Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Drug Metab Dispos ; 47(8): 907-918, 2019 08.
Article in English | MEDLINE | ID: mdl-31147315

ABSTRACT

Cytochrome P450s CYP1A1 and CYP1A2 can metabolize a broad range of foreign compounds and drugs. However, these enzymes have significantly overlapping substrate specificities. To establish their relative contribution to drug metabolism in vivo, we used a combination of mice humanized for CYP1A1 and CYP1A2 together with mice nulled at the Cyp1a1 and Cyp1a2 gene loci. CYP1A2 was constitutively expressed in the liver, and both proteins were highly inducible by 2,3,7,8-tetrachlorodibenzodioxin (TCDD) in a number of tissues, including the liver, lung, kidney, and small intestine. Using the differential inhibition of the human enzymes by quinidine, we developed a method to distinguish the relative contribution of CYP1A1 or CYP1A2 in the metabolism of drugs and foreign compounds. Both enzymes made a significant contribution to the hepatic metabolism of the probe compounds 7-methoxy and 7-ehthoxyresorufin in microsomal fractions from animals treated with TCDD. This enzyme kinetic approach allows modeling of the CYP1A1, CYP1A2, and non-CYP1A contribution to the metabolism of any substrate at any substrate, inhibitor, or enzyme concentration and, as a consequence, can be integrated into a physiologically based pharmacokinetics model. The validity of the model can then be tested in humanized mice in vivo. SIGNIFICANCE STATEMENT: Human CYP1A1 and CYP1A2 are important in defining the efficacy and toxicity/carcinogenicity of drugs and foreign compounds. In light of differences in substrate specificity and sensitivity to inhibitors, it is of central importance to understand their relative role in foreign compound metabolism. To address this issue, we have generated mice humanized or nulled at the Cyp1a gene locus and, through the use of these mouse lines and selective inhibitors, developed an enzyme kinetic-based model to enable more accurate prediction of the fate of new chemicals in humans and which can be validated in vivo using mice humanized for cytochrome P450-mediated metabolism.


Subject(s)
Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Drug Evaluation, Preclinical/methods , Oxazines/pharmacokinetics , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Gene Knock-In Techniques , Liver/metabolism , Mice, Knockout , Models, Animal , Oxazines/administration & dosage
2.
Drug Metab Dispos ; 47(6): 601-615, 2019 06.
Article in English | MEDLINE | ID: mdl-30910785

ABSTRACT

Species differences in drug metabolism and disposition can confound the extrapolation of in vivo PK data to man and also profoundly compromise drug efficacy studies owing to differences in pharmacokinetics, in metabolites produced (which are often pharmacologically active), and in differential activation of the transcription factors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), which regulate the expression of such enzymes as P450s and drug transporters. These differences have gained additional importance as a consequence of the use of genetically modified mouse models for drug-efficacy testing and also patient-derived xenografts to predict individual patient responses to anticancer drugs. A number of humanized mouse models for cytochrome P450s, CAR, and PXR have been reported. However, the utility of these models has been compromised by the redundancy in P450 reactions across gene families, whereby the remaining murine P450s can metabolize the compounds being tested. To remove this confounding factor and create a mouse model that more closely reflects human pathways of drug disposition, we substituted 33 murine P450s from the major gene families involved in drug disposition, together with Car and Pxr, for human CAR, PXR, CYP1A1, CYP1A2, CYP2C9, CYP2D6, CYP3A4, and CYP3A7. We also created a mouse line in which 34 P450s were deleted from the mouse genome. Using model compounds and anticancer drugs, we demonstrated how these mouse lines can be applied to predict drug-drug interactions in patients and discuss here their potential application in the more informed design of clinical trials and the personalized treatment of cancer.


Subject(s)
Drug Interactions/physiology , Pharmaceutical Preparations/metabolism , Signal Transduction/physiology , Animals , Cell Line , Clinical Trials as Topic , Female , Mice
3.
J Fish Biol ; 89(3): 1905-12, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27456225

ABSTRACT

The influence of two cryptic, seagrass-inhabiting omnivorous fishes, the fan-bellied leatherjacket Monacanthus chinensis and the variable sabretoothed blenny Petroscirtes variabilis, on seagrass epiphyte biomass are described. Overall, M. chinensis significantly reduced epiphyte biomass by 35·1% after 18 h in experimental aquaria, whilst P. variabilis showed a non-significant 15·7% reduction. It is concluded that some cryptic omnivorous species play an important role in epiphyte removal in seagrass beds.


Subject(s)
Ecosystem , Herbivory , Perciformes , Tetraodontiformes , Animals , Biomass , Fishes , Plants
4.
Fish Physiol Biochem ; 42(6): 1521-1526, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27178186

ABSTRACT

Samples for the analysis of stable isotopes, genetics and other tissue sampling methods of small fish are often taken via lethal techniques. The aim of this study was to determine the suitability of a non-lethal sampling method for removing muscle tissue from fish under 30 cm total length (TL). A 4-mm biopsy punch was used to remove muscle tissue from two different species, Lutjanus russelli (Lutjanidae) and Lethrinus laticaudis (Lethrinidae). Several scales were removed from the sampling location, and then the biopsy punch was inserted to remove the muscle tissue. Immediately following this, a mixture of Fish Bandage™ and three drops of Betadine™ antiseptic solution was applied to the wound to reduce the chance of infection. The biopsy punch removes an average of 8 mg of muscle tissue from the fish, more than is required for stable isotope and genetic analyses (1 mg). The condition of fish subjected to our three treatments, biopsied fish, a handling controls and a tank control, was compared via gill beat counts at the time of biopsy and 3, 6, 9, 12 and 24 h after treatment. Overall, no instances of mortality were recorded across the study for all species and all treatments. This method has been found to be a suitable non-lethal method in the removal of muscle tissue from these two fish species, potentially also other species under 30 cm TL, by eliminating mortality and minimising stress responses in sampled fishes.


Subject(s)
Muscles/surgery , Perciformes/surgery , Animals , Biopsy
5.
Mol Neurobiol ; 46(2): 475-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22773138

ABSTRACT

Mitochondrial dysfunction and oxidative stress are implicated in the neurodegenerative process in Parkinson's disease (PD). Moreover, c-Jun N-terminal kinase (JNK) plays an important role in dopaminergic neuronal death in substantia nigra pars compacta. Tauroursodeoxycholic acid (TUDCA) acts as a mitochondrial stabilizer and anti-apoptotic agent in several models of neurodegenerative diseases. Here, we investigated the role of TUDCA in preventing 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurodegeneration in a mouse model of PD. We evaluated whether TUDCA modulates MPTP-induced degeneration of dopaminergic neurons in the nigrostriatal axis, and if that can be explained by regulation of JNK phosphorylation, reactive oxygen species (ROS) production, glutathione S-transferase (GST) catalytic activation, and Akt signaling, using C57BL/6 glutathione S-transferase pi (GSTP) null mice. TUDCA efficiently protected against MPTP-induced dopaminergic degeneration. We have previously demonstrated that exacerbated JNK activation in GSTP null mice resulted in increased susceptibility to MPTP neurotoxicity. Interestingly, pre-treatment with TUDCA prevented MPTP-induced JNK phosphorylation in mouse midbrain and striatum. Moreover, the anti-oxidative role of TUDCA was demonstrated in vivo by impairment of ROS production in the presence of MPTP. Finally, results herein suggest that the survival pathway activated by TUDCA involves Akt signaling, including downstream Bad phosphorylation and NF-κB activation. We conclude that TUDCA is neuroprotective in an in vivo model of PD, acting mainly by modulation of JNK activity and cellular redox thresholds, together with activation of the Akt pro-survival pathway. These results open new perspectives for the pharmacological use of TUDCA, as a modulator of neurodegeneration in PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Parkinson Disease/pathology , Parkinson Disease/prevention & control , Taurochenodeoxycholic Acid/pharmacology , Animals , Cell Death/drug effects , Disease Models, Animal , HSP27 Heat-Shock Proteins/metabolism , I-kappa B Proteins/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/enzymology , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Taurochenodeoxycholic Acid/therapeutic use , bcl-Associated Death Protein/metabolism
6.
Pharmacology ; 83(2): 99-109, 2009.
Article in English | MEDLINE | ID: mdl-19088497

ABSTRACT

BACKGROUND AND AIMS: The 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole prodrug Phortress exerts potent and selective antitumour activity in vitro and in vivo. Preclinical toxicokinetic studies in 2 rodent species were undertaken to determine Phortress' maximum tolerated dose and advise a safe starting dose for clinical evaluation. METHODS: Plasma pharmacokinetic parameters were determined by high-performance liquid chromatography and fluorescence detection following Phortress administration to mice (10 mg/kg, intravenously on days 1 and 8). Phortress (20 mg/kg, on days 1 and 8) was administered to CYP1A1/betaGAL reporter mice; tissues were examined macro- and microscopically. Toxicological and pharmacodynamic endpoints were examined in organs of rodents receiving Phortress (10 mg/kg or 20 mg/kg, on days 1 and 8). CYP1A1 expression and Phortress-derived DNA adducts were determined in lungs and livers (on days 11 and 36). RESULTS: No accumulation of Phortress was detected in murine plasma. beta-Galactosidase activity inferred Phortress-derived induction of cyp1a1 transcription in the livers of transgenic mice; no total body weight loss was encountered in these animals. However, a fall in lung:body weight and kidney:body weight ratios, raised serum alkaline phosphatase levels and hepatic histopathological disturbances in animals receiving 20 mg/kg Phortress indicate organ sites of potential toxicity. CYP1A1 protein was induced transiently in the lungs of both species and in the livers of rats. Elimination of hepatic DNA adducts and rat pulmonary adducts was evident; however, murine pulmonary adducts persisted. CONCLUSION: Rodent preclinical toxicology established that mice represent the more sensitive rodent species, resolving a maximum tolerated dose of 10 mg/kg Phortress.


Subject(s)
Prodrugs/pharmacokinetics , Prodrugs/toxicity , Thiazoles/pharmacokinetics , Thiazoles/toxicity , Alkaline Phosphatase/blood , Animals , Body Weight , Cytochrome P-450 CYP1A1/metabolism , DNA Adducts/drug effects , DNA Adducts/pharmacokinetics , Drug Evaluation, Preclinical , Female , Genes, Reporter/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Lung/drug effects , Lung/pathology , Male , Maximum Tolerated Dose , Mice , Mice, Inbred ICR , Organ Size , Rats , Rats, Sprague-Dawley , Thiazoles/blood , beta-Galactosidase/metabolism
7.
Cell Death Differ ; 12(9): 1240-54, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16094404

ABSTRACT

Several studies have indicated that proteasome inhibitors (PIs) are promising anticancer agents. We have discovered that PIs have the unique ability to activate effector caspases through a mitochondrial Bcl-2 inhibitable but caspase-9 independent pathway. Stabilization of released Smac induced by blockade of the proteasome could explain the apoptosome-independent cell death induced by PIs. In fact, Smac/DIABLO critically supports this PIs-dependent caspase activation. By using a new assay, we confirm that at a single cell level both Smac and PIs can activate caspases in the absence of the apoptosome. Moreover, we have observed two PIs-induced kinetics of caspase activation, with caspase-9 being still required for the rapid caspase activation in response to mitochondrial depolarization, but dispensable for the slow DEVDase activation. In summary, our data indicate that PIs can activate downstream caspases at least in part through Smac/DIABLO stabilization.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Proteasome Inhibitors , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Carrier Proteins , Caspase 9 , Caspases/metabolism , Cell Death , Cytochromes c/metabolism , Cytosol/metabolism , Enzyme Activation , Etoposide/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted , Intracellular Signaling Peptides and Proteins , Kinetics , Membrane Potentials , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Proteins , Plasmids/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Retroviridae/genetics , Time Factors , Transfection
8.
Aust N Z J Public Health ; 28(1): 32-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15108744

ABSTRACT

OBJECTIVE: To compare the risk of death in a cohort of Western Australian released prisoners with the risk experienced by the general population of Western Australia. METHODS: A cohort study of prisoners in Western Australia whose last date of release ranged from 1 January 1994 to 1 January 1999. Overall mortality and cause of death were determined by data linkage to the Registrar General's record of deaths. RESULTS: Aboriginal prisoners had a significantly lower survival rate after release than non-Aboriginal prisoners (p < 0.0001). When compared with their peers in the Western Australian community, both Aboriginal and non-Aboriginal prisoners were found to have an increased relative risk of death. Female non-Aboriginal released prisoners aged between 20 and 40 years were 17.8 (95% CI 8.1-27.5) times more likely to die than other female non-Aboriginals in Western Australia in the same age range. Male non-Aboriginal prisoners aged 20-40 years were 6.3 (95% CI 5.2-7.4) times more likely to die than their counterparts in the WA community. Female Aboriginal released prisoners were 3.4 (95% CI 1.2-5.6) times more likely to die than their peers, while male Aboriginal released prisoners were 2.9 (95% CI 2.2-3.5) times more likely to die. In their first six months after release, female non-Aboriginal prisoners aged 20 to 40 years were 69.1 (95% CI 17.9-120.3) times more likely to die than their counterparts in the WA community. The main causes of excess death were related to drug and alcohol abuse. CONCLUSION: All prisoners were at greater than expected relative risk of death after release from prison, with female non-Aboriginal prisoners at particularly high relative risk.


Subject(s)
Mortality , Native Hawaiian or Other Pacific Islander/statistics & numerical data , Prisoners/statistics & numerical data , Adult , Cause of Death , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Prisons , Risk Assessment , Sex Distribution , Survival Analysis , Western Australia/epidemiology
9.
Neurology ; 61(6): 834-6, 2003 Sep 23.
Article in English | MEDLINE | ID: mdl-14504333

ABSTRACT

A unique case of amyloidoma presenting as a dumbbell-shaped tumor of a spinal root without bony erosion is described. Amyloid was also present in the facial nerve. DNA analysis for transthyretin was negative. Isolated amyloid fibers contained lambda light chains, and although plasma and urine immunoelectrophoresis performed by immunofixation was normal, it is possible the tumor may have been derived from an isolated plasmacytoma.


Subject(s)
Amyloidosis/pathology , Nerve Compression Syndromes/etiology , Spinal Nerve Roots/pathology , Amino Acid Sequence , Amyloidosis/complications , Amyloidosis/diagnosis , Amyloidosis/surgery , Cervical Vertebrae/surgery , Decompression, Surgical , Diagnostic Errors , Facial Nerve Diseases/pathology , Facial Nerve Diseases/surgery , Facial Paralysis/etiology , Humans , Hypesthesia/etiology , Immunoglobulin lambda-Chains/chemistry , Laminectomy , Male , Middle Aged , Molecular Sequence Data , Multiple Sclerosis/diagnosis , Nerve Compression Syndromes/diagnosis , Nerve Compression Syndromes/surgery , Sequence Homology, Amino Acid , Trigeminal Nerve/physiopathology
10.
Toxicol In Vitro ; 17(2): 201-5, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12650674

ABSTRACT

In these experiments precision-cut tissue slices from two existing transgenic mouse strains, with transgenes that couple promoting or binding elements to a reporter protein, were used for determination of reporter induction. This approach combines the power of transgenic animals with the practicality of in vitro systems to investigate the biological impact of xenobiotics. Additionally, the normal cellular architecture and heterogeneity is retained in precision-cut tissue slices. Two transgenic mouse strains, one of which couples the promoting region of CYP 1A1 to beta-galactosidase, and another which couples two forward and two backward 12-O-tetradecanoyl phorbol-13-acetate (TPA) repeat elements (TRE) to luciferase (termed AP-1/luciferase), were used to determine the feasibility of this approach. Precision-cut kidney and liver slices from both transgenic strains remain viable as determined by slice K(+) ion content and LDH enzyme release. Liver slices harvested from the CYP 1A1/beta-galactosidase transgenic mice exhibit a 14-fold increase in beta-galactosidase activity when incubated with beta-napthoflavone for 24 h. Kidney and liver slices obtained from the AP-1/luciferase transgenic mice demonstrate induction of luciferase (up to 2.5-fold) when incubated with phorbol myristate acetate (PMA or TPA) up to 4 h. These data indicate that precision-cut tissue slices from transgenic mice offer a novel in vitro method for toxicity evaluation while maintaining normal cell heterogeneity.


Subject(s)
Microtomy , Toxicity Tests/methods , Animals , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction/drug effects , In Vitro Techniques , Kidney/drug effects , Kidney/enzymology , Kidney/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Luciferases/biosynthesis , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Transcription Factor AP-1/genetics , beta-Galactosidase/biosynthesis
11.
AIHAJ ; 62(4): 428-33, 2001.
Article in English | MEDLINE | ID: mdl-11549136

ABSTRACT

A novel procedure is described to establish knife steeling schedules for poultry and meat- processing operations based on increased force due to knife dullness from repetitive use to minimize operator exertions and physical stress associated with work-related musculoskeletal disorders. Knife dullness was quantified using a novel apparatus described in this article that measures the area cut by a knife into a carrageenan gel target for a controlled dynamic load at the knife handle. Two meat-cleaning jobs in a poultry-processing plant were studied. One job required significantly more force and a greater number of cuts than the other. Eight experienced operators participated in the study. Four freshly ground and honed knives were randomly used by each operator for 4, 45, 75, or 125 cutting cycles, measured for dullness and reconditioned by the operator using a steel sharpening rod. An empirical model for knife dulling and reconditioning was developed, and the corresponding increase in force was predicted for various cutting and reconditioning frequencies. The model showed that it took 57 and 125 cutting cycles for the high- and low-force jobs, respectively, to achieve a similar reduction in target surface area of 30%. This reduction in target surface area corresponded to a similar percentage increase in force needed for the same cut in carrageenan gel as compared to a freshly honed knife as measured using strain gages. This method may be used in meat processing plants for determining effective reconditioning schedules that reduce operator exertions with minimum effect on quality and productivity.


Subject(s)
Cooking and Eating Utensils , Food-Processing Industry , Occupational Health , Adult , Female , Humans , Male , Steel
12.
J Pharmacol Exp Ther ; 298(1): 339-45, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408560

ABSTRACT

Glutathione S-transferase P1-1 (GSTpi) is an abundant and ubiquitously expressed protein in normal and malignant mammalian tissues and possesses catalytic and ligand binding properties. Our present data suggest that the protein contributes to the regulation of cell proliferation. Mouse embryo fibroblasts (MEFs) isolated from mice with a GSTP1-1 [glutathione S-transferase P1-1 (isozyme in nonhepatic tissue)] null genotype (GSTpi(-/-)) doubled their population in 26.2 h versus 33.6 h for the wild type (GSTpi(+/+)). Retroviral transfection of GSTP1-1 into GSTpi(-/-) MEF cells slowed the doubling time to 30.4 h. Both early passage and immortalized MEF cells from GSTpi(-/-) animals expressed significantly elevated activity of extracellular signal-regulated kinases ERK1/ERK2, kinases linked to cell proliferation pathways. In vivo, GSTpi(-/-) mice had higher basal levels of circulating white blood cells compared with GSTpi(+/+). Administration of a peptidomimetic inhibitor of GSTP1-1, TLK199, (gamma-glutamyl-S-(benzyl)cysteinyl-R-phenyl glycine diethyl ester), stimulated both lymphocyte production and bone marrow progenitor (colony-forming unit-granulocyte macrophage) proliferation, but only in GSTpi(+/+) and not in GSTpi(-/-) animals. Selection of a resistant clone of an HL60 tumor cell line through chronic exposure to TLK199 resulted in cells with elevated activities of c-Jun NH2 terminal kinase (JNK1) and ERK1/ERK2, and allowed the cells to proliferate under stress conditions that induced high levels of apoptosis in the wild type cells. The in vitro and in vivo data are consistent with the principle that GSTP1-1 influences cell proliferation.


Subject(s)
Enzyme Inhibitors/pharmacology , Glutathione Transferase/drug effects , Glutathione/analogs & derivatives , Glutathione/pharmacology , Isoenzymes/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinases/drug effects , Animals , Fibroblasts/drug effects , Fibroblasts/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HL-60 Cells/drug effects , HL-60 Cells/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Superoxides/metabolism , Transfection
13.
J Clin Gastroenterol ; 32(5): 435-8, 2001.
Article in English | MEDLINE | ID: mdl-11319318

ABSTRACT

Collagenous colitis is a form of microscopic colitis that results in chronic watery diarrhea. The disorder predominantly affects middle-aged women, and its course tends to be benign. It is not thought to be a precursor of overt inflammatory bowel disease; however, apparent progression to ulcerative colitis has been reported on one previous occasion. We describe two further patients with symptoms and histologic features of collagenous colitis who subsequently developed ulcerative colitis. The first patient developed ulcerative colitis 13 months after diagnosis of collagenous colitis, although she gave a 23-year history of profuse watery diarrhea, which had not been adequately investigated. In the second patient, collagenous colitis was diagnosed soon after the onset of watery diarrhea, and 12 months later, progression to ulcerative colitis was documented. Both patients tested positive for perinuclear antineutrophil cytoplasmic antibody after they developed ulcerative colitis; the first patient was initially negative. In conclusion, these two cases, in addition to the one other in the literature, suggest that collagenous colitis and ulcerative colitis may represent extremes in the spectrum of inflammatory bowel disease and that collagenous colitis may evolve to ulcerative colitis. Therefore, progression to ulcerative colitis should be considered in any patient with known collagenous colitis whenever bloody diarrhea occurs, or if red cells, as well as white cells, are noted on stool microscopy.


Subject(s)
Colitis, Ulcerative/physiopathology , Colitis/pathology , Aged , Colitis, Ulcerative/pathology , Collagen , Colonoscopy , Disease Progression , Female , Humans , Middle Aged
14.
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
15.
Proc Natl Acad Sci U S A ; 97(23): 12741-5, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11058152

ABSTRACT

Overdose of acetaminophen, a widely used analgesic drug, can result in severe hepatotoxicity and is often fatal. This toxic reaction is associated with metabolic activation by the P450 system to form a quinoneimine metabolite, N-acetyl-p-benzoquinoneimine (NAPQI), which covalently binds to proteins and other macromolecules to cause cellular damage. At low doses, NAPQI is efficiently detoxified, principally by conjugation with glutathione, a reaction catalyzed in part by the glutathione S-transferases (GST), such as GST Pi. To assess the role of GST in acetaminophen hepatotoxicity, we examined acetaminophen metabolism and liver damage in mice nulled for GstP (GstP1/P2((-/-))). Contrary to our expectations, instead of being more sensitive, GstP null mice were highly resistant to the hepatotoxic effects of this compound. No significant differences between wild-type (GstP1/P2((+/+))) mice and GstP1/P2((-/-)) nulls in either the rate or route of metabolism, particularly to glutathione conjugates, or in the levels of covalent binding of acetaminophen-reactive metabolites to cellular protein were observed. However, although a similar rapid depletion of hepatic reduced glutathione (GSH) was found in both GstP1/P2((+/+)) and GstP1/P2((-/-)) mice, GSH levels only recovered in the GstP1/P2((-/-)) mice. These data demonstrate that GstP does not contribute in vivo to the formation of glutathione conjugates of acetaminophen but plays a novel and unexpected role in the toxicity of this compound. This study identifies new ways in which GST can modulate cellular sensitivity to toxic effects and suggests that the level of GST Pi may be an important and contributing factor in the sensitivity of patients with acetaminophen-induced hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Glutathione Transferase/physiology , Isoenzymes/physiology , Liver/drug effects , Acetaminophen/administration & dosage , Acetaminophen/metabolism , Acetaminophen/pharmacokinetics , Alanine Transaminase/metabolism , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/metabolism , Analgesics, Non-Narcotic/pharmacokinetics , Animals , Benzoquinones/metabolism , Glutathione/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Imines/metabolism , Isoenzymes/genetics , Liver/injuries , Liver/metabolism , Male , Mice , Mice, Knockout
16.
Phys Med Biol ; 45(10): N133-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11049193

ABSTRACT

Different methodologies are used for calibrating polyacrylamide gel (PAG) dosimeters. One methodology involves injecting nitrogen-filled glass vials with polymer gel. Due to the vials being pre-filled with nitrogen, a nitrogen-filled space remains in the glass vial above the gel. The glass vial is then irradiated using ionizing radiation to polymerize the PAG. Monte Carlo simulations were performed to examine the effects on the radiation field due to the glass vial and the lack of backscatter material due to the nitrogen-filled space. Results for two different formulations of PAG indicated that the influence of the glass and the nitrogen-filled space are negligible.


Subject(s)
Acrylic Resins , Glass , Radiometry/instrumentation , Radiometry/methods , Scattering, Radiation , Computer Simulation , Monte Carlo Method , Nitrogen
17.
Mol Pharmacol ; 58(1): 167-74, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10860939

ABSTRACT

TER286 [gamma-glutamyl-alpha-amino-beta(2-ethyl-N,N,N', N'-tetrakis(2-chloroethyl)phosphorodiamidate)-sulfonyl-propionyl-( R)- (-) phenylglycine] is a novel nitrogen mustard prodrug that is preferentially activated by glutathione S-transferase P1-1 (GSTP1-1). A human promyelocytic leukemia /TER286-resistant cell line was selected by chronic, long-term exposure to the prodrug. Although resistance was not readily achieved, eventually a 5-fold resistant clone was isolated. Cross-resistance to melphalan occurred, but not to doxorubicin (Adriamycin), taxol, and gamma-glutamyl-S-(benzyl)cysteinyl-R(-)-phenyl glycine diethyl ester, a GSTP1-1 inhibitor. The protein and transcript levels and enzymatic activity of GSTP1-1 were reduced significantly in the selected resistant line. GSTalpha levels were unchanged, and GSTmu was undetectable. Although glutathione levels were elevated in human promyelocytic leukemia/TER286 cells, no changes in the expression of thiol-related genes including gamma-glutamylcysteine synthetase, gamma-glutamyl transpeptidase, or multidrug resistance protein were found. A 7-fold increase in catalase expression in the resistant cell line indicated an adaptive response to oxidative and electrophilic stress, and this was also reflected in the lower prevalence of drug-induced DNA single-strand breaks in the resistant cells. Mouse embryo fibroblast GSTP1-1(-/-) cells exhibited 2-fold resistance to TER286 compared with GSTP1-1(+/+) cells. NIH3T3 cells transfected with combinations of gamma-GCS and multidrug resistance protein exhibited enhanced resistance to TER286, although the degree of resistance was impaired by cotransfection of GSTP1-1. These results are consistent with responses in the TER286-resistant cells indicative of GSTP1-1-mediated mechanism of activation. In consequence, these data support the rationale that tumors expressing high levels of GSTP1-1 will be more sensitive to the cytotoxic effects of the drug.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Cytotoxins/metabolism , Glutathione Transferase/metabolism , Glutathione/analogs & derivatives , Glutathione/metabolism , Isoenzymes/metabolism , Prodrugs/metabolism , 3T3 Cells , Animals , Antineoplastic Agents, Alkylating/pharmacology , Cytotoxins/pharmacology , Drug Screening Assays, Antitumor , Glutathione/pharmacology , Glutathione S-Transferase pi , HL-60 Cells , Humans , Inhibitory Concentration 50 , Mice , Prodrugs/pharmacology , Tumor Cells, Cultured
18.
Pathology ; 32(2): 142-6, 2000 May.
Article in English | MEDLINE | ID: mdl-10840837

ABSTRACT

Ectopic hamartomatous thymoma is a rare and distinctive tumor found in the deep soft tissues of the neck, which is characterized histopathologically by a mixture of spindle, epithelial, and adipose cell elements. We present a case of this lesion occurring in a 39-year-old male. The characteristic histochemical and immunohistochemical findings of these tumors are demonstrated. In addition, review of the reported histological, immunohistochemical and ultrastructural findings is presented together with a table of clinical findings in the tumors so far described. A brief discussion of theories of histogenesis and possible differential diagnosis is included.


Subject(s)
Choristoma/pathology , Hamartoma/pathology , Head and Neck Neoplasms/pathology , Soft Tissue Neoplasms/pathology , Thymoma/pathology , Thymus Neoplasms/pathology , Adult , Biomarkers, Tumor/analysis , Hamartoma/chemistry , Head and Neck Neoplasms/chemistry , Humans , Immunoenzyme Techniques , Male , Neoplasm Proteins/analysis , Soft Tissue Neoplasms/chemistry , Thymoma/chemistry , Thymus Neoplasms/chemistry
19.
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
20.
Biochem Soc Trans ; 28(2): 42-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10816096

ABSTRACT

The cytochrome P450 mono-oxygenase system represents a major defence against chemical challenge from the environment, constituting part of an adaptive response mounted by an organism following exposure to harmful agents. Cytochrome P450s are also able to catalyse the activation of compounds to toxic products, and participate in a variety of essential 'housekeeping' functions, such as biosynthesis of steroid hormones and fatty acid oxidation. It is clear that the modulation of expression of these enzymes can have a significant effect on chemical toxicity, carcinogenicity and mutagenicity. The concept of cancer chemoprevention, i.e. the administration of a (non-toxic) chemical or dietary component in order to prevent neoplastic disease or to inhibit its progression, is an attractive one. Despite this, relatively little work has been done to characterize the ability of putative chemopreventive agents to modulate P450 expression, or to understand the interaction between P450s and chemopreventive agents. Before chemopreventive treatment can become a reality, it is essential that this complex issue is addressed; for instance, it is likely that any single chemopreventive agent will induce more than one P450 isoenzyme, and while altered expression of a particular P450 may attenuate the effects of one toxic agent, the effects of others might well be potentiated. Our laboratory has created a transgenic mouse line in which the rat CYP1A1 promoter drives expression of the beta-galactosidase gene. These mice can be used to define which compounds act via the Ah receptor, in which tissues, and at which stage of development. We are currently developing another mouse line in which beta1-galactosidase expression is controlled by the mouse GstA1 promoter, allowing us to define the role of the antioxidant responsive element in the action of chemopreventive agents. Finally, using cre-loxP transgenic technology, we have generated a mouse line in which P450 reductase can be deleted in a conditional, i.e. tissue-specific, manner, permitting us to investigate the role of P450s in chemoprevention in a more defined manner.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cytochrome P-450 Enzyme System/physiology , Animals , Cytochrome P-450 CYP1A1/genetics , Gene Expression Regulation/drug effects , Kidney/drug effects , Liver/drug effects , Lung/drug effects , Male , Mice , Mice, Transgenic , Models, Biological , Promoter Regions, Genetic , Rats , Rats, Inbred F344 , Tissue Distribution , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL