Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
Add more filters

Publication year range
1.
Stem Cells ; 39(2): 170-182, 2021 02.
Article in English | MEDLINE | ID: mdl-33159685

ABSTRACT

Heart transplantation is a life-saving therapy for end-stage organ failure. Organ deterioration during transportation limits storage to 4 hours, limiting hearts available. Approaches ameliorating organ damage could increase the number of hearts acceptable for transplantation. Prior studies show that adipose-derived stem/stromal cell secretome (ASC-S) rescues tissues from postischemic damage in vivo. This study tested whether ASC-S preserved the function of mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes (iCM) exposed to organ transportation and transplantation conditions. Hearts were subjected to cold University of Wisconsin (UW) cardioplegic solution ± ASC-S for 6 hours followed by analysis using the Langendorff technique. In parallel, the effects of ASC-S on the recovery of iCM from UW solution were examined when provided either during or after cold cardioplegia. Exposure of hearts and iCM to UW deteriorated contractile activity and caused cell apoptosis, worsening in iCM as a function of exposure time; these were ameliorated by augmenting with ASC-S. Silencing of superoxide dismutase 3 and catalase expression prior to secretome generation compromised the ASC-S cardiomyocyte-protective effects. In this study, a novel in vitro iCM model was developed to complement a rodent heart model in assessing efficacy of approaches to improve cardiac preservation. ASC-S displays strong cardioprotective activity on iCM either with or following cold cardioplegia. This effect is associated with ASC-S-mediated cellular clearance of reactive oxygen species. The effect of ASC-S on the temporal recovery of iCM function supports the possibility of lengthening heart storage by augmenting cardioplegic transport solution with ASC-S, expanding the pool of hearts for transplantation.


Subject(s)
Cardioplegic Solutions/toxicity , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Organ Preservation Solutions/toxicity , Recovery of Function/physiology , Adenosine/toxicity , Allopurinol/toxicity , Animals , Glutathione/toxicity , Humans , Induced Pluripotent Stem Cells/drug effects , Insulin/toxicity , Isolated Heart Preparation/methods , Male , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Raffinose/toxicity , Recovery of Function/drug effects
2.
Drug Dev Ind Pharm ; 46(5): 717-731, 2020 May.
Article in English | MEDLINE | ID: mdl-32249604

ABSTRACT

Objectives: l-Glutathione (GSH) is an endogenous tripeptide with super antioxidant properties. In this study, preformulation parameters of GSH and its degradation products were fully investigated.Significance: To date, no experimental preformulation data is available for GSH. Therefore, to the author's knowledge, this is the first study to experimentally determine the preformulation parameters of GSH, which can be considered more reliable for further studies.Methods: An HPLC method for GSH was optimized and validated to accurately quantify the GSH amount in solution, used to investigate GSH's solubility and Log P. Differential Scanning Calorimeter and Thermogravimetric Analyzer were used to evaluate the thermal properties of GSH. Polarized microscope and Fourier-transform Infrared Spectroscopy were used to determine GSH's crystal habits and functional groups, respectively. Forced degradation kinetics and the degradation products were investigated and identified by LC-MS, respectively. GSH's cellular cytotoxicity on fibroblasts was investigated by MTT assay.Results: It was determined that GSH has high aqueous solubility (252.7 mg/mL), low Log P (-3.1), a melting endotherm of 195 °C and decomposition at 210°C, negligible moisture content, and a rectangular/cylindrical-shaped crystalline form. Seven degradation products were identified; one of the major degradation products of GSH under different conditions is first order kinetic oxidation into glutathione disulfide. No cytotoxicity was observed when fibroblasts were treated with GSH (0.005-10.000 mg/mL).Conclusions: Precise preformulation parameters of GSH were obtained, and these are imperative for the development and optimization of advanced GSH formulations.


Subject(s)
Chemistry, Pharmaceutical/methods , Cytotoxins/chemistry , Cytotoxins/toxicity , Glutathione/chemistry , Glutathione/toxicity , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chemical Phenomena/drug effects , Cytotoxins/analysis , Dose-Response Relationship, Drug , Drug Compounding/methods , Fibroblasts/drug effects , Fibroblasts/pathology , Glutathione/analysis , Humans , Kinetics , Tandem Mass Spectrometry/methods , X-Ray Diffraction/methods
3.
Drug Metab Dispos ; 47(8): 809-817, 2019 08.
Article in English | MEDLINE | ID: mdl-31097424

ABSTRACT

Vildagliptin (VG), a dipeptidyl peptidase-4 inhibitor, is used for treating type 2 diabetes. On rare occasions, VG causes liver injury as an adverse reaction. One case report suggested the involvement of immune responses in the hepatotoxicity, but the underlying mechanisms are unknown. We recently reported that VG binds covalently in vitro to l-cysteine to produce a thiazoline acid metabolite, M407, implying that the covalent binding may trigger the immune-mediated hepatotoxicity. There was no evidence, however, that such a thiazoline acid metabolite was formed in vivo. In the present study, we administered a single oral dose of VG to male Sprague-Dawley rats, and detected M407 in plasma. The sum of urinary and fecal excretions of M407 reached approximately 2% of the dose 48 hours postdosing. Using bile duct-cannulated rats, we demonstrated that M407 was secreted into bile as a glucuronide, designated as M583. Another newly identified thiazoline metabolite of VG, the cysteinylglycine conjugate M464, was detected in urine, feces, and bile. The formation of M464 was confirmed by in vitro incubation of VG with glutathione even in the absence of metabolic enzymes. A glutathione adduct against the nitrile moiety M611 was also detected in vitro but not in vivo. In summary, we found three new thiazoline-containing thiol adduct metabolites in VG-administered rats. Nonenzymatic covalent binding of VG would likely occur in humans, and it may be relevant to predicting adverse reactions.


Subject(s)
Cysteine/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Glutathione/metabolism , Sulfhydryl Compounds/metabolism , Vildagliptin/pharmacokinetics , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Cysteine/chemistry , Cysteine/toxicity , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Glutathione/chemistry , Glutathione/toxicity , Humans , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/toxicity , Vildagliptin/administration & dosage , Vildagliptin/adverse effects
4.
J Toxicol Environ Health A ; 81(1-3): 37-52, 2018.
Article in English | MEDLINE | ID: mdl-29190187

ABSTRACT

Trichloroethylene (TCE) is a ubiquitous environmental toxicant that is a liver and kidney carcinogen. Conjugation of TCE with glutathione (GSH) leads to formation of nepthrotoxic and mutagenic metabolites postulated to be critical for kidney cancerdevelopment; however, relatively little is known regarding their tissue levels as previous analytical methods for their detection lacked sensitivity. Here, an LC-MS/MS-based method for simultaneous detection of S-(1,2-dichlorovinyl)-glutathione (DCVG), S-(1,2-dichlorovinyl)-L-cysteine (DCVC), and N-acetyl-S-(1,2-dichlorovinyl)-L-cysteine (NAcDCVC) in multiple mouse tissues was developed. This analytical method is rapid, sensitive (limits of detection (LOD) 3-30 fmol across metabolites and tissues), and robust to quantify all three metabolites in liver, kidneys, and serum. The method was used to characterize inter-tissue and inter-strain variability in formation of conjugative metabolites of TCE. Single oral dose of TCE (24, 240 or 800 mg/kg) was administered to male mice from 20 inbred strains of Collaborative Cross. Inter-strain variability in the levels of DCVG, DCVC, and NAcDCVC (GSD = 1.6-2.9) was observed. Whereas NAcDCVC was distributed equally among analyzed tissues, highest levels of DCVG were detected in liver and DCVC in kidneys. Evidence indicated that inter-strain variability in conjugative metabolite formation of TCE might affect susceptibility to adverse health effects and that this method might aid in filling data gaps in human health assessment of TCE.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine/analogs & derivatives , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione/toxicity , Trichloroethylene/metabolism , Trichloroethylene/toxicity , Acetylcysteine/metabolism , Acetylcysteine/toxicity , Animals , Cysteine/metabolism , Cysteine/toxicity , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Signal-To-Noise Ratio , Tissue Distribution
5.
Am J Physiol Renal Physiol ; 313(6): F1200-F1208, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28768661

ABSTRACT

Although all-trans-retinoic acid (ATRA) provides protection against a variety of conditions in vivo, particularly ischemia, the molecular mechanisms underpinning these effects remain unclear. The present studies were designed to assess potential mechanisms by which ATRA affords cytoprotection against renal toxicants in LLC-PK1 cells. Pretreatment of LLC-PK1 cells with ATRA (25 µM) for 24 h afforded cytoprotection against oncotic cell death induced by p-aminophenol (PAP), 2-(glutathion-S-yl)hydroquinone (MGHQ), and iodoacetamide but not against apoptotic cell death induced by cisplatin. Inhibition of protein synthesis with cycloheximide blunted ATRA protection, indicating essential cell survival pathways must be engaged before toxicant exposure to provide cytoprotection. Interestingly, ATRA did not prevent the PAP-induced generation of reactive oxygen species (ROS) nor did it alter glutathione levels. Moreover, ATRA had no significant effect on Nrf2 protein expression, and the Nrf2 inducers sulforaphane and MG132 did not influence ATRA cytoprotection, suggesting cytoprotective pathways beyond those that influence ROS levels contribute to ATRA protection. In contrast, ATRA rapidly (15 min) induced levels of the cellular stress kinases p-ERK and p-AKT at concentrations of ATRA (10 and 25 µM) required for cytoprotection. Consistent with a role for p-ERK in ATRA-mediated cytoprotection, inhibition of p-ERK with PD98059 reduced the ability of ATRA to afford protection against PAP toxicity. Collectively, these data suggest that p-ERK and its downstream targets, independent of ROS and antioxidant signaling, are important contributors to the cytoprotective effects of ATRA against oncotic cell death.


Subject(s)
Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney/drug effects , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology , Aminophenols/toxicity , Animals , Apoptosis/drug effects , Cisplatin/toxicity , Cytoprotection , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Glutathione/analogs & derivatives , Glutathione/toxicity , Iodoacetamide/toxicity , Kidney/enzymology , Kidney/pathology , LLC-PK1 Cells , Necrosis , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Swine , Time Factors
6.
Article in English | MEDLINE | ID: mdl-28418776

ABSTRACT

Many pyrrolizidine alkaloids (PAs) are hepatotoxic, genotoxic, and carcinogenic phytochemicals. Metabolism of PAs in vivo generates four (±)-6,7-dihydro-7-hydroxy-1-hydroxymethyl-5H-pyrrolizine (DHP)-DNA adducts that have been proposed to be responsible for PA-induced liver tumor formation in rats. In this present study, we determined that the same set of DHP-DNA adducts was formed upon the incubation of 7-glutathione-DHP and 7-cysteine-DHP with cultured human hepatocarcinoma HepG2 cells. These results suggest that 7-glutathione-DHP and 7-cysteine-DHP are reactive metabolites of PAs that can bind to cellular DNA to form DHP-DNA adducts in HepG2 cells, and can potentially initiate liver tumor formation.


Subject(s)
Carcinogens/toxicity , Cysteine/analogs & derivatives , Glutathione/analogs & derivatives , Pyrroles/toxicity , Pyrrolizidine Alkaloids/toxicity , Animals , Cysteine/metabolism , Cysteine/toxicity , DNA Adducts , Glutathione/metabolism , Glutathione/toxicity , Pyrrolizidine Alkaloids/metabolism , Rats , Rats, Inbred F344
7.
Mol Carcinog ; 55(8): 1243-50, 2016 08.
Article in English | MEDLINE | ID: mdl-26333016

ABSTRACT

Increased activity of B-Raf has been identified in approximately 7% of human cancers. Treatment of Eker rats (Tsc-2(EK/+) ), bearing a mutation in one allele of the tuberous sclerosis-2 (Tsc-2) gene, with the nephrocarcinogen 2,3,5-tris-(glutathion-S-yl) hydroquinone (TGHQ) results in loss of the wild-type allele of Tsc-2 in renal preneoplastic lesions and tumors. These tumors have increased protein expression of B-Raf, C-Raf (Raf-1), and increased expression and activity of ERK kinase. Similar changes are observed in Raf kinases following TGHQ-mediated transformation of primary renal epithelial cells derived from Tsc-2(EK/+) rats (QTRRE cells), cells that are also null for tuberin. Herein, we utilized LC-MS/MS to identify constitutive phosphorylation of S345 and S483 in both 100- and 95-kDa forms of B-Raf in QTRRE cells. Using microRotofor liquid-phase isoelectric focusing, we identified four fractions of B-Raf that contain different post-translational modification profiles in QTRRE cells. Amplification of the kinase domain of B-Raf from QTRRE cells, outer-stripe of the outer medulla of 8-month TGHQ- or vehicle-treated Tsc-2(+/+) and Tsc-2(EK/+) rats, as well as tumors excised from 8-month TGHQ-treated Tsc-2(EK/+) rats revealed three splice variants of B-Raf within the kinase domain. These splice variants differed by approximately 340, 544, and 600 bp; confirmed by sequencing. No point mutations within the kinase domain of B-Raf were identified. In addition, B-Raf/Raf-1/14-3-3 complex formation in the QTRRE cells was decreased by sorafenib, with concomitant selective decreases in p-ERK levels. Transcriptional and post-translational characterization of critical kinases, such as B-Raf, may contribute to the progression of tuberous sclerosis RCC. (246/250) © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinoma, Renal Cell/metabolism , Glutathione/analogs & derivatives , Hydroquinones/toxicity , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tuberous Sclerosis/metabolism , Animals , Carcinoma, Renal Cell/chemically induced , Carcinoma, Renal Cell/genetics , Cell Line , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Glutathione/toxicity , Humans , Kidney Neoplasms/chemically induced , Kidney Neoplasms/genetics , Male , Neoplasms, Experimental , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins c-raf , RNA Splicing/drug effects , Rats , Tuberous Sclerosis/chemically induced , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
8.
Bioconjug Chem ; 26(9): 1900-7, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26287808

ABSTRACT

Redox-responsive polymersomes were prepared by self-assembly of a hydrophobically modified keratin and employing a water addition/solvent evaporation method. Polyethylene glycol-40 stearate (PEG40ST) was chosen as hydrophobic block to be coupled to keratin via radical grafting. The amphiphilic polymer exhibited low critical aggregation concentration (CAC; 10 µg/mL), indicating a good thermodynamic stability. The polymeric vesicles loaded both hydrophilic methotrexate and hydrophobic curcumin with high entrapment efficiencies, and showed a GSH-dependent drug release rate. Confocal studies on HeLa cells revealed that the obtained polymersomes were efficiently internalized. Biocompatibility properties of the proposed delivery vehicle were assessed in HET-CAM test and Balb-3T3 mouse fibroblasts. Polymersomes loaded with either methotrexate or curcumin inhibited HeLa and CHO-K1 cancer cells proliferation. Overall, the proposed keratin polymersomes could be efficient nanocarriers for chemotherapeutic agents.


Subject(s)
Drug Carriers/chemistry , Drug Liberation , Glutathione/chemistry , Hydrophobic and Hydrophilic Interactions , Intracellular Space/metabolism , Keratins/chemistry , 3T3 Cells , Animals , Biological Transport , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , Drug Carriers/metabolism , Drug Carriers/toxicity , Drug Stability , Glutathione/metabolism , Glutathione/toxicity , HeLa Cells , Humans , Mice , Thermodynamics
9.
J Biol Inorg Chem ; 20(4): 687-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25783495

ABSTRACT

Selenium (Se) is an essential antioxidative micronutrient but can exert cancer-selective cytotoxicity if the nutritional levels are too high. Selenodiglutathione (GSSeSG) is a primary Se metabolite conjugated with two glutathione (GSH) moieties. GSSeSG has been suggested to be an important molecule for cytotoxicity. Here, we propose the underlying mechanisms for the potent cytotoxicity of GSSeSG: cellular intake; reductive metabolism; production of reactive oxygen species; oxidative damage to DNA; apoptosis induction. GSSeSG rather than selenite decreased cell viability and induced apoptosis accompanied by increases in intracellular Se contents. Therefore, GSSeSG-specific cytotoxicity may be ascribed to its preferable incorporation. Base oxidation and strand fragmentation in genomic DNA preceded cell death, suggesting that oxidative stress (including DNA damage) is crucial for GSSeSG cytotoxicity. Strand breaks of purified DNA were caused by the coexistence of GSSeSG and thiols (GSH, cysteine, homocysteine), but not the oxidized form or non-thiol reductants. This implies the important role of intracellular thiols in the mechanism of Se toxicity. GSH-assisted DNA strand breaks were inhibited by specific scavengers for hydrogen peroxide or hydroxyl radicals. The GSSeSG metabolite selenide induced some DNA strand breaks without GSH, whereas elemental Se did so only with GSH. These observations suggest involvement of Fenton-type reaction in the absence of transition metals and reactivation of inert elemental Se. Overall, our results suggest that chemical interactions between Se and the sulfur of thiols are crucial for the toxicity mechanisms of Se.


Subject(s)
Glutathione/analogs & derivatives , Organoselenium Compounds/chemistry , Organoselenium Compounds/pharmacology , Sulfhydryl Compounds/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA/drug effects , DNA/genetics , DNA Damage , Dose-Response Relationship, Drug , Glutathione/chemistry , Glutathione/metabolism , Glutathione/pharmacology , Glutathione/toxicity , Humans , MCF-7 Cells , Organoselenium Compounds/metabolism , Organoselenium Compounds/toxicity , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
10.
Analyst ; 139(24): 6357-60, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25343163

ABSTRACT

CdTe nanocrystals capped by cysteamine were synthesized to study Cr(V)-induced genotoxicity. On the surface of TiO2 thin films, the stepwise process of DNA breakage induced by Cr(V)-GSH complexes was vividly observed by using CdTe-DNA self-assembled fluorescent probes; in acetate buffer solution, an analytical method was developed to detect Cr(V)-induced genotoxicity with CdTe fluorescent probes.


Subject(s)
Cadmium Compounds/chemistry , Chromates/toxicity , DNA Fragmentation/drug effects , Fluorescent Dyes/chemistry , Glutathione/toxicity , Mutagens/toxicity , Nanoparticles/chemistry , Tellurium/chemistry , Animals , Cattle , DNA/ultrastructure , Microscopy, Fluorescence
11.
J Appl Toxicol ; 34(9): 968-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24038001

ABSTRACT

Acetaminophen overdose is the most frequent cause of acute liver injury. The main mechanism of acetaminophen toxicity has been attributed to oxidation of acetaminophen. The oxidation product is very reactive and reacts with glutathione generating acetaminophen-glutathione conjugate (APAP-SG). Although this conjugate has been recognized to be generally nontoxic, we have found recently that APAP-SG could produce a toxic effect. Therefore, the aim of our study was to estimate the toxicity of purified APAP-SG by characterizing the inhibitory effect in human glutathione reductase (GR) and comparing that to the inhibitory effect of the natural inhibitor reduced glutathione. We used two types of human GR: recombinant and freshly purified from red blood cells. Our results show that GR was significantly inhibited in the presence of both APAP-SG and reduced glutathione. For example, the enzyme activity of recombinant and purified GR was reduced in the presence of 4 mm APAP-SG (with 0.5 mm glutathione disulfide) by 28% and 22%, respectively. The type of enzyme inhibition was observed to be competitive in the cases of both APAP-SG and glutathione. As glutathione inhibits GR activity in cells under physiological conditions, the rate of enzyme inhibition ought to be weaker in the case of glutathione depletion that is typical of acetaminophen overdose. Notably, however, enzyme activity likely remains inhibited due to the presence of APAP-SG, which might enhance the pro-oxidative status in the cell. We conclude that our finding could reflect some other pathological mechanism that may contribute to the toxicity of acetaminophen.


Subject(s)
Acetaminophen/analogs & derivatives , Glutathione Reductase/metabolism , Glutathione/toxicity , Acetaminophen/toxicity , Dose-Response Relationship, Drug , Erythrocytes/enzymology , Glutathione Disulfide/metabolism , Glutathione Reductase/antagonists & inhibitors , Humans , Recombinant Proteins/metabolism
12.
Zhonghua Gan Zang Bing Za Zhi ; 22(11): 826-30, 2014 Nov.
Article in Zh | MEDLINE | ID: mdl-25531379

ABSTRACT

OBJECTIVE: To study expression of regucalcin (RGN) and prohibitin (PHB) genes in cirrhotic rat liver and to investigate the related effects of compound glutathione inosine injection (CGII) intervention. METHODS: Forty male Wistar rats were randomly divided into a control group (n=12) and a model group (n=28).The model was established by injecting sterile porcine serum (0.5 mL) into the rat abdominal cavity, twice weekly for 8 consecutive weeks; the control group rats were treated with physiological saline injection (0.5 mL) into the abdominal cavity with the same frequency and time span. During the modeling period, four rats from the model group were randomly selected at different time points to examine changes in liver pathology. Upon pathology confirmation of liver cirrhosis, the porcine serum injection was terminated. The remaining 24 rats in the model group were randomly divided into a fibrosis group and a CGII treatment group.The CGII group received CGII (intramuscular injection of 0.018 mL 100g-1 body weight) once a day for 6 continuous weeks; the fibrosis rats were treated with the same dosage of physiological saline with the same frequency and time span.Liver tissue morphology was examined by both hematoxylin-eosin and Masson's staining. RGN and PHB expression at the mRNA and protein levels in liver tissues were detected by real time RT-PCR and immunohistochemical staining, respectively. RESULTS: Both the mRNA and protein expression levels of RGN and PHB were significantly lower in the liver tissues of the fibrosis group than in the control group.CGII intervention led to significant alleviation of the liver fibrosis severity; moreover, the mRNA and protein expression levels of RGN and PHB were significantly higher than those in the fibrosis group. CONCLUSION: Down-regulation of regucalcin and prohibitin gene expression might contribute to the pathogenesis of liver cirrhosis.


Subject(s)
Calcium-Binding Proteins/genetics , Gene Expression , Glutathione/toxicity , Intracellular Signaling Peptides and Proteins/genetics , Liver Cirrhosis/genetics , Repressor Proteins/genetics , Animals , Carboxylic Ester Hydrolases , Down-Regulation , Inosine , Male , Prohibitins , Rats , Rats, Wistar
13.
J Drugs Dermatol ; 12(11): 1272-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24196336

ABSTRACT

Skin and soft tissue infections (SSTIs) are growing in prevalence in both the outpatient and inpatient settings and are some of the most common diseases seen by dermatologists, who are often the first point of care for these patients. Microbial resistance to antibiotics continues to rise as more virulent strains evolve, and strains predominantly found in the hospital setting are now being seen in the community. Therefore, innovative approaches to combat this trend are needed. Glutathione (GSH) is a well-described and established antioxidant. It participates in detoxification of xenobiotics, regulation of cellular growth, modulation of immune response, and maintenance of the thiol status of proteins and cellular cysteine levels. GSH is also known to have a regulatory effect on immune cells and even inherent antibacterial properties have been reported. To this end, the value of GSH as an antibiotic was evaluated by growing methicillin resistant S. aureus, E. coli, K. pneumoniae and P. aeruginosa strains isolated from human skin and soft tissue infection in the presence of GSH. At a physiologic concentration of 10 mM, GSH had no effect on bacterial growth. At concentrations above 50 mM, which created acidic conditions (pH < 4), bacterial growth was completely inhibited. When adjusted to physiologic pH, GSH exhibited a bacteriostatic effect in a concentration-dependent manner. Additionally, the cytotoxicity of GSH was evaluated in a murine cell line. GSH was relatively non-toxic to murine macrophages, even at the highest concentration tested (160 mM). These results suggest the potential utility of GSH for the prevention and/or as adjunctive treatment of infection, most significantly in disease states associated with GSH deficiency.


Subject(s)
Anti-Bacterial Agents/pharmacology , Glutathione/pharmacology , Skin Diseases, Bacterial/drug therapy , Soft Tissue Infections/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/toxicity , Antioxidants/administration & dosage , Antioxidants/pharmacology , Antioxidants/toxicity , Cell Line , Dose-Response Relationship, Drug , Glutathione/administration & dosage , Glutathione/toxicity , Humans , Hydrogen-Ion Concentration , Macrophages/drug effects , Macrophages/metabolism , Mice , Prevalence , Skin Diseases, Bacterial/microbiology , Soft Tissue Infections/microbiology , Toxicity Tests
14.
Nanomedicine ; 9(2): 257-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22772047

ABSTRACT

Gold nanoparticles are emerging as promising materials from which to construct nanoscale therapeutics and therapeutic delivery systems. However, animal studies have shown that gold nanoparticles modified with certain thiol monolayers such as tiopronin can cause renal complications and morbidity. Although these effects may be eliminated by coadsorbing small amounts of polyethylene glycol (PEG) onto the nanoparticle surface, PEG can also lower cellular internalization efficiency and binding interactions with protein disease targets, significantly reducing the potential for using gold nanoparticles as therapeutics. Using ICP-MS analysis of blood, urine, and several organs, we show in this article that glutathione-coated gold nanoparticles (1.2 nm ± 0.9 nm) cause no morbidity at any concentration up to and including 60 µM and target primary organs although providing gradual dissipation and clearance over time. This study suggests that glutathione may be an attractive alternative to PEG in the design of gold nanoparticle therapeutics. FROM THE CLINICAL EDITOR: This study describes the utility and toxicity of glutathione coated gold nanoparticles in comparison to PEGylated counterparts that are commonly used to increase "Stealth" properties and lower cytotoxicity. Too much PEG on the NPs can lead to lower cellular internalization efficiency and less efficient binding interactions with protein disease targets, significantly reducing the potential for using gold nanoparticles as therapeutics.


Subject(s)
Glutathione/toxicity , Gold/toxicity , Nanoparticles/toxicity , Animals , Erythrocyte Count , Glutathione/blood , Glutathione/chemistry , Glutathione/pharmacokinetics , Gold/blood , Gold/chemistry , Gold/pharmacokinetics , Kidney/drug effects , Kidney/metabolism , Leukocyte Count , Mice , Mice, Inbred BALB C , Nanoparticles/analysis , Nanoparticles/chemistry
15.
Nitric Oxide ; 26(3): 148-56, 2012 Mar 31.
Article in English | MEDLINE | ID: mdl-22326933

ABSTRACT

A comparative study of hypotensive effects of binuclear forms of dinitrosyl iron complexes (DNICs) with glutathione, S-nitrosoglutathione (GS-NO) and sodium nitrite (NaNO(2)) on rats has been carried out. The latter appeared to be the least efficient, viz., mean arterial pressure (MAP) decreased by 10 and 30 mmHg at 25 and 100 µmoles/kg of NaNO(2). In contrast, DNIC and GS-NO produced an appreciable hypotensive effect when used at much lower concentrations. GS-NO reduced MAP to the same extent, viz., to 90 mmHg, on a hundredfold dose scale (from 0.4 up to 50 µmoles/kg) with subsequent restoration of MAP within the next 6-15 min. A similar effect was observed for DNIC except that the amplitude of the MAP drop was lower and the duration of hypotension was essentially greater. DNIC with glutathione were selected as a basic material for pilot-scale production of a hypotensive drug (commercial name Oxacom®). Preliminary pharmacological testing of Oxacom did not establish any adverse or deleterious side effects. Clinical trials of Oxacom® were performed on 14 healthy male volunteers in whom single intravenous infusion of the drug (5mg/kg or 0.2 µmoles/kg of DNIC, respectively) evoked a characteristic response manifested as a 3-4 min drop by 24-27 mmHg of both diastolic and systolic AP with its subsequent slow restoration within the next 8-10h. The heart rate was quickly normalized after an initial increase. Cardiac output was unchanged despite reduced cardiac filling. A comprehensive analysis of clinical and biochemical data failed to establish any significant pathological changes in these parameters. The data obtained suggest that Oxacom® can be recommended for the second phase of clinical trials.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/toxicity , Blood Pressure/drug effects , Ferrous Compounds/pharmacology , Glutathione/analogs & derivatives , Adult , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/blood , Cardiac Output/drug effects , Embryo, Mammalian/drug effects , Female , Ferrous Compounds/adverse effects , Ferrous Compounds/blood , Ferrous Compounds/toxicity , Glutathione/adverse effects , Glutathione/blood , Glutathione/pharmacology , Glutathione/toxicity , Hemostasis/drug effects , Hormones/blood , Humans , Male , Platelet Adhesiveness/drug effects , Pregnancy , Rats , Rats, Wistar , Toxicity Tests
16.
Chem Biol Interact ; 350: 109701, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34656557

ABSTRACT

Acrylamide (AA) is classified as a probable human carcinogen and is ubiquitous in foods processed at high temperatures. The carcinogenicity of AA has been attributed to its active metabolite, glycidamide (GA). Both AA and GA can spontaneously or enzymatically conjugate with glutathione (GSH) to form their corresponding GSH conjugates. Profiling AA-glutathione conjugate (AA-GSH) and GA-glutathione conjugates (2 isomers: GA2-GSH and GA3-GSH) in serum would better illustrate AA detoxification compared with urinary metabolite analysis. However, the lack of AA-, GA2, and GA3-GSH study remains a critical data gap. Our study aimed to investigate the toxicokinetics of AA-, GA2-and GA3-GSH in Sprague Dawley rats treated with 0.1 mg/kg, 1.0 mg/kg, or 5.0 mg/kg AA. Blood samples were collected for LC-MS/MS analysis of the GSH conjugate products. Within 24 h of treatment, we observed rapid formation, elimination, and linear kinetics of AA-, GA2-and GA3-GSH. The ∑GA-GSH AUC/AA-GSH AUC ratios were 0.14-0.29, similar to ∑GA/AA AUC in serum but different from ∑GA/AA-derived urinary mercapturic acids in rodents. Our analysis of AA- and GA-GSHs values represents direct detoxification of AA and GA in vivo. This study advances our understanding of sex and inter-species differences in AA detoxification and may refine the existing kinetic models for a more relevant risk extrapolation.


Subject(s)
Acrylamide/toxicity , Glutathione/analogs & derivatives , Acrylamide/chemistry , Acrylamide/metabolism , Animals , Carcinogens/chemistry , Carcinogens/metabolism , Carcinogens/toxicity , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Female , Glutathione/metabolism , Glutathione/toxicity , Humans , Male , Metabolic Networks and Pathways , Models, Biological , Rats , Rats, Sprague-Dawley , Toxicokinetics
17.
Chem Res Toxicol ; 23(10): 1544-6, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20879737

ABSTRACT

The mutagenicity and carcinogenicity of the important commodity chemical 1,3-butadiene are attributed to the epoxide products. We confirmed our previous work showing that expression of rat glutathione (GSH) transferase 5-5 enhances the mutagenicity of butadiene diepoxide in Salmonella typhimurium TA1535. A GSH-butadiene diepoxide conjugate was isolated and fully characterized by mass spectrometry and nuclear magnetic resonance as S-(2-hydroxy-3,4-epoxybutyl)GSH. The conjugate had a t(½) of 2.6 h (pH 7.4, 37 °C) and was considerably more mutagenic than butadiene diepoxide or monoepoxide in S. typhimurium. We propose that the GSH conjugate may be a major species involved in butadiene genotoxicity, not a detoxication product.


Subject(s)
Butadienes/chemistry , Epoxy Compounds/chemistry , Glutathione/analogs & derivatives , Glutathione/chemistry , Animals , DNA/metabolism , Epoxy Compounds/toxicity , Glutathione/toxicity , Glutathione Transferase/metabolism , Mutagenicity Tests , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism
18.
Physiol Res ; 59(2): 225-232, 2010.
Article in English | MEDLINE | ID: mdl-19537930

ABSTRACT

The aim of the present work was to investigate a new mechanism likely contributing to the toxic action of acetaminophen, especially to explore the possible inhibition of glutathione reductase through an acetaminophen-glutathione conjugate (APAP-SG). APAP-SG conjugate was synthesized by organic synthesis and purified by column chromatography. The inhibitory effect of the conjugate on two types of glutathione reductase (from yeasts and rat hepatocytes) was tested spectro-photometrically. We found that the enzyme activity was reduced similarly after the treatment with 2.96 mM acetaminophen-glutathione conjugate in both yeast and hepatocyte glutathione reductases (GR); the enzyme activity was inhibited to 52.7+/-1.5 % (2.4+/-0.3 mU/ml) in yeast GR (control activity was 5.6+/-0.3 mU/ml) and to 48.1+/-8.8 % (2.2+/-0.2 mU/ml) in rat hepatocytes lysate GR (control activity was 5.2+/-0.2 mU/ml). In addition, the enzyme activity (from hepatocytes lysate) was decreased to 79+/-7 %, 67+/-2 % and 39+/-7 %, in 0.37, 1.48 and 3.7 mM concentration of the conjugate, respectively. We found that glutathione reductase, the essential enzyme of the antioxidant system, was dose-dependently inhibited by the product of acetaminophen metabolism - the conjugate of acetaminophen and glutathione.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Glutathione Reductase/antagonists & inhibitors , Glutathione/toxicity , Hepatocytes/drug effects , Acetaminophen/chemical synthesis , Analgesics, Non-Narcotic/chemical synthesis , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Combinations , Enzyme Activation/drug effects , Glutathione/chemical synthesis , Glutathione Reductase/metabolism , Hepatocytes/enzymology , Hepatocytes/pathology , In Vitro Techniques , Male , Rats , Rats, Wistar
19.
Ecotoxicology ; 19(7): 1193-200, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20535553

ABSTRACT

Coal mining and incineration of solid residues of health services (SRHS) generate several contaminants that are delivered into the environment, such as heavy metals and dioxins. These xenobiotics can lead to oxidative stress overgeneration in organisms and cause different kinds of pathologies, including cancer. In the present study the concentrations of heavy metals such as lead, copper, iron, manganese and zinc in the urine, as well as several enzymatic and non-enzymatic biomarkers of oxidative stress in the blood (contents of lipoperoxidation = TBARS, protein carbonyls = PC, protein thiols = PT, α-tocopherol = AT, reduced glutathione = GSH, and the activities of glutathione S-transferase = GST, glutathione reductase = GR, glutathione peroxidase = GPx, catalase = CAT and superoxide dismutase = SOD), in the blood of six different groups (n = 20 each) of subjects exposed to airborne contamination related to coal mining as well as incineration of solid residues of health services (SRHS) after vitamin E (800 mg/day) and vitamin C (500 mg/day) supplementation during 6 months, which were compared to the situation before the antioxidant intervention (Ávila et al., Ecotoxicology 18:1150-1157, 2009; Possamai et al., Ecotoxicology 18:1158-1164, 2009). Except for the decreased manganese contents, heavy metal concentrations were elevated in all groups exposed to both sources of airborne contamination when compared to controls. TBARS and PC concentrations, which were elevated before the antioxidant intervention decreased after the antioxidant supplementation. Similarly, the contents of PC, AT and GSH, which were decreased before the antioxidant intervention, reached values near those found in controls, GPx activity was reestablished in underground miners, and SOD, CAT and GST activities were reestablished in all groups. The results showed that the oxidative stress condition detected previously to the antioxidant supplementation in both directly and indirectly subjects exposed to the airborne contamination from coal dusts and SRHS incineration, was attenuated after the antioxidant intervention.


Subject(s)
Air Pollutants, Occupational/toxicity , Antioxidants/therapeutic use , Coal Mining , Dietary Supplements , Oxidative Stress , Ascorbic Acid/administration & dosage , Ascorbic Acid/therapeutic use , Biomarkers/blood , Case-Control Studies , Environmental Exposure , Glutathione/blood , Glutathione/toxicity , Glutathione Reductase/blood , Glutathione Reductase/toxicity , Humans , Incineration , Lipid Peroxidation , Metals, Heavy/toxicity , Metals, Heavy/urine , Protein Carbonylation , Superoxide Dismutase/blood , Superoxide Dismutase/toxicity , Thiobarbituric Acid Reactive Substances/toxicity , Vitamin E/administration & dosage , Vitamin E/therapeutic use , alpha-Tocopherol/blood , alpha-Tocopherol/toxicity
20.
Talanta ; 211: 120699, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32070559

ABSTRACT

A progressive aggregation-induced emission (AIE) strategy is established based on two diverse stimulus-responsive patterns of copper nanoclusters (CuNCs) for imaging of aluminum ions (Al3+) in cellular microenvironment. The non-emissive CuNCs were facilely synthesized with l-glutathione (GSH) as both stabilizing agent and reducing agent, and demonstrated the excellent AIE characteristics in the ethanol/water mixture. Moreover, the dispersed CuNCs can be aggregated to give the AIE behavior in aqueous solutions by reducing the pH value, and could be further aggregated with 94-fold reinforce by introducing Al3+ ascribe to the strong coordination ability between Al3+ and the functional groups of GSH, demonstrating the progressive AIE process. Under endocytosis, the progressive AIE strategy can be employed to distinguish the Al3+ in the locations of lysosome against other organelles due to the acidic microenvironment of lysosome. The progressive AIE advantages of CuNCs provide a new concept for signal transduction, and have the promising applications in decoding the functions of intracellular biomolecules.


Subject(s)
Aluminum/metabolism , Cellular Microenvironment , Copper/chemistry , Glutathione/chemistry , Nanostructures/chemistry , Copper/toxicity , Glutathione/toxicity , HeLa Cells , Humans , Lysosomes/metabolism , Microscopy, Confocal , Nanostructures/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL