Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters










Publication year range
1.
Cancer Epidemiol Biomarkers Prev ; 10(3): 191-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11303587

ABSTRACT

Alpha-difluoromethylornithine (DFMO) is an irreversible inhibitor of ornithine decarboxylase, the first enzyme in polyamine synthesis. Previous work showed simultaneous administration of DFMO and a zinc-deficient (ZD) diet to weanling rats from the beginning inhibited the onset of zinc-deficiency-induced esophageal cell proliferation by activating apoptosis and reduced the incidence of N-nitrosomethylbenzylamine (NMBA)-induced esophageal cancer. Because esophageal cancer initiation by NMBA is very rapid in ZD rats, this study determined whether DFMO is effective in preventing esophageal carcinogenesis when administered after the establishment of a carcinogenic environment. Weanling rats were given a ZD diet for 5 weeks to establish sustained increased esophageal cell proliferation and then an intragastric dose of NMBA. Thereafter, 20 rats were switched to DFMO-containing water while nine control ZD animals remained on deionized water; all of the animals continued on the ZD diet. Esophagi were collected 15 weeks later. The upper portion was processed for immunohistochemical analysis of cell proliferation, apoptosis, and expression of related genes, and the lower was processed for polyamine content. DFMO substantially reduces the levels of esophageal putrescine and spermidine and esophageal tumor incidence from 89 to 10% in ZD rats. Importantly, DFMO-treated ZD esophagi display increased rate of apoptosis accompanied by intense bax expression and greatly reduced cell proliferation by proliferating cell nuclear antigen expression. In addition, the p16(ink4a)/retinoblastoma control at G1 to S, deregulated in ZD esophagi, is restored after DFMO treatment. These results demonstrate that DFMO, a highly effective chemopreventive agent in esophageal carcinogenesis, reverses and counteracts esophageal cell proliferation/cancer initiation in ZD animals by way of stimulating apoptosis.


Subject(s)
Apoptosis/drug effects , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/prevention & control , Proto-Oncogene Proteins c-bcl-2 , Analysis of Variance , Animals , Blotting, Western , Cell Division/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/analysis , Diet , Dimethylnitrosamine/analogs & derivatives , Disease Models, Animal , Drug Interactions , Esophageal Neoplasms/chemically induced , Genes, bcl-2 , Immunohistochemistry , Male , Polyamines/analysis , Proliferating Cell Nuclear Antigen , Proto-Oncogene Proteins/analysis , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Zinc/deficiency , bcl-2-Associated X Protein
2.
Cancer Res ; 60(16): 4589-95, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969811

ABSTRACT

The p16ink4a-cyclin D1/cyclin-dependent kinase 4 (Cdk4)-retinoblastoma (Rb) pathway has emerged as a critical target in oncogenesis. The zinc-deficient (ZD), N-nitrosomethylbenzylamine (NMBA)-induced rat esophageal cancer model provides a tool to study cell proliferation and cell cycle control in cancer initiation. Weanling rats were fed a ZD or zinc-sufficient (ZS) diet for 5 weeks, and then given a dose of NMBA. After 14 weeks, esophageal tumor incidence was 88% in ZD rats with highly proliferative esophagi versus 0% in ZS rats. Expression of p16ink4a, cyclin D1, Cdk4, and Rb in relation to that of proliferating cell nuclear antigen was characterized in esophagi by immunohistochemistry at 0, 24, and 48 h, and 1, 3, 7, 10, and 14 weeks after NMBA treatment. As early as 24 h, proliferating cell nuclear antigen-positive focal hyperplastic lesions were detected in the suprabasal layers of ZD esophagi. At the same time, overexpression of cyclin D1, Cdk4, and Rb was found in the corresponding lesion in adjacent esophageal sections. By contrast, p16ink4a expression was reduced or absent. At all time points, p16ink4a showed reduced nuclear staining in ZD esophagi compared with that in ZS esophagi. In addition, increased expression of the hyperphosphorylated forms of Rb was detected in ZD esophagi by immunoblotting. Importantly, tumors were consistently observed in ZD esophagi at very early time points. These data, obtained using a unique in vivo model for esophageal cancer with rapid tumor induction, provide strong evidence for a link between deregulation of the p16ink4a-cyclin D1/Cdk4-Rb pathway and the initiation of esophageal tumors.


