrf2 protein expression to inhibit RNA Polymerase II recruitment to transcription start site of proinflammatory cytokine genes to repress cytokine transcription, and protect cells from ferroptosis. Furthermore, simultaneously injection of RSL3 and Fer-1 ameliorated LPS-induced neuroinflammation in in vivo conditions. CONCLUSIONS: These data revealed the proinflammatory role of ferroptosis in microglia and macrophages, identified RSL3 as a novel inhibitor of LPS-induced inflammation, and uncovered the molecular regulation of microglia and macrophage sensitivity to ferroptosis. Thus, targeting ferroptosis in diseases by using RSL3 should consider both the pro-ferroptosis effect and the anti-inflammation effect to achieve optimal outcome.


Carbolines/pharmacology , Ferroptosis/physiology , Inflammation Mediators/metabolism , Macrophages/metabolism , Microglia/metabolism , NF-E2-Related Factor 2/biosynthesis , Animals , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Female , Ferroptosis/drug effects , Gene Expression , Inflammation Mediators/antagonists & inhibitors , Lipopolysaccharides/toxicity , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , NF-E2-Related Factor 2/genetics , RAW 264.7 Cells
6.
FASEB J ; 35(8): e21758, 2021 08.
Article En | MEDLINE | ID: mdl-34245611

After birth, the alveolar epithelium is exposed to environmental pathogens and high O2 tensions. The alveolar type II cells may protect this epithelium through surfactant production. Surfactant protein, SP-A, an immune modulator, is developmentally upregulated in fetal lung with surfactant phospholipid synthesis. Herein, we observed that the redox-regulated transcription factor, NRF2, and co-regulated C/EBPß and PPARγ, were markedly induced during cAMP-mediated differentiation of cultured human fetal lung (HFL) epithelial cells. This occurred with enhanced expression of immune modulators, SP-A, TDO2, AhR, and NQO1. Like SP-A, cAMP induction of NRF2 was prevented when cells were exposed to hypoxia. NRF2 knockdown inhibited induction of C/EBPß, PPARγ, and immune modulators. Binding of endogenous NRF2 to promoters of SP-A and other immune modulator genes increased during HFL cell differentiation. In mouse fetal lung (MFL), a developmental increase in Nrf2, SP-A, Tdo2, Ahr, and Nqo1 and decrease in Keap1 occurred from 14.5 to 18.5 dpc. Developmental induction of Nrf2 in MFL was associated with increased nuclear localization of NF-κB p65, a decline in p38 MAPK phosphorylation, increase in the MAPK phosphatase, DUSP1, induction of the histone acetylase, CBP, and decline in the histone deacetylase, HDAC4. Thus, together with surfactant production, type II cells protect the alveolar epithelium through increased expression of NRF2 and immune modulators to prevent inflammation and oxidative stress. Our findings further suggest that lung cancer cells have usurped this developmental pathway to promote immune tolerance and enhance survival.


Gene Expression Regulation, Developmental/immunology , Lung , NF-E2-Related Factor 2 , Animals , Female , Humans , Lung/embryology , Lung/immunology , Mice , Mice, Inbred ICR , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/immunology
7.
Life Sci ; 282: 119807, 2021 Oct 01.
Article En | MEDLINE | ID: mdl-34245771

AIM: This study was designed to investigate the changes of liver injury and Nrf2 signaling pathway in the process of sepsis. We also aimed to examine the role of Nrf2 in resisting oxidative stress and relieving inflammation in sepsis-induced hepatic injury. MAIN METHODS: By operating cecal ligation and puncture (CLP) on Nrf2-/- mice and wild type mice, a sepsis-induced hepatic injury model was established. We compared and contrasted the wild type mice with the Nrf2-/- mice during sepsis-induced hepatic injury, and evaluated the liver damage by biochemical analyses and staining hematoxylin-eosin (HE). Western blot or real-time PCR was performed to detect Nrf2 and its regulated genes NQO-1, GCLM and HO-1. Additionally, we detected the expressions and secretion of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), IL-1ß and anti-inflammatory cytokines IL-10. We assessed the oxidative stress through the levels of MDA and NO. KEY FINDINGS: The results showed that Nrf2 expressions at mRNA and protein levels were increased 1 day after CLP, namely the early stage of sepsis. Compared with wild type mice after CLP, Nrf2-/- mice showed more severe liver injury, accompanied by higher expression of inflammatory cytokines and oxidative stress. Notably, Nrf2-regulated genes GCLM and NQO-1, were strongly downregulated in Nrf2-/- mice. SIGNIFICANCE: Nrf2 was probably implicated in decreasing inflammatory cytokine levels and counteracting oxidative stress to alleviate sepsis-induced hepatic injury, mainly through regulating GCLM and NQO-1 in the early stage after CLP.


Gene Expression Regulation , Liver Diseases/prevention & control , Liver , NF-E2-Related Factor 2/biosynthesis , Oxidative Stress , Sepsis/metabolism , Animals , Liver/injuries , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/genetics , Liver Diseases/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Sepsis/complications , Sepsis/genetics
8.
J Neurochem ; 158(5): 1110-1130, 2021 09.
Article En | MEDLINE | ID: mdl-34254317

Bone cancer pain (BCP) is a clinical pathology that urgently needs to be solved, but research on the mechanism of BCP has so far achieved limited success. Nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) has been shown to be involved in pain, but its involvement in BCP and the specific mechanism have yet to be examined. This study aimed to test the hypothesis that BCP induces the transfer of Nrf2 from the cytoplasm to the nucleus and further promotes nuclear transcription to activate heme oxygenase-1 (HO-1) and inhibit the activation of nuclear factor-kappa B (NF-κB) signalling, ultimately regulating the neuroinflammatory response. Von-Frey was used for behavioural analysis in rats with BCP, whereas western blotting, real-time quantitative PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect molecular expression changes, and immunofluorescence was used to detect cellular localization. We demonstrated that BCP induced increased Nrf2 nuclear protein expression with decreased cytoplasmic protein expression in the spinal cord. Further increases in Nrf2 nuclear protein expression can alleviate hyperalgesia and activate HO-1 to inhibit the expression of NF-κB nuclear protein and inflammatory factors. Strikingly, intrathecal administration of the corresponding siRNA reversed the above effects. In addition, the results of double immune labelling revealed that Nrf2 and NF-κB were coexpressed in spinal cord neurons of rats with BCP. In summary, these findings suggest that the entry of Nrf2 into the nucleus promotes the expression of HO-1, inhibiting activation of the NF-κB signalling pathway, reducing neuroinflammation and ultimately exerting an anti-nociceptive effect.


Bone Neoplasms/metabolism , Cancer Pain/metabolism , Hyperalgesia/metabolism , NF-E2-Related Factor 2/biosynthesis , NF-kappa B/metabolism , Spinal Cord/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Bone Neoplasms/pathology , Cancer Pain/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Hyperalgesia/pathology , NF-kappa B/antagonists & inhibitors , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology
9.
Iran Biomed J ; 25(4): 303-7, 2021 07 01.
Article En | MEDLINE | ID: mdl-34217161

Background: In animal models of inflammatory diseases, Mammalian sterile 20-like kinase 1 (Mst1) facilitates the programmed cell death as a novel pro-apoptotic kinase. This research aimed to determine the expression level of Mst1 gene in a rat model of SCI treated with valproic acid (VPA). Methods: Severe rat model contusion was used for evaluation of the neuroprotective effect of valproic acid. The Basso-Beattie-Bresnahan test, was performed to determine locomotor functions. Hematoxylin/eosin staining and TUNEL assay were performed to detect cavity formation and apoptosis, respectively. The mRNA levels of the genes Mst1, nuclear factor (erythroid-derived 2)-like 2, and B-cell lymphoma 2 were evaluated, using quantitative real-time PCR acute spinal cord injury (RT-PCR). Results: The results revealed that Mst1 gene expression and TUNEL-positive cells in the VPA-treated group were significantly reduced as compared to the untreated group (p ≤ 0.05). Conclusion: Our findings indicate that VPA has therapeutic potential and can be a candidate for the treatment of neurodegenerative disorders and traumatic injury as a promising drug.


Locomotion/drug effects , NF-E2-Related Factor 2/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Spinal Cord Injuries/metabolism , Valproic Acid/pharmacology , Animals , Female , Gene Expression , Locomotion/physiology , NF-E2-Related Factor 2/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/genetics , Valproic Acid/therapeutic use
10.
Life Sci ; 283: 119856, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34329667

AIM: Hepatotoxicity is a critical consequence of the iron overload conditions such as hemochromatosis and blood transfusion-requiring anemia. Iron induces hepatotoxicity largely through disruption of cellular redox homeostasis and induction of inflammatory responses. The present work explored the hepatoprotective activity of the bio-active flavone galangin against iron-evoked hepatotoxicity. MAIN METHODS: Iron overload model was established in male Wistar rats via intraperitoneal injection of 150 mg/kg iron-dextran subdivided over a ten-day experimental period. Galangin was administered in a daily oral dose of 15 mg/kg throughout the experimental period. Blood and liver tissue samples were collected on day eleven and subjected to biochemical and molecular investigations. KEY FINDINGS: Galangin significantly reduced liver iron content and serum ferritin level, and alleviated the iron-evoked oxidative stress. It enhanced the liver cell integrity as reflected by decreased serum activity of the liver enzymes. Mechanistically, galangin up-regulated the redox-regulating transcription factor Nrf2 and its responsive proteins HO-1 and NQO1. Interestingly, galangin up-regulated the antioxidant and anti-inflammatory protein PPARγ and serum hepcidin levels under the iron overload conditions. Equally important, it diminished the nuclear shift of the inflammatory transcription factor NF-κB p65 and down-regulated the levels of the pro-inflammatory cytokines TNF-α and IL-1ß. SIGNIFICANCE: The results of the present study highlight the mitigating activity of galangin against iron-induced hepatotoxicity. The study accentuated targeting of Nrf2, PPARγ, and NF-κB signaling as potential contributing mechanisms. While clinical studies are still required, the current study supports the possible implementation of galangin in controlling iron overload-associated hepatotoxicity.


Flavonoids/pharmacology , Iron Overload , Liver Diseases , NF-E2-Related Factor 2/biosynthesis , PPAR gamma/biosynthesis , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Iron Overload/drug therapy , Iron Overload/metabolism , Iron Overload/pathology , Liver Diseases/drug therapy , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Rats , Rats, Wistar
11.
Biomed Pharmacother ; 141: 111801, 2021 Sep.
Article En | MEDLINE | ID: mdl-34146850

Plant-based natural extracts contain several nutrients and bioactive compounds, such as phenolics and flavonoids, that possess various health-promoting activities. This study investigated the effects of polyphenols from Pterocarpus santalinus hydroalcoholic extract (PSHE) against gamma radiation-induced derangements via the upregulation of Nrf2. Ultra High Performance Liquid Chromatography Coupled to High Resolution Mass Spectrometry (UHPLC-HRMS/MS) analysis was performed to identify the possible radioprotectors. In vivo and in vitro studies, namely Real-Time-PCR (RT-PCR) analysis, Reactive Oxygen Species (ROS) scavenging activity, lipid peroxidation and GSH levels, DNA damage and cell death studies, anti-inflammatory (Sandwich ELISA), immunomodulatory studies (antibody staining), and model free radical scavenging assays, were performed. Vanillic acid, protocatechuic acid, para-hydroxybenzoic acid, chlorogenic acid, TNF-α inhibitor (Eudesmin), isoflavone (Daidzein 7-o-glucoside), astragalin (Kaempferol 3-o-glycoside), and other polyphenols were identified in PSHE using UHPLC-HRMS/MS analysis. Prophylactic administration of PSHE (-1 h) rendered more than 33% survival in mice exposed to 8 Gy whole-body-irradiation with increased mice survival and recovery of bone marrow and spleen cellularity. Real-time RT-PCR analysis showed that PSHE treatment (50 µg/mL) upregulated Nrf2, HO-1, and GPX-1 in mice splenocytes. At 50 µg/mL, PSHE reduced ROSscavenging activity, mitochondrial and spleen membrane lipid peroxidation levels, DNA damage, and cell death, and increased GSH levels. At 10 µg/mL, PSHE treatment diminished the content of IL-6 and TNF-α. At 50 µg/mL, PSHE suppressed lymphocyte proliferation. These findings indicate that polyphenols of PSHE possess marked antioxidant, anti-inflammatory, and immunomodulatory capacities, which play important roles in the prevention of radiation damage.


Gamma Rays/adverse effects , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/radiation effects , Plant Extracts/pharmacology , Pterocarpus , Animals , Dose-Response Relationship, Drug , Free Radical Scavengers/isolation & purification , Free Radical Scavengers/pharmacology , Male , Mice , Mice, Inbred BALB C , Plant Extracts/isolation & purification , RAW 264.7 Cells , Up-Regulation/drug effects , Up-Regulation/radiation effects
12.
Brain Res Bull ; 174: 41-52, 2021 09.
Article En | MEDLINE | ID: mdl-34087360

Sleep deprivation (SD) is a hallmark of modern society and associated with many neuropsychiatric disorders, including depression and anxiety. However, the cellular and molecular mechanisms underlying SD-associated depression and anxiety remain elusive. Does the neuroinflammation play a role in mediating the effects of SD? In this study, we investigated SD-induced cellular and molecular alterations in the hippocampus and asked whether treatment with an anti-inflammatory drug, minocycline, could attenuate these alterations. We found that SD animals exhibit activated microglia and decreased levels of Keap1 and Nrf2 (antioxidant and anti-inflammatory factors) in the hippocampus. In vivo local field potential recordings show decreased theta and beta oscillations, but increased high gamma oscillations, as a result of SD. Behavioral analysis revealed increased immobility time in the forced swim and tail suspension tests, and decreased sucrose intake in SD mice, all indicative of depressive-like behavior. Moreover, open field test and elevated plus maze test results indicated that SD increases anxiety-like behavior. Interestingly, treatment with the microglial modulator minocycline prevented SD-induced microglial activation, restored Keap1 and Nrf2 levels, normalized neuronal oscillations, and alleviated depressive-like and anxiety-like behavior. The present study reveals that microglial activation and Keap1-Nrf2 signaling play a crucial role in SD-induced behavioral alteration, and that minocycline treatment has a protective effect on these alterations.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Kelch-Like ECH-Associated Protein 1/biosynthesis , Microglia/drug effects , Microglia/immunology , Minocycline/therapeutic use , NF-E2-Related Factor 2/biosynthesis , Sleep Deprivation/complications , Animals , Anxiety/prevention & control , Behavior, Animal/drug effects , Depression/prevention & control , Depression/psychology , Electroencephalography/drug effects , Female , Hindlimb Suspension , Hippocampus/metabolism , Hippocampus/pathology , Kelch-Like ECH-Associated Protein 1/genetics , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Sleep Deprivation/psychology , Swimming/psychology
13.
Neurotoxicology ; 85: 186-200, 2021 07.
Article En | MEDLINE | ID: mdl-34077701

Formononetin is an isoflavone, found in herbs like Trifolium pratense, which executes a variety of physiological activities including anti-neurodegenerative effect. However, the molecular mechanism of formononetin-mediated neuroprotection remains unclear. In this study, we investigated the protective effect of formononetin on hydrogen peroxide (H2O2)-induced death of human neuroblastoma SH-SY5Y cells and its underlying molecular mechanism. Formononetin suppressed H2O2-induced cytotoxicity. H2O2-induced increase in the intracellular reactive oxygen species (ROS) levels was decreased by formononetin, together with the enhanced expression of the antioxidant genes. H2O2-induced elevation of the Bax/Bcl-2 ratio and cleaved caspase-3 and caspase-7 levels were lowered by formononetin treatment. Moreover, formononetin repressed H2O2-induced phosphorylation of mitogen-activated protein kinases (MAPKs). Nuclear factor erythroid 2-related factor 2 (Nrf2) siRNA decreased antioxidant gene expression and elevated the H2O2-induced ROS level in the formononetin-treated cells. Furthermore, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling is involved in the activation of the nuclear translocation of Nrf2. These results indicate that the neuroprotective effect of formononetin against H2O2-induced cell death is due to a decrease in the ROS level with the enhanced expression of the antioxidant genes through activation of the PI3K/Akt-Nrf2 signaling. In addition, formononetin suppressed apoptosis through inhibition of phosphorylation of MAPKs in SH-SY5Y cells. Thus, formononetin is a potential therapeutic agent for the treatment of neurodegenerative diseases.


Cell Death/drug effects , Hydrogen Peroxide/toxicity , Isoflavones/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurons/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Neurons/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Phytoestrogens/pharmacology , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Reactive Oxygen Species/metabolism
14.
Mediators Inflamm ; 2021: 6636152, 2021.
Article En | MEDLINE | ID: mdl-33953643

Alcohol-induced liver injury implicates inflammation and oxidative stress as important mediators. Despite rigorous research, there is still no Food and Drug Administration (FDA) approved therapies for any stage of alcoholic liver disease (ALD). Interestingly, metformin (Met) and several probiotic strains possess the potential of inhibiting alcoholic liver injury. Therefore, we investigated the effectiveness of combination therapy using a mixture of eight strains of lactic acid-producing bacteria, commercialized as Visbiome® (V) and Met in preventing the ethanol-induced hepatic injury using in vitro and in vivo models. Human HepG2 cells and male Wistar rats were exposed to ethanol and simultaneously treated with probiotic V or Met alone as well as in combination. Endoplasmic reticulum (ER) stress markers, inflammatory markers, lipid metabolism, reactive oxygen species (ROS) production, and oxidative stress were evaluated, using qRT-PCR, Oil red O staining, fluorimetry, and HPLC. In vitro, probiotic V and Met in combination prevented ethanol-induced cellular injury, ER stress, oxidative stress, and regulated lipid metabolism as well as inflammatory response in HepG2 cells. Probiotic V and Met also promoted macrophage polarization towards the M2 phenotype in ethanol-exposed RAW 264.7 macrophage cells. In vivo, combined administration of probiotic V and Met ameliorated the histopathological changes, inflammatory response, hepatic markers (liver enzymes), and lipid metabolism induced by ethanol. It also improved the antioxidant markers (HO-1 and Nrf-2), as seen by their protein levels in both HepG2 cells as well as liver tissue using ELISA. Hence, probiotic V may act, in addition to the Met, as an effective preventive treatment against ethanol-induced hepatic injury.


Chemical and Drug Induced Liver Injury/drug therapy , Ethanol/toxicity , Inflammation/drug therapy , Metformin/administration & dosage , Oxidative Stress/drug effects , Probiotics/administration & dosage , Animals , Endoplasmic Reticulum Stress/drug effects , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Macrophages/physiology , Mice , NF-E2-Related Factor 2/biosynthesis , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
15.
J Biochem Mol Toxicol ; 35(8): e22821, 2021 Aug.
Article En | MEDLINE | ID: mdl-34036678

The preservation of the redox homeostasis is critical for cell survival and functionality. Redox imbalance is an essential inducer of several pathological states. CD4+ /helper T cells are highly dependent on the redox state of their surrounding milieu. The potential of the aryl hydrocarbon receptor (AhR) engagement in controlling CD4+ T-cell fate during redox alteration is still challenging. C57BL/6 mice were treated with AhR agonist 6-formylindolo[3,2-b]carbazole (FICZ), AhR antagonist CH223191, an inhibitor of glutathione biosynthesis buthionine sulfoximine (BSO), and the antioxidant N-acetylcysteine (NAC) alone or in combination. Six days later, splenocytes were evaluated for the expression of the redox-related genes and the possible changes in T-cell subsets. FICZ like BSO significantly elevated the expression of HMOX1, GCLC, and GCLM genes but it failed to increase the expression of the Nrf2 gene. Moreover, FICZ + BSO increased while FICZ + CH223191 or NAC decreased the expression of these genes. FICZ also significantly increased Th1 cell numbers but decreased Tregs in a dose-dependent manner. Furthermore, a high dose of FICZ + CH223191 + NAC significantly enhanced Th1, Th17, and Treg cells but its low dose in such a situation increased Th2 and Th17 while decreased Treg cells. AhR engagement during redox alteration can determine the fate of CD4 + T cells, so, AhR agonists or antagonists might be useful in assessing immune responses. However, these results need further verifications in vitro and in animal models of various diseases.


Receptors, Aryl Hydrocarbon , T-Lymphocytes, Helper-Inducer/metabolism , Acetylcysteine/pharmacology , Animals , Azo Compounds/pharmacology , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Membrane Proteins/biosynthesis , Mice , NF-E2-Related Factor 2/biosynthesis , Oxidation-Reduction/drug effects , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism
16.
Toxicology ; 457: 152820, 2021 06 15.
Article En | MEDLINE | ID: mdl-34023435

Prolonged exposure to therapeutic hyperoxia can induce severe side effects on intestinal epithelial cells. Meanwhile, interleukin (IL)-17D secreted by intestinal epithelial cells, plays an important role in the mucosal immune system. Therefore, this study aimed to investigate the changes of IL-17D, IL-4 and IL-6 and the regulatory effect of nuclear factor erythroid 2-related factor 2 (Nrf2) on IL-17D, IL-4 and IL-6 under hyperoxia in human intestinal epithelial cells. To achieve this, NCM460 cells were exposed to an atmosphere containing 85 % oxygen (hyperoxia) for 24 h, 48 h, or 72 h; tert-butylhydroquinone (tBHQ) and ML385 were used as an Nrf2 activator and inhibitor, respectively. Immunohistochemical staining, western blot, and reverse transcription-quantitative polymerase chain reaction were performed to detect the expression levels of IL-17D, Nrf2, Kelch-like ECH-associated protein 1 (Keap1), IL-6, and IL-4 in NCM460 cells. Results showed that hyperoxia significantly increased the expression of IL-17D, Nrf2, IL-6, and IL-4, while decreasing that of Keap1. tBHQ further activated Nrf2 and promoted the expression of IL-17D, IL-6, and IL-4. Additionally, tBHQ aggravated hyperoxia-induced inflammation caused by hyperoxia. In contrast, ML385 completely inhibited the expression of Nrf2 and IL-17D, transiently inhibited IL-6 and IL-4 expression, and did not influence Keap1 expression. These results cumulatively demonstrate that hyperoxia aggravates the inflammatory response in intestinal epithelial cells by activating the Nrf2/IL-17D axis.


Cytokines/biosynthesis , Hyperoxia/metabolism , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Intestinal Mucosa/metabolism , NF-E2-Related Factor 2/biosynthesis , Cell Line , Cytokines/genetics , Gene Expression , Humans , Hyperoxia/pathology , Interleukin-17/genetics , Intestinal Mucosa/pathology , NF-E2-Related Factor 2/genetics
17.
Neurotox Res ; 39(4): 1323-1337, 2021 Aug.
Article En | MEDLINE | ID: mdl-33999357

The present study aimed to evaluate the neuroprotective effects and underlying mechanisms of pinocembrin-7-methylether (PME), a natural bioflavonoid, in 6-hydroxydopamine (6-OHDA)-induced models of Parkinson's disease in vivo and in vitro. First, we found that PME decreased apoptosis in 6-OHDA-intoxicated SH-SY5Y cells. PME also blocked several 6-OHDA-induced mitochondrial apoptotic cascades, including loss of mitochondrial membrane potential, caspase 3 and PARP activation, and a decrease in the Bcl-2/Bax ratio. Also, PME suppressed 6-OHDA-induced oxidative stress while increasing antioxidant enzymatic activity. Further investigations indicated that PME significantly enhanced nuclear accumulation of Nrf2, improved ARE promoter activity, and upregulated HO-1 and NQO1 expression levels. In addition, siRNA-mediated Nrf2 knockdown abolished PME-induced anti-oxidative and anti-apoptotic effects. Interestingly, we found that PME promoted phosphorylation of AKT and ERK, whereas pharmacological inhibition of AKT or ERK pathways diminished PME-induced Nrf2 activation and protective actions. Moreover, PME attenuated 6-OHDA-induced loss of dopaminergic neurons and ameliorated locomotor deficiency in zebrafish, supporting the neuroprotective actions of PME in vivo. In summary, we found that PME conferred neuroprotection against 6-OHDA-induced neurotoxicity in PD models in vivo and in vitro. Taken together, our findings suggest that activation of Nrf2/ARE/HO-1 signaling cascades contributes to PME-induced anti-oxidative and neuroprotective actions, which are at least partially mediated by AKT and ERK pathways.


Flavanones/pharmacology , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/biosynthesis , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Proto-Oncogene Proteins c-akt/biosynthesis , Zebrafish Proteins/biosynthesis , Animals , Cell Line, Tumor , DNA Fragmentation/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Flavanones/therapeutic use , Humans , MAP Kinase Signaling System/physiology , Neuroprotective Agents/therapeutic use , Zebrafish
18.
Mol Cell Biochem ; 476(7): 2719-2727, 2021 Jul.
Article En | MEDLINE | ID: mdl-33677805

Hypercholesterolemia is one of the risk factors associated with increased morbidity and mortality in cardiovascular disorders. Chrysin (Chy) is reported to exhibit anti-inflammatory, anti-cancerous, anti-oxidative, anti-aging, and anti-atherogenic properties. In the present study, we aimed to investigate whether Chy would mediate the cardioprotective effect against hypercholesterolemia-triggered myocardial oxidative stress. Male Sprague Dawley rats were divided into different groups as control and fed with high-fat diet (HFD) followed by oral administration of Chy (100 mg/kg b.wt), atorvastatin (Atv) (10 mg/kg b.wt), and L-NAME (10 mg/kg b.wt) for 30 days. At the end of the experimental period, the rats were sacrificed and tissues were harvested. Biochemical results showed a significant increase of cardiac disease marker enzymes (ALT, AST, and CKMB), lipid peroxidation, and lipid profile (TC, TG, LDL, and VLDL) in HFD-fed rat tissues when compared to control, whereas oral administration of Chy significantly reduced the activities of these marker enzymes and controlled the lipid profile. qRT-PCR studies revealed that Chy administration significantly increased the expression of endothelial nitric oxide synthase (eNOS), and Nrf2 target genes such as SOD, catalase, and GCL3 in left ventricular heart tissue of HFD-challenged rats. Immunohistochemistry results also showed that Chy treatment increased myocardial protein expression of eNOS and Nrf2 in HFD-challenged rats. Concluding the results of the present study, the Chy could mediate the cardioprotective effect through the activation of eNOS and Nrf2 signaling against hypercholesterolemia-induced oxidative stress. Thus, the administration of Chy would provide a promising therapeutic strategy for the prevention of HFD-induced oxidative stress-mediated myocardial complications.


Diet, High-Fat/adverse effects , Flavonoids/pharmacology , Myocardium/metabolism , NF-E2-Related Factor 2/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Oxidative Stress/drug effects , Up-Regulation/drug effects , Animals , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley
19.
Neurochem Int ; 145: 105007, 2021 05.
Article En | MEDLINE | ID: mdl-33675841

Diabetic retinopathy is the most common cause of vision loss among diabetic patients. Although hyperglycemia produces retinal oxidative stress in long-standing diabetes, the pathogenesis mechanism is unknown. The Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a central role in cell responses against oxidative damage. We used adult Long Evans rats where diabetes was induced by streptozotocin. Normal and treated rats were sacrificed at 7, 20, and 45 days after streptozotocin injection. We analyzed Nrf2 and Keap1 expression in retinal homogenates, cytoplasmic, and nuclear retinal fractions. Normal retina showed Nrf2 expression in all retina nuclear layers. We found a transitory decrease of Nrf2 mRNA and protein expression at 7 and 20 days after the streptozotocin injection that recovered later on: moreover, the protein level increased after 45 days. Keap1 immunoprecipitation revealed similar levels as Nrf2 in normal and diabetic rat retinas, indicating that the diabetic condition did not lead to dissociation of the Keap1-Nrf2 complex. Indeed, glutathione levels and superoxide dismutase activity were not altered in the treated rat retinas. These results do not support oxidative stress in the retina shortly after diabetes induction.


Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , NF-E2-Related Factor 2/biosynthesis , Retina/metabolism , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/chemically induced , Diabetic Retinopathy/genetics , Female , Gene Expression , NF-E2-Related Factor 2/genetics , Rats , Rats, Long-Evans , Retina/drug effects , Streptozocin
20.
Inflammation ; 44(4): 1629-1642, 2021 Aug.
Article En | MEDLINE | ID: mdl-33709265

Multiple sclerosis is a chronic inflammatory neurodegenerative disease of the central nervous system which injures the myelin sheath. Telmisartan and nifedipine are antihypertensive drugs that recently showed neuroprotective properties against neurodegenerative diseases. This study evaluated the neuroprotective effect of telmisartan or nifedipine in cuprizone-induced demyelination in mice by examining the underlying mechanisms. C57BL/6 mice received a diet containing 0.7% (w/w) cuprizone for 7 days followed by 3 weeks on a 0.2% cuprizone diet. Telmisartan (5 mg/kg/day, p.o.) or nifedipine (5 mg/kg/day, p.o.) was administered for 3 weeks starting from the second week. Telmisartan or nifedipine improved locomotor activity and enhanced motor coordination as demonstrated by open field, rotarod, and grip strength tests. Furthermore, telmisartan or nifedipine restored myelin basic protein mRNA and protein expression and increased luxol fast blue-staining intensity. Telmisartan or nifedipine attenuated cuprizone-induced oxidative stress and apoptosis by decreasing brain malondialdehyde and caspase-3 along with restoring reduced glutathione and brain-derived neurotrophic factor levels. Telmisartan or nifedipine exerted an anti-inflammatory effect by reducing the expression of nuclear factor kappa B (NF-κB p65) as well as pro-inflammatory cytokines and elevating the expression of IκB-α. In parallel, telmisartan or nifedipine upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and the levels of heme oxygenase-1 and NADPH quinone oxidoreductase 1 enzymes. In conclusion, the current study provides evidence for the protective effect of telmisartan and nifedipine in cuprizone-induced demyelination and behavioral dysfunction in mice possibly by modulating NF-κB and Nrf2 signaling pathways.


Cuprizone/toxicity , Demyelinating Diseases/prevention & control , NF-E2-Related Factor 2/agonists , NF-kappa B/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Nifedipine/therapeutic use , Telmisartan/therapeutic use , Animals , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Hand Strength/physiology , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/biosynthesis , NF-kappa B/biosynthesis , Neuroprotective Agents/pharmacology , Nifedipine/pharmacology , Telmisartan/pharmacology
...