Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.282
Filter
1.
Cells ; 13(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39120303

ABSTRACT

Human NAD(P)H-quinone oxidoreductase1 (HNQO1) is a two-electron reductase antioxidant enzyme whose expression is driven by the NRF2 transcription factor highly active in the prooxidant milieu found in human malignancies. The resulting abundance of NQO1 expression (up to 200-fold) in cancers and a barely detectable expression in body tissues makes it a selective marker of neoplasms. NQO1 can catalyze the repeated futile redox cycling of certain natural and synthetic quinones to their hydroxyquinones, consuming NADPH and generating rapid bursts of cytotoxic reactive oxygen species (ROS) and H2O2. A greater level of this quinone bioactivation due to elevated NQO1 content has been recognized as a tumor-specific therapeutic strategy, which, however, has not been clinically exploited. We review here the natural and new quinones activated by NQO1, the catalytic inhibitors, and the ensuing cell death mechanisms. Further, the cancer-selective expression of NQO1 has opened excellent opportunities for distinguishing cancer cells/tissues from their normal counterparts. Given this diagnostic, prognostic, and therapeutic importance, we and others have engineered a large number of specific NQO1 turn-on small molecule probes that remain latent but release intense fluorescence groups at near-infrared and other wavelengths, following enzymatic cleavage in cancer cells and tumor masses. This sensitive visualization/quantitation and powerful imaging technology based on NQO1 expression offers promise for guided cancer surgery, and the reagents suggest a theranostic potential for NQO1-targeted chemotherapy.


Subject(s)
NAD(P)H Dehydrogenase (Quinone) , Neoplasms , Humans , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Neoplasms/pathology , Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Quinones/pharmacology , Quinones/metabolism , Molecular Targeted Therapy
2.
Nutrients ; 16(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999824

ABSTRACT

Parkinson's disease (PD) is a degenerative neurological disorder defined by the deterioration and loss of dopamine-producing neurons in the substantia nigra, leading to a range of motor impairments and non-motor symptoms. The underlying mechanism of this neurodegeneration remains unclear. This research examined the neuroprotective properties of Ecklonia cava polyphenols (ECPs) in mitigating neuronal damage induced by rotenone via the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway. Using human neuroblastoma SH-SY5Y cells and PD model mice, we found that ECP, rich in the antioxidant polyphenol phlorotannin, boosted the gene expression and functionality of the antioxidant enzyme NAD(P)H quinone oxidoreductase-1. ECP also promoted Nrf2 nuclear translocation and increased p62 expression, suggesting that p62 helps sustain Nrf2 activation via a positive feedback loop. The neuroprotective effect of ECP was significantly reduced by Compound C (CC), an AMP-activated protein kinase (AMPK) inhibitor, which also suppressed Nrf2 nuclear translocation. In PD model mice, ECPs improved motor functions impaired by rotenone, as assessed by the pole test and wire-hanging test, and restored intestinal motor function and colon tissue morphology. Additionally, ECPs increased tyrosine hydroxylase expression in the substantia nigra, indicating a protective effect on dopaminergic neurons. These findings suggest that ECP has a preventative effect on PD.


Subject(s)
NF-E2-Related Factor 2 , Neuroprotective Agents , Parkinson Disease , Polyphenols , Rotenone , Animals , Humans , Male , Mice , Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Cell Line, Tumor , Disease Models, Animal , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents/pharmacology , NF-E2-Related Factor 2/metabolism , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , Parkinson Disease/drug therapy , Polyphenols/pharmacology , Signal Transduction/drug effects , Kelp/chemistry
3.
Nutr Res ; 128: 14-23, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002358

ABSTRACT

Sesamolin, a lignan isolated from sesame oils, has been found to possess neuroprotective, anticancer, and free radical scavenging properties. We hypothesized that sesamolin could stimulate the activity of nuclear factor erythroid-derived 2-like 2 (Nrf2) and inhibit adipocyte differentiation of preadipocytes. The objective of this study was to investigate effects of sesamolin on adipocyte differentiation and its underlying molecular mechanisms. In this study, we determined the effects of treatment with 25 to 100 µM sesamolin on adipogenesis in cell culture systems. Sesamolin inhibited lipid accumulation and suppressed the expression of adipocyte markers during adipocyte differentiation of C3H10T1/2, 3T3-L1, and primary preadipocytes. Mechanism studies revealed that sesamolin increased Nrf2 protein expression without inducing its mRNA, leading to an increase in the expression of Nrf2 target genes such as heme oxygenase 1 and NAD(P)H:quinone oxidoreductase 1 (Nqo1) in C3H10T1/2 adipocytes and mouse embryonic fibroblasts. These effects were significantly attenuated in Nrf2 knockout (KO) mouse embryonic fibroblasts, indicating that effects of sesamolin were dependent on Nrf2. In H1299 human lung cancer cells with KO of Kelch like-ECH-associated protein 1 (Keap1), a negative regulator of Nrf2, sesamolin failed to further increase Nrf2 protein expression. However, upon reexpressing Keap1 in Keap1 KO cells, the ability of sesamolin to elevate Nrf2 protein expression was restored, highlighting the crucial role of Keap1 in sesamolin-induced Nrf2 activation. Taken together, these findings show that sesamolin can inhibit adipocyte differentiation through Keap1-mediated Nrf2 activation.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Kelch-Like ECH-Associated Protein 1 , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Animals , Mice , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Cell Differentiation/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Dioxoles/pharmacology , Mice, Knockout , Lignans/pharmacology , Humans , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics
4.
ACS Appl Mater Interfaces ; 16(30): 39021-39034, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39033517

ABSTRACT

Chemodynamic therapy (CDT), employing metal ions to transform endogenous H2O2 into lethal hydroxyl radicals (•OH), has emerged as an effective approach for tumor treatment. Yet, its efficacy is diminished by glutathione (GSH), commonly overexpressed in tumors. Herein, a breakthrough strategy involving extracellular vesicle (EV) mimetic nanovesicles (NVs) encapsulating iron oxide nanoparticles (IONPs) and ß-Lapachone (Lapa) was developed to amplify intracellular oxidative stress. The combination, NV-IONP-Lapa, created through a serial extrusion from ovarian epithelial cells showed excellent biocompatibility and leveraged magnetic guidance to enhance endocytosis in ovarian cancer cells, resulting in selective H2O2 generation through Lapa catalysis by NADPH quinone oxidoreductase 1 (NQO1). Meanwhile, the iron released from IONPs ionization under acidic conditions triggered the conversion of H2O2 into •OH by the Fenton reaction. Additionally, the catalysis process of Lapa eliminated GSH in tumor, further amplifying oxidative stress. The designed NV-IONP-Lapa demonstrated exceptional tumor targeting, facilitating MR imaging, and enhanced tumor suppression without significant side effects. This study presents a promising NV-based drug delivery system for exploiting CDT against NQO1-overexpressing tumors by augmenting intratumoral oxidative stress.


Subject(s)
Naphthoquinones , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Cell Line, Tumor , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Hydrogen Peroxide/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry , Oxidative Stress/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glutathione/metabolism , Glutathione/chemistry , Drug Delivery Systems
5.
J Phys Chem Lett ; 15(30): 7681-7693, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39038219

ABSTRACT

Accurate prediction of Drug-Target Interactions (DTI) is crucial for drug development. Current state-of-the-art deep learning methods have significantly advanced the field; however, these methods exhibit limitations in predictive performance and the propensity for false negatives. Therefore, we propose EADTN, a simple and efficient ensemble model. We have designed an innovative feature adaptation technique to automatically extract local weights of drugs and targets, and we utilize clustering-enhanced parameter fine-tuning to overcome the issue of false negatives, thereby enhancing its reliability in drug discovery. Based on EADTN, we also propose a Shapley value-based method for identifying key drug substructures, effectively enhancing the model's interpretability. Additionally, we utilized EADTN to reveal potential interactions between NQO1 targets and the drugs SIRT-IN-1 and LY2183240, which were subsequently validated through wet-lab experiments. Experimental evidence demonstrates that EADTN consistently outperforms existing best-performing models across various data sets, promising significant benefits in fields such as drug repositioning.


Subject(s)
Deep Learning , NAD(P)H Dehydrogenase (Quinone)/metabolism , Sirtuin 1/metabolism , Drug Discovery , Humans
6.
J Pathol Clin Res ; 10(4): e12390, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38992928

ABSTRACT

Oxidative stress and the immune microenvironment both contribute to the pathogenesis of esophageal squamous cell carcinoma (ESCC). However, their interrelationships remain poorly understood. We aimed to examine the status of key molecules involved in oxidative stress and the immune microenvironment, as well as their relationships with each other and with clinicopathological features and prognosis in ESCC. The expression of programmed death-ligand 1 (PD-L1), CD8, nuclear factor erythroid-2 related factor-2 (NRF2), and NAD(P)H quinone oxidoreductase 1 (NQO1) was detected using immunohistochemistry in tissue samples from 176 patients with ESCC. We employed both combined positive score (CPS) and tumor proportion score (TPS) to evaluate PD-L1 expression and found a positive correlation between CPS and TPS. Notably, PD-L1 expression, as assessed by either CPS or TPS, was positively correlated with both NRF2 nuclear score and NQO1 score in stage II-IV ESCC. We also observed a positive correlation between the density of CD8+ T cells and PD-L1 expression. Furthermore, high levels of PD-L1 CPS, but not TPS, were associated with advanced TNM stage and lymph node metastases. Moreover, both PD-L1 CPS and the nuclear expression of NRF2 were found to be predictive of shorter overall survival in stage II-IV ESCC. By using the Mandard-tumor regression grading (TRG) system to evaluate the pathological response of tumors to neoadjuvant chemotherapy (NACT), we found that the TRG-5 group had higher NRF2 nuclear score, PD-L1 CPS, and TPS in pre-NACT biopsy samples compared with the TRG-3 + 4 group. The NQO1 scores of post-NACT surgical specimens were significantly higher in the TRG-5 group than in the TRG 3 + 4 group. In conclusion, the expression of PD-L1 is associated with aberrant NRF2 signaling pathway, advanced TNM stage, lymph node metastases, and unfavorable prognosis. The dysregulation of PD-L1 and aberrant activation of the NRF2 signaling pathway are implicated in resistance to NACT. Our findings shed light on the complex interrelationships between oxidative stress and the immune microenvironment in ESCC, which may have implications for personalized therapies and improved patient outcomes.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Oxidative Stress , Tumor Microenvironment , Humans , NF-E2-Related Factor 2/metabolism , B7-H1 Antigen/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Male , Female , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , Middle Aged , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Adult , Neoplasm Staging , Lymphocytes, Tumor-Infiltrating/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Immunohistochemistry
7.
Anal Chem ; 96(32): 13308-13316, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39078110

ABSTRACT

NAD(P)H: quinone oxidoreductase-1 (NQO1) plays critical roles in antioxidation and abnormally overexpresses in tumors. Developing a fast and sensitive method of monitoring NQO1 will greatly promote cancer diagnosis in clinical practice. This study introduces a transformative colorimetric detection strategy for NQO1, harnessing an innovative competitive substrate mechanism between NQO1 and a new NADH oxidase (NOX) mimic, cobalt-nitrogen-doped carbon nanozyme (CoNC). This method ingeniously exploits the differential consumption of NADH in the presence of NQO1 to modulate the generation of H2O2 from CoNC catalysis, which is then quantified through a secondary, peroxidase-mimetic cascade reaction involving Prussian blue (PB) nanoparticles. This dual-stage reaction framework not only enhances the sensitivity of NQO1 detection, achieving a limit of detection as low as 0.67 µg mL-1, but also enables the differentiation between cancerous and noncancerous cells by their enzymatic activity profiles. Moreover, CoNC exhibits exceptional catalytic efficiency, with a specific activity reaching 5.2 U mg-1, significantly outperforming existing NOX mimics. Beyond mere detection, CoNC serves a dual role, acting as both a robust mimic of cytochrome c reductase (Cyt c) and a cornerstone for enzymatic regeneration, thereby broadening the scope of its biological applications. This study not only marks a significant step forward in the bioanalytical application of nanozymes but also sets the stage for their expanded use in clinical diagnostics and therapeutic monitoring.


Subject(s)
Colorimetry , NAD(P)H Dehydrogenase (Quinone) , NADH, NADPH Oxidoreductases , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/chemistry , Humans , NADH, NADPH Oxidoreductases/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Multienzyme Complexes/metabolism , Multienzyme Complexes/chemistry , Cobalt/chemistry , Carbon/chemistry , Biomimetics , Limit of Detection , Nitrogen/chemistry , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Ferrocyanides/chemistry , NAD/metabolism , NAD/chemistry
8.
Phytomedicine ; 132: 155876, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032284

ABSTRACT

BACKGROUND: Ferroptosis, an emerging nonapoptotic, modulated cell death process characterized by iron accumulation and subsequent lipid peroxidation, has been intimately implicated in the development and progression of ovarian cancer (OC). Daphnetin (Daph), a natural product isolated from Daphne Korean Nakai, exhibits anticancer efficacy against various solid tumors. However, the specific role and potential mechanism underlying Daph-mediated modulation of ferroptosis in OC cells remain elusive. PURPOSE: This study aims to analyze the proferroptotic impacts of Daph on OC cells and to further explore the underlying mechanisms involved. STUDY DESIGN AND METHODS: We used CCK-8, wound healing and Transwell assays to assess whether Daph can inhibit the proliferation and migration of OC cells. Additionally, transmission electron microscopy (TEM), iron measurement, reactive oxygen species (ROS) analysis, lipid peroxidation assays, qRT-PCR and western blotting were utilized to evaluate the impact of Daph on ferroptosis and elucidate the potential underlying mechanism. Furthermore, RNA sequencing analysis, molecular docking analysis, cellular thermal shift assays (CETSAs) and NQO1 activity assays were used to predict and validate the binding and mechanistic interactions between Daph and NQO1. Subcutaneous tumorigenesis models were utilized to examine the effectiveness of Daph (and/or cisplatin) in vivo. RESULTS: Daph exerted antitumor effects by inducing the death and suppressing the migration of A2780 and SKOV3 cells. Further, Daph induced ferroptosis in OC cells, as evidenced by the accumulation of intracellular ferrous iron (Fe2+), ROS and lipid peroxides, as well as the decreases in the glutathione/oxidized glutathione disulfide (GSH/GSSG) ratio and the expression of ferroptosis indicators (SLC7A11 and GPX4). RNA sequencing and molecular docking analyses revealed that the direct interaction between NQO1 and Daph reduced both the activity and expression of NQO1. Importantly, NQO1 overexpression effectively alleviated the effects of Daph on proliferation, migration, and ferroptosis in vitro and in vivo. Interestingly, we also found that combination treatment with Daph, a negative regulator of NQO1, and cisplatin synergistically induced cytotoxicity in OC cells. CONCLUSION: Our findings are the firstly demonstrated that Daph acts as a novel ferroptosis inducer in OC cells by specifically targeting NQO1 and is thus a promising candidate agent for OC treatment.