Subject(s)
Carrier Proteins/biosynthesis , Cyclin D1/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Dimethylnitrosamine/analogs & derivatives , Esophageal Neoplasms/pathology , Proto-Oncogene Proteins , Retinoblastoma Protein/biosynthesis , Signal Transduction/physiology , Zinc/deficiency , Animals , Carcinogens , Cell Division/physiology , Cocarcinogenesis , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16 , Esophageal Neoplasms/etiology , Esophageal Neoplasms/metabolism , Esophagus/cytology , Esophagus/drug effects , Esophagus/metabolism , G1 Phase/drug effects , G1 Phase/physiology , Immunohistochemistry , Male , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , S Phase/drug effects , S Phase/physiology , Signal Transduction/drug effects
3.
Cancer Lett ; 143(1): 63-9, 1999 Aug 23.
Article in English | MEDLINE | ID: mdl-10465339

ABSTRACT

The effect of zinc deficiency on N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumor formation in rats has been well documented. Our previous work showed that zinc deficiency and its associated increased esophageal cell proliferation were of paramount importance in esophageal tumor development in the NMBA-rat model. However, there has been no report concerning zinc deficiency and NMBA-induced esophageal tumor formation in mice. In this study, weanling C57BL/6 mice were fed ad libitum with either a zinc-sufficient or a zinc-deficient diet containing 3-4 ppm of zinc, and received six intragastric doses of NMBA (2 mg/kg; twice weekly for 3 weeks). The animals were sacrificed 46 weeks later after in vivo bromodeoxyuridine (BrDU) labeling followed by immunohistochemical detection of cells in S-phase. At 46 weeks, the tumor incidences in zinc-deficient mice were 57, 100, and 100% respectively, in the esophagus, forestomach and squamocolumnar junction with the glandular stomach (SCJ), as compared to 17, 39, and 67% in the corresponding tissue of zinc-sufficient mice. The difference between the two dietary groups was significant at P < 0.02 for the esophagus, and P < 0.001 for the forestomach and the SCJ. BrDU labeling revealed that the esophageal labeling index and the number of labeled cells were increased by zinc deficiency. These results support a role of increased cell proliferation in esophageal carcinogenesis in the mouse.


Subject(s)
Carcinogens/toxicity , Dimethylnitrosamine/analogs & derivatives , Epithelial Cells/pathology , Esophageal Neoplasms/pathology , Esophagus/pathology , Zinc/deficiency , Animals , Body Weight/drug effects , Bromodeoxyuridine/metabolism , Cell Division , Dimethylnitrosamine/toxicity , Eating/drug effects , Esophageal Neoplasms/chemically induced , Male , Mice , Mice, Inbred C57BL , Zinc/blood
4.
Cancer Res ; 58(23): 5380-8, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850069

ABSTRACT

Sustained, increased cell proliferation induced by dietary zinc deficiency in rats plays a critical role in esophageal carcinogenesis. It is the determining factor that converts an otherwise nontumorigenic dose of N-nitrosomethylbenzylamine (NMBA) into a highly tumorigenic one. We studied whether the increased esophageal cell proliferation and susceptibility to NMBA-induced carcinogenesis induced by zinc deficiency can be inhibited by alpha-difluoromethylornithine (DFMO), an enzyme-activated, irreversible inhibitor of ornithine decarboxylase (the first enzyme in polyamine synthesis). Weanling rats were divided into four groups: Zn+/DFMO-, Zn+/DFMO+, Zn-/DFMO-, and Zn-/DFMO+. They were fed ad libitum either a zinc-sufficient (Zn+, 75 ppm zinc) or a zinc-deficient (Zn-, 4 ppm zinc) diet and given either deionized water (DFMO-) or 1% DFMO in deionized water (DFMO+). After 5 weeks, 5-19 animals from each group were sacrificed after in vivo 5-bromo-2'-deoxyuridine labeling to detect cells in S phase. The remaining animals in each group were given a single intragastric dose of NMBA at 2 mg/kg and sacrificed 12 weeks later for tumor incidence analysis. At week 5, DFMO treatment greatly decreased (by 48-82%) the levels of putrescine and spermidine in rat esophagus, colon, and liver, irrespective of dietary zinc intake. The increased esophageal cell proliferation induced by dietary zinc deficiency, as measured by the labeling index, the number of labeled cells, and the total number of cells, was substantially reduced by DFMO. This was accompanied by an increase in the rate of apoptosis. In addition, the expression of bax protein, an apoptosis accelerator, was markedly stronger in esophagi from Zn-/DFMO+ animals that showed increased apoptosis, whereas increased expression of bcl-2, an inhibitor of apoptosis, was only seen in the highly proliferative, zinc-deficient esophagus (Zn-/DFMO-). At week 12 after NMBA dosing, DFMO reduced the incidence of esophageal tumors from 80 to 4% in zinc-deficient rats. Our data showed that DFMO effectively inhibited the increased esophageal cell proliferation induced by dietary zinc deficiency and reduced the incidence of esophageal tumors induced by a single dose of NMBA in zinc-deficient animals. Our results also indicate a role for increased apoptosis in the mechanism(s) whereby DFMO brings about the inhibition of cell proliferation and tumor induction. These findings support a role for DFMO as a chemopreventive agent.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Carcinogens/toxicity , Cocarcinogenesis , Dimethylnitrosamine/analogs & derivatives , Eflornithine/therapeutic use , Esophageal Neoplasms/prevention & control , Esophagus/drug effects , Zinc/deficiency , Animals , Biogenic Polyamines/metabolism , Cell Division/drug effects , Diet , Dimethylnitrosamine/antagonists & inhibitors , Dimethylnitrosamine/toxicity , Esophageal Neoplasms/chemically induced , Esophageal Neoplasms/pathology , Esophagus/cytology , Esophagus/metabolism , Male , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein
5.
Carcinogenesis ; 19(9): 1591-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9771929

