Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Oncogene ; 36(44): 6204-6212, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28692050

ABSTRACT

Homeodomain interacting protein kinase-2 (HIPK2) is a member of the HIPK family of stress-responsive kinases that modulates cell growth, apoptosis, proliferation and development. HIPK2 has several well-characterised tumour suppressor roles, but recent studies suggest it can also contribute to tumour progression, although the underlying mechanisms are unknown. Herein, we have identified novel crosstalk between HIPK2 and the cytoprotective transcription factor NRF2. We show that HIPK2 is a direct transcriptional target of NRF2, identifying a functional NRF2 binding site in the HIPK2 gene locus and demonstrating for the first time a transcriptional mode of regulation for this kinase. In addition, HIPK2 is required for robust NRF2 responsiveness in cells and in vivo. By using both gain-of-function and loss-of-function approaches, we demonstrate that HIPK2 can elicit a cytoprotective response in cancer cells via NRF2. Our results have uncovered a new downstream effector of HIPK2, NRF2, which is frequently activated in human tumours correlating with chemoresistance and poor prognosis. Furthermore, our results suggest that modulation of either HIPK2 levels or activity could be exploited to impair NRF2-mediated signalling in cancer cells, and thus sensitise them to chemotherapeutic drugs.


Subject(s)
Carrier Proteins/genetics , Cell Proliferation/drug effects , Lung Neoplasms/genetics , NF-E2-Related Factor 2/genetics , Protein Serine-Threonine Kinases/genetics , A549 Cells , Animals , Apoptosis/drug effects , Binding Sites , Carrier Proteins/metabolism , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , NF-E2-Related Factor 2/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Oncogene ; 32(32): 3765-81, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-22964642

ABSTRACT

Identification of regulatable mechanisms by which transcription factor NF-E2 p45-related factor 2 (Nrf2) is repressed will allow strategies to be designed that counter drug resistance associated with its upregulation in tumours that harbour somatic mutations in Kelch-like ECH-associated protein-1 (Keap1), a gene that encodes a joint adaptor and substrate receptor for the Cul3-Rbx1/Roc1 ubiquitin ligase. We now show that mouse Nrf2 contains two binding sites for ß-transducin repeat-containing protein (ß-TrCP), which acts as a substrate receptor for the Skp1-Cul1-Rbx1/Roc1 ubiquitin ligase complex. Deletion of either binding site in Nrf2 decreased ß-TrCP-mediated ubiquitylation of the transcription factor. The ability of one of the two ß-TrCP-binding sites to serve as a degron could be both increased and decreased by manipulation of glycogen synthase kinase-3 (GSK-3) activity. Biotinylated-peptide pull-down assays identified DSGIS(338) and DSAPGS(378) as the two ß-TrCP-binding motifs in Nrf2. Significantly, our pull-down assays indicated that ß-TrCP binds a phosphorylated version of DSGIS more tightly than its non-phosphorylated counterpart, whereas this was not the case for DSAPGS. These data suggest that DSGIS, but not DSAPGS, contains a functional GSK-3 phosphorylation site. Activation of GSK-3 in Keap1-null mouse embryonic fibroblasts (MEFs), or in human lung A549 cells that contain mutant Keap1, by inhibition of the phosphoinositide 3-kinase (PI3K)-protein kinase B (PKB)/Akt pathway markedly reduced endogenous Nrf2 protein and decreased to 10-50% of normal the levels of mRNA for prototypic Nrf2-regulated enzymes, including the glutamate-cysteine ligase catalytic and modifier subunits, glutathione S-transferases Alpha-1 and Mu-1, haem oxygenase-1 and NAD(P)H:quinone oxidoreductase-1. Pre-treatment of Keap1(-/-) MEFs or A549 cells with the LY294002 PI3K inhibitor or the MK-2206 PKB/Akt inhibitor increased their sensitivity to acrolein, chlorambucil and cisplatin between 1.9-fold and 3.1-fold, and this was substantially attenuated by simultaneous pre-treatment with the GSK-3 inhibitor CT99021.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , NF-E2-Related Factor 2/physiology , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Kelch-Like ECH-Associated Protein 1 , Mice , NF-E2-Related Factor 2/analysis , NF-E2-Related Factor 2/chemistry , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Structure, Tertiary , Ubiquitination , beta-Transducin Repeat-Containing Proteins/chemistry , p38 Mitogen-Activated Protein Kinases/physiology
3.
Cell Death Differ ; 19(5): 779-87, 2012 May.
Article in English | MEDLINE | ID: mdl-22095276

ABSTRACT

The glial environment is an important determinant of neuronal health in experimental models of neurodegeneration. Specifically, astrocytes have been shown, dependent on context, to be both injurious and protective. Human pluripotent stem cells offer a powerful new system to improve our understanding of the mechanisms underlying astrocyte-mediated neuroprotection. Here, we describe a human embryonic stem cell (HESC)-based system to assess the scope and mechanism of human astrocyte-mediated neuroprotection. We first report the generation of enriched and functional HESC-derived astrocytes, by combining BMP-mediated Smad and LIF-mediated JAK-STAT signalling. These astrocytes promote the protection of HESC-derived neurons against oxidative insults. Moreover, their neuroprotective capacity can be greatly enhanced by treatment with the nuclear factor-erythroid 2-related factor 2 (Nrf2)-activating triterpenoid 1[2-Cyano-3,12-dioxool-eana-1,9(11)-dien-28-oyl] trifluoroethylamide (CDDO(TFEA)). Activation of the transcription factor Nrf2 in human astrocytes by CDDO(TFEA) treatment induced expression of the glutamate-cysteine ligase (GCL) catalytic subunit, leading to enhanced GCL activity and glutathione production, and strong neuroprotection against H(2)O(2). This enhanced neuroprotection was found to be dependent on astrocytic GCL activity, unlike the basal neuroprotection afforded by untreated astrocytes. Direct treatment of HESC-derived neurons with CDDO(TFEA) elicited no induction of Nrf2 target genes, nor any neuroprotection. Thus, human astrocytes can mediate neuroprotection through glutathione-dependent and glutathione-independent mechanisms, and represent a therapeutic target for human disorders associated with neuronal oxidative stress.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Embryonic Stem Cells/cytology , NF-E2-Related Factor 2/metabolism , Animals , Astrocytes/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Mice , Mice, Mutant Strains , NF-E2-Related Factor 2/genetics , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology
4.
Biochem Pharmacol ; 62(11): 1511-9, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11728387

ABSTRACT

The regulation of the aldo-keto reductase AKR7A1 was examined in the livers of male and female rats during development by using Western blots, and its contribution to carbonyl metabolism was assessed by using enzyme assays. Hepatic levels of AKR7A1 are low in fetal rats and rise to a peak at around 6 weeks of age in animals of both sexes. Higher levels of the enzyme are found in adult male rat liver than in adult female rat liver. The reductase, therefore, appears to be subject to sex-specific regulation. The effect of growth hormone in mediating this difference in expression was examined by using hypophysectomized animals whose serum growth hormone levels had been feminized by continuous administration. Results demonstrate that such treatment leads to a reduction in AKR7A1 expression. AKR7A1 was found to be constitutively expressed in rat tissues such as liver, kidney, small intestine, and testis, but it was not detected in nasal mucosa, skeletal muscle, heart, adrenal gland, brain, or spleen. However, AKR7A1 was inducible by the synthetic antioxidant ethoxyquin in liver, kidney, and small intestine, but not in the other tissues examined. These results show that levels of this important detoxication enzyme vary considerably according to age and sex and that dietary antioxidants can also influence its level in several tissues.


Subject(s)
Alcohol Oxidoreductases/metabolism , Aldehyde Reductase/metabolism , Liver/enzymology , Age Factors , Aldo-Keto Reductases , Animals , Female , Gene Expression , Immunohistochemistry , Male , Organ Specificity , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Sex Factors
6.
Cancer Lett ; 174(2): 103-13, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11689285

ABSTRACT

This article provides an overview of the mechanisms by which cancer chemopreventive blocking agents increase the expression of detoxication and antioxidant genes. These agents all appear capable of transcriptionally activating a gene battery that includes NAD(P)H:quinone oxidoreductase, aldo-keto reductases, glutathione S-transferases, gamma-glutamylcysteine synthetase, glutathione synthetase and heme oxygenase. Gene induction occurs through the antioxidant responsive element (ARE), a process that is dependent on the Nuclear Factor-Erythroid 2p45-related factors, Nrf1 and Nrf2. Under basal conditions, these basic region leucine zipper (bZIP) transcription factors are located in the cytoplasm of the cell bound to Keap1, and upon challenge with inducing agents, they are released from Keap1 and translocate to the nucleus. Within the nucleus, Nrf1 and Nrf2 are recruited to the ARE as heterodimers with either small Maf proteins, FosB, c-Jun, JunD, activating transcription factor 2 (ATF2) or ATF4. The role of protein kinases in transducing chemical stress signals to the bZIP factors that affect gene induction through the ARE is discussed.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/prevention & control , Trans-Activators/metabolism , Animals , Antioxidants/metabolism , Humans , Leucine Zippers/physiology , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2 , Neoplasms/metabolism , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Oxidoreductases/metabolism
7.
Biochem J ; 359(Pt 3): 507-16, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672424

ABSTRACT

GSH-dependent prostaglandin D(2) synthase (PGDS) enzymes represent the only vertebrate members of class Sigma glutathione S-transferases (GSTs) identified to date. Complementary DNA clones encoding the orthologous human and rat GSH-dependent PGDS (hPGDS and rPGDS, respectively) have been expressed in Escherichia coli, and the recombinant proteins isolated by affinity chromatography. The purified enzymes were both shown to catalyse specifically the isomerization of prostaglandin (PG) H(2) to PGD(2). Each transferase also exhibited GSH-conjugating and GSH-peroxidase activities. The ability of hPGDS to catalyse the conjugation of aryl halides and isothiocyanates with GSH was found to be less than that of the rat enzyme. Whilst there is no difference between the enzymes with respect to their K(m) values for 1-chloro-2,4-dinitrobenzene, marked differences were found to exist with respect to their K(m) for GSH (8 mM versus 0.3 mM for hPGDS and rPGDS, respectively). Using molecular modelling techniques, amino acid substitutions have been identified in the N-terminal domain of these enzymes that lie outside the proposed GSH-binding site, which may explain these catalytic differences. The tissue-specific expression of PGDS also varies significantly between human and rat; amongst the tissues examined, variation in expression between the two species was most apparent in spleen and bone marrow. Differences in catalytic properties and tissue-specific expression of hPGDS and rPGDS appears to reflect distinct physiological roles for class Sigma GST between species. The evolution of divergent functions for the hPGDS and rPGDS is discussed in the context of the orthologous enzyme from chicken.


Subject(s)
Glutathione Transferase/metabolism , Intramolecular Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Catalysis , Glutathione Transferase/classification , Glutathione Transferase/genetics , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Isoenzymes/metabolism , Lipocalins , Models, Molecular , Molecular Sequence Data , Organ Specificity , Protein Conformation , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Sequence Alignment
8.
Brain Res ; 916(1-2): 229-38, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11597610

ABSTRACT

Elevated levels of oxidative stress or decreased antioxidant defense mechanisms may underlie the regionally increased oxidative damage to brain observed in many neurodegenerative disorders. Phase I detoxification pathways for reactive aldehydes generated from lipid peroxidation include aldehyde dehydrogenases, alcohol dehydrogenases and aldo-keto reductases (AKR). In the present study, we examined the cellular expression of AKR family member, succinic semialdehyde reductase (AKR7A2) that reduces toxic aldehydes as well as catalyzing the biosynthesis of the neuromodulator gamma-hydroxybutyrate (GHB). Our results show that in the cerebral cortex and hippocampus, AKR7A2 is primarily localized to glial cells, astrocytes and microglia. In the midbrain, AKR7A2 was found in glia and neuromelanin-containing neurons of the substantia nigra, and the periaqueductal gray. In sections of cerebral cortex and hippocampus from patients with AD and DLB, AKR7A2 immunoreactivity was elevated in reactive astrocytes and microglial cells. Furthermore, total AKR7A2 protein levels were elevated in the cerebral cortex of patients with AD versus control individuals. Our data suggest that reactive gliosis, as a response to injury, may affect GHB neuromodulatory pathways in neurodegenerative disease and elevate aldehyde detoxification pathways.


Subject(s)
Brain/enzymology , Hydroxybutyrate Dehydrogenase/metabolism , Neurodegenerative Diseases/enzymology , Neuroglia/enzymology , Neurons/enzymology , Up-Regulation/physiology , Aged , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Astrocytes/enzymology , Astrocytes/pathology , Brain/pathology , Brain/physiopathology , Cell Compartmentation/physiology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Hippocampus/embryology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Immunohistochemistry , Lewy Body Disease/enzymology , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Male , Microglia/enzymology , Microglia/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroglia/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Sodium Oxybate/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Cancer Res ; 61(16): 6120-30, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507062

ABSTRACT

The natural indoles 3,3'-diindolylmethane (DIM), ascorbigen (ASG), indole-3-carbinol (I3C), and indolo[3,2-b]carbazole (ICZ), as well as the natural isothiocyanates sulforaphane (SUL), benzyl isothiocyanate (BITC) and phenethyl isothiocyanate (PEITC), all possess cancer chemopreventive properties. It is now shown that DIM, ICZ, SUL, and BITC can each stimulate apoptosis in human colon adenocarcinoma LS-174 and Caco-2 cells. Treatment of LS-174 cells with nontoxic doses of DIM, ASG, I3C, or ICZ affected an increase of up to 21-fold in cytochrome P450 1A1 (CYP1A1). None of these indoles caused an elevation in either aldo-keto reductase 1C1 (AKR1C1) or the gamma-glutamylcysteine synthetase heavy subunit (GCS(h)), but DIM, I3C, and ICZ produced a very modest increase in NAD(P)H:quinone oxidoreductase 1 (NQO1). By contrast, nontoxic doses of SUL, BITC, or PEITC failed to induce expression of CYP1A1 in LS-174 cells, but caused an increase of between 11- and 17-fold in the protein levels of AKR1C1, NQO1, and GCS(h). Treatment of the colon cell line with ICZ or SUL caused increases in the levels of mRNA for CYP1A1, AKR1C1, and NQO1 that were consistent with the enzyme data. Exposure of Caco-2 cells to media containing indoles or isothiocyanates gave similar results to those obtained using LS-174 cells. Evidence is presented that the ability of indoles and isothiocyanates to stimulate either xenobiotic response element- or antioxidant response element-driven gene expression accounts for the two groups of phytochemicals inducing different gene batteries. Pretreatment of LS-174 cells for 24 h with ICZ and SUL before exposure for 24 h to benzo(a)pyrene (BaP) reduced to <20% the number of single-strand DNA breaks produced by the carcinogen. Neither ICZ alone nor SUL alone were able to confer the same degree of protection against DNA damage produced by BaP as they achieved in combination. Similar results were obtained with H(2)O(2) as the genotoxic agent. Together, these phytochemicals may prevent colon tumorigenesis by both stimulating apoptosis and enhancing intracellular defenses against genotoxic agents.


Subject(s)
Apoptosis/drug effects , Colon/drug effects , Colon/enzymology , DNA Damage , Indoles/pharmacology , Isothiocyanates/pharmacology , 20-Hydroxysteroid Dehydrogenases/biosynthesis , 20-Hydroxysteroid Dehydrogenases/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator , Brassicaceae/chemistry , Caco-2 Cells/cytology , Caco-2 Cells/drug effects , Caco-2 Cells/enzymology , Cell Line , Cell Line, Transformed , Colon/cytology , Colonic Neoplasms/prevention & control , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Enzyme Induction/drug effects , FMN Reductase , Gene Expression Regulation, Enzymologic/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Humans , Indoles/toxicity , Isothiocyanates/toxicity , NADH, NADPH Oxidoreductases/biosynthesis , NADH, NADPH Oxidoreductases/genetics , NF-E2-Related Factor 2 , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-maf , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/biosynthesis , Receptors, Aryl Hydrocarbon/genetics , Stimulation, Chemical , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Vegetables/chemistry
10.
Chem Res Toxicol ; 14(6): 727-37, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11409944

ABSTRACT

Oxidation of the mycotoxin aflatoxin (AF) B1 yields the 8,9-epoxide, which nonenzymatically hydrolyzes rapidly to a dihydrodiol that in turn undergoes slow, base-catalyzed ring opening to a dialdehyde [Johnson, W. W., Harris, T. M., and Guengerich F. P. (1996) J. Am. Chem. Soc. 118, 8213-8220]. AFB1 dialdehyde does not bind to DNA but can react with protein lysine groups. One enzyme induced by cancer chemopreventive agents is AFB1 aldehyde reductase (AFAR), which catalyzes the NADPH-dependent reduction of the dialdehyde to a dialcohol. AFB1 dialdehyde is known to convert nonenzymatically to AFB1 dihydrodiol at neutral pH, and we reinvestigated the enzymatic reaction by preparing AFB1 dialdehyde at pH 10 and then used this to initiate reactions (at neutral pH) with rat and human AFAR isozymes. Two monoalcohols were identified as products, and their identities were established by NaB2H4 reduction, chemical cleavage, and mass spectrometry. The monoalcohol corresponding to reduction at C-8 formed first in reactions catalyzed by either the rat or the human AFAR. This C-8 monoalcohol was further reduced to AFB1 dialcohol by AFAR. The other monoalcohol (C-6a) was formed but not reduced to the dialcohol rapidly. Steady-state kinetic parameters were estimated for the reduction of AFB1 dialdehyde by rat and human AFAR to the monoalcohols. The apparent k(cat) and K(m) values were not adequate to rationalize the observed DeltaA(340) spectral changes in a kinetic model. Simulation fitting was done and yielded parameters indicative of greater enzyme efficiency. A survey of 12 human liver cytosol samples showed a variation of 2.3-fold in AFAR activity. Rats treated with AFB1 excreted the dialcohol and a monoalcohol in urine. The results of these studies are consistent with a role of (rat and human) AFAR in protection against AFB1 toxicity.


Subject(s)
Aflatoxin B1/metabolism , Aldehyde Reductase/metabolism , Aflatoxin B1/adverse effects , Aflatoxin B1/pharmacokinetics , Aldehydes/adverse effects , Aldehydes/metabolism , Aldehydes/pharmacokinetics , Animals , Humans , Hydrogen-Ion Concentration , Kinetics , Male , Oxidation-Reduction , Rats , Rats, Inbred F344
11.
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
12.
Anal Chem ; 73(5): 987-96, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11289446

ABSTRACT

Sol-gel chemistry was successfully used for the fabrication of open tubular columns with surface-bonded octadecylsilane (ODS) stationary-phase coating for capillary electrochromatography (OT-CEC). Following column preparations, a series of experiments were performed to investigate the performance of the sol-gel coated ODS columns in OT-CEC. The incorporation of N-octadecyldimethyl[3-(trimethoxysilyl)propyl]ammonium chloride as one of the sol-gel precursors played an important role in the electrochromatographic performance of the prepared columns. This chemical reagent possesses a chromatographically favorable, bonded ODS moiety, in conjunction with three methoxy groups allowing for sol-gel reactivity. In addition, a positively charged nitrogen atom is present in the molecular structure of this reagent and provides a positively charged capillary surface responsible for the reversed electroosmotic flow (EOF) in the columns during CEC operation. Comparative studies involving the EOF within such sol-gel ODS coated and uncoated capillaries were performed using acetonitrile and methanol as the organic modifiers in the mobile phase. The use of a deactivating reagent, phenyldimethylsilane, in the sol-gel solution was evaluated. Efficiency values of over 400,000 theoretical plates per meter were achieved in CEC on a 64 cm x 25 microm i.d. sol-gel ODS open tubular column. Test mixtures of polycyclic aromatic hydrocarbons, benzene derivatives, and aromatic aldehydes and ketones were used to evaluate the CEC performances of both nondeactivated and deactivated open tubular sol-gel columns. The effects of mobile-phase organic modifier contents and pH on EOF in such columns were evaluated. The prepared sol-gel ODS columns are characterized by switchable electroosmotic flow. A pH value of approximately 8.5 was found correspond to the isoelectric point for the prepared sol-gel ODS coatings.

13.
Pharmacology ; 61(3): 154-66, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10971201

ABSTRACT

Two supergene families encode proteins with glutathione S-transferase (GST) activity: the family of soluble enzymes comprises at least 16 genes; the separate family of microsomal enzymes comprises at least 6 genes. These two GST families are believed to exert a critical role in cellular protection against oxidative stress and toxic foreign chemicals. They detoxify a variety of electrophilic compounds, including oxidized lipid, DNA and catechol products generated by reactive oxygen species-induced damage to intracellular molecules. An increasing number of GST genes are being recognized as polymorphic. Certain alleles, particularly those that confer impaired catalytic activity (e.g. GSTM1(*)0, GSTT1(*)0), may be associated with increased sensitivity to toxic compounds. GST polymorphisms may be disease modifying; for example, in subgroups of patients with basal cell carcinoma or bronchial hyper-responsiveness, certain GST appear to exert a statistically significant and biologically relevant impact on disease susceptibility.


Subject(s)
Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Polymorphism, Genetic/genetics , Animals , Humans
14.
Food Chem Toxicol ; 38(8): 707-16, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10908818

ABSTRACT

A physiologically-based pharmacokinetic (PB-PK) model was developed for ethylene dibromide (1,2-dibromoethane, EDB) for rats and humans, partly based on previously published in vitro data (Ploemen et al., 1997). In the present study, this PB-PK model has been validated for the rat. In addition, new data were used for the human class ThetaGST T1-1. Validation experiments are described in order to test the predictive value of kinetics to describe "whole-body" metabolism. For the validation experiments, groups of cannulated rats were dosed orally or intravenously with different doses of EDB. Obtained blood concentration-time curves of EDB for all dosing groups were compared to model predictions. It appeared that metabolism, which previously was assumed to be restricted to the liver, was underestimated. Therefore, we extended the PB-PK model to include all the extrahepatic organs, in which the enzymes involved in EDB metabolism have been detected and quantified. With this extended model, the blood concentrations were much more accurately described compared to the predictions of the "liver-model". Therefore, extrahepatic metabolism was also included in the human model. The present study illustrates the potential application of in vitro metabolic parameters in risk assessment, as well as the use of PB-PK modelling as a tool to understand and predict in vivo data.


Subject(s)
Ethylene Dibromide/pharmacokinetics , Liver/metabolism , Administration, Oral , Animals , Area Under Curve , Biotransformation , Chromatography, Gas , Cytochrome P-450 Enzyme System/metabolism , Ethylene Dibromide/administration & dosage , Ethylene Dibromide/toxicity , Glutathione Transferase/metabolism , Humans , Injections, Intravenous , Liver/drug effects , Male , Models, Biological , Rats , Rats, Wistar , Risk Assessment
15.
Biochem J ; 348 Pt 2: 389-400, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10816434

ABSTRACT

Antiserum raised against human aflatoxin B(1) aldehyde reductase 1 (hAFAR1) has been used to identify a previously unrecognized rat aldo-keto reductase (AKR). This novel enzyme is designated rat aflatoxin B(1) aldehyde reductase 2 (rAFAR2) and it characteristically migrates faster during SDS/PAGE than does the archetypal ethoxyquin-inducible rAFAR protein (now called rAFAR1). Significantly, rAFAR2 is essentially unreactive with polyclonal antibodies raised against rAFAR1. Besides its distinct electrophoretic and immunochemical properties, rAFAR2 appears to be regulated differently from rAFAR1 as it is expressed in most rat tissues and does not appear to be induced by ethoxyquin. Multiple forms of rAFAR2 have been identified. Anion-exchange chromatography on Q-Sepharose, followed by adsorption chromatography on columns of Matrex Orange A and Cibacron Blue, have been employed to purify rAFAR2 from rat liver cytosol. The Q-Sepharose chromatography step resulted in the resolution of rAFAR2 into three peaks of AKR activity, two of which were purified and shown to be capable of catalysing the reduction of 2-carboxybenzaldehyde, succinic semialdehyde, 4-nitrobenzaldehyde and 9,10-phenathrenequinone. The two most highly purified rAFAR2-containing preparations eluted from the Cibacron Blue column were 91 and 98% homogeneous. Analysis of these by SDS/PAGE indicated that the least anionic (peak CBA5) comprised a polypeptide of 37.0 kDa, whereas the most anionic (peak CBA6) contained two closely migrating polypeptides of 36.8 and 37.0 kDa; by contrast, in the present study, rAFAR1 was estimated by SDS/PAGE to be composed of 38.0 kDa subunits. Final purification of the 37 kDa polypeptide in CBA5 and CBA6 was accomplished by reversed-phase HPLC. Partial proteolysis of the two preparations of the 37 kDa polypeptide with Staphylococcus aureus V8 protease yielded fragments of identical size, suggesting that they represent the product of a single gene. Furthermore, the peptide maps from CBA5 and CBA6 differed substantially from that yielded by rAFAR1, indicating that they are genetically distinct from the inducible reductase. A peptide generated by CNBr digestion of the 37 kDa polypeptide from CBA6 was shown by Edman degradation to share 88% sequence identity with residues Tyr(168)-Leu(183) of rAFAR1. This provides evidence that the rat protein identified by its cross-reactivity with anti-hAFAR1 serum is an additional member of the AKR7 family.


Subject(s)
Aldehyde Reductase/isolation & purification , Liver/enzymology , Aldehyde Reductase/chemistry , Aldehyde Reductase/genetics , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Cloning, Molecular , Cytosol/enzymology , Female , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/isolation & purification , Kinetics , Liver Neoplasms, Experimental/enzymology , Male , Molecular Sequence Data , Organ Specificity , Peptide Fragments/chemistry , Rats , Rats, Inbred F344 , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Tumor Cells, Cultured
16.
Biochem Soc Trans ; 28(2): 33-41, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10816095

ABSTRACT

An overview is provided of the cancer chemoprevention actions of phenolic antioxidants and 6-ethoxy-1,2-dihydro-2,2,4-trimethylquinoline (ethoxyquin). These agents principally appear to exert their beneficial effects through induction of phase II drug-metabolizing enzymes such as glutathione S-transferase (GST). The requirement for oxidative metabolism of the synthetic antioxidants to carbonyl-containing compounds, including quinones, in order that they can induce gene expression is discussed. Previous work has shown that the basic leucine zipper transcription factor Nrf2 is involved in induction of GST by the phenolic antioxidant butylated hydroxyanisole (BHA). Evidence is provided from a mouse possessing a targeted disruption of the Nrf2 gene that, in murine liver, the transcription factor regulates basal expression of several class Alpha and class Mu GST subunits, but not class Pi GST. In the Nrf2 knock-out mouse, hepatic induction of class Alpha and class Mu GST by BHA and the synthetic antioxidant ethoxyquin is similarly impaired, suggesting that these agents affect gene activation by a related mechanism. Significantly, residual induction of GST by antioxidants is apparent in the Nrf2 mutant mouse, indicating the existence of an alternative mechanism of gene activation.


Subject(s)
Antioxidants/pharmacology , Butylated Hydroxyanisole/pharmacology , DNA-Binding Proteins/metabolism , Ethoxyquin/pharmacology , Glutathione Transferase/biosynthesis , Liver/metabolism , Trans-Activators/metabolism , Animals , Blotting, Western , DNA-Binding Proteins/genetics , Enzyme Induction , Male , Mice , Mice, Mutant Strains , NF-E2-Related Factor 2 , Trans-Activators/genetics , Transcription Factors/metabolism
17.
Cancer Res ; 60(4): 957-69, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706111

ABSTRACT

Structurally diverse compounds can confer resistance to aflatoxin B1 (AFB1) hepatocarcinogenesis in the rat. Treatment with either phytochemicals [benzyl isothiocyanate, coumarin (CMRN), or indole-3-carbinol] or synthetic antioxidants and other drugs (butylated hydroxyanisole, diethyl maleate, ethoxyquin, beta-naphthoflavone, oltipraz, phenobarbital, or trans-stilbene oxide) has been found to increase hepatic aldo-keto reductase activity toward AFB1-dialdehyde and glutathione S-transferase (GST) activity toward AFB1-8,9-epoxide in both male and female rats. Under the conditions used, the natural benzopyrone CMRN was a major inducer of the AFB1 aldehyde reductase (AFAR) and the aflatoxin-conjugating class-alpha GST A5 subunit in rat liver, causing elevations of between 25- and 35-fold in hepatic levels of these proteins. Induction was not limited to AFAR and GSTA5: treatment with CMRN caused similar increases in the amount of the class-pi GST P1 subunit and NAD(P)H: quinone oxidoreductase in rat liver. Immunohistochemistry demonstrated that the overexpression of AFAR, GSTA5, GSTP1, and NAD(P)H:quinone oxidoreductase affected by CMRN is restricted to the centrilobular (periacinar) zone of the lobule, sometimes extending almost as far as the portal tract. This pattern of induction was also observed with ethoxyquin, oltipraz, and trans-stilbene oxide. By contrast, induction of these proteins by beta-naphthoflavone and diethyl maleate was predominantly periportal. Northern blotting showed that induction of these phase II drug-metabolizing enzymes by CMRN was accompanied by similar increases in the levels of their mRNAs. To assess the biological significance of enzyme induction by dietary CMRN, two intervention studies were performed in which the ability of the benzopyrone to inhibit either AFB1-initiated preneoplastic nodules (at 13 weeks) or AFB1-initiated liver tumors (at 50 weeks) was investigated. Animals pretreated with CMRN for 2 weeks prior to administration of AFB1, and with continued treatment during exposure to the carcinogen for a further 11 weeks, were protected completely from development of hepatic preneoplastic lesions by 13 weeks. In the longer-term dietary intervention, treatment with CMRN before and during exposure to AFB1 for a total of 24 weeks was found to significantly inhibit the number and size of tumors that subsequently developed by 50 weeks. These data suggest that consumption of a CMRN-containing diet provides substantial protection against the initiation of AFB1 hepatocarcinogenesis in the rat.


Subject(s)
Aflatoxin B1/toxicity , Aldehyde Reductase/biosynthesis , Coumarins/pharmacology , Glutathione Transferase/biosynthesis , Liver Neoplasms, Experimental/prevention & control , Quinone Reductases/biosynthesis , Animals , Enzyme Induction/drug effects , Female , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Male , Precancerous Conditions/prevention & control , Rabbits , Rats , Rats, Inbred F344 , Sex Factors
18.
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
19.
Biochem J ; 343 Pt 2: 487-504, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10510318

ABSTRACT

Complementary DNA clones encoding human aflatoxin B(1) aldehyde reductase (AKR7A2), aldehyde reductase (AKR1A1), aldose reductase (AKR1B1), dihydrodiol dehydrogenase 1 (AKR1C1) and chlordecone reductase (AKR1C4) have been expressed in Escherichia coli. These members of the aldo-keto reductase (AKR) superfamily have been purified from E. coli as recombinant proteins. The recently identified AKR7A2 was shown to differ from the AKR1 isoenzymes in being able to catalyse the reduction of 2-carboxybenzaldehyde. Also, AKR7A2 was found to exhibit a narrow substrate specificity, with activity being restricted to succinic semialdehyde (SSA), 2-nitrobenzaldehyde, pyridine-2-aldehyde, isatin, 1,2-naphthoquinone (1,2-NQ) and 9,10-phenanthrenequinone. In contrast, AKR1A1 reduces a broad spectrum of carbonyl-containing compounds, displaying highest specific activity for SSA, 4-carboxybenzaldehyde, 4-nitrobenzaldehyde, pyridine-3-aldehyde, pyridine-4-aldehyde, 4-hydroxynonenal, phenylglyoxal, methylglyoxal, 2,3-hexanedione, 1, 2-NQ, 16-ketoestrone and d-glucuronic acid. Comparison between the kinetic properties of AKR7A2 and AKR1A1 showed that both recombinant enzymes exhibited roughly similar k(cat)/K(m) values for SSA, 1,2-NQ and 16-ketoestrone. Many of the compounds which are substrates for AKR1A1 also serve as substrates for AKR1B1, though the latter enzyme was shown to display a specific activity significantly less than that of AKR1A1 for most of the aromatic and aliphatic aldehydes studied. Neither AKR1C1 nor AKR1C4 was found to possess high reductase activity towards aliphatic aldehydes, aromatic aldehydes, aldoses or dicarbonyls. However, unlike AKR1A1 and AKR1B1, both AKR1C1 and AKR1C4 were able to catalyse the oxidation of 1-acenaphthenol and, in addition, AKR1C4 could oxidize di- and tri-hydroxylated bile acids. Specific antibodies raised against AKR7A2, AKR1A1, AKR1B1, AKR1C1 and AKR1C4 have been used to show the presence of all of the reductases in human hepatic cytosol; the levels of AKR1B1 and AKR1C1 were markedly elevated in livers with alcohol-associated injury, and indeed AKR1B1 was only detectable in livers with evidence of alcoholic liver disease. Western blotting of extracts from brain, heart, kidney, liver, lung, prostate, skeletal muscle, small intestine, spleen and testis showed that AKR7A2 is present in all of the organs examined, and AKR1B1 is similarly widely distributed in human tissues. These experiments revealed however, that the expression of AKR1A1 is restricted primarily to brain, kidney, liver and small intestine. The AKR1C family members proved not to be as widely expressed as the other reductases, with AKR1C1 being observed in only kidney, liver and testis, and AKR1C4 being found in liver alone. As human kidney is a rich source of AKR, the isoenzymes in this organ have been studied further. Anion-exchange chromatography of human renal cytosol on Q-Sepharose allowed resolution of AKR1A1, AKR1B1, AKR1C1 and AKR7A2, as identified by substrate specificity and Western blotting. Immunohistochemistry of human kidney demonstrated that AKR7A2 is expressed in a similar fashion to the AKR1 family members in proximal and distal convoluted renal tubules. Furthermore, both AKR7A2 and AKR1 members were expressed in renal carcinoma cells, suggesting that these groups of isoenzymes may be engaged in related physiological functions.


Subject(s)
Alcohol Oxidoreductases/metabolism , Aldehyde Reductase/metabolism , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Aldehyde Reductase/chemistry , Aldehyde Reductase/genetics , Blotting, Western , Catalysis , Cloning, Molecular , Drug Resistance , Escherichia coli/genetics , Humans , Immunohistochemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney/enzymology , Kinetics , Liver/enzymology , Molecular Sequence Data , Molecular Weight , Organ Specificity , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Thermodynamics
20.
J Biol Chem ; 274(42): 30215-21, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10514513

ABSTRACT

Following tissue injury, a fibrin network formed at the wound site serves as a scaffold supporting the early migration of stromal cells needed for wound healing. Growth factors such as insulin-like growth factor-I (IGF-I) concentrate in wounds to stimulate stromal cell function and proliferation. The ability of IGF-binding proteins (IGFBPs) such as IGFBP-3 to reduce the rate of IGF-I clearance from wounds suggests that IGFBP-3 might bind directly to fibrinogen/fibrin. Studies presented here show that IGFBP-3 does indeed bind to fibrinogen and fibrin immobilized on immunocapture plates, with K(d) values = 0.67 and 0.70 nM, respectively, and competitive binding studies suggest that the IGFBP-3 heparin binding domain may participate in this binding. IGF-I does not compete for IGFBP-3 binding; instead, IGF-I binds immobilized IGFBP-3.fibrinogen and IGFBP-3.fibrin complexes with affinity similar to that of IGF-I for the type I IGF receptor. In the presence of plasminogen, most IGFBP-3 binds directly to fibrinogen, although 35-40% of the IGFBP-3 binds to fibrinogen-bound plasminogen. IGFBP-3 also binds specifically to native fibrin clots, and addition of exogenous IGFBP-3 increases IGF-I binding. These studies suggest that IGF-I can concentrate at wound sites by binding to fibrin-immobilized IGFBP-3, and that the lower IGF affinity of fibrin-bound IGFBP-3 allows IGF-I release to type I IGF receptors of stromal cells migrating into the fibrin clot.


Subject(s)
Fibrin/metabolism , Fibrinogen/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Iodine Radioisotopes , Plasminogen/metabolism , Protein Binding , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL