Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 105(2): 246-54, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21712828

ABSTRACT

BACKGROUND: The epidermal growth factor receptor-targeted monoclonal antibody cetuximab (Erbitux) was recently introduced for the treatment of metastatic colorectal cancer. Treatment response is dependent on Kirsten-Ras (K-Ras) mutation status, in which the majority of patients with tumour-specific K-Ras mutations fail to respond to treatment. Mutations in the oncogenes B-Raf and PIK3CA (phosphoinositide-3-kinase) may also influence cetuximab response, highlighting the need for a sensitive, accurate and quantitative assessment of tumour mutation burden. METHODS: Mutations in K-Ras, B-Raf and PIK3CA were identified by both dideoxy and quantitative pyrosequencing-based methods in a cohort of unselected colorectal tumours (n=102), and pyrosequencing-based mutation calls correlated with various clinico-pathological parameters. RESULTS: The use of quantitative pyrosequencing-based methods allowed us to report a 13.7% increase in mutation burden, and to identify low-frequency (<30% mutation burden) mutations not routinely detected by dideoxy sequencing. K-Ras and B-Raf mutations were mutually exclusive and independently associated with a more advanced tumour phenotype. CONCLUSION: Pyrosequencing-based methods facilitate the identification of low-frequency tumour mutations and allow more accurate assessment of tumour mutation burden. Quantitative assessment of mutation burden may permit a more detailed evaluation of the role of specific tumour mutations in the pathogenesis and progression of colorectal cancer and may improve future patient selection for targeted drug therapies.


Subject(s)
Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Mutational Analysis/methods , Individuality , Mutation , Oncogenes/genetics , Adult , Aged , Aged, 80 and over , Carcinoma/pathology , Carcinoma/surgery , Cohort Studies , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation/physiology , Sequence Analysis, DNA/methods
2.
Toxicol Lett ; 186(1): 18-21, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19022363

ABSTRACT

Although toxicogenomics originated as a field of primarily preclinical investigation, a variety of genomic approaches can also be employed during or after clinical development to identify biomarkers linked to drug exposure and/or drug safety. Comparing and contrasting the different pharmacogenomic approaches according to their scale (targeted, focused or exploratory) illustrates the potential utility of each type of strategy in characterizing the genetic determinants that may play roles in various aspects of drug activity. Examples of targeted ADME genotyping, focused SNP panels, and exploratory whole genome association studies are briefly reviewed to provide an overview of the range of pharmacogenetic options available to the research community to support the ongoing efforts to identify biomarkers predictive of drug exposure and/or safety in human subjects.


Subject(s)
Biomedical Research/methods , Drug-Related Side Effects and Adverse Reactions , Toxicogenetics/methods , Animals , Biomarkers/metabolism , Drug Evaluation, Preclinical , Drug Industry , Gene Expression/drug effects , Genetic Variation , Humans , Pharmaceutical Preparations/metabolism , Treatment Outcome
3.
Biochemistry ; 40(36): 10901-10, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11535067

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are metabolized to trans-dihydrodiol proximate carcinogens by human epoxide hydrolase (EH) and CYP1A1. Human dihydrodiol dehydrogenase isoforms (AKR1C1-AKR1C4), members of the aldo-keto reductase (AKR) superfamily, activate trans-dihydrodiols by converting them to reactive and redox-active o-quinones. We now show that the constitutively and widely expressed human AKR, aldehyde reductase (AKR1A1), will oxidize potent proximate carcinogen trans-dihydrodiols to their corresponding o-quinones. cDNA encoding AKR1A1 was isolated from HepG2 cells, overexpressed in Escherichia coli, purified to homogeneity, and characterized. AKR1A1 oxidized the potent proximate carcinogen (+/-)-trans-7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene with a higher utilization ratio (V(max)/K(m)) than any other human AKR. AKR1A1 also displayed a high V(max)/K(m) for the oxidation of 5-methylchrysene-7,8-diol, benz[a]anthracene-3,4-diol, 7-methylbenz[a]anthracene-3,4-diol, and 7,12-dimethylbenz[a]anthracene-3,4-diol. AKR1A1 displayed rigid regioselectivity by preferentially oxidizing non-K-region trans-dihydrodiols. The enzyme was stereoselective and oxidized 50% of each racemic PAH trans-dihydrodiol tested. The absolute stereochemistries of the reactions were assigned by circular dichroism spectrometry. AKR1A1 preferentially oxidized the metabolically relevant (-)-benzo[a]pyrene-7(R),8(R)-dihydrodiol. AKR1A1 also preferred (-)-benz[a]anthracene-3(R),4(R)-dihydrodiol, (+)-7-methylbenz[a]anthracene-3(S),4(S)-dihydrodiol, and (-)-7,12-dimethylbenz[a]anthracene-3(R),4(R)-dihydrodiol. The product of the AKR1A1-catalyzed oxidation of (+/-)-trans-7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene was trapped with 2-mercaptoethanol and characterized as a thioether conjugate of benzo[a]pyrene-7,8-dione by LC/MS. Multiple human tissue expression array analysis showed coexpression of AKR1A1, CYP1A1, and EH, indicating that trans-dihydrodiol substrates are formed in the same tissues in which AKR1A1 is expressed. The ability of this general metabolic enzyme to divert trans-dihydrodiols to o-quinones suggests that this pathway of PAH activation may be widespread in human tissues.