ABSTRACT

Previous work has shown that sustained increased and decreased cell proliferation, induced by dietary zinc deficiency and caloric restriction respectively, influence the course of N-nitrosomethylbenzylamine (NMBA)-induced esophageal carcinogenesis in rats. The present study considered whether the increased cell proliferation and esophageal tumor incidence induced by zinc deficiency are reversed upon zinc replenishment. Weanling rats were maintained initially on a deficient diet containing 4 p.p.m. zinc. After 5 weeks, carcinogen-treated animals were given six intragastric doses of NMBA (2 mg/kg twice weekly). Controls were untreated. After the second NMBA dose, the rats were divided into three dietary groups. One group was continued on the deficient diet, while the other two groups were switched to diets containing either 75 or 200 p.p.m. zinc, with half of the members in each group fed ad libitum and half pair-fed with deficient rats. NMBA-untreated controls were similarly replenished. At various time points, esophageal cell proliferation was assessed in five animals from each group by immunohistochemical detection of cells in S phase, with in vivo 5-bromo-2'deoxyuridine labeling. At 11 weeks after the first dose, esophageal tumor incidence was greatly reduced, from 100% in the deficient group to 26 and 14% respectively in the replenished groups fed ad libitum 75 and 200 p.p.m. zinc and to 14 and 11% respectively in the replenished groups pair-fed 75 and 200 p.p.m. zinc. In addition, the number of tumors per esophagus was reduced from 9.93 +/- 4.25 in deficient rats, to a range of 0.11 +/- 0.31-0.30 +/- 0.54 in replenished animals. Following zinc replenishment, esophageal cell proliferation, as measured by labeling index (LI), the number of labeled cells and the total number of cells, was markedly decreased in NMBA-untreated and -treated esophagi as compared with those in corresponding deficient esophagi. Thus, the esophageal cell proliferation induced by zinc deficiency is reversed by zinc replenishment and replenished animals have a markedly lower incidence of esophageal tumors.


Subject(s)
Carcinogens/toxicity , Dimethylnitrosamine/analogs & derivatives , Esophageal Neoplasms/prevention & control , Esophagus/drug effects , Zinc/pharmacology , Animals , Cell Division/drug effects , Dimethylnitrosamine/toxicity , Esophageal Neoplasms/etiology , Esophagus/pathology , Male , Rats , Rats, Sprague-Dawley , Zinc/deficiency
6.
Carcinogenesis ; 18(8): 1477-84, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9276619

ABSTRACT

Dietary zinc deficiency in rats induces hyperplasia in the esophagus and increases N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumor incidence. Previous work showed a direct relationship between epithelial cell proliferation and esophageal tumor incidence in rats given multiple doses of NMBA. We investigated the effects of single low doses of NMBA in zinc-deficient rats since a single dose of 5.0 mg/kg was reported to be non-carcinogenic in rats. Zinc-sufficient and deficient rats received a single i.g. dose of NMBA at 0.5 or 2.0 mg/kg. At week 14, tumor incidence was 50% with 0.8 +/- 1.0 tumors/rat, and 80% with 2.2 +/- 1.9 tumors/rat, in deficient groups, D(0.5) and D(2.0), that received the lower and higher dose, respectively. In addition, two small papillomas were found in one out of eight untreated zinc-deficient rats. None of the NMBA-treated or untreated zinc-sufficient rats had any tumors. Esophageal cell proliferation, as determined by proliferating cell nuclear antigen (PCNA) immunohistochemistry, showed that, irrespective of NMBA treatment, deficient esophagi had significant increases in the number of labeled cells, the total number of cells, and the labeling index, as compared with zinc-sufficient ones. Mutations in Ha-ras and p53 genes in esophageal tumors were detected by single strand conformation polymorphism (SSCP) analysis. DNA sequencing of variant conformers revealed a point mutation (GGA-->GAA, codon 12) in Ha-ras in 4/5 (80%) and 5/8 (63%) tumors, from D(0.5) and D(2.0) rats, respectively. Three out of eight tumors from D(2.0) rats exhibited SSCP mobility shifts within p53 exons 5 and 7: two tumors (2/8, 25%) had missense mutations and the third, a silent mutation. Of the two tumors with p53 mutations, one had a double mutation (transition at codon 164, TCA-->TTA; transversion at codon 241, AGT-->TGT), and the other tumor, a transition at codon 172 (AGA-->GGA), with amino acid changes in all cases. In parallel with PCNA expression, elevated p53 expression was associated with hyperplastic and dysplastic regions, as well as with tumors, in deficient esophagi. In short, these data indicate that dietary zinc deficiency, with its associated sustained increased cell proliferation in the esophagus, can drive an otherwise non-tumorigenic dose of NMBA into a highly tumorigenic one.