Subject(s)
Cell Proliferation , Ferroptosis , Molecular Docking Simulation , NAD(P)H Dehydrogenase (Quinone) , Ovarian Neoplasms , Reactive Oxygen Species , Umbelliferones , Ferroptosis/drug effects , Ovarian Neoplasms/drug therapy , Female , Humans , Cell Line, Tumor , Animals , Reactive Oxygen Species/metabolism , Umbelliferones/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Cisplatin/pharmacology , Mice , Lipid Peroxidation/drug effects , Mice, Nude , Mice, Inbred BALB C , Daphne/chemistry , Antineoplastic Agents, Phytogenic/pharmacology
9.
Kaohsiung J Med Sci ; 40(8): 710-721, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837857

ABSTRACT

The proinflammatory properties of high-mobility group box protein 1 (HMGB1) in sepsis have been extensively studied. This study aimed to investigate the impact of HMGB1 on ferroptosis and its molecular mechanism in sepsis-induced acute lung injury (ALI). A septic mouse model was established using the cecal ligation and puncture method. Blocking HMGB1 resulted in improved survival rates, reduced lung injury, decreased levels of ferroptosis markers (reactive oxygen species, malondialdehyde, and Fe2+), and enhanced antioxidant enzyme activities (superoxide dismutase and catalase) in septic mice. In addition, knockdown of HMGB1 reduced cellular permeability, ferroptosis markers, and raised antioxidant enzyme levels in lipopolysaccharide (LPS)-stimulated MLE-12 cells. Silencing of HMGB1 led to elevations in the expressions of ferroptosis core-regulators in LPS-treated MLE-12 cells, such as solute carrier family 7 member 11 (SLC7A11), solute carrier family 3 member A2 (SLC3A2), and glutathione peroxidase 4. Furthermore, blocking HMGB1 did not alter ferroptosis, oxidative stress-related changes, and permeability in LPS-treated MLE-12 cells that were pretreated with ferrostatin-1 (a ferroptosis inhibitor). HMGB1 inhibition also led to elevated expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream targets, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in LPS-treated MLE-12 cells and lung tissues from septic mice. The Nrf2-specific inhibitor ML385 reversed the effects of HMGB1 silencing on ferroptosis and cell permeability in LPS-treated MLE-12 cells. Our findings indicated that the inhibition of HMGB1 restrains ferroptosis and oxidative stress, thereby alleviating sepsis-induced ALI through the activation of Nrf2 signaling.


Subject(s)
Acute Lung Injury , Ferroptosis , HMGB1 Protein , Lipopolysaccharides , NF-E2-Related Factor 2 , Oxidative Stress , Sepsis , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , HMGB1 Protein/metabolism , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Acute Lung Injury/drug therapy , Ferroptosis/drug effects , Sepsis/complications , Sepsis/metabolism , Sepsis/drug therapy , Oxidative Stress/drug effects , Mice , Lipopolysaccharides/toxicity , Signal Transduction/drug effects , Male , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Cyclohexylamines/pharmacology , Cell Line , Disease Models, Animal , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Amino Acid Transport System y+
10.
Oncoimmunology ; 13(1): 2363000, 2024.
Article in English | MEDLINE | ID: mdl-38846085

ABSTRACT

NAD(P)H:quinone oxidoreductase 1 (NQO1) is overexpressed in most solid cancers, emerging as a promising target for tumor-selective killing. ß-Lapachone (ß-Lap), an NQO1 bioactivatable drug, exhibits significant antitumor effects on NQO1-positive cancer cells by inducing immunogenic cell death (ICD) and enhancing tumor immunogenicity. However, the interaction between ß-Lap-mediated antitumor immune responses and neutrophils, novel antigen-presenting cells (APCs), remains unknown. This study demonstrates that ß-Lap selectively kills NQO1-positive murine tumor cells by significantly increasing intracellular ROS formation and inducing DNA double strand breaks (DSBs), resulting in DNA damage. Treatment with ß-Lap efficiently eradicates immunocompetent murine tumors and significantly increases the infiltration of tumor-associated neutrophils (TANs) into the tumor microenvironment (TME), which plays a crucial role in the drug's therapeutic efficacy. Further, the presence of ß-Lap-induced antigen medium leads bone marrow-derived neutrophils (BMNs) to directly kill murine tumor cells, aiding in dendritic cells (DCs) recruitment and significantly enhancing CD8+ T cell proliferation. ß-Lap treatment also drives the polarization of TANs toward an antitumor N1 phenotype, characterized by elevated IFN-ß expression and reduced TGF-ß cytokine expression, along with increased CD95 and CD54 surface markers. ß-Lap treatment also induces N1 TAN-mediated T cell cross-priming. The HMGB1/TLR4/MyD88 signaling cascade influences neutrophil infiltration into ß-Lap-treated tumors. Blocking this cascade or depleting neutrophil infiltration abolishes the antigen-specific T cell response induced by ß-Lap treatment. Overall, this study provides comprehensive insights into the role of tumor-infiltrating neutrophils in the ß-Lap-induced antitumor activity against NQO1-positive murine tumors.


