Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 392
Filter
1.
Int J Mol Sci ; 25(1)2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38203785

ABSTRACT

Propolis is a natural resin produced by honeybees with plenty of pharmacologic properties, including antioxidant activity. Oxidative stress disrupts germ cell development and sperm function, with demonstrated harmful effects on male reproduction. Several natural antioxidants have been shown to reduce oxidative damage and increase sperm fertility potential; however, little is known about the effects of propolis. This work evaluated the role of propolis in protecting spermatogonial cells from oxidative damage. Propolis' phytochemical composition and antioxidant potential were determined, and mouse GC-1spg spermatogonial cells were treated with 0.1-500 µg/mL propolis (12-48 h) in the presence or absence of an oxidant stimulus (tert-butyl hydroperoxide, TBHP, 0.005-3.6 µg/mL, 12 h). Cytotoxicity was assessed by MTT assays and proliferation by Ki-67 immunocytochemistry. Apoptosis, reactive oxygen species (ROS), and antioxidant defenses were evaluated colorimetrically. Propolis presented high phenolic and flavonoid content and moderate antioxidant activity, increasing the viability of GC-1spg cells and counteracting TBHP's effects on viability and proliferation. Additionally, propolis reduced ROS levels in GC-1spg, regardless of the presence of TBHP. Propolis decreased caspase-3 and increased glutathione peroxidase activity in TBHP-treated GC-1spg cells. The present study shows the protective action of propolis against oxidative damage in spermatogonia, opening the possibility of exploiting its benefits to male fertility.


Subject(s)
Ascomycota , Propolis , Male , Bees , Animals , Mice , Spermatogonia , Antioxidants/pharmacology , Propolis/pharmacology , tert-Butylhydroperoxide/toxicity , Reactive Oxygen Species , Seeds , Oxidative Stress
2.
Mol Biol (Mosk) ; 57(4): 689-691, 2023.
Article in Russian | MEDLINE | ID: mdl-37528789

ABSTRACT

Ras proteins are small GTPases and function as molecular switches to regulate cellular homeostasis. Ras-dependent signalling pathways regulate several essential processes such as cell cycle progression, growth, migration, apoptosis, and senescence. The dysregulation of Ras signaling pathway has been linked to several pathological outcomes. A potential role of RAS in regulating the redox signalling pathway has been established that includes the manipulation of ROS levels to provide a redox milieu that might be conducive to carcinogenesis. Reactive oxygen species (ROS) and mitochondrial impairment have been proposed as major factors affecting the physiology of cells and implicated in several pathologies. The present study was conducted to evaluate the role of Ras1, tert Butyl hydroperoxide (tBHP), and antimycin A in oxidative stress response in Schizosaccharomyces pombe cells. We observed decreased cell survival, higher levels of ROS, and mitochondrial dysfunctionality in ras1Δ cells and tBHP as well as respiratory inhibitor, antimycin A treated wild type cells. Furthermore, these defects were more profound in ras1Δ cells treated with tBHP or antimycin A. Additionally, Ras1 also has been shown to regulate the expression and activity of several antioxidant enzymes like glutathione peroxidase (GSH-Px), glutathione-S-transferase (GST), and catalase. Together, these results suggest the potential role of S. pombe Ras1 in mitigating oxidative stress response.


Subject(s)
Schizosaccharomyces , Reactive Oxygen Species/metabolism , tert-Butylhydroperoxide/toxicity , tert-Butylhydroperoxide/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Antimycin A/pharmacology , Antimycin A/metabolism , Oxidative Stress , Oxidation-Reduction
3.
Int J Mol Sci ; 23(8)2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35457223

ABSTRACT

Autophagy is the process by which intracellular components are degraded by lysosomes. It is also activated by oxidative stress; hence, autophagy is thought to be closely related to oxidative stress, one of the major causes of diabetic neuropathy. We previously reported that docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) induced antioxidant enzymes and protected Schwann cells from oxidative stress. However, the relationship between autophagy and oxidative stress-induced cell death in diabetic neuropathy has not been elucidated. Treatment with tert-butyl hydroperoxide (tBHP) decreased the cell survival rate, as measured by an MTT assay in immortalized Fischer rat Schwann cells 1 (IFRS1). A DHA pretreatment significantly prevented tBHP-induced cytotoxicity. tBHP increased autophagy, which was revealed by the ratio of the initiation markers, AMP-activated protein kinase, and UNC51-like kinase phosphorylation. Conversely, the DHA pretreatment suppressed excessive tBHP-induced autophagy signaling. Autophagosomes induced by tBHP in IFRS1 cells were decreased to control levels by the DHA pretreatment whereas autolysosomes were only partially decreased. These results suggest that DHA attenuated excessive autophagy induced by oxidative stress in Schwann cells and may be useful to prevent or reduce cell death in vitro. However, its potentiality to treat diabetic neuropathy must be validated in in vivo studies.


Subject(s)
Diabetic Neuropathies , Docosahexaenoic Acids , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy , Cell Death , Diabetic Neuropathies/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Oxidative Stress , Rats , Rats, Inbred F344 , Schwann Cells/metabolism , Signal Transduction , tert-Butylhydroperoxide/toxicity
4.
J Cell Physiol ; 236(4): 2725-2739, 2021 04.
Article in English | MEDLINE | ID: mdl-32892384

ABSTRACT

Ferroptosis is a necrotic form of regulated cell death that was associated with lipid peroxidation and free iron-mediated Fenton reactions. It has been reported that iron deficiency had been implicated in the pathogenesis of intervertebral disc degeneration (IVDD) by activating apoptosis. However, the role of ferroptosis in the process of IVDD has not been illuminated. Here, we demonstrate the involvement of ferroptosis in IVDD pathogenesis. Our in vitro models show the changes in protein levels of ferroptosis marker and enhanced lipid peroxidation level during oxidative stress. Safranin O staining, hematoxylin-eosin staining, and immunohistochemical were used to assess the IVDD after 8 weeks of surgical procedure in vivo. Treatment with ferrostatin-1, deferoxamine, and RSL3 demonstrate the role of ferroptosis in tert-butyl hydroperoxide (TBHP)-treated annulus fibrosus cells (AFCs) and nucleus pulposus cells (NPCs). Ferritinophagy, nuclear receptor coactivator 4 (NCOA4)-mediated ferritin selective autophagy, is originated during the process of ferroptosis in response to TBHP treatment. Knockdown and overexpression NCOA4 further prove TBHP may induce ferroptosis of AFCs and NPCs in an autophagy-dependent way. These findings support a role for oxidative stress-induced ferroptosis in the pathogenesis of IVDD.


Subject(s)
Annulus Fibrosus/metabolism , Ferroptosis , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/metabolism , Oxidative Stress , Animals , Annulus Fibrosus/drug effects , Annulus Fibrosus/ultrastructure , Autophagy , Carbolines/toxicity , Case-Control Studies , Cells, Cultured , Deferoxamine/pharmacology , Disease Models, Animal , Ferroptosis/drug effects , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/prevention & control , Lipid Peroxidation , Male , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Nucleus Pulposus/drug effects , Nucleus Pulposus/ultrastructure , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Siderophores/pharmacology , Signal Transduction , tert-Butylhydroperoxide/toxicity
5.
Exp Eye Res ; 207: 108586, 2021 06.
Article in English | MEDLINE | ID: mdl-33891955

ABSTRACT

Oxidative stress plays an important role in the pathogenesis of human retinal diseases. Ginkgo biloba products are widely consumed herbal supplements that contain ingredients with anti-oxidant potentials. However, the active agents in ginkgo biloba extracts (GBE) are unclear. This study assessed the anti-oxidant effects of 19 natural compounds isolated from GBE to provide a rational basis for their use in preventing retinal diseases. The compounds were tested in retinal pigment epithelial (RPE) cells subjected to tert-butyl hydroperoxide (t-BHP)-induced oxidative stress. Cell viability and intracellular reactive oxygen species (ROS) were assessed and flow cytometry was used to delineate the cell death profile. The expression of nuclear factor erythroid 2-related factor-2 (Nrf2) was activated in RPE cells by t-BHP accompanied with an activation of Erk1/2 signaling. GBE-derived rutin and procyanidin B2 ameliorated t-BHP-induced cell death and promoted cell viability by suppressing intracellular ROS generation. These agents also enhanced Nrf2 expression with activating Erk1/2 signaling in RPE cells. In contrast, the other compounds tested were minimally active and did not prevent the loss of cell viability elicited by t-BHP. The present findings suggest that rutin and procyanidin B2 may have potential therapeutic values in the prevention of retinal diseases induced by oxidative damage.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Ginkgo biloba/chemistry , MAP Kinase Signaling System/physiology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Plant Extracts/chemistry , Proanthocyanidins/pharmacology , Retinal Pigment Epithelium/drug effects , Rutin/pharmacology , Antioxidants/pharmacology , Blotting, Western , Cell Survival , Cells, Cultured , Flow Cytometry , Humans , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , tert-Butylhydroperoxide/toxicity
6.
Arch Toxicol ; 95(10): 3417-3424, 2021 10.
Article in English | MEDLINE | ID: mdl-34458933

ABSTRACT

The identification of genotoxic agents and their potential for genotoxic alterations in an organism is crucial for risk assessment and approval procedures of the chemical and pharmaceutical industry. Classically, testing strategies for DNA or chromosomal damage focus on in vitro and in vivo (mainly rodent) investigations. In cell culture systems, the alkaline unwinding (AU) assay is one of the well-established methods for detecting the percentage of double-stranded DNA (dsDNA). By establishing a reliable lysis protocol, and further optimization of the AU assay for the model organism Caenorhabditis elegans (C. elegans), we provided a new tool for genotoxicity testing in the niche between in vitro and rodent experiments. The method is intended to complement existing testing strategies by a multicellular organism, which allows higher predictability of genotoxic potential compared to in vitro cell line or bacterial investigations, before utilizing in vivo (rodent) investigations. This also allows working within the 3R concept (reduction, refinement, and replacement of animal experiments), by reducing and possibly replacing animal testing. Validation with known genotoxic agents (bleomycin (BLM) and tert-butyl hydroperoxide (tBOOH)) proved the method to be meaningful, reproducible, and feasible for high-throughput genotoxicity testing, and especially preliminary screening.


