Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 12(3): e2202045, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36239177

ABSTRACT

Doxorubicin (Dox)-mediated generation of reactive oxygen radicals (ROS) for mitochondrial apoptosis is identified as a new cytotoxic mechanism in addition to the well-established one via nuclear DNA replication interference. However, this mechanism contributes far less than the latter to Dox therapy. This newly identified pathway to make Dox therapy function like the combination of chemodynamic therapy (CDT) and chemotherapy-mediated by Dox alone would be amplified. One-pot nanoconstruction (HEBD) is fabricated based on the chemical reactions driven assemblies among epigallocatechin gallate (EGCG), buthionine sulfoximine (BSO) and formaldehyde in aqueous mediums followed by Dox adsorption. Acid tumor microenvironments allow the liberation of EGCG, BSO, and Dox due to the breakage of Schiff base bonds. EGCG component in HEBD is responsible for targeting mitochondria and disrupting mitochondrial electron transport chain (mETC) to compel electrons leakage in favor of their capture by Dox to produce more ROS. EGCG-induced mETC disruption results in mitochondrial respiration inhibition with alleviated hypoxia in tumor cells while BSO inhibits glutathione biosynthesis to protect ROS from redox depletion, further boosting Dox-induced CDT. This strategy of amplifying CDT pathway for the Dox-mediated combined therapy could largely improve antitumor effect, extend lifespan of tumor-bearing mice, reduce risks of cardiotoxicity and metastasis.


Subject(s)
Apoptosis , Doxorubicin , Mice , Animals , Reactive Oxygen Species/metabolism , Doxorubicin/pharmacology , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Mitochondria
2.
Arch Toxicol ; 96(12): 3315-3329, 2022 12.
Article in English | MEDLINE | ID: mdl-36057886

ABSTRACT

The persistence of hepatotoxicity induced by N-acetyl-para-aminophenol (Acetaminophen or Paracetamol, abbreviated as APAP) as the most common cause of acute liver failure in the United States, despite the availability of N-acetylcysteine, illustrates the clinical relevance of additional therapeutic approaches. While human mesenchymal stem cells (MSCs) have shown protection in mouse models of liver injury, the MSCs used are generally not cleared for human use and it is unclear whether these effects are due to xenotransplantation. Here we evaluated GMP manufactured clinical grade human Wharton's Jelly mesenchymal stem cells (WJMSCs), which are currently being investigated in human clinical trials, in a mouse model of APAP hepatotoxicity in comparison to human dermal fibroblasts (HDFs) to address these issues. C57BL6J mice were treated with a moderate APAP overdose (300 mg/kg) and WJMSCs were administered 90 min later. Liver injury was evaluated at 6 and 24 h after APAP. WJMSCs treatment reduced APAP-induced liver injury at both time points unlike HDFs, which showed no protection. APAP-induced JNK activation as well as AIF and Smac release from mitochondria were prevented by WJMSCs treatment without influencing APAP bioactivation. Mechanistically, WJMSCs treatment upregulated expression of Gclc and Gclm to enhance recovery of liver GSH levels to attenuate mitochondrial dysfunction and accelerated recovery of pericentral hepatocytes to re-establish liver zonation and promote liver homeostasis. Notably, preventing GSH resynthesis with buthionine sulfoximine prevented the protective effects of WJMSCs. These data indicate that these GMP-manufactured WJMCs could be a clinically relevant therapeutic approach in the management of APAP hepatotoxicity in humans.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Mesenchymal Stem Cells , Wharton Jelly , Humans , Mice , Animals , Acetaminophen/metabolism , Acetylcysteine/pharmacology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Liver , Hepatocytes , Disease Models, Animal , Fibroblasts , Mice, Inbred C57BL
3.
Toxicol Appl Pharmacol ; 443: 116007, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35367474

ABSTRACT

8-Epidiosbulbin E acetate (EEA), a furan-containing diterpenoid lactone, is one of main component of Dioscorea bulbifera L. (DBL). It has been reported that EEA induces severe hepatotoxicity in mice and that its hepatotoxicity is associated with metabolic activation. The present study demonstrated that exposure to EEA (50, 100 or 200 µM) induced DNA damage, including significant DNA fragmentation, increases of tail DNA and olive tail moment, H2AX phosphorylation and PARP-1 activation, in cultured mouse primary hepatocytes. Similar observation was obtained in mice administered EEA at 50, 100 or 200 mg/kg. Pre-treatment with 10 µM ketoconazole (KTC), 200 µM vitamin C (VC), or 200 µM glutathione ethyl ester (GSH-OEt) reversed the over-production of reactive oxygen species (ROS) induced by EEA and attenuated susceptibility of hepatocytes to EEA-induced cytotoxicity and DNA damage in mouse primary hepatocytes. In contrast, pre-treatment with 1.0 mM L-buthionine sulfoximine (BSO) potentiated over-production of ROS, cytotoxicity and DNA damage induced by EEA. In summary, EEA induced DNA damage in cultured primary hepatocytes and the liver of mice. ROS, possibly along with DNA alkylation, participated in the observed DNA damage.


Subject(s)
Chemical and Drug Induced Liver Injury , Diterpenes , Activation, Metabolic , Animals , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , DNA/metabolism , DNA Damage , Diterpenes/pharmacology , Glutathione/metabolism , Mice , Reactive Oxygen Species
4.
Biotechnol Bioeng ; 117(11): 3448-3458, 2020 11.
Article in English | MEDLINE | ID: mdl-32662871

ABSTRACT

Glutathione (GSH) plays a central role in the redox balance maintenance in mammalian cells. Previous studies of industrial Chinese hamster ovary cell lines have demonstrated a relationship between GSH metabolism and clone productivity. However, a thorough investigation is required to understand this relationship and potentially highlight new targets for cell engineering. In this study, we have modulated the GSH intracellular content of an industrial cell line under bioprocess conditions to further elucidate the role of the GSH synthesis pathway. Two strategies were used: the variation of cystine supply and the direct inhibition of the GSH synthesis using buthionine sulfoximine (BSO). Over time of the bioprocess, a correlation between intracellular GSH and product titer has been observed. Analysis of metabolites uptake/secretion rates and proteome comparison between BSO-treated cells and nontreated cells has highlighted a slowdown of the tricarboxylic acid cycle leading to a secretion of lactate and alanine in the extracellular environment. Moreover, an adaptation of the GSH-related proteome has been observed with an upregulation of the regulatory subunit of glutamate-cysteine ligase and a downregulation of a specific GSH transferase subgroup, the Mu family. Surprisingly, the main impact of BSO treatment was observed on a global downregulation of the cholesterol synthesis pathways. As cholesterol is required for protein secretion, it could be the missing piece of the puzzle to finally elucidate the link between GSH synthesis and productivity.


Subject(s)
Buthionine Sulfoximine/metabolism , Cholesterol/metabolism , Glutathione/metabolism , Proteome/metabolism , Animals , CHO Cells/metabolism , Cricetulus , Proteome/analysis , Proteomics
5.
Mol Biol Rep ; 46(4): 4423-4435, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31147858

ABSTRACT

Parkinson disease (PD) is characterized by the loss of dopaminergic (DAergic) neurons linked to environmental toxicants that cause oxidative stress (OS). The aim of this investigation was to establish the molecular response of human mesenchymal stroma cells (MSCs) depleted of glutathione (GSH) by the specific inhibitor L-buthionine-sulfoximine (BSO) to 6-hydroxydopamine (6-OHDA) and/or N-acetylcysteine (NAC) co-treatment. We found that treatment with BSO (10 mM) plus 6-OHDA (200 µM) induced apoptosis in MSCs through an oxidative stress (OS) mechanism involving H2O2, reflected by the detection of dichlorofluorescein-positive (DCF+) cells and oxidation of DJ-1 Cys106-SH into DJ-1 Cys106-SO3; an almost complete reduction in glutathione peroxidase 1 (GPX1) expression; activation of the transcription factor c-JUN, the pro-apoptotic protein BAX and BH-3-only protein PUMA; loss of mitochondrial membrane potential (∆Ψm); activation of the protease caspase-3 (CASP3) and apoptosis-inducing factor (AIF); chromatin condensation; and DNA fragmentation. Strikingly, co-treatment of MSCs with NAC (5 mM) and BSO + 6-OHDA significantly reduced the expression of OS and cell death markers but were unable to restore the expression of GPX1 compared to the expression in untreated or treated cells with NAC only. These findings highlighted the importance of the maintenance of the GSH-dependent (e.g., GPX1, GSH synthesis) and -independent (e.g., ROS scavenger molecules and thiol reducing activity) antioxidant systems (e.g., NAC) in the protection of MSCs from detrimental stress stimuli, thereby increasing the survival of stromal cells.


Subject(s)
Acetylcysteine/pharmacology , Apoptosis/drug effects , Mesenchymal Stem Cells/drug effects , Antioxidants/metabolism , Buthionine Sulfoximine/metabolism , Cell Death/drug effects , Glutathione/metabolism , Glutathione Peroxidase , Humans , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidopamine/metabolism , Reactive Oxygen Species/metabolism , Glutathione Peroxidase GPX1
6.
Free Radic Biol Med ; 135: 1-14, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30790656

ABSTRACT

Elaborate antioxidant pathways have evolved to minimize the threat of excessive reactive oxygen species (ROS) and to regulate ROS as signaling entities. ROS are chemically and functionally similar to reactive sulfur species (RSS) and both ROS and RSS have been shown to be metabolized by the antioxidant enzymes, superoxide dismutase and catalase. Here we use fluorophores to examine the effects of a variety of inhibitors of antioxidant pathways on metabolism of two important RSS, hydrogen sulfide (H2S with AzMC) and polysulfides (H2Sn, where n = 2-7, with SSP4) in HEK293 cells. Cells were exposed to inhibitors for up to 5 days in normoxia (21% O2) and hypoxia (5% O2), conditions also known to affect ROS production. Decreasing intracellular glutathione (GSH) with l-buthionine-sulfoximine (BSO) or diethyl maleate (DEM) decreased H2S production for 5 days but did not affect H2Sn. The glutathione reductase inhibitor, auranofin, initially decreased H2S and H2Sn but after two days H2Sn increased over controls. Inhibition of peroxiredoxins with conoidin A decreased H2S and increased H2Sn, whereas the glutathione peroxidase inhibitor, tiopronin, increased H2S. Aminoadipic acid, an inhibitor of cystine uptake did not affect either H2S or H2Sn. In buffer, the glutathione reductase and thioredoxin reductase inhibitor, 2-AAPA, the glutathione peroxidase mimetic, ebselen, and tiopronin variously reacted directly with AzMC and SSP4, reacted with H2S and H2S2, or optically interfered with AzMC or SSP4 fluorescence. Collectively these results show that antioxidant inhibitors, generally known for their ability to increase cellular ROS, have various effects on cellular RSS. These findings suggest that the inhibitors may affect cellular sulfur metabolism pathways that are not related to ROS production and in some instances they may directly affect RSS or the methods used to measure them. They also illustrate the importance of carefully evaluating RSS metabolism when biologically or pharmacologically attempting to manipulate ROS.


Subject(s)
Antioxidants/metabolism , Hydrogen Sulfide/metabolism , Sulfides/metabolism , Sulfur/metabolism , Buthionine Sulfoximine/metabolism , Catalase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , HEK293 Cells , Humans , Maleates/metabolism , Metabolic Networks and Pathways , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
7.
Free Radic Biol Med ; 102: 1-15, 2017 01.
Article in English | MEDLINE | ID: mdl-27847240

ABSTRACT

RATIONALE: Chlorine gas (Cl2) is a potent oxidant and trigger of irritant induced asthma. We explored NF-E2-related factor 2 (Nrf2)-dependent mechanisms in the asthmatic response to Cl2, using Nrf2-deficient mice, buthionine sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis and sulforaphane (SFN), a phytochemical regulator of Nrf2. METHODS: Airway inflammation and airway hyperresponsiveness (AHR) were assessed 24 and 48h after a 5-min nose-only exposure to 100ppm Cl2 of Nrf2-deficient and wild type Balb/C mice treated with BSO or SFN. Animals were anesthetized, paralyzed and mechanically ventilated (FlexiVent™) and challenged with aerosolized methacholine. Bronchoalveolar lavage (BAL) was performed and lung tissues were harvested for assessment of gene expression. RESULTS: Cl2 exposure induced a robust AHR and an intense neutrophilic inflammation that, although similar in Nrf2-deficient mice and wild-type mice at 24h after Cl2 exposure, were significantly greater at 48h post exposure in Nrf2-deficient mice. Lung GSH and mRNA for Nrf2-dependent phase II enzymes (NQO-1 and GPX2) were significantly lower in Nrf2-deficient than wild-type mice after Cl2 exposure. BSO reduced GSH levels and promoted Cl2-induced airway inflammation in wild-type mice, but not in Nrf2-deficient mice, whereas SFN suppressed Cl2-induced airway inflammation in wild-type but not in Nrf2-deficient mice. AHR was not affected by either BSO or SFN at 48h post Cl2 exposure. CONCLUSIONS: Nrf2-dependent phase II enzymes play a role in the resolution of airway inflammation and AHR after Cl2 exposure. Moderate deficiency of GSH affects the magnitude of acute inflammation but not AHR.


Subject(s)
Inflammation/metabolism , Lung/metabolism , NF-E2-Related Factor 2/genetics , Respiratory Hypersensitivity/metabolism , Animals , Bronchoalveolar Lavage , Buthionine Sulfoximine/metabolism , Chlorine/toxicity , Gene Expression Regulation/genetics , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Isothiocyanates/metabolism , Lung/drug effects , Lung/physiopathology , Methacholine Chloride/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , RNA, Messenger/genetics , Respiratory Hypersensitivity/physiopathology , Sulfoxides
8.
PLoS One ; 11(10): e0163214, 2016.
Article in English | MEDLINE | ID: mdl-27788147

ABSTRACT

BACKGROUND: Although reduced glutathione (rGSH) is decreased in obese mice and humans, block of GSH synthesis by buthionine sulfoximine (BSO) results in a lean, insulin-sensitive phenotype. Data is lacking about the effect of BSO on GSH precursors, cysteine and glutamate. Plasma total cysteine (tCys) is positively associated with stearoyl-coenzyme A desaturase (SCD) activity and adiposity in humans and animal models. OBJECTIVE: To explore the phenotype, amino acid and fatty acid profiles in BSO-treated mice. DESIGN: Male C3H/HeH mice aged 11 weeks were fed a high-fat diet with or without BSO in drinking water (30 mmol/L) for 8 weeks. Amino acid and fatty acid changes were assessed, as well as food consumption, energy expenditure, locomotor activity, body composition and liver vacuolation (steatosis). RESULTS: Despite higher food intake, BSO decreased particularly fat mass but also lean mass (both P<0.001), and prevented fatty liver vacuolation. Physical activity increased during the dark phase. BSO decreased plasma free fatty acids and enhanced insulin sensitivity. BSO did not alter liver rGSH, but decreased plasma total GSH (tGSH) and rGSH (by ~70%), and liver tGSH (by 82%). Glutamate accumulated in plasma and liver. Urine excretion of cysteine and its precursors was increased by BSO. tCys, rCys and cystine decreased in plasma (by 23-45%, P<0.001 for all), but were maintained in liver, at the expense of decreased taurine. Free and total plasma concentrations of the SCD products, oleic and palmitoleic acids were decreased (by 27-38%, P <0.001 for all). CONCLUSION: Counterintuitively, block of GSH synthesis decreases circulating tCys, raising the question of whether the BSO-induced obesity-resistance is linked to cysteine depletion. Cysteine-supplementation of BSO-treated mice is warranted to dissect the effects of cysteine and GSH depletion on energy metabolism.


Subject(s)
Amino Acids/metabolism , Body Weight , Fatty Acids/metabolism , Glutathione/deficiency , Phenotype , Sulfhydryl Compounds/metabolism , Adipose Tissue/cytology , Amino Acids/blood , Animals , Body Composition , Buthionine Sulfoximine/metabolism , Eating , Energy Metabolism , Fatty Acids/blood , Glutathione/urine , Insulin/metabolism , Liver/cytology , Liver/metabolism , Locomotion , Male , Mice , Sulfhydryl Compounds/blood
9.
BMC Pharmacol Toxicol ; 16: 37, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26666373

ABSTRACT

BACKGROUND: Previously, we have demonstrated that human ABCB5 is a full-sized ATP-binding cassette transporter that shares strong homology with ABCB1/P-glycoprotein. ABCB5-transfected cells showed resistance to taxanes and anthracyclines. Herein, we further screened ABCB5 substrates, and explored the mechanism of resistance. METHODS: Sensitivity of the cells to test compounds was evaluated using cell growth inhibition assay. Cellular levels of buthionine sulfoximine (BSO), glutathione and amino acids were measured using HPLC and an enzyme-based assay. Cellular and vesicular transport of glutathione was evaluated by a radiolabeled substrate. Expression levels of glutathione-metabolizing enzymes were assessed by RT-PCR. RESULTS: Human ABCB5-transfected 293/B5-11 cells and murine Abcb5-transfected 293/mb5-8 cells showed 6.5- and 14-fold higher resistance to BSO than the mock-transfected 293/mock cells, respectively. BSO is an inhibitor of gamma-glutamylcysteine ligase (GCL), which is a key enzyme of glutathione synthesis. 293/B5-11 and 293/mb5-8 cells also showed resistance to methionine sulfoximine, another GCL inhibitor. A cellular uptake experiment revealed that BSO accumulation in 293/B5-11 and 293/mb5-8 cells was similar to that in 293/mock cells, suggesting that BSO is not an ABCB5 substrate. The cellular glutathione content in 293/B5-11 and 293/mb5-8 cells was significantly higher than that in 293/mock cells. Evaluation of the BSO effect on the cellular glutathione content showed that compared with 293/mock cells the BSO concentration required for a 50 % reduction in glutathione content in 293/B5-11 and 293/mb5-8 cells was approximately 2- to 3-fold higher. This result suggests that the BSO resistance of the ABCB5- and Abcb5-transfected cells can be attributed to the reduced effect of BSO on the transfectants. Cellular and vesicular transport assays showed that the transport of radiolabeled glutathione in 293/B5-11 cells was similar to that in 293/mock cells. The mRNA expression of genes encoding glutathione-metabolizing enzymes in 293/B5-11 cells was similar to that in 293/mock cells. The cellular content of Glu, a precursor of glutathione, in 293/B5-11 and 293/mb5-8 cells was higher than that in 293/mock cells. CONCLUSIONS: ABCB5/Abcb5-transfected cells showed resistance to BSO, which is not a substrate of ABCB5. Our results suggest that ABCB5/Abcb5 upregulates cellular glutathione levels to protect cells from various poisons.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Glutathione/metabolism , Up-Regulation , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Amino Acids/metabolism , Animals , Blotting, Western , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Drug Resistance/genetics , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Glutathione Synthase/genetics , Glutathione Synthase/metabolism , HEK293 Cells , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
Mol Biochem Parasitol ; 201(1): 16-25, 2015 May.
Article in English | MEDLINE | ID: mdl-26024834

ABSTRACT

Previously, we have studied the effect of the gold-compound auranofin (AF) on both thioredoxin-glutathione reductasa (TGR) activity and viability of Taenia crassiceps cysticerci. It was demonstrated that micromolar concentrations of AF were high enough to fully inhibit TGR and kill the parasites. In this work, the dynamics of changes in the glutathione pool of T. crassiceps cysticerci following the addition of AF, was analyzed. A dose-dependent decrease in the internal glutathione concentration, concomitant with an increase in ROS production was observed. These changes were simultaneous with the formation of glutathione-protein complexes and the export of glutathione disulfide (GSSG) to the culture medium. Incubation of cysticerci in the presence of both AF and N-acetyl cysteine (NAC) prevents all the above changes, maintaining cysticerci viability. By contrast, the presence of both AF and buthionine sulfoximine (BSO) resulted in a potentiation of the effects of the gold compound, jeopardizing cysticerci viability. These results suggest the lethal effect of AF on T. crassiceps cysticerci, observed at micromolar concentrations, can be explained as a consequence of major changes in the glutathione status, which results in a significant increase in the oxidative stress of the parasites.


Subject(s)
Auranofin/toxicity , Glutathione/analysis , Oxidants/toxicity , Oxidative Stress , Taenia/chemistry , Taenia/drug effects , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Buthionine Sulfoximine/metabolism , Cysticercus/chemistry , Cysticercus/drug effects , Cysticercus/physiology , Reactive Oxygen Species/analysis , Survival Analysis , Taenia/physiology
11.
Chem Biol Interact ; 225: 13-20, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25450236

ABSTRACT

Glycyrrhizae radix (G. radix) has been demonstrated to have hepatoprotective properties. This study determined the therapeutic effects of isoliquiritigenin (isoLQ) in G. radix, against liver injury induced by CCl4 in rats. CCl4 (0.5 ml/kg/d, twice) or CCl4 plus buthionine sulfoximine exerted severe liver damage assessed by increased plasma levels of alanine aminotransferase and aspartate aminotransferase, in addition to hepatic degeneration and necrosis. These pathological changes were markedly protected by pretreatment with isoLQ (5, 20 mg/kg/d, p.o.) for 3 consecutive days. In addition, pretreatment with isoLQ inhibited CCl4-induced reduction of cytochrome P450 2E1 protein and mRNA expression as well as activity in the liver. Moreover, isoLQ pretreatment reversed the decrease in hepatic antioxidant capacity induced by CCl4 as well as suppressed expression of tumor necrosis factor-alpha and cyclooxigenase-2 in the liver. These results suggest that isoLQ has a protective effect against CCl4-induced liver damage through induction of antioxidant and anti-inflammatory activities.


Subject(s)
Buthionine Sulfoximine/metabolism , Carbon Tetrachloride/metabolism , Chalcones/pharmacology , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 CYP2E1/metabolism , Enzyme Inhibitors/pharmacology , Alanine Transaminase/blood , Alanine Transaminase/genetics , Animals , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/genetics , Buthionine Sulfoximine/toxicity , Carbon Tetrachloride/toxicity , Chalcones/therapeutic use , Chemical and Drug Induced Liver Injury/enzymology , Enzyme Inhibitors/therapeutic use , Histocytochemistry , Male , RNA, Messenger/chemistry , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
12.
Food Chem Toxicol ; 56: 425-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23500778

ABSTRACT

Here, we investigated whether hirsutenone, a compound isolated from Alnus japonica, was able to attenuate oxidative stress-induced death in transformed retinal ganglion (RGC-5) cells. Hirsutenone effectively protected RGC-5 cells from oxidative insult induced by, l-buthionine-(S,R)-sulfoximine (BSO) plus glutamate in a concentration-dependent manner, as demonstrated by propidium iodide (PI)/Hoechst 33342 double staining, flow cytometry, and MTT assays. Moreover, hirsutenone inhibited the increase in apoptotic protein expression resulting from BSO plus glutamate. Hirsutenone also effectively inhibited sodium nitroprusside (SNP)-induced lipid peroxidation in rat brain homogenates. To investigate the effects of hirsutenone in vivo, we used N-methyl-d-aspartate (NMDA) as a negative insult on the retinas of rats. NMDA affects the thinning of the inner plexiform layer (IPL) and causes an increase in the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive ganglion cells in the ganglion cell layer (GCL). Hirsutenone treatment led to a reduction in NMDA-induced IPL and TUNEL staining of the GCL. In conclusion, hirsutenone isolated from A. japonica may act as neuroprotective agent for conditions such as glaucoma.


Subject(s)
Alnus/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Apoptosis/drug effects , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/metabolism , Catechols/pharmacology , Cell Survival/drug effects , DNA Nucleotidylexotransferase/metabolism , Diarylheptanoids/pharmacology , Glaucoma/drug therapy , Glaucoma/prevention & control , Glutamic Acid/adverse effects , Glutamic Acid/metabolism , In Situ Nick-End Labeling , Lipid Peroxidation/drug effects , Male , N-Methylaspartate/adverse effects , N-Methylaspartate/antagonists & inhibitors , Nitroprusside/adverse effects , Nitroprusside/metabolism , Propidium/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Thiobarbituric Acid Reactive Substances/chemistry
13.
Environ Pollut ; 165: 18-24, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398017

ABSTRACT

The rootless duckweed Wolffia globosa can accumulate and tolerate relatively large amounts of arsenic (As); however, the underlying mechanisms were unknown. W. globosa was exposed to different concentrations of arsenate with or without l-buthionine sulphoximine (BSO), a specific inhibitor of γ-glutamylcysteine synthetase. Free thiol compounds and As(III)-thiol complexes were identified and quantified using HPLC - high resolution ICP-MS - accurate mass ESI-MS. Without BSO, 74% of the As accumulated in the duckweed was complexed with phytochelatins (PCs), with As(III)-PC(4) and As(III)-PC(3) being the main species. BSO was taken up by the duckweed and partly deaminated. The BSO treatment completely suppressed the synthesis of PCs and the formation of As(III)-PC complexes, and also inhibited the reduction of arsenate to arsenite. BSO markedly decreased both As accumulation and As tolerance in W. globosa. The results demonstrate an important role of PCs in detoxifying As and enabling As accumulation in W. globosa.


Subject(s)
Araceae/physiology , Arsenic/metabolism , Phytochelatins/metabolism , Water Pollutants, Chemical/metabolism , Adaptation, Physiological , Arsenic/analysis , Biodegradation, Environmental , Buthionine Sulfoximine/metabolism , Water Pollutants, Chemical/analysis
14.
Biomacromolecules ; 12(6): 2407-15, 2011 Jun 13.
Article in English | MEDLINE | ID: mdl-21557536

ABSTRACT

Novel redox-responsive polyphosphate nanosized assemblies based on amphiphilic hyperbranched multiarm copolyphosphates (HPHSEP-star-PEP(x)) with backbone redox-responsive, good biocompatibility, and biodegradability simultaneously have been designed and prepared successfully. The hydrophobic core and hydrophilic multiarm of HPHSEP-star-PEP(x) are composed of hyperbranched and linear polyphosphates, respectively. Benefiting from the amphiphilicity, HPHSEP-star-PEP(x) can self-assemble into spherical micellar nanoparticles in aqueous media with tunable size from about 70 to 100 nm via adjusting the molecular weight of PEP multiarm. Moreover, HPHSEP-star-PEP(x) micellar structure can be destructed under reductive environment and result in a triggered drug release behavior. The glutathione-mediated intracellular drug delivery was investigated against a HeLa human cervical carcinoma cell line, and the results indicate that doxorubicin-loaded (DOX-loaded) HPHSEP-star-PEP(x) micelles show higher cellular proliferation inhibition against glutathione monoester pretreated HeLa cells than that of the nonpretreated ones. In contrast, the DOX-loaded micelles exhibit lower inhibition against buthionine sulfoximine pretreated HeLa cells. These results suggest that such redox-responsive polyphosphate micelles can rapidly deliver anticancer drugs into the nuclei of tumor cells enhancing the inhibition of cell proliferation and provide a favorable platform to construct excellent drug delivery systems for cancer therapy.


Subject(s)
Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Carriers/chemical synthesis , Drug Delivery Systems/methods , Glutathione/analogs & derivatives , Polyphosphates/chemical synthesis , Surface-Active Agents/chemical synthesis , Uterine Cervical Neoplasms/drug therapy , Animals , Antimetabolites/metabolism , Antimetabolites/pharmacology , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Doxorubicin/metabolism , Drug Carriers/metabolism , Female , Flow Cytometry , Glutathione/metabolism , Glutathione/pharmacology , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Mice , Micelles , NIH 3T3 Cells , Nanoparticles/chemistry , Oxidation-Reduction , Polyphosphates/metabolism , Surface-Active Agents/metabolism , Uterine Cervical Neoplasms/pathology
15.
Cell Immunol ; 270(1): 53-61, 2011.
Article in English | MEDLINE | ID: mdl-21550026

ABSTRACT

The characterization of oxidant (glutathione)-dependent regulation of MAPK(p38/RK)-mediated TNF-α secretion was undertaken in vitro, and the ramifications of the influence of a redox microenvironment were unraveled. Intermittent exposure of alveolar epithelial cells (FATEII) to LPS (endotoxin) transiently and temporally induced the expression of MAPK(p38/RK). This upregulation was associated with the activation of MAPKAP-K(2), manifested by the specific phosphorylation of the downstream heat-shock protein (Hsp)-27. Selective blockading of the MAPK(p38/RK) pathway using the pyridinyl imidazole SB-203580 abrogated the LPS-dependent release of TNF-α. N-acetyl-l-cysteine (NAC), a precursor of glutathione, reduced TNF-α secretion and increased [GSH]. Conversely, l-buthionine-(S,R)-sulfoximine (BSO), an irreversible inhibitor of γ-glutamylcysteine synthetase (γ-GCS), the rate-limiting enzyme in the pathway mediating GSH biosynthesis, augmented the secretion of TNF-α and [GSSG] accumulation. Whereas NAC abrogated the phosphorylation of MAPK(p38/RK), BSO reversibly amplified this effect. Furthermore, intermittent exposure of FATEII cells to the exogenous oxidants X/XO and H(2)O(2) upregulated the secretion of pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α; this upregulation was correlated with increasing activity of key glutathione-related enzymes, closely involved with maintaining the cyclic GSH/GSSG equilibrium. These results indicate that a redox microenvironment plays a major role in regulating MAPK-dependent production of cytokines in the alveolar epithelium.


Subject(s)
Alveolar Epithelial Cells/metabolism , Glutathione Disulfide/metabolism , Pulmonary Alveoli/metabolism , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Enzyme Activation , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/metabolism , Glutathione Disulfide/pharmacology , Humans , Imidazoles/pharmacology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , MAP Kinase Signaling System/drug effects , Oxidation-Reduction , Phosphorylation , Pulmonary Alveoli/cytology , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Int J Oncol ; 38(6): 1731-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21455570

ABSTRACT

The potential of arsenic trioxide (As2O3) as a novel therapy against ovarian cancer has been progressively recognized. Its prospective usefulness for treatment of this malignancy either alone or in combination with other chemotherapeutic agents has been increasingly explored. In this study, we attempted to enhance the cytotoxicity of As2O3 in ovarian cancer cells through manipulation of cellular glutathione (GSH) levels using either buthionine sulfoximine (BSO) or ascorbic acid (AA). Results from our studies showed that combinatorial therapies using As2O3 with either low dose BSO or only pharmacological doses of AA acted synergistically to enhance the cytotoxicity of As2O3 in ovarian tumor cells. With these regimens, therapeutic selectivity was observed with preferential killing of ovarian tumor cells over normal fibroblast controls. Furthermore, contrary to previous reports, enhancement of As2O3-mediated cell killing by these two agents was propagated through different effects. With BSO, apoptotic and non-apoptotic cell death enhancement were mediated through increased arsenic accumulation and GSH depletion that occurred independently of reactive oxygen species. With pharmacological doses of AA, increase in cell death proceeded through non-apoptotic routes via an oxidative stress-related pathway independent of GSH levels. Taken together, these results indicate that GSH depleting agents or pro-oxidative chemicals have capabilities of improving the utility of As2O3 in ovarian cancer management.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Ascorbic Acid/pharmacology , Buthionine Sulfoximine/pharmacology , Ovarian Neoplasms/metabolism , Oxidative Stress/drug effects , Oxides/pharmacology , Apoptosis/drug effects , Arsenic Trioxide , Ascorbic Acid/metabolism , Buthionine Sulfoximine/metabolism , Cell Death/drug effects , Cell Line, Tumor , Drug Synergism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Humans , Intracellular Space/metabolism , Ovarian Neoplasms/drug therapy
17.
Int J Mol Med ; 27(4): 575-81, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21258765

ABSTRACT

Carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP) is an uncoupler of mitochondrial oxidative phosphorylation in eukaryotic cells. In the present study, we investigated the involvement of reactive oxygen species (ROS) and glutathione (GSH) in FCCP-induced As4.1 juxtaglomerular cell death. Intracellular ROS levels were decreased by FCCP at the early time points (10-150 min) and increased at 48 h. FCCP inhibited the activity of Mn-superoxide dismutase (Mn-SOD) via down-regulating its protein expression. Ebselen (an antioxidant) significantly attenuated ROS levels in FCCP-treated cells, but did not prevent FCCP-induced cell death. Moreover, intracellular GSH content was rapidly diminished within 10 min of FCCP treatment, which was accompanied by a reduction of the mitochondrial membrane potential [MMP (∆ψm)]. L-buthionine sulfoximine (BSO, a GSH synthesis inhibitor) significantly augmented As4.1 cell death by FCCP. However, N-acetylcysteine (NAC, a GSH precursor and antioxidant) attenuated GSH depletion, MMP (∆ψm) loss and cell death in FCCP-treated As4.1 cells. In addition, NAC increased Mn-SOD activity and decreased ROS levels in FCCP-treated As4.1 cells. In conclusion, these results suggest that compared to ROS levels, intracellular GSH levels are more closely linked to FCCP-induced apoptosis in As4.1 juxtaglomerular cells.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Glutathione/metabolism , Intracellular Space/metabolism , Juxtaglomerular Apparatus/drug effects , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Catalase/metabolism , Cell Death/drug effects , Cell Line , Juxtaglomerular Apparatus/metabolism , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
18.
Arch Toxicol ; 85(5): 525-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21046361

ABSTRACT

The clinical usefulness of anthracycline antineoplastic drugs is limited by their cardiotoxicity. Its mechanisms have not been fully understood, although the induction of oxidative stress is widely believed to play the principal role. Glutathione is the dominant cellular antioxidant, while glutathione peroxidase (GPx) together with glutathione reductase (GR) constitutes the major enzymatic system protecting the cardiac cells from oxidative damage. Therefore, this study aimed to assess their roles in anthracycline cardiotoxicity. Ten-week intravenous administration of daunorubicin (DAU, 3 mg/kg weekly) to rabbits induced heart failure, which was evident from decreased left ventricular ejection fraction and release of cardiac troponins to circulation. However, no significant changes in either total or oxidized glutathione contents or GR activity were detected in left ventricular tissue of DAU-treated rabbits when compared with control animals. GPx activity in the cardiac tissue significantly increased. In H9c2 rat cardiac cells, 24-h DAU exposure (0.1-10 µM) induced significant dose-dependent toxicity. Cellular content of reduced glutathione was insignificantly decreased, oxidized glutathione and GR activity were unaffected, and GPx activity was significantly increased. Neither buthionine sulfoximine (BSO, glutathione biosynthesis inhibitor) nor 2-oxo-4-thiazolidine-carboxylic acid (OTC, glutathione biosynthetic precursor) had significant effects on DAU cytotoxicity. This contrasted with model oxidative injury induced by hydrogen peroxide, which cytotoxicity was increased by BSO and decreased by OTC. In conclusion, our results suggest that the dysfunction of glutathione antioxidant system does not play a causative role in anthracycline cardiotoxicity.


Subject(s)
Antioxidants/pharmacology , Daunorubicin/adverse effects , Glutathione/pharmacology , Heart Diseases/chemically induced , Animals , Antibiotics, Antineoplastic , Buthionine Sulfoximine/metabolism , Cell Line , Dose-Response Relationship, Drug , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Heart/drug effects , Hydrogen Peroxide/toxicity , Male , Models, Animal , Oxidative Stress/drug effects , Pyrrolidonecarboxylic Acid/metabolism , Rabbits , Rats , Thiazolidines/metabolism
19.
J Biol Chem ; 285(42): 32192-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20682769

ABSTRACT

Splicing of plant organellar transcripts is facilitated by members of a large protein family, the pentatricopeptide repeat proteins. We have identified a pentatricopeptide repeat protein in a genetic screen for mutants resistant to inhibition of root growth by buthionine sulfoximine (BSO), an inhibitor of glutathione synthesis and consequently named BIR6 (BSO-insensitive roots 6). BIR6 is involved in splicing of intron 1 of the mitochondrial nad7 transcript. Loss-of-function mutations in BIR6 result in a strongly reduced accumulation of fully processed nad7 transcript. This affects assembly of Complex I and results in moderate growth retardation. In agreement with disruption of Complex I function, the genes encoding alternative NADH oxidizing enzymes are induced in the mutant, and the mutant plants are less sensitive to mannitol and salt stress. Mutation in the BIR6 gene allowed normal root growth in presence of BSO and strongly attenuated depletion of glutathione content at these conditions. The same phenotype was observed with other mutants affected in function of Complex I, thus reinforcing the importance of Complex I function for cellular redox homeostasis.


Subject(s)
Arabidopsis Proteins/metabolism , Genes, Plant , Introns , Mitochondria/genetics , RNA Splicing , Antimetabolites/metabolism , Arabidopsis/anatomy & histology , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Buthionine Sulfoximine/metabolism , Electron Transport Complex I/metabolism , Glutathione/genetics , Glutathione/metabolism , Mitochondria/metabolism , Plant Roots/drug effects , Plant Roots/genetics , Plant Roots/growth & development , Plant Roots/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
J Exp Biol ; 213(Pt 13): 2225-33, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20543121

ABSTRACT

Carotenoid and melanin pigments in the plumage of birds are hypothesized to be sensitive to oxidative stress. We manipulated oxidative status of captive greenfinches (Carduelis chloris L.) by the administration of buthionine sulfoximine (BSO), a selective inhibitor of the synthesis of glutathione (GSH), an intracellular antioxidant. Half of the birds in the treated group, as well as in the control group, also received dietary carotenoid (lutein) supplementation. BSO treatment reduced erythrocyte GSH levels and caused oxidative damage as indicated by the increased concentration of plasma malondialdehyde (MDA), an end product of lipid peroxidation. BSO treatment also reduced the brightness (i.e. increased blackness) of the tips of tail feathers grown during the experiment. These results show that a low systemic GSH level is required for development of eumelanin plumage coloration and that such a low GSH level is also potentially dangerous for the organism. Carotenoid supplementation increased plasma carotenoid levels and chroma of the yellow parts of the feathers grown during the experiment. However, carotenoid supplementation did not reduce plasma MDA levels. Manipulation of GSH did not affect plasma carotenoids or carotenoid-based plumage coloration. These findings argue against the antioxidant function of lutein in vivo and carotenoid signaling of antioxidant status.


Subject(s)
Feathers/metabolism , Passeriformes/metabolism , Pigmentation , Animals , Buthionine Sulfoximine/metabolism , Glutathione/metabolism , Lutein/metabolism , Male , Malondialdehyde/blood , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL
...