Subject(s)
Cocarcinogenesis , Esophageal Neoplasms/pathology , Genes, p53 , Genes, ras , Zinc/deficiency , Animals , Cell Division , Dimethylnitrosamine/analogs & derivatives , Dose-Response Relationship, Drug , Epithelium/chemistry , Esophageal Neoplasms/etiology , Esophageal Neoplasms/genetics , Immunohistochemistry , Male , Mutation , Polymorphism, Single-Stranded Conformational , Proliferating Cell Nuclear Antigen/analysis , Rats , Rats, Sprague-Dawley
7.
Carcinogenesis ; 17(9): 1841-8, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8824504

ABSTRACT

Target cell proliferation was investigated throughout the development of esophageal cancer induced by N-nitroso-methylbenzylamine (NMBA) in weanling rats maintained on zinc-deficient or sufficient diets. Deficient rats were fed ad libitum, while zinc-sufficient rats were either pair-fed to the deficient animals or fed ad libitum. After 5 weeks, half of the animals in each dietary group were given six intragastric doses of NMBA (2 mg/kg; twice weekly). The remaining rats were untreated by carcinogen. At weeks 1, 2, 3, 4, 5, 7, 9 and 11 post first dose, esophageal cell proliferation was assessed in rats from each group by in vivo bromodeoxyuridine (BrDU) labeling followed by immunohistochemical detection of cells in S-phase. At 11 weeks, the tumor incidence was 100, 23 and 6%, respectively, in the zinc-deficient, zinc-sufficient, ad libitum and pair-fed groups. In vivo BrDU labeling revealed that in the NMBA-untreated groups, the labeling index (LI), the number of labeled cells, and the total number of cells per cross section of entire esophagi were significantly increased by zinc deficiency at all time points; LI was lowest in zinc-sufficient, pair-fed rats. During NMBA treatment (weeks 6, 7 and 8), increased cell proliferation occurred in both groups of zinc-sufficient esophagi but only during week 6 in the deficient ones. In the weeks following the cessation of NMBA treatment, zinc-deficient esophagi showed significantly increased LI and greater number of labeled cells than the carcinogen treated, zinc-sufficient pair-fed or ad libitum fed groups. On the other hand, NMBA-treated zinc-sufficient pair-fed rats showed lower LI and smaller number of labeled cells than their zinc-sufficient ad libitum counterparts. Most importantly, esophageal papillomas were found in two zinc-deficient animals that had received no NMBA treatment, after 10-11 weeks of experimental diet. These data support a direct relationship between cell proliferation and tumor incidence, and also provide evidence that zinc deficiency and its associated cell proliferation could be carcinogenic.


Subject(s)
Carcinogens/toxicity , Dimethylnitrosamine/analogs & derivatives , Esophageal Neoplasms/pathology , Esophagus/pathology , Papilloma/pathology , Zinc/deficiency , Aging , Animals , Body Weight/drug effects , Bromodeoxyuridine , Cell Division/drug effects , Dimethylnitrosamine/toxicity , Esophageal Neoplasms/chemically induced , Esophageal Neoplasms/physiopathology , Esophagus/drug effects , Male , Mitotic Index/drug effects , Papilloma/chemically induced , Papilloma/physiopathology , Rats , Rats, Sprague-Dawley , Reference Values , S Phase , Time Factors , Weaning , Zinc/pharmacology
9.
Drug Metab Dispos ; 22(1): 43-9, 1994.
Article in English | MEDLINE | ID: mdl-8149888

ABSTRACT

Oral cotreatment of mice with ethanol results in increased tumors in extrahepatic organs caused by some nitrosamines. This action, attributed in part to inhibition of hepatic first-pass carcinogen metabolism by ethanol, has possible relevance to the enhancing effect of alcoholic beverage consumption on human cancer risk. In this study, the effects of ethanol on clearance of N-nitrosodimethylamine (NDMA) were quantified in Swiss female and strain A male mice. In Swiss mice, a 1.6 g/kg ig ethanol dose preceding 1 or 5 mg/kg iv NDMA resulted in 20- to 30-fold increases in area-under-the-blood-concentration-vs.-time curves, mean residence times, and clearance half-times, and similar decreases in clearance. For a 0.5 mg/kg ig NDMA dose, the pharmacokinetic parameters were altered 30-fold and 450-fold by simultaneous ethanol doses of 0.08 and 0.8 g/kg, respectively. With 5 mg NDMA/kg ig, 0.4, 0.8, and 1.6 g/kg ethanol resulted in 6-, 10-, and 20-fold changes in clearance parameters. Comparison of the data with results obtained previously with patas monkeys indicated comparable effects of ethanol on tissue exposure to NDMA in the two species, confirming potential human applicability. In experiments with strain A mice, NDMA concentrations were also monitored in lung and liver. NDMA amounts in lung paralleled those in blood, and were more than sufficient to account for the previously reported increases in DNA adducts and tumors in lungs of similarly treated strain A mice.


Subject(s)
Dimethylnitrosamine/pharmacokinetics , Ethanol/administration & dosage , Animals , Dimethylnitrosamine/blood , Ethanol/blood , Female , Liver/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred A
10.
Carcinogenesis ; 13(11): 2153-9, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1423888

ABSTRACT

Procarbazine (PCZ), an antineoplastic agent that produces methylated bases in DNA after metabolic activation, has been implicated in the development of secondary cancers in patients treated for a primary neoplasm. The repair of the important promutagenic lesion, O6-methylguanine (O6-meG) by O6-alkylguanine-DNA alkyl transferase (AGT) is believed to be crucial for the stability of O6-meG and for the tumorigenic outcome after exposure to methylating carcinogens. Using two different animal models, we investigated methyl DNA adduct dosimetry and DNA repair in (i) female rats given repeated doses of PCZ for 20 weeks under conditions of carcinogenicity, and (ii) female pigs administered repeated doses of PCZ for 4 weeks according to a regimen comparable to that given to human subjects undergoing cancer chemotherapy. After each successive week, four rats and three pigs were killed and tissues including blood, liver, mammary gland, spleen, thymus and lymph node were taken. The levels of O6-meG, 7-methylguanine (7-meG) in DNA and of AGT were determined in these tissues. In the rat, O6-meG in the liver DNA, and to a lesser degree in the spleen was efficiently removed throughout the 20 week dosing period, as indicated by the O6-meG/7-meG ratio being much less than 0.11. In the target organs, accumulation of O6-meG began in the mammary gland after 9 weeks, and in the lymph node and thymus after 3 weeks of dosing. Interestingly, the accumulation of O6-meG in the mammary gland correlates well with a concomitant decrease in AGT level as from week 10 and may be related to the induction of mammary gland tumors, first detected in two animals at week 10. In pigs, after a total dose of 750-4000 mg of PCZ, the range of 7-meG detected in leukocyte DNA was 21-66 mumol/mol G, which compares well with recent findings in cancer patients treated with PCZ. Similar levels of 7-meG were detected in the pig liver, thymus and lymph node. O6-MeG was only detectable in leukocyte DNA at week 4 with our present method. Compared with control pig tissues, a depressed AGT level was found in the leukocyte, lymph node and brain of the treated animals.


Subject(s)
DNA Damage , DNA Repair , Procarbazine/therapeutic use , Animals , Chromatography, High Pressure Liquid , DNA/chemistry , Female , Guanine/analogs & derivatives , Guanine/analysis , Methyltransferases/metabolism , O(6)-Methylguanine-DNA Methyltransferase , Procarbazine/administration & dosage , Procarbazine/toxicity , Rats , Rats, Sprague-Dawley , Species Specificity , Swine
11.
Cancer Res ; 52(6): 1463-8, 1992 Mar 15.
Article in English | MEDLINE | ID: mdl-1540953