Subject(s)
Aldehyde Reductase/metabolism , Carcinogens/pharmacokinetics , Quinones/pharmacokinetics , Biotransformation , Cloning, Molecular , Cytochrome P-450 CYP1A1/metabolism , Epoxide Hydrolases/metabolism , Escherichia coli , Humans , Isoenzymes/metabolism , Models, Chemical , Oxidation-Reduction , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Recombinant Proteins/metabolism , Substrate Specificity
4.
Drug Metab Dispos ; 29(9): 1243-50, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502735

ABSTRACT

The acceleration of drug discovery due to combinatorial chemistry and high-throughput screening methods has increased the numbers of candidate pharmaceuticals entering the drug development phase, and the capability to accurately predict whether drug candidates will induce various members of the drug-metabolizing cytochrome P450 (CYP) enzyme superfamily is currently of great interest to the pharmaceutical industry. In the present study, we describe the rapid and reliable analysis of CYP induction in a readily obtained model system (cultured rat hepatocytes) using both real-time quantitative reverse transcription-polymerase chain reaction (real-time RT-PCR) and the RNA invasive cleavage assay. The levels of members in the three primary inducible rat CYP subfamilies (CYP1A1, CYP2B1/2, and CYP3A1) were analyzed in untreated and induced (beta-naphthoflavone, phenobarbital, and hydrocortisone) hepatocyte cultures under various media conditions to screen for optimal CYP induction profiles. The fold inductions measured by real-time RT-PCR and the RNA invasive cleavage assay were also compared with enzyme activity measurements in parallel cultures using liquid chromatography/double mass spectrometry-based assays, and the sensitivity and the specificity of the two RNA analysis methods were compared. Using these techniques, various culture conditions were examined for optimizing induction of the three CYP subfamily members. Both real-time RT-PCR and the RNA invasive cleavage assay prove to be effective methods for determining the effects of drugs on specific CYPs in primary rat hepatocytes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Hepatocytes/enzymology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Animals , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Enzyme Induction , Male , Mass Spectrometry , Protein Folding , Rats , Rats, Sprague-Dawley
5.
Chem Biol Interact ; 130-132(1-3): 815-24, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11306097

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are metabolized to trans-dihydrodiol proximate carcinogens by CYP1A1 and epoxide hydrolase (EH). CYP1A1 or aldo-keto reductases (AKRs) from the 1C subfamily can further activate the trans-dihydrodiols by forming either anti-diol-epoxides or reactive and redox active o-quinones, respectively. To determine whether other AKR superfamily members can divert trans-dihydrodiols to o-quinones, the cDNA encoding human aldehyde reductase (AKR1A1) was isolated from hepatoma HepG2 cells using RT-PCR, subcloned into a prokaryotic expression vector, overexpressed in E. coli and purified to homogeneity in milligram amounts. Studies revealed that AKR1A1 preferentially oxidized the metabolically relevant (-)-[3R,4R]-dihydroxy-3,4-dihydrobenz[a]anthracene. AKR1A1 also displayed high utilization ratios (V(max)/K(m)) for the following PAH trans-dihydrodiols: (+/-)trans-3,4-dihydroxy-3,4-dihydro-7-methylbenz[a]anthracene, (+/-)trans-3,4-dihydroxy-3,4-dihydro-7,12-dimethylbenz[a]anthracene and (+/-)trans-7,8-dihydroxy-7,8-dihydro-5-methylchrysene. Multiple tissue expression (MTE) arrays were used to measure the co-expressed of CYP1A1, EH and AKR1A1. All the three enzymes co-expressed to sites of PAH activation. The high catalytic efficiency of AKR1A1 for potent proximate carcinogen trans-dihydrodiols and its presence in tissues that contain CYP1A1 and EH suggests that it plays an important role in this alternative pathway of PAH activation (supported by CA39504).


Subject(s)
Aldehyde Reductase/metabolism , Polycyclic Aromatic Hydrocarbons/metabolism , Aldehyde Reductase/genetics , Biotransformation , Cell Line , Cytochrome P-450 CYP1A1/metabolism , Epoxide Hydrolases/metabolism , Escherichia coli/genetics , Humans , In Vitro Techniques , Oxidation-Reduction , Polycyclic Aromatic Hydrocarbons/chemistry , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stereoisomerism
6.
Mol Cell Endocrinol ; 171(1-2): 137-49, 2001 Jan 22.
Article in English | MEDLINE | ID: mdl-11165022

ABSTRACT

17beta-Hydroxysteroid dehydrogenase (17beta-HSD) type 5 has been cloned from human prostate and is identical to type 2 3alpha-HSD and is a member of the aldo-keto reductase (AKR) superfamily; it is formally AKR1C3. In vitro the homogeneous recombinant enzyme expressed in Escherichia coli functions as a 3-keto-, 17-keto- and 20-ketosteroid reductase and as a 3alpha-, 17beta- and 20alpha-hydroxysteroid oxidase. The enzyme will reduce 5alpha-DHT, Delta(4)-androstene-3,17-dione, estrone and progesterone to produce 3alpha-androstanediol, testosterone, 17beta-estradiol and 20alpha-hydroxprogesterone, respectively. It will also oxidize 3alpha-androstanediol, testosterone, 17beta-estradiol and 20alpha-hydroxyprogesterone to produce 5alpha-androstane-3,17-dione, Delta(4)-androstene-3,17-dione, and progesterone, respectively. Many of these properties are shared by the related AKR1C1, AKR1C2 and AKR1C4 isoforms. RT-PCR shows that AKR1C3 is dominantly expressed in the human prostate and mammary gland. Examination of k(cat)/K(m) for these reactions indicates that as a reductase it prefers 5alpha-dihydrotestosterone and 5alpha-androstane-3,17-dione as substrates to Delta(4)-androstene-3,17-dione, suggesting that in the prostate it favors the formation of inactive androgens. Its concerted reductase activity may, however, lead to a pro-estrogenic state in the breast since it will convert estrone to 17beta-estradiol; convert Delta(4)-androstene-3,17-dione to testosterone (which can be aromatized to 17beta-estradiol); and it will reduce progesterone to its inactive metabolite 20alpha-hydroxyprogesterone. Drawing on detailed structure-function analysis of the related rat 3alpha-HSD (AKR1C9), which shares 69% sequence identity with AKR1C3, it is predicted that AKR1C3 catalyzes an ordered bi bi mechanism, that the rate determining step is k(chem), and that an oxyanion prevails in the transition state. Based on these relationships steroidal-based inhibitors that compete with the steroid product would be desirable since they would act as uncompetitive inhibitors. With regards to transition state analogs steroid carboxylates and pyrazoles may be preferred while 3alpha, 17beta or 20alpha-spiro-oxiranes may act as mechanism-based inactivators.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Enzyme Inhibitors/chemistry , Isoenzymes/metabolism , Structure-Activity Relationship , 17-Hydroxysteroid Dehydrogenases/analysis , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-alpha-Dihydroprogesterone/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) , Animals , Binding Sites , Breast Neoplasms/enzymology , Cloning, Molecular , Escherichia coli/genetics , Estradiol/metabolism , Estrone/metabolism , Female , Gene Expression , Gene Library , Humans , Isoenzymes/analysis , Isoenzymes/antagonists & inhibitors , Liver/enzymology , Male , Mammary Glands, Animal/enzymology , Mutagenesis, Site-Directed , Progesterone/metabolism , Prostate/enzymology , Prostatic Neoplasms/enzymology , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Uterus/enzymology
7.
J Biol Chem ; 276(4): 2890-7, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11060293