Subject(s)
Bleomycin/toxicity , Genomic Instability , Mutagenicity Tests/methods , tert-Butylhydroperoxide/toxicity , Animals , Caenorhabditis elegans , DNA Damage/drug effects , High-Throughput Screening Assays , Mutagens/toxicity , Reproducibility of Results
7.
Chem Res Toxicol ; 33(2): 426-435, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31858786

ABSTRACT

The transcription factor Nrf2a induces a cellular antioxidant response and provides protection against chemical-induced oxidative stress, as well as playing a critical role in development and disease. Zebrafish are a powerful model to study the role of Nrf2a in these processes but have been limited by reliance on transient gene knockdown techniques or mutants with only partial functional alteration. We developed several lines of zebrafish carrying different null (loss of function, LOF) or hyperactive (gain of function, GOF) mutations to facilitate our understanding of the Nrf2a pathway in protecting against oxidative stress. The mutants confirmed Nrf2a dependence for induction of the antioxidant genes gclc, gstp, prdx1, and gpx1a and identified a role for Nrf2a in the baseline expression of these genes, as well as for sod1. Specifically, the 4-fold induction of gstp by tert-butyl hydroperoxide (tBHP) in wild type fish was abolished in LOF mutants. In addition, baseline gstp expression in GOF mutants increased by 12.6-fold and in LOF mutants was 0.8-fold relative to wild type. Nrf2a LOF mutants showed increased sensitivity to the acute toxicity of cumene hydroperoxide (CHP) and tBHP throughout the first 4 days of development. Conversely, GOF mutants were less sensitive to CHP toxicity during the first 4 days of development and were protected against the toxicity of both hydroperoxides after 4 dpf. Neither gain nor loss of Nrf2a modulated the toxicity of R-(-)-carvone (CAR), despite the ability of this compound to potently induce Nrf2a-dependent antioxidant genes. Similar to other species, GOF zebrafish mutants exhibited significant growth and survival defects. In summary, these new genetic tools can be used to facilitate the identification of downstream gene targets of Nrf2a, better define the role of Nrf2a in the toxicity of environmental chemicals, and further the study of diseases involving altered Nrf2a function.


Subject(s)
Benzene Derivatives/toxicity , Clustered Regularly Interspaced Short Palindromic Repeats/drug effects , Gain of Function Mutation , Loss of Function Mutation , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Zebrafish Proteins/genetics , Zebrafish/genetics , tert-Butylhydroperoxide/toxicity , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Dose-Response Relationship, Drug , Gain of Function Mutation/drug effects , Loss of Function Mutation/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/genetics , Zebrafish Proteins/metabolism
8.
Cell Mol Neurobiol ; 40(4): 587-602, 2020 May.
Article in English | MEDLINE | ID: mdl-31828466

ABSTRACT

Neurodegenerative diseases all share several common features such as involvement of oxidative damage and mitochondrial dysfunction in pathogenesis. Oxidative stress induced by overproduction of mitochondrial reactive oxygen species (ROS) or impairment of the antioxidant deficiency results in mitochondrial dysfunction and initiation of the cell death cascade. Berberine (BBR), a traditional Chinese medicine, has been reported to exert anti-oxidative stress and anti-apoptosis effect in CNS diseases. However, the mechanism of BBR on regulating mitophagy and protecting mitochondrial function under oxidative stress remains unclear. In present study, we evaluated the beneficial effects of BBR on the tert-butyl hydroperoxide (t-BHP)-induced cytotoxicity. Furthermore, we explored the protective role of BBR in mitochondrial function and mitophagy under oxidative stress in PC-12 cells. Our results demonstrated that BBR effectively inhibited t-BHP-induced apoptosis which is associated with the decreased leakage of lactate dehydrogenase (LDH) and ROS overproduction. Moreover, BBR significantly suppressed cytochrome c expression, upregulated the ratio of Bcl-2/Bax, and ameliorated mitochondrial dysfunction by optimizing mitochondria membrane potential (ΔΨm) status and ATP production. In addition, BBR reduced the expression of autophagy-specific marker LC3, SQTM1/p62, and maintained lysosome normal function which involved the restoration of upstream signaling pathway AKT and mTOR phosphorylation level. Collectively, these findings suggested that BBR protects PC-12 cells from oxidative injury through inhibiting ROS level, mitochondria dysfunction, and mitophagy via PI3K/AKT/mTOR signaling pathways, which suggest a potential therapeutic strategy for oxidative stress and neurotoxic damages.


Subject(s)
Berberine/pharmacology , Mitochondria/pathology , Oxidative Stress/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Cell Death/drug effects , Chromones/pharmacology , Cytochromes c/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitophagy/drug effects , Morpholines/pharmacology , PC12 Cells , Rats , Signal Transduction/drug effects
9.
Mol Cell Biochem ; 475(1-2): 277-283, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32812103

ABSTRACT

Coumarin-pi, a new coumarin derivative isolated from the mushroom Paxillus involutus, has antioxidative activity, but the underlying mechanism against intracellular oxidative stress is still unclear. This study investigated its cytoprotective effects and the antioxidative mechanism in tert-butyl hydroperoxide (t-BHP)-induced HepG2 cells. The results demonstrated that coumarin-pi suppressed t-BHP-stimulated cytotoxicity, cell apoptosis, and generation of reactive oxygen species (ROS). Additionally, coumarin-pi promoted nuclear factor erythroid 2-related factor 2 (Nrf2) expression and upregulated the protein expression of antioxidantenzymes, including heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidase (NQO1), glutamyl cysteine ligase catalytic subunit (GCLC), and glutamate-cysteine ligase regulatory subunit (GCLM). After coumarin-pi treatment, transcriptome sequencing and bioinformatic analysis revealed that 256 genes were differentially expressed; interestingly, only 20 genes were downregulated, and the rest of the genes were upregulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional annotation were used to identify changes in metabolic pathways. Collectively, the results presented in this study indicate that coumarin-pi has a protective effect against t-BHP-induced cellular damage and oxidative stress.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Coumarins/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , tert-Butylhydroperoxide/toxicity , Antioxidants/metabolism , Cell Line , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hep G2 Cells , Humans , Reactive Oxygen Species/metabolism
10.
Cell Mol Biol (Noisy-le-grand) ; 66(2): 125-129, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32415938

ABSTRACT

The aim of this study was to investigate whether Jatrorrhizine hydrochloride (JAH) can attenuate oxidative damage of endothelial cells by regulating mitochondrial function and inflammatory response. It was found that JAH inhibited tert-butyl hydroperoxide (t-BHP)-induced oxidative damage in mouse brain endothelial cells (MBECs) by increasing cell viability and inhibiting cell apoptosis. Moreover, JAH significantly inhibited the production of reactive oxygen species (ROS) and lipid peroxidation. It enhanced mitochondrial membrane potential (MMP) and maintained ATP synthesis. In addition, JAH regulated the expressions of inflammatory cytokines and increased the activation of endothelial nitric oxide synthase (eNOS). The protective effect of JAH was related to the protein expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) gene. In conclusion, these results suggest that JAH may have therapeutic potential for ischemic stroke associated with endothelial dysfunction through its antioxidant and anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/analogs & derivatives , Oxidative Stress/drug effects , PPAR gamma/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Berberine/pharmacology , Brain/cytology , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide Synthase Type III/metabolism , PPAR gamma/genetics , Reactive Oxygen Species/metabolism , tert-Butylhydroperoxide/toxicity
11.
Biol Pharm Bull ; 43(3): 424-431, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31839625

ABSTRACT

Ferroptosis is characterized by the accumulation of iron-derived reactive oxygen species (ROS). Ferroptosis causes neuronal death in multiple neurological disorders. Dexmedetomidine (Dex), an extensively used anesthetic, has neuroprotective effects against ROS, but its effect on iron metabolism remains unknown. In this study, SK-N-SH cells were treated with Dex for 24 h before treatment with 100 µM tert-butyl hydroperoxide (t-BHP; an ROS inducer) for 1 h. Afterward, intracellular ROS and labile ferrous iron [Fe(II)] levels were assessed. Dex hindered the increase in cellular ROS and labile Fe(II) levels caused by t-BHP, although Dex alone had no effect on labile Fe(II) level. t-BHP increased the expression of iron importers, transferrin receptor-1 and divalent metal transporter-1, and iron regulatory protein 1 and 2. These effects were abrogated by Dex treatment and SP-1 knockdown. t-BHP increased the phosphorylation of c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 4 (STAT4), the primary up-stream activators of SP-1, but Dex decreased this. This study, for the first time, revealed that the antioxidative effect of Dex is partly associated to the inhibition of intracellular iron accumulation induced by t-BHP. Dex regulates iron metabolism by regulating iron importers and exporters through JNK/Sp1 and Stat4/Sp1 signaling. It is worth investigating whether Dex can protect neurons from ferroptosis.


Subject(s)
Dexmedetomidine/pharmacology , Iron/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Cell Survival/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/drug effects , Reactive Oxygen Species/metabolism , STAT4 Transcription Factor/metabolism , Sp1 Transcription Factor/metabolism , tert-Butylhydroperoxide/pharmacology , tert-Butylhydroperoxide/toxicity
12.
Molecules ; 25(8)2020 Apr 12.
Article in English | MEDLINE | ID: mdl-32290603

ABSTRACT

Kushenol C (KC) is a prenylated flavonoid isolated from the roots of Sophora flavescens aiton. Little is known about its anti-inflammatory and anti-oxidative stress activities. Here, we investigated the anti-inflammatory and anti-oxidative stress effects of KC in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages, and tert-butyl hydroperoxide (tBHP)-induced oxidative stress in HaCaT cells. The results demonstrated that KC dose-dependently suppressed the production of inflammatory mediators, including NO, PGE2, IL-6, IL1ß, MCP-1, and IFN-ß in LPS-stimulated RAW264.7 macrophages. The study demonstrated that the inhibition of STAT1, STAT6, and NF-κB activations by KC might have been responsible for the inhibition of NO, PGE2, IL-6, IL1ß, MCP-1, and IFN-ß in the LPS-stimulated RAW264.7 macrophages. KC also upregulated the expression of HO-1 and its activities in the LPS-stimulated RAW264.7 macrophages. The upregulation of Nrf2 transcription activities by KC in the LPS-stimulated RAW264.7 macrophages was demonstrated to be responsible for the upregulation of HO-1 expression and its activity in LPS-stimulated RAW264.7 macrophages. In HaCaT cells, KC prevented DNA damage and cell death by upregulating the endogenous antioxidant defense system involving glutathione, superoxide dismutase, and catalase, which prevented reactive oxygen species production from tert-butyl hydroperoxide (tBHP)-induced oxidative stress in HaCaT cells. The upregulated activation of Nrf2 and Akt in the PI3K-Akt signaling pathway by KC was demonstrated to be responsible for the anti-oxidative stress activity of KC in HaCaT cells. Collectively, the study suggests that KC can be further investigated as a potential anti-inflammatory candidate for the treatment of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Flavonoids/pharmacology , Plant Extracts/pharmacology , Plant Roots/chemistry , Sophora/chemistry , Animals , Catalase/metabolism , Cell Line , Flavonoids/chemistry , Glutathione/metabolism , HaCaT Cells , Humans , Lipopolysaccharides/toxicity , Macrophages/metabolism , Mice , Oxidative Stress/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , tert-Butylhydroperoxide/toxicity
13.
J Cell Mol Med ; 23(1): 177-193, 2019 01.
Article in English | MEDLINE | ID: mdl-30353656

ABSTRACT

Melatonin is reportedly associated with intervertebral disc degeneration (IDD). Endplate cartilage is vitally important to intervertebral discs in physiological and pathological conditions. However, the effects and mechanism of melatonin on endplate chondrocytes (EPCs) are still unclear. Herein, we studied the effects of melatonin on EPC apoptosis and calcification and elucidated the underlying mechanism. Our study revealed that melatonin treatment decreases the incidence of apoptosis and inhibits EPC calcification in a dose-dependent manner. We also found that melatonin upregulates Sirt1 expression and activity and promotes autophagy in EPCs. Autophagy inhibition by 3-methyladenine reversed the protective effect of melatonin on apoptosis and calcification, while the Sirt1 inhibitor EX-527 suppressed melatonin-induced autophagy and the protective effects of melatonin against apoptosis and calcification, indicating that the beneficial effects of melatonin in EPCs are mediated through the Sirt1-autophagy pathway. Furthermore, melatonin may ameliorate IDD in vivo in rats. Collectively, this study revealed that melatonin reduces EPC apoptosis and calcification and that the underlying mechanism may be related to Sirt1-autophagy pathway regulation, which may help us better understand the association between melatonin and IDD.


Subject(s)
Calcinosis/drug therapy , Chondrocytes/drug effects , Melatonin/pharmacology , Sirtuin 1/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy/physiology , Calcinosis/metabolism , Calcinosis/pathology , Carbazoles/pharmacology , Cells, Cultured , Chondrocytes/pathology , Disease Models, Animal , Female , Intervertebral Disc Degeneration/chemically induced , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/pathology , Male , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rats, Sprague-Dawley , Sirtuin 1/antagonists & inhibitors , tert-Butylhydroperoxide/toxicity
14.
Exp Eye Res ; 185: 107641, 2019 08.
Article in English | MEDLINE | ID: mdl-30980814

ABSTRACT

Dysfunction and eventual loss of retinal pigment epithelial (RPE) cells is a hallmark of atrophic age-related macular degeneration (AMD), and linked to oxidative and nitrosative damage. Herein, we use a high-throughput screen (HTS) to identify compounds that protect human RPE cells from oxidative stress-induced cell death and elucidate the possible mechanism of action. HTS was used to identify compounds that protect RPE cells from oxidative damage. We tested the identified compound(s) in models of RPE stress, including tert-butyl hydroperoxide (TBHP) exposure, ultraviolet-B (UV-B)-mediated light damage and nitrosative stress to the basement membrane using ARPE-19 cells, primary human RPE cells and induced-pluripotent stem cell (iPSC)-derived RPE cells from patients with AMD. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect gene expression of oxidative stress- and apoptosis-related genes and mitochondrial function was measured using a Seahorse XF96 analyzer to elucidate possible mechanisms of action. Five thousand and sixty-five compounds were screened, and of these, 12 compounds were active based on their ability to improve cell viability after exposure to TBHP. After chemical structure review, we identified ciclopirox olamine as a potent inhibitor of oxidative damage to RPE cells. Ciclopirox olamine increased cell viability in ARPE-19 cells treated with TBHP, UV-B light or on nitrite-modified extracellular matrix (ECM) by 1.68-fold, 1.54-fold and 4.3-fold, respectively (p < 0.01). Treatment with TBHP altered expression of genes related to oxidative stress and apoptosis, which was reversed by pretreatment with ciclopirox olamine. Ciclopirox olamine improved mitochondrial function in TBHP-exposed ARPE-19 cells and iPSC-derived RPE cells. Ciclopirox olamine protected primary human RPE cells and iPSC-derived RPE cells from the oxidative stress or damaged basement membrane. HTS of bioactive Food and Drug Administration (FDA)-approved libraries and follow-up studies can be used to identify small molecules (including ciclopirox olamine) that protect RPE cells exposed to various stressors associated with disease progression of AMD. This strategy can be used to identify potential compounds for treatment and prevention of AMD.


Subject(s)
Antifungal Agents/therapeutic use , Ciclopirox/therapeutic use , Induced Pluripotent Stem Cells/drug effects , Macular Degeneration/drug therapy , Oxidative Stress , Retinal Pigment Epithelium/drug effects , Apoptosis , Basement Membrane/drug effects , Basement Membrane/metabolism , Basement Membrane/pathology , Catalase/genetics , Catalase/metabolism , Cell Line , Cytoprotection , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Nitrosative Stress/physiology , Peroxiredoxin III/genetics , Peroxiredoxin III/metabolism , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Ultraviolet Rays/adverse effects , tert-Butylhydroperoxide/toxicity
15.
Can J Physiol Pharmacol ; 97(10): 999-1005, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31295412

ABSTRACT

Molecular hydrogen (H2) was believed to be an inert and nonfunctional molecule in mammalian cells; however, we overturned the concept by reporting the therapeutic effects of H2 against oxidative stress. Subsequently, extensive studies revealed multiple functions of H2 by exhibiting the efficacies of H2 in various animal models and clinical studies. Here, we investigated the effect of H2 on free-radical-induced cytotoxicity using tert-butyl hydroperoxide in a human acute monocytic leukemia cell line, THP-1. Cell membrane permeability was determined using lactate dehydrogenase release assay and Hoechst 33342 and propidium iodide staining. Fatty acid peroxidation and mitochondrial viability were measured using 2 kinds of fluorescent dyes, Liperfluo and C11-BODIPY, and using the alamarBlue assay based on the reduction of resazurin to resorufin by mainly mitochondrial succinate dehydrogenase, respectively. Mitochondrial membrane potential was evaluated using tetramethylrhodamine methyl ester. As a result, H2 protected the cultured cells against the cytotoxic effects induced by tert-butyl hydroperoxide; H2 suppressed cellular fatty acid peroxidation and cell membrane permeability, mitigated the decline in mitochondrial oxidoreductase activity and mitochondrial membrane potential, and protected cells against cell death evaluated using propidium iodide staining. These results suggested that H2 suppresses free-radical-induced cell death through protection against fatty acid peroxidation and mitochondrial dysfunction.


Subject(s)
Hydrogen/pharmacology , Mitochondria/drug effects , tert-Butylhydroperoxide/toxicity , Apoptosis/drug effects , Atherosclerosis/drug therapy , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Fatty Acids/metabolism , Humans , Hydrogen/therapeutic use , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects
16.
Arch Toxicol ; 93(5): 1265-1279, 2019 05.
Article in English | MEDLINE | ID: mdl-30798349

ABSTRACT

Ferroptosis is a recently discovered pathway of regulated necrosis dependent on iron and lipid peroxidation. It has gained broad attention since it is a promising approach to overcome resistance to apoptosis in cancer chemotherapy. We have recently identified tertiary-butyl hydroperoxide (t-BuOOH) as a novel inducer of ferroptosis. t-BuOOH is a widely used compound to induce oxidative stress in vitro. t-BuOOH induces lipid peroxidation and consequently ferroptosis in murine and human cell lines. t-BuOOH additionally results in a loss of mitochondrial membrane potential, formation of DNA double-strand breaks, and replication block. Here, we specifically address the question whether cell-cell contacts regulate t-BuOOH-induced ferroptosis and cellular damage. To this end, murine NIH3T3 or human HaCaT cells were seeded to confluence, but below their saturation density to allow the establishment of cell-cell contacts without inducing quiescence. Cells were then treated with t-BuOOH (50 or 200 µM, respectively). We revealed that cell-cell contacts reduce basal and t-BuOOH-triggered lipid peroxidation and consequently block ferroptosis. Similar results were obtained with the specific ferroptosis inducer erastin. Cell-cell contacts further protect against t-BuOOH-induced loss of mitochondrial membrane potential, and formation of DNA double-strand breaks. Interestingly, cell-cell contacts failed to prevent t-BuOOH-mediated replication block or formation of the oxidative base lesion 8-oxo-dG. Since evidence of protection against cell death was both (i) observed after treatment with hydrogen peroxide, methyl methanesulfonate or UV-C, and (ii) seen in several cell lines, we conclude that protection by cell-cell contacts is a widespread phenomenon. The impact of cell-cell contacts on toxicity might have important implications in cancer chemotherapy.


Subject(s)
Ferroptosis/drug effects , Membrane Potential, Mitochondrial/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Cell Communication/physiology , Cell Death/drug effects , Cell Line , DNA Breaks, Double-Stranded/drug effects , Dose-Response Relationship, Drug , Humans , Hydrogen Peroxide/administration & dosage , Lipid Peroxidation/drug effects , Mice , NIH 3T3 Cells , Oxidative Stress/drug effects , tert-Butylhydroperoxide/administration & dosage
17.
BMC Complement Altern Med ; 19(1): 139, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31221142

ABSTRACT

BACKGROUND: Several studies have found that caffeic acid (CA), a well-known phytochemical, displays important antioxidant and anti-cancer activities. However, no evidence exists on the protective effect and its mechanisms that CA treatment alone has against oxidative stress induced by tert-butyl hydroperoxide (t-BHP) in HepG2 cells. METHODS: Hepatoprotective activities such as cell viability, mRNA expression, and report gene assay were measured using HepG2 cell. Three types of genes and proteins related with detoxification in liver were used for measuring the hepatoprotective effects. Statistical analysis was performed using one-way ANOVA test and differences among groups were evaluated by Tukey's studentized range tests. RESULTS: The present study indicate that treatment with CA up-regulates heme oxygenase-1 (HO-1) and glutamate-cysteine ligase (GCL) mRNA and protein expressions in a CA-dose-dependent manner. In addition, translocation of nuclear factor-E2 p45-related factor (Nrf2) from the cytoplasm to the nucleus and phosphorylation of extracellular signal-regulated kinase, ERK and c-Jun N-terminal kinase, JNK which have been shown to be involved in mitogen-activated protein kinases, MAPKs are significantly enhanced by CA treatment. Furthermore, in cell nuclei, CA enhances the 5'-flanking regulatory region of human antioxidant response element (ARE) and activates the ARE binding site. CONCLUSION: Therefore, CA proved to be a stimulant of the expression of detoxification enzymes such as HO-1, GCLC, and GCLM through the ERK/Nrf2 pathway, and it may be an effective chemoprotective agent for protecting liver damage against oxidative damage.


Subject(s)
Caffeic Acids/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , tert-Butylhydroperoxide/toxicity , Antioxidant Response Elements/drug effects , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Liver Neoplasms/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism
18.
Molecules ; 24(21)2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31671915

ABSTRACT

Enhanced oxidative stress has been associated with muscle mitochondrial changes and metabolic disorders. Thus, it might be a good strategy to decrease oxidative stress and improve mitochondrial changes in skeletal muscle. In the present study, we investigate the role of the most biologically active metabolite of vitamin D, 1,25-dihyroxyvitamin D (1,25(OH)2D) in oxidative stress and mitochondrial changes in tertiary butyl-hydrogen (tBHP)-treated C2C12 muscle cells. Differentiated C2C12 muscle cells were pretreated with tBHP, followed by 1,25(OH)2D for additional 24 h. An exogenous inducer of oxidative stress, tBHP significantly increased oxidative stress, lipid peroxidation, intracellular damage, and cell death which were reversed by 1,25(OH)2D in C2C12 myotubes. 1.25(OH)2D improves tBHP-induced mitochondrial morphological changes such as swelling, irregular cristae, and smaller size and number, as observed by transmission electron microscope. In addition, 1,25(OH)2D treatment increases mtDNA contents as well as gene expression involved in mitochondrial biogenesis such as PGC1α, NRF1, and Tfam. Significant increments in mRNA levels related to antioxidant enzymes such as Nrf2, HMOX1, and TXNRD1, myogenic differentiation markers including myoglobin, muscle creatine kinase (MCK), and MHCІ and ІІ, and vitamin D metabolism such as CYP24, CYP27, and vitamin D receptor (VDR) were found in 1,25(OH)2D-treated myotubes. Moreover, upon t-BHP-induced oxidative stress, significant incremental changes in nicotinamide adenine dinucleotide (NAD) levels, activities of AMP-activated protein kinase (AMPK)/sirtulin 1 (SIRT1), and SIRT1 expression were noted in 1,25(OH)2D-treated C2C12 muscle cells. Taken together, these results suggest the observed potent inhibitory effect of 1,25(OH)2D on muscle oxidative stress and mitochondrial dynamics might be at least involved in the activation of AMPK and SIRT1 activation in muscle cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Muscle Cells/pathology , Oxidative Stress/drug effects , Sirtuin 1/metabolism , Vitamin D/analogs & derivatives , tert-Butylhydroperoxide/toxicity , Animals , Cell Line , Cell Survival/drug effects , DNA, Mitochondrial/genetics , Gene Expression Regulation/drug effects , Lipid Peroxidation/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Muscle Cells/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Vitamin D/pharmacology
19.
J Cell Mol Med ; 22(6): 3086-3096, 2018 06.
Article in English | MEDLINE | ID: mdl-29575654

ABSTRACT

Spermidine has therapeutic effects in many diseases including as heart diastolic function, myopathic defects and neurodegenerative disorders via autophagy activation. Autophagy has been found to mitigate cell apoptosis in intervertebral disc degeneration (IDD). Accordingly, we theorize that spermidine may have beneficial effects on IDD via autophagy stimulation. In this study, spermidine's effect on IDD was evaluated in tert-butyl hydroperoxide (TBHP)-treated nucleus pulposus cells of SD rats in vitro as well as in a puncture-induced rat IDD model. We found that autophagy was actuated by spermidine in nucleus pulposus cells. In addition, spermidine treatment weakened the apoptotic effects of TBHP in nucleus pulposus cells. Spermidine increased the expression of anabolic proteins including Collagen-II and aggrecan and decreased the expression of catabolic proteins including MMP13 and Adamts-5. Additionally, autophagy blockade using 3-MA reversed the beneficial impact of spermidine against nucleus pulposus cell apoptosis. Autophagy was thus important for spermidine's therapeutic effect on IDD. Spermidine-treated rats had an accentuated T2-weighted signal and a diminished histological degenerative grade than vehicle-treated rats, showing that spermidine inhibited intervertebral disc degeneration in vivo. Thus, spermidine protects nucleus pulposus cells against apoptosis through autophagy activation and improves disc, which may be beneficial for the treatment of IDD.


Subject(s)
Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc/drug effects , Nucleus Pulposus/drug effects , Spermidine/administration & dosage , Animals , Apoptosis/drug effects , Autophagy/drug effects , Disease Models, Animal , Extracellular Matrix/drug effects , Humans , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/chemically induced , Intervertebral Disc Degeneration/physiopathology , Nucleus Pulposus/pathology , Primary Cell Culture , Rats , tert-Butylhydroperoxide/toxicity
20.
Cell Physiol Biochem ; 45(5): 1915-1926, 2018.
Article in English | MEDLINE | ID: mdl-29518773

ABSTRACT

BACKGROUND/AIMS: Oxidants are important human toxicants. They have been implicated in the occurrence and development of liver diseases. Increased intracellular tert-butylhydroperoxide (t-BHP) may be critical for oxidant toxicity, and is commonly used for evaluating mechanisms involving oxidative stress, but the method remains controversial. METHODS: Primary cultures of hepatocytes as well as human Hep G2 and mouse FL83B liver cells were obtained. Cell viability was measured by annexin V-FITC/propidium iodide and DAPI staining to determine the effects of t-BHP treatment on acute liver injury. A proteomic assay provided information that was used to identify the differentially expressed proteins following t-BHP treatment; immunohistochemistry and western blotting were performed to detect the expression of PDIA6 activity in apoptotic and endoplasmic reticulum (ER) stress pathways. RESULTS: Our results demonstrate that t-BHP treatment of liver cells increased cell cytotoxicity and the generation of reactive oxygen species. This treatment also increased the level of PDIA6; this was validated in vitro and in vivo based on a comparison of t-BHP-treated and -untreated groups. Treatment of mouse liver FL83B cells with t-BHP activated caspase 3, increased the expression of apoptotic molecules, caused cytochrome c release, and induced Bcl-2, Bax and IRE1α/TRAF2 complex formation. t-BHP-dependent induction of apoptosis was accompanied by sustained phosphorylation of the IRE1α/ASK1/JNK1/2/p38 pathways and PDIA6 expression. Furthermore, t-BHP induced liver FL83B cell viability and apoptosis by upregulating the levels of PDIA6; this process could be involved in the activation of the IRE1α/ASK1/JNK1/2/p38 signalling pathways. CONCLUSIONS: We conclude that t-BHP induced an apoptosis cascade and ER stress in hepatocytes by upregulation of PDIA6, providing a new mechanism underlying the effects of t-BHP on liver injury.


Subject(s)
Protein Disulfide-Isomerases/metabolism , Proteomics , Up-Regulation/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Apoptosis/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Multienzyme Complexes/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL