1
|
Degradation of aflatoxin B1 by water-assisted microwave irradiation: Kinetics, products, and pathways. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.112310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
2
|
Murcia HW, Diaz GJ. Protective effect of glutathione S-transferase enzyme activity against aflatoxin B 1 in poultry species: relationship between glutathione S-transferase enzyme kinetic parameters, and resistance to aflatoxin B 1. Poult Sci 2021; 100:101235. [PMID: 34214746 PMCID: PMC8258694 DOI: 10.1016/j.psj.2021.101235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/31/2021] [Accepted: 04/24/2021] [Indexed: 11/26/2022] Open
Abstract
Comparative studies designed to investigate the role of glutathione S-transferase (GST) activity on the enzyme catalyzed trapping of aflatoxin B1-8,9-epoxide (AFBO) with glutathione, and the relationship with aflatoxin B1 (AFB1) resistance have not been conducted in poultry. Hepatic cytosolic fractions of chickens, quail, turkeys and ducks were used to measure in vitro the enzymatic parameters maximal velocity (Vmax), Michaelis-Menten constant (Km) and intrinsic clearance (CLint) for GST activity. AFB1 used ranged from 2.0 to 157.5 µM and the AFB1-GSH produced was identified and quantitated by HPLC. Significant differences were found in GST Vmax values, being the highest in chickens, followed by quail, ducks and turkeys. The Km values were also significantly different, with chickens < ducks < turkeys < quail. Chickens had the higher CLint value in contrast to ducks. Differences by sex showed that duck females had a higher CLint value than the turkey and quail, whereas duck males had a CLint close to that of turkey. The ratio "AFBO production /AFB1-GSH production" follows the order duck>turkey>quail>chicken, in agreement with the known poultry sensitivity. The extremely high "AFB1 epoxidation activity/ GST activity" ratio observed in ducks might be the explanation for the development of hepatocellular carcinoma in this species.
Collapse
Affiliation(s)
- Hansen W Murcia
- Laboratorio de Toxicología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia, Bogotá D.C., Colombia.
| | - Gonzalo J Diaz
- Laboratorio de Toxicología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| |
Collapse
|
3
|
Ghallab A, Hassan R, Myllys M, Albrecht W, Friebel A, Hoehme S, Hofmann U, Seddek AL, Braeuning A, Kuepfer L, Cramer B, Humpf HU, Boor P, Degen GH, Hengstler JG. Subcellular spatio-temporal intravital kinetics of aflatoxin B 1 and ochratoxin A in liver and kidney. Arch Toxicol 2021; 95:2163-2177. [PMID: 34003344 PMCID: PMC8166722 DOI: 10.1007/s00204-021-03073-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022]
Abstract
Local accumulation of xenobiotics in human and animal tissues may cause adverse effects. Large differences in their concentrations may exist between individual cell types, often due to the expression of specific uptake and export carriers. Here we established a two-photon microscopy-based technique for spatio-temporal detection of the distribution of mycotoxins in intact kidneys and livers of anesthetized mice with subcellular resolution. The mycotoxins ochratoxin A (OTA, 10 mg/kg b.w.) and aflatoxin B1 (AFB1, 1.5 mg/kg b.w.), which both show blue auto-fluorescence, were analyzed after intravenous bolus injections. Within seconds after administration, OTA was filtered by glomeruli, and enriched in distal tubular epithelial cells (dTEC). A striking feature of AFB1 toxicokinetics was its very rapid uptake from sinusoidal blood into hepatocytes (t1/2 ~ 4 min) and excretion into bile canaliculi. Interestingly, AFB1 was enriched in the nuclei of hepatocytes with zonal differences in clearance. In the cytoplasm of pericentral hepatocytes, the half-life (t1/2~ 63 min) was much longer compared to periportal hepatocytes of the same lobules (t1/2 ~ 9 min). In addition, nuclear AFB1 from periportal hepatocytes cleared faster compared to the pericentral region. These local differences in AFB1 clearance may be due to the pericentral expression of cytochrome P450 enzymes that activate AFB1 to protein- and DNA-binding metabolites. In conclusion, the present study shows that large spatio-temporal concentration differences exist within the same tissues and its analysis may provide valuable additional information to conventional toxicokinetic studies.
Collapse
Affiliation(s)
- Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany.
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| | - Reham Hassan
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Maiju Myllys
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Adrian Friebel
- Institute of Computer Science, Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107, Leipzig, Germany
| | - Stefan Hoehme
- Institute of Computer Science, Saxonian Incubator for Clinical Research (SIKT), University of Leipzig, Haertelstraße 16-18, 04107, Leipzig, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376, Stuttgart, Germany
| | - Abdel-Latif Seddek
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Lars Kuepfer
- Institute of Systems Medicine with Focus on Organ Interactions, University Hospital RWTH Aachen, Pauwelsstr. 19, 52074, Aachen, Germany
| | - Benedikt Cramer
- Institute of Food Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 45, 48149, Münster, Germany
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 45, 48149, Münster, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Gisela H Degen
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany.
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Ardeystr. 67, 44139, Dortmund, Germany.
| |
Collapse
|
4
|
Insights into Aflatoxin B1 Toxicity in Cattle: An In Vitro Whole-Transcriptomic Approach. Toxins (Basel) 2020; 12:toxins12070429. [PMID: 32610656 PMCID: PMC7404968 DOI: 10.3390/toxins12070429] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
Aflatoxins, and particularly aflatoxin B1 (AFB1), are toxic mycotoxins to humans and farm animal species, resulting in acute and chronic toxicities. At present, AFB1 is still considered a global concern with negative impacts on health, the economy, and social life. In farm animals, exposure to AFB1-contaminated feed may cause several untoward effects, liver damage being one of the most devastating ones. In the present study, we assessed in vitro the transcriptional changes caused by AFB1 in a bovine fetal hepatocyte-derived cell line (BFH12). To boost the cellular response to AFB1, cells were pre-treated with the co-planar PCB 3,3′,4,4′,5-pentachlorobiphenyl (PCB126), a known aryl hydrocarbon receptor agonist. Three experimental groups were considered: cells exposed to the vehicle only, to PCB126, and to PCB126 and AFB1. A total of nine RNA-seq libraries (three replicates/group) were constructed and sequenced. The differential expression analysis showed that PCB126 induced only small transcriptional changes. On the contrary, AFB1 deeply affected the cell transcriptome, the majority of significant genes being associated with cancer, cellular damage and apoptosis, inflammation, bioactivation, and detoxification pathways. Investigating mRNA perturbations induced by AFB1 in cattle BFH12 cells will help us to better understand AFB1 toxicodynamics in this susceptible and economically important food-producing species.
Collapse
|
5
|
Aflatoxin B1 metabolism: Regulation by phase I and II metabolizing enzymes and chemoprotective agents. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 778:79-89. [DOI: 10.1016/j.mrrev.2018.10.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 10/26/2018] [Indexed: 01/13/2023]
|
6
|
Ilic Z, Mondal TK, Guest I, Crawford DR, Sell S. Participation of liver stem cells in cholangiocarcinogenesis after aflatoxin B1 exposure of glutathione S-transferase A3 knockout mice. Tumour Biol 2018; 40:1010428318777344. [DOI: 10.1177/1010428318777344] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aflatoxin B1, arguably the most potent human carcinogen, induces liver cancer in humans, rats, trout, ducks, and so on, but adult mice are totally resistant. This resistance is because of a detoxifying enzyme, mouse glutathione S-transferase A3, which binds to and inactivates aflatoxin B1 epoxide, preventing the epoxide from binding to DNA and causing mutations. Glutathione S-transferase A3 or its analog has not been detected in any of the sensitive species, including humans. The generation of a glutathione S-transferase A3 knockout (represented as KO or -/-) mice has allowed us to study the induction of liver cancer in mice by aflatoxin B1. In contrast to the induction of hepatocellular carcinomas in other species, aflatoxin B1 induces cholangiocarcinomas in GSTA3-/- mice. In other species and in knockout mice, the induction of liver cancer is preceded by extensive proliferation of small oval cells, providing additional evidence that oval cells are bipolar stem cells and may give rise to either hepatocellular carcinoma or cholangiocarcinoma depending on the nature of the hepatocarcinogen and the species of animal. The recent development of mouse oval cell lines in our laboratory from aflatoxin B1-treated GSTA3-/- mice should provide a new venue for study of the properties and potential of putative mouse liver stem cells.
Collapse
Affiliation(s)
- Zoran Ilic
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Tapan K Mondal
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Ian Guest
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | | | - Stewart Sell
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
7
|
Wang H, Muhammad I, Li W, Sun X, Cheng P, Zhang X. Sensitivity of Arbor Acres broilers and chemoprevention of aflatoxin B 1-induced liver injury by curcumin, a natural potent inducer of phase-II enzymes and Nrf2. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 59:94-104. [PMID: 29550706 DOI: 10.1016/j.etap.2018.03.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/10/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
In this study, we scrutinized the effects of curcumin and AFB1 supplemented diet alone or in combination on phase-ӀӀ enzymes. Histopathological examination showed that after 28 days, AFB1 (5.0 mg/kg diet) induced liver injury in broilers, but curcumin supplementation partially ameliorated liver injury in a dose-dependent manner. RT-PCR data revealed that AFB1 significantly (p < 0.01) down-regulated Nrf2 and its downstream genes mRNA expression level. Moreover, Western blot analysis showed that Nrf2, GSTM2, and GSTA3 protein expression level was markedly (p < 0.01) reduced in AFB1-fed group. However, curcumin supplementation ameliorated AFB1-induced liver injury via enhancing phase-ӀӀ enzymes expressions and activity. HPLC results showed that curcumin increased AFB1-GSH conjugation in-vitro in liver cytosol. Surprisingly, similar trends were noted in mRNA, protein expression level of Nrf2 and its downstream genes at day 35, one week after the withdrawal of AFB1 and curcumin from the diet, showing the preventive effects of curcumin.
Collapse
Affiliation(s)
- He Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China
| | - Ishfaq Muhammad
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China
| | - Wei Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China
| | - Xiaoqi Sun
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China
| | - Ping Cheng
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China
| | - Xiuying Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development. Faculty of Basic Veterinary Science, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, PR China.
| |
Collapse
|
8
|
Soler L, Oswald I. The importance of accounting for sex in the search of proteomic signatures of mycotoxin exposure. J Proteomics 2018; 178:114-122. [DOI: 10.1016/j.jprot.2017.12.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022]
|
9
|
Kutsuno Y, Itoh T, Tukey RH, Fujiwara R. Glucuronidation of drugs and drug-induced toxicity in humanized UDP-glucuronosyltransferase 1 mice. Drug Metab Dispos 2014; 42:1146-52. [PMID: 24764149 PMCID: PMC4053997 DOI: 10.1124/dmd.114.057083] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/24/2014] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are phase II drug-metabolizing enzymes that catalyze glucuronidation of various drugs. Although experimental rodents are used in preclinical studies to predict glucuronidation and toxicity of drugs in humans, species differences in glucuronidation and drug-induced toxicity have been reported. Humanized UGT1 mice in which the original Ugt1 locus was disrupted and replaced with the human UGT1 locus (hUGT1 mice) were recently developed. In this study, acyl-glucuronidations of etodolac, diclofenac, and ibuprofen in liver microsomes of hUGT1 mice were examined and compared with those of humans and regular mice. The kinetics of etodolac, diclofenac, and ibuprofen acyl-glucuronidation in hUGT1 mice were almost comparable to those in humans, rather than in mice. We further investigated the hepatotoxicity of ibuprofen in hUGT1 mice and regular mice by measuring serum alanine amino transferase (ALT) levels. Because ALT levels were increased at 6 hours after dosing in hUGT1 mice and at 24 hours after dosing in regular mice, the onset pattern of ibuprofen-induced liver toxicity in hUGT1 mice was different from that in regular mice. These data suggest that hUGT1 mice can be valuable tools for understanding glucuronidations of drugs and drug-induced toxicity in humans.
Collapse
Affiliation(s)
- Yuki Kutsuno
- School of Pharmacy, Kitasato University, Tokyo, Japan (Y.K., T.I., R.F.); and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Tomoo Itoh
- School of Pharmacy, Kitasato University, Tokyo, Japan (Y.K., T.I., R.F.); and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Robert H Tukey
- School of Pharmacy, Kitasato University, Tokyo, Japan (Y.K., T.I., R.F.); and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| | - Ryoichi Fujiwara
- School of Pharmacy, Kitasato University, Tokyo, Japan (Y.K., T.I., R.F.); and Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California (R.H.T.)
| |
Collapse
|
10
|
Diao E, Shan C, Hou H, Wang S, Li M, Dong H. Structures of the ozonolysis products and ozonolysis pathway of aflatoxin B1 in acetonitrile solution. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9364-9370. [PMID: 22920447 DOI: 10.1021/jf302528e] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The ozonolysis of aflatoxin B(1) (400 μg/mL) in acetonitrile solution was conducted with an ozone concentration of 6.28 mg/L at the flow rate of 60 mL/min for different times. The results showed that ozone was an effective detoxification agent because of its powerful oxidative role. Thin-layer chromatography and liquid chromatography-quadrupole time-of-flight mass spectra were applied to confirm and identify the ozonolysis products of aflatoxin B(1). A total of 13 products were identified, and 6 of them were main products. The structural identification of these products provided effective information for understanding the ozonolysis pathway of aflatoxin B(1). Two ozonolysis pathways were proposed on the basis of the accurate mass and molecular formulas of these product ions. Nine ozonolysis products came from the first oxidative pathway based on the Criegee mechanism, and the other four products were produced from the second pathway based on the oxidative and electrophilic reactions of ozone. According to the toxicity mechanism of aflatoxin B(1) to animals, the toxicity of aflatoxin B(1) was significantly reduced because of the disappearance of the double bond on the terminal furan ring or the lactone moiety on the benzene ring.
Collapse
Affiliation(s)
- Enjie Diao
- College of Food Science and Engineering, Shandong Agricultural University, Tai'an 271018, People's Republic of China
| | | | | | | | | | | |
Collapse
|
11
|
Ilic Z, Crawford D, Vakharia D, Egner PA, Sell S. Glutathione-S-transferase A3 knockout mice are sensitive to acute cytotoxic and genotoxic effects of aflatoxin B1. Toxicol Appl Pharmacol 2009; 242:241-6. [PMID: 19850059 DOI: 10.1016/j.taap.2009.10.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 10/06/2009] [Accepted: 10/07/2009] [Indexed: 12/29/2022]
Abstract
Aflatoxin B1 (AFB1) is a major risk factor for hepatocellular carcinoma (HCC) in humans. However, mice, a major animal model for the study of AFB1 carcinogenesis, are resistant, due to high constitutive expression, in the mouse liver, of glutathione S-transferase A3 subunit (mGSTA3) that is lacking in humans. Our objective was to establish that a mouse model for AFB1 toxicity could be used to study mechanisms of toxicity that are relevant for human disease, i.e., an mGSTA3 knockout (KO) mouse that responds to toxicants such as AFB1 in a manner similar to humans. Exons 3-6 of the mGSTA3 were replaced with a neomycin cassette by homologous recombination. Southern blotting, RT-PCR, Western blotting, and measurement of AFB1-N(7)-DNA adduct formation were used to evaluate the mGSTA3 KO mice. The KO mice have deletion of exons 3-6 of the mGSTA3 gene, as expected, as well as a lack of mGSTA3 expression at the mRNA and protein levels. Three hours after injection of 5 mg/kg AFB1, mGSTA3 KO mice have more than 100-fold more AFB1-N(7)-DNA adducts in their livers than do similarly treated wild-type (WT) mice. In addition, the mGSTA3 KO mice die of massive hepatic necrosis, at AFB1 doses that have minimal toxic effects in WT mice. We conclude that mGSTA3 KO mice are sensitive to the acute cytotoxic and genotoxic effects of AFB1, confirming the crucial role of GSTA3 subunit in protection of normal mice against AFB1 toxicity. We propose the mGSTA3 KO mouse as a useful model with which to study the interplay of risk factors leading to HCC development in humans, as well as for testing of additional possible functions of mGSTA3.
Collapse
Affiliation(s)
- Zoran Ilic
- Ordway Research Institute, Albany, NY 12208, USA.
| | | | | | | | | |
Collapse
|
12
|
Jhee EC, Ho LL, Tsuji K, Gopalan P, Lotlikar PD. Mechanism of Inhibition of Aflatoxin B1- Dna Binding in the Liver by Butylated Hydroxyanisole Pretreatment of Rats. ACTA ACUST UNITED AC 2008. [DOI: 10.3109/15569548909059746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Lotlikar PD. Metabolic Basis for Susceptibility and Resistance to Aflatoxin B1Hepatocarcinogenesis in Rodents. ACTA ACUST UNITED AC 2008. [DOI: 10.3109/15569548909059742] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
14
|
Gopalan P, Santhanam K, Jhee EC, Jensen DE, Lotlikar PD. Microsome Mediated Aflatoxin B1-8,9-Epoxide Conjugation to Glutathione in Presence of Purified Hepatic Glutathione S-Transferases from Control and Butylated Hydroxyanisole Pretreated Rats. ACTA ACUST UNITED AC 2008. [DOI: 10.3109/15569548909059747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Bedard LL, Massey TE. Aflatoxin B1-induced DNA damage and its repair. Cancer Lett 2006; 241:174-83. [PMID: 16458422 DOI: 10.1016/j.canlet.2005.11.018] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2005] [Revised: 11/16/2005] [Accepted: 11/18/2005] [Indexed: 01/09/2023]
Abstract
Aflatoxin B(1) (AFB(1))-N(7)-guanine is the predominant adduct formed upon the reaction of AFB(1)-8,9-exo-epoxide with guanine residues in DNA. AFB(1)-N(7)-guanine can convert to the ring-opened formamidopyrimidine, or the adducted strand can undergo depurination. AFB(1)-N(7)-guanine and AFB(1)-formamidopyrimidine are thought to be predominantly repaired by nucleotide excision repair in bacteria, yeast and mammals. Although AFB(1)-formamidopyrimidine is removed less efficiently than AFB(1)-N(7)-guanine in mammals, both lesions are repaired with equal efficiencies in bacteria, reflecting differences in damage recognition between bacterial and mammalian repair systems. Furthermore, DNA repair activity and modulation of repair by AFB(1) seem to be major determinants of susceptibility to AFB(1)-induced carcinogenesis.
Collapse
Affiliation(s)
- Leanne L Bedard
- Department of Pharmacology and Toxicology, Queen's University, Botterell Hall, Kingston, Ont., Canada K7L 3N6
| | | |
Collapse
|
16
|
Fatemi F, Allameh A, Dadkhah A, Forouzandeh M, Kazemnejad S, Sharifi R. Changes in hepatic cytosolic glutathione S-transferase activity and expression of its class-P during prenatal and postnatal period in rats treated with aflatoxin B1. Arch Toxicol 2006; 80:572-9. [PMID: 16501953 DOI: 10.1007/s00204-006-0076-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2005] [Accepted: 01/23/2006] [Indexed: 10/25/2022]
Abstract
The effect of aflatoxin B1 (AFB1) on the expression of glutathione S-transferase-P (GST-P) which is the major isoform of GST in developmental stages has been investigated in rat liver during prenatal and postnatal stages. Following administration of AFB1 (0, 0.5, 1.0, 2.0, 3.0 or 4.0 mg/kg bw) injected I.P on day 8.5 of gestation the number of dead or reabsorbed fetuses and malformed embryos were recorded. Then the fetal livers were processed for measurement of total GST and GST-P activities, using 1-chloro-2,4-dinitrobenzene (CDNB) and ethacrynic acid (ETA) as substrates respectively. RT-PCR using rat GST-P specific primers was performed on mRNA extracted from livers. Besides, the effects of AFB1 on hepatic GST and GST-P were assessed in groups of suckling rats directly injected with the toxin. The results show that a single dose of AFB1 (1.0 or 2.0 mg/kg bw) caused approximately 50-60% depletion in fetal liver GST towards CDNB. Postnatal experiments revealed that liver GST (using CDNB as substrate) was significantly induced (approximately 40%) in suckling rats injected with a single dose of AFB1 (3.0 mg AFB1/kg) 24 h before killing. Liver GST-P expression was unaffected due to AFB1 exposures of rats before and after the birth. This finding was substantiated by western blotting and RT-PCR techniques. These data suggest that AFB1-related induction in rat liver total GST after birth may be implicated in protective mechanisms against AFB1. In contrast, inhibition of this enzyme in fetal liver following placental transfer of the carcinogen may explain high susceptibility of fetal cells to trans-plancental aflatoxins. Furthermore, lack of influence of AFB1 on GST-P expression in developmental stages can role out the involvement of this class of GST in AFB1 biotransformation.
Collapse
Affiliation(s)
- Faezeh Fatemi
- Department of Biochemistry Faculty of Medical Science, Tarbiat Modarres University, P.O. Box 14115-331, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
17
|
Tulayakul P, Sakuda S, Dong KS, Kumagai S. Comparative activities of glutathione-S-transferase and dialdehyde reductase toward aflatoxin B1 in livers of experimental and farm animals. Toxicon 2005; 46:204-9. [PMID: 15964045 DOI: 10.1016/j.toxicon.2005.03.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Accepted: 03/25/2005] [Indexed: 11/25/2022]
Abstract
In order to gain a better understanding of the relative activities of glutathione-S-transferase (GST) and aldehyde reductase toward aflatoxin B1 (AFB1) in relation to the variation of species susceptibilities, we studied the in vitro cytosolic GST and reductase activities in liver tissues from male Fischer rats, ICR mice and golden hamsters, adult male rainbow trouts and female piglets. The GST activity was determined by incubating the liver cytosol with glutathione (GSH) and AFB1 in the presence of the hamster liver microsomes to metabolize AFB1 to AFB1-8, 9-epoxide. The reaction product, AFB1 and GSH conjugate (AFB1-GSH), was quantified with HPLC. The reductase activity was determined by incubating liver cytosol with AFB1-dialdehyde, followed by the quantification of the metabolic product, AFB1-dialcohol, with HPLC. All the animal species possessed the GST activities, and AFB1-GSH formed increasingly with the increase of the AFB1 concentration according to the model of first-order enzyme reaction kinetics. The V(max) and K(m) values of the GST activities in rodent species were higher and lower, respectively, than those in the trout and pig, being consistent with the relative susceptibilities to AFB1 of these animal species. However, no relationship was noted between the reductase activity and species susceptibility. Thus, the result of this study shows that GST toward AFB1, but not aldehyde reductase, is a determinant of the variation of species susceptibilities.
Collapse
Affiliation(s)
- P Tulayakul
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | |
Collapse
|
18
|
Thornton AS, Oda Y, Stuart GR, Holcroft J, de Boer JG. The dioxin TCDD protects against aflatoxin-induced mutation in female rats, but not in male rats. Mutat Res 2004; 561:147-52. [PMID: 15238239 DOI: 10.1016/j.mrgentox.2004.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2003] [Revised: 04/30/2004] [Accepted: 05/04/2004] [Indexed: 04/30/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental contaminant and a potent carcinogen in laboratory rodents. When combined with other environmental toxins, it has been shown to increase the (geno)toxicity of some compounds. In this study, the effect of TCDD on the mutagenicity of aflatoxin-B1 (AFB1) was examined in the rat liver using a lacI transgenic rodent mutation assay. AFB1 induces GC-->TA transversions. Since TCDD is known to have a differential effect in male and female rodents, both sexes were studied. The data showed that a 6-week pre-exposure to TCDD had no significant effect on the frequency of aflatoxin-induced mutation in the liver of male rats. However, the TCDD treatment completely prevented the aflatoxin-induced transversion mutations in female animals.
Collapse
Affiliation(s)
- A S Thornton
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada V8W 3N5
| | | | | | | | | |
Collapse
|
19
|
Vassallo JD, Hicks SM, Daston GP, Lehman-McKeeman LD. Metabolic Detoxification Determines Species Differences in Coumarin-Induced Hepatotoxicity. Toxicol Sci 2004; 80:249-57. [PMID: 15141102 DOI: 10.1093/toxsci/kfh162] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hepatotoxicity of coumarin is attributed to metabolic activation to an epoxide intermediate, coumarin 3,4-epoxide (CE). However, whereas rats are most susceptible to coumarin-induced hepatotoxicity, formation of CE is greatest in mouse liver microsomes, a species showing little evidence of hepatotoxicity. Therefore, the present work was designed to test the hypothesis that detoxification of CE is a major determinant of coumarin hepatotoxicity. CE can either rearrange spontaneously to o-hydroxyphenylacetaldehyde (o-HPA) or be conjugated with gluatathione (GSH). o-HPA is hepatotoxic and is further detoxified by oxidation to o-hydroxyphenylacetic acid (o-HPAA). In vitro experiments were conducted using mouse liver microsomes to generate a constant amount of CE, and cytosols from F344 rats, B6C3F1 mice, and human liver were used to characterize CE detoxification. All metabolites were quantified by HPLC methods with UV detection. In rats and mice, GSH conjugation occurred non-enzymatically and through glutathione-S-transferases (GSTs), and the kinetics of GSH conjugation were similar in rats and mice. In rat liver cytosol, oxidation of o-HPA to o-HPAA was characterized with a high affinity K(m) of approximately 12 microM, and a V(max) of approximately 1.5 nmol/min/mg protein. In contrast, the K(m) and V(max) for o-HPA oxidation in mouse liver cytosol were approximately 1.7 microM and 5 nmol/min/mg protein, respectively, yielding a total intrinsic clearance through oxidation to o-HPAA that was 20 times higher in mouse than in rats. Human cytosols (two separate pools) detoxified CE through o-HPA oxidation with an apparent K(m) of 0.84 microM and a V(max) of 5.7 nmol/min/mg protein, for a net intrinsic clearance that was more than 50 times higher than the rat. All species also reduced o-HPA to o-hydroxyphenylethanol (o-HPE), but this was only a major reaction in rats. In the presence of a metabolic reaction replete with all necessary cofactors, GSH conjugation accounted for nearly half of all CE metabolites in rat and mouse, whereas the GSH conjugate represented only 10% of the metabolites in human cytosol. In mouse, o-HPAA represented the major ring-opened metabolite, accounting for the remaining 50% of metabolites, and in human cytosol, o-HPAA was the major metabolite, representing nearly 90% of all CE metabolites. In contrast, no o-HPAA was detected in rats, whereas o-HPE represented a major metabolite. Collectively, these in vitro data implicate o-HPA detoxification through oxidation to o-HPAA as the major determinant of species differences in coumarin-induced hepatotoxicity.
Collapse
Affiliation(s)
- Jeffrey D Vassallo
- Miami Valley Laboratories, The Procter and Gamble Company, 11810 East Miami River Road, Cincinnati, Ohio 45252, USA.
| | | | | | | |
Collapse
|
20
|
Klein PJ, Van Vleet TR, Hall JO, Coulombe RA. Effects of dietary butylated hydroxytoluene on aflatoxin B1-relevant metabolic enzymes in turkeys. Food Chem Toxicol 2003; 41:671-8. [PMID: 12659720 DOI: 10.1016/s0278-6915(02)00332-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We have shown previously that the extreme sensitivity of turkeys to aflatoxin B(1) (AFB(1)) is due to a combination of efficient AFB(1) activation by cytochrome P450s (CYPs) 1A and deficient detoxification by glutathione S-transferases (GSTs). Phenolic antioxidants such as butylated hydroxytoluene (BHT) have been shown to be chemoprotective in some animal models due, in part, to modulation of AFB(1)-relevant phase I and/or phase II activities, and we wished to determine whether BHT has a similar effect in turkeys. Ten-day-old male turkeys were maintained on diets amended with 1000 or 4000 ppm of BHT for 10 days, then sampled. Hepatic microsomal CYP 1A activity as well as conversion of AFB(1) to the putative toxic metabolite, the exo-AFB(1)-8,9-epoxide (AFBO), were significantly lower compared with control. Conversely, dietary BHT significantly increased activities of several isoforms of hepatic cytosolic GST, as well quinone oxidoreductase (QOR). Western immunoblotting confirmed that dietary BHT increased expression of homologues to rodent GST isoforms Yc1, Yc2 and Ya. There was, however, no observable BHT-related increase in GST-mediated specific conjugation with microsomally-generated AFBO. In total, our data indicates that dietary BHT modulates a variety of AFB(1)-relevant phase I and phase II enzymes, while having no measurable effect towards specific AFB(1) detoxification by GST.
Collapse
Affiliation(s)
- P J Klein
- Department of Veterinary Sciences, and Graduate Program in Toxicology, Utah State University, Logan 84322-4620, USA
| | | | | | | |
Collapse
|
21
|
Esaki H, Kumagai S. Glutathione-S-transferase activity toward aflatoxin epoxide in livers of mastomys and other rodents. Toxicon 2002; 40:941-5. [PMID: 12076648 DOI: 10.1016/s0041-0101(02)00090-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In order to study the liver glutathione-S-transferase (GST) activity toward aflatoxin B1 (AFB1) epoxide in mastomys in comparison with other rodents, we performed in vitro studies of the cytosolic GST activity toward AFB1-epoxide using mastomys, rat, mouse and hamster liver. Also AFB1 metabolism by liver microsomes including formation of AFB1-DNA adducts was studied. Cytosolic GST activity toward AFB1-epoxide was highest in mastomys liver, and higher in the hamster and mouse livers than in the rat liver, correlating well with the differences of the sensitivity of these species to the toxicity of AFB1. However, no relationship was noted between the sensitivity of a given species to the toxicity of AFB1 and the microsomal activity of binding of AFB1 to DNA or metabolizing AFB1 to AFM1, AFQ1 and AFP1. These results demonstrate the importance of the GST mediated AFB1-epoxide conjugation with glutathione in determining the differing sensitivities of these species to AFB1 toxicity. The extremely high activity of GST in mastomys indicates that this species would be a good model animal for studying GST toward AFB1-epoxide.
Collapse
Affiliation(s)
- H Esaki
- Graduate School of Agricultural and Life Sciences, University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Japan
| | | |
Collapse
|
22
|
Klein PJ, Van Vleet TR, Hall JO, Coulombe RA. Dietary butylated hydroxytoluene protects against aflatoxicosis in Turkeys. Toxicol Appl Pharmacol 2002; 182:11-9. [PMID: 12127258 DOI: 10.1006/taap.2002.9433] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Turkeys are among the most sensitive species to the toxic effects of the mycotoxin aflatoxin B(1) (AFB(1)). In mammals, dietary antioxidants, such as butylated hydroxytoluene (BHT), have been shown to lessen the toxic effects of AFB(1) by various mechanisms. To test whether BHT protects against aflatoxicosis in turkeys, we supplemented the feed of 10-day-old male white turkeys with low (1000 ppm) and high (4000 ppm) BHT for 20 days. AFB(1) (1 ppm) was then added to the diets and continued for another 10 days. Birds in the AFB(1)-only group had a lower weight gain, a condition that had returned to near control in groups fed diets containing AFB(1) + BHT. Significant elevations in serum aspartate transaminase, alanine aminotransferase, and lactate dehydrogenase, which were evident in the AFB(1) group, were reversed in the AFB(1) + BHT groups. Histopathology revealed hepatic submassive necrotic lesions and biliary hyperplasia, the severity of which was lessened in the AFB(1) + BHT-treated birds. Hepatocellular hydropic degeneration was observed in the BHT-only group, but not in the AFB(1) + BHT groups. This condition associated with BHT treatment was found in a separate study to be reversible and without any long-term adverse effects. These results indicate that BHT counteracts many of the deleterious effects caused by AFB(1) and that this antioxidant may prove to be a viable feed additive for the reduction of aflatoxicosis in turkeys.
Collapse
Affiliation(s)
- Patrick J Klein
- Department of Veterinary Scieces and Graduate Program in Toxicology, Utah State University, Lagan, Utah 84322-4620, USA
| | | | | | | |
Collapse
|
23
|
Klein PJ, Van Vleet TR, Hall JO, Coulombe RA. Biochemical factors underlying the age-related sensitivity of turkeys to aflatoxin B(1). Comp Biochem Physiol C Toxicol Pharmacol 2002; 132:193-201. [PMID: 12106896 DOI: 10.1016/s1532-0456(02)00065-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Poultry are some of the most sensitive species to the toxic effects of aflatoxin B(1) (AFB(1)), and younger poultry are more sensitive to this mycotoxin. To elucidate the mechanisms for this age-related susceptibility, various enzyme activities relevant to AFB(1) were measured in liver microsomes prepared from male turkeys 9, 41 and 65 days of age. Hepatic microsomal o-dealkylation of methoxy- and pentoxyresorufin significantly increased, while that of ethoxyresorufin decreased with age. Microsomal AFB(1) activation to the reactive AFB(1)-8,9-epoxide (AFBO) was most efficient in the youngest birds, with apparent K(m) and V(max) values of 168 and 19, 110 and 6, and 116 microM and 10 nmol/mg/min for 9, 41 and 65-day-old birds, respectively. The activity of hepatic cytosolic glutathione S-transferases (GSTs) was deficient in the youngest age group, but were higher in the older groups. There was also an age-related increase in the expression of GST isoforms Yc, Yc(2), as well as AFB(1)-aldehyde reductase (AFAR). However, livers from all ages lacked specific GST-mediated conjugation of AFBO, indicating that turkeys are deficient in this key AFB(1)-detoxification pathway. Our data indicate that efficient activation may underlie the extreme sensitivity of young turkeys to the toxic effects of AFB(1).
Collapse
Affiliation(s)
- Patrick J Klein
- Department of Veterinary Sciences, and Graduate Program in Toxicology, Utah State University, Logan, UT 84322-4620, USA
| | | | | | | |
Collapse
|
24
|
Dvorska JE, Surai PF, Speake BK, Sparks NH. Effect of the mycotoxin aurofusarin on the antioxidant composition and fatty acid profile of quail eggs. Br Poult Sci 2001; 42:643-9. [PMID: 11811917 DOI: 10.1080/00071660120088470] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
1. The effect of the mycotoxin aurofusarin on the antioxidant composition and fatty acid profile of quail eggs was investigated. 2. Thirty eight 45-d-old Japanese quails were divided into two groups (experimental and control, 15 females +4 males in each group) and were fed on a maize-soya diet balanced in all nutrients. The diet of the experimental quails was supplemented with aurofusarin at the level of 26.4 mg/kg feed in the form of Fusarium graminearum culture enriched with aurofusarin. At the beginning and after 2, 4 and 8 week supplementation periods, eggs were collected and analysed. After 8 weeks of supplementation, experimental quails were fed on unsupplemented diet during the next 4 weeks and eggs were collected after 2 and 4 weeks on such a diet and analysed. 3. Aurofusarin caused a significant (P<0.05) decrease in vitamins E, A, total carotenoid, lutein and zeaxanthin concentrations and significantly (P<0.05) increased egg yolk susceptibility to lipid peroxidation. During two weeks on the diet without aurofusarin the levels of carotenoids in the egg yolk returned to the initial level, vitamins A and E returned to the initial level during 4 weeks on the same unsupplemented diet. 4. Dietary supplementation with aurofusarin was associated with a significant (P<0.01) decrease in the docosahexaenoic acid proportion in the phospholipid, cholesteryl ester and free fatty acid fractions of the egg yolk. At the same time the proportion of linoleic acid in the phospholipid, free fatty acid and triacylglycerol fractions significantly (P<0.05) increased. 5. It is concluded that mycotoxin aurofusarin is detrimental to the nutritional quality of eggs.
Collapse
|
25
|
Yatim AM, Sachan DS. Carnitine alters binding of aflatoxin to DNA and proteins in rat hepatocytes and cell-free systems. J Nutr 2001; 131:1903-8. [PMID: 11435505 DOI: 10.1093/jn/131.7.1903] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The objective of this study was to determine effects of L-carnitine on aflatoxin B(1) (AFB(1))-DNA adduct formation in isolated rat hepatocytes, its dose response, specificity and mode of action. All experiments were conducted in either freshly isolated rat hepatocytes or cell-free systems. There was negative linear correlation between the dosage of carnitine and formation of [(3)H]AFB(1)-DNA adducts in the hepatocytes; however, the partitioning of AFB(1) into cellular compartments was not affected by carnitine. The attenuating effect of carnitine on AFB(1)-DNA adduct formation was also present in a cell-free system, but there was lack of specificity because acetylcarnitine and gamma-aminobutyric acid (GABA) were equally effective. Carnitine appears to interfere with bioactivation of AFB(1) and binding of AFB(1)-epoxide to DNA. On the contrary, carnitine enhanced the binding of AFB(1) and its epoxide to microsomal proteins, plasma proteins and bovine serum albumin. These results indicate that carnitine diverts AFB(1)-epoxide away from DNA by promoting binding to proteins. We conclude that modulation of AFB(1) binding to proteins and DNA by carnitine alters the carcinogenic and hepatotoxic potential of AFB(1) and poses concerns about the human AFB(1)-exposure data based on the AFB(1)-albumin adduct concentrations as a biomarker.
Collapse
Affiliation(s)
- A M Yatim
- Department of Nutrition, College of Human Ecology, University of Tennessee, Knoxville, TN 37996-1900, USA
| | | |
Collapse
|
26
|
Chen HW, Yang JJ, Tsai CW, Wu JJ, Sheen LY, Ou CC, Lii CK. Dietary fat and garlic oil independently regulate hepatic cytochrome p(450) 2B1 and the placental form of glutathione S-transferase expression in rats. J Nutr 2001; 131:1438-43. [PMID: 11340096 DOI: 10.1093/jn/131.5.1438] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The individual and combined effects of dietary fat and garlic oil on two drug-metabolizing enzymes, cytochrome P(450) 2B1 and the placental form of glutathione (GSH) S-transferase (PGST), in rat liver were examined in this study. Rats were fed a low corn oil, high corn oil or high fish oil diet and received various amount of garlic oil (0, 30, 80, 200 mg/kg body) orally three times per week for 6 wk. The fat energy in the low and high fat diets accounted for 11.6 and 45.7% of total energy, respectively. Final body weights did not differ among the three dietary fat groups and were not affected by garlic oil treatment. The fatty acid profile in hepatic phospholipids revealed higher eicosapentaenoic acid [20:5(n-3)] and docosahexaenoic acid [22:6(n-3)] levels in the fish oil-fed group than in the low and high corn oil-fed groups (P < 0.05). In contrast, the corn oil-fed groups had greater hepatic phospholipid arachidonic acid [20:4(n-6)] levels (P < 0.05). Both dietary fat and garlic oil significantly affected hepatic cytochrome 7-pentoxyresorufin O-dealkylase (PROD) activity and GST activity toward ethacrynic acid. Rats fed the high fish oil diet had 85 and 51% higher PROD activity compared with those fed the low or the high corn oil diet, respectively (P < 0.05). The GST activity in the high fish oil and the high corn oil groups was 33 and 18% higher than that in the low corn oil group (P < 0.05), respectively, and the GST activity in rats fed the high fish oil diet was higher than in those fed the high corn oil diet (P < 0.05). Garlic oil dose-dependently increased GST activity. No interaction between dietary fat and garlic oil on PROD or GST activity was noted. Northern and Western blot analysis revealed that dietary fish oil increased both cytochrome P(450) 2B1 and PGST mRNA and protein levels. Cytochrome P(450) 2B1 and PGST mRNA and protein levels were also dose-dependently increased by garlic oil treatment. The effects of garlic oil and dietary fat on P(450) 2B1 and PGST mRNA and protein expression were independent. These results indicate that dietary fat and garlic oil independently modulate P(450) 2B1 and PGST expression at transcriptional and/or post-transcriptional stages.
Collapse
Affiliation(s)
- H W Chen
- Department of Nutrition, Chung Shan Medical College, National Chung Cheng University, Min-Hsiung 621, Taiwan
| | | | | | | | | | | | | |
Collapse
|
27
|
Allameh A, Farahani M, Zarghi A. Kinetic studies of aflatoxin B1-glutathione conjugate formation in liver and kidneys of adult and weanling rats. Mech Ageing Dev 2000; 115:73-83. [PMID: 10854630 DOI: 10.1016/s0047-6374(00)00105-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aflatoxin B1(AFB1)-glutathione(GSH) conjugation is the major pathway for the detoxification of aflatoxin metabolites. This reaction is catalysed by GSH S-transferase (GST) and play a major role in modulation of AFB1 adduct formation to nuclear DNA. Changes recorded in hepatic GST activity during development of rats can alter the balance between AFB1-GSH conjugation and AFB1-DNA adduct formation. Measurment of cytosolic GST using 1-chloro-2,4-dinitrobenzene (CDNB) as the substrate showed that the enzyme activity is initially lower in weanling tissues as compared to that of adults. But nevertheless hepatic and renal cytosolic GST activity is increased significantly in growing rats pretreated with AFB1. Kinetic studies of AFB1-GSH conjugate formation in kidneys and livers of the two-age groups of rats treated with a single i.p. dose of AFB1 (400 microg/kg b.w.) revealed that at the end of 24 h of AFB1 administration the rate of the conjugate formation in kidneys of immature rats was approximately twice of that measured in adults. Age-related differences in the GST activity as well as AFB1-GSH conjugation was more pronounced in kidneys. The conjugate formation in kidneys of growing rats during 6-24 h following AFB1 administration shows that urinary excretion of aflatoxin metabolites is relatively rapid in growing rats. The major portion of the AFB1-GSH is formed in liver but contribution of the renal tissue to the formation of detoxification metabolites can not be ruled out. These data demonstrate that aflatoxin metabolites are eliminated more efficiently from kidneys of a growing rat. AFB1-induced GST induction in renal tissues of growing animals during 24 h of the carcinogen administration could be considered as an important mechanism for GSH conjugate formation and aflatoxin detoxification. Therefore GST induction in response to hepatotoxic drugs can confer resistance to young animals being exposed for the first time to such drugs. It is also worthmentioning that the GST activity measured before AFB1 administration does not reflect the rate of AFB1 detoxification via GSH conjugation.
Collapse
Affiliation(s)
- A Allameh
- Department of Biochemistry, Faculty of Medical Sciences, Tarbiat Modaress University, PO Box 14115-111, Tehran, Iran.
| | | | | |
Collapse
|
28
|
Klein PJ, Buckner R, Kelly J, Coulombe RA. Biochemical basis for the extreme sensitivity of turkeys to aflatoxin B(1). Toxicol Appl Pharmacol 2000; 165:45-52. [PMID: 10814552 DOI: 10.1006/taap.2000.8926] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Poultry are the most susceptible food animal species to the toxic effects of the mycotoxin aflatoxin B(1) (AFB(1)). Feed contaminated with even small amounts of AFB(1) results in significant adverse health effects in poultry. The purpose of this study was to explain the biochemical mechanism(s) for this extreme sensitivity. We measured microsomal activation of AFB(1) to the AFB(1)-8,9-epoxide (AFBO), the putative toxic intermediate, as well as cytosolic glutathione S-transferase (GST)-mediated detoxification of AFBO, in addition to other hepatic phase I and phase II enzyme activities, in 3-week-old male Oorlop strain turkeys. Liver microsomes prepared from these turkeys activated AFB(1) in vitro with an apparent K(m) of 109 microM and a V(max) of 1.25 nmol/mg/min. Preliminary evidence for the involvement of cytochromes P450 (CYP) 1A2 and, to a lesser extent, 3A4 for AFB(1) activation was assessed by the use of specific mammalian CYP inhibitors. The possible presence of avian orthologues of these CYPs was supported by activity toward ethoxyresorufin and nifedipine, as well as by Western immunoblotting using antibodies to human CYPs. Cytosol prepared from turkey livers exhibited GST-mediated conjugation of 1-chloro-2,4-dinitrobenzene (CDNB) and 3,4-dichloronitrobenzene (DCNB), but at a much lower rate than that observed in other species. Western immunoblotting indicated the presence of alpha and sigma class GSTs and another AFB(1)-detoxifying enzyme, AFB(1)-aldehyde reductase (AFAR). Turkey liver cytosol also had quinone oxidoreductase (QOR) activity. Importantly, cytosol exhibited no measurable GST-mediated detoxification of microsomally activated AFB(1), indicating that turkeys are deficient in the most crucial AFB(1)-detoxification pathway. In total, our data indicate that the extreme sensitivity of turkeys to AFB(1) may be attributed to a combination of efficient AFB(1) activation and deficient detoxification by phase II enzymes, such as GSTs.
Collapse
Affiliation(s)
- P J Klein
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, 84322-4620, USA
| | | | | | | |
Collapse
|
29
|
Kumagai S, Sugita-Konishi Y, Hara-Kudo Y, Ito Y, Noguchi Y, Yamamoto Y, Ogura A. The fate and acute toxicity of aflatoxin B1 in the mastomys and rat. Toxicon 1998; 36:179-88. [PMID: 9604291 DOI: 10.1016/s0041-0101(97)00071-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The fate and acute toxicity of aflatoxin B1 (AFB1) were studied in the mastomys (Praomys coucha) and compared with Fischer rats. The experiment regarding the fate of [3H]AFB1 showed that the radioactivity was excreted mainly through the feces, more rapidly in the mastomys than in the rat, regardless of whether [3H]AFB1 was given orally or intravenously. The levels of radioactivity bound to the liver DNA were lower in the mastomys than in the rat, indicating that the levels of AFB1 binding to the macromolecules in the liver were lower in the mastomys. Consistent with such differences in the fate of AFB1 between the two species, the mastomys were far more resistent to the acute effects of AFB1 than were the rats. Oral administration of AFB1 at a dose of 1.0 mg/kg to rats caused marked microscopic changes in the liver, involving hepatic necrosis and proliferation of bile ducts, but at a dose of 4.0 mg/kg to mastomys caused no pathological changes in the liver or kidneys, and at a dose of 10.0 mg/kg caused only glycogen deposition in hepatic cells in a limited area. The observed differences in susceptibility to the toxic effects of AFB1 and in the fate of AFB1 between the two species are in accord with our previous finding that liver cytosol in the mastomys inhibits microsome-mediated AFB1-DNA binding in vitro more strongly than in rat liver.
Collapse
Affiliation(s)
- S Kumagai
- Department of Biomedical Food Research, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Yanagimoto T, Itoh S, Sawada M, Kamataki T. Mouse cytochrome P450 (Cyp3a11): predominant expression in liver and capacity to activate aflatoxin B1. Arch Biochem Biophys 1997; 340:215-8. [PMID: 9143324 DOI: 10.1006/abbi.1997.9900] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
S1 mapping analysis for the expression of Cyp3a11 and Cyp3a13 indicated that Cyp3a11 mRNA is predominantly expressed in mouse liver, compared with that of Cyp3a13. In addition, all of six inducers, such as dexamethasone, 3-methylcholanthrene, phenobarbital, polychlorinated biphenyl, pregnenolone 16 alpha-carbonitrile, and rifampicin, increased the expression of the Cyp3a11 mRNA more extensively than that of Cyp3a13. The level of mRNAs corresponding to Cyp3a11 and Cyp3a13 reached the maximum level between 4 and 8 weeks after birth. Cyp3a11 enzyme was expressed into CR119 cells which had been established as a cell line stably expressing NADPH-cytochrome P450 reductase cDNA of guinea pigs. These transformants showed aflatoxin B1-dependent cytotoxicity in proportion to the amounts of Cyp3a11 mRNA. This cytotoxicity was enhanced by 7,8-benzoflavone, a known activator of CYP3A protein. Based on these results, we confirm that CYP3A in the mouse, which is an animal species known to be relatively insensitive to aflatoxin B1 genotoxicity, can activate this mycotoxin efficiently.
Collapse
Affiliation(s)
- T Yanagimoto
- Division of Drug Metabolism, Faculty of Pharmaceutical Sciences, Hokkaido University, Japan
| | | | | | | |
Collapse
|
31
|
Hiruma S, Kimura M, Lehmann K, Gopalan-Kriczky P, Qin GZ, Shinozuka H, Sato K, Lotlikar PD. Potentiation of aflatoxin B1-induced hepatocarcinogenesis in the rat by pretreatment with buthionine sulfoximine. Cancer Lett 1997; 113:103-9. [PMID: 9065808 DOI: 10.1016/s0304-3835(97)04602-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A single i.p. dose of aflatoxin B1 (AFB1) (1.0 and 2.0 mg/kg body wt)-induced hepatocarcinogenesis with phenobarbital as a promoter has been examined in young male Fischer rats. Immunohistochemical method has been employed to detect AFB1-induced glutathione S-transferase placental form (GST-P)-positive hepatic foci observed from 3 week and 10 week to 40-48 week periods. With 2.0 mg AFB1 dosing, the number, area and volume occupied by GST-P-positive hepatic foci increased significantly and progressively from 3 week, 10 week and 48 week periods. In long term studies (40-48 weeks), 1.0 mg and 2.0 mg AFB1 dose levels yielded linear response in area and volume occupied by AFB1-induced hepatic foci. Pretreatment of rats with L-buthionine sulfoximine (BSO), a GSH depleter, at a dose of 4 mmol/kg body wt 4 and 2 h before 1.0 or 2.0 mg AFB1 treatment enhanced the number, area and volume of GST-P-positive hepatic foci, increases being the largest at shorter time periods (3 and 10 weeks) compared to longer time periods (40 and 48 weeks). This report appears to be the first example of an enhanced chemical induced hepatocarcinogenesis in a long term study in any experimental animals species by a GSH depleting agent.
Collapse
Affiliation(s)
- S Hiruma
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kono M, Kumagai S. In vitro microsome-mediated aflatoxin B1-DNA binding and its inhibition by cytosol of various organs of the hamster and quail. Mycopathologia 1995; 132:117-9. [PMID: 8819834 DOI: 10.1007/bf01103784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We studied the in vitro activation of aflatoxin B1 (B1) by microsomes and its inactivation by the cytosol of various quail and hamster organs, using B1-DNA binding as an index. The microsomal activity of the liver to bind B1 to DNA was not largely different between the two species and was higher than that of the other organs examined in either species. The microsomal activity of the kidney and lung was very low in the quail compared with the hamster, indicating the very small contribution of the lung and kidney microsomes to the activation of B1 in birds. Only the hamster liver cytosol showed strong inhibition of microsome-mediated B1-DNA binding.
Collapse
Affiliation(s)
- M Kono
- Department of Biomedical Food Research, National Institute of Health, Shinjuku-ku, Tokyo, Japan
| | | |
Collapse
|
33
|
Chelcheleh M, Allameh A. In vivo biotransformation of aflatoxin B1 and its interaction with cellular macromolecules in neonatal rats. Mech Ageing Dev 1995; 78:189-96. [PMID: 7596201 DOI: 10.1016/0047-6374(94)01536-u] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In this study, the ability of neonatal rat liver to metabolise [3H]aflatoxin B1 (AFB1) was compared to that of the adult animal. In order to make this comparison, neonatal and young adult rats were killed 2, 6, 12 and 24 h after injection with a single i.p. dose of AFB1. The rate of AFB1 adduct formation to nuclear DNA and protein was measured in hepatic and pulmonary tissues. The results demonstrated that AFB1 was epoxidized more rapidly by the adult's liver and lungs 2 h after the toxin administration, compared with those of the neonatal's (adult 30 pmol and neonatal 12 pmol AFB1 bound/mg DNA). However, these differences were more pronounced in hepatic than in pulmonary tissues. The same differences between AFB1-DNA adducts were also observed at different time points. These changes are certainly related to the level of hepatic cytochrome P-450. The delayed cytochrome P-450-dependent AFB1 activation in neonatal's liver provides time enough for de-epoxidation of slowly generated epoxide. The rate of AFB1-epoxide formation at this age was consistent with the activity of phase II metabolism of AFB1 (glutathione conjugation). In addition, the hydrolysis of AFB1-DNA adducts at a relatively higher rate by neonatal's liver may also contribute to the quick removal of the adducts. In spite of the aforementioned evidence which shows the capability of neonatal liver to handle AFB1, the fate of large amounts of free (non-metabolised) AFB1 deposited in neonatal's liver is not well understood.
Collapse
Affiliation(s)
- M Chelcheleh
- Department of Biochemistry, School of Medical Sciences, Tarbiat Modaress University, Tehran, Iran
| | | |
Collapse
|
34
|
Gopalan-Kriczky P, Jensen DE, Lotlikar D. Conjugation of microsome generated and synthetic aflatoxin B1-8,9 epoxide and styrene oxide to glutathione by purified glutathione S-transferases from hamster and mouse livers. Cancer Lett 1994; 86:83-90. [PMID: 7954359 DOI: 10.1016/0304-3835(94)90183-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glutathione (GSH) conjugation of microsome-mediated and synthetic aflatoxin B1 (AFB1)-epoxide and styrene oxide has been studied with purified glutathione transferases (GSTs) from mouse and hamster liver cytosols. In hamster, with microsomally activated epoxide, the alpha group of GSTs show about 10-fold more activity than the mu group. With the synthetic AFB1 epoxide, the mu enzymes designated H3B and C show considerable activity although less than alpha, whereas H3A and D demonstrate similar ranges of activity as the alpha group. The pi class of GST could not be assayed due to its absence in the hamster liver. The mouse liver cytosols show 3.6-fold greater activity than hamster cytosol in microsome mediated assay system. The mouse alpha and mu enzymes have similar levels of activity in the microsome mediated system; this activity could not be determined with the pi GST due to shortage of this enzyme. The alpha group has 2- and 5-fold higher activity than mu and pi group of GSTs, respectively, with the synthetic epoxide of AFB1. With styrene oxide, the purified GSTs from hamster liver show total loss of activity whereas in the mouse alpha, mu and pi classes of GSTs have similar range of activity as the cytosol. The role of alpha and mu isozymes of GST in rendering these animals resistant to hepatocarcinogenecity is suggested.
Collapse
Affiliation(s)
- P Gopalan-Kriczky
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140
| | | | | |
Collapse
|
35
|
McLellan LI, Judah DJ, Neal GE, Hayes JD. Regulation of aflatoxin B1-metabolizing aldehyde reductase and glutathione S-transferase by chemoprotectors. Biochem J 1994; 300 ( Pt 1):117-24. [PMID: 8198522 PMCID: PMC1138132 DOI: 10.1042/bj3000117] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Ingestion of aflatoxin B1 (AFB1) represents a major risk factor in the aetiology of human hepatocellular carcinoma. In the rat, the harmful effects of AFB1 can be prevented by the administration of certain drugs which induce hepatic detoxification enzymes. We have previously shown that treatment of rats with the chemoprotector ethoxyquin (EQ) results in a marked increase in expression of the Alpha-class glutathione S-transferase (GST) Yc2 subunit which has high activity towards AFB1-8,9-epoxide [Hayes, Judah, McLellan, Kerr, Peacock and Neal (1991) Biochem. J. 279, 385-398]. To allow an assessment of whether the increased expression of GST Yc2 represents a general adaptive resistance mechanism to chemical stress, that is invoked by both chemoprotectors and carcinogens, we have examined the effects of EQ, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), phenobarbital (PB), AFB1, 3-methylcholanthrene (3-MC) and clofibrate on the AFB1-glutathione-conjugating activity and the GST subunit levels in rat liver. In addition, the effect of these drugs on the hepatic levels of an aldehyde reductase (AFB1-AR) that metabolizes the cytotoxic dialdehydic form of AFB1 has been studied as this enzyme also appears to be important in chemoprotection. Administration of the antioxidants EQ, BHA or BHT, as well as PB, led to a marked increase in levels of the GST Yc2 subunit in rat liver, and this increase coincided with a substantial rise in the GST activity towards AFB1-8,9-epoxide; neither AFB1, 3-MC nor clofibrate caused induction of Yc2 or any of the GST subunits examined. Among the xenobiotics studied, EQ was found to be the most effective inducing agent for the Yc2 subunit as well as Yc1, Yb1 and Yf. However, PB was equally as effective as EQ in increasing levels of the Ya-type subunits, although it was not found to be as potent an inducer of the other GST subunits, including Yc2. In addition to induction of GST, EQ caused a substantial increase in the hepatic content of AFB1-AR. Both BHA and BHT were also able to induce this enzyme but, by contrast, PB was found to be a poor inducer of AFB1-AR. AFB1, 3-MC and clofibrate were unable to serve as inducers of this reductase. The presence of Alpha-class GST, including the Yc2 subunit, was examined in various rat tissues. Constitutive expression of Yc2 was found in the epididymis at levels comparable with that observed in the liver from EQ-treated rats.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- L I McLellan
- Department of Clinical Biochemistry, University of Edinburgh, Royal Infirmary, U.K
| | | | | | | |
Collapse
|
36
|
Müller K, Kasper P, Müller L. Species differences in the time-dependent induction of unscheduled DNA synthesis in primary hepatocytes. Toxicol In Vitro 1994; 8:99-108. [DOI: 10.1016/0887-2333(94)90213-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/1992] [Revised: 03/16/1993] [Indexed: 10/27/2022]
|
37
|
Gopalan-Kriczky P, Hiruma S, Lotlikar PD. Effect of glutathione levels on aflatoxin B1-DNA binding in livers and kidneys of male rats and hamsters pretreated with buthionine sulfoximine and dimethylmaleate. Cancer Lett 1994; 76:25-30. [PMID: 8124663 DOI: 10.1016/0304-3835(94)90130-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The effects of pretreatment of buthionine sulfoximine (BSO) alone or in combination with diethylmaleate (DEM) on glutathione (GSH) levels and aflatoxin B1 (AFB1)-DNA binding have been examined in livers and kidneys of young male Fischer rats and Syrian golden hamsters 2 h after an intraperitoneal injection of [3H]AFB1 (0.4 mg/kg body wt.). Animals were treated with BSO (4 mmol/kg body wt.) alone at 4 h and 2 h or with DEM (3 mmol/kg body wt.) at 4 h and BSO at 2 h before AFB1 injection. Hepatic AFB1-DNA binding was about 29.0 and 6.0 pmol/mg DNA in rats and hamsters, respectively. In rats, BSO increased AFB1-DNA binding by about 40% with a drop in GSH by 70%. Treatment with DEM-BSO increased AFB1-DNA binding by about 80% with a concomitant drop in GSH in both species. In hamsters, BSO increased AFB1-DNA binding by only 10% with a 50% drop in GSH. The kidneys of both species have lower GSH levels and AFB1-DNA binding than their respective liver tissues. The effect of BSO alone or in combination with DEM on both GSH levels and AFB1-DNA binding are comparable even though BSO alone is less effective in both species. The role of modulation of GSH levels on AFB1-DNA binding and hence biological effects of AFB1 in these two species is discussed.
Collapse
Affiliation(s)
- P Gopalan-Kriczky
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140
| | | | | |
Collapse
|
38
|
Cassand P, Decoudu S, Lévêque F, Daubèze M, Narbonne JF. Effect of vitamin E dietary intake on in vitro activation of aflatoxin B1. Mutat Res 1993; 319:309-16. [PMID: 7504205 DOI: 10.1016/0165-1218(93)90020-e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The molecular mechanism of action of vitamin E on mammalian cells remains to be elucidated. In this study, vitamin E dietary intake was assessed for its effects on the initiation phase of carcinogenesis. We have conducted a dose-effect relationship between vitamin E dietary intake and aflatoxin B1 (AFB1) genotoxicity measured in vitro. Thus AFB1 induced mutagenesis in Salmonella typhimurium TA98 was investigated and compared to effect of vitamin E dietary intake on hepatic microsomal P-450 content and specific activities involved in AFB1 metabolism. Rats were fed ad libitum a diet containing 0, 0.05, 0.5 or 5 IU of alpha-tocopherol for 8 weeks. Modulation of vitamin E level in postmitochondrial and microsomal fractions resulted in nutritional effects. Cytochrome P-450 content was not modified by the level of vitamin E in the diet. The microsomal P-450 activities, P-450 IIB1 and IIIA, were decreased in the deficient group to -35% and -16%, respectively, as compared with control diet (0.05 IU). Diet supplemented with 0.5 IU of vitamin E increased P-450 IIB and IIIA activities (+28% and +37%, respectively) whereas a diet highly supplemented in vitamin E (5 IU) reduced these specific P-450 activities. Lipid peroxidation, estimated by the formation of thiobarbituric acid reactive products, increased in the dietary vitamin E free diet (+20%) and strongly decreased in the supplemented group (-99%). This study establishes that in vivo, dietary vitamin E protects directly membrane against damage induced by lipid peroxidation and indirectly hepatic microsomal monooxygenase activities. However, vitamin E accumulation seems to alter membrane structure and function. The nutritional effect of vitamin E on hepatic microsomal cytochrome P-450 activities modified the AFB1 genotoxicity measured in vitro.
Collapse
Affiliation(s)
- P Cassand
- Laboratoire de Toxicologie Alimentaire, Université Bordeaux I, Talence, France
| | | | | | | | | |
Collapse
|
39
|
Biswas G, Raj HG, Allameh A, Saxena M, Srivastava N, Mukerji KG. Comparative kinetic studies on aflatoxin B1 binding to pulmonary and hepatic DNA of rat and hamster receiving the carcinogen intratracheally. TERATOGENESIS, CARCINOGENESIS, AND MUTAGENESIS 1993; 13:259-68. [PMID: 7903827 DOI: 10.1002/tcm.1770130603] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Several epidemiological studies have discussed the outcome of inhalation of airborne aflatoxins by humans. Metabolism of aflatoxin B1 (AFB1) by lung parenchyma leading to DNA binding is reported here. The tissue distribution pattern of [3H]AFB1 radioactivity revealed the lungs to be the second most important organ after the liver to retain a considerable amount of the radioactivity (66%). The lung indicated a selective activation of AFB1 as it showed only 7.7% binding of [3H]AFB1 to pulmonary DNA. Rats and hamsters were dosed with [3H]AFB1 (2 microCi containing 40 micrograms AFB1/100 g body wt.) intratracheally (i.t.) and sacrificed at different intervals after toxin treatment. Peak binding occurred at 0.5, 1, and 2 h in case of hamster lung, rat lung, and alveolar macrophages of both the species, respectively. At the end of 24 h, the relative AFB1-DNA binding (percentage of peak binding) in hamster lung was 72% while that in rat was 24%. The relative binding in rat lung alveolar macrophages (AMs) was generally higher than that of the hamster. AFB1 binding to hepatic DNA of both the species approached the peak at 1 h after the toxin administration i.t. Under these conditions, binding of AFB1 (or its metabolites translocated to liver) to hepatic DNA of both the species progressively diminished with time in contrast to lung, as revealed by the relative binding values at 12 h for rat and hamster lung, which were 48 and 67%, respectively, while for the rat and hamster liver they were 28 and 24%, respectively. Binding of i.t. administered [3H]AFB1 to rat liver DNA is only marginally higher than that observed with hamster liver, in contrast to the wide difference observed in animals receiving AFB1 intraperitoneally. These results highlight the persistence of AFB1 binding to pulmonary DNA, and the extent of translocated AFB1 binding to hepatic DNA presents an interesting difference from that observed when the toxin was administered through a gastrointestinal route. It is worth concluding that AMs unlike many other xenobiotics, possess specific mixed function oxidase activity to epoxidize AFB1.
Collapse
Affiliation(s)
- G Biswas
- Biochemistry Department, V. Patel Chest Institute, University of Delhi, India
| | | | | | | | | | | |
Collapse
|
40
|
Decoudu S, Cassand P, Daubèze M, Frayssinet C, Narbonne JF. Effect of vitamin A dietary intake on in vitro and in vivo activation of aflatoxin B1. Mutat Res 1992; 269:269-78. [PMID: 1383711 DOI: 10.1016/0027-5107(92)90209-k] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The mechanism by which vitamin A prevents or delays in chemical carcinogenesis remains unclear. In the present study, we assess the suggestive role of vitamin A in the initiation phase of carcinogenesis. We have conducted a dose-effect relationship between vitamin A dietary intake and aflatoxin B1 (AFB1) genotoxicity measured both in vitro and in vivo. Thus AFB1-induced mutagenesis in Salmonella typhimurium TA98 was investigated and compared to AFB1-induced single-strand breaks (SSBs) in DNA of rat hepatocytes. Rats were fed ad libitum with diet containing 0, 5, 50 or 500 IU of retinyl palmitate for 8 weeks. The AFB1-treated rats were injected i.p. with 1 mg/kg body weight. In the Ames test conditions TA98 back-reversion was negatively correlated with the log of vitamin A concentration in liver S9 fractions from experimental groups. However, the activities of metabolizing enzymes which specifically activate or deactivate AFB1 were found to be significantly decreased in vitamin A-deficient animals and weakly modified in vitamin A-supplemented animals. For in vivo experiments, the DNA elution rate of both AFB1-treated and untreated rats was increased in vitamin A deficiency condition (+79% and +17% respectively) and was reduced with the higher vitamin A dietary level (-44% and -53% respectively). DNA damage measured in vivo showed a significant positive correlation with mutagenic activity measured in the Ames test. These results confirm that the vitamin A status of animals can influence AFB1 genotoxic activity in vitro and indicate that this phenomenon also occurs in vivo. Thus a similar mechanism may be considered for the protective action of vitamin A both in vitro and in vivo. However, this mechanism is unlikely to involve modulation of the microsomal enzyme system responsible for AFB1 metabolism. Therefore a protective mechanism at the cytosolic or nuclear levels may be suggested.
Collapse
Affiliation(s)
- S Decoudu
- Laboratoire de Toxicologie Alimentaire, Université de Bordeaux I, Réseau de Toxicologie Nutritionnelle MRES, Talence, France
| | | | | | | | | |
Collapse
|
41
|
Behroozikha M, Saidee M, Allameh A. Comparison of aflatoxin B1-DNA binding and glutathione conjugate formation by liver preparations from rats of different ages. Cancer Lett 1992; 66:69-76. [PMID: 1451098 DOI: 10.1016/0304-3835(92)90282-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The capability of the newborn rat liver to detoxify aflatoxin B1 (AFB1), a potent hepatocarcinogen is not well understood. Our present results show that immature rats are deficient in the hepatic key factors involved in biotransformation of AFB1. The activities of cytosolic glutathione S-transferases and microsomal cytochrome P-450 along with cellular glutathione (GSH) content show postnatal developmental changes. The ability of hepatic subcellular preparation from newborn rats to convert AFB1 to its reactive epoxide form, is reported for the first time in this communication. Epoxidation of [3H]AFB1 in the presence of liver microsomes from different age-groups as measured by its adduct formation to calf thymus DNA in vitro shows that newborn rats are capable of catalyzing only minimal AFB1-DNA binding compared with that of adults. Addition of cytosolic fraction of various age groups to the system suggests that young rats are less efficient in modulating the binding as compared with adults. The amount of AFB1-GSH conjugate formed is also significantly higher when adult GSH S-transferase is involved in the system. These observations show that immature liver is less efficient than a mature organ in handling a chemical carcinogen and the metabolism of AFB1 by neonatal liver differs from that in the adult.
Collapse
Affiliation(s)
- M Behroozikha
- Department of Biochemistry, Faculty of Medical Sciences, Tarbiat Modaress University, Tehran, Iran
| | | | | |
Collapse
|
42
|
Gopalan P, Jensen DE, Lotlikar PD. Glutathione conjugation of microsome-mediated and synthetic aflatoxin B1-8,9-oxide by purified glutathione S-transferases from rats. Cancer Lett 1992; 64:225-33. [PMID: 1638515 DOI: 10.1016/0304-3835(92)90047-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glutathione (GSH) conjugation of microsome-mediated and synthetic aflatoxin B1 (AFB1)-epoxide and styrene oxide has been investigated with purified GSH S-transferases (GSTs) from rats. Both styrene oxide and AFB1-epoxide were conjugated preferentially by millimicrons GSTs 3-3, 3-4 and 4-4 as compared to alpha GSTs 1-1, 1-2 and 2-2. The highest catalytic activity with styrene oxide conjugation was associated with GST 4-4. The highest catalytic activity with microsome-mediated AFB1-epoxide conjugation was observed with GST 3-3 whereas with the synthetic AFB1-epoxide conjugation was seen with GST 4-4. The catalytic activity of pi GST 7-7 was intermediate to millimicrons and alpha GSTs. It is suggested that GST 3-3 may play an important role in inactivation of AFB1-epoxide generated in vivo in the rat.
Collapse
Affiliation(s)
- P Gopalan
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA
| | | | | |
Collapse
|
43
|
Hayes JD, Judah DJ, Neal GE, Nguyen T. Molecular cloning and heterologous expression of a cDNA encoding a mouse glutathione S-transferase Yc subunit possessing high catalytic activity for aflatoxin B1-8,9-epoxide. Biochem J 1992; 285 ( Pt 1):173-80. [PMID: 1637297 PMCID: PMC1132762 DOI: 10.1042/bj2850173] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Resistance to the carcinogenic effects of aflatoxin B1 (AFB1) in the mouse is due to the constitutive expression of an Alpha-class glutathione S-transferase (GST), YcYc, with high detoxification activity towards AFB1-8,9-epoxide. A cDNA clone (pmusGST Yc) for a murine GST Yc polypeptide has been isolated. Sequencing has shown the cDNA insert of pmusGST Yc to be 922 bp in length, with an open reading frame of 663 bp that encodes a polypeptide of M(r) 25358. The primary structure of the murine GST Yc subunit predicted by pmusGST Yc is in complete agreement with the partial amino acid sequence of the aflatoxin-metabolizing mouse liver GST described previously [McLellan, Kerr, Cronshaw & Hayes (1991) Biochem. J. 276, 461-469]. A plasmid, termed pKK-musGST Yc, which permits the expression of the murine Yc subunit in Escherichia coli, has been constructed. The murine GST expressed in E. coli was purified and found to be catalytically active towards several GST substrates, including AFB1-8,9-epoxide. This enzyme was also found to possess electrophoretic and immunochemical properties closely similar to those of the GST Yc subunit from mouse liver. However, the GST synthesized in E. coli and the constitutive mouse liver Alpha-class GST exhibited small differences in their chromatographic behaviour during reverse-phase h.p.l.c. Automated Edman degradation revealed alanine to be the N-terminal amino acid in the GST Yc subunit expressed in E. coli, whereas the enzyme in mouse liver possesses a blocked N-terminus. Although sequencing showed that the purified Yc subunit from E. coli lacked the initiator methionine, the amino acid sequence obtained over the first eleven N-terminal residues agreed with that predicted from the cDNA clone, pmusGST Yc. Comparison of the deduced amino acid sequence of the mouse Yc polypeptide with the primary structures of the rat Alpha-class GST enzymes revealed that it is more closely related to the ethoxyquin-induced rat liver Yc2 subunit than to the constitutively expressed rat liver Yc1 subunit. The significance of the fact that both mouse Yc and rat Yc2 exhibit high catalytic activity towards AFB1-8,9-epoxide, whereas rat Yc1 possesses little activity towards this compound, is discussed in terms of structure/function.
Collapse
Affiliation(s)
- J D Hayes
- Department of Molecular Biology, Merck Frosst Center for Therapeutic Research, Merck Frosst Canada Inc, Pointe Claire-Dorval, Quebec, Canada
| | | | | | | |
Collapse
|
44
|
Borroz KI, Ramsdell HS, Eaton DL. Mouse strain differences in glutathione S-transferase activity and aflatoxin B1 biotransformation. Toxicol Lett 1991; 58:97-105. [PMID: 1910216 DOI: 10.1016/0378-4274(91)90195-c] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Previous studies have suggested that mice are resistant to the carcinogenic effects of aflatoxin B1 (AFB1) and that this resistance is largely the result of expression of an isoenzyme of glutathione S-transferase (GST) with high activity toward AFB1-8,9-epoxide. Significant interstrain differences in cytosolic GST activities toward a variety of substrates have been reported in mice. If such differences exist for the conjugation of AFB1-8,9-epoxide, then there may be significant mouse strain differences in susceptibility to AFB1-induced hepatocarcinogenicity. The hepatic microsomal and cytosolic biotransformation of AFB1 was studied in 8 different strains of mice fed a purified diet. GST-mediated conjugation of AFB1-8,9-epoxide with glutathione and GST activity toward 1-chloro-2,4-dinitrobenzene (CDNB), 1,2-dichloro-4-nitrobenzene (DCNB), ethacrynic acid (ECA) and cumene hydroperoxide (CHP) were determined with cytosolic fractions from 8-10 pooled livers. Specific activities of cytochrome-P-450-mediated oxidation of AFB1 to aflatoxin Q1 (AFQ1), aflatoxin M1 (AFM1), and aflatoxin P1 (AFP1), as well as the reactive intermediate AFB1-8,9-epoxide, were determined with hepatic microsomal fractions from each mouse strain. No striking differences in specific activity between mouse strains were observed for any of the P-450- or GST-mediated enzymatic pathways measured, although some statistically significant differences were found. GST specific activities toward AFB1-8,9-epoxide, CDNB, DCNB, ECA and CHP ranged from 1.5-2.1, 2,830-5,370, 81-144, 38-69 and 32-73 nmol/mg protein/min, respectively. The rate of formation of AFB1-8,9-epoxide ranged from 208 to 465 pmol/mg protein/min. The specific activities of AFQ1,AFM1, and AFP1 formation by microsomes ranged from 36-70, 161-326, and 252-426 pmol/mg protein/min, respectively. Mice fed a standard rodent chow diet showed evidence of microsomal and cytosolic enzyme induction when compared to mice fed a purified diet. The lack of substantial differences in enzyme specific activities between mouse strains suggests that interstrain variations in the hepatocarcinogenic effects of AFB1 in mice should not be large.
Collapse
Affiliation(s)
- K I Borroz
- Department of Environmental Health, University of Washington, Seattle 98195
| | | | | |
Collapse
|
45
|
Hayes JD, Judah DJ, McLellan LI, Neal GE. Contribution of the glutathione S-transferases to the mechanisms of resistance to aflatoxin B1. Pharmacol Ther 1991; 50:443-72. [PMID: 1754606 DOI: 10.1016/0163-7258(91)90053-o] [Citation(s) in RCA: 89] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The harmful effects of Aflatoxin B1 (AFB1) are a consequence of it being metabolized to AFB1-8,9-epoxide, a compound that serves as an alkylating agent and mutagen. The toxicity of AFB1 towards different cells varies substantially; sensitivity can change significantly during development, can be modulated by treatment with xenobiotics and is decreased markedly in preneoplastic lesions as well as in tumors. Three types of resistance, namely intrinsic, inducible and acquired, can be identified. The potential resistance mechanisms include low capacity to form AFB1-8,9-epoxide, high detoxification activity, increase in AFB1 efflux from cells and high DNA repair capacity. Circumstantial evidence exists that amongst these mechanisms the glutathione S-transferases, through their ability to detoxify AFB1-8,9-epoxide, play a major role in determining the sensitivity of cells to AFB1.
Collapse
Affiliation(s)
- J D Hayes
- University Department of Clinical Chemistry, Royal Infirmary, Edinburgh, Scotland, U.K
| | | | | | | |
Collapse
|
46
|
d'Arville CN, Johnson PJ. Growth factors, endocrine aspects and hormonal treatment in hepatocellular carcinoma--an overview. J Steroid Biochem Mol Biol 1990; 37:1007-12. [PMID: 2178361 DOI: 10.1016/0960-0760(90)90458-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Several clinical observations suggest that hepatocellular carcinoma (HCC or "hepatoma") may be a hormone-dependent tumour; the apparent relation to anabolic steroids and oral contraceptive preparations, and the striking male predominance particularly among patients with cirrhosis. In many animal models thyroid hormones, prolactin and testosterone stimulate tumour growth, and the latter may enhance the progression of chemically-induced hyperplastic nodules to frank malignancy. In animals and humans, both oestrogen and androgen receptors have been reported in normal and malignant liver tissue though some of the evidence is conflicting and the amounts detected vary widely. From a therapeutic standpoint, we failed to show any advantage from the addition of tamoxifen to adriamycin, in a controlled trial although other workers have, more recently, reported prolonged survival using tamoxifen alone. About 20% of HCC patients receiving the antiandrogen cyproterone acetate showed a clinical response.
Collapse
Affiliation(s)
- C N d'Arville
- Liver Unit, King's College Hospital, London, England
| | | |
Collapse
|
47
|
|
48
|
Lampe MA, Kammerer RC. Species differences in the metabolism and macromolecular binding of methapyrilene: a comparison of rat, mouse and hamster. Xenobiotica 1990; 20:1269-80. [PMID: 2075747 DOI: 10.3109/00498259009046626] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
1. The metabolism of methapyrilene (MPH) by rat, hamster and mouse liver microsomes in vitro was investigated together with the binding of 14C-MPH to calf thymus DNA after metabolic activation. 2. Both quantitative and qualitative differences in MPH metabolism were observed in these three species. Mouse liver microsomes catalyse the formation of two novel isomers of hydroxypyrdylmethapyrilene (hydroxypyridyl-MPH) as determined by mass spectral analysis. N,N'-Didesmethylmethapyrilene (didesmethyl-MPH) was formed in detectable quantities only when hamster liver microsomes were used. 3. Incubation of liver microsomes from all three species catalysed the binding of 14C-MPH to exogenous DNA, which was quantitatively similar for all three species. The effect of the cytochrome P-450 inhibitor, 2,4-dichloro-6-phenylphenoxyethylamine (DPEA), and methimazole, a flavin-dependent monooxygenase inhibitor, on binding differed significantly for the three species studied.
Collapse
Affiliation(s)
- M A Lampe
- Department of Pharmacology, School of Medicine, University of California, Los Angeles 90024
| | | |
Collapse
|
49
|
Teel RW, Strother A. Glucose alters rat liver S9-mediated mutagenesis, metabolism and DNA-binding of aflatoxin B1. Cancer Lett 1990; 54:163-9. [PMID: 2121329 DOI: 10.1016/0304-3835(90)90039-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The administration of 30% glucose in drinking water to rats for 48 h caused a significant increase in the hepatic S9-mediated mutagenicity of aflatoxin B1, in Salmonella typhimurium TA100 and in the binding of alfatoxin B1, to calf thymus DNA in vitro. These effects correlated with a reduction in the metabolism and detoxification of aflatoxin B1, by S9 from glucose-treated rats and suggest that the oral intake of sugar may affect the hepatocarcinogenicity of aflatoxin B1.
Collapse
Affiliation(s)
- R W Teel
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA 92350
| | | |
Collapse
|
50
|
Forrester LM, Neal GE, Judah DJ, Glancey MJ, Wolf CR. Evidence for involvement of multiple forms of cytochrome P-450 in aflatoxin B1 metabolism in human liver. Proc Natl Acad Sci U S A 1990; 87:8306-10. [PMID: 2122459 PMCID: PMC54944 DOI: 10.1073/pnas.87.21.8306] [Citation(s) in RCA: 111] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Liver cancer is a major cause of premature death in many areas of Africa and Asia and its incidence is strongly correlated with exposure to aflatoxin B1 (AFB1). Because AFB1 requires metabolic activation to achieve a biological response, there is a need for detailed knowledge of the mechanism of activation to assess individual risk. We have carried out an extensive study using a total of 19 human liver samples to determine the individual variability in the metabolism of the toxin to mutagenic or detoxification products and to identify the specific cytochrome P-450 forms involved in these processes. Metabolism to the toxic 8,9-epoxide or to products mutagenic in the Ames test was found to exhibit very large individual variation. The rates of metabolic activation were highly correlated with both the level of proteins of the P450IIIA gene family and with the total cytochrome P-450 content of the microsomes. In agreement with this, antibodies reacting with P450IIIA proteins were strong inhibitors of both the metabolism and mutagenicity in the majority of the samples. However, the inhibition varied between 50% and 100%. The expression of a protein in the P450IIC gene family also correlated with AFB1 metabolism and mutagenicity. This result therefore indicated the involvement of cytochromes other than P450IIIA in the activation of AFB1 by human liver microsomes. This hypothesis was strongly supported by the finding that antibodies to P450IA2 and P450IIA1 were also effective inhibitors of metabolism in many of the samples. These data demonstrate that, although P450IIIA probably plays an important role in AFB1 activation, several other cytochrome P-450 forms have the capacity to activate the toxin. Similar considerations apply to detoxifying metabolism to aflatoxin Q1 and aflatoxin M1. The levels of expression of many of the forms of cytochrome P-450 involved in AFB1 metabolism are known to be highly sensitive to environmental factors. This indicates that such factors will be an important determinant in individual susceptibility to the tumorigenic action of AFB1.
Collapse
Affiliation(s)
- L M Forrester
- Imperial Cancer Research Fund, University Department of Biochemistry, Edinburgh, United Kingdom
| | | | | | | | | |
Collapse
|