ABSTRACT

The human aldo-keto reductase AKR1C1 (20alpha(3alpha)-hydroxysteroid dehydrogenase) is induced by electrophilic Michael acceptors and reactive oxygen species (ROS) via a presumptive antioxidant response element (Burczynski, M. E., Lin, H. K., and Penning, T. M. (1999) Cancer Res. 59, 607-614). Physiologically, AKR1C1 regulates progesterone action by converting the hormone into its inactive metabolite 20alpha-hydroxyprogesterone, and toxicologically this enzyme activates polycyclic aromatic hydrocarbon trans-dihydrodiols to redox-cycling o-quinones. However, the significance of its potent induction by Michael acceptors and oxidative stress is unknown. 4-Hydroxy-2-nonenal (HNE) and other alpha,beta-unsaturated aldehydes produced during lipid peroxidation were reduced by AKR1C1 with high catalytic efficiency. Kinetic studies revealed that AKR1C1 reduced HNE (K(m) = 34 microm, k(cat) = 8.8 min(-1)) with a k(cat)/K(m) similar to that for 20alpha-hydroxysteroids. Six other homogeneous recombinant AKRs were examined for their ability to reduce HNE. Of these, AKR1C1 possessed one of the highest specific activities and was the only isoform induced by oxidative stress and by agents that deplete glutathione (ethacrynic acid). Several hydroxysteroid dehydrogenases of the AKR1C subfamily catalyzed the reduction of HNE with higher activity than aldehyde reductase (AKR1A1). NMR spectroscopy identified the product of the NADPH-dependent reduction of HNE as 1,4-dihydroxy-2-nonene. The K(m) of recombinant AKR1C1 for nicotinamide cofactors (K(m) NADPH approximately 6 microm, K(m)(app) NADH >6 mm) suggested that it is primed for reductive metabolism of HNE. Isoform-specific reverse transcription-polymerase chain reaction showed that exposure of HepG2 cells to HNE resulted in elevated levels of AKR1C1 mRNA. Thus, HNE induces its own metabolism via AKR1C1, and this enzyme may play a hitherto unrecognized role in a response mounted to counter oxidative stress. AKRs represent alternative GSH-independent/NADPH-dependent routes for the reductive elimination of HNE. Of these, AKR1C1 provides an inducible cytosolic barrier to HNE following ROS exposure.


Subject(s)
20-Hydroxysteroid Dehydrogenases/metabolism , Aldehydes/metabolism , Alkenes/metabolism , Dehydroascorbic Acid/metabolism , Oxidative Stress/physiology , 20-Hydroxysteroid Dehydrogenases/genetics , Enzyme Induction , Glutathione/metabolism , Glutathione Transferase/metabolism , Humans , NAD/metabolism , NADP/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Recombinant Proteins/metabolism
8.
In Vitr Mol Toxicol ; 14(3): 177-90, 2001.
Article in English | MEDLINE | ID: mdl-11846991

ABSTRACT

Nile Red is a fluorescent dye used extensively to study fat accumulation in many types of cells; unfortunately protocols that work well for most cells are not effective for studying drug-induced lipid accumulation in cultured liver cells and hepatocyte-derived cell lines. Using human hepatoma (HepG2) cells, we have developed a simple Nile Red binding assay as a screen for steatosis-inducing compounds. Increases in Nile Red binding in response to known hepatotoxic compounds were observed after incubating treated cells with 1 microM Nile Red for several hours, washing away free Nile Red, and then allowing redistribution, and/or clearance of the lipid-indicator dye. Several compounds known to cause hepatic fat accumulation in vivo were examined and most robustly increased Nile Red binding in HepG2 cells. These include estrogen and other steroids, ethionine, cyclosporin A, and valproic acid. Required concentrations for increased Nile Red binding were generally three-fold or more lower than the cytotoxic concentration determined by a resazurin reduction assay in the same cells. Qualitatively similar Nile Red binding results were obtained when primary canine or rat hepatocytes were used. Morphological differences in Nile Red staining were observed by confocal fluorescence microscopy in HepG2 cells after treatment with different compounds and likely reflect distinct toxicological mechanisms.


Subject(s)
Fatty Liver/chemically induced , Fluorescent Dyes , Lipids/analysis , Oxazines , Steroids/toxicity , Xanthenes , Animals , Cell Survival , Dogs , Fatty Liver/metabolism , Fatty Liver/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , In Vitro Techniques , Indicators and Reagents/pharmacokinetics , Microscopy, Fluorescence , Oxazines/pharmacokinetics , Oxidation-Reduction , Rats , Tumor Cells, Cultured
9.
Toxicol Sci ; 58(2): 399-415, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099651

ABSTRACT

The rapid discovery of sequence information from the Human Genome Project has exponentially increased the amount of data that can be retrieved from biomedical experiments. Gene expression profiling, through the use of microarray technology, is rapidly contributing to an improved understanding of global, coordinated cellular events in a variety of paradigms. In the field of toxicology, the potential application of toxicogenomics to indicate the toxicity of unknown compounds has been suggested but remains largely unsubstantiated to date. A major supposition of toxicogenomics is that global changes in the expression of individual mRNAs (i.e., the transcriptional responses of cells to toxicants) will be sufficiently distinct, robust, and reproducible to allow discrimination of toxicants from different classes. Definitive demonstration is still lacking for such specific "genetic fingerprints," as opposed to nonspecific general stress responses that may be indistinguishable between compounds and therefore not suitable as probes of toxic mechanisms. The present studies demonstrate a general application of toxicogenomics that distinguishes two mechanistically unrelated classes of toxicants (cytotoxic anti-inflammatory drugs and DNA-damaging agents) based solely upon a cluster-type analysis of genes differentially induced or repressed in cultured cells during exposure to these compounds. Initial comparisons of the expression patterns for 100 toxic compounds, using all approximately 250 genes on a DNA microarray ( approximately 2.5 million data points), failed to discriminate between toxicant classes. A major obstacle encountered in these studies was the lack of reproducible gene responses, presumably due to biological variability and technological limitations. Thus multiple replicate observations for the prototypical DNA damaging agent, cisplatin, and the non-steroidal anti-inflammatory drugs (NSAIDs) diflunisal and flufenamic acid were made, and a subset of genes yielding reproducible inductions/repressions was selected for comparison. Many of the "fingerprint genes" identified in these studies were consistent with previous observations reported in the literature (e. g., the well-characterized induction by cisplatin of p53-regulated transcripts such as p21(waf1/cip1) and PCNA [proliferating cell nuclear antigen]). These gene subsets not only discriminated among the three compounds in the learning set but also showed predictive value for the rest of the database ( approximately 100 compounds of various toxic mechanisms). Further refinement of the clustering strategy, using a computer-based optimization algorithm, yielded even better results and demonstrated that genes that ultimately best discriminated between DNA damage and NSAIDs were involved in such diverse processes as DNA repair, xenobiotic metabolism, transcriptional activation, structural maintenance, cell cycle control, signal transduction, and apoptosis. The determination of genes whose responses appropriately group and dissociate anti-inflammatory versus DNA-damaging agents provides an initial paradigm upon which to build for future, higher throughput-based identification of toxic compounds using gene expression patterns alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinoma, Hepatocellular/genetics , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Algorithms , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , Humans , Liver Neoplasms/drug therapy , Reproducibility of Results , Tumor Cells, Cultured
10.
Biochem J ; 351(Pt 1): 67-77, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10998348

ABSTRACT

The kinetic parameters, steroid substrate specificity and identities of reaction products were determined for four homogeneous recombinant human 3alpha-hydroxysteroid dehydrogenase (3alpha-HSD) isoforms of the aldo-keto reductase (AKR) superfamily. The enzymes correspond to type 1 3alpha-HSD (AKR1C4), type 2 3alpha(17beta)-HSD (AKR1C3), type 3 3alpha-HSD (AKR1C2) and 20alpha(3alpha)-HSD (AKR1C1), and share at least 84% amino acid sequence identity. All enzymes acted as NAD(P)(H)-dependent 3-, 17- and 20-ketosteroid reductases and as 3alpha-, 17beta- and 20alpha-hydroxysteroid oxidases. The functional plasticity of these isoforms highlights their ability to modulate the levels of active androgens, oestrogens and progestins. Salient features were that AKR1C4 was the most catalytically efficient, with k(cat)/K(m) values for substrates that exceeded those obtained with other isoforms by 10-30-fold. In the reduction direction, all isoforms inactivated 5alpha-dihydrotestosterone (17beta-hydroxy-5alpha-androstan-3-one; 5alpha-DHT) to yield 5alpha-androstane-3alpha,17beta-diol (3alpha-androstanediol). However, only AKR1C3 reduced Delta(4)-androstene-3,17-dione to produce significant amounts of testosterone. All isoforms reduced oestrone to 17beta-oestradiol, and progesterone to 20alpha-hydroxy-pregn-4-ene-3,20-dione (20alpha-hydroxyprogesterone). In the oxidation direction, only AKR1C2 converted 3alpha-androstanediol to the active hormone 5alpha-DHT. AKR1C3 and AKR1C4 oxidized testosterone to Delta(4)-androstene-3,17-dione. All isoforms oxidized 17beta-oestradiol to oestrone, and 20alpha-hydroxyprogesterone to progesterone. Discrete tissue distribution of these AKR1C enzymes was observed using isoform-specific reverse transcriptase-PCR. AKR1C4 was virtually liver-specific and its high k(cat)/K(m) allows this enzyme to form 5alpha/5beta-tetrahydrosteroids robustly. AKR1C3 was most prominent in the prostate and mammary glands. The ability of AKR1C3 to interconvert testosterone with Delta(4)-androstene-3,17-dione, but to inactivate 5alpha-DHT, is consistent with this enzyme eliminating active androgens from the prostate. In the mammary gland, AKR1C3 will convert Delta(4)-androstene-3,17-dione to testosterone (a substrate aromatizable to 17beta-oestradiol), oestrone to 17beta-oestradiol, and progesterone to 20alpha-hydroxyprogesterone, and this concerted reductive activity may yield a pro-oesterogenic state. AKR1C3 is also the dominant form in the uterus and is responsible for the synthesis of 3alpha-androstanediol which has been implicated as a parturition hormone. The major isoforms in the brain, capable of synthesizing anxiolytic steroids, are AKR1C1 and AKR1C2. These studies are in stark contrast with those in rat where only a single AKR with positional- and stereo-specificity for 3alpha-hydroxysteroids exists.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Steroids/metabolism , 3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/isolation & purification , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific) , Androgens/chemistry , Androgens/metabolism , Binding Sites , Catalysis , Cloning, Molecular , Estrogens/chemistry , Estrogens/metabolism , Female , Gene Expression Profiling , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Ketones/metabolism , Kinetics , Male , Organ Specificity , Progestins/chemistry , Progestins/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Steroids/biosynthesis , Substrate Specificity
11.
Cancer Res ; 60(4): 908-15, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706104

ABSTRACT

Procarcinogenic polycyclic aromatic hydrocarbons (PAHs) induce their own metabolism and activation by binding to the cytosolic aryl hydrocarbon receptor (AhR), which then translocates to the nucleus and activates CYP1A1 gene transcription via xenobiotic response elements (XREs). Although the AhR demonstrates a strict specificity for planar aromatics, nonplanar (+/-)-trans-7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene also induced CYP1A1 expression in HepG2 cells over a delayed timecourse (approximately 6-12 h), suggesting a requirement for (+/-)trans-7,8-dihydrobenzo(a)pyrene metabolism. Aldo-keto reductase (AKR) inhibitors blocked this effect, suggesting that benzo(a)pyrene-7,8-dione (BPQ), a planar PAH o-quinone generated by AKRs, was the downstream inducer. BPQ was found to be a potent and rapid inducer of CYP1A1, with an EC50 value in HepG2 cells identical to that of the parent benzo(a)pyrene. BPQ was a more potent inducer of CYP1A1 when compared with the 1,6-, 3,6-, and 6,12-benzo(a)pyrene-diones. Multiple PAH o-quinones caused induction of CYP1A1, demonstrating that this was a general property of AKR-generated PAH o-quinones. HepG2-101L cells stably transfected with a XRE-luciferase construct showed that BPQ activated CYP1A1 transcription via a XRE-dependent mechanism. BPQ failed to induce CYP1A1 in AhR-deficient and AhR nuclear translocator-deficient murine hepatoma cell lines and confirmed that induction of CYP1A1 was AhR and AhR nuclear translocator-dependent. Electrophoretic mobility shift assays demonstrated the specific appearance of BPQ-activated AhR in the nucleus, and immunofluorescence studies confirmed that BPQ mediated nuclear translocation of the AhR. Classical bifunctional inducers elevate CYP1A1 expression via a XRE and are subsequently converted by CYP1A1 to electrophiles that induce phase II enzymes via an electrophilic response element/antioxidant response element PAH o-quinones represent a novel class of bifunctional inducer because they are electrophiles produced by phase II enzymes that simultaneously induce phase I enzymes via a XRE and phase II enzymes via a electrophilic response element/antioxidant response element (see also M. E. Burczynski et al., Cancer Res., 59: 607-614, 1999). This study shows that the AhR provides the only known mechanism by which genotoxic PAH o-quinones generated in the cytosol can be targeted to the nucleus with specificity.


Subject(s)
Alcohol Oxidoreductases/physiology , Cell Nucleus/metabolism , Cytochrome P-450 CYP1A1/biosynthesis , Quinones/toxicity , Receptors, Aryl Hydrocarbon/physiology , Alcohol Oxidoreductases/antagonists & inhibitors , Aldehyde Reductase , Aldo-Keto Reductases , Benzo(a)pyrene/toxicity , Biological Transport , Enzyme Induction , Humans , Polychlorinated Dibenzodioxins/toxicity , Transcriptional Activation , Tumor Cells, Cultured
12.
Cancer Res ; 59(3): 607-14, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973208

ABSTRACT

Human dihydrodiol dehydrogenase (DD) isoforms are aldo-keto reductases (AKRs) that activate polycyclic aromatic hydrocarbons (PAHs) by oxidizing trans-dihydrodiol proximate carcinogens to reactive and redox-active ortho-quinones. Of these, human AKR1C1 (DD1) and AKR1C2 (DD2) oxidize trans-7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene to the cytotoxic and genotoxic metabolite benzo[a]pyrene-7,8-dione (BPQ) with the highest catalytic efficiency. Exposure of HepG2 cells to a panel of inducers revealed that mRNA encoding one or more human AKR1C member(s) was induced (3- to 10-fold) by benzo[a]pyrene and other polycyclic aromatic compounds (bi-functional inducers), electrophilic Michael acceptors and phenolic antioxidants (monofunctional inducers), and reactive oxygen species (ROS). The induction of AKR1C mRNA by bifunctional inducers was delayed with respect to the induction of CYP1A1 mRNA, and AKR1C mRNA was not induced by the nonmetabolizable aryl hydrocarbon receptor ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). These data suggest that, in contrast to the CYPs, induction of AKR1C member(s) by PAHs and other bifunctional inducers is mediated indirectly via an antioxidant response element rather than a xenobiotic response element. Immunoblot and enzymatic assays confirmed that the increases in AKR1C mRNA were faithfully translated into functional AKR1C protein(s). The increased DD activity in HepG2 lysates was inhibited only by high concentrations of ursodeoxycholate, which suggested that AKR1C2 (DD2, bile-acid-binding protein) was not the isoform induced. RNase protection assays identified AKR1C1 (DD1) mRNA as the transcript which was up-regulated by mono- and bi-functional inducers and ROS in both human hepatoma (HepG2) and colon carcinoma (HT29) cells. BPQ, the electrophilic and redox-cycling product of the AKR1C1 reaction, also induced AKR1C1 expression. Thus, BPQ formation by AKR1C1 results in both a chemical (redox-cycling) and a genetic (AKR1C1 induction) amplification of ROS in PAH-exposed cells. Because ROS have been implicated in both tumor initiation and tumor promotion, the amplification of ROS by this pathway may play a significant role in PAH carcinogenesis.


Subject(s)
Alcohol Oxidoreductases/biosynthesis , Alcohol Oxidoreductases/metabolism , Isoenzymes/biosynthesis , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases , Polycyclic Aromatic Hydrocarbons/pharmacology , Polycyclic Aromatic Hydrocarbons/pharmacokinetics , Alcohol Oxidoreductases/antagonists & inhibitors , Aldehyde Reductase , Aldo-Keto Reductases , Benzopyrenes/pharmacology , Benzoquinones/pharmacology , Biotransformation , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP1A1/biosynthesis , Enzyme Induction , Enzyme Inhibitors/pharmacology , HT29 Cells/enzymology , Humans , Indicators and Reagents/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Ligands , Oxidation-Reduction , Oxidative Stress/physiology , Polychlorinated Dibenzodioxins/metabolism , Polychlorinated Dibenzodioxins/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Ribonucleases/metabolism , Xenobiotics/pharmacology , beta-Naphthoflavone/pharmacology
14.
Biochemistry ; 37(19): 6781-90, 1998 May 12.
Article in English | MEDLINE | ID: mdl-9578563

ABSTRACT

The bioactivation of polycyclic aromatic hydrocarbons (PAHs) to their ultimate carcinogenic forms proceeds via the formation of proximate carcinogen trans-dihydrodiols. Previous studies demonstrated that rat liver 3 alpha-hydroxysteroid dehydrogenase/dihydrodiol dehydrogenase (3 alpha-HSD/DD), a member of the aldo-keto reductase (AKR) superfamily, oxidizes PAH trans-dihydrodiols to redox-cycling o-quinones. Multiple closely related AKRs exist in human liver; however, it is unclear which, if any, participate in PAH activation by catalyzing the NADP+ -dependent oxidation of PAH trans-dihydrodiols. In this study, cDNAs encoding four human DD isoforms were isolated from HepG2 cells using isoform-selective RT-PCR. The recombinant proteins were overexpressed in Escherichia coli, purified to homogeneity, and kinetically characterized. Calculation of KM and kcat values of each isoform for model substrates revealed that they possessed enzymatic activities assigned to native human liver DD1, DD2, DD4, and type 2 3alpha-HSD (DDX) proteins. The ability of human DDs to oxidize the potent proximate carcinogen (+/-)-trans-7,8-dihydroxy-7, 8-dihydrobenzo[a]pyrene (BP-diol) was then examined. A reverse phase HPLC radiochemical assay demonstrated that all four isoforms oxidize (+/-)-BP-diol in the following rank order: DD2 > DD1 > DD4 > DDX. Each DD consumed the entire racemic BP-diol mixture, indicating that both the minor (+)-S,S- and major (-)-R,R-stereoisomers formed in vivo are substrates. First-order decay plots showed that DD1 and DD2 displayed preferences for one of the stereoisomers, and circular dichroism spectroscopy indicated that this isomer was the (+)-7S, 8S-enantiomer. The products of these reactions were trapped as either glycine or thiol ether conjugates of benzo[a]pyrene-7,8-dione (BPQ), indicating that the initial oxidation product was the reactive BPQ. Thus, human liver possesses multiple AKRs which contribute to PAH activation by catalyzing the NADP+-dependent oxidation of PAH trans-dihydrodiols to redox-active o-quinones.


Subject(s)
Benzopyrenes/metabolism , Dihydroxydihydrobenzopyrenes/metabolism , Isoenzymes/biosynthesis , Oxidoreductases/biosynthesis , Recombinant Proteins/biosynthesis , Benzopyrenes/chemistry , Biotransformation , Carcinoma, Hepatocellular , Catalysis , DNA, Complementary/isolation & purification , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Vectors/metabolism , Glycine/chemistry , Humans , Isoenzymes/genetics , Isoenzymes/isolation & purification , Kinetics , Liver/enzymology , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases/isolation & purification , Phospholipid Ethers/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Stereoisomerism , Tumor Cells, Cultured
15.
Biochemistry ; 35(42): 13664-72, 1996 Oct 22.
Article in English | MEDLINE | ID: mdl-8885846

ABSTRACT

A novel pathway of polycyclic aromatic hydrocarbon metabolism involves the oxidation of non-K-region trans-dihydrodiols to yield o-quinones, a reaction catalyzed by dihydrodiol dehydrogenase (DD). We have recently shown that in isolated rat hepatocytes (+/-)-trans-7,8-dihydroxy-7,8-dihydrobenzo-[a] pyrene (BP-diol) was oxidized by this route to yield benzo [a] pyrene-7,8-dione (BPQ). We now report the disposition of BPQ and its mutagenic and genotoxic properties. Using [3H]BPQ it was found that 30% of the radioactivity was sequestered by rat hepatocytes into the cell pellet. Isolation of hepatocyte DNA provided evidence for a low level of covalent incorporation of BPQ into DNA (30 +/- 17 adducts/ 10(6) base pairs). Examination of the hepatocellular DNA by agarose gel electrophoresis following treatment with BPQ indicated that extensive fragmentation had occurred. DNA fragmentation was also observed when hepatocytes were treated with BP-diol and this effect was attenuated by indomethacin, a DD inhibitor. Hepatocytes treated with either BP-diol or BPQ were found to produce large quantities of superoxide anion radical (O2.-). The amount of O2.- generated by BP-diol was blocked by DD inhibitors. These data suggest that by diverting BP-diol to BPQ reactive oxygen species (ROS) were generated which caused DNA fragmentation. The ability of BPQ to cause DNA strand scission was further studied using supercoiled phi X174 DNA. It was found that BPQ caused concentration-dependent (0.05-10 microM) strand scission in the presence of 1 mM NADPH (which promoted redox-cycling) provided CuCl2 (10 microM) was present. Complete destruction of the DNA was observed using 10 microM BPQ. This strand scission was prevented by catalase and hydroxyl radical scavengers but not by superoxide dismutase. These data indicate that ROS were responsible for the destruction of the DNA. Using 20 microM (+/-)-anti-7,8-dihydroxy-9,10-epoxy-7,8,9,10-tetrahydrobenzo [a]pyrene [(+/-)-anti-BPDE] only single nicks in the DNA were observed indicating that BPQ was the more potent chemical nuclease. BPQ was also found to be a direct-acting mutagen in the Ames test using Salmonella typhimurium tester strains TA97a, TA98, TA100, TA102, and TA104, but was 10-5500-fold less efficient as a mutagen than (+/-)-anti-BPDE. Our data indicate that DD suppresses the mutagenicity of (+/-)-anti-BPDE by producing BPQ, but in doing so a potent chemical nuclease is produced which causes extensive DNA fragmentation via the generation of ROS.


Subject(s)
Benzopyrenes/metabolism , Benzopyrenes/toxicity , DNA Damage , Liver/metabolism , Mutagens/toxicity , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , Animals , Benzoquinones/metabolism , Cells, Cultured , DNA Adducts/metabolism , DNA Fragmentation , DNA, Superhelical/metabolism , DNA, Viral/metabolism , Dihydroxydihydrobenzopyrenes/metabolism , Liver/cytology , Male , Mutagenicity Tests , Oxidoreductases/metabolism , Polycyclic Aromatic Hydrocarbons/metabolism , Polycyclic Aromatic Hydrocarbons/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL