Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(8): 3299-307, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309284

ABSTRACT

Northern blotting has shown that mouse small intestine contains relatively large amounts of the nuclear factor-E2 p45-related factor (Nrf) 2 transcription factor but relatively little Nrf1. Regulation of intestinal antioxidant and detoxication enzymes by Nrf2 has been assessed using a mouse line bearing a targeted disruption of the gene encoding this factor. Both Nrf2-/- and Nrf2+/+ mice were fed a control diet or one supplemented with either synthetic cancer chemopreventive agents [butylated hydroxyanisole (BHA), ethoxyquin (EQ), or oltipraz] or phytochemicals [indole-3-carbinol, cafestol and kahweol palmitate, sulforaphane, coumarin (CMRN), or alpha-angelicalactone]. The constitutive level of NAD(P)H:quinone oxidoreductase (NQO) and glutathione S-transferase (GST) enzyme activities in cytosols from small intestine was typically found to be between 30% and 70% lower in samples prepared from Nrf2 mutant mice fed a control diet than in equivalent samples from Nrf2+/+ mice. Most of the chemopreventive agents included in this study induced NQO and GST enzyme activities in the small intestine of Nrf2+/+ mice. Increases of between 2.7- and 6.2-fold were observed in wild-type animals fed diets supplemented with BHA or EQ; increases of about 2-fold were observed with a mixture of cafestol and kahweol palmitate, CMRN, or alpha-angelicalactone; and increases of 1.5-fold were measured with sulforaphane. Immunoblotting confirmed that in the small intestine, the constitutive level of NQO1 is lower in the Nrf2-/- mouse, and it also showed that induction of the oxidoreductase was substantially diminished in the mutant mouse. Immunoblotting class-alpha and class-mu GST showed that constitutive expression of most transferase subunits is also reduced in the small intestine of Nrf2 mutant mice. Significantly, induction of class-alpha and class-mu GST by EQ, BHA, or CMRN is apparent in the gene knockout animal. No consistent change in the constitutive levels of the catalytic heavy subunit of gamma-glutamylcysteinyl synthetase (GCS(h)) was observed in the small intestine of Nrf2-/- mice. However, although the expression of GCS(h) was found to be increased dramatically in the small intestine of Nrf2+/+ mice by dietary BHA or EQ, this induction was essentially abolished in the knockout mice. It is apparent that Nrf2 influences both constitutive and inducible expression of intestinal antioxidant and detoxication proteins in a gene-specific fashion. Immunohistochemistry revealed that induction of NQO1, class-alpha GST, and GCS(h) occurs primarily in epithelial cells of the small intestine. This suggests that the variation in inducibility of NQO1, Gsta1/2, and GCS(h) in the mutant mouse is not attributable to the expression of the enzymes in distinct cell types but rather to differences in the dependency of these genes on Nrf2 for induction.


Subject(s)
DNA-Binding Proteins/physiology , Glutathione Transferase/biosynthesis , Intestine, Small/enzymology , Leucine Zippers/physiology , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Diet , Enzyme Induction/drug effects , Erythroid-Specific DNA-Binding Factors , Gene Expression , Glutamate-Cysteine Ligase/biosynthesis , Glutathione Transferase/metabolism , Inactivation, Metabolic , Intestine, Small/drug effects , Leucine Zippers/genetics , Male , Mice , Mice, Knockout , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , NF-E2-Related Factor 2 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/genetics
2.
Carcinogenesis ; 21(10): 1827-34, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023540

ABSTRACT

Certain dietary constituents can protect against chemically induced carcinogenesis in rodents. A principal mechanism by which these chemopreventive compounds exert their protective effects is likely to be via induction of carcinogen detoxification. This can be mediated by conjugation with glutathione, which is synthesized by the sequential actions of glutamate-cysteine ligase (GLCL) and glutathione synthetase. We have demonstrated that dietary administration of the naturally occurring chemopreventive agents, ellagic acid, coumarin or alpha-angelicalactone caused an increase in GLCL activity of between approximately 3- and 5-fold in rat liver. Treatment with the synthetic antioxidant ethoxyquin or the classic inducer phenobarbital caused < 2-fold induction of GLCL activity in rat liver, which was not found to be significant. The increases in GLCL activity were accompanied by increases (between 2- and 4-fold) in levels of both the catalytic heavy subunit (GLCLC) and regulatory light subunit (GLCLR). No substantial induction of GLCL was observed in rat kidney. The glutathione S-transferase (GST) subunits A1, A3, A4, A5, P1 and M1 were all found to be inducible in rat liver by most of the agents. The greatest levels of induction were observed for GST P1, following treatment with coumarin (20-fold), alpha-angelicalactone (10-fold) or ellagic acid (6-fold), and GST A5, following treatment with coumarin (7-fold), alpha-angelicalactone (6-fold) and ethoxyquin (6-fold). Glutathione synthetase was induced approximately 1.5-fold by coumarin, alpha-angelicalactone, ellagic acid and ethoxyquin. The expression of glutathione-related enzymes was also examined in preneoplastic lesions induced in rat liver by aflatoxin B(1). The majority of gamma-glutamyltranspeptidase (GGT)-positive preneoplastic foci contained increased levels of GLCLC relative to the surrounding tissue. This was usually found to be accompanied by an increase in GLCLR. Cells in the inner cortex of rat kidney were found to contain the highest levels of both GLCLC and GLCLR. The same cells showed the strongest staining for GGT activity.


Subject(s)
4-Butyrolactone/analogs & derivatives , Aflatoxin B1/toxicity , Anticarcinogenic Agents/pharmacology , Carcinogens/toxicity , Glutamate-Cysteine Ligase/metabolism , Liver Neoplasms, Experimental/enzymology , Precancerous Conditions/enzymology , 4-Butyrolactone/pharmacology , Aflatoxin B1/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Carcinogens/antagonists & inhibitors , Catalytic Domain/genetics , Coumarins/pharmacology , Diet , Ellagic Acid/pharmacology , Enzyme Induction/drug effects , Ethoxyquin/pharmacology , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Inactivation, Metabolic , Liver/drug effects , Liver/enzymology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Male , Phenobarbital/pharmacology , Precancerous Conditions/chemically induced , Rats , Rats, Inbred F344 , Rats, Wistar
3.
Hepatology ; 32(2): 321-33, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915739

ABSTRACT

Exposure of cells to toxic chemical species can result in reduced glutathione (GSH) depletion, generation of free radicals, and/or binding to critical cell determinants. Chemical stress is usually followed by a concerted cellular response aimed at restoring homeostasis, although the precise initial stimulus for the response is unclear. We have focused on one component of this stress response, the up-regulation of gamma-glutamylcysteine synthetase (gamma-GCS) and the preceding molecular events involved in its regulation in an in vivo mouse model. Male CD-1 mice received buthionine sulphoximine (BSO; 7.2 mmol/kg), diethyl maleate (DEM; 4.2 mmol/kg), paracetamol (APAP; 3.5 and 1.0 mmol/kg), or carbon tetrachloride (CCl(4); 1.0 and 0.2 mmol/kg). Biochemical (serum transaminase and hepatic GSH levels) and molecular (c-jun and c-fos messenger RNA [mRNA] levels and activator protein 1 [AP-1] DNA binding activity) parameters were measured, as well as the consequent effects on gamma-GCS levels and activity. All compounds produced GSH depletion, but only the higher doses of APAP and CCl(4) caused liver damage. DEM, APAP, and CCl(4) increased c-jun and c-fos mRNA levels, together with an increase in AP-1 binding; BSO failed to induce AP-1 despite an increase in c-fos. Interestingly, the effects on gamma-GCS varied markedly according to the compound: BSO and DEM increased gamma-GCS enzyme activity, although only DEM, but not BSO, resulted in an increase in gamma-GCS(h) mRNA and protein. In contrast, APAP and CCl(4) both increased gamma-GCS(h) mRNA and protein; however, there was a marked dose-dependent decrease in gamma-GCS activity. These data indicate that the effect of chemical stress on the liver is compound specific and is not merely dependent on depletion of GSH.


Subject(s)
Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Liver/drug effects , Acetaminophen/toxicity , Animals , Buthionine Sulfoximine/toxicity , Carbon Tetrachloride/toxicity , Genes, fos , Genes, jun , Glutamate-Cysteine Ligase/genetics , Liver/metabolism , Liver/pathology , Male , Maleates/toxicity , Mice , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/metabolism
4.
FEBS Lett ; 467(2-3): 337-40, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10675565

ABSTRACT

gamma-Glutamylcysteine synthetase (GCS) catalyses a critical, rate-limiting step in glutathione synthesis. In this study we describe the isolation and characterisation of a GCS cDNA (pDmGCS4.3. 3) from Drosophila melanogaster by functional complementation of a Saccharomyces cerevisiae gsh1 mutant. Expression of pDmGCS4.3.3 in the yeast mutant partially restored glutathione levels and conferred resistance to methylglyoxal. The pDmGCS4.3.3 cDNA was found to be approx. 4.6 kb in length, containing a 2 kb fragment encoding an open reading frame with a high degree of deduced amino acid sequence identity with previously reported GCS sequences. In situ hybridisation revealed that the Drosophila GCS gene maps to 7D6-9 on the X chromosome.


Subject(s)
Drosophila melanogaster/genetics , Glutamate-Cysteine Ligase/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/chemistry , Drosophila melanogaster/enzymology , Genetic Complementation Test , Glutathione/biosynthesis , Molecular Sequence Data , Mutation , Phylogeny , Plasmids , Saccharomyces cerevisiae/genetics , Sequence Alignment
5.
Free Radic Res ; 31(4): 273-300, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10517533

ABSTRACT

Increases in the intracellular levels of reactive oxygen species (ROS), frequently referred to as oxidative stress, represents a potentially toxic insult which if not counteracted will lead to membrane dysfunction, DNA damage and inactivation of proteins. Chronic oxidative stress has numerous pathological consequences including cancer, arthritis and neurodegenerative disease. Glutathione-associated metabolism is a major mechanism for cellular protection against agents which generate oxidative stress. It is becoming increasingly apparent that the glutathione tripeptide is central to a complex multifaceted detoxification system, where there is substantial inter-dependence between separate component members. Glutathione participates in detoxification at several different levels, and may scavenge free radicals, reduce peroxides or be conjugated with electrophilic compounds. Thus, glutathione provides the cell with multiple defences not only against ROS but also against their toxic products. This article discusses how glutathione biosynthesis, glutathione peroxidases, glutathione S-transferases and glutathione S-conjugate efflux pumps function in an integrated fashion to allow cellular adaption to oxidative stress. Co-ordination of this response is achieved, at least in part, through the antioxidant responsive element (ARE) which is found in the promoters of many of the genes that are inducible by oxidative and chemical stress. Transcriptional activation through this enhancer appears to be mediated by basic leucine zipper transcription factors such as Nrf and small Maf proteins. The nature of the intracellular sensor(s) for ROS and thiol-active chemicals which induce genes through the ARE is described. Gene activation through the ARE appears to account for the enhanced antioxidant and detoxification capacity of normal cells effected by many cancer chemopreventive agents. In certain instances it may also account for acquired resistance of tumours to cancer chemotherapeutic drugs. It is therefore clear that determining the mechanisms involved in regulation of ARE-driven gene expression has enormous medical implications.


Subject(s)
Antioxidants , Glutathione , Oxidative Stress , Animals , Antioxidants/metabolism , Glutathione/metabolism , Humans
6.
Biochim Biophys Acta ; 1446(1-2): 47-56, 1999 Jul 07.
Article in English | MEDLINE | ID: mdl-10395918

ABSTRACT

gamma-Glutamylcysteine synthetase (GCS) is of major importance in glutathione homeostasis. The GCS heterodimer is composed of catalytic (heavy subunit, GCSh) and regulatory (light subunit, GCSl) subunits. Regulation of the human GCSl subunit gene (GLCLR) expression was studied as GCSl has a critical role in glutathione synthesis. The minimal basal expression of GLCLR was found to be mediated by a region between nt -205 and -318. The major transcriptional start site in HT29 cells was located within this region at nt -283. A region between nt -411 and -447 was identified as having a potential involvement in the negative regulation of GLCLR expression. In order to study the transcriptional regulation of GCSl by oxidant stress, HepG2 cells were treated with sodium nitroprusside (SNP). SNP (1.5 mM) was found to increase glutathione levels by 2-fold, as well as GCS activity by 6-fold. This is accompanied by a co-ordinate increase in the levels of the both the GCSl and GCSh subunits, each by approximately 2-fold. The transcriptional activity of the GLCLR gene was increased by approximately 2.5-fold in SNP-treated cells.


Subject(s)
Glutamate-Cysteine Ligase/genetics , Base Sequence , Binding Sites , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/chemistry , Glutamate-Cysteine Ligase/metabolism , HT29 Cells , Humans , Molecular Sequence Data , Nitroprusside/pharmacology , Plasmids , Transcription, Genetic , Tumor Cells, Cultured
7.
Biochem J ; 337 ( Pt 3): 559-66, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9895302

ABSTRACT

gamma-Glutamylcysteine synthetase (GCS) is reported to catalyse the rate-limiting step in glutathione biosynthesis, and is a heterodimer composed of a catalytic subunit [heavy subunit (GCSh) of Mr 73000] and a regulatory subunit [light subunit (GCSl) of Mr 31000]. In the present study, we have demonstrated for the first time a potential role for GCSl in resistance towards doxorubicin and cadmium chloride. Addition of recombinant GCSl to HeLa cell extracts in vitro was found to result in an increase in GCS activity of between 2- and 3-fold. Transient transfections of COS-1 cells with the GCSl cDNA cause an increase in GCS activity of approx. 2-fold, and a small but significant (P=0.008) increase in glutathione levels from 126.9+/-4. 2 nmol/mg protein to 178.8+/-19.1 nmol/mg protein. We proceeded to make a HeLa cell line (LN73), which stably overexpresses GCSl. These cells overexpress GCSl approx. 20-fold above basal levels. LN73 was found to have a 2-fold increase in GCS activity (437.3+/-85.2 pmol/min per mg) relative to the control cell line, HL9 (213.4+/-71. 8 pmol/min per mg). In contrast with the transient transfections in COS-1 cells, stable overexpression of GCSl was found not to be associated with an increase in glutathione content. However, when the LN73 and HL9 cells were treated with the glutathione-depleting agent, diethylmaleate, the LN73 cells were found to have an enhanced ability to regenerate glutathione, compared with HL9 cells. The cell lines were treated with various anti-cancer drugs, and their cytotoxicity was examined. No obvious differences in toxicity were observed between the different cell lines following treatment with cisplatin and melphalan. The redox-cycling agent doxorubicin, however, was found to be more toxic (approx. 2-fold) to the HL9 cells than the LN73 cells. When the cells were treated with the carcinogenic transition-metal compound, cadmium chloride, LN73 cells were found to be approx. 3-fold more resistant than HL9 cells.


Subject(s)
Glutamate-Cysteine Ligase/metabolism , Animals , Antineoplastic Agents/pharmacology , COS Cells , Cadmium Chloride/toxicity , Carcinogens/toxicity , Cell Line , Cell Survival/drug effects , DNA, Complementary/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Glutathione/biosynthesis , HeLa Cells , Humans , Maleates/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
Biochem J ; 336 ( Pt 3): 535-9, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9841862

ABSTRACT

Mutation analysis of putative regulatory elements located in the 5'-flanking region of the gene encoding the regulatory subunit of gamma-glutamylcysteine synthetase (GLCLR) revealed that neither an antioxidant-responsive element (ARE) nor an activator protein-1 (AP-1) site regulates inducible expression by t-butylhydroquinone (tBHQ). The AP-1 site was found to modulate basal expression of GLCLR. A 42 bp region between nucleotides -303 and -344, not containing an ARE, was found to regulate inducible expression of GLCLR by tBHQ.


Subject(s)
Antioxidants/pharmacology , Glutamate-Cysteine Ligase/biosynthesis , Hydroquinones/pharmacology , Base Sequence , DNA/metabolism , Enhancer Elements, Genetic , Enzyme Induction , Glutamate-Cysteine Ligase/genetics , Humans , Molecular Sequence Data , Protein Conformation , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
9.
Br J Haematol ; 99(3): 625-31, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9401076

ABSTRACT

Ineffective haemopoiesis in the myelodysplastic syndromes (MDS) is mediated, at least in part, by apoptosis, though the mechanisms of apoptotic induction are unclear. Tumour necrosis factor-alpha (TNF-alpha) promotes apoptosis via intracellular oxygen free radical production, oxidation of DNA and proteins, and is increasingly implicated in the pathogenesis of MDS. Using single-cell gel electrophoresis, we have identified oxidized pyrimidine nucleotides in the progenitor-enriched bone marrow CD34+ compartment from MDS patients (P=0.039), which are absent in both CD34- MDS cells (P=0.53) and also CD34+ cells from normal subjects (P=0.55). MDS CD34+ blood cells also showed oxidized pyrimidine nucleotides compared with CD34- cells (P=0.029). Within normal subjects no differences were seen between CD34+ and CD34- bone marrow cell compartments. CD34+ bone marrow cell oxidized pyrimidines were strongly associated with elevated plasma TNF-alpha and low bone marrow mononuclear cell glutathione concentrations (5/6 patients) and the inverse relationship was also found (3/4 patients). This data implies a role for intracellular oxygen free radical production, perhaps mediated by TNF-alpha, in the pathogenesis of ineffective haemopoiesis in MDS and provides a rationale for the bone marrow stimulatory effects of antioxidants such as Amifostine in MDS.


Subject(s)
DNA Damage , Myelodysplastic Syndromes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Aged , Aged, 80 and over , Antigens, CD34 , Apoptosis , Enzyme-Linked Immunosorbent Assay , Glutathione/metabolism , Humans , Middle Aged , Myelodysplastic Syndromes/pathology , Oxidation-Reduction , Pyrimidine Nucleosides/metabolism , Sulfhydryl Compounds/metabolism
10.
Biochem J ; 328 ( Pt 1): 99-104, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9359839

ABSTRACT

We have shown that in HepG2 cells treatment with 75 microM t-butylhydroquinone (tBHQ) results in a 2.5-fold increase in glutathione concentration, as part of an adaptive response to chemical stress. In these cells the elevation in intracellular glutathione level was found to be accompanied by an increase of between 2-fold and 3-fold in the level of the 73 kDa catalytic subunit of gamma-glutamylcysteine synthetase (heavy subunit, GCSh) and the 31 kDa regulatory subunit (light subunit, GCSl). Levels of GCSh and GCSl mRNA were increased by up to 5-fold in HepG2 cells in response to tBHQ. To study the transcriptional regulation of GCSl, we subcloned 6.7 kb of the upstream region of the human GCSl gene (GLCLR) from a genomic clone isolated from a P1 lymphoblastoid cell line genomic library. HepG2 cells were transfected with GLCLR promoter reporter constructs and treated with tBHQ. This resulted in an induction of between 1.5-fold and 3.5-fold in reporter activity, indicating that transcriptional regulation of GLCLR is likely to contribute to the induction of GCSl by tBHQ in HepG2 cells. Sequence analysis of the promoter region demonstrated the presence of putative enhancer elements including AP-1 sites and an antioxidant-responsive element, which might be involved in the observed induction of the GLCLR promoter.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Glutamate-Cysteine Ligase/biosynthesis , Liver Neoplasms/enzymology , Amino Acid Sequence , Base Sequence , Carcinoma, Hepatocellular/genetics , Catalysis , Cloning, Molecular , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glutamate-Cysteine Ligase/chemistry , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/isolation & purification , Humans , Hydroquinones/pharmacology , Liver Neoplasms/genetics , Molecular Sequence Data , Promoter Regions, Genetic/physiology , Sequence Analysis, DNA , Tumor Cells, Cultured
11.
IARC Sci Publ ; (139): 175-87, 1996.
Article in English | MEDLINE | ID: mdl-8923030

ABSTRACT

A number of xenobiotics, including the synthetic antioxidant ethoxyquin, inhibit aflatoxin B1 (AFB1)-induced hepatocarcinogenesis in the rat. Two detoxification enzymes that mediate ethoxyquin-induced chemoprotection against AFB1 have been identified by protein purification: a glutathione S-transferase (GST) Yc2 subunit with at least 100-fold greater activity towards AFB1-8,9-epoxide than previously studied transferases, and a unique aldehyde reductase with activity towards the dialdehydic form of AFB1-8,9-dihydrodiol. Molecular cloning has revealed that the Yc2 subunit is a class alpha GST and that the aflatoxin-metabolizing aldehyde reductase (AFAR) is a distant member of the aldo-keto reductase superfamily. Enzyme assay and western blotting have shown that many chemoprotectors, such as ethoxyquin, butylated hydroxyanisole, butylated hydroxytoluene, oltipraz and indole-3-carbinol, that inhibit AFB1-mediated hepatocarcinogenesis induce both GST Yc2 and AFAR. However, western blotting suggests that these enzymes are not always coordinately regulated, as treatment with phenobarbital and beta-naphthoflavone results in differences in the relative increase in hepatic GST Yc2 and AFAR. These findings indicate that GST Yc2 and AFAR represent important resistance mechanisms against AFB1 in the rat. This conclusion is supported by the observation that GST Yc2 and AFAR are overexpressed in rat liver preneoplastic nodules, which display pleiotropic drug resistance.


Subject(s)
Aflatoxin B1/toxicity , Aldehyde Reductase/drug effects , Anticarcinogenic Agents/pharmacology , Carcinogens/toxicity , Glutathione Transferase/drug effects , Aflatoxin B1/pharmacokinetics , Aldehyde Reductase/metabolism , Animals , Biotransformation , Carcinogens/pharmacokinetics , Drug Resistance , Ethoxyquin/pharmacology , Glutathione Transferase/metabolism , Humans , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Rats
12.
Carcinogenesis ; 16(9): 2099-106, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7554060

ABSTRACT

Modulation of cellular thiols has been used to ameliorate the toxic side effects associated with cancer chemotherapy and is currently being investigated as a novel therapeutic strategy in cancer treatment. One of the most extensively studied modulators of thiol levels is N-acetylcysteine (NAC), a cytoprotective drug with multiple therapeutic applications, including use as an adjunct to cancer chemotherapy. Tissue-specific protective effects have previously been observed when NAC has been used in conjunction with chemotherapeutic alkylating agents, but the basis for this was unknown. In view of the contrasting cytoprotective effects of NAC in bladder and bone marrow we examined the effect of this compound on mouse liver, bladder and bone marrow glutathione (GSH) levels, as well as the disposition of 14C-labelled NAC. Radiolabelled NAC was taken up by the majority of tissues at varying rates and levels, except for the brain and spinal cord. The bladder, bone marrow and liver all took up the drug or its metabolites within 15 min of injection. NAC was not found to alter GSH concentrations in the liver, but increased GSH levels in the bladder approximately 2-fold. In contrast, the GSH content of bone marrow was found to decrease by 70-50% after NAC administration. When separate bone marrow cell populations were examined the decrease in GSH was associated with granulocytes, as opposed to lymphocytes, whose GSH levels remained unchanged. These findings provide a possible explanation for the differential cytoprotective effects of NAC.


Subject(s)
Acetylcysteine/pharmacokinetics , Glutathione/metabolism , Acetylcysteine/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells , Carbon Radioisotopes , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred CBA , Tissue Distribution , Urinary Bladder/drug effects , Urinary Bladder/metabolism
13.
Drug Metab Dispos ; 22(3): 392-9, 1994.
Article in English | MEDLINE | ID: mdl-8070315

ABSTRACT

Aflatoxin B1 (AFB1), a metabolite of the grain mold Aspergillus flavus, is a potent hepatocarcinogen and widespread contaminant of human food supplies. AFB1-induced tumors or preneoplastic lesions in experimental animals can be inhibited by cotreatment with several compounds, including indole-3-carbinol (I3C), a component of cruciferous vegetables, and the well-known Ah receptor agonist beta-naphthoflavone (BNF). This study examines the influence of these two agents on the AFB1-glutathione detoxication pathway and AFB1-DNA adduction in rat liver. After 7 days of feeding approximately equally inhibitory doses of I3C (0.2%) or BNF (0.04%) alone or in combination, male Fischer 344 rats were administered [3H]AFB1 (0.5 mg/kg, 480 microCi/kg) intraperitoneally and killed 2 hr later. All three experimental diets inhibited in vivo AFB1-DNA adduction (BNF, 46%; I3C, 68%; combined, 51%). Based on Western blots using antibodies specific for the glutathione S-transferase (GST), subunit Yc2 (subunit 10) appeared to be substantially elevated by the diets containing I3C (I3C diet, 4.0-fold increase in band density; combined diet, 2.8-fold). The BNF diet appeared to elevate Yc2 to a lesser extent (2.2-fold increase in band density).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Aflatoxin B1/analogs & derivatives , Aflatoxin B1/metabolism , Anticarcinogenic Agents/pharmacology , DNA Adducts , DNA/metabolism , Glutathione Transferase/biosynthesis , Glutathione Transferase/drug effects , Indoles/pharmacology , Liver/drug effects , Liver/enzymology , Animals , Benzoflavones/pharmacology , Blotting, Western , Cytosol/enzymology , Enzyme Induction , Glutathione Transferase/metabolism , Isomerism , Liver/metabolism , Macromolecular Substances , Male , Rats , Rats, Inbred F344 , beta-Naphthoflavone
14.
Biochem J ; 300 ( Pt 1): 117-24, 1994 May 15.
Article in English | MEDLINE | ID: mdl-8198522

ABSTRACT

Ingestion of aflatoxin B1 (AFB1) represents a major risk factor in the aetiology of human hepatocellular carcinoma. In the rat, the harmful effects of AFB1 can be prevented by the administration of certain drugs which induce hepatic detoxification enzymes. We have previously shown that treatment of rats with the chemoprotector ethoxyquin (EQ) results in a marked increase in expression of the Alpha-class glutathione S-transferase (GST) Yc2 subunit which has high activity towards AFB1-8,9-epoxide [Hayes, Judah, McLellan, Kerr, Peacock and Neal (1991) Biochem. J. 279, 385-398]. To allow an assessment of whether the increased expression of GST Yc2 represents a general adaptive resistance mechanism to chemical stress, that is invoked by both chemoprotectors and carcinogens, we have examined the effects of EQ, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), phenobarbital (PB), AFB1, 3-methylcholanthrene (3-MC) and clofibrate on the AFB1-glutathione-conjugating activity and the GST subunit levels in rat liver. In addition, the effect of these drugs on the hepatic levels of an aldehyde reductase (AFB1-AR) that metabolizes the cytotoxic dialdehydic form of AFB1 has been studied as this enzyme also appears to be important in chemoprotection. Administration of the antioxidants EQ, BHA or BHT, as well as PB, led to a marked increase in levels of the GST Yc2 subunit in rat liver, and this increase coincided with a substantial rise in the GST activity towards AFB1-8,9-epoxide; neither AFB1, 3-MC nor clofibrate caused induction of Yc2 or any of the GST subunits examined. Among the xenobiotics studied, EQ was found to be the most effective inducing agent for the Yc2 subunit as well as Yc1, Yb1 and Yf. However, PB was equally as effective as EQ in increasing levels of the Ya-type subunits, although it was not found to be as potent an inducer of the other GST subunits, including Yc2. In addition to induction of GST, EQ caused a substantial increase in the hepatic content of AFB1-AR. Both BHA and BHT were also able to induce this enzyme but, by contrast, PB was found to be a poor inducer of AFB1-AR. AFB1, 3-MC and clofibrate were unable to serve as inducers of this reductase. The presence of Alpha-class GST, including the Yc2 subunit, was examined in various rat tissues. Constitutive expression of Yc2 was found in the epididymis at levels comparable with that observed in the liver from EQ-treated rats.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aflatoxin B1/metabolism , Aldehyde Reductase/biosynthesis , Anticarcinogenic Agents/pharmacology , Glutathione Transferase/biosynthesis , Isoenzymes/biosynthesis , Aldehyde Reductase/metabolism , Animals , Butylated Hydroxyanisole/pharmacology , Butylated Hydroxytoluene/pharmacology , Clofibrate/pharmacology , Enzyme Induction/drug effects , Ethoxyquin/pharmacology , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Liver/drug effects , Liver/enzymology , Male , Methylcholanthrene/pharmacology , Phenobarbital/pharmacology , Rats , Rats, Inbred F344 , Tissue Distribution , Xenobiotics/pharmacology
15.
Toxicol Appl Pharmacol ; 125(1): 27-33, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8128492

ABSTRACT

Rat hepatocytes on culturing rapidly lose the ability to metabolize many xenobiotics including the mycotoxin aflatoxin B1 (AFB1). Rat hepatocytes cultured for 5 days on plastic retain < 10% of the initial cytochrome P450 content compared with 34% in cells grown on matrigel alone and 74% in cells cultured on matrigel plus phenobarbital (PB). Although microsomal activation of AFB1 was preserved in matrigel cultured hepatocytes and increased by PB in all the culturing conditions examined, the AFB1 macromolecular binding capacity was reduced to < 10% of that present in the initial cultures. This lack of correlation between cytochrome P450 levels, microsomal activation, and AFB1 binding involved cytosolic glutathione-S-transferase activity (GST). This increased in matrigel cultured cells and was further enhanced by PB. Western blotting of the cytosols showed that the expression of the alpha class GST subunit Yc2 correlated with AFB1-GSH conjugate formation. By contrast, expression of the alpha class GST Ya-type subunit showed no similar close correlation. The expression of pi class GST Yf subunit, which is associated with dedifferentiation in hepatocytes, was suppressed by culturing on matrigel. The results obtained using hepatocytes cultured on matrigel indicate that despite limitations, this comprises a potentially useful model for the in vivo situation.


Subject(s)
Aflatoxin B1/metabolism , Liver/metabolism , Animals , Biocompatible Materials , Biotransformation , Blotting, Western , Cells, Cultured , Collagen , Culture Media , Cytochrome P-450 Enzyme System/metabolism , Drug Combinations , Glutathione Transferase/metabolism , Laminin , Liver/cytology , Male , Microsomes, Liver/metabolism , Phenobarbital/pharmacology , Proteoglycans , Rats , Rats, Inbred F344
16.
Carcinogenesis ; 13(12): 2255-61, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1473231

ABSTRACT

Induction of glutathione S-transferases (GST) by the anticarcinogen butylated hydroxyanisole (BHA) has been examined in lung, kidney and small intestine of male and female BALB/c mice. BHA produced maximal induction of GST in the gut and although it increased GST levels in the kidney, it had little effect on pulmonary GST. Dietary BHA induced Alpha (Ya and Yk), Mu (Yb) and Pi (Yf) class GST subunits at least 10-fold in the small intestine but, by contrast, selenium-dependent glutathione peroxidase activity was reduced by approximately 4-fold in this organ following BHA treatment. In the kidney, all of the GST subunits, apart from Yk in males, showed modest levels of induction by BHA. However, a pronounced sex difference in the expression of renal alpha class subunits in both control and BHA-treated mice was observed, with female mice expressing approximately 4-fold greater levels of Ya and Yk than male mice. All renal GST were localized primarily in the proximal tubules. Dietary BHA was found to have the least inductive effect in the lung, where the GST were localized solely in the bronchi. The pulmonary Mu class GST subunits were induced approximately 2-fold by BHA; the expression of other GST was marginally increased by this inducer. Alpha class GST was also subject to sexual differentiation in the lung with female mice possessing higher levels of Yc and Yk than males. The Ya-type subunit was not detected in the lung nor was it induced by BHA.


Subject(s)
Anticarcinogenic Agents/pharmacology , Butylated Hydroxyanisole/pharmacology , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Intestine, Small/enzymology , Kidney/enzymology , Lung/enzymology , Animals , Electrophoresis, Polyacrylamide Gel , Female , Intestine, Small/drug effects , Kidney/drug effects , Lung/drug effects , Male , Mice , Mice, Inbred BALB C , Sex Factors
17.
Biochem J ; 279 ( Pt 2): 385-98, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-1953636

ABSTRACT

A purification scheme has been devised for two ethoxyquin-inducible Alpha-class glutathione S-transferases (GSTs) which possess at least 25-fold greater activity towards aflatoxin B1 (AFB1)-8,9-epoxide than that exhibited by the GSTs (i.e. F, L, B and AA) that have been described previously. These two enzymes are both heterodimers and both contain a subunit of Mr 25,800. This subunit has been isolated from both of the GST isoenzymes and, after cleavage with CNBr, it has been subjected to automated amino acid sequencing. The primary structure of the Mr 25,800 subunit revealed that it forms part of a subfamily of Alpha-class GSTs which possess closest identity (about 92%) with the Yc subunit of apparent Mr 27,500, which is encoded by the recombinant cDNA clone pGTB42 [Telakowski-Hopkins, Rodkey, Bennett, Lu & Pickett (1985) J. Biol. Chem. 260, 5820-5825]. As these two GSTs possess less than 70% sequence identity with the Ya1 and Ya2 subunits, both of Mr 25,500, the constitutively expressed Yc subunit of Mr 27,500 has been renamed Yc1 and the ethoxyquin-inducible GST of Mr 25,800 has been designated Yc2. Using this nomenclature, the two GSTs with high activity for AFB1-8,9-epoxide are Ya1Yc2 and Yc1Yc2. Although evidence suggests that induction of Yc2 is responsible for the high detoxification capacity of livers from ethoxyquin-treated rats for AFB1-8,9-epoxide, resistance towards AFB1 may be multifactorial in this instance as dietary ethoxyquin also induces the Ya1, Ya2 and Yc1 subunits about 2.2-, 10.9- and 2.7-fold respectively. Besides the induction of GST by ethoxyquin, activity towards AFB1-8,9-epoxide is also elevated in the livers of neonatal rats and in livers that contain preneoplastic nodules. Western blotting experiments show that Yc2 is not present in hepatic cytosol from adult rats fed on normal diets but is expressed in neonatal rat livers and in the livers of adult rats that contain preneoplastic nodules that have arisen as a consequence of consuming diets contaminated with AFB1.


Subject(s)
Aflatoxin B1/analogs & derivatives , Aflatoxin B1/pharmacology , Ethoxyquin/pharmacology , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Liver/enzymology , Aflatoxin B1/metabolism , Aflatoxin B1/pharmacokinetics , Amino Acid Sequence , Animals , Drug Resistance , Enzyme Induction/drug effects , Glutathione Transferase/chemistry , Immunohistochemistry , Isoenzymes/chemistry , Liver/drug effects , Macromolecular Substances , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Rats , Rats, Inbred F344 , Substrate Specificity
18.
Biochem J ; 277 ( Pt 2): 501-12, 1991 Jul 15.
Article in English | MEDLINE | ID: mdl-1859377

ABSTRACT

Induction of glutathione S-transferases (GSTs) is believed to represent an important mechanism whereby butylated hydroxyanisole inhibits chemical carcinogenesis. The soluble hepatic GSTs expressed by mice fed on normal diets are all homodimers comprising Ya3 (Mr 25,800), Yb1 (Mr 26,400) and Yf (Mr 24,800) subunits. In addition to these constitutively expressed GSTs, we have identified enzymes containing Ya1 (Mr 25,600), Ya2 (Mr 25,600), Yb2 (Mr 26,200) and Yb5 (Mr 26,500) subunits from the livers of Balb/c mice fed on diets containing butylated hydroxyanisole (BHA). Gradient affinity elution of GSH-Sepharose has been used to resolve the mouse liver enzymes into several discrete pools of activity from which GSTs were purified by cation-exchange chromatography. The inducible Mu-class Yb2 and Yb5 subunits were separately isolated as the heterodimers GST Yb1Yb2 and GST Yb1Yb5 and their catalytic properties are described; this showed that 1,2-dichloro-4-nitrobenzene and trans-4-phenylbut-3-en-2-one are marker substrates for the mouse Yb1 and Yb2 subunits respectively, but no discriminating model substrate was found that allows the identification of the Yb5 subunit. Individual GST subunits were resolved by reverse-phase h.p.l.c. and their amino acid compositions were determined. Certain subunits (Yb1, Yb2, Yb5 and Yf) were also subjected to automated amino acid sequence analysis, and this demonstrated that the Yb5 subunit has a blocked N-terminus. The mouse Yb1, Yb2 and Yb5 subunits from the major inducible Mu-class heterodimers were cleaved with CNBr and purified peptides from the Yb2 and Yb5 subunits were sequenced. These data show that the Yb2 subunit is distinct from the GSTs that are encoded by the cDNAs that have been cloned from mouse liver cDNA libraries but possesses identity with the protein that is encoded by pmGT2, a cDNA isolated from a mouse fibroblast cell line by Townsend, Goldsmith, Pickett & Cowan [(1989) J. Biol. Chem. 264. 21582-21590]. The sequence data also show that the cDNA encoding the mouse Yb5 subunit has not, to date, been cloned, and the relationship between this subunit and Mu-class GSTs in other species that possess a blocked N-terminus (e.g. rat GST YoYo) is discussed.


Subject(s)
Butylated Hydroxyanisole/pharmacology , Diet , Glutathione Transferase/biosynthesis , Liver/enzymology , Amino Acid Sequence , Animals , Chromatography, Affinity/methods , Chromatography, High Pressure Liquid , Cytosol/enzymology , Enzyme Induction , Female , Glutathione , Glutathione Transferase/isolation & purification , Humans , Isoenzymes/biosynthesis , Isoenzymes/isolation & purification , Liver/drug effects , Macromolecular Substances , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molecular Weight , Peptide Fragments/isolation & purification , Rats , Reference Values , Sequence Homology, Nucleic Acid
19.
Biochem J ; 276 ( Pt 2): 461-9, 1991 Jun 01.
Article in English | MEDLINE | ID: mdl-2049074

ABSTRACT

Liver cytosol from mice fed on a normal diet contains Alpha-class glutathione S-transferase (GST) subunits of Mr 25,800, Mu-class GST subunits of Mr 26,400 and Pi-class GST subunits of Mr 24,800. Feeding female mice with a diet containing the anticarcinogenic antioxidant butylated hydroxyanisole (BHA) causes induction of the constitutively expressed Mu-class and Pi-class subunits. BHA also induces an Alpha-class GST comprising subunits of Mr 25,600, which is not expressed at detectable levels in normal mouse liver [McLellan & Hayes (1989) Biochem. J. 263, 393-402]. Data are now presented that show that administration of the anticarcinogen beta-naphthoflavone (BNF), like BHA, induces the Alpha-class 25,600-Mr subunits but not the constitutive Alpha-class GST with subunits of Mr 25,800. The effects of BNF on expression of hepatic GST were studied in both DBA/2 and C57BL/6 mice; these studies revealed a preferential induction of the Alpha-class 25,600-Mr subunits and of the Pi-class 24,800-Mr subunits in those mice in possession of a functional Ah receptor. The BHA/BNF-inducible Alpha-class GST can be resolved into two separate, non-interconvertible peaks by reverse-phase h.p.l.c. Automated amino acid sequence analysis of CNBr-derived peptides from each of these h.p.l.c.-purified peaks showed that the peaks contained at least two very similar subunits. These have been named Ya1 and Ya2. The amino acid sequence of the Ya1 subunit was compared with sequences deduced from a genomic clone, lambda mYa1 (Daniel, Sharon, Tichauer & Sarid (1987) DNA 6, 317-324], and a cDNA clone, pGT41 [Pearson, Reinhart, Sisk, Anderson & Adler (1988) J. Biol. Chem. 263, 13324-13332]. Our data suggest that the Ya1 subunit represents the subunit encoded by the genomic clone, lambda mYa1. Sequence analysis of the constitutive Alpha-class Ya3 subunit (Mr 25,800) shows that, although it is a member of the same gene family as the Ya1 and Ya2 subunits, it represents a distinct sub-family of Alpha-class GST, containing subunits that are more similar to rat Yc. Our data indicate that, of these Alpha-class GST subunits, the two with Mr 25,600 (Ya1 and Ya2) are selectively induced by BHA or BNF in mouse liver; neither BHA nor BNF induces significantly the GST subunit with Mr 25,800 (Ya3).


Subject(s)
Antioxidants/pharmacology , Benzoflavones/pharmacology , Butylated Hydroxyanisole/pharmacology , Glutathione Transferase/metabolism , Isoenzymes/metabolism , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Cytosol/enzymology , Enzyme Induction , Female , Glutathione Transferase/biosynthesis , Glutathione Transferase/genetics , Glutathione Transferase/isolation & purification , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/isolation & purification , Kinetics , Macromolecular Substances , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Molecular Weight , beta-Naphthoflavone
20.
Pharmacol Ther ; 50(3): 443-72, 1991.
Article in English | MEDLINE | ID: mdl-1754606

ABSTRACT

The harmful effects of Aflatoxin B1 (AFB1) are a consequence of it being metabolized to AFB1-8,9-epoxide, a compound that serves as an alkylating agent and mutagen. The toxicity of AFB1 towards different cells varies substantially; sensitivity can change significantly during development, can be modulated by treatment with xenobiotics and is decreased markedly in preneoplastic lesions as well as in tumors. Three types of resistance, namely intrinsic, inducible and acquired, can be identified. The potential resistance mechanisms include low capacity to form AFB1-8,9-epoxide, high detoxification activity, increase in AFB1 efflux from cells and high DNA repair capacity. Circumstantial evidence exists that amongst these mechanisms the glutathione S-transferases, through their ability to detoxify AFB1-8,9-epoxide, play a major role in determining the sensitivity of cells to AFB1.


Subject(s)
Aflatoxin B1/toxicity , Glutathione Transferase/physiology , Aflatoxin B1/analogs & derivatives , Aflatoxin B1/metabolism , Amino Acid Sequence , Animals , Drug Resistance/physiology , Humans , Liver/enzymology , Liver/metabolism , Molecular Sequence Data , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...