Subject(s)
NAD(P)H Dehydrogenase (Quinone) , Naphthoquinones , Neutrophils , Tumor Microenvironment , Animals , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/immunology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Cell Line, Tumor , Neutrophil Infiltration/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Female , Phenotype
11.
Chem Res Toxicol ; 37(6): 1044-1052, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38833663

ABSTRACT

The aim of the present study was to evaluate the cardiotoxic effects of alcohol and its potential toxic mechanism on ferroptosis in mice and H9c2 cells. Mice were intragastrically treated with three different concentrations of alcohol, 7, 14, and 28%, each day for 14 days. Body weight and electrocardiography (ECG) were recorded over the 14 day period. Serum creatine kinase (CK), lactic dehydrogenase (LDH), MDA, tissue iron, and GSH levels were measured. Cardiac tissues were examined histologically, and ferroptosis was assessed. In H9c2 cardiomyocytes, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. The proteins of ferroptosis were evaluated by the western blot technique in vivo and in vitro. The results showed that serum CK, LDH, MDA, and tissue iron levels significantly increased in the alcohol treatment group in a dose-dependent manner. The content of GSH decreased after alcohol treatment. ECG and histological examinations showed that alcohol impaired cardiac function and structure. In addition, the levels of ROS and LIP increased, and MMP levels decreased after alcohol treatment. Ferrostatin-1 (Fer-1) protected cells from lipid peroxidation. Western blotting analysis showed that alcohol downregulated the expression of Nrf2, NQO1, HO-1, and GPX4. The expressions of P53 and TfR were upregulated in vivo and in vitro. Fer-1 significantly alleviated alcohol-induced ferroptosis. In conclusion, the study showed that Nrf2/NQO1-dependent ferroptosis played a vital role in the cardiotoxicity induced by alcohol.


Subject(s)
Cardiotoxicity , Ethanol , Ferroptosis , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Animals , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Mice , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Male , Reactive Oxygen Species/metabolism , Rats , Mice, Inbred C57BL , Cell Survival/drug effects
12.
J Appl Toxicol ; 44(9): 1454-1465, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38812125

ABSTRACT

Ochratoxin A (OTA) is a type of mycotoxin commonly found in raw and processed foods. It is essential to be aware of this toxin, as it can harm your health if consumed in high quantities. OTA can induce toxic effects in various cell models. However, a more comprehensive understanding of the harmful effects of OTA on human astrocytes is required. This study evaluated OTA's neurotoxic effects on the Gibco® Human Astrocyte (GHA) cell line, its underlying mechanisms, and the antioxidant N-acetylcysteine (NAC) ability to prevent them. OTA exposure within 5-30 µM has induced concentration-dependent cytotoxicity. In the OTA-treated cells, the levels of reactive oxygen species (ROS) were found to be significantly increased, while the glutathione (GSH) contents were found to decrease considerably. The western blotting of OTA-treated cells has revealed increased Bax, cleaved caspase-9/caspase-3 protein levels, and increased Bax/Bcl-2 ratio. In addition, exposure to OTA has resulted in the induction of antioxidant responses associated with the protein expressions of Nrf2, HO-1, and NQO1. On the other hand, the pretreatment with NAC has partially alleviated the significant toxic effects of OTA. In conclusion, our findings suggest that oxidative stress and apoptosis are involved in the OTA-induced cytotoxicity in GHA cells. NAC could act as a protective agent against OTA-induced oxidative damage.


Subject(s)
Acetylcysteine , Apoptosis , Astrocytes , Glutathione , Ochratoxins , Oxidative Stress , Reactive Oxygen Species , Ochratoxins/toxicity , Humans , Astrocytes/drug effects , Acetylcysteine/pharmacology , Reactive Oxygen Species/metabolism , Cell Line , Apoptosis/drug effects , Oxidative Stress/drug effects , Glutathione/metabolism , Cell Survival/drug effects , Antioxidants/pharmacology , NF-E2-Related Factor 2/metabolism , Caspase 3/metabolism , Heme Oxygenase-1/metabolism , bcl-2-Associated X Protein/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism
13.
Free Radic Biol Med ; 221: 181-187, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38772511

ABSTRACT

Sulforaphane (SFN), found in cruciferous vegetables, is a known activator of NRF2 (master regulator of cellular antioxidant responses). Patients with chronic kidney disease (CKD) present an imbalance in the redox state, presenting reduced expression of NRF2 and increased expression of NF-κB. Therefore, this study aimed to evaluate the effects of SFN on the mRNA expression of NRF2, NF-κB and markers of oxidative stress in patients with CKD. Here, we observed a significant increase in the mRNA expression of NRF2 (p = 0.02) and NQO1 (p = 0.04) in the group that received 400 µg/day of SFN for 1 month. Furthermore, we observed an improvement in the levels of phosphate (p = 0.02), glucose (p = 0.05) and triglycerides (p = 0.02) also in this group. On the other hand, plasma levels of LDL-c (p = 0.04) and total cholesterol (p = 0.03) increased in the placebo group during the study period. In conclusion, 400 µg/day of SFN for one month improves the antioxidant system and serum glucose and phosphate levels in non-dialysis CKD patients.


Subject(s)
Isothiocyanates , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Oxidative Stress , RNA, Messenger , Renal Insufficiency, Chronic , Sulfoxides , Humans , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Male , Middle Aged , Female , RNA, Messenger/genetics , RNA, Messenger/metabolism , Oxidative Stress/drug effects , Antioxidants/metabolism , Antioxidants/pharmacology , Triglycerides/blood , Triglycerides/metabolism , Blood Glucose/metabolism , Up-Regulation/drug effects , Adult , Aged , NF-kappa B/metabolism , NF-kappa B/genetics
14.
Zhongguo Zhong Yao Za Zhi ; 49(9): 2316-2325, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812132

ABSTRACT

This study aimed to investigate the intervention effect of tetramethylpyrazine(TMP) combined with transplantation of neural stem cells(NSCs) on middle cerebral artery occlusion(MCAO) rat model and to explore the mechanism of TMP combined with NSCs transplantation on ischemic stroke based on the regulation of stem cell biological behavior. MCAO rats were randomly divided into a model group, a TMP group, an NSCs transplantation group, and a TMP combined with NSCs transplantation group according to neurological function scores. A sham group was set up at the same time. The neurological function score was used to evaluate the improvement of neurological function in MCAO rats after TMP combined with NSCs transplantation. The proliferation, migration, and differentiation of NSCs were evaluated by BrdU, BrdU/DCX, BrdU/NeuN, and BrdU/GFAP immunofluorescence labeling. The protein expression of stromal cell-derived factor 1(SDF-1), C-X-C motif chemokine receptor 4(CXCR4), as well as oxidative stress pathway proteins nuclear factor erythroid 2-related factor 2(Nrf2), Kelch-like ECH-associated protein 1(KEAP1), heme oxygenase 1(HO-1), NAD(P)H quinone oxidoreductase 1(NQO1) was detected by Western blot to study the migration mechanism of TMP combined with NSCs. The results showed that TMP combined with NSCs transplantation significantly improved the neurological function score in MCAO rats. Immunofluorescence staining showed a significant increase in the number of BrdU~+, BrdU~+/DCX~+, BrdU~+/NeuN~+, and BrdU~+/GFAP~+ cells in the TMP, NSCs transplantation, and combined treatment groups, with the combined treatment group showing the most significant increase. Further Western blot analysis revealed significantly elevated expression of CXCR4 protein in the TMP, NSCs transplantation, and combined treatment groups, along with up-regulated protein expression of Nrf2, HO-1, and NQO1, and decreased KEAP1 protein expression. This study showed that both TMP and NSCs transplantation can promote the recovery of neurological function by promoting the proliferation, migration, and differentiation of NSCs, and the effect of TMP combined with NSCs transplantation is superior. The mechanism of action may be related to the activation of the Nrf2/HO-1/CXCR4 pathway.


Subject(s)
Brain Ischemia , Doublecortin Protein , NF-E2-Related Factor 2 , Neural Stem Cells , Pyrazines , Rats, Sprague-Dawley , Receptors, CXCR4 , Animals , Pyrazines/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Neural Stem Cells/metabolism , Rats , Male , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Brain Ischemia/therapy , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Stem Cell Transplantation/methods , Cell Proliferation/drug effects , Cell Movement/drug effects , Humans , Reperfusion Injury/therapy , Reperfusion Injury/metabolism , Infarction, Middle Cerebral Artery/therapy , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics
15.
Life Sci ; 349: 122714, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38735366

ABSTRACT

AIMS: Non-alcoholic fatty liver disease (NAFLD) has risen as a significant global public health issue, for which vertical sleeve gastrectomy (VSG) has become an effective treatment method. The study sought to elucidate the processes through which PIM1 mitigates the advancement of NAFLD. The Pro-viral integration site for Moloney murine leukemia virus 1 (PIM1) functions as a serine/threonine kinase. Bioinformatics analysis revealed that reduced PIM1 expression in NAFLD. METHODS: To further prove the role of PIM1 in NAFLD, an in-depth in vivo experiment was performed, in which male C57BL/6 mice were randomly grouped to receive a normal or high-fat diet for 24 weeks. They were operated or delivered the loaded adeno-associated virus which the PIM1 was overexpressed (AAV-PIM1). In an in vitro experiment, AML12 cells were treated with palmitic acid to induce hepatic steatosis. KEY FINDINGS: The results revealed that the VSG surgery and virus delivery of mice alleviated oxidative stress, and apoptosis in vivo. For AML12 cells, the levels of oxidative stress, apoptosis, and lipid metabolism were reduced via PIM1 upregulation. Moreover, ML385 treatment resulted in the downregulation of the NRF2/HO-1/NQO1 signaling cascade, indicating that PIM1 mitigates NAFLD by targeting this pathway. SIGNIFICANCE: PIM1 alleviated mice liver oxidative stress and NAFLD induced by high-fat diet by regulating the NRF2/HO-1/NQO1 signaling Pathway.


Subject(s)
Heme Oxygenase-1 , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Non-alcoholic Fatty Liver Disease , Oxidative Stress , Proto-Oncogene Proteins c-pim-1 , Animals , Proto-Oncogene Proteins c-pim-1/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Male , Mice , NF-E2-Related Factor 2/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Heme Oxygenase-1/metabolism , Diet, High-Fat/adverse effects , Liver/metabolism , Liver/pathology , Signal Transduction , Apoptosis , Membrane Proteins/metabolism , Membrane Proteins/genetics
16.
Sci Rep ; 14(1): 10696, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38730068

ABSTRACT

COVID-19, caused by SARS-CoV-2, affects neuronal cells, causing several symptoms such as memory loss, anosmia and brain inflammation. Curcuminoids (Me08 e Me23) and curcumin (CUR) are derived from Curcuma Longa extract (EXT). Many therapeutic actions have been linked to these compounds, including antiviral action. Given the severe implications of COVID-19, especially within the central nervous system, our study aims to shed light on the therapeutic potential of curcuminoids against SARS-CoV-2 infection, particularly in neuronal cells. Here, we investigated the effects of CUR, EXT, Me08 and Me23 in human neuroblastoma SH-SY5Y. We observed that Me23 significantly decreased the expression of plasma membrane-associated transmembrane protease serine 2 (TMPRSS2) and TMPRSS11D, consequently mitigating the elevated ROS levels induced by SARS-CoV-2. Furthermore, Me23 exhibited antioxidative properties by increasing NRF2 gene expression and restoring NQO1 activity following SARS-CoV-2 infection. Both Me08 and Me23 effectively reduced SARS-CoV-2 replication in SH-SY5Y cells overexpressing ACE2 (SH-ACE2). Additionally, all of these compounds demonstrated the ability to decrease proinflammatory cytokines such as IL-6, TNF-α, and IL-17, while Me08 specifically reduced INF-γ levels. Our findings suggest that curcuminoid Me23 could serve as a potential agent for mitigating the impact of COVID-19, particularly within the context of central nervous system involvement.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Antiviral Agents , COVID-19 Drug Treatment , Curcumin , SARS-CoV-2 , Humans , Curcumin/pharmacology , Curcumin/analogs & derivatives , Antioxidants/pharmacology , Antiviral Agents/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Curcuma/chemistry , Serine Endopeptidases/metabolism , COVID-19/virology , COVID-19/metabolism , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Cytokines/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/virology
17.
Redox Rep ; 29(1): 2332038, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38563333

ABSTRACT

OBJECTIVES: Gentamicin is one of the most common ototoxic drugs that can lower patients' quality of life. Oxidative stress is a key factors inducing sensory hair cell death during gentamicin administration. So far, there are no effective drugs to prevent or treat gentamicin- induced hearing loss. A recent study found cystic fibrosis transmembrane conductance regulator (CFTR) as a new target to modulate cellular oxidative balance. The objective of this study was to estimate the effect of the CFTR activator ivacaftor on gentamicin-induced ototoxicity and determine its mechanism. METHODS: The hair cell count was analyzed by Myosin 7a staining. Apoptosis was analyzed by TUNEL Apoptosis Kit. Cellular reactive oxygen species (ROS) level was detected by DCFH-DA probes. The Nrf2 related proteins expression levels were analyzed by western blot. RESULTS: An in vitro cochlear explant model showed that gentamicin caused ROS accumulation in sensory hair cells and induced apoptosis, and this effect was alleviated by pretreatment with ivacaftor. Western blotting showed that ivacaftor administration markedly increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO1), and NAD(P)H:quinone oxidoreductase 1 (NQO1). The protective effect of ivacaftor was abolished by the Nrf2 inhibitor ML385. DISCUSSION: Our results indicate the protective role of the CFTR-Nrf2-HO1/NQO1 pathway in gentamicin-induced ototoxicity. Ivacaftor may be repositioned or repurposed towards aminoglycosides-induced hearing loss.


Subject(s)
Aminophenols , Hearing Loss , Ototoxicity , Quinolones , Humans , Gentamicins/toxicity , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/pharmacology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/pharmacology , Quality of Life , Oxidative Stress , Apoptosis , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/pharmacology
18.
J Pharm Pharmacol ; 76(7): 851-860, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38625054

ABSTRACT

OBJECTIVES: The study aimed to investigate the protective effects of dexmedetomidine (DEX) on renal injury caused by acute stress in rats and explore the protective pathways of DEX on rat kidneys in terms of oxidative stress. METHODS: An acute restraint stress model was utilized, where rats were restrained for 3 hours after a 15-minute swim. Biochemical tests and histopathological sections were conducted to evaluate renal function, along with the measurement of oxidative stress and related pathway proteins. KEY FINDINGS: The open-field experiments validated the successful establishment of the acute stress model. Acute stress-induced renal injury led to increased NADPH oxidase 4 (NOX4) protein expression and decreased expression levels of nuclear transcription factor 2 (Nrf2), heme oxygenase-1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1). Following DEX treatment, there was a significant reduction in renal NOX4 expression. The DEX-treated group exhibited normalized renal biochemical results and less damage observed in pathological sections compared to the acute stress group. CONCLUSIONS: The findings suggest that DEX treatment during acute stress can impact the NOX4/Nrf2/HO-1/NQO1 signaling pathway and inhibit oxidative stress, thereby preventing acute stress-induced kidney injury. Additionally, DEX shows promise for clinical applications in stress syndromes.


Subject(s)
Antioxidants , Dexmedetomidine , Kidney , NAD(P)H Dehydrogenase (Quinone) , NADPH Oxidase 4 , NF-E2-Related Factor 2 , Oxidative Stress , Rats, Sprague-Dawley , Signal Transduction , Animals , NADPH Oxidase 4/metabolism , NF-E2-Related Factor 2/metabolism , Dexmedetomidine/pharmacology , Oxidative Stress/drug effects , Male , Antioxidants/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Rats , Signal Transduction/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Heme Oxygenase-1/metabolism , Disease Models, Animal , Heme Oxygenase (Decyclizing)
19.
Eur J Pharmacol ; 973: 176511, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38604545

ABSTRACT

Lung cancer is one of the most lethal cancers with high incidence worldwide. The prevention of lung cancer is of great significance to reducing the social harm caused by this disease. An in-depth understanding of the molecular changes underlying precancerous lesions is essential for the targeted chemoprevention against lung cancer. Here, we discovered an increased NQO1 level over time within pulmonary premalignant lesions in both the KrasG12D-driven and nicotine-derived nitrosamine ketone (NNK)-induced mouse models of lung cancer, as well as in KrasG12D-driven and NNK-induced malignant transformed human bronchial epithelial cells (BEAS-2B and 16HBE). This suggests a potential correlation between the NQO1 expression and lung carcinogenesis. Based on this finding, we utilized ß-Lapachone (ß-Lap), an NQO1 bioactivatable drug, to suppress lung tumorigenesis. In this study, the efficacy and safety of low-dose ß-Lap were demonstrated in preventing lung tumorigenesis in vivo. In conclusion, our study suggests that long-term consumption of low-dose ß-Lap could potentially be an effective therapeutic strategy for the prevention of lung premalignant lesions. However, further studies and clinical trials are necessary to validate our findings, determine the safety of long-term ß-Lap usage in humans, and promote the use of ß-Lap in high-risk populations.


Subject(s)
Lung Neoplasms , NAD(P)H Dehydrogenase (Quinone) , Naphthoquinones , Animals , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Humans , Mice , Carcinogenesis/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Female , Cell Line
20.
Biosci Trends ; 18(2): 153-164, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38599881

ABSTRACT

NAD(P)H-quinone oxidoreductase 1 (NQO1) is an essential redox enzyme responsible for redox balance and energy metabolism. Despite of its importance, the brain contains high capacity of polyunsaturated fatty acids and maintains low levels of NQO1 expression. In this study, we examined how levels of NQO1 expression affects cell survival in response to toxic insults causing mitochondrial dysfunction and ferroptosis, and whether NQO1 has a potential as a biomarker in different stressed conditions. Following treatment with rotenone, overexpressed NQO1 in SH-SY5Y cells improved cell survival by reducing mitochondrial reductive stress via increased NAD+ supply without mitochondrial biogenesis. However, NQO1 overexpression boosted lipid peroxidation following treatment with RSL3 and erastin. A lipid droplet staining assay showed increased lipid droplets in cells overexpressing NQO1. In contrast, NQO1 knockdown protected cells against ferroptosis by increasing GPX4, xCT, and the GSH/GSSG system. Also, NQO1 knockdown showed lower iron contents and lipid droplets than non-transfectants and cells overexpressing NQO1, even though it could not attenuate cell death when exposed to rotenone. In summary, our study suggests that different NQO1 levels may have advantages and disadvantages depending on the surrounding environments. Thus, regulating NQO1 expression could be a potential supplementary tool when treating neuronal diseases.


Subject(s)
Ferroptosis , Mitochondria , NAD(P)H Dehydrogenase (Quinone) , Rotenone , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Ferroptosis/drug effects , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Rotenone/toxicity , Rotenone/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Lipid Peroxidation/drug effects , Piperazines/pharmacology , Carbolines
SELECTION OF CITATIONS
SEARCH DETAIL