ABSTRACT

Low concentrations of N-nitrosodimethylamine are metabolized in rodent and human liver by cytochrome P450IIE1, an activity competitively inhibitable by ethanol. In rodents coadministration of ethanol with N-nitrosodimethylamine results in increased tumorigenicity in extrahepatic organs, probably as a result of reduced hepatic clearance. To test this concept in a primate, the effects of ethanol cotreatment on the pharmacokinetics of N-nitrosodimethylamine were measured in male patas monkeys. Ethanol, 1.2 g/kg given p.o. before i.v. N-nitrosodimethylamine (1 mg/kg) or concurrently with an intragastric dose resulted in a 10-50-fold increase in the area under the blood concentration versus time curves and a 4-13-fold increase in mean residence times for N-nitrosodimethylamine. Isopropyl alcohol, 3.2 g/kg 24 h before N-nitrosodimethylamine, also increased these parameters 7-10-fold; this effect was associated with persistence of isopropyl alcohol and its metabolic product acetone, both IIE1 inhibitors, in the blood. While no N-nitrosodimethylamine was detected in expired air, trace amounts were found in urine. Ethanol and isopropyl alcohol pretreatment increased the maximum urinary N-nitrosodimethylamine concentration 15-50-fold and the percentage of the dose excreted in the urine by 100-800-fold. Thus ethanol and isopropyl alcohol greatly increase systemic exposure of extrahepatic organs to N-nitrosodimethylamine in a primate.


Subject(s)
1-Propanol/pharmacology , Dimethylnitrosamine/pharmacokinetics , Ethanol/pharmacology , 1-Propanol/blood , Acetone/blood , Animals , Dimethylnitrosamine/blood , Dimethylnitrosamine/urine , Erythrocebus patas , Ethanol/blood , Male , Premedication
13.
Drug Metab Dispos ; 18(5): 626-31, 1990.
Article in English | MEDLINE | ID: mdl-1981711

ABSTRACT

The disposition, metabolism, and endogenous formation of N-nitrosodimethylamine (NDMA) from nitrosatable precursors was studied in the intact pig and in animals with cannulated hepatic and portal veins and catheterized bile ducts. Rates of disappearance of NDMA from peripheral venous and arterial blood after iv injections were virtually identical and the compound appeared in bile after a lag time of about 1 hr, with a subsequent decline in biliary concentration at about the same rate as in circulating blood. Measurements of NDMA in portal and hepatic vein blood after oral doses of 10, 1.0 and 0.1 mg/kg, respectively, showed progressively greater hepatic extraction with levels in the hepatic vein approaching the limits of detection after the lowest dose. Both halothane and ethanol virtually abolished the hepatic extraction of NDMA, presumably due to their known inhibitory action on its metabolism in the liver. Endogenous formation of NDMA and N-nitrosomorpholine after oral doses of the amines plus nitrite was demonstrated by their detection and measurement in the portal vein blood. Morpholine was nitrosated more effectively than dimethylamine and inhibited the nitrosation of the latter when the two amines were given together. NDMA was found in the portal blood after sequential oral administration of aminopyrine and nitrite, the concentration being considerably greater after fasting for 24 hr than after a 2-hr fast when much food was present in the stomach.


Subject(s)
Dimethylnitrosamine/metabolism , Liver/metabolism , Aminopyrine/metabolism , Animals , Chromatography, Gas , Dimethylnitrosamine/pharmacokinetics , Ethanol/pharmacology , Halothane/pharmacology , In Vitro Techniques , Morpholines/pharmacology , Nitrosamines/metabolism , Swine , Tissue Distribution
14.
Cancer Res ; 50(14): 4366-70, 1990 Jul 15.
Article in English | MEDLINE | ID: mdl-2364390

ABSTRACT

The pharmacokinetics of N-nitrosodimethylamine was studied in patas monkeys following i.v. doses of 0.5, 1.0, and 5.0 mg/kg and a p.o. dose of 1.0 mg/kg, and in Swiss mice at i.v. doses of 1.0 and 2.0 mg/kg. In the patas monkey the pharmacokinetics was linear over the i.v. dose range studied. The mean clearance (Cl), steady-state volume of distribution (Vss), mean residence time, and elimination half-life (t 1/2) were 103.3 +/- 26.7 (SD) ml/min, 3061 +/- 821 ml, 30.8 +/- 10.8 min, and 21.1 +/- 8.5 min, respectively. Assuming that the pharmacokinetics was linear at the p.o. dose used, the p.o. bioavailability of N-nitrosodimethylamine in the monkey was 49%. The pharmacokinetics was also linear in mice, and the average Cl, Vss, mean residence time, and t 1/2 were 3.81 ml/min, 21.0 ml, 5.5 min, and 11.9 min, respectively. These data and data for rats, hamsters, rabbits, dogs, and pigs taken from the literature were used to scale Cl and Vss to body weight using the allometric equation. The resulting equation for Cl was Cl = 49.7B0.998 and the equation for Vss was Vss = 748B1.05 where B is body weight in kg. The fit of the data to the equation was excellent in both cases. Using these equations and assuming a body weight of 70 kg for humans, the Cl and Vss for N-nitrosodimethylamine in humans are estimated to be 3450 ml/min and 64,800 ml, respectively.


Subject(s)
Dimethylnitrosamine/pharmacokinetics , Animals , Cricetinae , Dogs , Erythrocebus patas , Half-Life , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Rabbits , Rats , Species Specificity , Swine
15.
Carcinogenesis ; 11(3): 411-7, 1990 Mar.
Article in English | MEDLINE | ID: mdl-2311184

ABSTRACT

The metabolism of the carcinogenic antitumor drug procarbazine (PCZ) is complex with the ultimate production, among other metabolites, of a methyldiazonium ion which is also the ultimate carcinogenic species of the DNA-methylating N-nitroso compounds including N-methylnitrosourea (MNU). This suggests a similar mechanism of carcinogenic action. Following a single oral dose of [14C]PCZ (50 mg/rat) to 50 day old female Sprague-Dawley rats under the reported conditions of mammary gland carcinogenicity, the DNA adducts 7-methylguanine (7-meG) and O6-methylguanine (O6-meG) were determined in target (mammary gland) and non-target organs. The degree of DNA methylation was similar in all the organs considered. In the mammary gland, lung, spleen, small intestine and stomach the O6-meG/7-meG ratio was close to 0.11. At a lower dose of PCZ (26 mg/rat), the levels of 7-meG in the tissues were 40-60% of those produced by the higher dose. Eighty percent of the rats given the higher dose versus 37% of those given the lower dose developed mammary tumors after 20 weeks. With the higher dose of MNU (50 mg/kg body wt) DNA methylation was more or less uniform in all the organs including the mammary gland, with slightly greater yields in the liver. At a lower MNU dose (25 mg/kg) the levels of 7-meG were 40-48% of those produced by the higher dose. Fifty seven percent of the rats given the higher dose versus 21% of the animals given the lower dose developed mammary gland tumors after 20 weeks. On a mol/kg body wt basis, PCZ was approximately 5-times less active than MNU in the production of 7-meG in mammary gland but only approximately 2-times less active than MNU in the production of mammary gland tumors. The O6-alkylguanine-DNA alkyltransferase (AGT) levels in the liver, kidney, spleen and lung of PCZ or MNU treated rats were approximately 9-28% (expressed relative to protein content) and 10-33% (expressed relative to homogenate DNA content) of those in the corresponding organs of the saline-treated controls. However, the AGT levels of the mammary gland and brain were in the range of 45-61% (expressed relative to protein content) and 39-54% (expressed relative to homogenate DNA content) of those of the saline-treated controls. Also the mammary gland of the 50 day old female rats has the lowest AGT activity (expressed relative to DNA content).(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
DNA/metabolism , Mammary Neoplasms, Experimental/metabolism , Methylnitrosourea/toxicity , Methyltransferases/analysis , Procarbazine/toxicity , Animals , Female , Mammary Neoplasms, Experimental/chemically induced , Methylation , O(6)-Methylguanine-DNA Methyltransferase , Rats , Rats, Inbred Strains
16.
Nutr Cancer ; 14(1): 57-67, 1990.
Article in English | MEDLINE | ID: mdl-2142277

ABSTRACT

Because long-term oral administration of the adrenal steroid dehydroepiandrosterone (DHEA) has previously been shown to inhibit the development of spontaneous breast cancer and chemically induced lung, colon, skin, and liver tumors in various mouse and rat strains, the effect of DHEA on the development of rat kidney tumors by a single dose of 30 mg/kg dimethylnitrosamine (DMN) was tested. DHEA was administered in the diet for a 26-week period commencing 2 weeks after DMN treatment. DHEA administration caused a reduction in body weight gain in accordance with its known antiobesity activity. However, it did not exert any inhibitory effect on either renal mesenchymal or cortical epithelial tumor induction by DMN, nor did it alter the average survival time. There was a statistically significant increase in the incidence of renal adenocarcinomas in the DHEA-treated group but not of renal adenomas. The results were discussed in relation to the mesodermal origin of kidney and the potency of single-dose systems of experimental cancer induction.


Subject(s)
Dehydroepiandrosterone/pharmacology , Diet , Dimethylnitrosamine , Kidney Neoplasms/chemically induced , Adenocarcinoma/chemically induced , Adenocarcinoma/mortality , Adenoma/chemically induced , Adenoma/mortality , Animals , Body Weight/drug effects , Cocarcinogenesis , Dimethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Kidney Neoplasms/mortality , Male , Organ Size/drug effects , Rats , Rats, Inbred Strains
17.
Biochem J ; 262(3): 985-8, 1989 Sep 15.
Article in English | MEDLINE | ID: mdl-2531577

ABSTRACT

The influence of short-term treatment with dehydroepiandrosterone (DHEA), a naturally occurring adrenal steroid, on hepatic metabolism and macromolecular interactions of the hepatocarcinogen dimethylnitrosamine (NDMA) was investigated in male Sprague-Dawley rats. Liver weight, total tissue protein (P less than 0.05), microsomal and cytosolic proteins and cytochrome P-450 (P less than 0.001) were all significantly increased in rats treated orally with DHEA (300 mg/kg body wt., suspended in 1.0 ml of sesame oil). The hepatic DNA content was not altered, however. Methylation of DNA by NDMA was reduced significantly in DHEA-treated rats (P less than 0.05). The binding of [14C]NDMA to hepatic proteins was greater in DHEA-treated rats. The results suggest that short-term treatment of rats with DHEA enhances the binding of NDMA-derived metabolites to hepatic proteins, resulting in the protection of DNA from the damaging effects of NDMA.


Subject(s)
Dehydroepiandrosterone/pharmacology , Dimethylnitrosamine/metabolism , Liver/metabolism , Animals , Cytochrome P-450 Enzyme System/biosynthesis , DNA/metabolism , Male , Microsomes, Liver/metabolism , Organ Size , Protein Binding/drug effects , Protein Biosynthesis , Rats , Rats, Inbred Strains
18.
Drug Chem Toxicol ; 12(3-4): 327-35, 1989.
Article in English | MEDLINE | ID: mdl-2534380

ABSTRACT

The effects of feeding two compounds, dehydroepiandrosterone (DHEA), an adrenal steroid, and clofibrate (CLOF) to rats (which are both hypolipidemic, hepatomegaly inducing and hepatic peroxisome proliferating agents) on the binding of 7,12-dimethylbenz(a)anthracene (DMBA) to hepatic DNA in vivo is compared. Male Sprague Dawley rats (two-three months old) were fed either DHEA or CLOF for 14 days at a dietary level of 0.8%. Control rats were pair fed. An increase in liver weight followed by increases per whole liver in total protein, without much change in DNA content was observed. Subsequently, all the animals were given a single intraperitoneal dose of [3H]DMBA (133 mumol/kg body weight, 102 microCi/rat) in 250 microliters dimethyl sulfoxide. Forty-eight hours later, binding of DMBA to hepatic DNA was determined. The results indicate that DMBA binding to DNA was reduced by 67% in DHEA-fed rats whereas in clofibrate-fed rats it was not significantly different from that of the controls.


Subject(s)
Carrier Proteins/metabolism , Clofibrate/pharmacology , DNA/metabolism , Dehydroepiandrosterone/pharmacology , Liver/metabolism , Animals , Body Weight/drug effects , Male , Organ Size/drug effects , Rats , Rats, Inbred Strains
19.
Carcinogenesis ; 10(5): 953-5, 1989 May.
Article in English | MEDLINE | ID: mdl-2523253

ABSTRACT

The influence of dehydroepiandrosterone (DHEA), an adrenal steroid, on the biotransformation of the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA) in rats has been investigated. Male Sprague-Dawley rats (2-3 months old) were fed DHEA for 14 days at a dietary level of 0.8%. There was an increase in liver weights with increases per whole liver, in total protein, microsomal and cytosolic protein and cytochrome P-450, and cytosolic glutathione transferase activity in DHEA fed rats. DNA content of the liver, however, remained constant. Forty-eight hours after a single i.p. dose of [3H]DMBA (133 mumol/kg body weight, 102 muCi/rat) binding of DMBA derived metabolites to DNA decreased significantly both per unit of DNA (605 versus 194 pmol/mg DNA) as well as per whole liver DNA (25.4 versus 8.5 nmol) in DHEA fed rats. However, a significantly higher amount of DMBA-derived metabolites were bound to total hepatic protein (455 versus 288 nmol) in the steroid fed rats. Microsome mediated binding of DMBA to DNA was 3-fold higher in DHEA fed rats. Excretion of DMBA-derived metabolites in urine was 2-fold higher in DHEA fed rats. The results of this study demonstrate that DHEA inhibits binding of DMBA to hepatic DNA in vivo in spite of the increased metabolic activation of the carcinogen perhaps due to increased detoxification and competitive binding of its active species to proteins.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/metabolism , Dehydroepiandrosterone/pharmacology , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Cytosol/enzymology , DNA/drug effects , DNA/metabolism , Glutathione Transferase/metabolism , Liver/drug effects , Liver/metabolism , Male , Microsomes, Liver/metabolism , Proteins/metabolism , Rats , Rats, Inbred Strains , Reference Values , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...